Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
ACS Sens ; 9(1): 272-282, 2024 01 26.
Article in English | MEDLINE | ID: mdl-38214491

ABSTRACT

Extracellular vesicles (EVs) are vectors of biomolecular cargo that play essential roles in intercellular communication across a range of cells. Protein, lipid, and nucleic acid cargo harbored within EVs may serve as biomarkers at all stages of disease; however, the choice of methodology may challenge the specificity and reproducibility of discovery. To address these challenges, the integration of rigorous EV purification methods, cutting-edge spectroscopic technologies, and data analysis are critical to uncover diagnostic signatures of disease. Herein, we demonstrate an EV isolation and analysis pipeline using surface-enhanced Raman spectroscopy (SERS) and mass spectrometry (MS) techniques on plasma samples obtained from umbilical cord blood, healthy donor (HD) plasma, and plasma from women with early stage high-grade serous carcinoma (HGSC). Plasma EVs were purified by size exclusion chromatography and analyzed by surface-enhanced Raman spectroscopy (SERS), mass spectrometry (MS), and atomic force microscopy. After determining the fraction of highest EV purity, SERS and MS were used to characterize EVs from HDs, pooled donors with noncancerous gynecological ailments (n = 6), and donors with early stage [FIGO (I/II)] with HGSC. SERS spectra were subjected to different machine learning algorithms such as PCA, logistic regression, support vector machine, naïve Bayes, random forest, neural network, and k nearest neighbors to differentiate healthy, benign, and HGSC EVs. Collectively, we demonstrate a reproducible workflow with the potential to serve as a diagnostic platform for HGSC.


Subject(s)
Extracellular Vesicles , Neoplasms , Humans , Female , Tandem Mass Spectrometry , Bayes Theorem , Reproducibility of Results , Extracellular Vesicles/metabolism , Neoplasms/metabolism , Biomarkers, Tumor/analysis
2.
PLoS Pathog ; 15(10): e1008111, 2019 10.
Article in English | MEDLINE | ID: mdl-31626661

ABSTRACT

The herpes simplex virus virion host shutoff (vhs) RNase destabilizes cellular and viral mRNAs and blunts host innate antiviral responses. Previous work demonstrated that cells infected with vhs mutants display enhanced activation of the host double-stranded RNA (dsRNA)-activated protein kinase R (PKR), implying that vhs limits dsRNA accumulation in infected cells. Confirming this hypothesis, we show that partially complementary transcripts of the UL23/UL24 and UL30/31 regions of the viral genome increase in abundance when vhs is inactivated, giving rise to greatly increased levels of intracellular dsRNA formed by annealing of the overlapping portions of these RNAs. Thus, vhs limits accumulation of dsRNA at least in part by reducing the levels of complementary viral transcripts. We then asked if vhs also destabilizes dsRNA after its initial formation. Here, we used a reporter system employing two mCherry expression plasmids bearing complementary 3' UTRs to produce defined dsRNA species in uninfected cells. The dsRNAs are unstable, but are markedly stabilized by co-expressing the HSV dsRNA-binding protein US11. Strikingly, vhs delivered by super-infecting HSV virions accelerates the decay of these pre-formed dsRNAs in both the presence and absence of US11, a novel and unanticipated activity of vhs. Vhs binds the host RNA helicase eIF4A, and we find that vhs-induced dsRNA decay is attenuated by the eIF4A inhibitor hippuristanol, providing evidence that eIF4A participates in the process. Our results show that a herpesvirus host shutoff RNase destabilizes dsRNA in addition to targeting partially complementary viral mRNAs, raising the possibility that the mRNA destabilizing proteins of other viral pathogens dampen the host response to dsRNA through similar mechanisms.


Subject(s)
RNA Stability/genetics , RNA, Double-Stranded/metabolism , RNA, Messenger/metabolism , RNA, Viral/metabolism , Ribonucleases/metabolism , Simplexvirus/genetics , Viral Proteins/metabolism , Animals , Cell Line , Chlorocebus aethiops , DNA-Directed DNA Polymerase/metabolism , Eukaryotic Initiation Factor-4F/antagonists & inhibitors , Eukaryotic Initiation Factor-4F/metabolism , Exodeoxyribonucleases/metabolism , HeLa Cells , Humans , Nuclear Proteins/metabolism , RNA-Binding Proteins/metabolism , Vero Cells
4.
J Virol ; 90(13): 6049-6057, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27099317

ABSTRACT

UNLABELLED: The herpes simplex virus (HSV) virion host shutoff (vhs) RNase destabilizes cellular and viral mRNAs, suppresses host protein synthesis, dampens antiviral responses, and stimulates translation of viral mRNAs. vhs mutants display a host range phenotype: translation of viral true late mRNAs is severely impaired and stress granules accumulate in HeLa cells, while translation proceeds normally in Vero cells. We found that vhs-deficient virus activates the double-stranded RNA-activated protein kinase R (PKR) much more strongly than the wild-type virus does in HeLa cells, while PKR is not activated in Vero cells, raising the possibility that PKR might play roles in stress granule induction and/or inhibiting translation in restrictive cells. We tested this possibility by evaluating the effects of inactivating PKR. Eliminating PKR in HeLa cells abolished stress granule formation but had only minor effects on viral true late protein levels. These results document an essential role for PKR in stress granule formation by a nuclear DNA virus, indicate that induction of stress granules is the consequence rather than the cause of the translational defect, and are consistent with our previous suggestion that vhs promotes translation of viral true late mRNAs by preventing mRNA overload rather than by suppressing eIF2α phosphorylation. IMPORTANCE: The herpes simplex virus vhs RNase plays multiple roles during infection, including suppressing PKR activation, inhibiting the formation of stress granules, and promoting translation of viral late mRNAs. A key question is the extent to which these activities are mechanistically connected. Our results demonstrate that PKR is essential for stress granule formation in the absence of vhs, but at best, it plays a secondary role in suppressing translation of viral mRNAs. Thus, the ability of vhs to promote translation of viral mRNAs can be largely uncoupled from PKR suppression, demonstrating that this viral RNase modulates at least two distinct aspects of RNA metabolism.


Subject(s)
Cytoplasmic Granules/metabolism , Herpesvirus 1, Human/metabolism , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Viral/genetics , Viral Proteins/metabolism , Virion/metabolism , eIF-2 Kinase/metabolism , Animals , Chlorocebus aethiops , Enzyme Activation , Eukaryotic Initiation Factor-2/metabolism , HEK293 Cells , HeLa Cells , Host-Pathogen Interactions , Humans , RNA, Double-Stranded/genetics , RNA, Double-Stranded/metabolism , RNA, Messenger/metabolism , RNA, Viral/metabolism , Vero Cells , Viral Proteins/genetics , Virus Replication , eIF-2 Kinase/genetics
5.
J Virol ; 88(21): 12727-39, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25142597

ABSTRACT

UNLABELLED: In a previous study, it was observed that cells infected with herpes simplex virus 2 (HSV-2) failed to accumulate stress granules (SGs) in response to oxidative stress induced by arsenite treatment. As a follow-up to this observation, we demonstrate here that disruption of arsenite-induced SG formation by HSV-2 is mediated by a virion component. Through studies on SG formation in cells infected with HSV-2 strains carrying defective forms of UL41, the gene that encodes vhs, we identify vhs as a virion component required for this disruption. Cells infected with HSV-2 strains producing defective forms of vhs form SGs spontaneously late in infection. In addition to core SG components, these spontaneous SGs contain the viral immediate early protein ICP27 as well as the viral serine/threonine kinase Us3. As part of these studies, we reexamined the frameshift mutation known to reside within the UL41 gene of HSV-2 strain HG52. We demonstrate that this mutation is unstable and can rapidly revert to restore wild-type UL41 following low-multiplicity passaging. Identification of the involvement of virion-associated vhs in the disruption of SG formation will enable mechanistic studies on how HSV-2 is able to counteract antiviral stress responses early in infection. In addition, the ability of Us3 to localize to stress granules may indicate novel roles for this viral kinase in the regulation of translation. IMPORTANCE: Eukaryotic cells respond to stress by rapidly shutting down protein synthesis and storing mRNAs in cytoplasmic stress granules (SGs). Stoppages in protein synthesis are problematic for all viruses as they rely on host cell machinery to synthesize viral proteins. Thus, many viruses target SGs for disruption or modification. Infection by herpes simplex virus 2 (HSV-2) was previously observed to disrupt SG formation induced by oxidative stress. In this follow-up study, we identify virion host shutoff protein (vhs) as a viral protein involved in this disruption. The identification of a specific viral protein involved in disrupting SG formation is a key step toward understanding how HSV-2 interacts with these antiviral structures. Additionally, this understanding may provide insights into the biology of SGs that may find application in studies on human motor neuron degenerative diseases, like amyotrophic lateral sclerosis (ALS), which may arise as a result of dysregulation of SG formation.


Subject(s)
Arsenic/toxicity , Cytoplasmic Granules/metabolism , Herpesvirus 2, Human/enzymology , Host-Pathogen Interactions , Oxidative Stress , Ribonucleases/metabolism , Viral Proteins/metabolism , Virion/enzymology , Animals , Cell Line , Humans
6.
J Virol ; 88(17): 9624-32, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24920814

ABSTRACT

UNLABELLED: We recently demonstrated that the virion host shutoff (vhs) protein, an mRNA-specific endonuclease, is required for efficient herpes simplex virus 1 (HSV-1) replication and translation of viral true-late mRNAs, but not other viral and cellular mRNAs, in many cell types (B. Dauber, J. Pelletier, and J. R. Smiley, J. Virol. 85:5363-5373, 2011, http://dx.doi.org/10.1128/JVI.00115-11). Here, we evaluated whether the structure of true-late mRNAs or the timing of their transcription is responsible for the poor translation efficiency in the absence of vhs. To test whether the highly structured 5' untranslated region (5'UTR) of the true-late gC mRNA is the primary obstacle for translation initiation, we replaced it with the less structured 5'UTR of the γ-actin mRNA. However, this mutation did not restore translation in the context of a vhs-deficient virus. We then examined whether the timing of transcription affects translation efficiency at late times. To this end, we engineered a vhs-deficient virus mutant that transcribes the true-late gene US11 with immediate-early kinetics (IEUS11-ΔSma). Interestingly, IEUS11-ΔSma showed increased translational activity on the US11 transcript at late times postinfection, and US11 protein levels were restored to wild-type levels. These results suggest that mRNAs can maintain translational activity throughout the late stage of infection if they are present before translation factors and/or ribosomes become limiting. Taken together, these results provide evidence that in the absence of the mRNA-destabilizing function of vhs, accumulation of viral mRNAs overwhelms the capacity of the host translational machinery, leading to functional exclusion of the last mRNAs that are made during infection. IMPORTANCE: The process of mRNA translation accounts for a significant portion of a cell's energy consumption. To ensure efficient use of cellular resources, transcription, translation, and mRNA decay are tightly linked and highly regulated. However, during virus infection, the overall amount of mRNA may increase drastically, possibly overloading the capacity of the translation apparatus. Our results suggest that the HSV-1 vhs protein, an mRNA-specific endoribonuclease, prevents mRNA overload during infection, thereby allowing translation of late viral mRNAs. The requirement for vhs varies between cell types. Further studies of the basis for this difference likely will offer insights into how cells regulate overall mRNA levels and access to the translational apparatus.


Subject(s)
Herpesvirus 1, Human/physiology , Protein Biosynthesis , RNA, Messenger/metabolism , RNA, Viral/metabolism , Ribonucleases/metabolism , Viral Proteins/biosynthesis , Animals , Chlorocebus aethiops , HeLa Cells , Herpesvirus 1, Human/genetics , Humans , Vero Cells , Viral Proteins/metabolism
7.
J Infect Dis ; 204(10): 1483-90, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21917878

ABSTRACT

BACKGROUND: Both pandemic and interpandemic influenza is associated with high morbidity and mortality worldwide. Seasonal epidemics are caused by both influenza A and B virus strains that cocirculate with varying predominance and may give rise to severe illness equally. According to World Health Organization recommendations, current annual vaccines are composed of 2 type A and 1 type B virus-specific component. METHODS: As a novel attenuated live vaccine against influenza B virus, we generated a hemagglutinin cleavage site mutant of strain B/Lee/40 by replacing the common monobasic cleavage site recognized by trypsinlike proteases with an elastase-sensitive site, and we investigated the in vitro properties, attenuation, humoral responses, and efficacy in mice. RESULTS: This mutant virus replicated in cell culture equally well as the wild type but in a strictly elastase-dependent manner. In contrast to the mouse-pathogenic parental virus, the cleavage site mutant was fully attenuated in mice and not detectable in their lungs. After 1 intranasal immunization, the animals survived lethal challenge with wild-type virus without weight loss or any other signs of disease. Furthermore, no challenge virus could be reisolated from the lungs of vaccinated mice. CONCLUSIONS: These findings demonstrate that proteolytic activation mutants can serve as live vaccine against influenza B virus.


Subject(s)
Influenza B virus/genetics , Influenza Vaccines/immunology , Mutation , Animals , Antibodies, Viral/blood , Female , Hemagglutination, Viral/genetics , Influenza B virus/chemistry , Influenza B virus/immunology , Influenza Vaccines/genetics , Mice , Mice, Inbred BALB C , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
8.
J Virol ; 85(11): 5363-73, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21430045

ABSTRACT

The herpes simplex virus 1 (HSV-1) virion host shutoff protein (vhs) degrades viral and cellular mRNAs. Here, we demonstrate for the first time that vhs also boosts translation of viral true late mRNAs in a cell type-dependent manner and that this effect determines the viral growth phenotype in the respective cell type. Our study was prompted by the detection of stress granules, indicators of stalled translation initiation, in cells infected with vhs mutants but not in wild-type-virus-infected cells. Accumulation of true late-gene products gC and US11 was strongly reduced in the absence of vhs in HeLa cells and several other restrictive cell lines but not in Vero and other permissive cells and was independent of phosphorylation of the α subunit of eukaryotic initiation factor 2 (eIF2α). Polysome analysis showed that gC and US11 transcripts were poorly translated in vhs-null-virus-infected HeLa cells, while translation of a cellular mRNA was not affected. Interestingly, hippuristanol, an eIF4A inhibitor, produced a similar phenotype in HeLa cells infected with wild-type HSV-1, while Vero cells were much more resistant to the inhibitor. These results suggest that translation of true late-gene transcripts is particularly sensitive to conditions of limited access to translation factors and that vhs is able either to prevent the limiting conditions or to facilitate translation initiation under these conditions. The varied permissivity of cell lines to vhs-null infection may stem from differences in the resilience of the translation machinery or the ability to control the accumulation of mRNAs.


Subject(s)
Herpesvirus 1, Human/physiology , Protein Biosynthesis , Ribonucleases/metabolism , Viral Proteins/biosynthesis , Animals , Cell Line , Eukaryotic Initiation Factor-2/metabolism , Gene Deletion , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/growth & development , Humans , Mutant Proteins/genetics , Mutant Proteins/metabolism , Polyribosomes/metabolism , RNA, Messenger/metabolism , RNA, Viral/metabolism , Ribonucleases/genetics , Viral Proteins/genetics , Viral Proteins/metabolism
9.
J Virol ; 84(4): 2122-33, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20007264

ABSTRACT

A reassortant avian influenza virus (designated FPV NS GD), carrying the NS-segment of the highly pathogenic avian influenza virus (HPAIV) strain A/Goose/Guangdong/1/96 (GD; H5N1) in the genetic background of the HPAIV strain A/FPV/Rostock/34 (FPV; H7N1), was rescued by reverse genetics. Remarkably, in contrast to the recombinant wild-type FPV (rFPV), the reassortant virus was able to replicate more efficiently in different human cell lines and primary mouse epithelia cells without prior adaptation. Moreover, FPV NS GD caused disease and death in experimentally infected mice and was detected in mouse lungs; in contrast, rFPV was not able to replicate in mice effectively. These results indicated an altered host range and increased virulence. Furthermore FPV NS GD showed pronounced pathogenicity in chicken embryos. In an attempt to define the molecular basis for the apparent differences, we determined that NS1 proteins of the H5N1 and H7N1 strains bound the antiviral kinase PKR and the F2F3 domain of cleavage and polyadenylation specificity factor 30 (CPSF30) with comparable efficiencies in vitro. However, FPV NS GD infection resulted in (i) increased expression of NS1, (ii) faster and stronger PKR inhibition, and (iii) stronger beta interferon promoter inhibition than rFPV. Taken together, the results shed further light on the importance of the NS segment of an H5N1 strain for viral replication, molecular pathogenicity, and host range of HPAIVs and the possible consequences of a reassortment between naturally occurring H7 and H5 type HPAIVs.


Subject(s)
Influenza A Virus, H5N1 Subtype/physiology , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza A virus/physiology , Influenza A virus/pathogenicity , Viral Nonstructural Proteins/physiology , Animals , Base Sequence , Birds , Cell Line , Chick Embryo , DNA, Viral/genetics , Dogs , Female , Genes, Viral , Humans , Influenza A Virus, H5N1 Subtype/genetics , Influenza A virus/genetics , Interferon-beta/biosynthesis , Mice , Mice, Inbred C57BL , Reassortant Viruses/genetics , Reassortant Viruses/pathogenicity , Reassortant Viruses/physiology , Viral Nonstructural Proteins/genetics , Virulence/genetics , Virulence/physiology , Virus Replication/genetics , Virus Replication/physiology , eIF-2 Kinase/metabolism
10.
PLoS Pathog ; 5(6): e1000473, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19521506

ABSTRACT

Activation of the latent kinase PKR is a potent innate defense reaction of vertebrate cells towards viral infections, which is triggered by recognition of viral double-stranded (ds) RNA and results in a translational shutdown. A major gap in our understanding of PKR's antiviral properties concerns the nature of the kinase activating molecules expressed by influenza and other viruses with a negative strand RNA genome, as these pathogens produce little or no detectable amounts of dsRNA. Here we systematically investigated PKR activation by influenza B virus and its impact on viral pathogenicity. Biochemical analysis revealed that PKR is activated by viral ribonucleoprotein (vRNP) complexes known to contain single-stranded RNA with a 5'-triphosphate group. Cell biological examination of recombinant viruses showed that the nucleo-cytoplasmic transport of vRNP late in infection is a strong trigger for PKR activation. In addition, our analysis provides a mechanistic explanation for the previously observed suppression of PKR activation by the influenza B virus NS1 protein, which we show here to rely on complex formation between PKR and NS1's dsRNA binding domain. The high significance of this interaction for pathogenicity was revealed by the finding that attenuated influenza viruses expressing dsRNA binding-deficient NS1 proteins were rescued for high replication and virulence in PKR-deficient cells and mice, respectively. Collectively, our study provides new insights into an important antiviral defense mechanism of vertebrates and leads us to suggest a new model of PKR activation by cytosolic vRNP complexes, a model that may also be applicable to other negative strand RNA viruses.


Subject(s)
Influenza B virus/physiology , Influenza B virus/pathogenicity , Ribonucleoproteins/metabolism , Viral Proteins/metabolism , eIF-2 Kinase/metabolism , Active Transport, Cell Nucleus , Animals , Cell Line , Cell Line, Tumor , Cells, Cultured , Cytoplasm/metabolism , Enzyme Activation , Female , Humans , Influenza B virus/genetics , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Models, Biological , Phosphorylation , RNA, Double-Stranded/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication
11.
J Virol ; 83(2): 701-11, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18987144

ABSTRACT

Many proteins that function in the transcription, maturation, and export of metazoan mRNAs are concentrated in nuclear speckle domains, indicating that the compartment is important for gene expression. Here, we show that the NS1 protein of influenza B virus (B/NS1) accumulates in nuclear speckles and causes rounding and morphological changes of the domains, indicating a disturbance in their normal functions. This property was located within the N-terminal 90 amino acids of the B/NS1 protein and was shown to be independent of any other viral gene product. Within this protein domain, we identified a monopartite importin alpha binding nuclear localization signal. Reverse-genetic analysis of this motif indicated that nuclear import and speckle association of the B/NS1 protein are required for the full replication capacity of the virus. In the late phase of virus infection, the B/NS1 protein relocated to the cytoplasm, which occurred in a CRM1-independent manner. The interaction of the B/NS1 protein with nuclear speckles may reflect a recruitment function to promote viral-gene expression. To our knowledge, this is the first functional description of a speckle-associated protein that is encoded by a negative-strand RNA virus.


Subject(s)
Influenza B virus/physiology , Viral Nonstructural Proteins/metabolism , Virus Replication , Animals , Cell Line , Cell Nucleus/chemistry , Cytoplasm/chemistry , Humans , Nuclear Localization Signals , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Spodoptera , Viral Nonstructural Proteins/genetics
12.
Viruses ; 1(3): 523-44, 2009 12.
Article in English | MEDLINE | ID: mdl-21994559

ABSTRACT

The interferon-induced double-stranded (ds)RNA-dependent protein kinase (PKR) limits viral replication by an eIF2α-mediated block of translation. Although many negative-strand RNA viruses activate PKR, the responsible RNAs have long remained elusive, as dsRNA, the canonical activator of PKR, has not been detected in cells infected with such viruses. In this review we focus on the activating RNA molecules of different virus families, in particular the negative-strand RNA viruses. We discuss the recently identified non-canonical activators 5'-triphosphate RNA and the vRNP of influenza virus and give an update on strategies of selected RNA and DNA viruses to prevent activation of PKR.

13.
Cell Microbiol ; 9(4): 930-8, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17140406

ABSTRACT

Influenza A virus causes epidemics of respiratory diseases in humans leading to thousands of death annually. One of its major virulence factors, the non-structural protein 1 (NS1), exhibits interferon-antagonistic properties. While epithelial cells of the respiratory tract are the primary targets of influenza virus, the virus-sensing mechanisms in these cells eventually leading to IFNbeta production are incompletely understood. Here we show that infection of epithelial cells with NS1-deficient influenza A virus upregulated expression of two molecules that have been previously implicated in sensing of RNA viruses, the retinoic acid-inducible gene I (RIG-I) and the melanoma differentiation-associated gene 5 (MDA5). Gene silencing and overexpression experiments demonstrated that RIG-I, its adapter interferon-beta promoter stimulator 1 (IPS-1) and interferon-regulated factor 3 (IRF3) were involved in influenza A virus-mediated production of the antiviral IFNbeta. In addition, we showed that the NS1 protein is capable to inhibit the RIG-I-induced signalling, a mechanism which corresponded to the observation that only NS1-deficient but not the wild-type virus induced high-level production of IFNbeta. In conclusion, we demonstrated a critical involvement of RIG-I, IPS-1 and IRF3 in influenza A virus infection of epithelial cells.


Subject(s)
DEAD-box RNA Helicases/physiology , Influenza A virus/growth & development , Interferon-beta/metabolism , Viral Nonstructural Proteins/physiology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/physiology , Cell Line , Cell Line, Tumor , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/virology , Humans , Immunoblotting , Influenza A virus/genetics , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/physiology , Interferon-Induced Helicase, IFIH1 , Interferon-beta/genetics , Mutation , Promoter Regions, Genetic/genetics , RNA Interference , Receptors, Immunologic , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/physiology , Viral Nonstructural Proteins/genetics
14.
J Virol ; 80(23): 11667-77, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16987984

ABSTRACT

Expression of alpha/beta interferon (IFN-alpha/beta) in virus-infected vertebrate cells is a key event in the establishment of a sustained antiviral response, which is triggered by double-stranded RNA (dsRNA) produced during viral replication. These antiviral cytokines initiate the expression of cellular proteins with activities that limit the replication and spread of the invading viruses. Within this response, the dsRNA-dependent protein kinase R (PKR) that is expressed at constitutive levels and upregulated by IFN-alpha/beta acts as an important antiviral effector that can block the cellular translational machinery. We previously demonstrated that efficient replication of influenza B virus depends on the viral dsRNA-binding NS1 protein that inhibits the transcriptional activation of IFN-alpha/beta genes. Here we tested the postulate that the viral NS1 protein counteracts antiviral responses through sequestering intracellular dsRNA by analyzing a collection of recombinant influenza B viruses. As expected, viruses expressing dsRNA-binding-defective NS1 proteins were strongly attenuated for replication in IFN-competent hosts. Interestingly, these virus mutants failed to prevent activation of PKR but could effectively limit IFN induction. Conversely, a mutant virus expressing the N-terminal dsRNA-binding domain of NS1 prevented PKR activation, but not IFN induction, suggesting an important role for the NS1 C-terminal part in silencing the activation route of IFN-alpha/beta genes. Thus, our findings indicate an unexpected mechanistic dichotomy of the influenza B virus NS1 protein in the suppression of antiviral responses, which involves at least one activity that is largely separable from dsRNA binding.


Subject(s)
Influenza B virus/chemistry , Interferon-alpha/metabolism , Interferon-beta/metabolism , RNA-Binding Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/pharmacology , eIF-2 Kinase/antagonists & inhibitors , Cell Line , Gene Expression Regulation, Viral , Interferon-alpha/genetics , Interferon-beta/genetics , RNA-Binding Proteins/metabolism , Viral Nonstructural Proteins/metabolism
15.
Proc Natl Acad Sci U S A ; 102(38): 13640-5, 2005 Sep 20.
Article in English | MEDLINE | ID: mdl-16155125

ABSTRACT

Expression of the antiviral cytokines IFN-alpha/beta is among the most potent innate defenses of higher vertebrates to virus infections, which is controlled by the inducible transcription factor IFN regulatory factor (IRF)3. Borna disease virus (BDV) establishes persistent noncytolytic infections in animals and tissue culture cells, indicating that it can circumvent this antiviral reaction by an unexplained activity. In this study, we identify the BDV P protein as microbial gene product that associates with and inhibits the principal regulatory kinase of IRF3, Traf family member-associated NF-kappaB activator (TANK)-binding kinase 1 (TBK-1). We demonstrate that the P protein counteracts TBK-1-dependent IFN-beta expression in cells and, hence, the establishment of an antiviral state. Furthermore, our data show that the BDV P protein itself is phosphorylated by TBK-1, suggesting that P functions as a viral decoy substrate that prevents activation of cellular target proteins of TBK-1. Thus, our findings provide evidence for a previously undescribed mechanism by which a viral protein interferes with the induction of the antiviral IFN cascade.


Subject(s)
Borna Disease/metabolism , Borna disease virus/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Viral Proteins/metabolism , Viral Structural Proteins/metabolism , Animals , Borna disease virus/genetics , Cell Line , DNA-Binding Proteins/metabolism , Dogs , Humans , Interferon Regulatory Factor-3 , Interferon-alpha/biosynthesis , Interferon-beta/biosynthesis , Phosphoproteins/genetics , Phosphorylation , Transcription Factors/metabolism , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism , Viral Proteins/genetics , Viral Structural Proteins/genetics
16.
J Virol ; 78(21): 11574-82, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15479798

ABSTRACT

The NS1 proteins of influenza A and B viruses (A/NS1 and B/NS1 proteins) have only approximately 20% amino acid sequence identity. Nevertheless, these proteins show several functional similarities, such as their ability to bind to the same RNA targets and to inhibit the activation of protein kinase R in vitro. A critical function of the A/NS1 protein is the inhibition of synthesis of alpha/beta interferon (IFN-alpha/beta) during viral infection. Recently, it was also found that the B/NS1 protein inhibits IFN-alpha/beta synthesis in virus-infected cells. We have now found that the expression of the B/NS1 protein complements the growth of an influenza A virus with A/NS1 deleted. Expression of the full-length B/NS1 protein (281 amino acids), as well as either its N-terminal RNA-binding domain (amino acids 1 to 93) or C-terminal domain (amino acids 94 to 281), in the absence of any other influenza B virus proteins resulted in the inhibition of IRF-3 nuclear translocation and IFN-beta promoter activation. A mutational analysis of the truncated B/NS1(1-93) protein showed that RNA-binding activity correlated with IFN-beta promoter inhibition. In addition, a recombinant influenza B virus with NS1 deleted induces higher levels of IRF-3 activation, as determined by its nuclear translocation, and of IFN-alpha/beta synthesis than wild-type influenza B virus. Our results support the hypothesis that the NS1 protein of influenza B virus plays an important role in antagonizing the IRF-3- and IFN-induced antiviral host responses to virus infection.


Subject(s)
Active Transport, Cell Nucleus , DNA-Binding Proteins/antagonists & inhibitors , Influenza B virus/physiology , Interferon-beta/antagonists & inhibitors , Promoter Regions, Genetic , Transcription Factors/antagonists & inhibitors , Viral Nonstructural Proteins/physiology , Animals , Cell Line , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Humans , Influenza B virus/chemistry , Interferon Regulatory Factor-3 , Interferon-beta/genetics , Protein Transport , RNA/metabolism , Transcription Factors/metabolism , Viral Nonstructural Proteins/chemistry
17.
J Virol ; 78(4): 1865-72, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14747551

ABSTRACT

We analyzed the functions of the influenza B virus nonstructural NS1-B protein, both by utilizing a constructed mutant virus (Delta NS1-B) lacking the NS1 gene and by testing the activities of the protein when expressed in cells. The mutant virus replicated to intermediate levels in 6-day-old embryonated chicken eggs that contain an immature interferon (IFN) system, whereas older eggs did not support viral propagation to a significant extent. The Delta NS1-B virus was a substantially stronger inducer of beta IFN (IFN-beta) transcripts in human lung epithelial cells than the wild type, and furthermore, transiently expressed NS1-B protein efficiently inhibited virus-dependent activation of the IFN-beta promoter. Interestingly, replication of the Delta NS1-B knockout virus was attenuated by more than 4 orders of magnitude in tissue culture cells containing or lacking functional IFN-alpha/beta genes. These findings show that the NS1-B protein functions as a viral IFN antagonist and indicate a further requirement of this protein for efficient viral replication that is unrelated to blocking IFN effects.


Subject(s)
Influenza B virus/growth & development , Interferon-beta/antagonists & inhibitors , Interferon-beta/biosynthesis , Viral Nonstructural Proteins/metabolism , Animals , Cell Line , Chick Embryo , Chlorocebus aethiops , Dogs , Humans , Influenza B virus/genetics , Influenza B virus/immunology , Mutation , Vero Cells , Viral Nonstructural Proteins/genetics , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...