Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 114
Filter
1.
J Virol ; 96(7): e0020022, 2022 04 13.
Article in English | MEDLINE | ID: mdl-35293768

ABSTRACT

Spleen tyrosine kinase (Syk) has recently come forth as a critical regulator of innate immune response. Previous studies identify Syk as a key kinase for STAT1 activation at the early stage of influenza A virus (IAV) infection that is involved in initial antiviral immunity. However, the involvement of Syk in host antiviral immunity during the late phase of IAV infection and its effect on pathogenesis of the virus remain unknown. Here, we found through time course studies that Syk restrained antiviral immune response at the late stage of IAV infection, thereby promoting viral replication. Depletion of Syk suppressed IAV replication in vitro, whereas ectopic expression of Syk facilitated viral replication. Moreover, Syk-deficient mice were employed, and we observed that knockout of Syk rendered mice more resistant to IAV infection, as evidenced by a lower degree of lung injury, slower body weight loss, and an increased survival rate of Syk knockout mice challenged with IAV. Furthermore, we revealed that Syk repressed the interferon response at the late stage of viral infection. Loss of Syk potentiated the expression of type I and III interferons in both Syk-depleted cells and mice. Mechanistically, Syk interacted with TBK1 and modulated its phosphorylation status, thereby impeding TBK1 activation and restraining innate immune signaling that governs interferon response. Together, these findings unveil a role of Syk in temporally regulating host antiviral immunity and advance our understanding of complicated mechanisms underlying regulation of innate immunity against viral invasion. IMPORTANCE Innate immunity must be tightly controlled to eliminate invading pathogens while avoiding autoimmune or inflammatory diseases. Syk is essential for STAT1 activation at the early stage of IAV infection, which is critical for initial antiviral responses. Surprisingly, here a time course study showed that Syk suppressed innate immunity during late phases of IAV infection and thereby promoted IAV replication. Syk deficiency enhanced the expression of type I and III interferons, inhibited IAV replication, and rendered mice more resistant to IAV infection. Syk impaired innate immune signaling through impeding TBK1 activation. These data reveal that Syk participates in the initiation of antiviral defense against IAV infection and simultaneously contributes to the restriction of innate immunity at the late stage of viral infection, suggesting that Syk serves a dual function in regulating antiviral responses. This finding provides new insights into complicated mechanisms underlying interaction between virus and host immune system.


Subject(s)
Immunity, Innate , Influenza A virus , Orthomyxoviridae Infections , Animals , Antiviral Agents/metabolism , Host Microbial Interactions/immunology , Humans , Interferons/metabolism , Mice , Orthomyxoviridae Infections/enzymology , Orthomyxoviridae Infections/immunology , Syk Kinase/genetics , Syk Kinase/immunology , Virus Replication
2.
Proc Natl Acad Sci U S A ; 119(10): e2110647119, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35238649

ABSTRACT

SignificanceAn immunosuppressant protein (MTX), which facilitates virus infection by inhibiting leukotriene A4 hydrolase (LTA4H) to produce the lipid chemoattractant leukotriene B4 (LTB4), was identified and characterized from the submandibular salivary glands of the bat Myotis pilosus. To the best of our knowledge, this is a report of an endogenous LTA4H inhibitor in animals. MTX was highly concentrated in the bat salivary glands, suggesting a mechanism for the generation of immunological privilege and immune tolerance and providing evidence of viral shedding through oral secretions. Moreover, given that the immunosuppressant MTX selectively inhibited the proinflammatory activity of LTA4H, without affecting its antiinflammatory activity, MTX might be a potential candidate for the development of antiinflammatory drugs by targeting the LTA4-LTA4H-LTB4 inflammatory axis.


Subject(s)
Enzyme Inhibitors/metabolism , Epoxide Hydrolases , Influenza A Virus, H1N1 Subtype/metabolism , Leukotriene A4/metabolism , Orthomyxoviridae Infections/enzymology , Salivary Glands , Salivary Proteins and Peptides/metabolism , Virus Diseases , Animals , Chiroptera , Epoxide Hydrolases/antagonists & inhibitors , Epoxide Hydrolases/metabolism , Mice , Salivary Glands/enzymology , Salivary Glands/virology
3.
Biomolecules ; 11(6)2021 05 29.
Article in English | MEDLINE | ID: mdl-34072389

ABSTRACT

Influenza virus (IV) infections are considered to cause severe diseases of the respiratory tract. Beyond mild symptoms, the infection can lead to respiratory distress syndrome and multiple organ failure. 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. Interestingly, the virus-supportive function of the cellular phosphatidylinositol 3-kinase (PI3K) suggests that this signaling module may be a potential target for antiviral intervention. In the sense of repurposing existing drugs for new indications, we used Pictilisib, a known PI3K inhibitor to investigate its effect on IV infection, in mono-cell-culture studies as well as in a human chip model. Our results indicate that Pictilisib is a potent inhibitor of IV propagation already at early stages of infection. In a murine model of IV pneumonia, the in vitro key findings were verified, showing reduced viral titers as well as inflammatory response in the lung after delivery of Pictilisib. Our data identified Pictilisib as a promising drug candidate for anti-IV therapies that warrant further studying. These results further led to the conclusion that the repurposing of previously approved substances represents a cost-effective and efficient way for development of novel antiviral strategies.


Subject(s)
Indazoles/pharmacology , Influenza A virus/metabolism , Lung , Orthomyxoviridae Infections , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Pneumonia, Viral , Sulfonamides/pharmacology , A549 Cells , Animals , Dogs , Humans , Lung/enzymology , Lung/virology , Madin Darby Canine Kidney Cells , Mice , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/enzymology , Orthomyxoviridae Infections/virology , Pneumonia, Viral/drug therapy , Pneumonia, Viral/enzymology , Pneumonia, Viral/virology
4.
Int J Mol Sci ; 22(5)2021 Feb 27.
Article in English | MEDLINE | ID: mdl-33673431

ABSTRACT

In this study, we have introduced newly synthesized substituted benzothiazole based berberine derivatives that have been analyzed for their in vitro and in silico biological properties. The activity towards various kinds of influenza virus strains by employing the cytopathic effect (CPE) and sulforhodamine B (SRB) assay. Several berberine-benzothiazole derivatives (BBDs), such as BBD1, BBD3, BBD4, BBD5, BBD7, and BBD11, demonstrated interesting anti-influenza virus activity on influenza A viruses (A/PR/8/34, A/Vic/3/75) and influenza B viral (B/Lee/40, and B/Maryland/1/59) strain, respectively. Furthermore, by testing neuraminidase activity (NA) with the neuraminidase assay kit, it was identified that BBD7 has potent neuraminidase activity. The molecular docking analysis further suggests that the BBD1-BBD14 compounds' antiviral activity may be because of interaction with residues of NA, and the same as in oseltamivir.


Subject(s)
Benzothiazoles/pharmacology , Berberine/pharmacology , Molecular Docking Simulation , Neuraminidase/antagonists & inhibitors , Orthomyxoviridae Infections/drug therapy , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Benzothiazoles/therapeutic use , Berberine/analogs & derivatives , Berberine/therapeutic use , Cell Line , Cytopathogenic Effect, Viral , Dogs , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Influenza A virus/drug effects , Influenza A virus/enzymology , Influenza B virus/drug effects , Influenza B virus/enzymology , Orthomyxoviridae Infections/enzymology , Viral Proteins/antagonists & inhibitors
5.
Oxid Med Cell Longev ; 2020: 8824934, 2020.
Article in English | MEDLINE | ID: mdl-33204399

ABSTRACT

Antigenic mismatch can cause influenza vaccines to be ineffective, and influenza viruses resistant to antiviral drugs are rising. Thus, development of antiviral agents against these viruses is an immediate need. Rhus verniciflua (RVS) has long been used in herbal medicine and as a nutritional supplement. The effect of RVS and its components on influenza virus has not, however, been reported. We found that RVS treatment significantly reduced viral replication when evaluated with green fluorescent protein- (GFP-) tagged virus (influenza A virus, A/PR/8/34-GFP) in Madin-Darby canine kidney (MDCK) cells. RVS showed significant inhibition of neuraminidase from A/PR/8/34. Subsequently, three fractions were prepared from an ethanolic crude extract of RVS. In vitro assays indicated that an ethyl acetate fraction (RVSE) was more potent than H2O and CHCl3 fractions. RVSE significantly suppressed influenza virus infection in MDCK cells via neuraminidase inhibition. Additionally, RVSE treatment inhibited expression of several virus proteins and decreased mortality of mice exposed to influenza A/PR/8/34 by 50% and reduced weight loss by 11.5%. Active components in RVSE were isolated, and 5-deoxyluteolin (5) and sulfuretin (7) demonstrate the highest neuraminidase inhibitory activity against influenza A virus. RVS, RVSE, and their constituents may be useful for the development of anti-influenza agents.


Subject(s)
Antiviral Agents/pharmacology , Enzyme Inhibitors/pharmacology , Influenza, Human/drug therapy , Neuraminidase/antagonists & inhibitors , Orthomyxoviridae Infections/drug therapy , Plant Extracts/pharmacology , Rhus/chemistry , A549 Cells , Acetates/chemistry , Animals , Dogs , Ethanol/chemistry , Female , Humans , Influenza A virus/drug effects , Influenza, Human/enzymology , Influenza, Human/virology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/enzymology , Orthomyxoviridae Infections/virology , Phytotherapy , Viral Proteins/metabolism , Virus Replication
6.
J Biol Chem ; 295(33): 11388-11407, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32303635

ABSTRACT

Cleavage of influenza virus hemagglutinin (HA) by host proteases is essential for virus infectivity. HA of most influenza A and B (IAV/IBV) viruses is cleaved at a monobasic motif by trypsin-like proteases. Previous studies have reported that transmembrane serine protease 2 (TMPRSS2) is essential for activation of H7N9 and H1N1pdm IAV in mice but that H3N2 IAV and IBV activation is independent of TMPRSS2 and carried out by as-yet-undetermined protease(s). Here, to identify additional H3 IAV- and IBV-activating proteases, we used RNA-Seq to investigate the protease repertoire of murine lower airway tissues, primary type II alveolar epithelial cells (AECIIs), and the mouse lung cell line MLE-15. Among 13 candidates identified, TMPRSS4, TMPRSS13, hepsin, and prostasin activated H3 and IBV HA in vitro IBV activation and replication was reduced in AECIIs from Tmprss2/Tmprss4-deficient mice compared with WT or Tmprss2-deficient mice, indicating that murine TMPRSS4 is involved in IBV activation. Multicycle replication of H3N2 IAV and IBV in AECIIs of Tmprss2/Tmprss4-deficient mice varied in sensitivity to protease inhibitors, indicating that different, but overlapping, sets of murine proteases facilitate H3 and IBV HA cleavages. Interestingly, human hepsin and prostasin orthologs did not activate H3, but they did activate IBV HA in vitro Our results indicate that TMPRSS4 is an IBV-activating protease in murine AECIIs and suggest that TMPRSS13, hepsin, and prostasin cleave H3 and IBV HA in mice. They further show that hepsin and prostasin orthologs might contribute to the differences observed in TMPRSS2-independent activation of H3 in murine and human airways.


Subject(s)
Influenza A Virus, H3N2 Subtype/physiology , Influenza B virus/physiology , Influenza, Human/enzymology , Orthomyxoviridae Infections/enzymology , Peptide Hydrolases/metabolism , Virus Activation , Animals , Cell Line , Dogs , Enzyme Activation/drug effects , Gene Expression Profiling , HEK293 Cells , Host-Pathogen Interactions/drug effects , Humans , Influenza A Virus, H3N2 Subtype/drug effects , Influenza B virus/drug effects , Influenza, Human/drug therapy , Influenza, Human/genetics , Influenza, Human/virology , Lung/enzymology , Lung/metabolism , Lung/virology , Madin Darby Canine Kidney Cells , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/virology , Peptide Hydrolases/genetics , Protease Inhibitors/pharmacology , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Virus Activation/drug effects
7.
J Virol ; 93(21)2019 11 01.
Article in English | MEDLINE | ID: mdl-31391268

ABSTRACT

Cleavage of influenza virus hemagglutinin (HA) by host cell proteases is essential for virus infectivity and spread. We previously demonstrated in vitro that the transmembrane protease TMPRSS2 cleaves influenza A virus (IAV) and influenza B virus (IBV) HA possessing a monobasic cleavage site. Subsequent studies revealed that TMPRSS2 is crucial for the activation and pathogenesis of H1N1pdm and H7N9 IAV in mice. In contrast, activation of H3N2 IAV and IBV was found to be independent of TMPRSS2 expression and supported by an as-yet-undetermined protease(s). Here, we investigated the role of TMPRSS2 in proteolytic activation of IAV and IBV in three human airway cell culture systems: primary human bronchial epithelial cells (HBEC), primary type II alveolar epithelial cells (AECII), and Calu-3 cells. Knockdown of TMPRSS2 expression was performed using a previously described antisense peptide-conjugated phosphorodiamidate morpholino oligomer, T-ex5, that interferes with splicing of TMPRSS2 pre-mRNA, resulting in the expression of enzymatically inactive TMPRSS2. T-ex5 treatment produced efficient knockdown of active TMPRSS2 in all three airway cell culture models and prevented proteolytic activation and multiplication of H7N9 IAV in Calu-3 cells and H1N1pdm, H7N9, and H3N2 IAV in HBEC and AECII. T-ex5 treatment also inhibited the activation and spread of IBV in AECII but did not affect IBV activation in HBEC and Calu-3 cells. This study identifies TMPRSS2 as the major HA-activating protease of IAV in human airway cells and IBV in type II pneumocytes and as a potential target for the development of novel drugs to treat influenza infections.IMPORTANCE Influenza A viruses (IAV) and influenza B viruses (IBV) cause significant morbidity and mortality during seasonal outbreaks. Cleavage of the viral surface glycoprotein hemagglutinin (HA) by host proteases is a prerequisite for membrane fusion and essential for virus infectivity. Inhibition of relevant proteases provides a promising therapeutic approach that may avoid the development of drug resistance. HA of most influenza viruses is cleaved at a monobasic cleavage site, and a number of proteases have been shown to cleave HA in vitro This study demonstrates that the transmembrane protease TMPRSS2 is the major HA-activating protease of IAV in primary human bronchial cells and of both IAV and IBV in primary human type II pneumocytes. It further reveals that human and murine airway cells can differ in their HA-cleaving protease repertoires. Our data will help drive the development of potent and selective protease inhibitors as novel drugs for influenza treatment.


Subject(s)
Influenza A virus/physiology , Influenza B virus/physiology , Influenza, Human/virology , Serine Endopeptidases/metabolism , Animals , Bronchi/cytology , Cells, Cultured , Epithelial Cells/virology , Gene Knockdown Techniques , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Host-Pathogen Interactions , Humans , Influenza, Human/enzymology , Influenza, Human/metabolism , Mice , Orthomyxoviridae Infections/enzymology , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology , Pulmonary Alveoli/cytology , Serine Endopeptidases/genetics , Up-Regulation , Virus Replication
8.
J Exp Med ; 216(2): 304-316, 2019 02 04.
Article in English | MEDLINE | ID: mdl-30683737

ABSTRACT

Broadly neutralizing antibodies (Abs) that bind the influenza virus hemagglutinin (HA) stem may enable universal influenza vaccination. Here, we show that anti-stem Abs sterically inhibit viral neuraminidase (NA) activity against large substrates, with activity inversely proportional to the length of the fibrous NA stalk that supports the enzymatic domain. By modulating NA stalk length in recombinant IAVs, we show that anti-stem Abs inhibit virus release from infected cells by blocking NA, accounting for their in vitro neutralization activity. NA inhibition contributes to anti-stem Ab protection in influenza-infected mice, likely due at least in part to NA-mediated inhibition of FcγR-dependent activation of innate immune cells by Ab bound to virions. Food and Drug Administration-approved NA inhibitors enhance anti-stem-based Fc-dependent immune cell activation, raising the possibility of therapeutic synergy between NA inhibitors and anti-stem mAb treatment in humans.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Hemagglutinin Glycoproteins, Influenza Virus , Influenza A virus , Neuraminidase , Orthomyxoviridae Infections , Animals , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/pharmacology , Antibodies, Viral/immunology , Antibodies, Viral/pharmacology , Dogs , Female , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Immunity, Innate/drug effects , Influenza A virus/enzymology , Influenza A virus/immunology , Madin Darby Canine Kidney Cells , Mice , Neuraminidase/antagonists & inhibitors , Neuraminidase/immunology , Neuraminidase/metabolism , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/enzymology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/pathology , Protein Domains , Receptors, IgG/immunology , Receptors, IgG/metabolism
9.
DNA Cell Biol ; 37(10): 808-811, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30222366

ABSTRACT

The normal cellular prion protein, designated PrPC, is a membrane glycoprotein expressed most abundantly in brains, particularly by neurons, and to a lesser extent in non-neuronal tissues including lungs. Conformational conversion of PrPC into the amyloidogenic isoform is a key pathogenic event in prion diseases. We recently found that PrPC has a protective role against infection with influenza A viruses (IAVs) in mice by reducing reactive oxygen species in the lungs after infection with IAVs. The antioxidative activity of PrPC is probably attributable to its function to activate antioxidative enzyme Cu/Zn-superoxide dismutase, or SOD1, through regulating Cu content in lungs infected with IAVs. Oxidative stress could play a pivotal role in the pathogenesis of a wide range of viral infections. Here, we introduce our and others' studies on the role of PrPC in viral infections, and raise the attractive possibility that PrPC might be a novel target molecule for development of antioxidative therapeutics against not only IAV infection but also other viral infections.


Subject(s)
Gene Expression Regulation/immunology , Heat-Shock Proteins/immunology , Lung/immunology , Orthomyxoviridae Infections/genetics , PrPC Proteins/immunology , Superoxide Dismutase-1/immunology , Animals , Copper/immunology , Copper/metabolism , Enzyme Activation , Epithelial Cells/enzymology , Epithelial Cells/immunology , Epithelial Cells/virology , Heat-Shock Proteins/genetics , Influenza A virus/pathogenicity , Influenza A virus/physiology , Lung/enzymology , Lung/virology , Mice , Orthomyxoviridae Infections/enzymology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Oxidative Stress , PrPC Proteins/genetics , Protective Factors , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Superoxide Dismutase-1/genetics
10.
Biochem Biophys Res Commun ; 503(4): 2872-2877, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30100058

ABSTRACT

Protein glycosylation, attaching glycans covalently onto amino acid side chains of protein by various glycosyltransferase, is the most common post-translational modification. The UDP-GalNAc transferase 3 (GANLT3), encoded by Galnt3, transfers N-acetyl-d-galactosamine to hydroxyl groups of the side chains of Ser/Thr residues, initiating mucin type O-glycosylation of proteins. Most researches as yet focus on the involvement and abnormal expression of GALNT3 in various tumors. In this study, we found that GALNT3 was significantly decreased in the lungs after influenza A virus (IAV) infection in mice. Overexpression of GALNT3 in cell lines markedly inhibited IAV replication. Further experiments demonstrated that GALNT3 inhibited NF-κB signaling by preventing the translocation of phosphorylated P65 into nucleus. Therefore, our results reveal an important role of GALNT3 in regulating host responses during IAV infection, indicating the broad functions of the GALNT family, and the direct involvement of GALNTs during viral infections.


Subject(s)
Influenza A virus , N-Acetylgalactosaminyltransferases/physiology , Orthomyxoviridae Infections/enzymology , Active Transport, Cell Nucleus , Animals , Cell Line , Mice , NF-kappa B/metabolism , Orthomyxoviridae Infections/metabolism , Signal Transduction , Transcription Factor RelA/metabolism , Virus Replication , Polypeptide N-acetylgalactosaminyltransferase
11.
J Virol ; 92(13)2018 07 01.
Article in English | MEDLINE | ID: mdl-29643248

ABSTRACT

Influenza B virus (IBV) is one of the human respiratory viruses and one of the targets of seasonal vaccination. However, the bifurcation of two antigenically distinct lineages of IBVs makes it difficult to arrange proper medical countermeasures. Moreover, compared with pathogenicity-related molecular markers known for influenza A virus, little has been known for IBVs. To understand pathogenicity caused by IBVs, we investigated the molecular determinants of IBV pathogenicity in animal models. After serial lung-to-lung passages of Victoria lineage B/Brisbane/60/2008 (Vc_BR60) and Yamagata lineage B/Wisconsin/01/2010 (Ym_WI01) viruses in BALB/c mice, we identified the mouse-adapted Vc_BR60 (maVc_BR60) and Ym_WI01 (maYm_WI01) viruses, respectively. To find a molecular clue(s) to the increased pathogenicity of maVc_BR60 and maYm_WI01, we determined their genetic sequences. Several amino acid mutations were identified in the PB2, PB1, PA, BM2, and/or NS1 protein-coding regions, and one concurrent lysine (K)-to-arginine (R) mutation in PA residue 338 (PA K338R) was found in both maVc_BR60 and maYm_WI01 viruses. When analyzed using viruses rescued through reverse genetics, it was shown that PA K338R alone could increase the pathogenicity of both IBVs in mice and viral replication in the respiratory tracts of ferrets. In a subsequent minireplicon assay, the effect of PA K338R was highlighted by the enhancement of viral polymerase complex activity of both Vc_BR60 and Ym_WI01 viruses. These results suggest that the PA K338R mutation may be a molecular determinant of IBV pathogenicity via modulating the viral polymerase function of IBVs.IMPORTANCE To investigate molecular pathogenic determinants of IBVs, which are one of the targets of seasonal influenza vaccines, we adapted both Victoria and Yamagata lineage IBVs independently in mice. The recovered mouse-adapted viruses exhibited increased virulence, and of the various mutations identified from both mouse-adapted viruses, a concurrent amino acid mutation was found in the PA protein-coding region. When analyzed using viruses rescued through reverse genetics, the PA mutation alone appeared to contribute to viral pathogenicity in mice within the compatible genetic constellation between the IBV lineages and to the replication of IBVs in ferrets. Regarding the potential mechanism of increased viral pathogenicity, it was shown that the PA mutation could upregulate the viral polymerase complex activity of both IBV lineages. These results indicate that the PA mutation could be a newly defined molecular pathogenic determinant of IBVs that substantiates our understanding of the viral pathogenicity and public health risks of IBVs.


Subject(s)
DNA-Directed DNA Polymerase/metabolism , Influenza B virus/pathogenicity , Orthomyxoviridae Infections/virology , Viral Proteins/metabolism , Virus Replication , Animals , DNA-Directed DNA Polymerase/genetics , Female , Ferrets , Influenza B virus/enzymology , Male , Mice , Mice, Inbred BALB C , Mutation , Orthomyxoviridae Infections/enzymology , Viral Proteins/genetics
12.
Am J Physiol Lung Cell Mol Physiol ; 315(1): L52-L58, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29516781

ABSTRACT

Infection with seasonal influenza A virus (IAV) leads to lung inflammation and respiratory failure, a main cause of death in influenza-infected patients. Previous experiments in our laboratory indicate that Bruton's tyrosine kinase (Btk) plays a substantial role in regulating inflammation in the respiratory region during acute lung injury in mice; therefore, we sought to determine if blocking Btk activity has a protective effect in the lung during influenza-induced inflammation. The Btk inhibitor ibrutinib (also known as PCI-32765) was administered intranasally to mice starting 72 h after lethal infection with IAV. Our data indicate that treatment with the Btk inhibitor not only reduced weight loss and led to survival, but also had a dramatic effect on morphological changes to the lungs, in IAV-infected mice. Attenuation of lung inflammation indicative of acute lung injury, such as alveolar hemorrhage, interstitial thickening, and the presence of alveolar exudate, together with reduced levels of the inflammatory mediators TNFα, IL-1ß, IL-6, KC, and MCP-1, strongly suggests amelioration of the pathological immune response in the lungs to promote resolution of the infection. Finally, we observed that blocking Btk specifically in the alveolar compartment led to significant attenuation of neutrophil extracellular traps released into the lung in vivo and neutrophil extracellular trap formation in vitro. Our innovative findings suggest that Btk may be a new drug target for influenza-induced lung injury, and, in general, that immunomodulatory treatment may be key in treating lung dysfunction driven by excessive inflammation.


Subject(s)
Acute Lung Injury/enzymology , Agammaglobulinaemia Tyrosine Kinase/metabolism , Influenza A Virus, H1N1 Subtype/metabolism , Macrophages, Alveolar/enzymology , Orthomyxoviridae Infections/enzymology , Acute Lung Injury/drug therapy , Acute Lung Injury/pathology , Acute Lung Injury/virology , Adenine/analogs & derivatives , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Animals , Cytokines/metabolism , Extracellular Traps/metabolism , Macrophages, Alveolar/pathology , Mice , Orthomyxoviridae Infections/pathology , Piperidines , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology
13.
J Am Chem Soc ; 139(18): 6390-6395, 2017 05 10.
Article in English | MEDLINE | ID: mdl-28426220

ABSTRACT

A conceptually new type of enzymatic cleavage assay is reported that utilizes in situ supramolecular capture of the fluorescent product. A squaraine-derived substrate with large blocking groups at each end of its structure cannot be threaded by a tetralactam macrocycle until the blocking groups are removed by enzyme cleavage. A prototype design responds to viral neuraminidase, an indicator of influenza infection, and also measures susceptibility of the sample to neuraminidase inhibitor drugs. The substrate structure incorporates three key features: (a) a bis(4-amino-3-hydroxyphenyl)squaraine core with bright deep-red fluorescence and excellent photostability, (b) an N-methyl group at each end of the squaraine core that ensures fast macrocycle threading kinetics, and (c) sialic acid blocking groups that prevent macrocycle threading until they are removed by viral neuraminidase. The enzyme assay can be conducted in aqueous solution where dramatic colorimetric and fluorescence changes are easily observed by the naked eye. Alternatively, affinity capture beads coated with macrocycle can be used to immobilize the liberated squaraine and enable a range of heterogeneous analysis options. With further optimization, this new type of neuraminidase assay may be useful in a point of care clinic to rapidly diagnose influenza infection and also determine which of the approved antiviral inhibitor drugs is likely to be the most effective treatment for an individual patient. The assay design is generalizable and can be readily modified to monitor virtually any type of enzyme-catalyzed cleavage reaction.


Subject(s)
Cyclobutanes/chemistry , Cyclobutanes/metabolism , Enzyme Assays/methods , Fluorescence , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Neuraminidase/chemistry , Neuraminidase/metabolism , Orthomyxoviridae Infections/enzymology , Phenols/chemistry , Phenols/metabolism , Animals , Macromolecular Substances/chemistry , Macromolecular Substances/metabolism , Orthomyxoviridae Infections/diagnosis
14.
Viral Immunol ; 30(7): 542-544, 2017 09.
Article in English | MEDLINE | ID: mdl-28402179

ABSTRACT

While upregulation of 2,3-dioxygenase (IDO) accompanied by degradation of tryptophan along the kynurenine pathway have been reported to exert antimicrobial effects against a wide range of infectious agents, its role in the replication of influenza A virus remains uncertain. We performed experiments using influenza A/WSN/33 virus infection of mouse fibroblast cell-line (NIH-3T3) to study the effects of IDO on viral replication. Influenza infection resulted in prominent elevations of transcripts encoding IDO, interferon (IFN)-ß, and segment 8 of the virus in NIH-3T3 cells. Introduction of siRNA targeted against IDO followed by infection resulted in further increased levels of viral RNA without altering IFN-ß expression. Inhibition of IDO during the infection also resulted in reduction of virus-driven upregulation of 3-hydroxyanthranilate 3,4-dioxygenase (HAAO), but not kynurenine 3-monooxygenase (KMO), which are enzymes downstream in the kynurenine pathway. Thus, induction of IDO appears to contribute to limiting replication of the WSN/33 strain of influenza A virus in murine NIH-3T3 cells.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/pharmacology , Influenza A virus/drug effects , Orthomyxoviridae Infections/enzymology , Viral Proteins/genetics , Virus Replication/drug effects , 3-Hydroxyanthranilate 3,4-Dioxygenase/genetics , Animals , Antiviral Agents/pharmacology , Gene Expression Regulation, Viral , Gene Knockdown Techniques , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Influenza A virus/physiology , Interferon-beta/genetics , Mice , NIH 3T3 Cells , Orthomyxoviridae Infections/virology , Tryptophan/metabolism
15.
Nature ; 541(7635): 117-121, 2017 01 05.
Article in English | MEDLINE | ID: mdl-28002402

ABSTRACT

The heterotrimeric influenza polymerase (FluPol), comprising subunits PA, PB1 and PB2, binds to the conserved 5' and 3' termini (the 'promoter') of each of the eight single-stranded viral RNA (vRNA) genome segments and performs both transcription and replication of vRNA in the infected cell nucleus. To transcribe viral mRNAs, FluPol associates with cellular RNA polymerase II (Pol II), which enables it to take 5'-capped primers from nascent Pol II transcripts. Here we present a co-crystal structure of bat influenza A polymerase bound to a Pol II C-terminal domain (CTD) peptide mimic, which shows two distinct phosphoserine-5 (SeP5)-binding sites in the polymerase PA subunit, accommodating four CTD heptad repeats overall. Mutagenesis of the SeP5-contacting basic residues (PA K289, R454, K635 and R638) weakens CTD repeat binding in vitro without affecting the intrinsic cap-primed (transcription) or unprimed (replication) RNA synthesis activity of recombinant polymerase, whereas in cell-based minigenome assays the same mutations substantially reduce overall polymerase activity. Only recombinant viruses with a single mutation in one of the SeP5-binding sites can be rescued, but these viruses are severely attenuated and genetically unstable. Several previously described mutants that modulate virulence can be rationalized by our results, including a second site mutation (PA(C453R)) that enables the highly attenuated mutant virus (PA(R638A)) to revert to near wild-type infectivity. We conclude that direct binding of FluPol to the SeP5 Pol II CTD is fine-tuned to allow efficient viral transcription and propose that the CTD-binding site on FluPol could be targeted for antiviral drug development.


Subject(s)
Chiroptera/virology , Orthomyxoviridae/enzymology , RNA Polymerase II/chemistry , RNA Polymerase II/metabolism , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/metabolism , Amino Acid Sequence , Animals , Antiviral Agents/pharmacology , Binding Sites/drug effects , Binding Sites/genetics , Crystallography, X-Ray , Influenza A virus/enzymology , Influenza B virus/enzymology , Models, Molecular , Molecular Targeted Therapy , Mutation , Orthomyxoviridae/genetics , Orthomyxoviridae/growth & development , Orthomyxoviridae/pathogenicity , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/enzymology , Orthomyxoviridae Infections/virology , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Phosphoserine/metabolism , Protein Binding/drug effects , Protein Domains , Protein Subunits , RNA-Dependent RNA Polymerase/genetics , Virulence/genetics , Virus Replication
16.
Am J Physiol Lung Cell Mol Physiol ; 309(11): L1313-22, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26432867

ABSTRACT

Extracellular nucleotides and nucleosides are important signaling molecules in the lung. Nucleotide and nucleoside concentrations in alveolar lining fluid are controlled by a complex network of surface ectonucleotidases. Previously, we demonstrated that influenza A/WSN/33 (H1N1) virus resulted in increased levels of the nucleotide ATP and the nucleoside adenosine in bronchoalveolar lavage fluid (BALF) of wild-type (WT) C57BL/6 mice. Influenza-induced acute lung injury (ALI) was highly attenuated in A1-adenosine receptor-knockout mice. Because AMP hydrolysis by the ecto-5'-nucleotidase (CD73) plays a central role in and is rate-limiting for generation of adenosine in the normal lung, we hypothesized that ALI would be attenuated in C57BL/6-congenic CD73-knockout (CD73-KO) mice. Infection-induced hypoxemia, bradycardia, viral replication, and bronchoconstriction were moderately increased in CD73-KO mice relative to WT controls. However, postinfection weight loss, pulmonary edema, and parenchymal dysfunction were not altered. Treatment of WT mice with the CD73 inhibitor 5'-(α,ß-methylene) diphosphate (APCP) also had no effect on infection-induced pulmonary edema but modestly attenuated hypoxemia. BALF from CD73-KO and APCP-treated WT mice contained more IL-6 and CXCL-10/IFN-γ-induced protein 10, less CXCL-1/keratinocyte chemoattractant, and fewer neutrophils than BALF from untreated WT controls. BALF from APCP-treated WT mice also contained fewer alveolar macrophages and more transforming growth factor-ß than BALF from untreated WT mice. These results indicate that CD73 is not necessary for development of ALI following influenza A virus infection and suggest that tissue-nonspecific alkaline phosphatase may be responsible for increased adenosine generation in the infected lung. However, they do suggest that CD73 has a previously unrecognized immunomodulatory role in influenza.


Subject(s)
5'-Nucleotidase/metabolism , Acute Lung Injury/enzymology , Acute Lung Injury/immunology , Immunity, Innate , Influenza, Human/immunology , Orthomyxoviridae Infections/enzymology , Orthomyxoviridae Infections/immunology , 5'-Nucleotidase/genetics , Acute Lung Injury/complications , Acute Lung Injury/virology , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Alkaline Phosphatase/metabolism , Animals , Bronchoalveolar Lavage Fluid/cytology , Chemokines/metabolism , Compliance , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Expression Regulation/drug effects , Heart/drug effects , Heart/physiopathology , Humans , Immunity, Innate/drug effects , Influenza A Virus, H1N1 Subtype/immunology , Leukocyte Count , Lung/drug effects , Lung/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/cytology , Neutrophils/drug effects , Orthomyxoviridae Infections/complications , Orthomyxoviridae Infections/virology , Pulmonary Edema/etiology , Pulmonary Edema/pathology , Pulmonary Edema/physiopathology , Virus Replication/drug effects
17.
Drug Metab Dispos ; 43(8): 1169-80, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25994032

ABSTRACT

Members of the cytochrome P450 CYP2J subfamily are expressed in multiple tissues in mice and humans. These enzymes are active in the metabolism of fatty acids to generate bioactive compounds. Herein we report new methods and results for quantitative polymerase chain reaction (qPCR) analysis for the seven genes (Cyp2j5, Cyp2j6, Cyp2j8, Cyp2j9, Cyp2j11, Cyp2j12, and Cyp2j13) of the mouse Cyp2j subfamily. SYBR Green primer sets were developed and compared with commercially available TaqMan primer/probe assays for specificity toward mouse Cyp2j cDNA, and analysis of tissue distribution and regulation of Cyp2j genes. Each TaqMan primer/probe set and SYBR Green primer set were shown to be specific for their intended mouse Cyp2j cDNA. Tissue distribution of the mouse Cyp2j isoforms confirmed similar patterns of expression between the two qPCR methods. Cyp2j5 and Cyp2j13 were highly expressed in male kidneys, and Cyp2j11 was highly expressed in both male and female kidneys. Cyp2j6 was expressed in multiple tissues, with the highest expression in the small intestine and duodenum. Cyp2j8 was detected in various tissues, with highest expression found in the skin. Cyp2j9 was highly expressed in the brain, liver, and lung. Cyp2j12 was predominately expressed in the brain. We also determined the Cyp2j isoform expression in Cyp2j5 knockout mice to determine whether there was compensatory regulation of other Cyp2j isoforms, and we assessed Cyp2j isoform regulation during various inflammatory models, including influenza A, bacterial lipopolysaccharide, house dust mite allergen, and corn pollen. Both qPCR methods detected similar suppression of Cyp2j6 and Cyp2j9 during inflammation in the lung.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Animals , Cytochrome P-450 CYP2J2 , Cytochrome P-450 Enzyme System/biosynthesis , DNA Primers , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Female , Gene Expression Regulation, Enzymologic/genetics , Hypersensitivity/enzymology , Hypersensitivity/genetics , Kidney/enzymology , Lung/enzymology , Male , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/enzymology , Pollen/immunology , Polymerase Chain Reaction , Tissue Distribution , Zea mays/immunology
18.
PLoS One ; 10(5): e0124422, 2015.
Article in English | MEDLINE | ID: mdl-25938456

ABSTRACT

Genetically similar H5N1 viruses circulating in the avian reservoir exhibit different levels of pathogenicity in mice. In this study, we characterized two highly pathogenic H5N1 avian isolates--A/Hunan/316/2005 (HN05), which is highly pathogenic in mice, and A/Hubei/489/2004 (HB04), which is nonpathogenic. In mammalian cells, HN05 replicates more efficiently than HB04, although both viruses have similar growth kinetics in avian cells. We used reverse genetics to generate recombinant H5N1 strains containing genes from HN05 and HB04 and examined their virulence. HN05 genes encoding the polymerase complex determine pathogenicity and viral replication ability both in vitro and in vivo. The PB2 subunit plays an important role in enhancing viral replication, and the PB1 and PA subunits contribute mainly to pathogenicity in mice. These results can be used to elucidate host-range expansion and the molecular basis of the high virulence of H5N1 viruses in mammalian species.


Subject(s)
DNA-Directed RNA Polymerases/metabolism , Influenza A Virus, H5N1 Subtype/physiology , Influenza A Virus, H5N1 Subtype/pathogenicity , Orthomyxoviridae Infections/enzymology , Orthomyxoviridae Infections/virology , Protein Subunits/metabolism , Virus Replication , Amino Acids/metabolism , Animals , Cell Line , Chickens/virology , Female , Humans , Influenza A Virus, H5N1 Subtype/growth & development , Influenza A Virus, H5N1 Subtype/isolation & purification , Mice, Inbred C57BL , Reassortant Viruses/pathogenicity , Recombination, Genetic/genetics , Ribonucleoproteins/metabolism , Virulence
19.
Mol Immunol ; 63(2): 227-34, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25124144

ABSTRACT

Several viruses, including Orthomyxovirus, utilize cellular reactive oxygen species (ROS) for viral genomic replication and survival within host cells. However, the role of ROS in early events of viral entry and signal induction has not been elucidated. Here, we show that ISA virus (ISAV) induces ROS production very early during infection of CHSE-214 and SHK-1Ycells, and that production is sustained over the observed 24h post-infection. The mitogen-activated protein kinase (MAPK) family is responsible for important signaling pathways. In this study, we report that ISAV activates ERK and p38 in Salmo salar. In salmonid macrophages, while ERK was required for SOD, GLURED, p47phox expression, p38 regulated the ROS production by the NADPH oxidase complex activation. These results, together with the presence of several consensus target motifs for p38 MAPK in the promoter of the S. salar p47phox gene, suggest that p38 MAPK regulates p47phox gene expression in fish through the activation of this key transcription factor.


Subject(s)
Isavirus/physiology , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , Salmo salar/immunology , Salmo salar/virology , p38 Mitogen-Activated Protein Kinases/metabolism , Amino Acid Sequence , Animals , Antioxidants/metabolism , Base Sequence , Kinetics , Molecular Sequence Data , NADPH Oxidases/chemistry , NADPH Oxidases/genetics , Orthomyxoviridae Infections/enzymology , Orthomyxoviridae Infections/immunology , Promoter Regions, Genetic/genetics , Virus Replication , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
20.
Nat Immunol ; 16(1): 67-74, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25419628

ABSTRACT

Immune responses are tightly regulated to ensure efficient pathogen clearance while avoiding tissue damage. Here we report that Setdb2 was the only protein lysine methyltransferase induced during infection with influenza virus. Setdb2 expression depended on signaling via type I interferons, and Setdb2 repressed expression of the gene encoding the neutrophil attractant CXCL1 and other genes that are targets of the transcription factor NF-κB. This coincided with occupancy by Setdb2 at the Cxcl1 promoter, which in the absence of Setdb2 displayed diminished trimethylation of histone H3 Lys9 (H3K9me3). Mice with a hypomorphic gene-trap construct of Setdb2 exhibited increased infiltration of neutrophils during sterile lung inflammation and were less sensitive to bacterial superinfection after infection with influenza virus. This suggested that a Setdb2-mediated regulatory crosstalk between the type I interferons and NF-κB pathways represents an important mechanism for virus-induced susceptibility to bacterial superinfection.


Subject(s)
Histone-Lysine N-Methyltransferase/immunology , NF-kappa B/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae/immunology , Pneumonia/immunology , Superinfection/immunology , Animals , Chemokine CXCL1/immunology , Disease Susceptibility , Female , Interferon Type I/immunology , Male , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Orthomyxoviridae Infections/enzymology , Orthomyxoviridae Infections/virology , Pneumonia/enzymology , Pneumonia/virology , RNA/chemistry , RNA/genetics , Real-Time Polymerase Chain Reaction , Specific Pathogen-Free Organisms , Superinfection/enzymology , Superinfection/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...