Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Cancers (Basel) ; 13(14)2021 Jul 09.
Article in English | MEDLINE | ID: mdl-34298655

ABSTRACT

Despite the introduction of epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) to treat advanced lung cancer harboring EGFR-activating mutations, the prognosis remains unfavorable because of intrinsic and/or acquired resistance. We generated a new state-of-the-art mouse strain harboring the human EGFRT790M/L858R oncogene and MET overexpression (EGFR/MET strain) that mimics the MET amplification occurring in one out of five patients with EGFR-mutated lung cancer that relapsed after treatment with osimertinib, a third-generation anti-EGFR TKI. We found that survival was reduced in EGFR/MET mice compared with mice harboring only EGFRT790M/L858R (EGFR strain). Moreover, EGFR/MET-driven lung tumors were resistant to osimertinib, recapitulating the phenotype observed in patients. Conversely, as also observed in patients, the crizotinib (anti-MET TKI) and osimertinib combination improved survival and reduced tumor burden in EGFR/MET mice, further validating the model's value for preclinical studies. We also found that in EGFR/MET mice, MET overexpression negatively regulated EGFR activity through MIG6 induction, a compensatory mechanism that allows the coexistence of the two onco-genic events. Our data suggest that single EGFR or MET inhibition might not be a good therapeutic option for EGFR-mutated lung cancer with MET amplification, and that inhibition of both pathways should be the best clinical choice in these patients.

2.
Biochem J ; 477(5): 1033-1047, 2020 03 13.
Article in English | MEDLINE | ID: mdl-32091571

ABSTRACT

Hepatocellular carcinoma (HCC) is the most frequent primary liver cancer in adults. Among the altered pathways leading to HCC, an increasing role is attributed to abnormal epigenetic regulation. Members of the Heterochromatin Protein (HP1) 1 family are key players in chromatin organisation, acting as docking sites for chromatin modifiers. Here, we inactivated HP1α in HepG2 human liver carcinoma cells and showed that HP1α participated in cell proliferation. HP1α-depleted cells have a global decrease in DNA methylation and consequently a perturbed chromatin organisation, as exemplified by the reactivation of transcription at centromeric and pericentromeric regions, eventhough the protein levels of chromatin writers depositing methylation marks, such as EZH2, SETDB1, SUV39H1, G9A and DNMT3A remained unaltered. This decrease was attributed mainly to a low S-Adenosyl Methionine (SAM) level, a cofactor involved in methylation processes. Furthermore, we showed that this decrease was due to a modification in the Methionine adenosyl transferase 2A RNA (MAT2A) level, which modifies the ratio of MAT1A/MAT2A, two enzymes that generate SAM. Importantly, HP1α reintroduction into HP1α-depleted cells restored the MAT2A protein to its initial level. Finally, we demonstrated that this transcriptional deregulation of MAT2A in HP1α-depleted cells relied on a lack of recruitment of HP1ß and HP1γ to MAT2A promoter where an improper non-CpG methylation site was promoted in the vicinity of the transcription start site where HP1ß and HP1γ bound. Altogether, these results highlight an unanticipated link between HP1 and the SAM synthesis pathway, and emphasise emerging functions of HP1s as sensors of some aspects of liver cell metabolism.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Chromosomal Proteins, Non-Histone/deficiency , Liver Neoplasms/metabolism , S-Adenosylmethionine/metabolism , Biosynthetic Pathways/physiology , Chromobox Protein Homolog 5 , HEK293 Cells , Hep G2 Cells , Humans
3.
J Clin Invest ; 130(2): 612-624, 2020 02 03.
Article in English | MEDLINE | ID: mdl-31671073

ABSTRACT

EGFR-mutated lung adenocarcinoma patients treated with gefitinib and osimertinib show a therapeutic benefit limited by the appearance of secondary mutations, such as EGFRT790M and EGFRC797S. It is generally assumed that these secondary mutations render EGFR completely unresponsive to the inhibitors, but contrary to this, we uncovered here that gefitinib and osimertinib increased STAT3 phosphorylation (p-STAT3) in EGFRT790M and EGFRC797S tumoral cells. Interestingly, we also found that concomitant Notch inhibition with gefitinib or osimertinib treatment induced a p-STAT3-dependent strong reduction in the levels of the transcriptional repressor HES1. Importantly, we showed that tyrosine kinase inhibitor-resistant tumors, with EGFRT790M and EGFRC797S mutations, were highly responsive to the combined treatment of Notch inhibitors with gefitinib or osimertinib, respectively. Finally, in patients with EGFR mutations treated with tyrosine kinase inhibitors, HES1 protein levels increased during relapse and correlated with shorter progression-free survival. Therefore, our results offer a proof of concept for an alternative treatment to chemotherapy in lung adenocarcinoma osimertinib-treated patients after disease progression.


Subject(s)
Acrylamides/pharmacology , Adenocarcinoma of Lung , Aniline Compounds/pharmacology , Drug Resistance, Neoplasm/drug effects , ErbB Receptors , Gefitinib/pharmacology , Lung Neoplasms , Mutation, Missense , Neoplasm Proteins , Protein Kinase Inhibitors/pharmacology , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/pathology , Amino Acid Substitution , Animals , Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Transgenic , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Transcription Factor HES-1/genetics , Transcription Factor HES-1/metabolism
4.
Sci Data ; 4: 170150, 2017 10 17.
Article in English | MEDLINE | ID: mdl-29039845

ABSTRACT

RNA-Seq enables the generation of extensive transcriptome information providing the capability to characterize transcripts (including alternative isoforms and polymorphism), to quantify expression and to identify differential regulation in a single experiment. To reveal the capacity of new anti-HIV ABX464 candidate in modulating the expression of genes, datasets were generated and validated using RNA-seq approach. This comprehensive dataset will be useful to deepen the comprehensive understanding of the progression of human immunodeficiency virus (HIV) associated with mucosal damage in the gastrointestinal (GI) tract and subsequent inflammation, providing an opportunity to generate new therapies, diagnoses, and preventive strategies.


Subject(s)
Anti-HIV Agents/adverse effects , Macrophage Activation/drug effects , Macrophage Activation/genetics , Quinolines/adverse effects , Gastroenteritis/complications , Gastroenteritis/drug therapy , HIV Infections/complications , HIV Infections/drug therapy , Humans , Sequence Analysis, RNA
5.
Sci Rep ; 7(1): 4860, 2017 07 07.
Article in English | MEDLINE | ID: mdl-28687795

ABSTRACT

The progression of human immunodeficiency virus (HIV) is associated with mucosal damage in the gastrointestinal (GI) tract. This damage enables bacterial translocation from the gut and leads to subsequent inflammation. Dextran sulfate sodium (DSS-exposure) is an established animal model for experimental colitis that was recently shown to recapitulate the link between GI-tract damage and pathogenic features of SIV infection. The current study tested the protective properties of ABX464, a first-in-class anti-HIV drug candidate currently in phase II clinical trials. ABX464 treatment strongly attenuated DSS-induced colitis in mice and produced a long-term protection against prolonged DSS-exposure after drug cessation. Consistently, ABX464 reduced the colonic production of the inflammatory cytokines IL-6 and TNFα as well as that of the chemoattractant MCP-1. However, RNA profiling analysis revealed the capacity of ABX464 to induce the expression of IL-22, a cytokine involved in colitis tissue repair, both in DSS-treated mice and in LPS-stimulated bone marrow-derived macrophages. Importantly, anti-IL-22 antibodies significantly reduced the protective effect of ABX464 on colitis in DSS-treated mice. Because reduced IL-22 production in the gut mucosa is an established factor of HIV and DSS-induced immunopathogenesis, our data suggest that the anti-inflammatory properties of ABX464 warrant exploration in both HIV and inflammatory ulcerative colitis (UC) disease.


Subject(s)
Anti-HIV Agents/administration & dosage , Anti-Inflammatory Agents/administration & dosage , Colitis/drug therapy , Interleukins/metabolism , Macrophages/immunology , Quinolines/administration & dosage , Animals , Anti-HIV Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Colitis/chemically induced , Colitis/pathology , Cytokines/analysis , Dextran Sulfate/administration & dosage , Dextran Sulfate/toxicity , Disease Models, Animal , Gene Expression Profiling , Interleukins/genetics , Intestinal Mucosa/pathology , Macrophages/drug effects , Mice , Quinolines/pharmacology , Treatment Outcome , Interleukin-22
6.
PLoS One ; 12(3): e0172603, 2017.
Article in English | MEDLINE | ID: mdl-28273108

ABSTRACT

ICAT (Inhibitor of ß-CAtenin and TCF) is a small acidic protein that negatively regulates ß-catenin co-transcriptional activity by competing with TCF/LEF factors in their binding to ß-catenin superhelical core. In melanoma cells, ICAT competes with LEF1 to negatively regulate the M-MITF and NEDD9 target genes. The structure of ICAT consists of two domains: the 3-helix bundle N-terminal domain binds to ß-catenin Armadillo (Arm) repeats 10-12 and the C-terminal tail binds to Arm repeats 5-9. To elucidate the structural mechanisms governing ICAT/ß-catenin interactions in melanoma cells, three ICAT residues Y15, K19 and V22 in the N-terminal domain, contacting hydrophobic ß-catenin residue F660, were mutated and interaction was assessed by immunoprecipitation. Despite the moderate hydrophobicity of the contact, its removal completely abolished the interaction. In the ICAT C-terminal tail consensus sequence, neutralization of the electrostatic interactions between residues D66, E75 and ß-catenin residues K435, K312, coupled to deletion of the hydrophobic contact between F71 and ß-catenin R386, markedly reduced, but failed to abolish the ICAT-mediated negative regulation of M-MITF and NEDD9 promoters. We conclude that in melanoma cells, anchoring of ICAT N-terminal domain to ß-catenin through the hook made by residue F660, trapped in the pincers formed by ICAT residues Y15 and V22, is crucial for stabilizing the ICAT/ß-catenin complex. This is a prerequisite for binding of the consensus peptide to Arm repeats 5-9 and competition with LEF1. Differences between ICAT and LEF1 in their affinity for ß-catenin may rely on the absence in ICAT of hydrophilic residues between D66 and F71.


Subject(s)
Gene Expression Regulation , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Mutation , Transcriptional Activation , beta Catenin/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Binding, Competitive , Cell Line, Tumor , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Lymphoid Enhancer-Binding Factor 1/metabolism , Melanocytes/metabolism , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Models, Molecular , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs
7.
Arthritis Res Ther ; 17: 193, 2015 Jul 29.
Article in English | MEDLINE | ID: mdl-26220665

ABSTRACT

INTRODUCTION: We previously reported that low ratio of osteoprotegerin (OPG) to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) was associated with Disease Activity Score in 28 joints (DAS28) remission at 6 months in patients with early rheumatoid arthritis (RA). Here, we aimed to evaluate the value of baseline OPG/TRAIL ratio in predicting clinical and radiological outcomes in patients with early RA in the ESPOIR cohort. METHODS: OPG and TRAIL serum concentrations were assessed in the ESPOIR cohort patients. Patients with definite RA were included in this study. Patients were excluded if they had high erosion score at baseline (>90(th) percentile) or received biological therapy during the first 2 years of follow-up. Data were analyzed by univariate analysis and multivariate logistic regression to predict 1-year DAS28 remission and 2-year radiographic disease progression. RESULTS: On univariate analysis of 399 patients, OPG/TRAIL ratio at baseline was significantly lower in patients with than without remission at 1 year (p = 0.015). On multivariate logistic regression including age, gender, body mass index and DAS28, low OPG/TRAIL ratio was independently associated with remission at 1 year (odds ratio 1.68 [95 % confidence interval 1.01-2.79]). On univariate analysis, high OPG/TRAIL ratio at baseline was associated with rapid progression of erosion at 2 years (p = 0.041), and on multivariate logistic regression including age, anti-citrullinated protein antibody positivity and C-reactive protein level, OPG/TRAIL ratio independently predicted rapid progression of erosion at 2 years. CONCLUSIONS: OPG/TRAIL ratio at baseline was an independent predictor of 1-year remission and 2-year rapid progression of erosion for patients with early rheumatoid arthritis. Thus, OPG/TRAIL ratio could be included in matrix prediction scores to predict rapid radiographic progression. Further confirmation in an independent cohort is warranted.


Subject(s)
Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/diagnosis , Osteoprotegerin/blood , TNF-Related Apoptosis-Inducing Ligand/blood , Adult , Biomarkers/blood , Cohort Studies , Female , Humans , Male , Middle Aged , Prognosis , Prospective Studies
8.
EMBO J ; 33(19): 2247-60, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25180231

ABSTRACT

TTLL3 and TTLL8 are tubulin glycine ligases catalyzing posttranslational glycylation of microtubules. We show here for the first time that these enzymes are required for robust formation of primary cilia. We further discover the existence of primary cilia in colon and demonstrate that TTLL3 is the only glycylase in this organ. As a consequence, colon epithelium shows a reduced number of primary cilia accompanied by an increased rate of cell division in TTLL3-knockout mice. Strikingly, higher proliferation is compensated by faster tissue turnover in normal colon. In a mouse model for tumorigenesis, lack of TTLL3 strongly promotes tumor development. We further demonstrate that decreased levels of TTLL3 expression are linked to the development of human colorectal carcinomas. Thus, we have uncovered a novel role for tubulin glycylation in primary cilia maintenance, which controls cell proliferation of colon epithelial cells and plays an essential role in colon cancer development.


Subject(s)
Cell Proliferation , Cilia/metabolism , Colon/metabolism , Colonic Neoplasms/metabolism , Glycine/metabolism , Peptide Synthases/physiology , Tubulin/physiology , Animals , Blotting, Western , Carcinogens/toxicity , Cells, Cultured , Colon/pathology , Colonic Neoplasms/chemically induced , Colonic Neoplasms/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Disease Models, Animal , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Immunoenzyme Techniques , Mice , Mice, Knockout , Microtubules/metabolism , Protein Processing, Post-Translational , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
9.
Arthritis Rheumatol ; 66(12): 3289-99, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25078097

ABSTRACT

OBJECTIVE: Injection of agonistic anti-Fas antibody has been shown to decrease disease symptoms in mouse models of arthritis. Additionally, membrane-bound FasL (mFasL) has been shown to induce cell death in fibroblast-like synoviocytes (FLS) from rheumatoid arthritis (RA) patients. However, levels of soluble FasL (sFasL) are increased in the joints of RA patients and have been associated with disease severity, indicating that mFasL and sFasL play opposing roles in RA. The purpose of this study was to analyze the effects of FasL on RA FLS responses. METHODS: The responses of FLS from RA and osteoarthritis (OA) patients to soluble and oligomeric FasL, the latter mimicking mFasL, were analyzed by fluorescence-activated cell sorting and proliferation assays, using 3 different FasL variants. The signaling pathways that trigger FasL responses were characterized by Western blotting. RESULTS: We found that mFasL and sFasL have distinct roles in RA FLS. Crosslinked FasL preferentially induced apoptosis, whereas sFasL stimulated proliferation. Moreover, sFasL activated several signaling pathways in RA FLS, such as ERK-1/2, phosphatidylinositol 3-kinase, caspase 8, and JNK, with a prominent role of JNK, since only the blockade of this pathway rendered FLS more susceptible to FasL-induced apoptosis. Crosslinked FasL induced apoptosis in FLS from OA patients, but sFasL failed to stimulate their proliferation. CONCLUSION: Our findings suggest that sFasL is a disease promoter in RA, a finding consistent with previous reports describing a tumor-promoting role of FasL. Therefore, blocking of sFasL could be a therapeutic strategy for RA.


Subject(s)
Apoptosis/physiology , Arthritis, Rheumatoid/metabolism , Cell Proliferation/physiology , Fas Ligand Protein/metabolism , Fibroblasts/metabolism , Synovial Membrane/metabolism , Adult , Aged , Apoptosis/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Fas Ligand Protein/drug effects , Female , Fibroblasts/drug effects , Flow Cytometry , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Male , Middle Aged , Osteoarthritis/metabolism , Phosphatidylinositol 3-Kinases/drug effects , Phosphatidylinositol 3-Kinases/physiology , Synovial Membrane/cytology , Synovial Membrane/drug effects
10.
Cancer Res ; 74(7): 1983-95, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24514042

ABSTRACT

Inhibitor of ß-catenin and TCF (ICAT) inhibits ß-catenin transcriptional activity by competing with T-cell factor/lymphoid enhancer factor. We documented high ICAT levels in human melanoma cells, in which ß-catenin signaling is frequently deregulated, finding a correlation with the capacity to form metastases in nude mice. Ectopic expression of ICAT in melanoma cells did not affect their proliferation but increased cell motility and Matrigel invasion of metastatic cells in a manner relying upon stable ICAT-ß-catenin interaction. This effect was associated with conversion of an elongated/mesenchymal phenotype to a round/amoeboid phenotype in the absence of similar effects on elongated morphology of nonmetastatic melanoma cells. Transition from mesenchymal to amoeboid movement was associated with decreased levels of NEDD9 and activated Rac1, a positive regulator of mesenchymal movement. Ectopic ICAT promoted colonization of melanoma cells in the lungs of nude mice, suggesting an increase in metastatic potential. Together, our results showed that by downregulating Rac signaling in metastatic melanoma cells, ICAT increased their invasive motility by promoting a morphologic variation that facilitates a favorable adaptation to their microenvironment.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , Melanoma/pathology , Adaptor Proteins, Signal Transducing/physiology , Animals , Cell Line, Tumor , Cell Movement , Humans , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Melanoma/mortality , Melanoma/secondary , Mice , Mice, Nude , Neoplasm Invasiveness , Phosphoproteins/physiology , beta Catenin/physiology
11.
Ann Rheum Dis ; 72(8): 1367-74, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23178293

ABSTRACT

BACKGROUND: The tumour necrosis factor (TNF)-family members B cell activating factor (BAFF) and A PRoliferation-Inducing Ligand (APRIL) play important roles in B cell biology, and share binding to B cell maturation antigen and transmembrane activator and cyclophilin ligand interactor, both receptors of the TNF-family. However, while it is reported that BAFF can break B cell tolerance, the role of APRIL in autoimmunity remains elusive. OBJECTIVE: To evaluate the role of APRIL on collagen-induced arthritis (CIA). METHODS: CIA was induced in APRIL-transgenic (Tg) DBA/1 mice and littermates. Disease progression was evaluated by clinical and histological signs of arthritis. In another experimental setting mice were exposed to the collagen antibody-induced arthritis. In addition, we tested T cell dependent humoral responses in APRIL-Tg mice. RESULTS: We found that APRIL-Tg displayed a strongly reduced incidence and severity of CIA compared with littermates, with decreases in collagen-specific autoantibody titres, immune complex deposition and downstream mast cell activation in joints. Notably, ectopic APRIL-expression was also found to negatively regulate T cell dependent humoral responses. The lower autoantibody production in APRIL-Tg mice during CIA appears to be crucial, as arthritis induced by administration of anti-collagen antibodies developed similar in APRIL-Tg and control mice, thus demonstrating that the downstream effector pathways induced by anti-collagen antibodies remain intact in APRIL-Tg mice. This protective effect was specifically mediated by APRIL, as adenoviral delivery of APRIL decreased CIA in a therapeutic setting. CONCLUSIONS: Collectively, our data identify APRIL as a negative regulator of CIA by regulating autoantibody production. These findings are of important clinical relevance, as the therapeutic potential of transmembrane activator and cyclophilin ligand interactor-Fc (atacicept) is presently evaluated in clinical trials.


Subject(s)
Arthritis, Experimental/genetics , Gene Expression Regulation , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics , Animals , Antigen-Antibody Complex/metabolism , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Autoantibodies , Cell Degranulation/immunology , Disease Progression , Hindlimb , Immunity, Humoral/genetics , Immunity, Humoral/immunology , Immunoglobulin G/blood , Mast Cells/immunology , Mast Cells/pathology , Mice , Mice, Inbred DBA , Mice, Transgenic , Stifle/immunology , Stifle/metabolism , Stifle/pathology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism
12.
Virology ; 393(2): 311-8, 2009 Oct 25.
Article in English | MEDLINE | ID: mdl-19733381

ABSTRACT

RNA helicase-like receptors MDA-5 but not RIG-I has been shown to be essential for triggering innate immune responses against picornaviruses. However, virus-host co-evolution has selected for viruses capable of replicating despite host cells antiviral defences. In this report, we demonstrate that RIG-I is degraded during encephalomyocarditis virus (EMCV) infection. This effect is mediated by both the viral-encoded 3C protease and caspase proteinase. In addition, we show that RIG-I overexpression confers IFN-beta promoter activation during EMCV infection, in MDA-5 knockout (MDA-5(-/-)) mouse embryo fibroblasts. This induction is followed by a strong inhibition reflecting the ability of EMCV to disrupt RIG-I signalling. Taken together, our data strongly suggest that during evolution RIG-I has been involved for triggering innate immune response to picornavirus infections.


Subject(s)
Cardiovirus Infections/immunology , DEAD-box RNA Helicases/metabolism , Encephalomyocarditis virus/immunology , Immunity, Innate , Interferon-beta/immunology , 3C Viral Proteases , Animals , Cardiovirus Infections/virology , Caspases/immunology , Caspases/metabolism , Cysteine Endopeptidases/immunology , Cysteine Endopeptidases/metabolism , DEAD Box Protein 58 , DEAD-box RNA Helicases/immunology , Encephalomyocarditis virus/metabolism , Gene Expression Regulation , HeLa Cells , Humans , Interferon-Induced Helicase, IFIH1 , Mice , Promoter Regions, Genetic , Viral Proteins/immunology , Viral Proteins/metabolism
13.
J Gen Virol ; 90(Pt 3): 536-545, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19218198

ABSTRACT

The interferon (IFN) system is a major effector of the innate immunity that allows time for the subsequent establishment of an adaptive immune response against a wide-range of pathogens. Their diverse biological actions are thought to be mediated by the products of specific but usually overlapping sets of cellular genes induced in the target cells. Ubiquitin ligase members of the tripartite motif (TRIM) protein family have emerged as IFN-induced proteins involved in both innate and adaptive immunity. In this report, we provide evidence that TRIM22 is a functional E3 ubiquitin ligase that is also ubiquitinated itself. We demonstrate that TRIM22 expression leads to a viral protection of HeLa cells against encephalomyocarditis virus infections. This effect is dependent upon its E3 ubiquitinating activity, since no antiviral effect was observed in cells expressing a TRIM22-deletion mutant defective in ubiquitinating activity. Consistent with this, TRIM22 interacts with the viral 3C protease (3C(PRO)) and mediates its ubiquitination. Altogether, our findings demonstrate that TRIM22 E3 ubiquitin ligase activity represents a new antiviral pathway induced by IFN against picornaviruses.


Subject(s)
Antiviral Agents/metabolism , Cysteine Endopeptidases/metabolism , Encephalomyocarditis virus/drug effects , Repressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Viral Proteins/metabolism , 3C Viral Proteases , Antiviral Agents/pharmacology , Cell Line , Cysteine Endopeptidases/genetics , Encephalomyocarditis virus/enzymology , Encephalomyocarditis virus/genetics , HeLa Cells , Humans , Minor Histocompatibility Antigens , Repressor Proteins/genetics , Repressor Proteins/pharmacology , Tripartite Motif Proteins , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/pharmacology , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...