Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Vaccines (Basel) ; 11(12)2023 Dec 08.
Article in English | MEDLINE | ID: mdl-38140236

ABSTRACT

Virus-specific antibodies are crucial for protective immunity against SARS-CoV-2. Assessing functional antibodies through conventional or pseudotyped virus neutralisation tests (pVNT) requires high biosafety levels. Alternatively, the virus-free surrogate virus neutralisation test (sVNT) quantifies antibodies interfering with spike binding to angiotensin-converting enzyme 2. We evaluated secreted nanoluciferase-tagged spike protein fragments as diagnostic antigens in the sVNT in a vaccination cohort. Initially, spike fragments were tested in a capture enzyme immunoassay (EIA), identifying the receptor binding domain (RBD) as the optimal diagnostic antigen. The sensitivity of the in-house sVNT applying the nanoluciferase-labelled RBD equalled or surpassed that of a commercial sVNT (cPass, GenScript Diagnostics) and an in-house pVNT four weeks after the first vaccination (98% vs. 94% and 72%, respectively), reaching 100% in all assays four weeks after the second and third vaccinations. When testing serum reactivity with Omicron BA.1 spike, the sVNT and pVNT displayed superior discrimination between wild-type- and variant-specific serum reactivity compared to a capture EIA. This was most pronounced after the first and second vaccinations, with the third vaccination resulting in robust, cross-reactive BA.1 construct detection. In conclusion, utilising nanoluciferase-labelled antigens permits the quantification of SARS-CoV-2-specific inhibitory antibodies. Designed as flexible modular systems, the assays can be readily adjusted for monitoring vaccine efficacy.

2.
PLoS Pathog ; 19(7): e1010986, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37440521

ABSTRACT

Influenza A virus (IAV), like any other virus, provokes considerable modifications of its host cell's metabolism. This includes a substantial increase in the uptake as well as the metabolization of glucose. Although it is known for quite some time that suppression of glucose metabolism restricts virus replication, the exact molecular impact on the viral life cycle remained enigmatic so far. Using 2-deoxy-d-glucose (2-DG) we examined how well inhibition of glycolysis is tolerated by host cells and which step of the IAV life cycle is affected. We observed that effects induced by 2-DG are reversible and that cells can cope with relatively high concentrations of the inhibitor by compensating the loss of glycolytic activity by upregulating other metabolic pathways. Moreover, mass spectrometry data provided information on various metabolic modifications induced by either the virus or agents interfering with glycolysis. In the presence of 2-DG viral titers were significantly reduced in a dose-dependent manner. The supplementation of direct or indirect glycolysis metabolites led to a partial or almost complete reversion of the inhibitory effect of 2-DG on viral growth and demonstrated that indeed the inhibition of glycolysis and not of N-linked glycosylation was responsible for the observed phenotype. Importantly, we could show via conventional and strand-specific qPCR that the treatment with 2-DG led to a prolonged phase of viral mRNA synthesis while the accumulation of genomic vRNA was strongly reduced. At the same time, minigenome assays showed no signs of a general reduction of replicative capacity of the viral polymerase. Therefore, our data suggest that the significant reduction in IAV replication by glycolytic interference occurs mainly due to an impairment of the dynamic regulation of the viral polymerase which conveys the transition of the enzyme's function from transcription to replication.


Subject(s)
Influenza A virus , Influenza A virus/genetics , Virus Replication/physiology , Transcription, Genetic , Nucleotidyltransferases/metabolism , Genomics , Glycolysis , RNA, Viral/genetics , RNA, Viral/metabolism
3.
Pharmaceuticals (Basel) ; 15(5)2022 Apr 25.
Article in English | MEDLINE | ID: mdl-35631357

ABSTRACT

For almost two years, the COVID-19 pandemic has constituted a major challenge to human health, particularly due to the lack of efficient antivirals to be used against the virus during routine treatment interventions. Multiple treatment options have been investigated for their potential inhibitory effect on SARS-CoV-2. Natural products, such as plant extracts, may be a promising option, as they have shown an antiviral activity against other viruses in the past. Here, a quantified extract of Hypericum perforatum was tested and found to possess a potent antiviral activity against SARS-CoV-2. The antiviral potency of the extract could be attributed to the naphtodianthrones hypericin and pseudohypericin, in contrast to other tested ingredients of the plant material, which did not show any antiviral activity. Hypericum perforatum and its main active ingredient hypericin were also effective against different SARS-CoV-2 variants (Alpha, Beta, Delta, and Omicron). Concerning its mechanism of action, evidence was obtained that Hypericum perforatum and hypericin may hold a direct virus-blocking effect against SARS-CoV-2 virus particles. Taken together, the presented data clearly emphasize the promising antiviral activity of Hypericum perforatum and its active ingredients against SARS-CoV-2 infections.

4.
Am J Infect Control ; 50(4): 420-426, 2022 04.
Article in English | MEDLINE | ID: mdl-34562528

ABSTRACT

BACKGROUND: During shortages of filtering face pieces (FFP) in a pandemic, it is necessary to implement a method for safe reuse or extended use. Our aim was to develop a simple, inexpensive and ecological method for decontamination of disposable FFPs that preserves filtration efficiency and material integrity. MATERIAL AND METHODS: Contamination of FFPs (3M Aura 9320+) with SARS-CoV-2 (1.15 × 104 PFUs), Enterococcus faecium (>106 CFUs), and physiological nasopharyngeal flora was performed prior to decontamination by submersion in a solution of 6 % acetic acid and 6 % hydrogen peroxide (6%AA/6%HP solution) over 30 minutes. Material integrity was assessed by testing the filtering efficiency, loss of fit and employing electron microscopy. RESULTS AND DISCUSSION: Decontamination with the 6%AA/6%HP solution resulted in the complete elimination of SARS-CoV-2, E. faecium and physiological nasopharyngeal flora. Material characterization post-treatment showed neither critical material degradation, loss of fit or reduction of filtration efficiency. Electron microscopy revealed no damage to the fibers, and the rubber bands' elasticity was not affected by the decontamination procedure. No concerning residuals of the decontamination procedure were found. CONCLUSION: The simple application and widespread availability of 6%AA/6%HP solution for decontaminating disposable FFPs make this solution globally viable, including developing and third world countries.


Subject(s)
COVID-19 , Pandemics , COVID-19/prevention & control , Decontamination/methods , Equipment Reuse , Humans , Pandemics/prevention & control , Peracetic Acid/pharmacology , SARS-CoV-2 , Ventilators, Mechanical
5.
Viruses ; 13(10)2021 10 14.
Article in English | MEDLINE | ID: mdl-34696497

ABSTRACT

Respiratory viruses are known to be the most frequent causative mediators of lung infections in humans, bearing significant impact on the host cell signaling machinery due to their host-dependency for efficient replication. Certain cellular functions are actively induced by respiratory viruses for their own benefit. This includes metabolic pathways such as glycolysis, fatty acid synthesis (FAS) and the tricarboxylic acid (TCA) cycle, among others, which are modified during viral infections. Here, we summarize the current knowledge of metabolic pathway modifications mediated by the acute respiratory viruses respiratory syncytial virus (RSV), rhinovirus (RV), influenza virus (IV), parainfluenza virus (PIV), coronavirus (CoV) and adenovirus (AdV), and highlight potential targets and compounds for therapeutic approaches.


Subject(s)
Citric Acid Cycle/physiology , Energy Metabolism/physiology , Fatty Acids/biosynthesis , Glycolysis/physiology , Respiratory Tract Infections/pathology , Respiratory Tract Infections/virology , Adenoviridae/metabolism , Coronavirus/metabolism , Humans , Orthomyxoviridae/metabolism , Parainfluenza Virus 1, Human/metabolism , Respiratory Syncytial Viruses/metabolism , Rhinovirus/metabolism
6.
Article in English | MEDLINE | ID: mdl-31871235

ABSTRACT

Cells respond to extracellular agents by activation of intracellular signaling pathways. Viruses can be regarded as such agents, leading to a firework of signaling inside the cell, primarily induced by pathogen-associated molecular patterns (PAMPs) that provoke safeguard mechanisms to defend from the invader. In the constant arms race between pathogen and cellular defense, viruses not only have evolved mechanisms to suppress or misuse supposedly antiviral signaling processes for their own benefit but also actively induce signaling to promote replication. This creates viral dependencies that may be exploited for novel strategies of antiviral intervention. Here, we will summarize the current knowledge of activation and function of influenza virus-induced signaling pathways with a focus on nuclear factor (NF)-κB signaling, mitogen-activated protein kinase cascades, and the phosphatidylinositol-3-kinase pathway. We will discuss the opportunities and drawbacks of targeting these signaling pathways for antiviral intervention.


Subject(s)
Orthomyxoviridae/drug effects , Orthomyxoviridae/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Animals , Antiviral Agents/pharmacology , Apoptosis/drug effects , Humans , Mitogen-Activated Protein Kinases/physiology , NF-kappa B/physiology , Orthomyxoviridae/growth & development , Viral Nonstructural Proteins/physiology , Virus Replication/drug effects
7.
J Infect Dis ; 223(10): 1806-1816, 2021 05 28.
Article in English | MEDLINE | ID: mdl-32959872

ABSTRACT

BACKGROUND: Secondary bacterial coinfections are ranked as a leading cause of hospitalization and morbid conditions associated with influenza. Because vitamin A deficiency (VAD) and insufficiency are frequent in both developed and developing countries, we asked how VAD influences coinfection severity. METHODS: VAD and control mice were infected with influenza virus for evaluation of inflammatory cytokines, cellular immune responses, and viral clearance. Influenza-infected mice were coinfected with Streptococcus pneumoniae to study weight loss and survival. RESULTS: Naive VAD mouse lungs exhibited dysregulated immune function. Neutrophils were enhanced in frequency and there was a significant reduction in RANTES (regulated on activation of normal T cells expressed and secreted), a chemokine instrumental in T-cell homing and recruitment. After influenza virus infection, VAD mice experienced failures in CD4+ T-cell recruitment and B-cell organization into lymphoid structures in the lung. VAD mice exhibited higher viral titers than controls and slow viral clearance. There were elevated levels of inflammatory cytokines and innate cell subsets in the lungs. However, arginase, a marker of alternatively activated M2 macrophages, was rare. When influenza-infected VAD animals were exposed to bacteria, they experienced a 100% mortality rate. CONCLUSION: Data showed that VAD dysregulated the immune response. Consequently, secondary bacterial infections were 100% lethal in influenza-infected VAD mice.


Subject(s)
Coinfection , Orthomyxoviridae Infections , Pneumococcal Infections/complications , Vitamin A Deficiency , Animals , Cytokines , Immunity , Lung , Mice , Mice, Inbred C57BL , Orthomyxoviridae , Orthomyxoviridae Infections/complications , Pneumococcal Infections/mortality , Streptococcus pneumoniae , Vitamin A Deficiency/complications
8.
Microorganisms ; 8(12)2020 Dec 15.
Article in English | MEDLINE | ID: mdl-33333815

ABSTRACT

Human beings are exposed to microorganisms every day. Among those, diverse commensals and potential pathogens including Staphylococcus aureus (S. aureus) compose a significant part of the respiratory tract microbiota. Remarkably, bacterial colonization is supposed to affect the outcome of viral respiratory tract infections, including those caused by influenza viruses (IV). Since 30% of the world's population is already colonized with S. aureus that can develop metabolically inactive dormant phenotypes and seasonal IV circulate every year, super-infections are likely to occur. Although IV and S. aureus super-infections are widely described in the literature, the interactions of these pathogens with each other and the host cell are only scarcely understood. Especially, the effect of quasi-dormant bacterial subpopulations on IV infections is barely investigated. In the present study, we aimed to investigate the impact of S. aureus small colony variants on the cell intrinsic immune response during a subsequent IV infection in vitro. In fact, we observed a significant impact on the regulation of pro-inflammatory factors, contributing to a synergistic effect on cell intrinsic innate immune response and induction of harmful cell death. Interestingly, the cytopathic effect, which was observed in presence of both pathogens, was not due to an increased pathogen load.

9.
PLoS One ; 15(5): e0233052, 2020.
Article in English | MEDLINE | ID: mdl-32413095

ABSTRACT

Severe influenza virus (IV) infections still represent a major challenge to public health. To combat IV infections, vaccines and antiviral compounds are available. However, vaccine efficacies vary with very limited to no protection against newly emerging zoonotic IV introductions. In addition, the development of resistant virus variants against currently available antivirals can be rapidly detected, in consequence demanding the design of novel antiviral strategies. Virus supportive cellular signaling cascades, such as the NF-κB pathway, have been identified to be promising antiviral targets against IV in in vitro and in vivo studies and clinical trials. While administration of NF-κB pathway inhibiting agents, such as LASAG results in decreased IV replication, it remained unclear whether blocking of NF-κB might sensitize cells to secondary bacterial infections, which often come along with viral infections. Thus, we examined IV and Staphylococcus aureus growth during LASAG treatment. Interestingly, our data reveal that the presence of LASAG during superinfection still leads to reduced IV titers. Furthermore, the inhibition of the NF-κB pathway resulted in decreased intracellular Staphylococcus aureus loads within epithelial cells, indicating a dependency on the pathway for bacterial uptake. Unfortunately, so far it is not entirely clear if this phenomenon might be a drawback in bacterial clearance during infection.


Subject(s)
Antiviral Agents/adverse effects , Aspirin/analogs & derivatives , Bacterial Infections/etiology , Glycine/adverse effects , Influenza, Human/drug therapy , Lysine/analogs & derivatives , NF-kappa B/antagonists & inhibitors , A549 Cells , Aspirin/adverse effects , Drug Combinations , Epithelial Cells/drug effects , Epithelial Cells/microbiology , Gene Knockdown Techniques , Humans , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/physiology , Influenza, Human/complications , Influenza, Human/virology , Lysine/adverse effects , Methicillin-Resistant Staphylococcus aureus/drug effects , Signal Transduction/drug effects , Staphylococcal Infections/etiology , Superinfection/etiology , Transcription Factor RelA/antagonists & inhibitors , Transcription Factor RelA/genetics , Virus Replication/drug effects
10.
Cell Microbiol ; 21(1): e12955, 2019 01.
Article in English | MEDLINE | ID: mdl-30223301

ABSTRACT

Activation of the Raf/MEK/ERK cascade is required for efficient propagation of several RNA and DNA viruses, including human respiratory syncytial virus (RSV). In RSV infection, activation of the Raf/MEK/ERK cascade is biphasic. An early induction within minutes after infection is associated with viral attachment. Subsequently, a second activation occurs with, so far, unknown function in the viral life cycle. In this study, we aimed to characterise the role of Raf/MEK/ERK-mediated signalling during ongoing RSV infection. Our data show that inhibition of the kinase MEK after the virus has been internalised results in a reduction of viral titers. Further functional investigations revealed that the late-stage activation of ERK is required for a specific step in RSV replication, namely, the secretory transport of the RSV fusion protein F. Thus, MEK inhibition resulted in impaired surface accumulation of the F protein. F protein surface expression is essential for efficient replication as it is involved in viral filament formation, cell fusion, and viral transmission. In summary, we provide detailed insights of how host cell signalling interferes with RSV replication and identified the Raf/MEK/ERK kinase cascade as potential target for novel anti-RSV strategies.


Subject(s)
Cell Membrane/metabolism , Host-Pathogen Interactions , MAP Kinase Signaling System , Respiratory Syncytial Virus, Human/physiology , Viral Fusion Proteins/metabolism , Virus Replication , Animals , Cells, Cultured , Humans , Protein Transport
11.
J Gen Virol ; 99(9): 1187-1198, 2018 09.
Article in English | MEDLINE | ID: mdl-30084768

ABSTRACT

The haemagglutinin (HA) of H1N1 and H3N2 influenza A virus (IAV) subtypes has to be activated by host proteases. Previous studies showed that H1N1 virus cannot replicate efficiently in Tmprss2-/- knock-out mice whereas H3N2 viruses are able to replicate to the same levels in Tmprss2-/- as in wild type (WT) mice. Here, we investigated the sequence requirements for the HA molecule that allow IAV to replicate efficiently in the absence of TMPRSS2. We showed that replacement of the H3 for the H1-loop sequence (amino acids 320 to 329, at the C-terminus of HA1) was not sufficient for equal levels of virus replication or severe pathology in Tmprss2-/- knock-out mice compared to WT mice. However, exchange of a distant amino acid from H1 to H3 sequence (E31D) in addition to the HA-loop substitution resulted in virus replication in Tmprss2-/- knock-out mice that was comparable to WT mice. The higher virus replication and lung damage was associated with increased epithelial damage and higher mortality. Our results provide further evidence and insights into host proteases as a promising target for therapeutic intervention of IAV infections.


Subject(s)
Hemagglutinins/metabolism , Influenza A virus/physiology , Orthomyxoviridae Infections/virology , Serine Endopeptidases/metabolism , Virus Replication/physiology , Amino Acid Substitution , Animals , Cloning, Molecular , Dogs , Gene Expression Regulation, Viral/physiology , Hemagglutinins/chemistry , Madin Darby Canine Kidney Cells , Mice , Mice, Knockout , Models, Molecular , Mutagenesis , Protein Conformation , Serine Endopeptidases/genetics
12.
Cell Rep ; 11(10): 1591-603, 2015 Jun 16.
Article in English | MEDLINE | ID: mdl-26051937

ABSTRACT

Incursions of new pathogenic viruses into humans from animal reservoirs are occurring with alarming frequency. The molecular underpinnings of immune recognition, host responses, and pathogenesis in this setting are poorly understood. We studied pandemic influenza viruses to determine the mechanism by which increasing glycosylation during evolution of surface proteins facilitates diminished pathogenicity in adapted viruses. ER stress during infection with poorly glycosylated pandemic strains activated the unfolded protein response, leading to inflammation, acute lung injury, and mortality. Seasonal strains or viruses engineered to mimic adapted viruses displaying excess glycans on the hemagglutinin did not cause ER stress, allowing preservation of the lungs and survival. We propose that ER stress resulting from recognition of non-adapted viruses is utilized to discriminate "non-self" at the level of protein processing and to activate immune responses, with unintended consequences on pathogenesis. Understanding this mechanism should improve strategies for treating acute lung injury from zoonotic viral infections.


Subject(s)
Acute Lung Injury/immunology , Acute Lung Injury/virology , Endoplasmic Reticulum Stress/immunology , Acute Lung Injury/metabolism , Animals , Dogs , Glycosylation , HEK293 Cells , Humans , Immunity, Innate/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza, Human/immunology , Influenza, Human/metabolism , Influenza, Human/virology , Madin Darby Canine Kidney Cells , Mice , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology , Signal Transduction
13.
J Infect Dis ; 211(9): 1418-28, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25367299

ABSTRACT

BACKGROUND: Nonstructural protein 1 (NS1) proteins from avian influenza viruses like the 1918 pandemic NS1 are capable of inhibiting the key signaling integrator c-Abl (Abl1), resulting in massive cytopathic cell alterations. METHODS: In the current study, we addressed the consequences of NS1-mediated alteration of c-Abl on acute lung injury and pathogenicity in an in vivo mouse model. RESULTS: Comparing isogenic strains that differ only in their ability to inhibit c-Abl, we observed elevated pathogenicity for the c-Abl-inhibiting virus. NS1-mediated blockade of c-Abl resulted in severe lung pathology and massive edema formation and facilitated secondary bacterial pneumonia. This phenotype was independent of differences in replication and immune responses, defining it as an NS1 virulence mechanism distinct from its canonical functions. Microarray analysis revealed extensive downregulation of genes involved in cell integrity and vascular endothelial regulation. CONCLUSIONS: NS1 protein-mediated blockade of c-Abl signaling drives acute lung injury and primes for bacterial coinfections revealing potential insights into the pathogenicity of the 1918 pandemic virus.


Subject(s)
Acute Lung Injury/etiology , Bacterial Infections/etiology , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza Pandemic, 1918-1919 , Orthomyxoviridae Infections/virology , Proto-Oncogene Proteins c-abl/metabolism , Viral Nonstructural Proteins/metabolism , Acute Lung Injury/complications , Acute Lung Injury/pathology , Animals , Biomarkers , Humans , Lung/pathology , Lung/virology , Mice , Proto-Oncogene Proteins c-abl/genetics
14.
Nat Commun ; 5: 5645, 2014 Dec 09.
Article in English | MEDLINE | ID: mdl-25487526

ABSTRACT

The type I interferon (IFN) response represents the first line of defence to invading pathogens. Internalized viral ribonucleoproteins (vRNPs) of negative-strand RNA viruses induce an early IFN response by interacting with retinoic acid inducible gene I (RIG-I) and its recruitment to mitochondria. Here we employ three-dimensional stochastic optical reconstruction microscopy (STORM) to visualize incoming influenza A virus (IAV) vRNPs as helical-like structures associated with mitochondria. Unexpectedly, an early IFN induction in response to vRNPs is not detected. A distinct amino-acid motif in the viral polymerases, PB1/PA, suppresses early IFN induction. Mutation of this motif leads to reduced pathogenicity in vivo, whereas restoration increases it. Evolutionary dynamics in these sequences suggest that completion of the motif, combined with viral reassortment can contribute to pandemic risks. In summary, inhibition of the immediate anti-viral response is 'pre-packaged' in IAV in the sequences of vRNP-associated polymerase proteins.


Subject(s)
DEAD-box RNA Helicases/chemistry , Influenza A virus/pathogenicity , Interferon Type I/immunology , Mitochondria/virology , Virion/chemistry , Amino Acid Motifs , Animals , Cell Line, Tumor , DEAD Box Protein 58 , Evolution, Molecular , Female , Humans , Imaging, Three-Dimensional , Lung/immunology , Lung/virology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , RNA Viruses/pathogenicity , Receptors, Immunologic , Viral Load , Viral Proteins/chemistry , Virulence
15.
Cell Microbiol ; 16(12): 1854-74, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25052580

ABSTRACT

The non-structural protein 1 (NS1) of influenza A viruses (IAV) encodes several src homology (SH) binding motifs (bm) (one SH2bm, up to two SH3bm), which mediate interactions with host cell proteins. In contrast to NS1 of human IAV, NS1 of avian strains possess the second SH3bm (SH3(II)bm) consensus sequence. Since our former studies demonstrated an NS1-CRK interaction, mediated by this motif, here, we addressed the regulatory properties of this SH3bm for cellular signalling. Initially, we observed a reduced basal CRK phosphorylation upon infection with avian IAV harbouring an NS1 with an SH3(II)bm in contrast to human IAV. Reduced activity of the tyrosine kinase c-Abl was identified to be responsible for reduced CRK phosphorylation. Further, binding of NS1 to c-Abl was determined, and mutational manipulation of the SH3(II)bm illustrated the necessity of this motif for c-Abl inhibition. Interestingly, Abl kinase inhibition resulted in impaired avian IAV propagation and pathogenicity and mutational analysis linked the pronounced inhibition of c-Abl to cytopathogenic cell alterations upon avian IAV infections. Taken together, NS1 proteins of avian IAV interfere with the kinase activity of c-Abl, a major cellular signalling integrator that controls multiple signalling processes and cell fate regulations apparently including IAV infections.


Subject(s)
Host-Pathogen Interactions , Influenza A virus/physiology , Influenza in Birds/virology , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Viral Nonstructural Proteins/metabolism , Animals , Birds , Cell Line , Humans , Influenza A virus/isolation & purification , Protein Binding
16.
J Virol ; 88(16): 9038-48, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24899174

ABSTRACT

UNLABELLED: Influenza A virus (IAV) replication depends on the interaction of virus proteins with host factors. The viral nonstructural protein 1 (NS1) is essential in this process by targeting diverse cellular functions, including mRNA splicing and translation, cell survival, and immune defense, in particular the type I interferon (IFN-I) response. In order to identify host proteins targeted by NS1, we established a replication-competent recombinant IAV that expresses epitope-tagged forms of NS1 and NS2, which are encoded by the same gene segment, allowing purification of NS proteins during natural cell infection and analysis of interacting proteins by quantitative mass spectrometry. We identified known NS1- and NS2-interacting proteins but also uncharacterized proteins, including PACT, an important cofactor for the IFN-I response triggered by the viral RNA-sensor RIG-I. We show here that NS1 binds PACT during virus replication and blocks PACT/RIG-I-mediated activation of IFN-I, which represents a critical event for the host defense. Protein interaction and interference with IFN-I activation depended on the functional integrity of the highly conserved RNA binding domain of NS1. A mutant virus with deletion of NS1 induced high levels of IFN-I in control cells, as expected; in contrast, shRNA-mediated knockdown of PACT compromised IFN-I activation by the mutant virus, but not wild-type virus, a finding consistent with the interpretation that PACT (i) is essential for IAV recognition and (ii) is functionally compromised by NS1. Together, our data describe a novel approach to identify virus-host protein interactions and demonstrate that NS1 interferes with PACT, whose function is critical for robust IFN-I production. IMPORTANCE: Influenza A virus (IAV) is an important human pathogen that is responsible for annual epidemics and occasional devastating pandemics. Viral replication and pathogenicity depends on the interference of viral factors with components of the host defense system, particularly the type I interferon (IFN-I) response. The viral NS1 protein is known to counteract virus recognition and IFN-I production, but the molecular mechanism is only partially defined. We used a novel proteomic approach to identify host proteins that are bound by NS1 during virus replication and identified the protein PACT, which had previously been shown to be involved in virus-mediated IFN-I activation. We find that NS1 prevents PACT from interacting with an essential component of the virus recognition pathway, RIG-I, thereby disabling efficient IFN-I production. These observations provide an important piece of information on how IAV efficiently counteracts the host immune defense.


Subject(s)
Antiviral Agents/metabolism , Influenza A virus/metabolism , RNA-Binding Proteins/metabolism , Viral Nonstructural Proteins/metabolism , Animals , Cell Line , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Dogs , HEK293 Cells , Humans , Influenza A virus/genetics , Interferon-beta/genetics , Interferon-beta/metabolism , Madin Darby Canine Kidney Cells , Promoter Regions, Genetic/genetics , Proteomics/methods , RNA, Viral/genetics , RNA-Binding Proteins/genetics , Receptors, Immunologic , Viral Nonstructural Proteins/genetics , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication/genetics
17.
J Virol ; 88(16): 8843-52, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24872593

ABSTRACT

UNLABELLED: A hallmark cell response to influenza A virus (IAV) infections is the phosphorylation and activation of c-jun N-terminal kinase (JNK). However, so far it is not fully clear which molecules are involved in the activation of JNK upon IAV infection. Here, we report that the transfection of influenza viral-RNA induces JNK in a retinoic acid-inducible gene I (RIG-I)-dependent manner. However, neither RIG-I-like receptors nor MyD88-dependent Toll-like receptors were found to be involved in the activation of JNK upon IAV infection. Viral JNK activation may be blocked by addition of cycloheximide and heat shock protein inhibitors during infection, suggesting that the expression of an IAV-encoded protein is responsible for JNK activation. Indeed, the overexpression of nonstructural protein 1 (NS1) of certain IAV subtypes activated JNK, whereas those of some other subtypes failed to activate JNK. Site-directed mutagenesis experiments using NS1 of the IAV H7N7, H5N1, and H3N2 subtypes identified the amino acid residue phenylalanine (F) at position 103 to be decisive for JNK activation. Cleavage- and polyadenylation-specific factor 30 (CPSF30), whose binding to NS1 is stabilized by the amino acids F103 and M106, is not involved in JNK activation. Conclusively, subtype-specific sequence variations in the IAV NS1 protein result in subtype-specific differences in JNK signaling upon IAV infection. IMPORTANCE: Influenza A virus (IAV) infection leads to the activation or modulation of multiple signaling pathways. Here, we demonstrate for the first time that the c-jun N-terminal kinase (JNK), a long-known stress-activated mitogen-activated protein (MAP) kinase, is activated by RIG-I when cells are treated with IAV RNA. However, at the same time, nonstructural protein 1 (NS1) of IAV has an intrinsic JNK-activating property that is dependent on IAV subtype-specific amino acid variations around position 103. Our findings identify two different and independent pathways that result in the activation of JNK in the course of an IAV infection.


Subject(s)
Amino Acid Sequence/genetics , Influenza A virus/genetics , Influenza, Human/genetics , JNK Mitogen-Activated Protein Kinases/genetics , Orthomyxoviridae Infections/genetics , Viral Nonstructural Proteins/genetics , Animals , Cell Line , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Dogs , HEK293 Cells , Humans , Influenza A virus/metabolism , Influenza, Human/metabolism , Influenza, Human/virology , JNK Mitogen-Activated Protein Kinases/metabolism , Madin Darby Canine Kidney Cells , Mutagenesis, Site-Directed/methods , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology , Signal Transduction/genetics , Tretinoin/metabolism , Viral Nonstructural Proteins/metabolism
18.
J Infect Dis ; 209(4): 532-41, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-23983213

ABSTRACT

The identification of amino acid motifs responsible for increased virulence and/or transmission of influenza viruses is of enormous importance to predict pathogenicity of upcoming influenza strains. We phenotypically and genotypically compared 2 variants of influenza virus A/PR/8/34 with different passage histories. The analysis revealed differences in virulence due to an altered type I interferon (IFN) induction, as evidenced by experiments using IFNAR(-/-) mice. Interestingly, these differences were not due to altered functions of the well-known viral IFN antagonists NS1 or PB1-F2. Using reassortant viruses, we showed that differences in the polymerase proteins and nucleoprotein determined the altered virulence. In particular, changes in PB1 and PA contributed to an altered host type I IFN response, indicating IFN antagonistic properties of these proteins. Thus, PB1 and PA appear to harbor previously unknown virulence markers, which may prove helpful in assessing the risk potential of emerging influenza viruses.


Subject(s)
Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza, Human/virology , Amino Acid Substitution , Animals , Cell Line , Chickens , Cytokines/analysis , Dogs , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human/immunology , Interferon Type I/metabolism , Kaplan-Meier Estimate , Lung/chemistry , Lung/virology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Receptor, Interferon alpha-beta/genetics , STAT1 Transcription Factor , Viral Nonstructural Proteins/metabolism , Viral Proteins/metabolism , Virulence/immunology
19.
Antiviral Res ; 99(3): 336-44, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23811282

ABSTRACT

The appearance of pandemic H1N1 and highly pathogenic avian H5N1 viruses in humans as well as the emergence of seasonal H1N1 variants resistant against neuraminidase inhibitors highlight the urgent need for new and amply available antiviral drugs. We and others have demonstrated that influenza virus misuses the cellular IKK/NF-kappaB signaling pathway for efficient replication suggesting that this module may be a suitable target for antiviral intervention. Here, we show that the novel NF-kappaB inhibitor SC75741 significantly protects mice against infection with highly pathogenic avian influenza A viruses of the H5N1 and H7N7 subtypes. Treatment was efficient when SC75741 was given intravenously in a concentration of 5mg/kg/day. In addition, application of SC75741 via the intraperitoneal route resulted in a high bioavailability and was also efficient against influenza when given 15 mg/kg/day or 7.5 mg/kg/twice a day. Protection was achieved when SC75741 was given for seven consecutive days either prior to infection or as late as four days after infection. SC75741 treatment showed no adverse effects in the concentrations required to protect mice against influenza virus infection. Although more pre-clinical studies are needed SC75741 might be a promising candidate for a novel antiviral drug against influenza viruses that targets the host cell rather than the virus itself.


Subject(s)
Antiviral Agents/administration & dosage , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H5N1 Subtype/drug effects , Influenza A Virus, H7N7 Subtype/drug effects , Influenza, Human/drug therapy , NF-kappa B/antagonists & inhibitors , Animals , Birds , Drug Evaluation, Preclinical , Female , Humans , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza A Virus, H5N1 Subtype/physiology , Influenza A Virus, H7N7 Subtype/pathogenicity , Influenza A Virus, H7N7 Subtype/physiology , Influenza in Birds/virology , Influenza, Human/genetics , Influenza, Human/metabolism , Influenza, Human/virology , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Virulence
20.
Cell Microbiol ; 15(7): 1198-211, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23320394

ABSTRACT

Ongoing human infections with highly pathogenic avian H5N1 viruses and the emergence of the pandemic swine-origin influenza viruses (IV) highlight the permanent threat elicited by these pathogens. Occurrence of resistant seasonal and pandemic strains against the currently licensed antiviral medications points to the urgent need for new and amply available anti-influenza drugs. The recently identified virus-supportive function of the cellular IKK/NF-κB signalling pathway suggests this signalling module as a potential target for antiviral intervention. We characterized the NF-κB inhibitor SC75741 as a broad and efficient blocker of IV replication in non-toxic concentrations. The underlying molecular mechanism of SC75741 action involves impaired DNA binding of the NF-κB subunit p65, resulting in reduced expression of cytokines, chemokines, and pro-apoptotic factors, subsequent inhibition of caspase activation and block of caspase-mediated nuclear export of viralribonucleoproteins. SC75741 reduces viral replication and H5N1-induced IL-6 and IP-10 expression in the lung of infected mice. Besides its virustatic effect the drug suppresses virus-induced overproduction of cytokines and chemokines, suggesting that it might prevent hypercytokinemia that is discussed to be an important pathogenicity determinant of highly pathogenic IV. Importantly the drug exhibits a high barrier for development of resistant virus variants. Thus, SC75741-derived drugs may serve as broadly non-toxic anti-influenza agents.


Subject(s)
Antiviral Agents/pharmacology , Influenza A Virus, H5N1 Subtype/physiology , NF-kappa B/antagonists & inhibitors , Virus Replication/drug effects , Animals , Antiviral Agents/therapeutic use , Cell Line , Disease Models, Animal , Humans , Lung/virology , Mice , Orthomyxoviridae Infections/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...