Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
Add more filters










Publication year range
1.
J Proteome Res ; 9(7): 3527-36, 2010 Jul 02.
Article in English | MEDLINE | ID: mdl-20459142

ABSTRACT

Innate immunity has evolved complex molecular pathways to protect organisms from viral infections. One pivotal line of cellular defense is the induction of the antiviral effect of interferon. To circumvent this primary response and achieve their own replication, viruses have developed complex molecular strategies. Here, we provide a systems-level study of the human type I interferon system subversion by the viral proteome, by reconstructing the underlying protein-protein interaction network. At this network level, viruses establish a massive and a gradual attack, from receptors to transcription factors, by interacting preferentially with highly connected and central proteins as well as interferon-induced proteins. We also demonstrate that viruses significantly target 22% of the proteins directly interacting with the type I interferon system network, suggesting the relevance of our network-based method to identify new candidates involved in the regulation of the antiviral response. Finally, based on the comparative analysis of interactome profiles across four viral families, we provide evidence of common and differential targeting strategies.


Subject(s)
Host-Pathogen Interactions/immunology , Interferon Type I/immunology , Protein Interaction Mapping/methods , Systems Biology/methods , Viruses/immunology , Databases, Genetic , Flaviviridae/immunology , Herpesviridae/immunology , Humans , Papillomaviridae/immunology , Retroviridae/immunology , Signal Transduction , Statistics, Nonparametric
2.
J Bone Miner Res ; 25(3): 661-72, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19775202

ABSTRACT

Dendritic cells (DCs), mononuclear cells that initiate immune responses, and osteoclasts (OCs), multinucleated bone-resorbing cells, are hematopoietic cells derived from monocytic precursor cells. Using in vitro generated dendritic cells, we previously showed that human and murine DCs could transdifferentiate into resorbing osteoclasts in the presence of macrophage colony-stimulating factor (M-CSF) and receptor activator of NF-kappaB ligand (RANKL). In this study we globally compared by transcriptomic profiling this new osteoclast differentiation pathway from DCs with the canonical differentiation pathway from monocytes. DNA chip data revealed that starting from two very distinct cell types, treatment with M-CSF and RANKL generated two highly similar types of osteoclast. In particular, DC-derived osteoclasts expressed all the characteristic marker genes of monocyte-derived osteoclasts. Two major molecular events could be observed during osteoclastogenesis: downregulation of a large set of monocyte or DC specific markers, together with upregulation of characteristic osteoclast marker genes. Most interestingly, our transcriptomic data showed a closer molecular profile between DCs and OCs than between monocytes and OCs. Our data establish DCs as a new osteoclast precursor able to generate OCs more efficiently than monocytes.


Subject(s)
Bone Resorption , Cell Differentiation , Dendritic Cells/cytology , Monocytes/cytology , Osteoclasts/cytology , Antigens, Surface/genetics , Antigens, Surface/metabolism , Cells, Cultured , Flow Cytometry , Gene Expression Regulation , Genome-Wide Association Study , Humans , Reverse Transcriptase Polymerase Chain Reaction
3.
Nucleic Acids Res ; 38(Database issue): D371-8, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20007148

ABSTRACT

Large collections of protein-encoding open reading frames (ORFs) established in a versatile recombination-based cloning system have been instrumental to study protein functions in high-throughput assays. Such 'ORFeome' resources have been developed for several organisms but in virology, plasmid collections covering a significant fraction of the virosphere are still needed. In this perspective, we present ViralORFeome 1.0 (http://www.viralorfeome.com), an open-access database and management system that provides an integrated set of bioinformatic tools to clone viral ORFs in the Gateway(R) system. ViralORFeome provides a convenient interface to navigate through virus genome sequences, to design ORF-specific cloning primers, to validate the sequence of generated constructs and to browse established collections of virus ORFs. Most importantly, ViralORFeome has been designed to manage all possible variants or mutants of a given ORF so that the cloning procedure can be applied to any emerging virus strain. A subset of plasmid constructs generated with ViralORFeome platform has been tested with success for heterologous protein expression in different expression systems at proteome scale. ViralORFeome should provide our community with a framework to establish a large collection of virus ORF clones, an instrumental resource to determine functions, activities and binding partners of viral proteins.


Subject(s)
Computational Biology/methods , Databases, Genetic , Databases, Nucleic Acid , Databases, Protein , Genes, Viral , Open Reading Frames , Cloning, Molecular , Computational Biology/trends , Genetic Techniques , Genome, Viral , Information Storage and Retrieval/methods , Internet , Protein Structure, Tertiary , Software , User-Computer Interface
4.
Mol Syst Biol ; 4: 230, 2008.
Article in English | MEDLINE | ID: mdl-18985028

ABSTRACT

A proteome-wide mapping of interactions between hepatitis C virus (HCV) and human proteins was performed to provide a comprehensive view of the cellular infection. A total of 314 protein-protein interactions between HCV and human proteins was identified by yeast two-hybrid and 170 by literature mining. Integration of this data set into a reconstructed human interactome showed that cellular proteins interacting with HCV are enriched in highly central and interconnected proteins. A global analysis on the basis of functional annotation highlighted the enrichment of cellular pathways targeted by HCV. A network of proteins associated with frequent clinical disorders of chronically infected patients was constructed by connecting the insulin, Jak/STAT and TGFbeta pathways with cellular proteins targeted by HCV. CORE protein appeared as a major perturbator of this network. Focal adhesion was identified as a new function affected by HCV, mainly by NS3 and NS5A proteins.


Subject(s)
Hepatitis C/metabolism , Viral Proteins/metabolism , Hepacivirus/metabolism , Hepacivirus/physiology , Humans , Protein Binding , Two-Hybrid System Techniques
5.
J Virol ; 81(23): 12859-71, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17898060

ABSTRACT

Measles virus (MeV) infection is characterized by the formation of multinuclear giant cells (MGC). We report that beta interferon (IFN-beta) production is amplified in vitro by the formation of virus-induced MGC derived from human epithelial cells or mature conventional dendritic cells. Both fusion and IFN-beta response amplification were inhibited in a dose-dependent way by a fusion-inhibitory peptide after MeV infection of epithelial cells. This effect was observed at both low and high multiplicities of infection. While in the absence of virus replication, the cell-cell fusion mediated by MeV H/F glycoproteins did not activate any IFN-alpha/beta production, an amplified IFN-beta response was observed when H/F-induced MGC were infected with a nonfusogenic recombinant chimerical virus. Time lapse microscopy studies revealed that MeV-infected MGC from epithelial cells have a highly dynamic behavior and an unexpected long life span. Following cell-cell fusion, both of the RIG-I and IFN-beta gene deficiencies were trans complemented to induce IFN-beta production. Production of IFN-beta and IFN-alpha was also observed in MeV-infected immature dendritic cells (iDC) and mature dendritic cells (mDC). In contrast to iDC, MeV infection of mDC induced MGC, which produced enhanced amounts of IFN-alpha/beta. The amplification of IFN-beta production was associated with a sustained nuclear localization of IFN regulatory factor 3 (IRF-3) in MeV-induced MGC derived from both epithelial cells and mDC, while the IRF-7 up-regulation was poorly sensitive to the fusion process. Therefore, MeV-induced cell-cell fusion amplifies IFN-alpha/beta production in infected cells, and this indicates that MGC contribute to the antiviral immune response.


Subject(s)
Dendritic Cells/virology , Epithelial Cells/virology , Giant Cells/virology , Interferon Type I/biosynthesis , Measles virus/immunology , Measles virus/physiology , Animals , Cell Fusion , Cell Line , Cell Nucleus/chemistry , Chlorocebus aethiops , Dendritic Cells/immunology , Epithelial Cells/immunology , Giant Cells/cytology , Giant Cells/immunology , Humans , Interferon Regulatory Factor-3/analysis , Interferon Regulatory Factor-7/analysis , Measles virus/genetics , Microscopy, Video , Viral Fusion Proteins/immunology , Viral Fusion Proteins/physiology , Viral Proteins/immunology , Viral Proteins/physiology
6.
J Gen Virol ; 86(Pt 6): 1771-1784, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15914856

ABSTRACT

Measles virus (MV) nucleoprotein (N) is a cytosolic protein that is released into the extracellular compartment after apoptosis and/or secondary necrosis of MV-infected cells in vitro. Thus, MV-N becomes accessible to inhibitory cell-surface receptors: FcgammaRIIB and an uncharacterized nucleoprotein receptor (NR). MV-N is composed of two domains: NCORE (aa 1-400) and NTAIL (aa 401-525). To assess the contribution of MV-N domains and of these two receptors in suppression of cell proliferation, a human melanoma HT144 cell line expressing (HT144IIB1) or lacking FcgammaRIIB1 was used as a model. Specific and exclusive NCORE-FcgammaRIIB1 and NTAIL-NR interactions were shown. Moreover, NTAIL binding to human NR predominantly led to suppression of cell proliferation by arresting cells in the G0/G1 phases of the cell cycle, rather than to apoptosis. NCORE binding to HT144IIB1 cells primarily triggered caspase-3 activation, in contrast to HT144IIB1/IC- cells lacking the FcgammaRIIB1 intra-cytoplasmic tail, thus demonstrating the specific inhibitory effect of the NCORE-FcgammaRIIB1 interaction. MV-N- and NCORE-mediated apoptosis through FcgammaRIIB1 was inhibited by the pan-caspase inhibitor zVAD-FMK, indicating that apoptosis was dependent on caspase activation. By using NTAIL deletion proteins, it was also shown that the region of NTAIL responsible for binding to human NR and for cell growth arrest maps to one of the three conserved boxes (Box1, aa 401-420) found in N of Morbilliviruses. This work unveils novel mechanisms by which distinct domains of MV-N may display different immunosuppressive activities, thus contributing to our comprehension of the immunosuppressive state associated with MV infection. Finally, MV-N domains may be good tools to target tumour cell proliferation and/or apoptosis.


Subject(s)
Antigens, CD/metabolism , Measles virus/physiology , Nucleoproteins/metabolism , Receptors, Cell Surface/metabolism , Receptors, IgG/metabolism , Receptors, Virus/metabolism , Viral Proteins/metabolism , Virus Replication , Animals , Apoptosis , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Gene Deletion , Humans , Measles virus/metabolism , Nucleocapsid Proteins , Nucleoproteins/genetics , Protein Structure, Tertiary/genetics , Viral Proteins/genetics
8.
Curr Top Microbiol Immunol ; 276: 103-23, 2003.
Article in English | MEDLINE | ID: mdl-12797445

ABSTRACT

Measles virus (MV) infection induces both an efficient MV-specific immune response and a transient but profound immunosuppression characterised by a panlymphopenia that occasionally results in opportunistic infections responsible for a high rate of mortality in children. On the basis of in vitro studies, the putative roles of dendritic cells (DCs) in MV infection are discussed. (1) DCs could participate in anti-MV innate immunity because MV turns on TNF-related apoptosis-inducing ligand (TRAIL)-mediated DC cytotoxicity. (2) Cross-priming by non-infected DCs might be the route of MV adaptive immune response. (3) After CD40-ligand activation in secondary lymphoid organs, MV-infected DCs could initiate the formation of Warthin-Finkeldey multinucleated giant cells, replicating MV and responsible for in vivo spreading of MV. (4) We review how integrated viral attack of the host immune system also targets DCs: Progress in understanding the immunobiology of MV-infected DCs that could account for MV-induced immunosuppression observed in vivo is presented and their potential role in lymphopenia is underlined. In conclusion, future research directions are proposed.


Subject(s)
Cytotoxicity, Immunologic/immunology , Dendritic Cells/immunology , Immunosuppression Therapy , Measles virus/immunology , Measles/immunology , Apoptosis , Apoptosis Regulatory Proteins , CD40 Antigens/immunology , CD40 Ligand/immunology , Dendritic Cells/virology , Humans , Measles virus/physiology , Membrane Glycoproteins/immunology , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/immunology , Virus Replication
9.
J Immunol ; 167(12): 6780-5, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11739493

ABSTRACT

Efficient T cell activation requires at least two signals, one mediated by the engagement of the TCR-CD3 complex and another one mediated by a costimulatory molecule. We recently showed that CD46, a complement regulatory receptor for C3b as well as a receptor for several pathogens, could act as a potent costimulatory molecule for human T cells, highly promoting T cell proliferation. Indeed, we show in this study that CD46/CD3 costimulation induces a synergistic activation of extracellular signal-related kinase mitogen-activated protein kinase. Furthermore, whereas T lymphocytes primarily circulate within the bloodstream, activation may induce their migration toward secondary lymphoid organs or other tissues to encounter APCs or target cells. In this study, we show that CD46/CD3 costimulation also induces drastic morphological changes of primary human T cells, as well as actin relocalization. Moreover, we show that the GTP/GDP exchange factor Vav is phosphorylated upon CD46 stimulation alone, and that CD46/CD3 costimulation induces a synergistic increase of Vav phosphorylation. These results prompted us to investigate whether CD46/CD3 costimulation induced the activation of GTPases from the Rho family. Indeed, we report that the small GTPase Rac is also activated upon CD46/CD3 costimulation, whereas no change of Rho and Cdc42 activity could be detected. Therefore, CD46 costimulation profoundly affects T cell behavior, and these results provide important data concerning the biology of primary human T cells.


Subject(s)
Antigens, CD/metabolism , CD3 Complex/metabolism , Cell Cycle Proteins , Lymphocyte Activation , MAP Kinase Signaling System , Membrane Glycoproteins/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Actins/analysis , Cells, Cultured , Cytoskeleton/ultrastructure , Enzyme Activation , Humans , Kinetics , Membrane Cofactor Protein , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-vav , T-Lymphocytes/ultrastructure , rac GTP-Binding Proteins/metabolism
10.
J Immunol ; 167(7): 3765-72, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11564793

ABSTRACT

The main function of dendritic cells (DCs) is to induce adaptive immune response through Ag presentation and specific T lymphocyte activation. However, IFN-alpha- or IFN-gamma-stimulated CD11c+ blood DCs and IFN-beta-stimulated monocyte-derived DCs were recently reported to express functional TNF-related apoptosis-inducing ligand (TRAIL), suggesting that DCs may become cytotoxic effector cells of innate immunity upon appropriate stimulation. In this study, we investigate whether dsRNA and CD40 ligand (CD40L), that were characterized as potent inducers of DC maturation, could also stimulate or modulate DC cytotoxicity toward tumoral cells. We observed that dsRNA, but not CD40L, is a potent inducer of TRAIL expression in human monocyte-derived DCs. As revealed by cytotoxicity assays, DCs acquire the ability to kill tumoral cells via the TRAIL pathway when treated with dsRNA. More precisely, dsRNA is shown to induce IFN-beta synthesis that consecutively mediates TRAIL expression by the DCs. In contrast, we demonstrate that TRAIL expression in dsRNA- or IFN-alpha-treated DCs is potently inhibited after CD40L stimulation. Unexpectedly, CD40L-activated DCs still developed cytotoxicity toward tumoral cells. This latter appeared to be partly mediated by TNF-alpha induction and a yet unidentified pathway. Altogether, these results demonstrate that dsRNA and CD40L, that were originally characterized as maturation signals for DCs, also stimulate their cytotoxicity that is mediated through TRAIL-dependent or -independent mechanisms.


Subject(s)
CD40 Ligand/physiology , Cytotoxicity, Immunologic , Dendritic Cells/immunology , RNA, Double-Stranded/pharmacology , Animals , Apoptosis Regulatory Proteins , CD40 Antigens/metabolism , Cells, Cultured , Cytotoxicity Tests, Immunologic , Dendritic Cells/drug effects , Humans , Interferon-beta/biosynthesis , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Mice , Monocytes/immunology , RNA, Messenger/biosynthesis , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/physiology
11.
Immunity ; 14(1): 69-79, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11163231

ABSTRACT

Measles virus (MV) causes profound immunosuppression, resulting in high infant mortality. The mechanisms are poorly understood, largely due to the lack of a suitable animal model. Here, we report that particular MV proteins, in the absence of MV replication, could generate a systemic immunosuppression in mice through two pathways: (1) via MV-nucleoprotein and its receptor FcgammaR on dendritic cells; and (2) via virus envelope glycoproteins and the MV-hemagglutinin cellular receptor, CD46. The effects comprise reduced hypersensitivity responses associated with impaired function of dendritic cells, decreased production of IL-12, and the loss of antigen-specific T cell proliferation. These results introduce a novel model for testing the immunosuppressive potential of anti-measles vaccines and reveal a specific mechanism of MV-induced modulation of inflammatory reactions.


Subject(s)
Antigens, CD/immunology , Hemagglutinins, Viral/immunology , Immunosuppressive Agents/immunology , Measles virus/immunology , Membrane Glycoproteins/immunology , Nucleoproteins/immunology , Receptors, IgG/immunology , Viral Fusion Proteins/immunology , Viral Proteins/immunology , Animals , Antigen-Presenting Cells/immunology , Antigens, CD/genetics , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Division , Dendritic Cells/immunology , Dermatitis, Contact/immunology , Dinitrofluorobenzene/immunology , Disease Models, Animal , Hemocyanins/immunology , Hypersensitivity, Delayed/chemically induced , Hypersensitivity, Delayed/immunology , Interleukin-12/biosynthesis , Lymph Nodes/immunology , Membrane Cofactor Protein , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Nucleocapsid Proteins , Ultraviolet Rays
12.
Immunobiology ; 204(5): 629-38, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11846228

ABSTRACT

Measle virus (MV) infection induces a transient but profound immunosuppression characterized by a panlymphopenia which occasionally results in opportunistic infections responsible for a high rate of mortality in malnourished children. MV can encounter human dendritic cells (DC) in the respiratory mucosa or in the secondary lymphoid organs. After a brief presentation of DCs, we review progress in understanding the immunobiology of MV-infected DCs that could account for MV-induced immunosuppression. In addition, we develop the newly described TRAIL-mediated cytotoxic function of DCs that is turned on by MV infection, but also by interferons or double-stranded RNA (poly (I:C)). Finally, we propose a model where the measles-associated lymphopenia could be mediated by TRAIL and the measles-induced immunosuppression could be transiently prolonged by Fas-mediated destruction of DCs.


Subject(s)
Cytotoxicity, Immunologic/immunology , Dendritic Cells/immunology , Immune Tolerance/immunology , Measles/immunology , Membrane Glycoproteins/immunology , Tumor Necrosis Factor-alpha/immunology , Adaptation, Physiological/immunology , Animals , Apoptosis Regulatory Proteins , Dendritic Cells/virology , Humans , Immunity, Active/immunology , Ligands , Measles virus/immunology , TNF-Related Apoptosis-Inducing Ligand
13.
Gene Ther ; 7(19): 1613-23, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11083469

ABSTRACT

We describe the generation and the characterization of new lentiviral vectors derived from SIVmac251, a simian immunodeficiency virus (SIV). A methodical approach was used to engineer both efficient and safe packaging constructs allowing the production of SIV viral core proteins. SIV-vectors encoding GFP (green fluorescent protein) were generated as VSV-G-pseudotyped particles upon transient expression of the vector construct and helper functions in 293 cells. The SIV vectors were able to transduce efficiently various target cell types at low multiplicity of infection, including monocyte-differentiated human dendritic cells (DCs) which retained their capacity to differentiate into mature DCs after gene transfer. Transduction of the DCs by the SIV vectors was prevented when infections were performed in the presence of AZT, a reverse-transcriptase inhibitor. After gene transfer, expression of the GFP in the target cells remained constant after several weeks, indicating that the vectors had been stably integrated into the genome of the host cells. Preparations of SIV vectors were systematically checked for the absence of replication-competent and recombinant retroviruses but remained negative, suggesting the innocuousness of these novel gene delivery vectors. Side-to-side comparisons with vectors derived from HIV-1 (human immunodeficiency virus) indicated that the SIV vectors were equally potent in transducing proliferating target cells. Finally, we have determined the infectivity of SIV vectors pseudotyped with surface glycoproteins of several membrane-enveloped viruses.


Subject(s)
Dendritic Cells/metabolism , Genetic Vectors , Simian Immunodeficiency Virus/genetics , Transfection/methods , Animals , Cell Line , Gene Expression , Genetic Engineering , Green Fluorescent Proteins , HIV-1/genetics , Humans , Luminescent Proteins/genetics , Virosomes
14.
EMBO J ; 19(13): 3304-13, 2000 Jul 03.
Article in English | MEDLINE | ID: mdl-10880443

ABSTRACT

Despite CD40's role in stimulating dendritic cells (DCs) for efficient specific T-cell stimulation, its signal transduction components in DCs are still poorly documented. We show that CD40 receptors on human monocyte-derived DCs associate with sphingolipid- and cholesterol-rich plasma membrane microdomains, termed membrane rafts. Following engagement, CD40 utilizes membrane raft-associated Lyn Src family kinase, and possibly other raft-associated Src family kinases, to initiate tyrosine phosphorylation of intracellular substrates. CD40 engagement also leads to a membrane raft-restricted recruitment of tumor necrosis factor (TNF) receptor-associated factor (TRAF) 3 and, to a lesser extent, TRAF2, to CD40's cytoplasmic tail. Thus, the membrane raft structure plays an integral role in proximal events of CD40 signaling in DCs. We demonstrate that stimulation of Src family kinase within membrane rafts initiates a pathway implicating ERK activation, which leads to interleukin (IL)-1alpha/beta and IL-1Ra mRNA production and contributes to p38-dependent IL-12 mRNA production. These results provide the first evidence that membrane rafts play a critical role in initiation of CD40 signaling in DCs, and delineate the outcome of CD40-mediated pathways on cytokine production.


Subject(s)
CD40 Antigens/metabolism , Dendritic Cells/metabolism , Signal Transduction , Cell Membrane/metabolism , Cells, Cultured , Dendritic Cells/immunology , Enzyme Activation , Enzyme Precursors/metabolism , Humans , Interleukin 1 Receptor Antagonist Protein , Interleukin-1/biosynthesis , Interleukin-12/biosynthesis , Intracellular Signaling Peptides and Proteins , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Sialoglycoproteins/biosynthesis , Syk Kinase , src-Family Kinases/metabolism
15.
J Immunol ; 164(12): 6091-5, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10843656

ABSTRACT

The widely expressed transmembrane molecule CD46 is the complement regulatory receptor for C3b as well as the receptor for several pathogens. Beside its binding functions, CD46 is also able to transduce signals. We showed that CD46 aggregation on human T cells induces p120CBL and linker for activation of T cells (LAT) phosphorylation. These two proteins are adaptor proteins known to regulate TCR signaling. p120CBL is a complex adaptor protein involved in negatively regulating signaling events, whereas LAT is a transmembrane adaptor protein found in glycolipid-enriched microdomains essential for T cell activation. Therefore, we investigated if a CD46/TCR costimulation would affect T cell activation. Indeed, CD46/CD3 costimulation strongly promotes T cell proliferation. Therefore, we propose that CD46 acts as a potent costimulatory molecule for human T cells.


Subject(s)
Adaptor Proteins, Signal Transducing , Antigens, CD/physiology , Carrier Proteins/metabolism , Membrane Glycoproteins/physiology , Membrane Proteins , Phosphoproteins/metabolism , Proto-Oncogene Proteins/metabolism , T-Lymphocytes/metabolism , Ubiquitin-Protein Ligases , CD3 Complex/physiology , Cell Line , Humans , Intracellular Fluid/metabolism , Lymphocyte Activation/immunology , Membrane Cofactor Protein , Phosphorylation , Proto-Oncogene Proteins c-cbl , T-Lymphocytes/immunology , Tyrosine/metabolism
16.
J Virol ; 74(10): 4672-8, 2000 May.
Article in English | MEDLINE | ID: mdl-10775604

ABSTRACT

A chimeric fusion protein encompassing the CD46 ectodomain linked to the C-terminal part of the C4b binding protein (C4bp) alpha chain (sCD46-C4bpalpha) was produced in eukaryotic cells. This protein, secreted as a disulfide-linked homo-octamer, was recognized by a panel of anti-CD46 antibodies with varying avidities. Unlike monomeric sCD46, the octameric sCD46-C4bpalpha protein was devoid of complement regulatory activity. However, sCD46-C4bpalpha was able to bind to the measles virus hemagglutinin protein expressed on murine cells with a higher avidity than soluble monomeric sCD46. Moreover, the octameric sCD46-C4bpalpha protein was significantly more efficient than monomeric sCD46 in inhibiting virus binding to CD46, in blocking virus induced cell-cell fusion, and in neutralizing measles virus in vitro. In addition, the octameric sCD46-C4bpalpha protein, but not the monomeric sCD46, fully protected CD46 transgenic mice against a lethal intracranial measles virus challenge.


Subject(s)
Antigens, CD/metabolism , Complement Inactivator Proteins , Glycoproteins , Measles virus/metabolism , Membrane Glycoproteins/metabolism , Receptors, Virus/metabolism , Animals , Antibodies, Viral/metabolism , Antigens, CD/chemistry , Antigens, CD/genetics , Antigens, CD/immunology , CHO Cells , Cell Fusion , Complement Activation , Cricetinae , Hemagglutinins, Viral/metabolism , Measles/prevention & control , Measles virus/immunology , Membrane Cofactor Protein , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Transgenic , Neutralization Tests , Receptors, Complement/chemistry , Receptors, Complement/genetics , Receptors, Complement/metabolism , Receptors, Virus/chemistry , Receptors, Virus/genetics , Receptors, Virus/immunology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism
17.
J Immunol ; 164(4): 1753-60, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10657621

ABSTRACT

Measles virus (MV) infection induces a profound immunosuppression responsible for a high rate of mortality in malnourished children. MV can encounter human dendritic cells (DCs) in the respiratory mucosa or in the secondary lymphoid organs. The purpose of this study was to investigate the consequences of DC infection by MV, particularly concerning their maturation and their ability to generate CD8+ T cell proliferation. We first show that MV-infected Langerhans cells or monocyte-derived DCs undergo a maturation process similarly to the one induced by TNF-alpha or LPS, respectively. CD40 ligand (CD40L) expressed on activated T cells is shown to induce terminal differentiation of DCs into mature effector DCs. In contrast, the CD40L-dependent maturation of DCs is inhibited by MV infection, as demonstrated by CD25, CD69, CD71, CD40, CD80, CD86, and CD83 expression down-regulation. Moreover, the CD40L-induced cytokine pattern in DCs is modified by MV infection with inhibition of IL-12 and IL-1alpha/beta and induction of IL-10 mRNAs synthesis. Using peripheral blood lymphocytes from CD40L-deficient patients, we demonstrate that MV infection of DCs prevents the CD40L-dependent CD8+ T cell proliferation. In such DC-PBL cocultures, inhibition of CD80 and CD86 expression on DCs was shown to require both MV replication and CD40 triggering. Finally, for the first time, MV was shown to inhibit tyrosine-phosphorylation level induced by CD40 activation in DCs. Our data demonstrate that MV replication modifies CD40 signaling in DCs, thus leading to impaired maturation. This phenomenon could play a pivotal role in MV-induced immunosuppression.


Subject(s)
CD40 Antigens/metabolism , Dendritic Cells/immunology , Dendritic Cells/virology , Measles virus/immunology , Membrane Glycoproteins/physiology , CD40 Ligand , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Cell Differentiation/immunology , Cell Division/immunology , Coculture Techniques , Cytokines/biosynthesis , Dendritic Cells/cytology , Dendritic Cells/metabolism , Humans , Immunophenotyping , Langerhans Cells/cytology , Langerhans Cells/immunology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/virology , Ligands , Lymphocyte Activation/immunology , Monocytes/cytology , Monocytes/immunology , Signal Transduction/immunology , Virus Replication/immunology
18.
J Virol ; 74(3): 1373-82, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10627548

ABSTRACT

Measles virus (MV) infection causes acute childhood disease, associated in certain cases with infection of the central nervous system (CNS) and development of neurological disease. To develop a murine model of MV-induced pathology, we generated several lines of transgenic mice ubiquitously expressing as the MV receptor a human CD46 molecule with either a Cyt1 or Cyt2 cytoplasmic tail. All transgenic lines expressed CD46 protein in the brain. Newborn transgenic mice, in contrast to nontransgenic controls, were highly sensitive to intracerebral infection by the MV Edmonston strain. Signs of clinical illness (lack of mobility, tremors, and weight loss) appeared within 5 to 7 days after infection, followed by seizures, paralysis, and death of the infected animals. Virus replication was detected in neurons from infected mice, and virus was reproducibly isolated from transgenic brain tissue. MV-induced apoptosis observed in different brain regions preceded the death of infected animals. Similar results were obtained with mice expressing either a Cyt1 or Cyt2 cytoplasmic tail, demonstrating the ability of different isoforms of CD46 to function as MV receptors in vivo. In addition, maternally transferred immunity delayed death of offspring given a lethal dose of MV. These results document a novel CD46 transgenic murine model where MV neuronal infection is associated with the production of infectious virus, similarly to progressive infectious measles encephalitis seen in immunocompromised patients, and provide a new means to study pathogenesis of MV infection in the CNS.


Subject(s)
Brain/pathology , Encephalitis, Viral/pathology , Measles virus/physiology , Measles/pathology , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Apoptosis , Brain/metabolism , Brain/virology , Disease Models, Animal , Encephalitis, Viral/virology , Female , Humans , Immunity, Maternally-Acquired , Measles/immunology , Measles/virology , Measles virus/isolation & purification , Membrane Cofactor Protein , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Transgenic , Pregnancy , Receptors, Virus/genetics , Receptors, Virus/metabolism , Transgenes , Virus Replication
19.
J Pept Res ; 56(6): 398-408, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11152299

ABSTRACT

A series of conformationally restricted analogs of the hen egg lysozyme (HEL) decapeptide 52-61 in which the conformationally flexible Tyr53 residue was replaced by several more constrained tyrosine and phenylalanine analogs was prepared. Among these tyrosine and phenylalanine analogs were 1,2,3,4-tetrahydro-7-hydroxyisoquinoline-3-carboxylic acid (Htc), 1,2,3,4-tetrahydroisoquinoline-3-carboxylic acid (Tic), 4-amino- 1,2,4,5-tetrahydro-8-hydroxy-2-benzazepine-3-one (Hba), 4-amino-1,2,4,5-tetrahydro-2-benzazepine-3-one (Aba), 2-amino-6-hydroxytetralin-2-carboxylic acid (Hat) and 2-amino-5-hydroxyindan-2-carboxylic acid (Hai) in which the rotations around Calpha-Cbeta and Cbeta-Cgamma were restricted because of cyclization of the side-chain to the backbone. Synthesis of Pht-Hba-Gly-OH using a modification of the Flynn and de Laszlo procedure is described. Analogs of beta-methyltyrosine (beta-MeTyr) in which the side-chains were biased to particular side-chain torsional angles because of substitution at the beta-hydrogens were also prepared. These analogs of HEL[52-61] peptide were tested for their ability to bind to the major histocompatibility complex class II I-Ak molecule and to be recognized in this context by two T-cell hybridomas, specific for the parent peptide HEL[52-61]. The data showed that the conformation and also the configuration of the Tyr53 residue influenced both the binding of the peptide to I-Ak and the recognition of the peptide/I-Ak complex by a T-cell receptor.


Subject(s)
Major Histocompatibility Complex , Peptides/chemistry , Receptors, Antigen, T-Cell/chemistry , Tyrosine/chemistry , Amino Acid Sequence , Animals , Antigen-Presenting Cells/chemistry , B-Lymphocytes/chemistry , Chickens , Mice , Models, Chemical , Molecular Sequence Data , Muramidase/chemistry , Peptide Biosynthesis , Phenylalanine/chemistry , Protein Binding , Protein Conformation , Temperature
20.
J Virol ; 74(9): 4387-93, 2000 May.
Article in English | MEDLINE | ID: mdl-10756053

ABSTRACT

Mortality from measles virus (MV) infection is caused mostly by secondary infections associated with a pronounced immunosuppression. Dendritic cells (DCs) represent a major target of MV and could be involved in immunosuppression. In this study, human monocyte-derived DCs were used to demonstrate that DC apoptosis in MV-infected DC-T-cell cocultures is Fas mediated, whereas apoptotic T cells could not be rescued by blocking the Fas pathway. Two novel consequences of DC apoptosis after MV infection were demonstrated. (i) Fas-mediated apoptosis of DCs facilitates MV release, while CD40 activation enhances MV replication in DCs. Indeed, detailed studies of infectious MV release and intracellular MV nucleoprotein (NP) showed that inhibition of CD40-CD40L ligand interaction blocks NP synthesis. We conclude that the CD40 ligand expressed by activated T cells first enhances MV replication in DCs, and then Fas ligand produced by activated T cells induces Fas-mediated apoptosis of DCs, thus facilitating MV release. (ii) Not only MV-infected DCs but also bystander uninfected DCs undergo a maturation process confirmed by CD1a, CD40, CD80, CD86, CD83, and major histocompatibility complex type II labeling. The bystander maturation effect results from contact and/or engulfment of MV-induced apoptotic DCs by uninfected DCs. A model is proposed to explain how both a specific immune response and immunosuppression can simultaneously occur after MV infection through Fas-mediated apoptosis and CD40 activation of DCs.


Subject(s)
Apoptosis , Dendritic Cells/virology , Measles virus/physiology , fas Receptor/metabolism , CD40 Antigens/immunology , Coculture Techniques , Dendritic Cells/cytology , Dendritic Cells/immunology , Humans , Measles virus/immunology , T-Lymphocytes/immunology , T-Lymphocytes/virology , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...