Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
PLoS Pathog ; 12(3): e1005489, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26939124

ABSTRACT

Sensing invading pathogens early in infection is critical for establishing host defense. Two cytosolic RIG-like RNA helicases, RIG-I and MDA5, are key to type I interferon (IFN) induction in response to viral infection. Mounting evidence suggests that another viral RNA sensor, protein kinase R (PKR), may also be critical for IFN induction during infection, although its exact contribution and mechanism of action are not completely understood. Using PKR-deficient cells, we found that PKR was required for type I IFN induction in response to infection by vaccinia virus lacking the PKR antagonist E3L (VVΔE3L), but not by Sendai virus or influenza A virus lacking the IFN-antagonist NS1 (FluΔNS1). IFN induction required the catalytic activity of PKR, but not the phosphorylation of its principal substrate, eIF2α, or the resulting inhibition of host translation. In the absence of PKR, IRF3 nuclear translocation was impaired in response to MDA5 activators, VVΔE3L and encephalomyocarditis virus, but not during infection with a RIG-I-activating virus. Interestingly, PKR interacted with both RIG-I and MDA5; however, PKR was only required for MDA5-mediated, but not RIG-I-mediated, IFN production. Using an artificially activated form of PKR, we showed that PKR activity alone was sufficient for IFN induction. This effect required MAVS and correlated with IRF3 activation, but no longer required MDA5. Nonetheless, PKR activation during viral infection was enhanced by MDA5, as virus-stimulated catalytic activity was impaired in MDA5-null cells. Taken together, our data describe a critical and non-redundant role for PKR following MDA5, but not RIG-I, activation to mediate MAVS-dependent induction of type I IFN through a kinase-dependent mechanism.


Subject(s)
DEAD-box RNA Helicases/metabolism , Encephalomyocarditis virus/immunology , Vaccinia virus/immunology , Vaccinia/immunology , eIF-2 Kinase/metabolism , Cell Line , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , Encephalomyocarditis virus/genetics , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Genes, Reporter , Humans , Interferon Type I/metabolism , Interferon-Induced Helicase, IFIH1 , Mutation , Phosphorylation , RNA, Viral/genetics , Receptors, Immunologic , Signal Transduction , Vaccinia/virology , Vaccinia virus/genetics , Viral Proteins/genetics , Viral Proteins/metabolism , eIF-2 Kinase/genetics
2.
RNA ; 18(7): 1338-46, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22635403

ABSTRACT

microRNAs (miRNAs) represent a class of noncoding RNAs that fine-tune gene expression through post-transcriptional silencing. While miRNA biogenesis occurs in a stepwise fashion, initiated by the nuclear microprocessor, rare noncanonical miRNAs have also been identified. Here we characterize the molecular components and unique attributes associated with the processing of virus-derived cytoplasmic primary miRNAs (c-pri-miRNAs). RNA in situ hybridization and inhibition of cellular division demonstrated a complete lack of nuclear involvement in c-pri-miRNA cleavage while genetic studies revealed that maturation still relied on the canonical nuclear RNase III enzyme, Drosha. The involvement of Drosha was mediated by a dramatic relocalization to the cytoplasm following virus infection. Deep sequencing analyses revealed that the cytoplasmic localization of Drosha does not impact the endogenous miRNA landscape during infection, despite allowing for robust synthesis of virus-derived miRNAs in the cytoplasm. Taken together, this research describes a unique function for Drosha in the processing of highly structured cytoplasmic RNAs in the context of virus infection.


Subject(s)
Cytoplasm/metabolism , MicroRNAs/metabolism , RNA Processing, Post-Transcriptional , Alphavirus Infections/metabolism , Cell Line , Humans , Influenza A virus/genetics , Influenza, Human/metabolism , RNA, Viral/biosynthesis , Ribonuclease III/metabolism , Sindbis Virus/genetics
3.
PLoS Pathog ; 8(1): e1002465, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22241991

ABSTRACT

Dengue virus (DENV) is a mosquito-borne pathogen for which no vaccine or specific therapeutic is available. Although it is well established that dendritic cells and macrophages are primary sites of DENV replication, it remains unclear whether non-hematopoietic cellular compartments serve as virus reservoirs. Here, we exploited hematopoietic-specific microRNA-142 (miR-142) to control virus tropism by inserting tandem target sites into the virus to restrict replication exclusively in this cell population. In vivo use of this virus restricted infection of CD11b+, CD11c+, and CD45+ cells, resulting in a loss of virus spread, regardless of the route of administration. Furthermore, sequencing of the targeted virus population that persisted at low levels, demonstrated total excision of the inserted miR-142 target sites. The complete conversion of the virus population under these selective conditions suggests that these immune cells are the predominant sources of virus amplification. Taken together, this work highlights the importance of hematopoietic cells for DENV replication and showcases an invaluable tool for the study of virus pathogenesis.


Subject(s)
Dengue Virus/physiology , Dengue/immunology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/virology , Virus Replication/physiology , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, CD/metabolism , Dengue/genetics , Dengue/metabolism , HEK293 Cells , Hematopoietic Stem Cells/metabolism , Humans , Mice , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/immunology , MicroRNAs/metabolism
4.
Virol J ; 11: 209, 2014 Dec 07.
Article in English | MEDLINE | ID: mdl-25481026

ABSTRACT

BACKGROUND: The outcome of a viral infection is regulated by complex interactions of viral and host factors. SARS coronavirus (SARS-CoV) engages and regulates several innate immune response pathways during infection. We have previously shown that the SARS-CoV Papain-like Protease (PLpro) inhibits type I interferon (IFN) by inhibiting IRF3 phosphorylation thereby blocking downstream Interferon induction. This finding prompted us to identify other potential mechanisms of inhibition of PLpro on IFN induction. METHODS: We have used plasmids expressing PLpro and IRF3 including an IRF3 mutant that is constitutively active, called IRF3(5D). In these experiments we utilize transfections, chromatin immunoprecipitation, Electro-mobility Shift Assays (EMSA) and protein localization to identify where IRF3 and IRF3(5D) are inhibited by PLpro. RESULTS: Here we show that PLpro also inhibits IRF3 activation at a step after phosphorylation and that this inhibition is dependent on the de-ubiquitination (DUB) activity of PLpro. We found that PLpro is able to block the type I IFN induction of a constitutively active IRF3, but does not inhibit IRF3 dimerization, nuclear localization or DNA binding. However, inhibition of PLpro's DUB activity by mutagenesis blocked the IRF3 inhibition activity of PLpro, suggesting a role for IRF3 ubiquitination in induction of a type I IFN innate immune response. CONCLUSION: These results demonstrate an additional mechanism that PLpro is able to inhibit IRF3 signaling. These data suggest novel innate immune antagonism activities of PLpro that may contribute to SARS-CoV pathogenesis.


Subject(s)
Cysteine Endopeptidases/metabolism , Interferon Regulatory Factor-3/antagonists & inhibitors , Severe acute respiratory syndrome-related coronavirus/enzymology , Viral Proteins/metabolism , Cell Line , Chromatin Immunoprecipitation , Coronavirus 3C Proteases , Electrophoretic Mobility Shift Assay , Humans , Transfection , Ubiquitin/metabolism
5.
Mol Ther ; 20(2): 367-75, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22086233

ABSTRACT

The discovery of microRNAs (miRNAs) revealed an unappreciated level of post-transcriptional control used by the cell to maintain optimal protein levels. This process has represented an attractive strategy for therapeutics that is currently limited by in vivo delivery constraints. Here, we describe the generation of a single-stranded, cytoplasmic virus of negative polarity capable of producing functional miRNAs. Cytoplasmic RNA virus-derived miRNAs accumulated to high levels in vitro, generated significant amounts of miRNA star strand, associated with the RNA-induced silencing complex (RISC), and conferred post transcriptional gene silencing in a sequence-specific manner. Furthermore, we demonstrate that these vectors could deliver miRNAs to a wide range of tissues, and sustain prolonged expression capable of achieving measurable knockdown of physiological targets in vivo. Taken together, these results validate noncanonical processing of cytoplasmic-derived miRNAs and provide a novel platform for small RNA delivery.


Subject(s)
Cytoplasm/metabolism , Gene Transfer Techniques , Genetic Vectors , MicroRNAs/genetics , RNA Viruses/genetics , Animals , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Cell Line , Gene Order , Humans , Mice , Mice, Inbred BALB C , Mice, Knockout , MicroRNAs/biosynthesis , MicroRNAs/metabolism , RNA-Induced Silencing Complex/metabolism
6.
RNA ; 16(11): 2068-74, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20841420

ABSTRACT

Cellular utilization of RNA interference (RNAi) as a mechanism to combat virus infection is thought to be restricted to plants and invertebrates. In vertebrates, antiviral defenses are largely dependent on interferons (IFNs), with the use of small RNAs restricted to microRNA (miRNA)-mediated targeting of host transcripts. Here we demonstrate that incorporation of a primary miRNA into a cytoplasmic virus results in the formation of a Dicer-dependent, DGCR8-independent, mature miRNA capable of conferring RNAi-like activity. Processing of the viral mirtron-like product (virtron) is indistinguishable from endogenous miRNA maturation and elicits post-transcriptional gene silencing, albeit at a reduced level. Furthermore, virtrons impose Dicer-dependent, microprocessor-independent, and IFN-independent interference on virus replication in a sequence-specific manner. Taken together, these results suggest the existence of a noncanonical, small-RNA-based activity capable of processing cytoplasmic hairpins and perhaps contributing to the cell's antiviral arsenal.


Subject(s)
Cytoplasm/genetics , MicroRNAs/genetics , Sindbis Virus/genetics , Animals , Base Sequence , Cell Line , Cytoplasm/chemistry , Cytoplasm/metabolism , DEAD-box RNA Helicases/deficiency , DEAD-box RNA Helicases/metabolism , Endoribonucleases/deficiency , Endoribonucleases/metabolism , Humans , Mice , Mice, Knockout , MicroRNAs/chemistry , MicroRNAs/metabolism , Molecular Sequence Data , Nucleic Acid Conformation , Ribonuclease III , Sindbis Virus/metabolism , Virus Replication
7.
J Immunol ; 184(6): 2908-17, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20164420

ABSTRACT

The dendritic cell (DC) is a master regulator of immune responses. Pathogenic viruses subvert normal immune function in DCs through the expression of immune antagonists. Understanding how these antagonists interact with the host immune system requires knowledge of the underlying genetic regulatory network that operates during an uninhibited antiviral response. To isolate and identify this network, we studied DCs infected with Newcastle disease virus, which is able to stimulate innate immunity and DC maturation through activation of RIG-I signaling, but lacks the ability to evade the human IFN response. To analyze this experimental model, we developed a new approach integrating genome-wide expression kinetics and time-dependent promoter analysis. We found that the genetic program underlying the antiviral cell-state transition during the first 18 h postinfection could be explained by a single convergent regulatory network. Gene expression changes were driven by a stepwise multifactor cascading control mechanism, where the specific transcription factors controlling expression changed over time. Within this network, most individual genes were regulated by multiple factors, indicating robustness against virus-encoded immune evasion genes. In addition to effectively recapitulating current biological knowledge, we predicted, and validated experimentally, antiviral roles for several novel transcription factors. More generally, our results show how a genetic program can be temporally controlled through a single regulatory network to achieve the large-scale genetic reprogramming characteristic of cell-state transitions.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Gene Expression Regulation, Viral/immunology , Newcastle disease virus/immunology , Transcription Factors/physiology , Up-Regulation/immunology , Conserved Sequence , Dendritic Cells/virology , Genes, Overlapping/immunology , Humans , Monocytes/immunology , Monocytes/metabolism , Monocytes/virology , Multigene Family/immunology , Newcastle disease virus/growth & development , Oligonucleotide Array Sequence Analysis , Predictive Value of Tests , Promoter Regions, Genetic/immunology , Reproducibility of Results , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transcription Factors/metabolism , Up-Regulation/genetics
8.
Cell Host Microbe ; 16(3): 314-327, 2014 Sep 10.
Article in English | MEDLINE | ID: mdl-25211074

ABSTRACT

To successfully establish infection, flaviviruses have to overcome the antiviral state induced by type I interferon (IFN-I). The nonstructural NS5 proteins of several flaviviruses antagonize IFN-I signaling. Here we show that yellow fever virus (YFV) inhibits IFN-I signaling through a unique mechanism that involves binding of YFV NS5 to the IFN-activated transcription factor STAT2 only in cells that have been stimulated with IFN-I. This NS5-STAT2 interaction requires IFN-I-induced tyrosine phosphorylation of STAT1 and the K63-linked polyubiquitination at a lysine in the N-terminal region of YFV NS5. We identified TRIM23 as the E3 ligase that interacts with and polyubiquitinates YFV NS5 to promote its binding to STAT2 and trigger IFN-I signaling inhibition. Our results demonstrate the importance of YFV NS5 in overcoming the antiviral action of IFN-I and offer a unique example of a viral protein that is activated by the same host pathway that it inhibits.


Subject(s)
Interferon-beta/metabolism , Viral Nonstructural Proteins/metabolism , Yellow Fever/metabolism , Yellow fever virus/metabolism , Amino Acid Motifs , Animals , Cell Line , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Host-Pathogen Interactions , Humans , Phosphorylation , Protein Binding , STAT1 Transcription Factor/chemistry , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/genetics , STAT2 Transcription Factor/metabolism , Signal Transduction , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , Yellow Fever/genetics , Yellow Fever/virology , Yellow fever virus/genetics
9.
Cell Host Microbe ; 12(2): 200-10, 2012 Aug 16.
Article in English | MEDLINE | ID: mdl-22901540

ABSTRACT

The life cycle of several viruses involves host or virally encoded small noncoding RNAs, which play important roles in posttranscriptional regulation. Small noncoding RNAs include microRNAs (miRNAs), which modulate the transcriptome, and small interfering RNAs (siRNAs), which are involved in pathogen defense in plants, worms, and insects. We show that insect and mammalian poxviruses induce the degradation of host miRNAs. The virally encoded poly(A) polymerase, which polyadenylates viral transcripts, also mediates 3' polyadenylation of host miRNAs, resulting in their degradation by the host machinery. In contrast, siRNAs, which are protected by 2'O-methylation (2'OMe), were not targeted by poxviruses. These findings suggest that poxviruses may degrade host miRNAs to promote replication and that virus-mediated small RNA degradation likely contributed to 2'OMe evolution.


Subject(s)
MicroRNAs/metabolism , Polynucleotide Adenylyltransferase/metabolism , Poxviridae Infections/metabolism , Poxviridae/enzymology , Viral Proteins/metabolism , Animals , Cell Line , Drosophila , Host-Pathogen Interactions , Humans , Methylation , Mice , MicroRNAs/chemistry , MicroRNAs/genetics , Moths , Polynucleotide Adenylyltransferase/genetics , Poxviridae/genetics , Poxviridae Infections/genetics , Poxviridae Infections/virology , RNA Stability , Viral Proteins/genetics
10.
Viruses ; 2(1): 55-72, 2010 Jan.
Article in English | MEDLINE | ID: mdl-21994600

ABSTRACT

The ability of a cell to combat an intracellular pathogen requires a mechanism to recognize the threat and elicit a transcriptional response against it. In the context of virus infection, the cell must take measures to inhibit viral replication, meanwhile, convey warning signals to neighboring cells of the imminent threat. This immune response is predominantly mediated by the production of cytokines, notably, interferon beta (IFNß). IFNß signaling results in the transcriptional induction of over one hundred antiviral gene products whose timely expression renders infected cells more capable of inhibiting virus replication, while providing the uninfected cells with the reinforcements to generate a less permissive cellular environment. Induction of IFNß and many aspects of the antiviral response pivot on the function of the IKK and IKK-related kinases. Despite sharing high levels of homology and some degree of functional redundancy, the classic IKK kinases: IKKα and IKKß, and the IKK-related kinases: TBK1 and IKKɛ, perform distinct roles in regulating the host antiviral defense. These kinases serve as molecular operators in their cooperative ability to integrate incoming cellular cues and act on a range of essential antiviral transcription factors to reshape the cellular transcriptome during infection.

11.
Nat Biotechnol ; 27(6): 572-6, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19483680

ABSTRACT

Influenza A virus leads to yearly epidemics and sporadic pandemics. Present prophylactic strategies focus on egg-grown, live, attenuated influenza vaccines (LAIVs), in which attenuation is generated by conferring temperature sensitivity onto the virus. Here we describe an alternative approach to attenuating influenza A virus based on microRNA-mediated gene silencing. By incorporating nonavian microRNA response elements (MREs) into the open-reading frame of the viral nucleoprotein, we generate reassortant LAIVs for H1N1 and H5N1 that are attenuated in mice but not in eggs. MRE-based LAIVs show a greater than two-log reduction in mortality compared with control viruses lacking MREs and elicit a diverse antibody response. This approach might be combined with existing LAIVs to increase attenuation and improve vaccine safety.


Subject(s)
Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/genetics , Influenza Vaccines , MicroRNAs , RNA Interference , Vaccines, Attenuated , Animals , Body Weight , Cell Line , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H5N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza Vaccines/genetics , Influenza Vaccines/immunology , Influenza, Human/immunology , Influenza, Human/prevention & control , Mice , MicroRNAs/genetics , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , RNA, Viral/genetics , Response Elements/genetics , Species Specificity , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology
SELECTION OF CITATIONS
SEARCH DETAIL