Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 42(15): 9700-16, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25092922

ABSTRACT

The Epstein-Barr virus (EBV) nuclear antigen 3 family of protein is critical for the EBV-induced primary B-cell growth transformation process. Using a yeast two-hybrid screen we identified 22 novel cellular partners of the EBNA3s. Most importantly, among the newly identified partners, five are known to play direct and important roles in transcriptional regulation. Of these, the Myc-interacting zinc finger protein-1 (MIZ-1) is a transcription factor initially characterized as a binding partner of MYC. MIZ-1 activates the transcription of a number of target genes including the cell cycle inhibitor CDKN2B. Focusing on the EBNA3A/MIZ-1 interaction we demonstrate that binding occurs in EBV-infected cells expressing both proteins at endogenous physiological levels and that in the presence of EBNA3A, a significant fraction of MIZ-1 translocates from the cytoplasm to the nucleus. Moreover, we show that a trimeric complex composed of a MIZ-1 recognition DNA element, MIZ-1 and EBNA3A can be formed, and that interaction of MIZ-1 with nucleophosmin (NPM), one of its coactivator, is prevented by EBNA3A. Finally, we show that, in the presence of EBNA3A, expression of the MIZ-1 target gene, CDKN2B, is downregulated and repressive H3K27 marks are established on its promoter region suggesting that EBNA3A directly counteracts the growth inhibitory action of MIZ-1.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p15/genetics , Epstein-Barr Virus Nuclear Antigens/metabolism , Gene Expression Regulation , Kruppel-Like Transcription Factors/metabolism , Repressor Proteins/metabolism , Transcription, Genetic , Alcohol Oxidoreductases/metabolism , Cell Nucleus/metabolism , Cyclin-Dependent Kinase Inhibitor p15/biosynthesis , DNA-Binding Proteins/metabolism , Down-Regulation , Epstein-Barr Virus Nuclear Antigens/chemistry , HEK293 Cells , HeLa Cells , Histones/metabolism , Humans , Kruppel-Like Transcription Factors/chemistry , Nuclear Proteins/metabolism , Nucleophosmin , Promoter Regions, Genetic , Protein Interaction Domains and Motifs , Proto-Oncogene Proteins c-myc/metabolism , Repressor Proteins/chemistry
2.
PLoS Pathog ; 9(9): e1003599, 2013.
Article in English | MEDLINE | ID: mdl-24086130

ABSTRACT

The interplay between autophagy and intracellular pathogens is intricate as autophagy is an essential cellular response to fight against infections, whereas numerous microbes have developed strategies to escape this process or even exploit it to their own benefit. The fine tuned timing and/or selective molecular pathways involved in the induction of autophagy upon infections could be the cornerstone allowing cells to either control intracellular pathogens, or be invaded by them. We report here that measles virus infection induces successive autophagy signallings in permissive cells, via distinct and uncoupled molecular pathways. Immediately upon infection, attenuated measles virus induces a first transient wave of autophagy, via a pathway involving its cellular receptor CD46 and the scaffold protein GOPC. Soon after infection, a new autophagy signalling is initiated which requires viral replication and the expression of the non-structural measles virus protein C. Strikingly, this second autophagy signalling can be sustained overtime within infected cells, independently of the expression of C, but via a third autophagy input resulting from cell-cell fusion and the formation of syncytia. Whereas this sustained autophagy signalling leads to the autophagy degradation of cellular contents, viral proteins escape from degradation. Furthermore, this autophagy flux is ultimately exploited by measles virus to limit the death of infected cells and to improve viral particle formation. Whereas CD150 dependent virulent strains of measles virus are unable to induce the early CD46/GOPC dependent autophagy wave, they induce and exploit the late and sustained autophagy. Overall, our work describes distinct molecular pathways for an induction of self-beneficial sustained autophagy by measles virus.


Subject(s)
Measles virus/metabolism , Measles virus/pathogenicity , Measles/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing , Antigens, CD/genetics , Antigens, CD/metabolism , Autophagy , Carrier Proteins/genetics , Carrier Proteins/metabolism , Giant Cells/metabolism , Giant Cells/pathology , Giant Cells/virology , Golgi Matrix Proteins , HeLa Cells , Humans , Measles/genetics , Measles/pathology , Measles virus/genetics , Membrane Cofactor Protein/genetics , Membrane Cofactor Protein/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Transport Proteins , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Signaling Lymphocytic Activation Molecule Family Member 1
3.
PLoS One ; 8(2): e56865, 2013.
Article in English | MEDLINE | ID: mdl-23441221

ABSTRACT

Dendritic cells initiate adaptive immune responses, leading either to control cancer by effector T cells or to exacerbate cancer by regulatory T cells that inhibit IFN-γ-mediated Th1-type response. Dendritic cells can also induce Th17-type immunity, mediated by IL-17A. However, the controversial role of this cytokine in cancer requires further investigations. We generated dendritic cells from peripheral blood monocytes to investigate lifespan, phenotype and chemoresistance of dendritic cells, treated with IL-17A with or without IFN-γ. Studying the expression of Bcl-2 family members, we demonstrated that dendritic cells constitutively express one pro-survival Bcl-2 member: MCL1. Immature dendritic cells were CD40(low)HLADR(low) CD1a(+) MCL1(+), did not express CD14, CD68 or BCL2A1, and displayed a short 2-day lifespan. IL-17A-treated DC exhibited a semi-mature (CD40(high) HLADR(low)) pre-M2 (CCL22(+) CD206(+) CD163(+) IL1RN(+) IL-10(-) CXCL10(-) IL-12(-)) mixed (CD1a(+) CD14+ CD68(+)) macrophage-dendritic cell phenotype. They efficiently exerted mannose receptor-mediated endocytosis and did not produce superoxide anions, in the absence of TLR engagement. Interestingly, IL-17A promoted a long-term survival of dendritic cells, beyond 12 days, that correlated to BCL2A1 induction, a pro-survival Bcl-2 family member. BCL2A1 transcription was activated by NF-κB, downstream of IL-17A transduction. Thus, immature dendritic cells only express MCL1, whereas IL-17A-treated dendritic cells concomitantly expressed two pro-survival Bcl-2 family members: MCL1 and BCL2A1. These latter developed chemoresistance to 11 of the 17 chemotherapy agents tested. However, high doses of either vinblastine or cytarabine decreased MCL1 expression and induced dendritic cell death. When IL-17A is produced in vivo, administration of anti-IL-17A biotherapy may impair dendritic cell survival by targeting BCL2A1 expression. Consequently, depending on the effector or regulatory role of dendritic cells, blocking IL-17A, may be either dangerous or beneficial for cancer outcomes, thus contributing to the apparent controversy around the role of IL-17A in cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Dendritic Cells/cytology , Dendritic Cells/drug effects , Drug Resistance, Neoplasm , Interleukin-17/pharmacology , Monocytes/cytology , Cell Differentiation , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Dendritic Cells/metabolism , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation/drug effects , Humans , Immunophenotyping , Interferon-gamma/pharmacology , Minor Histocompatibility Antigens , Myeloid Cell Leukemia Sequence 1 Protein , NF-kappa B/metabolism , Phenotype , Protein Transport/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Transcription, Genetic
4.
Methods ; 58(4): 349-59, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22898364

ABSTRACT

Comparative interactomics is a strategy for inferring potential interactions among orthologous proteins or "interologs". Herein we focus, in contrast to standard homology-based inference, on the divergence of protein interaction profiles among closely related organisms, showing that the approach can correlate specific traits to phenotypic differences. As a model, this new comparative interactomic approach was applied at a large scale to human papillomaviruses (HPVs) proteins. The oncogenic potential of HPVs is mainly determined by the E6 and E7 early proteins. We have mapped and overlapped the virus-host protein interaction networks of E6 and E7 proteins from 11 distinct HPV genotypes, selected for their different tropisms and pathologies. We generated robust and comprehensive datasets by combining two orthogonal protein interaction assays: yeast two-hybrid (Y2H), and our recently described "high-throughput Gaussia princeps protein complementation assay" (HT-GPCA). HT-GPCA detects protein interaction by measuring the interaction-mediated reconstitution of activity of a split G. princeps luciferase. Hierarchical clustering of interaction profiles recapitulated HPV phylogeny and was used to correlate specific virus-host interaction profiles with pathological traits, reflecting the distinct carcinogenic potentials of different HPVs. This comparative interactomics constitutes a reliable and powerful strategy to decipher molecular relationships in virtually any combination of microorganism-host interactions.


Subject(s)
Alphapapillomavirus/physiology , Host-Pathogen Interactions , Luciferases/genetics , Plant Proteins/genetics , Two-Hybrid System Techniques , Alphapapillomavirus/genetics , Arecaceae/enzymology , Biomarkers/metabolism , Cluster Analysis , Genotype , HEK293 Cells , Humans , Luciferases/biosynthesis , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins/genetics , Papillomavirus E7 Proteins/metabolism , Phylogeny , Plant Proteins/biosynthesis , Protein Binding , Protein Interaction Mapping , Protein Interaction Maps , Proteome/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Viral Tropism
5.
Autophagy ; 8(7): 1136-7, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22722598

ABSTRACT

Several intracellular pathogens have the ability to avoid or exploit the otherwise destructive process of autophagy. RNA viruses are constantly confronted with cellular autophagy, and several of them hijack autophagy during the infectious cycle to improve their own replication. Nevertheless, our knowledge of viral molecular strategies used to manipulate autophagy remains limited. Our study allowed the identification of molecular interactions between 44 autophagy-associated proteins and 83 viral proteins belonging to five different RNA virus families. This interactome revealed that the autophagy network machinery is highly targeted by RNA viruses. Interestingly, whereas some autophagy-associated proteins are targeted by only one RNA virus family, others are recurrent targets of several families. Among them, we found IRGM as the most targeted autophagy-associated protein. Downregulation of IRGM expression prevents autophagy induction by measles virus, HCV and HIV-1, and compromises viral replication. Our work combined interactomic and analytical approaches to identify potential pathogen virulence factors targeting autophagy.

6.
Nucleic Acids Res ; 40(14): 6834-49, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22505578

ABSTRACT

The Epstein-Barr Virus (EBV) protein EB2 (also called Mta, SM and BMLF1), is an essential nuclear protein produced during the replicative cycle of EBV. EB2 is required for the efficient cytoplasmic accumulation of viral mRNAs derived from intronless genes. EB2 is an RNA-binding protein whose expression has been shown to influence RNA stability, splicing, nuclear export and translation. Using a yeast two-hybrid screen, we have identified three SR proteins, SF2/ASF, 9G8 and SRp20, as cellular partners of EB2. Then, by using siRNA to deplete cells of specific SR proteins, we found that SRp20 plays an essential role in the processing of several model mRNAs: the Renilla luciferase reporter mRNA, the human ß-globin cDNA transcript and two EBV late mRNAs. These four mRNAs were previously found to be highly dependent on EB2 for their efficient cytoplasmic accumulation. Here, we show that SRp20 depletion results in an increase in the accumulation of these mRNAs, which correlates with an absence of additive effect of EB2, suggesting that EB2 functions by antagonizing SRp20. Moreover, by using RNA-immunoprecipitation assays we found that EB2 enhances the association of SRp20 with the ß-globin transcript suggesting that EB2 acts by stabilizing SRp20's labile interactions with the RNA.


Subject(s)
Phosphoproteins/metabolism , RNA, Messenger/metabolism , RNA, Viral/metabolism , RNA-Binding Proteins/metabolism , Trans-Activators/metabolism , Cell Nucleus/virology , Cytoplasm/metabolism , Cytoplasm/virology , Down-Regulation , HEK293 Cells , HeLa Cells , Humans , Luciferases, Renilla/genetics , Mutation , Nuclear Proteins/metabolism , Phosphoproteins/chemistry , Phosphoproteins/genetics , Protein Interaction Domains and Motifs , RNA-Binding Proteins/antagonists & inhibitors , Serine-Arginine Splicing Factors , Trans-Activators/chemistry , Trans-Activators/genetics , Two-Hybrid System Techniques , beta-Globins/genetics
7.
Methods Mol Biol ; 812: 103-20, 2012.
Article in English | MEDLINE | ID: mdl-22218856

ABSTRACT

Using global approaches and high-throughput technologies in virology brings a new vision of the infections physiology and allows the identification of cellular factors, mandatory for viral life cycle, that could be targeted by original therapeutic agents. It opens perspectives for the treatment of viral infections by acting on cellular pathways that the virus must use for its own replication. Combining these new molecules with classical antiviral drugs and immunomodulators diversifies and enlarges the antiviral arsenal and contributes to fight drug resistance. Our laboratory and others are constructing virus-human interactomes to propose a comprehensive analysis of viral infection at the cellular level. Studying these infection maps, where the viral infection can be visualized as perturbation of the human protein-protein interaction network, and identifying the biological functions that are impaired by these perturbations may lead to discovery of new therapeutic targets. These virus-human interaction maps are constructed in a stringent yeast two-hybrid system by screening human cDNA libraries with viral proteins as bait and integrating interactions mined from literature and public databases.


Subject(s)
Host-Pathogen Interactions , Two-Hybrid System Techniques , Viral Proteins/metabolism , Virus Physiological Phenomena , Viruses/metabolism , DNA, Viral/genetics , DNA, Viral/isolation & purification , Databases, Protein , Gene Library , Humans , Open Reading Frames/genetics , Transformation, Genetic , Viral Proteins/genetics , Viruses/genetics , Yeasts/cytology , Yeasts/genetics
8.
PLoS Pathog ; 7(12): e1002422, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22174682

ABSTRACT

Autophagy is a conserved degradative pathway used as a host defense mechanism against intracellular pathogens. However, several viruses can evade or subvert autophagy to insure their own replication. Nevertheless, the molecular details of viral interaction with autophagy remain largely unknown. We have determined the ability of 83 proteins of several families of RNA viruses (Paramyxoviridae, Flaviviridae, Orthomyxoviridae, Retroviridae and Togaviridae), to interact with 44 human autophagy-associated proteins using yeast two-hybrid and bioinformatic analysis. We found that the autophagy network is highly targeted by RNA viruses. Although central to autophagy, targeted proteins have also a high number of connections with proteins of other cellular functions. Interestingly, immunity-associated GTPase family M (IRGM), the most targeted protein, was found to interact with the autophagy-associated proteins ATG5, ATG10, MAP1CL3C and SH3GLB1. Strikingly, reduction of IRGM expression using small interfering RNA impairs both Measles virus (MeV), Hepatitis C virus (HCV) and human immunodeficiency virus-1 (HIV-1)-induced autophagy and viral particle production. Moreover we found that the expression of IRGM-interacting MeV-C, HCV-NS3 or HIV-NEF proteins per se is sufficient to induce autophagy, through an IRGM dependent pathway. Our work reveals an unexpected role of IRGM in virus-induced autophagy and suggests that several different families of RNA viruses may use common strategies to manipulate autophagy to improve viral infectivity.


Subject(s)
Autophagy/physiology , GTP-Binding Proteins/metabolism , RNA Virus Infections/metabolism , RNA Virus Infections/transmission , RNA Viruses/metabolism , Base Sequence , Blotting, Western , Computational Biology , GTP-Binding Proteins/genetics , HeLa Cells , Humans , Microscopy, Confocal , Molecular Sequence Data , Open Reading Frames/genetics , RNA Virus Infections/genetics , RNA Viruses/genetics , RNA, Small Interfering , Transfection , Two-Hybrid System Techniques , Viral Proteins/metabolism
9.
BMC Microbiol ; 11: 234, 2011 Oct 20.
Article in English | MEDLINE | ID: mdl-22014111

ABSTRACT

BACKGROUND: The genus Flavivirus encompasses more than 50 distinct species of arthropod-borne viruses, including several major human pathogens, such as West Nile virus, yellow fever virus, Japanese encephalitis virus and the four serotypes of dengue viruses (DENV type 1-4). Each year, flaviviruses cause more than 100 million infections worldwide, some of which lead to life-threatening conditions such as encephalitis or haemorrhagic fever. Among the viral proteins, NS3 and NS5 proteins constitute the major enzymatic components of the viral replication complex and are essential to the flavivirus life cycle. RESULTS: We report here the results of a high-throughput yeast two-hybrid screen to identify the interactions between human host proteins and the flavivirus NS3 and NS5 proteins. Using our screen results and literature curation, we performed a global analysis of the NS3 and NS5 cellular targets based on functional annotation with the Gene Ontology features. We finally created the first flavivirus NS3 and NS5 proteins interaction network and analysed the topological features of this network. Our proteome mapping screen identified 108 human proteins interacting with NS3 or NS5 proteins or both. The global analysis of the cellular targets revealed the enrichment of host proteins involved in RNA binding, transcription regulation, vesicular transport or innate immune response regulation. CONCLUSIONS: We proposed that the selective disruption of these newly identified host/virus interactions could represent a novel and attractive therapeutic strategy in treating flavivirus infections. Our virus-host interaction map provides a basis to unravel fundamental processes about flavivirus subversion of the host replication machinery and/or immune defence strategy.


Subject(s)
Host-Pathogen Interactions , Protein Interaction Mapping , Viral Nonstructural Proteins/metabolism , Flavivirus/pathogenicity , Flavivirus Infections/virology , HEK293 Cells , High-Throughput Screening Assays , Humans , RNA Helicases/metabolism , Serine Endopeptidases/metabolism , Two-Hybrid System Techniques
10.
J Virol ; 85(24): 13010-8, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21994455

ABSTRACT

The influenza virus transcribes and replicates its genome inside the nucleus of infected cells. Both activities are performed by the viral RNA-dependent RNA polymerase that is composed of the three subunits PA, PB1, and PB2, and recent studies have shown that it requires host cell factors to transcribe and replicate the viral genome. To identify these cellular partners, we generated a comprehensive physical interaction map between each polymerase subunit and the host cellular proteome. A total of 109 human interactors were identified by yeast two-hybrid screens, whereas 90 were retrieved by literature mining. We built the FluPol interactome network composed of the influenza virus polymerase (PA, PB1, and PB2) and the nucleoprotein NP and 234 human proteins that are connected through 279 viral-cellular protein interactions. Analysis of this interactome map revealed enriched cellular functions associated with the influenza virus polymerase, including host factors involved in RNA polymerase II-dependent transcription and mRNA processing. We confirmed that eight influenza virus polymerase-interacting proteins are required for virus replication and transcriptional activity of the viral polymerase. These are involved in cellular transcription (C14orf166, COPS5, MNAT1, NMI, and POLR2A), translation (EIF3S6IP), nuclear transport (NUP54), and DNA repair (FANCG). Conversely, we identified PRKRA, which acts as an inhibitor of the viral polymerase transcriptional activity and thus is required for the cellular antiviral response.


Subject(s)
Host-Pathogen Interactions , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H5N1 Subtype/pathogenicity , Protein Interaction Mapping , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/metabolism , Humans , Protein Binding , Two-Hybrid System Techniques , Virus Replication
11.
Med Sci (Paris) ; 27(1): 41-7, 2011 Jan.
Article in French | MEDLINE | ID: mdl-21299961

ABSTRACT

Autophagy is a highly conserved, self-degradative pathway for clearance and recycling of cytoplasmic contents. This ubiquitous cell intrinsic process can be used as a defence mechanism against intracellular pathogens. Indeed autophagy is increased upon pathogen detection, and experimental extinction in vitro and in vivo of this cellular process has been demonstrated as a crucial role to control intracellular pathogens. Co-evolution between host-cells and pathogens has selected numerous micoorganisms able to avoid or usurp autophagy to their own benefit. Understanding mechanisms underlying the anti-microbial properties of autophagy as well as those used by certain pathogens to escape this cellular process might be crucial to manipulate this cellular function in order to prevent or treat infectious diseases.


Subject(s)
Autophagy , Host-Pathogen Interactions/physiology , Animals , Bacterial Physiological Phenomena , Cells/microbiology , Cells/parasitology , Cells/virology , Eukaryotic Cells/physiology , HIV/physiology , Humans , Interferon Type I/physiology , Membrane Fusion , Models, Biological , Phagosomes/physiology , Proteasome Endopeptidase Complex/physiology , Receptors, Pattern Recognition/physiology , Selection, Genetic , Small Ubiquitin-Related Modifier Proteins/physiology , Unfolded Protein Response/physiology
12.
Curr Opin Virol ; 1(1): 6-12, 2011 Jul.
Article in English | MEDLINE | ID: mdl-22440562

ABSTRACT

Entry into a cell submits viruses to detection by pattern recognition receptors (PRRs) leading to an early innate anti-viral response. Several viruses evolved strategies to avoid or subvert PRR recognition at the step of virus entry to promote infection. Whereas viruses mostly escape from soluble PRR detection, endocytic/phagocytic PRRs, such as the mannose receptor or DC-SIGN, are commonly used for virus entry. Moreover, virion-incorporated proteins may also offer viruses a way to dampen anti-viral innate immunity upon virus entry, and entering viruses might usurp autophagy to improve their own infectivity.


Subject(s)
Immunity, Innate , Virus Diseases/immunology , Virus Diseases/virology , Virus Internalization , Animals , Humans , Receptors, Pattern Recognition/genetics , Receptors, Pattern Recognition/immunology , Receptors, Virus/genetics , Receptors, Virus/immunology , Viral Proteins/genetics , Viral Proteins/immunology , Viruses/genetics , Viruses/immunology
13.
Nucleic Acids Res ; 39(Database issue): D569-75, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21081560

ABSTRACT

Fast viral adaptation and the implication of this rapid evolution in the emergence of several new infectious diseases have turned this issue into a major challenge for various research domains. Indeed, viruses are involved in the development of a wide range of pathologies and understanding how viruses and host cells interact in the context of adaptation remains an open question. In order to provide insights into the complex interactions between viruses and their host organisms and namely in the acquisition of novel functions through exchanges of genetic material, we developed the PhEVER database. This database aims at providing accurate evolutionary and phylogenetic information to analyse the nature of virus-virus and virus-host lateral gene transfers. PhEVER (http://pbil.univ-lyon1.fr/databases/phever) is a unique database of homologous families both (i) between sequences from different viruses and (ii) between viral sequences and sequences from cellular organisms. PhEVER integrates extensive data from up-to-date completely sequenced genomes (2426 non-redundant viral genomes, 1007 non-redundant prokaryotic genomes, 43 eukaryotic genomes ranging from plants to vertebrates) and offers a clustering of proteins into homologous families containing at least one viral sequences, as well as alignments and phylogenies for each of these families. Public access to PhEVER is available through its webpage and through all dedicated ACNUC retrieval systems.


Subject(s)
Databases, Genetic , Evolution, Molecular , Host-Pathogen Interactions/genetics , Viruses/genetics , Cluster Analysis , Gene Transfer, Horizontal , Genes, Viral , Genome, Viral , Genomics , Phylogeny , Sequence Homology , User-Computer Interface , Viral Proteins/chemistry , Viral Proteins/classification , Viral Proteins/genetics , Viruses/classification
14.
Virus Res ; 152(1-2): 115-25, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20600391

ABSTRACT

While the antiviral response during measles virus (MeV) infection is documented, the contribution of the hosting cell type to the type I interferon (IFN-alpha/beta) response is still not clearly established. Here, we report that a signature heterogeneity of the IFN-alpha/beta response according to the cell type. The MeV tropism dictated by the expression of appropriate cellular receptor appeared to be crucial for epithelial cells. For conventional DCs (cDCs), the maturation state played a prominent role. In response to both wild type MeV isolates and laboratory/vaccine strains, immature cDCs produced higher levels of IFN-alpha than mature cDCs, despite the reduced expression levels of both CD46 and CD150 receptors by the former ones. While in epithelial cells and cDCs the MeV transcription was required to activate the IFN-alpha/beta response, plasmacytoid DCs (pDCs) rapidly produced large amounts of IFN-alpha mostly independently of the viral infection cycle. This argues for a significant contribution of pDCs in response to MeV infection and/or vaccination.


Subject(s)
Cell Differentiation , Dendritic Cells/immunology , Endocytosis , Epithelial Cells/immunology , Interferon Type I/immunology , Measles virus/physiology , Measles/immunology , Receptors, Virus/immunology , Antigens, CD/immunology , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/virology , Epithelial Cells/cytology , Epithelial Cells/virology , Humans , Measles/physiopathology , Measles/virology , Measles virus/genetics , Measles virus/immunology , Membrane Cofactor Protein/immunology , Receptors, Cell Surface/immunology , Signaling Lymphocytic Activation Molecule Family Member 1 , Viral Tropism
15.
Med Sci (Paris) ; 26(6-7): 603-9, 2010.
Article in French | MEDLINE | ID: mdl-20619162

ABSTRACT

Infection caused by pathogens kills millions of people every year. Comprehensive understanding of molecular pathogen-host interactions, i.e. the infectome, is one of the key steps towards the development of novel diagnostic, therapeutic and preventive strategies. In this quest, progress in high-throughput << omics >> technologies applied to pathogens, i.e. infectomics, opens new perspectives toward systemic understanding of perturbations induced during infection. Deciphering the pathogen-host system also relies on the analytical and predictive power of molecular systems biology and by developing in silico models taking into account the whole picture of the molecules and their interactions. In this context, we have reconstructed a prototype of the human virtual infected cell based on 30 years of intensive research in the field of molecular virology. This model contains more than one hundred viral infectomes, including major human pathogens (HCV, HBV, HIV, HHV, HPV) and has led to the generation of novel systems-level hypotheses that could be suitable for the development of innovative antiviral strategies based on the control of cellular functions.


Subject(s)
Antiviral Agents/chemical synthesis , Antiviral Agents/therapeutic use , User-Computer Interface , Virus Diseases/genetics , Genome, Viral , Humans , Models, Genetic , Proteome , Viral Proteins/genetics , Virus Diseases/drug therapy , Viruses/genetics
16.
Autophagy ; 6(2): 299-300, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20087059

ABSTRACT

Autophagy is a degradative mechanism involved in cell protection against invading pathogens. Although the autophagic process is well characterized, the molecular pathways leading to its activation upon pathogen binding remain poorly understood. Our recent work demonstrates that the cell surface pathogen receptor CD46 induces autophagy upon pathogen recognition. The molecular pathway linking CD46 to the autophagosome machinery relies on the scaffold protein GOPC and on the autophagosome formation complex Beclin 1/VPS34. The CD46-dependent autophagy is critical to an early control of infection.


Subject(s)
Autophagy/immunology , Membrane Cofactor Protein/immunology , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Host-Pathogen Interactions , Mice , Phagosomes/metabolism
17.
Cell Host Microbe ; 6(4): 354-66, 2009 Oct 22.
Article in English | MEDLINE | ID: mdl-19837375

ABSTRACT

Autophagy is a highly regulated self-degradative mechanism required at a basal level for intracellular clearance and recycling of cytoplasmic contents. Upon intracellular pathogen invasion, autophagy can be induced as an innate immune mechanism to control infection. Nevertheless, pathogens have developed strategies to avoid or hijack autophagy for their own benefit. The molecular pathways inducing autophagy in response to infection remain poorly documented. We report here that the engagement of CD46, a ubiquitous human surface receptor able to bind several different pathogens, is sufficient to induce autophagy. CD46-Cyt-1, one of the two C-terminal splice variants of CD46, is linked to the autophagosome formation complex VPS34/Beclin1 via its interaction with the scaffold protein GOPC. Measles virus and group A Streptococcus, two CD46-binding pathogens, induce autophagy through a CD46-Cyt-1/GOPC pathway. Thus, upon microorganism recognition, a cell surface pathogen receptor can directly trigger autophagy, a critical step to control infection.


Subject(s)
Autophagy , Measles virus/immunology , Membrane Cofactor Protein/immunology , Streptococcus pyogenes/immunology , Adaptor Proteins, Signal Transducing , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Carrier Proteins/metabolism , Golgi Matrix Proteins , Humans , Membrane Proteins/metabolism , Membrane Transport Proteins , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding , Protein Interaction Mapping
18.
BMC Res Notes ; 2: 220, 2009 Oct 29.
Article in English | MEDLINE | ID: mdl-19874608

ABSTRACT

BACKGROUND: High-throughput screening of protein-protein interactions opens new systems biology perspectives for the comprehensive understanding of cell physiology in normal and pathological conditions. In this context, yeast two-hybrid system appears as a promising approach to efficiently reconstruct protein interaction networks at the proteome-wide scale. This protein interaction screening method generates a large amount of raw sequence data, i.e. the ISTs (Interaction Sequence Tags), which urgently need appropriate tools for their systematic and standardised analysis. FINDINGS: We develop pISTil, a bioinformatics pipeline combined with a user-friendly web-interface: (i) to establish a standardised system to analyse and to annotate ISTs generated by two-hybrid technologies with high performance and flexibility and (ii) to provide high-quality protein-protein interaction datasets for systems-level approach. This pipeline has been validated on a large dataset comprising more than 11.000 ISTs. As a case study, a detailed analysis of ISTs obtained from yeast two-hybrid screens of Hepatitis C Virus proteins against human cDNA libraries is also provided. CONCLUSION: We have developed pISTil, an open source pipeline made of a collection of several applications governed by a Perl script. The pISTil pipeline is intended to laboratories, with IT-expertise in system administration, scripting and database management, willing to automatically process large amount of ISTs data for accurate reconstruction of protein interaction networks in a systems biology perspective. pISTil is publicly available for download at http://sourceforge.net/projects/pistil.

19.
Nucleic Acids Res ; 37(Database issue): D661-8, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18984613

ABSTRACT

Infectious diseases caused by viral agents kill millions of people every year. The improvement of prevention and treatment of viral infections and their associated diseases remains one of the main public health challenges. Towards this goal, deciphering virus-host molecular interactions opens new perspectives to understand the biology of infection and for the design of new antiviral strategies. Indeed, modelling of an infection network between viral and cellular proteins will provide a conceptual and analytic framework to efficiently formulate new biological hypothesis at the proteome scale and to rationalize drug discovery. Therefore, we present the first release of VirHostNet (Virus-Host Network), a public knowledge base specialized in the management and analysis of integrated virus-virus, virus-host and host-host interaction networks coupled to their functional annotations. VirHostNet integrates an extensive and original literature-curated dataset of virus-virus and virus-host interactions (2671 non-redundant interactions) representing more than 180 distinct viral species and one of the largest human interactome (10,672 proteins and 68,252 non-redundant interactions) reconstructed from publicly available data. The VirHostNet Web interface provides appropriate tools that allow efficient query and visualization of this infected cellular network. Public access to the VirHostNet knowledge-based system is available at http://pbildb1.univ-lyon1.fr/virhostnet.


Subject(s)
Databases, Protein , Host-Pathogen Interactions , Protein Interaction Mapping , Viral Proteins/metabolism , Internet , Proteome/metabolism , User-Computer Interface , Virus Diseases/metabolism , Virus Diseases/virology , Virus Physiological Phenomena
20.
Nat Med ; 14(1): 81-7, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18157139

ABSTRACT

IL-17A is a T cell-specific cytokine that is involved in chronic inflammations, such as Mycobacterium infection, Crohn's disease, rheumatoid arthritis and multiple sclerosis. Mouse models have explained the molecular basis of IL-17A production and have shown that IL-17A has a positive effect not only on granuloma formation and neurodegeneration through unknown mechanisms, but also on bone resorption through Receptor activator of NF-kappaB ligand (RANKL) induction in osteoblasts. Langerhans cell histiocytosis (LCH) is a rare disease of unknown etiology, lacking an animal model, that cumulates symptoms that are found separately in various IL-17A-related diseases, such as aggressive chronic granuloma formation, bone resorption and soft tissue lesions with occasional neurodegeneration. We examined IL-17A in the context of LCH and found that there were high serum levels of IL-17A during active LCH and unexpected IL-17A synthesis by dendritic cells (DCs), the major cell type in LCH lesions. We also found an IL-17A-dependent pathway for DC fusion, which was highly potentiated by IFN-gamma and led to giant cells expressing three major tissue-destructive enzymes: tartrate resistant acidic phosphatase and matrix metalloproteinases 9 and 12. IFN-gamma expression has been previously documented in LCH and observed in IL-17A-related diseases. Notably, serum IL-17A-dependent fusion activity correlates with LCH activity. Thus, IL-17A and IL-17A-stimulated DCs represent targets that may have clinical value in the treatment of LCH and other IL-17A-related inflammatory disorders.


Subject(s)
Dendritic Cells/metabolism , Histiocytosis, Langerhans-Cell/pathology , Interleukin-17/metabolism , Animals , Arthritis, Rheumatoid/metabolism , Cell Fusion , Humans , Inflammation , Interferon-gamma/metabolism , Lymphocyte Activation , Lymphocytes/metabolism , Mice , Monocytes/metabolism , Mycobacterium/metabolism , Oligonucleotide Array Sequence Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...