Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
Add more filters

Country/Region as subject
Publication year range
1.
PLoS Pathog ; 18(1): e1010200, 2022 01.
Article in English | MEDLINE | ID: mdl-35025968

ABSTRACT

The Epstein-Barr Virus (EBV) is involved in the etiology of multiple hematologic and epithelial human cancers. EBV+ tumors employ multiple immune escape mechanisms, including the recruitment of immunosuppressive regulatory T cells (Treg). Here, we show some EBV+ tumor cells express high levels of the chemokines CCL17 and CCL22 both in vitro and in vivo and that this expression mirrors the expression levels of expression of the EBV LMP1 gene in vitro. Patient samples from lymphoblastic (Hodgkin lymphoma) and epithelial (nasopharyngeal carcinoma; NPC) EBV+ tumors revealed CCL17 and CCL22 expression of both tumor cell-intrinsic and -extrinsic origin, depending on tumor type. NPCs grown as mouse xenografts likewise showed both mechanisms of chemokine production. Single cell RNA-sequencing revealed in vivo tumor cell-intrinsic CCL17 and CCL22 expression combined with expression from infiltrating classical resident and migratory dendritic cells in a CT26 colon cancer mouse tumor engineered to express LMP1. These data suggest that EBV-driven tumors employ dual mechanisms for CCL17 and CCL22 production. Importantly, both in vitro and in vivo Treg migration was effectively blocked by a novel, small molecule antagonist of CCR4, CCR4-351. Antagonism of the CCR4 receptor may thus be an effective means of activating the immune response against a wide spectrum of EBV+ tumors.


Subject(s)
Chemokine CCL17/immunology , Chemokine CCL22/immunology , Epstein-Barr Virus Infections/immunology , Neoplasms/immunology , Neoplasms/virology , T-Lymphocytes, Regulatory/immunology , Animals , Epstein-Barr Virus Infections/complications , Herpesvirus 4, Human , Heterografts , Hodgkin Disease/immunology , Hodgkin Disease/virology , Humans , Mice , Nasopharyngeal Carcinoma/immunology , Nasopharyngeal Carcinoma/virology , Nasopharyngeal Neoplasms/immunology , Nasopharyngeal Neoplasms/virology
2.
Int J Mol Sci ; 24(6)2023 Mar 09.
Article in English | MEDLINE | ID: mdl-36982351

ABSTRACT

Damage-associated molecular patterns (DAMPs) are endogenous molecules released from the necrotic cells dying after exposure to various stressors. After binding to their receptors, they can stimulate various signaling pathways in target cells. DAMPs are especially abundant in the microenvironment of malignant tumors and are suspected to influence the behavior of malignant and stromal cells in multiple ways often resulting in promotion of cell proliferation, migration, invasion, and metastasis, as well as increased immune evasion. This review will start with a reminder of the main features of cell necrosis, which will be compared to other forms of cell death. Then we will summarize the various methods used to assess tumor necrosis in clinical practice including medical imaging, histopathological examination, and/or biological assays. We will also consider the importance of necrosis as a prognostic factor. Then the focus will be on the DAMPs and their role in the tumor microenvironment (TME). We will address not only their interactions with the malignant cells, frequently leading to cancer progression, but also with the immune cells and their contribution to immunosuppression. Finally, we will emphasize the role of DAMPs released by necrotic cells in the activation of Toll-like receptors (TLRs) and the possible contributions of TLRs to tumor development. This last point is very important for the future of cancer therapeutics since there are attempts to use TLR artificial ligands for cancer therapeutics.


Subject(s)
Neoplasms , Tumor Microenvironment , Humans , Neoplasms/metabolism , Necrosis , Toll-Like Receptors/metabolism , Signal Transduction
3.
Int J Mol Sci ; 24(20)2023 Oct 17.
Article in English | MEDLINE | ID: mdl-37894949

ABSTRACT

Tumor necrosis is a recurrent characteristic of head and neck squamous cell carcinomas (HNSCCs). There is a need for more investigations on the influence of biomolecules released by these necrotic foci in the HNSCC tumor microenvironment. It is suspected that a fraction of the biomolecules released by necrotic cells are damage-associated molecular patterns (DAMPs), which are known to be natural endogenous ligands of Toll-like receptors (TLRs), including, among others, proteins and nucleic acids. However, there has been no direct demonstration that biomolecules released by HNSCC necrotic cells can activate TLRs. Our aim was to investigate whether some of these molecules could behave as agonists of the TLR3, either in vitro or in vivo. We chose a functional approach based on reporter cell exhibiting artificial TLR3 expression and downstream release of secreted alkaline phosphatase. The production of biomolecules activating TLR3 was first investigated in vitro using three HNSCC cell lines subjected to various pronecrotic stimuli (external irradiation, serum starvation, hypoxia and oxidative stress). TLR3 agonists were also investigated in necrotic tumor fluids from five oral cancer patients and three mouse tumor grafts. The release of biomolecules activating TLR3 was demonstrated for all three HNSCC cell lines. External irradiation was the most consistently efficient stimulus, and corresponding TLR3 agonists were conveyed in extracellular vesicles. TLR3-stimulating activity was detected in the fluids from all five patients and three mouse tumor grafts. In most cases, this activity was greatly reduced by RNAse pretreatment or TLR3 blocking antibodies. Our data indicate that TLR3 agonists are consistently present in necrotic fluids from HNSCC cells and mainly made of dsRNA fragments. These endogenous agonists may induce TLR3, which might lead to a protumorigenic effect. Regarding methodological aspects, our study demonstrates that direct investigations-including functional testing-can be performed on necrotic fluids from patient tumors.


Subject(s)
Head and Neck Neoplasms , Toll-Like Receptor 3 , Animals , Humans , Mice , Necrosis/metabolism , Squamous Cell Carcinoma of Head and Neck , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 9 , Toll-Like Receptors , Tumor Microenvironment
4.
Cancer Immunol Immunother ; 71(1): 13-24, 2022 Jan.
Article in English | MEDLINE | ID: mdl-33993319

ABSTRACT

Around 30-50% of classical Hodgkin lymphoma (cHL) cases in immunocompetent individuals from industrialized countries are associated with the B-lymphotropic Epstein-Barr virus (EBV). Although natural killer (NK) cells exhibit anti-viral and anti-tumoral functions, virtually nothing is known about quantitative and qualitative differences in NK cells in patients with EBV+ cHL vs. EBV- cHL. Here, we prospectively investigated 36 cHL patients without known immune suppression or overt immunodeficiency at diagnosis. All 10 EBV+ cHL patients and 25 out 26 EBV- cHL were seropositive for EBV antibodies, and EBV+ cHL patients presented with higher plasma EBV DNA levels compared to EBV- cHL patients. We show that the CD56dim CD16+ NK cell subset was decreased in frequency in EBV+ cHL patients compared to EBV- cHL patients. This quantitative deficiency translates into an impaired CD56dim NK cell mediated degranulation toward rituximab-coated HLA class 1 negative lymphoblastoid cells in EBV+ compared to EBV- cHL patients. We finally observed a trend to a decrease in the rituximab-associated degranulation and ADCC of in vitro expanded NK cells of EBV+ cHL compared to healthy controls. Our findings may impact on the design of adjunctive treatment targeting antibody-dependent cellular cytotoxicity in EBV+ cHL.


Subject(s)
Antibodies/immunology , CD56 Antigen/biosynthesis , Hodgkin Disease/metabolism , Hodgkin Disease/therapy , Receptors, IgG/biosynthesis , Adult , Aged , Antineoplastic Agents/pharmacology , Epstein-Barr Virus Infections/complications , Female , GPI-Linked Proteins/biosynthesis , Herpesvirus 4, Human/metabolism , Hodgkin Disease/complications , Humans , Immunotherapy , In Vitro Techniques , Killer Cells, Natural/metabolism , Leukocytes, Mononuclear/cytology , Lymphocytes/metabolism , Lysosomal-Associated Membrane Protein 1/biosynthesis , Male , Middle Aged , Phenotype , Prospective Studies , Rituximab/pharmacology
5.
Cancer Immunol Immunother ; 70(2): 323-336, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32737537

ABSTRACT

BACKGROUND: Nasopharyngeal carcinoma (NPC) is a highly malignant epithelial cancer linked to Epstein-Barr virus (EBV) infection. Tumors are characterized by a lymphomononuclear infiltrate and the number of natural killer (NK) cells in tumors appears to be of prognostic significance. Standard treatment for NPC in adolescents and young adults consists of induction chemotherapy followed by radiochemotherapy. Though survival rates are above 80%, the majority of patients suffer from long-term side-effects, mainly related to radiotherapy. The addition of immunotherapy to induction chemotherapy could improve tumor response. METHODS: We have investigated the killing of NPC cells by NK cells in the context of chemotherapy, using a panel of three nasopharyngeal carcinoma cell lines and a patient-derived xenograft. Cytotoxicity was measured using the calcein-release assay, while the contribution of different checkpoints and signaling pathways to killing was studied by siRNA-mediated gene silencing and chemical inhibitors. RESULTS: Chemotherapeutics cisplatin, 5-fluorouracil and gemcitabine sensitized NPC cells to killing by NK cells. Chemotherapeutics led to upregulation of PD-1 in NK cells and PD-L1 in NPC cells via NF-κB. Inhibition of the PD-L1/PD-1 checkpoint by an anti-PD-1 antibody or siRNA increased NK-cell cytotoxicity towards NPC cells. CONCLUSION: The addition of an anti-PD-1 antibody to chemotherapy in patients with NPC could increase the efficacy of induction chemotherapy. If confirmed in a clinical trial, more efficient induction therapy could allow the dose of radiotherapy to be reduced and thereby diminish severe late effects of such therapy.


Subject(s)
Immunotherapy/methods , Killer Cells, Natural/immunology , Nasopharyngeal Carcinoma/genetics , Programmed Cell Death 1 Receptor/therapeutic use , Animals , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Programmed Cell Death 1 Receptor/metabolism , Transfection , Up-Regulation
6.
Proc Natl Acad Sci U S A ; 114(36): 9683-9688, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28831010

ABSTRACT

Nasopharyngeal carcinoma (NPC) most frequently occurs in southern China and southeast Asia. Epidemiology studies link NPC to genetic predisposition, Epstein-Barr virus (EBV) infection, and environmental factors. Genetic studies indicate that mutations in chromatin-modifying enzymes are the most frequent genetic alterations in NPC. Here, we used H3K27ac chromatin immune precipitation followed by deep sequencing (ChIP-seq) to define the NPC epigenome in primary NPC biopsies, NPC xenografts, and an NPC cell line, and compared them to immortalized normal nasopharyngeal or oral epithelial cells. We identified NPC-specific enhancers and found these enhancers were enriched with nuclear factor κB (NF-κB), IFN-responsive factor 1 (IRF1) and IRF2, and ETS family members ETS1 motifs. Normal cell-specific enhancers were enriched with basic leucine zipper family members and TP53 motifs. NPC super-enhancers with extraordinarily broad and high H3K27ac signals were also identified, and they were linked to genes important for oncogenesis including ETV6. ETV6 was also highly expressed in NPC biopsies by immunohistochemistry. High ETV6 expression correlated with a poor prognosis. Furthermore, we defined the EBV episome epigenetic landscapes in primary NPC tissue.


Subject(s)
Carcinoma/genetics , Enhancer Elements, Genetic , Nasopharyngeal Neoplasms/genetics , Proto-Oncogene Proteins c-ets/genetics , Repressor Proteins/genetics , Adolescent , Adult , Aged , Animals , Azepines/pharmacology , Carcinoma/etiology , Carcinoma/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Enhancer Elements, Genetic/drug effects , Epigenesis, Genetic , Epstein-Barr Virus Infections/complications , Female , Genome, Viral , Heterografts , High-Throughput Nucleotide Sequencing , Histone Code/genetics , Humans , Immunohistochemistry , Male , Mice , Mice, Nude , Middle Aged , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/etiology , Nasopharyngeal Neoplasms/metabolism , Nuclear Proteins/antagonists & inhibitors , Prognosis , Proto-Oncogene Proteins c-ets/metabolism , Repressor Proteins/metabolism , Transcription Factors/antagonists & inhibitors , Triazoles/pharmacology , Young Adult , ETS Translocation Variant 6 Protein
7.
Cancer Immunol Immunother ; 68(8): 1317-1329, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31312900

ABSTRACT

BACKGROUND: Nasopharyngeal carcinoma (NPC) is an EBV-associated neoplasm occurring endemically in Southeast Asia and sporadically all over the world. In children and adolescents, high cure rates have been obtained using chemotherapy, radiochemotherapy and maintenance therapy with interferon beta (IFNß). The mechanism by which IFNß contributes to a low systemic relapse rate has not yet been fully revealed. PATIENTS AND METHODS: NK cells and serum samples from two patients with NPC were analyzed before and at different time points during IFNß therapy, for assessment of TRAIL expression and NK cell cytotoxicity. Cytotoxicity was measured using the calcein release assay and the contribution of different death effector pathways was analyzed using specific inhibitors. RESULTS: Treatment with IFNß induced TRAIL expression on patients' NK cells and increased their cytotoxicity against NPC targets in vitro. NK cell-mediated cytotoxicity was predominately mediated via TRAIL. IFNß also induced the production of soluble TRAIL (sTRAIL) by NK cells and its release upon contact with NPC cells. IFNß treatment increased serum levels of sTRAIL in patients. Moreover, sTRAIL concentrated from patients' serum samples induced apoptosis ex vivo in NPC cells from a patient-derived xenograft. CONCLUSION: Increased cytotoxicity of NK cells against NPC cells and increased serum levels of biologically active TRAIL in patients treated with IFNß could be a means to eliminate micrometastatic disease and explain the low systemic relapse rate in this patient group.


Subject(s)
Epstein-Barr Virus Infections/immunology , Herpesvirus 4, Human/physiology , Immunotherapy/methods , Interferon-beta/therapeutic use , Killer Cells, Natural/immunology , Nasopharyngeal Carcinoma/therapy , TNF-Related Apoptosis-Inducing Ligand/metabolism , Adolescent , Animals , Apoptosis , Cell Line, Tumor , Child , Cytotoxicity, Immunologic , Female , Humans , Mice , Mice, Nude , Nasopharyngeal Carcinoma/immunology , Neoplasm Recurrence, Local , Treatment Outcome , Xenograft Model Antitumor Assays
8.
PLoS Pathog ; 13(7): e1006503, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28732079

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are expanded in tumor microenvironments, including that of Epstein-Barr virus (EBV)-associated nasopharyngeal carcinoma (NPC). The link between MDSC expansion and EBV infection in NPC is unclear. Here, we show that EBV latent membrane protein 1 (LMP1) promotes MDSC expansion in the tumor microenvironment by promoting extra-mitochondrial glycolysis in malignant cells, which is a scenario for immune escape initially suggested by the frequent, concomitant detection of abundant LMP1, glucose transporter 1 (GLUT1) and CD33+ MDSCs in tumor sections. The full process has been reconstituted in vitro. LMP1 promotes the expression of multiple glycolytic genes, including GLUT1. This metabolic reprogramming results in increased expression of the Nod-like receptor family protein 3 (NLRP3) inflammasome, COX-2 and P-p65 and, consequently, increased production of IL-1ß, IL-6 and GM-CSF. Finally, these changes in the environment of malignant cells result in enhanced NPC-derived MDSC induction. One key step is the physical interaction of LMP1 with GLUT1 to stabilize the GLUT1 protein by blocking its K48-ubiquitination and p62-dependent autolysosomal degradation. This work indicates that LMP1-mediated glycolysis regulates IL-1ß, IL-6 and GM-CSF production through the NLRP3 inflammasome, COX-2 and P-p65 signaling pathways to enhance tumor-associated MDSC expansion, which leads to tumor immunosuppression in NPC.


Subject(s)
Carcinoma/physiopathology , Epstein-Barr Virus Infections/physiopathology , Herpesvirus 4, Human/metabolism , Myeloid-Derived Suppressor Cells/cytology , Nasopharyngeal Neoplasms/physiopathology , Viral Matrix Proteins/metabolism , Carcinoma/genetics , Carcinoma/metabolism , Carcinoma/virology , Cell Line, Tumor , Cell Proliferation , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Infections/virology , Gene Expression Regulation, Neoplastic , Glycolysis , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Herpesvirus 4, Human/genetics , Host-Pathogen Interactions , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Myeloid-Derived Suppressor Cells/metabolism , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/virology , Signal Transduction , Viral Matrix Proteins/genetics
9.
Protein Expr Purif ; 162: 44-50, 2019 10.
Article in English | MEDLINE | ID: mdl-31145974

ABSTRACT

BALF0/1 is a putative Epstein-Barr virus (EBV) protein that has been described as a modulator of apoptosis. So far, the lack of specific immunological reagents impaired the detection of native BALF0/1 in EBV-infected cells. This study describes the expression and purification of a truncated form of BALF0/1 (tBALF0) using a heterologous bacterial expression system. tBALF0 was further used as an antigen in an indirect Enzyme-linked Immunosorbent Assay (ELISA) that unraveled the presence of low titer IgGs to BALF0/1 during primary (10.0%) and past (13.3%) EBV infection. Conversely high-titer IgGs to BALF0/1 were detected in 33.3% of nasopharyngeal carcinoma (NPC) patients suggesting that BALF0/1 and/or humoral response against it may contribute to NPC pathogenesis.


Subject(s)
Antibodies, Viral/blood , Epstein-Barr Virus Infections/blood , Herpesvirus 4, Human/immunology , Immunoglobulin G/blood , Nasopharyngeal Carcinoma/blood , Viral Proteins/immunology , Antibodies, Viral/immunology , Enzyme-Linked Immunosorbent Assay , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/genetics , Humans , Immunity, Humoral , Immunoglobulin G/immunology , Nasopharyngeal Carcinoma/virology , Viral Proteins/genetics
10.
J Pathol ; 244(4): 394-407, 2018 04.
Article in English | MEDLINE | ID: mdl-29230817

ABSTRACT

Nasopharyngeal carcinoma (NPC) is a highly invasive epithelial malignancy that is prevalent in southern China and Southeast Asia. It is consistently associated with latent Epstein-Barr virus (EBV) infection. In NPC, miR-BARTs, the EBV-encoded miRNAs derived from BamH1-A rightward transcripts, are abundantly expressed and contribute to cancer development by targeting various cellular and viral genes. In this study, we establish a comprehensive transcriptional profile of EBV-encoded miRNAs in a panel of NPC patient-derived xenografts and an EBV-positive NPC cell line by small RNA sequencing. Among the 40 miR-BARTs, predominant expression of 22 miRNAs was consistently detected in these tumors. Among the abundantly expressed EBV-miRNAs, BART5-5p, BART7-3p, BART9-3p, and BART14-3p could negatively regulate the expression of a key DNA double-strand break (DSB) repair gene, ataxia telangiectasia mutated (ATM), by binding to multiple sites on its 3'-UTR. Notably, the expression of these four miR-BARTs represented more than 10% of all EBV-encoded miRNAs in tumor cells, while downregulation of ATM expression was commonly detected in all of our tested sequenced samples. In addition, downregulation of ATM was also observed in primary NPC tissues in both qRT-PCR (16 NP and 45 NPC cases) and immunohistochemical staining (35 NP and 46 NPC cases) analysis. Modulation of ATM expression by BART5-5p, BART7-3p, BART9-3p, and BART14-3p was demonstrated in the transient transfection assays. These findings suggest that EBV uses miRNA machinery as a key mechanism to control the ATM signaling pathway in NPC cells. By suppressing these endogenous miR-BARTs in EBV-positive NPC cells, we further demonstrated the novel function of miR-BARTs in inhibiting Zta-induced lytic reactivation. These findings imply that the four viral miRNAs work co-operatively to modulate ATM activity in response to DNA damage and to maintain viral latency, contributing to the tumorigenesis of NPC. © 2017 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/genetics , MicroRNAs/genetics , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Neoplasms/genetics , RNA, Viral/genetics , 3' Untranslated Regions , Animals , Ataxia Telangiectasia Mutated Proteins/biosynthesis , Binding Sites , Cell Line, Tumor , DNA Damage , Enzyme Repression , Epstein-Barr Virus Infections/diagnosis , Female , Gene Expression Regulation, Neoplastic , Heterografts , Host-Pathogen Interactions , Humans , Male , Mice, Nude , Middle Aged , Nasopharyngeal Carcinoma/enzymology , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Carcinoma/virology , Nasopharyngeal Neoplasms/enzymology , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/virology , Transcriptome , Virus Latency
11.
Lab Invest ; 98(8): 1093-1104, 2018 08.
Article in English | MEDLINE | ID: mdl-29769697

ABSTRACT

Epstein-Barr virus (EBV) infects more than 90% of the adult human population. Undifferentiated nasopharyngeal carcinoma (NPC) is common in Southeast Asia, with a particularly high incidence among southern Chinese. The EBV genome can be detected in practically all cancer cells in undifferentiated NPC. The role of EBV in pathogenesis of undifferentiated NPC remains elusive. NPC cell lines are known to be difficult to establish in culture. The EBV+ve NPC cell lines, even if established in culture, rapidly lost their EBV episomes upon prolonged propagation. At present, the C666-1 NPC cell line, which is defective in lytic EBV reactivation, is the only EBV+ve NPC cell line available for NPC and EBV research. The need to establish new and representative NPC cell lines is eminent for NPC and EBV research. In this study, we report the use of the Rho-associated kinase inhibitor (Y-27632) has facilitated the establishment of a new EBV+ve NPC cell line from an earlier established NPC xenograft, C17. The C17 cell line was tumorigenic in immune-deficient mice (NOD/SCID). It retained the EBV episomes and could be induced to undergo productive lytic reactivation of EBV to generate infectious virus particles. The C17 cell line represents a new investigative tool for NPC and EBV studies. The ability of C17 to undergo lytic reactivation is unique and opens up the opportunity to examine regulation of latent and lytic infection of EBV and their contributions to NPC pathogenesis.


Subject(s)
Epstein-Barr Virus Infections/pathology , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Neoplasms/pathology , Virus Activation , Animals , Cell Line, Tumor , Epstein-Barr Virus Infections/virology , Genome, Viral/genetics , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/physiology , Host-Pathogen Interactions , Humans , Karyotyping , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Nasopharyngeal Carcinoma/virology , Nasopharyngeal Neoplasms/virology , Transplantation, Heterologous , Tumor Burden
12.
Mol Carcinog ; 56(4): 1214-1226, 2017 04.
Article in English | MEDLINE | ID: mdl-27805282

ABSTRACT

Toll-like receptor 3 (TLR3) has a dual role in cancer; its activation can trigger apoptosis as well as stimulate cancer cell survival, proliferation, and progression. We have shown here that TLR3 activation can induce metabolic reprogramming in a pharyngeal cancer cell line, leading to increased aerobic glycolysis, cell migration, elevated levels of reactive oxidative species (ROS), and decreased anti-oxidative response. Key proteins in these signaling pathways are heterogeneous nuclear ribonucleoprotein A1 (hnRNPA1), pyruvate kinase M2 (PKM2), and CD44 variants, which were over-expressed after TLR3 stimulation. TLR3 activation also induced upregulation of different genes involved in cancer progression (VEGF, MMP9, uPAR) and enzymes involved in glycolytic pathway. Most of the observed effects were Myc-dependent; however, some of them were also connected with MAPK and HIF signaling pathways. Since TLR3 agonists are being investigated as potential novel cancer therapy adjuvants and apoptosis inducers, alone or in combination with other therapeutic options, data presented here suggest extreme caution before their introduction into clinical practice. The fact that TLR3 ligands [poly(I:C) and poly(A:U)] can also aid cancer survival and progression, through induction of metabolic reprogramming, emphasizes the need to investigate this particular topic. Our data suggest that the combination of TLR3 ligands with Myc or MAPK inhibitors may be a way to neutralize their undesirable effects while enhancing their anti-tumor effect. © 2016 Wiley Periodicals, Inc.


Subject(s)
Hypoxia-Inducible Factor 1/metabolism , MAP Kinase Signaling System , Pharyngeal Neoplasms/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , Toll-Like Receptor 3/metabolism , Cell Line, Tumor , Cell Movement , Glycolysis , Humans , Oxidative Stress , Pharyngeal Neoplasms/pathology , Pharynx/metabolism , Pharynx/pathology , Poly I-C/metabolism , Reactive Oxygen Species/metabolism
13.
J Biol Chem ; 290(27): 16797-811, 2015 Jul 03.
Article in English | MEDLINE | ID: mdl-25947381

ABSTRACT

Galectin-9 (gal-9) is a multifunctional ß-galactoside-binding lectin, frequently released in the extracellular medium, where it acts as a pleiotropic immune modulator. Despite its overall immunosuppressive effects, a recent study has reported bimodal action of gal-9 on human resting blood T cells with apoptosis occurring in the majority of them, followed by a wave of activation and expansion of Th1 cells in the surviving population. Our knowledge of the signaling events triggered by exogenous gal-9 in T cells remains limited. One of these events is cytosolic calcium (Ca(2+)) release reported in some murine and human T cells. The aim of this study was to investigate the contribution of Ca(2+) mobilization to apoptotic and nonapoptotic effects of exogenous gal-9 in human T cells. We found that the T cell receptor (TCR)-CD3 complex and the Lck kinase were required for Ca(2+) mobilization but not for apoptosis induction in Jurkat cells. These data were confirmed in human CD4(+) T cells from peripheral blood as follows: a specific Lck chemical inhibitor abrogated Ca(2+) mobilization but not apoptosis induction. Moreover, Lck activity was also required for the production of Th1-type cytokines, i.e. interleukin-2 and interferon-γ, which resulted from gal-9 stimulation in peripheral CD4(+) T cells. These findings indicate that gal-9 acts on T cells by two distinct pathways as follows: one mimicking antigen-specific activation of the TCR with a mandatory contribution of proximal elements of the TCR complex, especially Lck, and another resulting in apoptosis that is independent of this complex.


Subject(s)
Apoptosis , CD3 Complex/metabolism , Galectins/metabolism , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism , CD3 Complex/genetics , Calcium/metabolism , Cytokines/genetics , Cytokines/metabolism , Galectins/genetics , Humans , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/cytology
14.
Thorax ; 70(6): 537-42, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25855608

ABSTRACT

BACKGROUND: Sarcoidosis is a rare lung disease in children. The aim of the present study was to provide update information on disease presentation and progression, patient management and prognosis factors in a cohort of children with lung sarcoidosis. METHODS: With the network of the French Reference Centre for Rare Lung Diseases (RespiRare), we collected information on a large cohort of paediatric thoracic sarcoidosis to provide information on disease presentation, management and outcome. RESULTS: Forty-one patients were included with a median age at diagnosis of 11.8  years (1.1-15.8), mostly from Afro-Caribbean and Sub-Saharan origin. At diagnosis, 85% presented with a multi-organic disease, and no major differences were found regarding disease severity between the patients diagnosed before or after 10 years old. Corticosteroids were the most used treatment, with more intravenous pulses in the youngest patients. The 18-month outcome showed that patients diagnosed before 10 years old were more likely to recover (50% vs 29%), and presented fewer relapses (29% vs 58%). At 4-5 years of follow-up, relapses were mostly observed for patients diagnosed after 10 years old. DISCUSSION: In the included children, mostly of Afro-Caribbean and Sub-Saharan origin, sarcoidosis seems severe, with multi-organic involvement and foreground general symptoms. Common prognosis factors are not suitable in paediatric patients, and a young age at diagnosis does not seem to be associated with a poorer prognosis. The study is ongoing to provide further information on the very-long-term follow-up of paediatric sarcoidosis.


Subject(s)
Black People , Glucocorticoids/therapeutic use , Sarcoidosis, Pulmonary/drug therapy , Sarcoidosis, Pulmonary/ethnology , Adolescent , Africa South of the Sahara/ethnology , Black People/statistics & numerical data , Caribbean Region/ethnology , Child , Child, Preschool , Cohort Studies , Female , Follow-Up Studies , France/epidemiology , Humans , Infant , Infusions, Intravenous , Male , Rare Diseases , Recurrence , Sarcoidosis, Pulmonary/diagnosis , Severity of Illness Index , Treatment Outcome
15.
Mol Cancer ; 13: 184, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-25098679

ABSTRACT

BACKGROUND: As a distinctive type of head and neck cancers, nasopharyngeal carcinoma (NPC) is genesis from the clonal Epstein-Barr virus (EBV)-infected nasopharyngeal epithelial cells accumulated with multiple genetic lesions. Among the recurrent genetic alterations defined, loss of 9p21.3 is the most frequent early event in the tumorigenesis of EBV-associated NPC. In addition to the reported CDKN2A/p16, herein, we elucidated the role of a miRNA, miR-31 within this 9p21.3 region as NPC-associated tumor suppressor. METHODS: The expression and promoter methylation of miR-31 were assessed in a panel of NPC tumor lines and primary tumors. Its in vitro and in vivo tumor suppression function was investigated through the ectopic expression of miR-31 in NPC cells. We also determined the miR-31 targeted genes and its involvement in the growth in NPC. RESULTS: Downregulation of miR-31 expression was detected in almost all NPC cell line, patient-derived xenografts (PDXs) and primary tumors. Both homozygous deletion and promoter hypermethylation were shown to be major mechanisms for miR-31 silencing in this cancer. Strikingly, loss of miR-31 was also obviously observed in the dysplastic lesions of nasopharynx. Restoration of miR-31 in C666-1 cells inhibited the cell proliferation, colony-forming and migratory capacities. Dramatic reduction of in vitro anchorage-independent growth and in vivo tumorigenic potential were demonstrated in the stable clones expressing miR-31. Furthermore, we proved that miR-31 suppressed the NPC cell growth via targeting FIH1 and MCM2. CONCLUSIONS: The findings provide strong evidence to support miR-31 as a new NPC-associated tumor suppressor on 9p21.3 region. The inactivation of miR-31 may contribute to the early development of NPC.


Subject(s)
Carcinogenesis/pathology , Herpesvirus 4, Human/physiology , MicroRNAs/metabolism , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/virology , Carcinogenesis/genetics , Carcinoma , Cell Movement/genetics , Cell Proliferation , Cell Survival/genetics , Comparative Genomic Hybridization , DNA Methylation/genetics , Down-Regulation/genetics , Gene Deletion , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Homozygote , Humans , MicroRNAs/genetics , Minichromosome Maintenance Complex Component 2/metabolism , Mixed Function Oxygenases/metabolism , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/pathology , Phosphorylation , Promoter Regions, Genetic , Repressor Proteins/metabolism , Tumor Suppressor Protein p53/metabolism
16.
J Pathol ; 231(3): 311-22, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23868181

ABSTRACT

As a distinct type of head and neck cancer, non-keratinizing nasopharyngeal carcinoma (NPC) is closely associated with EBV infection and massive lymphoid infiltration. The unique histological features suggest that local inflammation plays an important role in NPC tumourigenesis. We comprehensively characterized NF-κB signalling, a key inflammatory pathway which might contribute to the tumourigenesis of this EBV-associated cancer. By EMSA, western blotting, and immunohistochemical staining, constitutive activation of distinct NF-κB complexes, either p50/p50/Bcl3 or p50/RelB, was found in almost all EBV-positive NPC tumours. siRNA or chemical inhibition of NF-κB signalling significantly inhibited the growth of EBV-positive NPC cells C666-1. Gene expression profiling identified a number of NF-κB target genes involved in cell proliferation, apoptosis, immune response, and transcription. We further confirmed that p50 signals modulate the expression of multiple oncogenes (MYB, BCL2), chemokines, and chemokine receptors (CXCL9, CXCL10, CX3CL1, and CCL20). The findings support a crucial role of these constitutively activated NF-κB signals in NPC tumourigenesis and local inflammation. In addition to expression of the viral oncoprotein LMP1, genetic alteration of several NF-κB regulators (eg TRAF3, TRAF2, NFKBIA, A20) also contributes to the aberrant NF-κB activation in EBV-associated NPC. Except for LMP1-expressing C15 cells, all NPC tumour lines harbour at least one of these genetic alterations. Importantly, missense mutations of TRAF3, TRAF2, and A20 were also detected in 3/33 (9.1%) primary tumours. Taken together with the reported LTBR amplification in 7.3% of primary NPCs, genetic alterations in NF-κB pathways occurred in at least 16% of cases of this cancer. The findings indicate that distinct NF-κB signals are constitutively activated in EBV-positive NPC cells by either multiple genetic changes or EBV latent genes.


Subject(s)
Cell Transformation, Viral , Epstein-Barr Virus Infections/metabolism , NF-kappa B/metabolism , Nasopharyngeal Neoplasms/metabolism , Signal Transduction , Antineoplastic Agents/pharmacology , Apoptosis , B-Cell Lymphoma 3 Protein , Base Sequence , Carcinoma , Cell Line, Tumor , Cell Proliferation , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/virology , Gene Expression Regulation, Neoplastic , Humans , Inflammation Mediators/metabolism , Molecular Sequence Data , Mutation , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , NF-kappa B p50 Subunit/metabolism , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/immunology , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/virology , Proto-Oncogene Proteins/metabolism , RNA Interference , Signal Transduction/drug effects , Time Factors , Transcription Factor RelB/metabolism , Transcription Factors/metabolism , Transfection
17.
J Pathol ; 231(2): 158-67, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23878065

ABSTRACT

Nasopharyngeal carcinoma (NPC) is a distinct type of head and neck cancer which is prevalent in southern China, south-east Asia and northern Africa. The development and stepwise progression of NPC involves accumulation of multiple gross genetic changes during the clonal expansion of Epstein-Barr virus (EBV)-infected nasopharyngeal epithelial cell population. Here, using paired-end whole-transcriptome sequencing, we discovered a number of chimeric fusion transcripts in a panel of EBV-positive tumour lines. Among these transcripts, a novel fusion of ubiquitin protein ligase E3 component n-recognin 5 (UBR5) on 8q22.3 and zinc finger protein 423 (ZNF423) on 16q12.1, identified from the NPC cell line C666-1, was recurrently detected in 12/144 (8.3%) of primary tumours. The fusion gene contains exon 1 of UBR5 and exons 7-9 of ZNF423 and produces a 94 amino acid chimeric protein including the original C-terminal EBF binding domain (ZF29-30) of ZNF423. Notably, the growth of NPC cells with UBR5-ZNF423 rearrangement is dependent on expression of this fusion protein. Knock-down of UBR5-ZNF423 by fusion-specific siRNA significantly inhibited the cell proliferation and colony-forming ability of C666-1 cells. The transforming ability of UBR5-ZNF423 fusion was also confirmed in NIH3T3 fibroblasts. Constitutive expression of UBR5-ZNF423 in NIH3T3 fibroblasts significantly enhanced its anchorage-independent growth in soft agar and induced tumour formation in a nude mouse model. These findings suggest that expression of UBR5-ZNF423 protein might contribute to the transformation of a subset of NPCs, possibly by altering the activity of EBFs (early B cell factors). Identification of the oncogenic UBR5-ZNF423 provides new potential opportunities for therapeutic intervention in NPC.


Subject(s)
DNA-Binding Proteins/genetics , Nasopharyngeal Neoplasms/genetics , Recombinant Fusion Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Amino Acid Sequence , Animals , Blotting, Western , Carcinoma , Cell Line, Tumor , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/genetics , Female , Humans , In Situ Hybridization, Fluorescence , Male , Mice , Mice, Nude , Middle Aged , Molecular Sequence Data , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/virology , Oncogenes/genetics , Proteins , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transcriptome , Transfection , Transplantation, Heterologous
18.
Semin Cancer Biol ; 22(2): 127-36, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22249142

ABSTRACT

Like other human solid tumors, nasopharyngeal carcinoma (NPC) is a tissue and a systemic disease as much as a cell disease. Tumor cell population in NPC is highly heterogeneous. Heavy infiltration by non-malignant leucocytes results at least in part from the production of abundant inflammatory cytokines by the malignant epithelial cells. There is indirect evidence that interactions between stromal and malignant cells contribute to tumor development. Peripheral blood samples collected from NPC patients contain multiple products derived from the tumor, including cytokines, non-cytokine tumor proteins, tumor exosomes and viral nucleic acids. These products represent a potential source of biomarkers for assessment of tumor aggressiveness, indirect exploration of cellular interactions and monitoring of tumor response to therapeutic agents. Most NPC patients are immunocompetent with evidence of active humoral and cellular immune responses against EBV-antigens at the systemic level. Tumor development is facilitated by local immunosuppressive factors which are not fully understood. Local accumulation of regulatory T-cells is probably one important factor. At least two NPC tumor products are suspected to contribute to their expansion, the cytokine CCL20 and the tumor exosomes carrying galectin 9. In the future, new therapeutic modalities will probably aim at breaking immune tolerance or at blocking cellular interactions critical for tumor growth.


Subject(s)
Cytokines/immunology , Nasopharyngeal Neoplasms/immunology , T-Lymphocytes/immunology , Tumor Microenvironment/immunology , Carcinoma , Chemokine CCL20/immunology , Chemokine CCL20/metabolism , Cytokines/metabolism , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/pathology , Exosomes/immunology , Exosomes/metabolism , Herpesvirus 4, Human/immunology , Humans , Models, Immunological , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/complications , Nasopharyngeal Neoplasms/pathology , T-Lymphocytes/metabolism
19.
Virol J ; 10: 119, 2013 Apr 16.
Article in English | MEDLINE | ID: mdl-23590857

ABSTRACT

BACKGROUND: Because latent Epstein Barr (EBV)-infection is a specific characteristic of malignant nasopharyngeal carcinoma (NPC), various molecules of viral origin are obvious candidate biomarkers in this disease. In a previous study, we could show in a few clinical samples that it was possible to detect a category of EBV microRNAs called miR-BARTs in the plasma of at least a fraction of NPC patients. The first aim of the present study was to investigate the status of circulating miR-BART17-5p (one of the miR-BARTs hereafter called miR-BART17) and EBV DNA in a larger series of NPC plasma samples. The second aim was to determine whether or not circulating miR-BART17 was carried by plasma exosomes. PATIENTS AND METHODS: Plasma samples were collected from 26 NPC patients and 10 control donors, including 9 patients with non-NPC Head and Neck squamous cell carcinoma and one healthy EBV carrier. Concentrations of miR-BART17 and two cellular microRNAs (hsa-miR-16 and -146a) were assessed by real-time quantitative PCR with spike-in normalization and absolute quantification. In addition, for 2 patients, exosome distributions of miR-BART17 and miR-16 were investigated following plasma lipoprotein fractionation by isopycnic density gradient ultrcentrifugation. RESULTS: The miR-BART17 was significantly more abundant in plasma samples from NPC patients compared to non-NPC donors. Above a threshold of 506 copies/mL, detection of miR-BART17 was highly specific for NPC patients (ROC curve analysis: AUC=0.87 with true positive rate = 0.77, false positive rate = 0.10). In this relatively small series, the concentration of plasma miR-BART17 and the plasma EBV DNA load were not correlated. When plasma samples were fractionated, miR-BART17 co-purified with a protein-rich fraction but not with exosomes. CONCLUSIONS: Detection of high concentrations of plasma miR-BART17 is consistent in NPC patients. This parameter is, at least in part, independent of the viral DNA load. Circulating miR-BART17 does not co-purify with exosomes.


Subject(s)
Biomarkers/blood , Herpesvirus 4, Human/genetics , MicroRNAs/blood , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/virology , Plasma/chemistry , RNA, Viral/blood , Adult , Aged , Biological Transport , Carcinoma , DNA, Viral/blood , Exosomes/virology , Female , Humans , Male , MicroRNAs/metabolism , Middle Aged , Nasopharyngeal Carcinoma , RNA, Viral/metabolism , Real-Time Polymerase Chain Reaction
20.
J Pathol ; 226(3): 471-81, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22009689

ABSTRACT

Nasopharyngeal carcinoma (NPC) is an EBV-associated epithelial malignancy which is prevalent in south-east Asia and southern China. Despite the multiple genetic and epigenetic changes reported, the contribution of dysregulated signalling pathways to this distinct type of head and neck cancer is not well understood. Here we demonstrate the up-regulation of NOTCH ligands (JAG1 or DLL4) and effector (HEY1) in the majority of EBV-positive tumour lines and primary tumours. Among the NOTCH receptors, NOTCH3 was over-expressed in all EBV-positive tumour lines and 92.5% of primary tumours. Aberrant activation of NOTCH3 signalling was consistently detected in all these samples. These findings imply that NOTCH3 may play an crucial role in the development of NPC. By NOTCH3 specific siRNA, NOTCH3 signalling was suppressed and thereby significant growth inhibition and apoptosis induction occurred in NPC cells. Down-regulation of a number of targets involved in cell proliferation, eg CCND1, C-MYC,NFKB1, and survival, eg BCL2, BCL-XL, SURVIVIN, was confirmed in the NOTCH3 knockdown NPC cells. Importantly, NOTCH3 knockdown highly enhanced the sensitivity of NPC cells to cisplatin treatment. Furthermore, we revealed that the ability of NPC cells to form spheroids in vitro and tumours in nude mice was also significantly decreased after knockdown of NICD3 expression. These findings indicate that activation of NOTCH3 pathway is a critical oncogenic event in NPC tumourigenesis. Targeting NOTCH3 signalling may serve as a potential therapeutic approach for treating patients suffering from EBV-associated NPC.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Epstein-Barr Virus Infections/complications , Nasopharyngeal Neoplasms/drug therapy , Receptors, Notch/antagonists & inhibitors , Signal Transduction/physiology , Animals , Apoptosis/physiology , Carcinoma , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/drug effects , Gene Knockdown Techniques , Genes, Neoplasm/physiology , Humans , Inhibitor of Apoptosis Proteins/metabolism , Mice , Mice, Nude , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/virology , Neoplasm Transplantation , RNA, Small Interfering/metabolism , Receptor, Notch3 , Receptors, Notch/metabolism , Spheroids, Cellular/physiology , Transfection , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL