Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Antiviral Res ; 225: 105840, 2024 May.
Article in English | MEDLINE | ID: mdl-38438015

ABSTRACT

Host targeting antiviral drugs (HTA) are directed against cellular mechanisms which can be exploited by viruses. These mechanisms are essential for viral replication, because missing functions cannot be compensated by the virus. However, this assumption needs experimental proof. Here we compared the HTA Zapnometinib (ZMN), with direct acting antivirals (DAA) (Remdesivir (RDV), Molnupiravir (MPV), Nirmatrelvir (NTV), Ritonavir (RTV), Paxlovid PAX)), in terms of their potency to induce reduced drug susceptibilities in SARS-CoV-2. During serial passage of δ-B1.617.2 adaptation to all DAAs occurred, while the inhibitory capacity of ZMN was not altered. Known single nucleotide polymorphisms (SNPs) responsible for partial resistances were found for RDV, NTV and PAX. Additionally, the high mutagenic potential of MPV was confirmed and decreased drug efficacies were found for the first time. Reduced DAA efficacy did not alter the inhibitory potential of ZMN. These results show that ZMN confers a high barrier towards the development of viral resistance and has the potential to act against partially DAA-insensitive viruses.


Subject(s)
COVID-19 , Cytidine/analogs & derivatives , Hepatitis C, Chronic , Hydroxylamines , Humans , Antiviral Agents , SARS-CoV-2 , Ritonavir
2.
Emerg Microbes Infect ; 12(1): 2212809, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37191590

ABSTRACT

Influenza A viruses (IAV) cause annual epidemics and occasional pandemics in humans. The most recent pandemic outbreak occurred in 2009 with H1N1pdm09. This virus, which most likely reassorted in swine before its transmission to humans, was reintroduced into the swine population and continues circulating ever since. In order to assess its potential to cause reassortants on a cellular level, human origin H1N1pdm09 and a recent Eurasian avian-like H1N1 swine IAV were (co-)passaged in the newly generated swine lung cell line C22. Co-infection with both viruses gave rise to numerous reassortants that additionally carry different mutations which can partially be found in nature as well. Reassortment most frequently affected the PB1, PA and NA segments with the swine IAV as recipient. These reassortants reached higher titers in swine lung cells and were able to replicate in genuine human lung tissue explants ex vivo, suggesting a possible zoonotic potential. Interestingly, reassortment and mutations in the viral ribonucleoprotein complex influence the viral polymerase activity in a cell type and species-specific manner. In summary, we demonstrate reassortment promiscuity of these viruses in a novel swine lung cell model and indicate a possible zoonotic potential of the reassortants.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza A virus , Influenza, Human , Orthomyxoviridae Infections , Swine Diseases , Animals , Humans , Swine , Orthomyxoviridae Infections/epidemiology , Orthomyxoviridae Infections/veterinary , Influenza A Virus, H1N1 Subtype/genetics , Reassortant Viruses/genetics , Influenza A virus/genetics , Genomics , Swine Diseases/epidemiology , Influenza, Human/epidemiology
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.
J Virol ; 95(20): e0067221, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34319782

ABSTRACT

Influenza A virus (IAV) is the causative agent of flu disease that results in annual epidemics and occasional pandemics. IAV alters several signaling pathways of the cellular host response in order to promote its replication. Therefore, some of these pathways can serve as targets for novel antiviral agents. Here, we show that c-Jun NH2-terminal kinase (JNK)-interacting protein 4 (JIP4) is dynamically phosphorylated in IAV infection. The lack of JIP4 resulted in higher virus titers, with significant differences in viral protein and mRNA accumulation as early as within the first replication cycle. In accordance, decreased IAV titers and protein accumulation were observed during the overexpression of JIP4. Strikingly, the antiviral function of JIP4 does not originate from modulation of JNK or p38 mitogen-activated protein kinase (MAPK) pathways or from altered expression of interferons or interferon-stimulated genes but rather originates from a direct reduction of viral polymerase activity. Furthermore, the interference of JIP4 with IAV replication seems to be linked to the phosphorylation of the serine at position 730 that is sufficient to impede the viral polymerase. Collectively, we provide evidence that JIP4, a host protein modulated in IAV infection, exhibits antiviral properties that are dynamically controlled by its phosphorylation at S730. IMPORTANCE Influenza A virus (IAV) infection is a world health concern, and current treatment options encounter high rates of resistance. Our group investigates host pathways modified in IAV infection as promising new targets. The host protein JIP4 is dynamically phosphorylated in IAV infection. JIP4 absence resulted in higher virus titers and viral protein and mRNA accumulation within the first replication cycle. Accordingly, decreased IAV titers and protein accumulation were observed during JIP4 overexpression. Strikingly, the antiviral function of JIP4 does not originate from modulation of JNK or p38 MAPK pathways or from altered expression of interferons or interferon-stimulated genes but rather originates from a reduction in viral polymerase activity. The interference of JIP4 with IAV replication is linked to the phosphorylation of serine 730. We provide evidence that JIP4, a host protein modulated in IAV infection, exhibits antiviral properties that are dynamically controlled by its phosphorylation at S730.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Influenza A virus/metabolism , A549 Cells , Animals , Chlorocebus aethiops , Dogs , Host-Pathogen Interactions/genetics , Humans , Immune Evasion/genetics , Immunity, Innate/genetics , Influenza A virus/physiology , Influenza, Human/virology , Interferons/genetics , Madin Darby Canine Kidney Cells , Phosphorylation , Signal Transduction/genetics , Vero Cells , Virus Replication/genetics , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Cell Microbiol ; 23(6): e13323, 2021 06.
Article in English | MEDLINE | ID: mdl-33655690

ABSTRACT

Nonsense-mediated mRNA decay (NMD) was identified as a process to degrade flawed cellular messenger RNA (mRNA). Within the last decades it was also shown that NMD carries virus-restricting capacities and thus could be considered a part of the cellular antiviral system. As this was shown to affect primarily positive-sense single stranded RNA ((+)ssRNA) viruses there is only scarce knowledge if this also applies to negative-sense single stranded RNA ((-)ssRNA) viruses. Influenza A viruses (IAVs) harbour a segmented (-)ssRNA genome. During their replication IAVs produce numerous RNA transcripts and simultaneously impair cellular transcription and translation. The viral mRNAs hold several molecular patterns which can elicit NMD and in turn would lead to their degradation. This, in consequence, may mitigate viral propagation. Thus, we examined if a knockdown or a pharmacological inhibition of NMD key components may influence IAV replication. Additionally, we performed similar experiments with respiratory syncytial virus (RSV), another (-)ssRNA virus, but with a non-segmented genome. Although it seemed that a knockdown of up-frameshift protein 1 (UPF1), the central NMD factor, slightly increased viral mRNA and protein levels, no significant alteration of viral replication could be observed, implying that the NMD machinery may not have restricting capacities against (-)ssRNA viruses.


Subject(s)
Host-Pathogen Interactions , Influenza A virus/genetics , Influenza A virus/physiology , Nonsense Mediated mRNA Decay , RNA, Messenger/metabolism , RNA, Viral/metabolism , A549 Cells , Gene Silencing , Humans , RNA Helicases/genetics , RNA, Viral/genetics , Respiratory Syncytial Viruses/genetics , Trans-Activators/genetics , Virus Replication
7.
J Virol ; 95(6)2021 02 24.
Article in English | MEDLINE | ID: mdl-33408177

ABSTRACT

Influenza A virus (IAV) nonstructural protein 1 (NS1) is a protein with multiple functions that are regulated by phosphorylation. Phosphoproteomic screening of H1N1 virus-infected cells revealed that NS1 was phosphorylated at serine 205 in intermediate stages of the viral life cycle. Interestingly, S205 is one of six amino acid changes in NS1 of post-pandemic H1N1 viruses currently circulating in humans compared to the original swine-origin 2009 pandemic (H1N1pdm09) virus, suggesting a role in host adaptation. To identify NS1 functions regulated by S205 phosphorylation, we generated recombinant PR8 H1N1 NS1 mutants with S205G (nonphosphorylatable) or S205N (H1N1pdm09 signature), as well as H1N1pdm09 viruses harboring the reverse mutation NS1 N205S or N205D (phosphomimetic). Replication of PR8 NS1 mutants was attenuated relative to wild-type (WT) virus replication in a porcine cell line. However, PR8 NS1 S205N showed remarkably higher attenuation than PR8 NS1 S205G in a human cell line, highlighting a potential host-independent advantage of phosphorylatable S205, while an asparagine at this position led to a potential host-specific attenuation. Interestingly, PR8 NS1 S205G did not show polymerase activity-enhancing functions, in contrast to the WT, which can be attributed to diminished interaction with cellular restriction factor DDX21. Analysis of the respective kinase mediating S205 phosphorylation indicated an involvement of casein kinase 2 (CK2). CK2 inhibition significantly reduced the replication of WT viruses and decreased NS1-DDX21 interaction, as observed for NS1 S205G. In summary, NS1 S205 is required for efficient NS1-DDX21 binding, resulting in enhanced viral polymerase activity, which is likely to be regulated by transient phosphorylation.IMPORTANCE Influenza A viruses (IAVs) still pose a major threat to human health worldwide. As a zoonotic virus, IAV can spontaneously overcome species barriers and even reside in new hosts after efficient adaptation. Investigation of the functions of specific adaptational mutations can lead to a deeper understanding of viral replication in specific hosts and can probably help to find new targets for antiviral intervention. In the present study, we analyzed the role of NS1 S205, a phosphorylation site that was reacquired during the circulation of pandemic H1N1pdm09 "swine flu" in the human host. We found that phosphorylation of human H1N1 virus NS1 S205 is mediated by the cellular kinase CK2 and is needed for efficient interaction with human host restriction factor DDX21, mediating NS1-induced enhancement of viral polymerase activity. Therefore, targeting CK2 activity might be an efficient strategy for limiting the replication of IAVs circulating in the human population.


Subject(s)
Influenza A virus/physiology , RNA-Dependent RNA Polymerase/metabolism , Serine/metabolism , Viral Nonstructural Proteins/metabolism , Adaptation, Physiological/genetics , Animals , Casein Kinase II/metabolism , Cell Line , DEAD-box RNA Helicases/metabolism , Host-Pathogen Interactions , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/metabolism , Influenza A Virus, H1N1 Subtype/physiology , Influenza A virus/genetics , Influenza A virus/metabolism , Mutation , Phosphorylation , Protein Binding , Swine , Viral Nonstructural Proteins/genetics , Virus Replication
8.
PLoS Pathog ; 16(8): e1008775, 2020 08.
Article in English | MEDLINE | ID: mdl-32866218

ABSTRACT

Small RNA viruses only have a very limited coding capacity, thus most viral proteins have evolved to fulfill multiple functions. The highly conserved matrix protein 1 (M1) of influenza A viruses is a prime example for such a multifunctional protein, as it acts as a master regulator of virus replication whose different functions have to be tightly regulated. The underlying mechanisms, however, are still incompletely understood. Increasing evidence points towards an involvement of posttranslational modifications in the spatio-temporal regulation of M1 functions. Here, we analyzed the role of M1 tyrosine phosphorylation in genuine infection by using recombinant viruses expressing M1 phosphomutants. Presence of M1 Y132A led to significantly decreased viral replication compared to wildtype and M1 Y10F. Characterization of phosphorylation dynamics by mass spectrometry revealed the presence of Y132 phosphorylation in M1 incorporated into virions that is most likely mediated by membrane-associated Janus kinases late upon infection. Molecular dynamics simulations unraveled a potential phosphorylation-induced exposure of the positively charged linker domain between helices 4 and 5, supposably acting as interaction platform during viral assembly. Consistently, M1 Y132A showed a defect in lipid raft localization due to reduced interaction with viral HA protein resulting in a diminished structural stability of viral progeny and the formation of filamentous particles. Importantly, reduced M1-RNA binding affinity resulted in an inefficient viral genome incorporation and the production of non-infectious virions that interferes with virus pathogenicity in mice. This study advances our understanding of the importance of dynamic phosphorylation as a so far underestimated level of regulation of multifunctional viral proteins and emphasizes the potential feasibility of targeting posttranslational modifications of M1 as a novel antiviral intervention.


Subject(s)
Influenza A virus/metabolism , Mutation, Missense , Viral Matrix Proteins/metabolism , A549 Cells , Amino Acid Substitution , Animals , Dogs , Female , HEK293 Cells , Humans , Influenza A virus/genetics , Madin Darby Canine Kidney Cells , Male , Mice , Mice, Transgenic , Phosphorylation , Viral Matrix Proteins/genetics
9.
Proc Natl Acad Sci U S A ; 117(28): 16557-16566, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32601201

ABSTRACT

Influenza viruses (IV) exploit a variety of signaling pathways. Previous studies showed that the rapidly accelerated fibrosarcoma/mitogen-activated protein kinase/extracellular signal-regulated kinase (Raf/MEK/ERK) pathway is functionally linked to nuclear export of viral ribonucleoprotein (vRNP) complexes, suggesting that vRNP export is a signaling-induced event. However, the underlying mechanism remained completely enigmatic. Here we have dissected the unknown molecular steps of signaling-driven vRNP export. We identified kinases RSK1/2 as downstream targets of virus-activated ERK signaling. While RSK2 displays an antiviral role, we demonstrate a virus-supportive function of RSK1, migrating to the nucleus to phosphorylate nucleoprotein (NP), the major constituent of vRNPs. This drives association with viral matrix protein 1 (M1) at the chromatin, important for vRNP export. Inhibition or knockdown of MEK, ERK or RSK1 caused impaired vRNP export and reduced progeny virus titers. This work not only expedites the development of anti-influenza strategies, but in addition demonstrates converse actions of different RSK isoforms.


Subject(s)
Influenza A virus/metabolism , Influenza, Human/virology , Ribonucleoproteins/metabolism , Active Transport, Cell Nucleus , Cell Nucleus/metabolism , Cell Nucleus/virology , Humans , Influenza A virus/genetics , Influenza, Human/genetics , Influenza, Human/metabolism , MAP Kinase Signaling System , Nuclear Export Signals , Ribonucleoproteins/genetics , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism
10.
Cell Microbiol ; 22(2): e13143, 2020 02.
Article in English | MEDLINE | ID: mdl-31711273

ABSTRACT

The innate immune system, in particular the type I interferon (IFN) response, is a powerful defence against virus infections. In turn, many if not all viruses have evolved various means to circumvent, resist, or counteract this host response to ensure efficient replication and propagation. Influenza viruses are no exception to this rule, and several viral proteins have been described to possess IFN-antagonistic functions. Although the viral nonstructural protein 1 appears to be a major antagonist in influenza A and B viruses (IAV and IBV), we have previously shown that a specific motif in the IAV polymerase proteins exerts an IFN-suppressive function very early in infection. The question remained whether a similar function would also exist in IBV polymerases. Here, we show that indeed a specific amino acid position (A523) of the PB1 protein in the IBV polymerase complex confers IFN-antagonistic properties. Mutation of this position leads to enhanced activation of the IFN-mediated signalling pathway after infection and subsequent reduction of virus titres. This indicates that inhibition of innate immune responses is a conserved activity shared by polymerase proteins of IAV and IBV.


Subject(s)
Influenza B virus , Interferon Type I/antagonists & inhibitors , Nucleocapsid Proteins/immunology , RNA-Dependent RNA Polymerase/immunology , Viral Proteins/immunology , A549 Cells , Animals , Chlorocebus aethiops , HEK293 Cells , Host-Pathogen Interactions , Humans , Immunity, Innate , Influenza B virus/enzymology , Influenza B virus/immunology , Influenza, Human/virology , Vero Cells
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 Microbiol ; 18(6): 784-91, 2016 06.
Article in English | MEDLINE | ID: mdl-26687707

ABSTRACT

Phosphorylation and dephosphorylation acts as a fundamental molecular switch that alters protein function and thereby regulates many cellular processes. The non-structural protein 1 (NS1) of influenza A virus is an important factor regulating virulence by counteracting cellular immune responses against viral infection. NS1 was shown to be phosphorylated at several sites; however, so far, no function has been conclusively assigned to these post-translational events yet. Here, we show that the newly identified phospho-site threonine 49 of NS1 is differentially phosphorylated in the viral replication cycle. Phosphorylation impairs binding of NS1 to double-stranded RNA and TRIM25 as well as complex formation with RIG-I, thereby switching off its interferon antagonistic activity. Because phosphorylation was shown to occur at later stages of infection, we hypothesize that at this stage other functions of the multifunctional NS1 beyond its interferon-antagonistic activity are needed.


Subject(s)
Influenza A Virus, H1N1 Subtype/physiology , Interferon-beta/metabolism , Threonine/metabolism , Viral Nonstructural Proteins/metabolism , Animals , Cell Line , DEAD Box Protein 58/metabolism , Dogs , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/pathogenicity , Interferon-beta/genetics , Madin Darby Canine Kidney Cells/virology , Mutation , Phosphorylation , Promoter Regions, Genetic , Receptors, Immunologic , Transcription Factors/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Viral Nonstructural Proteins/genetics , Virus Replication
13.
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
14.
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
15.
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
16.
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
17.
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
18.
Vaccine ; 30(30): 4480-9, 2012 Jun 22.
Article in English | MEDLINE | ID: mdl-22575164

ABSTRACT

The nucleoprotein (NP) of influenza A virus (IAV) is associated with many different functions including host range restriction. Multiple sequence alignment analyses of 748 NP gene sequences from GenBank revealed a highly conserved region of 60 nucleotides within the ORF at the 3'-ends of the cRNA, in some codons even silent mutations were not found. This suggests that the RNA structure integrity within this region is crucial for IAV replication. To explore the impact of these conserved nucleotides for viral replication we created mutant viruses with one or more silent mutations in the respective region of the NP gene of the IAV strain A/WSN/33 (H1N1) (WSN). Assessment of viral replication of these WSN mutant viruses showed significant growth disadvantages when compared to the corresponding parental strain. On the basis of these findings we tested whether the attenuation of IAV by introduction of silent mutations into the NP gene may serve as a strategy to create a live attenuated vaccine. Mice vaccinated with the attenuated WSN mutant survived a lethal challenge dose of wild type WSN virus or the mouse adapted pandemic H1N1v strain A/Hamburg/4/2009. Thus, introduction of silent mutations in the NP of IAV is a feasible approach for a novel vaccination strategy allowing attenuation of the master strain but leaves the antigenicity of the gene product unaltered. This principle is potentially applicable for all viruses with segmented genomes.


Subject(s)
Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza Vaccines/genetics , Orthomyxoviridae Infections/prevention & control , RNA-Binding Proteins/genetics , Viral Core Proteins/genetics , Animals , Base Sequence , Conserved Sequence , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/physiology , Mice , Mice, Inbred BALB C , Mutation , Nucleocapsid Proteins , RNA-Binding Proteins/immunology , Vaccines, Attenuated/genetics , Viral Core Proteins/immunology , Virus Replication
19.
Am J Pathol ; 180(6): 2361-74, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22525464

ABSTRACT

The nonstructural protein 1 (A/NS1) of influenza A viruses (IAV) harbors several src homology (SH)-binding motifs (bm) that mediate interactions with cellular proteins. In contrast to the sequence variability of the second SH3bm, tyrosine 89, within the SH2bm is a highly conserved residue among IAV strains. This prompted us to evaluate the necessity of this SH2bm for IAV virulence. In an in vivo mouse model, we observed drastic reductions in weight loss, mortality, and virus titers in lung and bronchoalveolar lavage fluid after infection with the mutant virus PR8 A/NS1-Y89F (PR8 Y89F) when compared with wild-type virus (PR8 wt). Concomitantly, we observed decreased inflammation and less severe pathologic changes, reflecting reduced levels of virus titers. At histologic analysis, lungs infected with PR8 wt virus showed widespread destruction of the bronchiolar epithelium, with extensive distribution of virus antigen within tracheal, bronchial, bronchiolar, and alveolar epithelium. In marked contrast, the bronchiolar epithelium after infection with the mutant PR8 Y89F virus was entirely intact, and the severity and extent of viral infection was reduced and strongly restricted to alveoli. These findings demonstrate that change of a single residue of the highly conserved SH2bm within the A/NS1 results in restricted virus spread in mouse lung and strongly reduced virulence, which illustrates the necessity of the SH2bm for IAV-induced pathogenicity.


Subject(s)
Influenza A virus/genetics , Orthomyxoviridae Infections/virology , Point Mutation , Viral Nonstructural Proteins/genetics , Viral Tropism/genetics , Animals , Bronchoalveolar Lavage Fluid/immunology , Bronchoalveolar Lavage Fluid/virology , Chemokines/biosynthesis , Cytokines/biosynthesis , Epithelial Cells/virology , Influenza A virus/isolation & purification , Influenza A virus/pathogenicity , Influenza A virus/physiology , Lung/immunology , Lung/pathology , Lung/virology , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/pathology , Protein Interaction Domains and Motifs/genetics , Virulence/genetics , Virus Replication/genetics , Weight Loss , src Homology Domains/genetics
20.
Biol Chem ; 392(12): 1135-44, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22050228

ABSTRACT

PB1-F2 is a nonstructural protein of influenza viruses encoded by the PB1 gene segment from a +1 open reading frame. It has been shown that PB1-F2 contributes to viral pathogenicity, although the underlying mechanisms are still unclear. Induction of type I interferon (IFN) and the innate immune response are the first line of defense against viral infection. Here we show that influenza A viruses (IAVs) lacking the PB1-F2 protein induce an enhanced expression of IFN-ß and IFN-stimulated genes in infected epithelial cells. Studying molecular mechanisms underlying the PB1-F2-mediated IFN antagonistic activity showed that PB1-F2 interferes with the RIG-I/MAVS protein complex thereby inhibiting the activation of the downstream transcription factor IFN regulatory factor 3. These findings were also reflected in in vivo studies demonstrating that infection with PR8 wild-type (wt) virus resulted in higher lung titers and a more severe onset of disease compared with infection with its PB1-F2-deficient counterpart. Accordingly, a much more pronounced infiltration of lungs with immune cells was detected in mice infected with the PB1-F2 wt virus. In summary, we demonstrate that the PB1-F2 protein of IAVs exhibits a type I IFN-antagonistic function by interfering with the RIG-I/MAVS complex, which contributes to an enhanced pathogenicity in vivo.


Subject(s)
Interferon Type I/antagonists & inhibitors , Viral Proteins/metabolism , Animals , Cell Line , Dogs , Humans , Interferon Type I/genetics , Interferon Type I/metabolism , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...