Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
J Virol ; 97(11): e0090623, 2023 Nov 30.
Article in English | MEDLINE | ID: mdl-37843369

ABSTRACT

IMPORTANCE: It is well known that influenza A viruses (IAV) initiate host cell infection by binding to sialic acid, a sugar molecule present at the ends of various sugar chains called glycoconjugates. These sugar chains can vary in chain length, structure, and composition. However, it remains unknown if IAV strains preferentially bind to sialic acid on specific glycoconjugate type(s) for host cell infection. Here, we utilized CRISPR gene editing to abolish sialic acid on different glycoconjugate types in human lung cells, and evaluated human versus avian IAV infections. Our studies show that both human and avian IAV strains can infect human lung cells by utilizing any of the three major sialic acid-containing glycoconjugate types, specifically N-glycans, O-glycans, and glycolipids. Interestingly, simultaneous elimination of sialic acid on all three major glycoconjugate types in human lung cells dramatically decreased human IAV infection, yet had little effect on avian IAV infection. These studies show that avian IAV strains effectively utilize other less prevalent glycoconjugates for infection, whereas human IAV strains rely on a limited repertoire of glycoconjugate types. The remarkable ability of avian IAV strains to utilize diverse glycoconjugate types may allow for easy transmission into new host species.


Subject(s)
Influenza A virus , Influenza, Human , Lung , Receptors, Cell Surface , Animals , Humans , Carrier Proteins/metabolism , Glycoconjugates/metabolism , Influenza A virus/metabolism , Lung/virology , N-Acetylneuraminic Acid/metabolism , Polysaccharides/metabolism , Sugars/metabolism , Influenza in Birds/metabolism , Receptors, Cell Surface/metabolism , Receptors, Virus/metabolism
2.
PLoS Pathog ; 18(8): e1010756, 2022 08.
Article in English | MEDLINE | ID: mdl-35926068

ABSTRACT

Reporter viruses provide powerful tools for both basic and applied virology studies, however, the creation and exploitation of reporter influenza A viruses (IAVs) have been hindered by the limited tolerance of the segmented genome to exogenous modifications. Interestingly, our previous study has demonstrated the underlying mechanism that foreign insertions reduce the replication/transcription capacity of the modified segment, impairing the delicate balance among the multiple segments during IAV infection. In the present study, we developed a "balance compensation" strategy by incorporating additional compensatory mutations during initial construction of recombinant IAVs to expand the tolerance of IAV genome. As a proof of concept, promoter-enhancing mutations were introduced within the modified segment to rectify the segments imbalance of a reporter influenza PR8-NS-Gluc virus, while directed optimization of the recombinant IAV was successfully achieved. Further, we generated recombinant IAVs expressing a much larger firefly luciferase (Fluc) by coupling with a much stronger compensatory enhancement, and established robust Fluc-based live-imaging mouse models of IAV infection. Our strategy feasibly expands the tolerance for foreign gene insertions in the segmented IAV genome, which opens up better opportunities to develop more versatile reporter IAVs as well as live attenuated influenza virus-based vaccines for other important human pathogens.


Subject(s)
Influenza A virus , Influenza, Human , Animals , Humans , Influenza A virus/genetics , Influenza, Human/genetics , Mice , Virus Replication/genetics
3.
J Virol ; 96(3): e0183721, 2022 02 09.
Article in English | MEDLINE | ID: mdl-34851142

ABSTRACT

Research activities with infectious severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are currently permitted only under biosafety level 3 (BSL3) containment. Here, we report the development of a single-cycle infectious SARS-CoV-2 virus replicon particle (VRP) system with a luciferase and green fluorescent protein (GFP) dual reporter that can be safely handled in BSL2 laboratories to study SARS-CoV-2 biology. The spike (S) gene of SARS-CoV-2 encodes the envelope glycoprotein, which is essential for mediating infection of new host cells. Through deletion and replacement of this essential S gene with a luciferase and GFP dual reporter, we have generated a conditional SARS-CoV-2 mutant (ΔS-VRP) that produces infectious particles only in cells expressing a viral envelope glycoprotein of choice. Interestingly, we observed more efficient production of infectious particles in cells expressing vesicular stomatitis virus (VSV) glycoprotein G [ΔS-VRP(G)] than in cells expressing other viral glycoproteins, including S. We confirmed that infection from ΔS-VRP(G) is limited to a single round and can be neutralized by anti-VSV serum. In our studies with ΔS-VRP(G), we observed robust expression of both luciferase and GFP reporters in various human and murine cell types, demonstrating that a broad variety of cells can support intracellular replication of SARS-CoV-2. In addition, treatment of ΔS-VRP(G)-infected cells with either of the anti-CoV drugs remdesivir (nucleoside analog) and GC376 (CoV 3CL protease inhibitor) resulted in a robust decrease in both luciferase and GFP expression in a drug dose- and cell-type-dependent manner. Taken together, our findings show that we have developed a single-cycle infectious SARS-CoV-2 VRP system that serves as a versatile platform to study SARS-CoV-2 intracellular biology and to perform high-throughput screening of antiviral drugs under BSL2 containment. IMPORTANCE Due to the highly contagious nature of SARS-CoV-2 and the lack of immunity in the human population, research on SARS-CoV-2 has been restricted to biosafety level 3 laboratories. This has greatly limited participation of the broader scientific community in SARS-CoV-2 research and thus has hindered the development of vaccines and antiviral drugs. By deleting the essential spike gene in the viral genome, we have developed a conditional mutant of SARS-CoV-2 with luciferase and fluorescent reporters, which can be safely used under biosafety level 2 conditions. Our single-cycle infectious SARS-CoV-2 virus replicon system can serve as a versatile platform to study SARS-CoV-2 intracellular biology and to perform high-throughput screening of antiviral drugs under BSL2 containment.


Subject(s)
Genetic Engineering , Recombination, Genetic , Replicon , SARS-CoV-2/genetics , COVID-19/virology , Cell Culture Techniques , Cell Line , Containment of Biohazards/standards , Genes, Reporter , Humans , Laboratories/standards , Viral Proteins/genetics , Virus Replication
4.
PLoS Pathog ; 17(9): e1009321, 2021 09.
Article in English | MEDLINE | ID: mdl-34473799

ABSTRACT

Influenza A virus [IAV] genomes comprise eight negative strand RNAs packaged into virions in the form of viral ribonucleoproteins [vRNPs]. Rab11a plays a crucial role in the transport of vRNPs from the nucleus to the plasma membrane via microtubules, allowing assembly and virus production. Here, we identify a novel function for Rab11a in the inter-cellular transport of IAV vRNPs using tunneling nanotubes [TNTs]as molecular highways. TNTs are F-Actin rich tubules that link the cytoplasm of nearby cells. In IAV-infected cells, Rab11a was visualized together with vRNPs in these actin-rich intercellular connections. To better examine viral spread via TNTs, we devised an infection system in which conventional, virion-mediated, spread was not possible. Namely, we generated HA-deficient reporter viruses which are unable to produce progeny virions but whose genomes can be replicated and trafficked. In this system, vRNP transfer to neighboring cells was observed and this transfer was found to be dependent on both actin and Rab11a. Generation of infectious virus via TNT transfer was confirmed using donor cells infected with HA-deficient virus and recipient cells stably expressing HA protein. Mixing donor cells infected with genetically distinct IAVs furthermore revealed the potential for Rab11a and TNTs to serve as a conduit for genome mixing and reassortment in IAV infections. These data therefore reveal a novel role for Rab11a in the IAV life cycle, which could have significant implications for within-host spread, genome reassortment and immune evasion.


Subject(s)
Cell Communication , Influenza A virus/pathogenicity , Influenza, Human/virology , rab GTP-Binding Proteins/metabolism , A549 Cells , Animals , Dogs , Humans , Influenza A virus/genetics , Influenza, Human/genetics , Madin Darby Canine Kidney Cells , Nanotubes
5.
PLoS Pathog ; 17(5): e1009517, 2021 05.
Article in English | MEDLINE | ID: mdl-33970958

ABSTRACT

It is well documented that influenza A viruses selectively package 8 distinct viral ribonucleoprotein complexes (vRNPs) into each virion; however, the role of host factors in genome assembly is not completely understood. To evaluate the significance of cellular factors in genome assembly, we generated a reporter virus carrying a tetracysteine tag in the NP gene (NP-Tc virus) and assessed the dynamics of vRNP localization with cellular components by fluorescence microscopy. At early time points, vRNP complexes were preferentially exported to the MTOC; subsequently, vRNPs associated on vesicles positive for cellular factor Rab11a and formed distinct vRNP bundles that trafficked to the plasma membrane on microtubule networks. In Rab11a deficient cells, however, vRNP bundles were smaller in the cytoplasm with less co-localization between different vRNP segments. Furthermore, Rab11a deficiency increased the production of non-infectious particles with higher RNA copy number to PFU ratios, indicative of defects in specific genome assembly. These results indicate that Rab11a+ vesicles serve as hubs for the congregation of vRNP complexes and enable specific genome assembly through vRNP:vRNP interactions, revealing the importance of Rab11a as a critical host factor for influenza A virus genome assembly.


Subject(s)
Genome, Viral , Influenza A virus/genetics , Influenza, Human/virology , Ribonucleoproteins/metabolism , Viral Proteins/metabolism , Virus Assembly , rab GTP-Binding Proteins/metabolism , A549 Cells , HEK293 Cells , Humans , Influenza A virus/isolation & purification , Influenza, Human/genetics , Ribonucleoproteins/genetics , Viral Proteins/genetics , Virus Replication , rab GTP-Binding Proteins/genetics
6.
PLoS Pathog ; 16(9): e1008842, 2020 09.
Article in English | MEDLINE | ID: mdl-32898178

ABSTRACT

Signaling through retinoic acid inducible gene I (RIG-I) like receptors (RLRs) is tightly regulated, with activation occurring upon sensing of viral nucleic acids, and suppression mediated by negative regulators. Under homeostatic conditions aberrant activation of melanoma differentiation-associated protein-5 (MDA5) is prevented through editing of endogenous dsRNA by RNA editing enzyme Adenosine Deaminase Acting on RNA (ADAR1). In addition, ADAR1 is postulated to play pro-viral and antiviral roles during viral infections that are dependent or independent of RNA editing activity. Here, we investigated the importance of ADAR1 isoforms in modulating influenza A virus (IAV) replication and revealed the opposing roles for ADAR1 isoforms, with the nuclear p110 isoform restricting versus the cytoplasmic p150 isoform promoting IAV replication. Importantly, we demonstrate that p150 is critical for preventing sustained RIG-I signaling, as p150 deficient cells showed increased IFN-ß expression and apoptosis during IAV infection, independent of RNA editing activity. Taken together, the p150 isoform of ADAR1 is important for preventing sustained RIG-I induced IFN-ß expression and apoptosis during viral infection.


Subject(s)
Adenosine Deaminase/metabolism , Apoptosis , DEAD Box Protein 58/metabolism , Influenza A virus/physiology , Influenza, Human/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction , Virus Replication , A549 Cells , Adenosine Deaminase/genetics , DEAD Box Protein 58/genetics , HEK293 Cells , Humans , Influenza, Human/genetics , Isoenzymes/genetics , Isoenzymes/metabolism , RNA-Binding Proteins/genetics , Receptors, Immunologic
7.
J Virol ; 94(9)2020 04 16.
Article in English | MEDLINE | ID: mdl-32075925

ABSTRACT

Seasonal influenza virus infections cause mild illness in healthy adults, as timely viral clearance is mediated by the functions of cytotoxic T cells. However, avian H5N1 influenza virus infections can result in prolonged and fatal illness across all age groups, which has been attributed to the overt and uncontrolled activation of host immune responses. Here, we investigate how excessive innate immune responses to H5N1 impair subsequent adaptive T cell responses in the lungs. Using recombinant H1N1 and H5N1 strains sharing 6 internal genes, we demonstrate that H5N1 (2:6) infection in mice causes higher stimulation and increased migration of lung dendritic cells to the draining lymph nodes, resulting in greater numbers of virus-specific T cells in the lungs. Despite robust T cell responses in the lungs, H5N1 (2:6)-infected mice showed inefficient and delayed viral clearance compared with H1N1-infected mice. In addition, we observed higher levels of inhibitory signals, including increased PD-1 and interleukin-10 (IL-10) expression by cytotoxic T cells in H5N1 (2:6)-infected mice, suggesting that delayed viral clearance of H5N1 (2:6) was due to the suppression of T cell functions in vivo Importantly, H5N1 (2:6)-infected mice displayed decreased numbers of tissue-resident memory T cells compared with H1N1-infected mice; however, despite the decreased number of tissue-resident memory T cells, H5N1 (2:6) was protected against a heterologous challenge from H3N2 virus (X31). Taken together, our study provides mechanistic insight for the prolonged viral replication and protracted illness observed in H5N1-infected patients.IMPORTANCE Influenza viruses cause upper respiratory tract infections in humans. In healthy adults, seasonal influenza virus infections result in mild disease. Occasionally, influenza viruses endemic in domestic birds can cause severe and fatal disease even in healthy individuals. In avian influenza virus-infected patients, the host immune system is activated in an uncontrolled manner and is unable to control infection in a timely fashion. In this study, we investigated why the immune system fails to effectively control a modified form of avian influenza virus. Our studies show that T cell functions important for clearing virally infected cells are impaired by higher negative regulatory signals during modified avian influenza virus infection. In addition, memory T cell numbers were decreased in modified avian influenza virus-infected mice. Our studies provide a possible mechanism for the severe and prolonged disease associated with avian influenza virus infections in humans.


Subject(s)
Influenza A Virus, H5N1 Subtype/genetics , Influenza, Human/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Birds , Humans , Immunity, Innate/immunology , Immunologic Memory/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H5N1 Subtype/metabolism , Influenza A virus/physiology , Influenza, Human/metabolism , Lung/virology , Mice , Orthomyxoviridae Infections/virology , Virus Replication/genetics
8.
Nature ; 505(7485): 691-5, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24284630

ABSTRACT

The type I interferon (IFN) response protects cells from viral infection by inducing hundreds of interferon-stimulated genes (ISGs), some of which encode direct antiviral effectors. Recent screening studies have begun to catalogue ISGs with antiviral activity against several RNA and DNA viruses. However, antiviral ISG specificity across multiple distinct classes of viruses remains largely unexplored. Here we used an ectopic expression assay to screen a library of more than 350 human ISGs for effects on 14 viruses representing 7 families and 11 genera. We show that 47 genes inhibit one or more viruses, and 25 genes enhance virus infectivity. Comparative analysis reveals that the screened ISGs target positive-sense single-stranded RNA viruses more effectively than negative-sense single-stranded RNA viruses. Gene clustering highlights the cytosolic DNA sensor cyclic GMP-AMP synthase (cGAS, also known as MB21D1) as a gene whose expression also broadly inhibits several RNA viruses. In vitro, lentiviral delivery of enzymatically active cGAS triggers a STING-dependent, IRF3-mediated antiviral program that functions independently of canonical IFN/STAT1 signalling. In vivo, genetic ablation of murine cGAS reveals its requirement in the antiviral response to two DNA viruses, and an unappreciated contribution to the innate control of an RNA virus. These studies uncover new paradigms for the preferential specificity of IFN-mediated antiviral pathways spanning several virus families.


Subject(s)
Immunity, Innate/genetics , Immunity, Innate/immunology , Interferons/immunology , Nucleotidyltransferases/immunology , Nucleotidyltransferases/metabolism , Viruses/immunology , Animals , Cluster Analysis , DNA Viruses/immunology , DNA Viruses/pathogenicity , Flow Cytometry , Gene Library , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-3/metabolism , Interferons/metabolism , Membrane Proteins/metabolism , Mice , Mice, Knockout , Nucleotidyltransferases/deficiency , Nucleotidyltransferases/genetics , RNA Viruses/immunology , RNA Viruses/pathogenicity , STAT1 Transcription Factor/metabolism , Substrate Specificity , Viruses/classification , Viruses/pathogenicity
9.
Bull Math Biol ; 82(3): 35, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32125535

ABSTRACT

It is difficult to determine whether an immune response or target cell depletion by the infectious agent is most responsible for the control of acute primary infection. Both mechanisms can explain the basic dynamics of an acute infection-exponential growth of the pathogen followed by control and clearance-and can also be represented by many different differential equation models. Consequently, traditional model comparison techniques using time series data can be ambiguous or inconclusive. We propose that varying the inoculum dose and measuring the subsequent infectious load can rule out target cell depletion by the pathogen as the main control mechanism. Infectious load can be any measure that is proportional to the number of infected cells, such as viraemia. We show that a twofold or greater change in infectious load is unlikely when target cell depletion controls infection, regardless of the model details. Analyzing previously published data from mice infected with influenza, we find the proportion of lung epithelial cells infected was 21-fold greater (95% confidence interval 14-32) in the highest dose group than in the lowest. This provides evidence in favor of an alternative to target cell depletion, such as innate immunity, in controlling influenza infections in this experimental system. Data from other experimental animal models of acute primary infection have a similar pattern.


Subject(s)
Models, Immunological , Virus Diseases/immunology , Virus Diseases/virology , Adaptive Immunity , Animals , Disease Models, Animal , Host Microbial Interactions/immunology , Humans , Immunity, Innate , Mathematical Concepts , Mice , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Viral Load
10.
PLoS Pathog ; 13(3): e1006270, 2017 03.
Article in English | MEDLINE | ID: mdl-28282445

ABSTRACT

The cellular and molecular mechanisms underpinning the unusually high virulence of highly pathogenic avian influenza H5N1 viruses in mammalian species remains unknown. Here, we investigated if the cell tropism of H5N1 virus is a determinant of enhanced virulence in mammalian species. We engineered H5N1 viruses with restricted cell tropism through the exploitation of cell type-specific microRNA expression by incorporating microRNA target sites into the viral genome. Restriction of H5N1 replication in endothelial cells via miR-126 ameliorated disease symptoms, prevented systemic viral spread and limited mortality, despite showing similar levels of peak viral replication in the lungs as compared to control virus-infected mice. Similarly, restriction of H5N1 replication in endothelial cells resulted in ameliorated disease symptoms and decreased viral spread in ferrets. Our studies demonstrate that H5N1 infection of endothelial cells results in excessive production of cytokines and reduces endothelial barrier integrity in the lungs, which culminates in vascular leakage and viral pneumonia. Importantly, our studies suggest a need for a combinational therapy that targets viral components, suppresses host immune responses, and improves endothelial barrier integrity for the treatment of highly pathogenic H5N1 virus infections.


Subject(s)
Endothelial Cells/virology , Influenza A Virus, H5N1 Subtype/physiology , Influenza A Virus, H5N1 Subtype/pathogenicity , Orthomyxoviridae Infections/pathology , Viral Tropism/physiology , Animals , Blotting, Western , Disease Models, Animal , Female , Ferrets , Flow Cytometry , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , Virulence
11.
PLoS Pathog ; 12(7): e1005754, 2016 07.
Article in English | MEDLINE | ID: mdl-27438481

ABSTRACT

Retinoic acid inducible gene-I (RIG-I) is an innate RNA sensor that recognizes the influenza A virus (IAV) RNA genome and activates antiviral host responses. Here, we demonstrate that RIG-I signaling plays a crucial role in restricting IAV tropism and regulating host immune responses. Mice deficient in the RIG-I-MAVS pathway show defects in migratory dendritic cell (DC) activation, viral antigen presentation, and priming of CD8+ and CD4+ T cell responses during IAV infection. These defects result in decreased frequency of polyfunctional effector T cells and lowered protection against heterologous IAV challenge. In addition, our data show that RIG-I activation is essential for protecting epithelial cells and hematopoietic cells from IAV infection. These diverse effects of RIG-I signaling are likely imparted by the actions of type I interferon (IFN), as addition of exogenous type I IFN is sufficient to overcome the defects in antigen presentation by RIG-I deficient BMDC. Moreover, the in vivo T cell defects in RIG-I deficient mice can be overcome by the activation of MDA5 -MAVS via poly I:C treatment. Taken together, these findings demonstrate that RIG-I signaling through MAVS is critical for determining the quality of polyfunctional T cell responses against IAV and for providing protection against subsequent infection from heterologous or novel pandemic IAV strains.


Subject(s)
Membrane Proteins/immunology , Nerve Tissue Proteins/immunology , Orthomyxoviridae Infections/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Adaptor Proteins, Signal Transducing/immunology , Animals , Coculture Techniques , Dendritic Cells/immunology , Disease Models, Animal , Flow Cytometry , Influenza A virus/immunology , Interferon Type I/biosynthesis , Interferon Type I/immunology , Lymphocyte Activation/immunology , Membrane Proteins/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , Polymerase Chain Reaction , Receptors, Cell Surface , T-Lymphocytes/metabolism
12.
J Immunol ; 196(11): 4739-49, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27183583

ABSTRACT

Dietary lipids and their metabolites activate members of the peroxisome proliferative-activated receptor (PPAR) family of transcription factors and are critical for colonic health. The PPARα isoform plays a vital role in regulating inflammation in various disease settings, but its role in intestinal inflammation, commensal homeostasis, and mucosal immunity in the gut are unclear. In this study, we demonstrate that the PPARα pathway in innate immune cells orchestrates gut mucosal immunity and commensal homeostasis by regulating the expression of IL-22 and the antimicrobial peptides RegIIIß, RegIIIγ, and calprotectin. Additionally, the PPARα pathway is critical for imparting regulatory phenotype in intestinal macrophages. PPARα deficiency in mice led to commensal dysbiosis in the gut, resulting in a microbiota-dependent increase in the expression of inflammatory cytokines and enhanced susceptibility to intestinal inflammation. Pharmacological activation of this pathway decreased the expression of inflammatory cytokines and ameliorated colonic inflammation. Taken together, these findings identify a new important innate immune function for the PPARα signaling pathway in regulating intestinal inflammation, mucosal immunity, and commensal homeostasis. Thus, the manipulation of the PPARα pathway could provide novel opportunities for enhancing mucosal immunity and treating intestinal inflammation.


Subject(s)
Gastrointestinal Microbiome/immunology , Homeostasis , Inflammation/prevention & control , PPAR alpha/metabolism , Signal Transduction , Animals , Cells, Cultured , Homeodomain Proteins/immunology , Inflammation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR alpha/deficiency
13.
Nature ; 483(7390): 428-33, 2012 Mar 14.
Article in English | MEDLINE | ID: mdl-22419161

ABSTRACT

Viral infection is commonly associated with virus-driven hijacking of host proteins. Here we describe a novel mechanism by which influenza virus affects host cells through the interaction of influenza non-structural protein 1 (NS1) with the infected cell epigenome. We show that the NS1 protein of influenza A H3N2 subtype possesses a histone-like sequence (histone mimic) that is used by the virus to target the human PAF1 transcription elongation complex (hPAF1C). We demonstrate that binding of NS1 to hPAF1C depends on the NS1 histone mimic and results in suppression of hPAF1C-mediated transcriptional elongation. Furthermore, human PAF1 has a crucial role in the antiviral response. Loss of hPAF1C binding by NS1 attenuates influenza infection, whereas hPAF1C deficiency reduces antiviral gene expression and renders cells more susceptible to viruses. We propose that the histone mimic in NS1 enables the influenza virus to affect inducible gene expression selectively, thus contributing to suppression of the antiviral response.


Subject(s)
Gene Expression Regulation , Histones/metabolism , Influenza A Virus, H3N2 Subtype/metabolism , Influenza, Human/genetics , Influenza, Human/immunology , Molecular Mimicry , Viral Nonstructural Proteins/metabolism , Amino Acid Sequence , Gene Expression Regulation/immunology , Histones/chemistry , Humans , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/pathogenicity , Influenza, Human/pathology , Influenza, Human/virology , Molecular Sequence Data , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Protein Binding , Transcription Factors , Transcription, Genetic/immunology , Viral Nonstructural Proteins/chemistry
14.
J Immunol ; 194(7): 3295-304, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25710911

ABSTRACT

Breakdown in immunological tolerance to self-Ags or uncontrolled inflammation results in autoimmune disorders. Dendritic cells (DCs) play an important role in regulating the balance between inflammatory and regulatory responses in the periphery. However, factors in the tissue microenvironment and the signaling networks critical for programming DCs to control chronic inflammation and promote tolerance are unknown. In this study, we show that wnt ligand-mediated activation of ß-catenin signaling in DCs is critical for promoting tolerance and limiting neuroinflammation. DC-specific deletion of key upstream (lipoprotein receptor-related protein [LRP]5/6) or downstream (ß-catenin) mediators of canonical wnt signaling in mice exacerbated experimental autoimmune encephalomyelitis pathology. Mechanistically, loss of LRP5/6-ß-catenin-mediated signaling in DCs led to an increased Th1/Th17 cell differentiation but reduced regulatory T cell response. This was due to increased production of proinflammatory cytokines and decreased production of anti-inflammatory cytokines such as IL-10 and IL-27 by DCs lacking LRP5/6-ß-catenin signaling. Consistent with these findings, pharmacological activation of canonical wnt/ß-catenin signaling delayed experimental autoimmune encephalomyelitis onset and diminished CNS pathology. Thus, the activation of canonical wnt signaling in DCs limits effector T cell responses and represents a potential therapeutic approach to control autoimmune neuroinflammation.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Th1 Cells/immunology , Th17 Cells/immunology , Transforming Growth Factor beta/metabolism , Wnt Signaling Pathway , Animals , Cell Differentiation , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Deletion , Gene Knockout Techniques , Inflammation Mediators/metabolism , Interleukin-10/metabolism , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Male , Mice , Mice, Transgenic , Signal Transduction , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Th1 Cells/cytology , Th1 Cells/metabolism , Th17 Cells/cytology , Th17 Cells/metabolism , Wnt Signaling Pathway/drug effects , Wnt3A Protein/metabolism , beta Catenin/metabolism
15.
J Virol ; 89(3): 1537-49, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25428869

ABSTRACT

UNLABELLED: In the extracellular environment, cell-free virions seek out naive host cells over long distances and between organisms. This is the primary mechanism of spread for most viruses. Here we provide evidence for an alternative pathway previously undescribed for orthomyxoviruses, whereby the spread of influenza A virus (IAV) infectious cores to neighboring cells can occur within intercellular connections. The formation of these connections requires actin dynamics and is enhanced by viral infection. Connected cells have contiguous membranes, and the core infectious viral machinery (RNP and polymerase) was present inside the intercellular connections. A live-cell movie of green fluorescent protein (GFP)-tagged NS1 of IAV shows viral protein moving from one cell to another through an intercellular connection. The movement of tagged protein was saltatory but overall traveled only in one direction. Infectious virus cores can move from one cell to another without budding and release of cell-free virions, as evidenced by the finding that whereas a neuraminidase inhibitor alone did not inhibit the development of IAV microplaques, the presence of a neuraminidase inhibitor together with drugs inhibiting actin dynamics or the microtubule stabilizer paclitaxel (originally named taxol) precluded microplaque formation. Similar results were also observed with parainfluenza virus 5 (PIV5), a paramyxovirus, when neutralizing antibody was used to block spread by cell-free virions. Intercellular spread of infectious core particles was unaffected or enhanced in the presence of nocodazole for IAV but inhibited for PIV5. The intercellular connections have a core of filamentous actin, which hints toward transport of virus particles through the use of a myosin motor. IMPORTANCE: Here we describe a new method by which influenza A virus (IAV) spreads from cell to cell: IAV uses intracellular connections. The formation of these connections requires actin dynamics and is enhanced by viral infection and the absence of microtubules. Connected cells appeared to have contiguous membranes, and the core infectious viral machinery (RNP and polymerase) was present inside the intercellular connections. Infectious virus cores can move from one cell to another without budding and release of cell-free virions. Similar results were also observed with parainfluenza virus 5 (PIV5).


Subject(s)
Influenza A virus/physiology , Intercellular Junctions/physiology , Intercellular Junctions/virology , Virus Internalization , Actins/metabolism , Animals , Cell Line , Humans , Microscopy, Fluorescence , Microscopy, Video , Parainfluenza Virus 5/physiology
17.
J Virol ; 88(22): 13436-46, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25210184

ABSTRACT

UNLABELLED: Influenza pandemics occur when influenza A viruses (IAV) adapted to other host species enter humans and spread through the population. Pandemics are relatively rare due to host restriction of IAV: strains adapted to nonhuman species do not readily infect, replicate in, or transmit among humans. IAV can overcome host restriction through reassortment or adaptive evolution, and these are mechanisms by which pandemic strains arise in nature. To identify mutations that facilitate growth of avian IAV in humans, we have adapted influenza A/duck/Alberta/35/1976 (H1N1) (dk/AB/76) virus to a high-growth phenotype in differentiated human tracheo-bronchial epithelial (HTBE) cells. Following 10 serial passages of three independent lineages, the bulk populations showed similar growth in HTBE cells to that of a human seasonal virus. The coding changes present in six clonal isolates were determined. The majority of changes were located in the polymerase complex and nucleoprotein (NP), and all isolates carried mutations in the PB2 627 domain and regions of NP thought to interact with PB2. Using reverse genetics, the impact on growth and polymerase activity of individual and paired mutations in PB2 and NP was evaluated. The results indicate that coupling of the mammalian-adaptive mutation PB2 E627K or Q591K to selected mutations in NP further augments the growth of the corresponding viruses. In addition, minimal combinations of three (PB2 Q236H, E627K, and NP N309K) or two (PB2 Q591K and NP S50G) mutations were sufficient to recapitulate the efficient growth in HTBE cells of dk/AB/76 viruses isolated after 10 passages in this substrate. IMPORTANCE: Influenza A viruses adapted to birds do not typically grow well in humans. However, as has been seen recently with H5N1 and H7N9 subtype viruses, productive and virulent infection of humans with avian influenza viruses can occur. The ability of avian influenza viruses to adapt to new host species is a consequence of their high mutation rate that supports their zoonotic potential. Understanding of the adaptation of avian viruses to mammals strengthens public health efforts aimed at controlling influenza. In particular, it is critical to know how readily and through mutation to which functional components avian influenza viruses gain the ability to grow efficiently in humans. Our data show that as few as three mutations, in the PB2 and NP proteins, support robust growth of a low-pathogenic, H1N1 duck isolate in primary human respiratory cells.


Subject(s)
Epithelial Cells/virology , Influenza A Virus, H1N1 Subtype/growth & development , Influenza A Virus, H1N1 Subtype/genetics , Mutation , RNA-Binding Proteins/genetics , RNA-Dependent RNA Polymerase/genetics , Viral Core Proteins/genetics , Viral Proteins/genetics , Adaptation, Biological , Animals , Cell Line , DNA Mutational Analysis , Ducks , Humans , Influenza A Virus, H1N1 Subtype/physiology , Influenza in Birds/virology , Mutant Proteins/genetics , Mutant Proteins/metabolism , Nucleocapsid Proteins , RNA-Binding Proteins/metabolism , RNA-Dependent RNA Polymerase/metabolism , Recombination, Genetic , Reverse Genetics , Serial Passage , Viral Core Proteins/metabolism , Viral Proteins/metabolism
18.
Sci Adv ; 10(8): eadk9004, 2024 Feb 23.
Article in English | MEDLINE | ID: mdl-38394202

ABSTRACT

Seasonal or pandemic illness caused by influenza A viruses (IAVs) is a major public health concern due to the high morbidity and notable mortality. Although there are several approved drugs targeting different mechanisms, the emergence of drug resistance calls for new drug candidates that can be used alone or in combinations. Small-molecule IAV entry inhibitor, ING-1466, binds to hemagglutinin (HA) and blocks HA-mediated viral infection. Here, we show that this inhibitor demonstrates preventive and therapeutic effects in a mouse model of IAV with substantial improvement in the survival rate. When administered orally it elicits a therapeutic effect in mice, even after the well-established infection. Moreover, the combination of ING-1466 with oseltamivir phosphate or baloxavir marboxil enhances the therapeutic effect in a synergistic manner. Overall, ING-1466 has excellent oral bioavailability and in vitro absorption, distribution, metabolism, excretion, and toxicity profile, suggesting that it can be developed for monotherapy or combination therapy for the treatment of IAV infections.


Subject(s)
Dibenzothiepins , Influenza A virus , Morpholines , Pyridones , Thiepins , Triazines , Animals , Mice , Oseltamivir/pharmacology , Oseltamivir/therapeutic use , Antiviral Agents/therapeutic use , Oxazines/pharmacology , Oxazines/therapeutic use , Pyridines , Thiepins/pharmacology , Thiepins/therapeutic use
19.
Commun Biol ; 7(1): 486, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38649430

ABSTRACT

The ongoing evolution of SARS-CoV-2 to evade vaccines and therapeutics underlines the need for innovative therapies with high genetic barriers to resistance. Therefore, there is pronounced interest in identifying new pharmacological targets in the SARS-CoV-2 viral life cycle. The small molecule PAV-104, identified through a cell-free protein synthesis and assembly screen, was recently shown to target host protein assembly machinery in a manner specific to viral assembly. In this study, we investigate the capacity of PAV-104 to inhibit SARS-CoV-2 replication in human airway epithelial cells (AECs). We show that PAV-104 inhibits >99% of infection with diverse SARS-CoV-2 variants in immortalized AECs, and in primary human AECs cultured at the air-liquid interface (ALI) to represent the lung microenvironment in vivo. Our data demonstrate that PAV-104 inhibits SARS-CoV-2 production without affecting viral entry, mRNA transcription, or protein synthesis. PAV-104 interacts with SARS-CoV-2 nucleocapsid (N) and interferes with its oligomerization, blocking particle assembly. Transcriptomic analysis reveals that PAV-104 reverses SARS-CoV-2 induction of the type-I interferon response and the maturation of nucleoprotein signaling pathway known to support coronavirus replication. Our findings suggest that PAV-104 is a promising therapeutic candidate for COVID-19 with a mechanism of action that is distinct from existing clinical management approaches.


Subject(s)
Antiviral Agents , Epithelial Cells , SARS-CoV-2 , Virus Replication , Humans , SARS-CoV-2/drug effects , SARS-CoV-2/physiology , Virus Replication/drug effects , Epithelial Cells/virology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Antiviral Agents/pharmacology , Virus Assembly/drug effects , COVID-19/virology , COVID-19 Drug Treatment
20.
J Virol ; 86(16): 8359-66, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22674996

ABSTRACT

PB1-F2 is a small, 87- to 90-amino-acid-long protein encoded by the +1 alternate open reading frame of the PB1 gene of most influenza A virus strains. It has been shown to contribute to viral pathogenicity in a host- and strain-dependent manner, and we have previously discovered that a serine at position 66 (66S) in the PB1-F2 protein increases virulence of the 1918 and H5N1 pandemic viruses. Recently, we have shown that PB1-F2 inhibits the induction of type I interferon (IFN) at the level of the MAVS adaptor protein. However, the molecular mechanism for the IFN antagonist function of PB1-F2 has remained unclear. In the present study, we demonstrated that the C-terminal portion of the PB1-F2 protein binds to MAVS in a region that contains the transmembrane domain. Strikingly, PB1-F2 66S was observed to bind to MAVS more efficiently than PB1-F2 66N. We also tested the effect of PB1-F2 on the IFN antagonist functions of the polymerase proteins PB1, PB2, and PA and observed enhanced IFN inhibition by the PB1 and PB2 proteins in combination with PB1-F2 but not by the PA protein. Using a flow cytometry-based assay, we demonstrate that the PB1-F2 protein inhibits MAVS-mediated IFN synthesis by decreasing the mitochondrial membrane potential (MMP). Interestingly, PB1-F2 66S affected the MMP more efficiently than wild-type PB1-F2. In summary, the results of our study identify the molecular mechanism by which the influenza virus PB1-F2 N66S protein increases virulence.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Host-Pathogen Interactions , Immune Evasion , Influenza A Virus, H1N1 Subtype/pathogenicity , Interferon Type I/antagonists & inhibitors , Membrane Potential, Mitochondrial , Viral Proteins/metabolism , Cell Line , Humans , Protein Binding , Protein Interaction Mapping
SELECTION OF CITATIONS
SEARCH DETAIL