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










Database
Language
Publication year range
1.
Arch Virol ; 156(2): 219-34, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21069396

ABSTRACT

We have modeled a 3D structure for the C-type lectin domain of the African swine fever virus protein EP153R, based on the structure of CD69, CD94 and Ly49A cell receptors, and this model predicts that a dimer of EP153R may establish an asymmetric interaction with one MHC-I molecule. A functional consequence of this interaction could be the modulation of MHC-I expression. By using both transfection and virus infection experiments, we demonstrate here that EP153R inhibits MHC-I membrane expression, most probably by impairing the exocytosis process, without affecting the synthesis or glycosylation of MHC antigens. Interestingly, the EP153-mediated control of MHC requires the intact configuration of the lectin domain of the viral protein, and specifically the R133 residue. Interference of EP153R gene expression during virus infection and studies using virus recombinants with the EP153R gene deleted further support the inhibitory role of the viral lectin on the expression of MHC-I antigens.


Subject(s)
African Swine Fever Virus/immunology , Histocompatibility Antigens Class I/metabolism , Lectins, C-Type/chemistry , Lectins, C-Type/immunology , Viral Proteins/chemistry , Viral Proteins/immunology , African Swine Fever Virus/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Line , DNA, Viral/genetics , Dimerization , Down-Regulation , Endoplasmic Reticulum/virology , Exocytosis , Genes, Viral , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class II/chemistry , Histocompatibility Antigens Class II/metabolism , Humans , Lectins, C-Type/genetics , Mice , Models, Molecular , Molecular Sequence Data , Protein Interaction Domains and Motifs , Protein Structure, Quaternary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Sequence Homology, Amino Acid , Static Electricity , Structural Homology, Protein , Swine , Viral Proteins/genetics
2.
PLoS Pathog ; 5(8): e1000562, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19714237

ABSTRACT

African swine fever virus (ASFV), like other complex DNA viruses, deploys a variety of strategies to evade the host's defence systems, such as inflammatory and immune responses and cell death. Here, we analyse the modifications in the translational machinery induced by ASFV. During ASFV infection, eIF4G and eIF4E are phosphorylated (Ser1108 and Ser209, respectively), whereas 4E-BP1 is hyperphosphorylated at early times post infection and hypophosphorylated after 18 h. Indeed, a potent increase in eIF4F assembly is observed in ASFV-infected cells, which is prevented by rapamycin treatment. Phosphorylation of eIF4E, eIF4GI and 4E-BP1 is important to enhance viral protein production, but is not essential for ASFV infection as observed in rapamycin- or CGP57380-treated cells. Nevertheless, eIF4F components are indispensable for ASFV protein synthesis and virus spread, since eIF4E or eIF4G depletion in COS-7 or Vero cells strongly prevents accumulation of viral proteins and decreases virus titre. In addition, eIF4F is not only activated but also redistributed within the viral factories at early times of infection, while eIF4G and eIF4E are surrounding these areas at late times. In fact, other components of translational machinery such as eIF2alpha, eIF3b, eIF4E, eEF2 and ribosomal P protein are enriched in areas surrounding ASFV factories. Notably, the mitochondrial network is polarized in ASFV-infected cells co-localizing with ribosomes. Thus, translation and ATP synthesis seem to be coupled and compartmentalized at the periphery of viral factories. At later times after ASFV infection, polyadenylated mRNAs disappear from the cytoplasm of Vero cells, except within the viral factories. The distribution of these pools of mRNAs is similar to the localization of viral late mRNAs. Therefore, degradation of cellular polyadenylated mRNAs and recruitment of the translation machinery to viral factories may contribute to the inhibition of host protein synthesis, facilitating ASFV protein production in infected cells.


Subject(s)
African Swine Fever Virus/physiology , African Swine Fever/metabolism , African Swine Fever/virology , Eukaryotic Initiation Factors/metabolism , Animals , COS Cells , Caspase 3/metabolism , Chlorocebus aethiops , Immunohistochemistry , Mitochondria/metabolism , Phosphoproteins/metabolism , Phosphorylation , RNA Cap-Binding Proteins/metabolism , RNA, Messenger/metabolism , RNA, Viral/metabolism , Ribosomal Proteins/metabolism , Vero Cells
3.
J Virol ; 83(2): 969-80, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19004945

ABSTRACT

During a viral infection, reprogramming of the host cell gene expression pattern is required to establish an adequate antiviral response. The transcriptional coactivators p300 and CREB binding protein (CBP) play a central role in this regulation by promoting the assembly of transcription enhancer complexes to specific promoters of immune and proinflammatory genes. Here we show that the protein A238L encoded by African swine fever virus counteracts the host cell inflammatory response through the control of p300 transactivation during the viral infection. We demonstrate that A238L inhibits the expression of the inflammatory regulators cyclooxygenase-2 (COX-2) and tumor necrosis factor alpha (TNF-alpha) by preventing the recruitment of p300 to the enhanceosomes formed on their promoters. Furthermore, we report that A238L inhibits p300 activity during the viral infection and that its amino-terminal transactivation domain is essential in the A238L-mediated inhibition of the inflammatory response. Importantly, we found that the residue serine 384 of p300 is required for the viral protein to accomplish its inhibitory function and that ectopically expressed PKC-theta completely reverts this inhibition, thus indicating that this signaling pathway is disrupted by A238L during the viral infection. Furthermore, we show here that A238L does not affect PKC-theta enzymatic activity, but the molecular mechanism of this viral inhibition relies on the lack of interaction between PKC-theta and p300. These findings shed new light on how viruses alter the host cell antiviral gene expression pattern through the blockade of the p300 activity, which represents a new and sophisticated viral mechanism to evade the inflammatory and immune defense responses.


Subject(s)
African Swine Fever Virus/immunology , African Swine Fever Virus/physiology , E1A-Associated p300 Protein/antagonists & inhibitors , Protein Kinase C/antagonists & inhibitors , Viral Proteins/metabolism , Animals , Chlorocebus aethiops , Cyclooxygenase 2/biosynthesis , Protein Binding , Protein Interaction Mapping , Tumor Necrosis Factor-alpha/biosynthesis , Vero Cells
4.
Antivir Ther ; 13(7): 909-17, 2008.
Article in English | MEDLINE | ID: mdl-19043925

ABSTRACT

BACKGROUND: Antiviral compounds are needed in the control of many animal and human diseases. METHODS: We analysed the effect of the antitumoural drug lauryl gallate on the infectivity of the African swine fever virus among other DNA (herpes simplex and vaccinia) and RNA (influenza, porcine transmissible gastroenteritis and Sindbis) viruses, paying attention to its effect on the viability of the corresponding host cells. RESULTS: Viral production was strongly inhibited in different cell lines at non-toxic concentrations of the drug (1-10 microM), reducing the titres 3->5 log units depending on the multiplicity of infection. In our model system (African swine fever virus in Vero cells), the addition of the drug 1 h before virus adsorption completely abolished virus productivity in a one-step growth virus cycle. Interestingly, no inhibitory effect was observed when lauryl gallate was added after 5-8 h post-infection. Both cellular and viral DNA synthesis and late viral transcription were inhibited by the drug; however, the early viral protein synthesis and the virus-mediated increase of p53 remained unaffected. Activation of the apoptotic effector caspase-3 was not detected after lauryl gallate treatment of Vero cells. Furthermore, the presence of the drug abrogated the activation of this protease induced by the virus infection. CONCLUSIONS: Lauryl gallate is a powerful antiviral agent against several pathogens of clinical and veterinary importance. The overall results indicate that a cellular factor or function might be the target of the antiviral action of alkyl gallates.


Subject(s)
Antiviral Agents/pharmacology , DNA Viruses/drug effects , Gallic Acid/analogs & derivatives , African Swine Fever Virus/drug effects , African Swine Fever Virus/physiology , Animals , Cell Line , Chlorocebus aethiops , Cricetinae , DNA Viruses/physiology , DNA, Viral/biosynthesis , DNA, Viral/drug effects , Gallic Acid/pharmacology , Proteins/drug effects , Proteins/genetics , Proteins/metabolism , RNA Viruses/drug effects , RNA Viruses/physiology , Vero Cells , Viral Proteins/drug effects , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication
5.
J Virol ; 80(21): 10487-96, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17041221

ABSTRACT

Uncontrolled generation of nitric oxide (NO) by inducible nitric-oxide synthase (iNOS) can cause damage to host cells and inflammation, two undesirable events for virus spreading. African swine fever virus (ASFV) infection regulates iNOS-induced gene expression through the synthesis of the A238L virus protein. We here explored the role of A238L, an NF-kappaB and NFAT inhibitor, in the regulation of iNOS transcription in macrophages. NO production and iNOS mRNA and protein levels as well as iNOS promoter activity after lipopolysaccharide (LPS)-gamma interferon (IFN-gamma) treatment were down-regulated both during ASFV infection and in Raw 264.7 cells stably expressing the viral protein. Overexpression of p300, but not of a histone acetyltransferase (HAT) defective mutant, reverted the A238L-mediated inhibition of both basal and LPS-IFN-gamma-induced iNOS promoter activity. Following stimulation with LPS-IFN-gamma, p65 and p300 interaction was abolished in Raw-A238L cells. Expression of A238L also inhibited p65/relA and p300 binding to the distal NF-kappaB sequence of the iNOS promoter together with p65 acetylation. Finally, A238L abrogated p300 transactivation mediated by a GAL4-p300 construction. These results provide evidence for an unique viral mechanism involved in transcriptional regulation of iNOS gene expression.


Subject(s)
African Swine Fever Virus/physiology , Nitric Oxide Synthase Type II/genetics , Viral Proteins/physiology , Acetylation , African Swine Fever Virus/genetics , African Swine Fever Virus/pathogenicity , Animals , Base Sequence , Cell Line , Cells, Cultured , DNA, Viral/genetics , Down-Regulation , Gene Expression Regulation, Enzymologic , Macrophages/enzymology , Macrophages/virology , Mice , NF-kappa B/metabolism , Nitric Oxide/biosynthesis , Promoter Regions, Genetic , Swine , Transcription Factor RelA/metabolism , Transcription, Genetic , Viral Proteins/genetics , p300-CBP Transcription Factors/metabolism
6.
J Allergy Clin Immunol ; 117(6): 1462-9, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16751014

ABSTRACT

BACKGROUND: X-linked agammaglobulinemia (XLA) is characterized by impaired B-cell differentiation caused by mutations in the Bruton's tyrosine kinase (Btk) gene. The natural disease model, the X-linked immunodeficiency mouse, shows a less severe phenotype, indicating a different requirement of Btk in human and mouse B cells. Btk is also expressed in the myeloid line and participates in LPS signaling. Deficient oxidative burst and myeloid differentiation have been reported in the X-linked immunodeficiency mouse, but the precise mechanism and relevance of Btk activity in human monocytes is poorly understood. OBJECTIVE: The apparent absence in XLA of clinical manifestations of myeloid deficiency prompted us to explore the relevance of complete Btk absence in human myeloid cells. METHODS: Seven patients with XLA with BTK mutations conditioning a null protein expression were included in the study. Monocyte LPS-induced mitogen-activated protein kinase activation, TNF-alpha and IL-6 production in monocytes, and oxidative burst in monocytes and granulocytes were analyzed by means of flow cytometry. RESULTS: We show that in response to LPS, Btk-null monocytes from patients with XLA induce early mitogen-activated protein kinase activation and intracellular TNF-alpha and IL-6 production with the same intensity as cells from age- and sex-matched control subjects. In addition, the oxidative burst in response to LPS and other stimulants was completely normal in Btk-null monocytes and neutrophils. CONCLUSION: Our results indicate that Btk is not essential for early LPS signaling in human monocytes and that different Btk dependency might exist between human and mouse myeloid cells. CLINICAL IMPLICATIONS: These findings provide a better understanding of XLA, and they show the differences between human XLA and murine Xid models.


Subject(s)
Lipopolysaccharides/immunology , Monocytes/immunology , Monocytes/metabolism , Protein-Tyrosine Kinases , Agammaglobulinaemia Tyrosine Kinase , Agammaglobulinemia/enzymology , Agammaglobulinemia/genetics , Agammaglobulinemia/immunology , Animals , Cells, Cultured , Child, Preschool , Chromosomes, Human, X/genetics , Disease Models, Animal , Humans , Infant , Mice , Mitogen-Activated Protein Kinases/metabolism , Monocytes/enzymology , Point Mutation , Protein-Tyrosine Kinases/biosynthesis , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...