Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
J Virol ; 97(10): e0127123, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37819131

ABSTRACT

IMPORTANCE: The respiratory tract of humans is constantly exposed to potentially harmful agents, such as small particles or pathogens, and thus requires protective measures. Respiratory mucus that lines the airway epithelia plays a major role in the prevention of viral infections by limiting the mobility of viruses, allowing subsequent mucociliary clearance. Understanding the interplay between respiratory mucus and viruses can help elucidate host and virus characteristics that enable the initiation of infection. Here, we tested a panel of primary influenza A viruses of avian or human origin for their sensitivity to mucus derived from primary human airway cultures and found that differences between virus strains can be mapped to viral neuraminidase activity. We also show that binding of influenza A viruses to decoy receptors on highly glycosylated mucus components constitutes the major inhibitory function of mucus against influenza A viruses.


Subject(s)
Influenza A virus , Influenza, Human , Mucus , Neuraminidase , Animals , Humans , Birds , Influenza A virus/metabolism , Mucus/metabolism , Neuraminidase/metabolism , Respiratory System/metabolism
2.
mSphere ; 8(5): e0022623, 2023 10 24.
Article in English | MEDLINE | ID: mdl-37594288

ABSTRACT

Multiple respiratory viruses, including influenza A virus (IAV), can be transmitted via expiratory aerosol particles, and aerosol pH was recently identified as a major factor influencing airborne virus infectivity. Indoors, small exhaled aerosols undergo rapid acidification to pH ~4. IAV is known to be sensitive to mildly acidic conditions encountered within host endosomes; however, it is unknown whether the same mechanisms could mediate viral inactivation within the more acidic aerosol micro-environment. Here, we identified that transient exposure to pH 4 caused IAV inactivation by a two-stage process, with an initial sharp decline in infectious titers mainly attributed to premature attainment of the post-fusion conformation of viral protein haemagglutinin (HA). Protein changes were observed by hydrogen-deuterium exchange coupled to mass spectrometry (HDX-MS) as early as 10 s post-exposure to acidic conditions. Our HDX-MS data are in agreement with other more labor-intensive structural analysis techniques, such as X-ray crystallography, highlighting the ease and usefulness of whole-virus HDX-MS for multiplexed protein analyses, even within enveloped viruses such as IAV. Additionally, virion integrity was partially but irreversibly affected by acidic conditions, with a progressive unfolding of the internal matrix protein 1 (M1) that aligned with a more gradual decline in viral infectivity with time. In contrast, no acid-mediated changes to the genome or lipid envelope were detected. Improved understanding of respiratory virus fate within exhaled aerosols constitutes a global public health priority, and information gained here could aid the development of novel strategies to control the airborne persistence of seasonal and/or pandemic influenza in the future. IMPORTANCE It is well established that COVID-19, influenza, and many other respiratory diseases can be transmitted by the inhalation of aerosolized viruses. Many studies have shown that the survival time of these airborne viruses is limited, but it remains an open question as to what drives their infectivity loss. Here, we address this question for influenza A virus by investigating structural protein changes incurred by the virus under conditions relevant to respiratory aerosol particles. From prior work, we know that expelled aerosols can become highly acidic due to equilibration with indoor room air, and our results indicate that two viral proteins are affected by these acidic conditions at multiple sites, leading to virus inactivation. Our findings suggest that the development of air treatments to quicken the speed of aerosol acidification would be a major strategy to control infectious bioburdens in the air.


Subject(s)
Influenza A virus , Influenza, Human , Humans , Influenza A virus/physiology , Respiratory Aerosols and Droplets , Hydrogen-Ion Concentration
3.
Environ Sci Technol ; 57(1): 486-497, 2023 01 10.
Article in English | MEDLINE | ID: mdl-36537693

ABSTRACT

Respiratory viruses, including influenza virus and SARS-CoV-2, are transmitted by the airborne route. Air filtration and ventilation mechanically reduce the concentration of airborne viruses and are necessary tools for disease mitigation. However, they ignore the potential impact of the chemical environment surrounding aerosolized viruses, which determines the aerosol pH. Atmospheric aerosol gravitates toward acidic pH, and enveloped viruses are prone to inactivation at strong acidity levels. Yet, the acidity of expiratory aerosol particles and its effect on airborne virus persistence have not been examined. Here, we combine pH-dependent inactivation rates of influenza A virus (IAV) and SARS-CoV-2 with microphysical properties of respiratory fluids using a biophysical aerosol model. We find that particles exhaled into indoor air (with relative humidity ≥ 50%) become mildly acidic (pH ∼ 4), rapidly inactivating IAV within minutes, whereas SARS-CoV-2 requires days. If indoor air is enriched with nonhazardous levels of nitric acid, aerosol pH drops by up to 2 units, decreasing 99%-inactivation times for both viruses in small aerosol particles to below 30 s. Conversely, unintentional removal of volatile acids from indoor air may elevate pH and prolong airborne virus persistence. The overlooked role of aerosol acidity has profound implications for virus transmission and mitigation strategies.


Subject(s)
Air Pollution, Indoor , COVID-19 , Respiratory Aerosols and Droplets , Humans , Hydrogen-Ion Concentration , SARS-CoV-2 , Virus Inactivation , Disease Transmission, Infectious
5.
Microbiol Spectr ; 10(4): e0092122, 2022 08 31.
Article in English | MEDLINE | ID: mdl-35867415

ABSTRACT

Influenza A virus (IAV) coopts numerous host factors for efficient replication. The cysteine protease cathepsin W (CTSW) has been identified as one host factor required for IAV entry, specifically for the escape of IAVs from late endosomes. However, the substrate specificity of CTSW and the proviral mechanism are thus far unknown. Here, we show that intracellular but not secreted CTSW promotes viral entry. We reveal 79 potential direct and 31 potential indirect cellular target proteins of CTSW using the high-throughput proteomic approach terminal amine isotopic labeling of substrates (TAILS) and determine the cleavage motif shared by the substrates of CTSW. Subsequent integration with data from RNA interference (RNAi) screens for IAV host factors uncovers first insights into the proviral function of CTSW. Notably, CTSW-deficient mice display a 25% increase in survival and a delay in mortality compared to wild-type mice upon IAV infection. Altogether, these findings support the development of drugs targeting CTSW as novel host-directed antiviral therapies. IMPORTANCE Influenza viruses are respiratory pathogens and pose a constant threat to human health. Although antiviral drugs are available for influenza, the emergence and spread of drug-resistant viruses is cause for concern. Therefore, the development of new antivirals with lower chances of their target viruses acquiring resistance is urgently needed to reduce the high morbidity and mortality caused by influenza. Promising alternatives to drugs targeting viral proteins are those directed against host factors required for viral replication. The cysteine protease cathepsin W (CTSW) is an important host factor for IAV replication, and its proteolytic activity is required for fusion of viral and endosomal membranes. In this work, we identify a number of hitherto unknown CTSW substrates, providing new insights into virus-host interactions, and reveal that CTSW might also play a proviral role in an in vivo model. These results support the development of CTSW as a drug target for next-generation antivirals against influenza.


Subject(s)
Influenza A virus , Influenza, Human , Animals , Antiviral Agents/pharmacology , Cathepsin W , Host-Pathogen Interactions , Humans , Influenza, Human/drug therapy , Mice , Proteomics
6.
ACS Infect Dis ; 8(7): 1265-1279, 2022 07 08.
Article in English | MEDLINE | ID: mdl-35766385

ABSTRACT

There is a pressing need for host-directed therapeutics that elicit broad-spectrum antiviral activities to potentially address current and future viral pandemics. Apratoxin S4 (Apra S4) is a potent Sec61 inhibitor that prevents cotranslational translocation of secretory proteins into the endoplasmic reticulum (ER), leading to anticancer and antiangiogenic activity both in vitro and in vivo. Since Sec61 has been shown to be an essential host factor for viral proteostasis, we tested Apra S4 in cellular models of viral infection, including SARS-CoV-2, influenza A virus, and flaviviruses (Zika, West Nile, and Dengue virus). Apra S4 inhibited viral replication in a concentration-dependent manner and had high potency particularly against SARS-CoV-2 and influenza A virus, with subnanomolar activity in human cells. Characterization studies focused on SARS-CoV-2 revealed that Apra S4 impacted a post-entry stage of the viral life-cycle. Transmission electron microscopy revealed that Apra S4 blocked formation of stacked double-membrane vesicles, the sites of viral replication. Apra S4 reduced dsRNA formation and prevented viral protein production and trafficking of secretory proteins, especially the spike protein. Given the potent and broad-spectrum activity of Apra S4, further preclinical evaluation of Apra S4 and other Sec61 inhibitors as antivirals is warranted.


Subject(s)
COVID-19 Drug Treatment , Influenza A virus , Zika Virus Infection , Zika Virus , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Depsipeptides , Humans , Pandemics , SARS-CoV-2 , Zika Virus Infection/drug therapy
7.
STAR Protoc ; 3(1): 101089, 2022 03 18.
Article in English | MEDLINE | ID: mdl-35535160

ABSTRACT

Due to the low stoichiometry and highly transient nature of protein phosphorylation it is challenging to capture the dynamics and complexity of phosphorylation events on a systems level. Here, we present an optimized protocol to measure virus-induced phosphorylation events with high sensitivity using label free quantification-based phosphoproteomics. Specifically, we describe filter assisted protein digestion (FASP), enrichment of phosphopeptides, mass spectrometry, and subsequent bioinformatic analysis. For complete details on the use and execution of this protocol, please refer to Hunziker et al. (2022).


Subject(s)
Phosphopeptides , Proteomics , Mass Spectrometry/methods , Phosphopeptides/analysis , Phosphorylation , Proteomics/methods , Signal Transduction
8.
Cell Rep ; 38(12): 110549, 2022 03 22.
Article in English | MEDLINE | ID: mdl-35320721

ABSTRACT

Host interferons (IFNs) powerfully restrict viruses through the action of several hundred IFN-stimulated gene (ISG) products, many of which remain uncharacterized. Here, using RNAi screening, we identify several ISG restriction factors with previously undescribed contributions to IFN-mediated defense. Notably, RABGAP1L, a Tre2/Bub2/Cdc16 (TBC)-domain-containing protein involved in regulation of small membrane-bound GTPases, robustly potentiates IFN action against influenza A viruses (IAVs). Functional studies reveal that the catalytically active TBC domain of RABGAP1L promotes antiviral activity, and the RABGAP1L proximal interactome uncovered its association with proteins involved in endosomal sorting, maturation, and trafficking. In this regard, RABGAP1L overexpression is sufficient to disrupt endosomal function during IAV infection and restricts an early post-attachment, but pre-fusion, stage of IAV cell entry. Other RNA viruses that enter cells primarily via endocytosis are also impaired by RABGAP1L, while entry promiscuous SARS-CoV-2 is resistant. Our data highlight virus endocytosis as a key target for host defenses.


Subject(s)
Antiviral Agents , COVID-19 , Cell Line , Endocytosis , Humans , SARS-CoV-2
9.
Cell Rep ; 38(4): 110306, 2022 01 25.
Article in English | MEDLINE | ID: mdl-35081340

ABSTRACT

Binding of influenza virus to its receptor triggers signaling cascades that reprogram the cell for infection. To elucidate global virus-induced changes to the cellular signaling landscape, we conducted a quantitative phosphoproteomic screen with human and avian influenza viruses. Proteins with functions in cell adhesion and cytoskeletal remodeling are overrepresented among the hits, and the majority of factors undergoing phosphorylation changes have a significant impact on infection efficiency. We show that influenza virus induces the formation of filopodia through Cdc42 signaling, which results in enhanced virus endocytosis. The host cell counteracts this mechanism with cortactin, a regulator of actin polymerization that becomes phosphorylated in response to virus binding and translocates to the cell cortex, where it limits filopodia formation and virus uptake. Overall, our study reveals the signaling cascades induced by influenza virus receptor engagement and uncovers virus-induced filopodia formation that is counteracted by the host cell.


Subject(s)
Cortactin/metabolism , Host-Pathogen Interactions/physiology , Influenza A virus/pathogenicity , Orthomyxoviridae Infections/metabolism , Pseudopodia/metabolism , Virus Internalization , Animals , Cell Line , Humans , Phosphorylation , Proteomics
10.
J Gen Virol ; 102(7)2021 07.
Article in English | MEDLINE | ID: mdl-34319869

ABSTRACT

Rapid repurposing of existing drugs as new therapeutics for COVID-19 has been an important strategy in the management of disease severity during the ongoing SARS-CoV-2 pandemic. Here, we used high-throughput docking to screen 6000 compounds within the DrugBank library for their potential to bind and inhibit the SARS-CoV-2 3 CL main protease, a chymotrypsin-like enzyme that is essential for viral replication. For 19 candidate hits, parallel in vitro fluorescence-based protease-inhibition assays and Vero-CCL81 cell-based SARS-CoV-2 replication-inhibition assays were performed. One hit, diclazuril (an investigational anti-protozoal compound), was validated as a SARS-CoV-2 3 CL main protease inhibitor in vitro (IC50 value of 29 µM) and modestly inhibited SARS-CoV-2 replication in Vero-CCL81 cells. Another hit, lenvatinib (approved for use in humans as an anti-cancer treatment), could not be validated as a SARS-CoV-2 3 CL main protease inhibitor in vitro, but serendipitously exhibited a striking functional synergy with the approved nucleoside analogue remdesivir to inhibit SARS-CoV-2 replication, albeit this was specific to Vero-CCL81 cells. Lenvatinib is a broadly-acting host receptor tyrosine kinase (RTK) inhibitor, but the synergistic effect with remdesivir was not observed with other approved RTK inhibitors (such as pazopanib or sunitinib), suggesting that the mechanism-of-action is independent of host RTKs. Furthermore, time-of-addition studies revealed that lenvatinib/remdesivir synergy probably targets SARS-CoV-2 replication subsequent to host-cell entry. Our work shows that combining computational and cellular screening is a means to identify existing drugs with repurposing potential as antiviral compounds. Future studies could be aimed at understanding and optimizing the lenvatinib/remdesivir synergistic mechanism as a therapeutic option.


Subject(s)
Adenosine Monophosphate/analogs & derivatives , Alanine/analogs & derivatives , COVID-19 Drug Treatment , COVID-19/virology , Chymases/antagonists & inhibitors , Phenylurea Compounds/pharmacology , Quinolines/pharmacology , SARS-CoV-2/drug effects , Adenosine Monophosphate/pharmacology , Alanine/pharmacology , Animals , Antiviral Agents/pharmacology , COVID-19/enzymology , Cells, Cultured , Drug Evaluation, Preclinical , Humans , Molecular Docking Simulation , Protein Kinase Inhibitors/pharmacology , SARS-CoV-2/isolation & purification , SARS-CoV-2/pathogenicity
11.
Nat Nanotechnol ; 16(8): 918-925, 2021 08.
Article in English | MEDLINE | ID: mdl-34083772

ABSTRACT

Minimizing the spread of viruses in the environment is the first defence line when fighting outbreaks and pandemics, but the current COVID-19 pandemic demonstrates how difficult this is on a global scale, particularly in a sustainable and environmentally friendly way. Here we introduce and develop a sustainable and biodegradable antiviral filtration membrane composed of amyloid nanofibrils made from food-grade milk proteins and iron oxyhydroxide nanoparticles synthesized in situ from iron salts by simple pH tuning. Thus, all the membrane components are made of environmentally friendly, non-toxic and widely available materials. The membrane has outstanding efficacy against a broad range of viruses, which include enveloped, non-enveloped, airborne and waterborne viruses, such as SARS-CoV-2, H1N1 (the influenza A virus strain responsible for the swine flu pandemic in 2009) and enterovirus 71 (a non-enveloped virus resistant to harsh conditions, such as highly acidic pH), which highlights a possible role in fighting the current and future viral outbreaks and pandemics.


Subject(s)
Amyloid/chemistry , Antiviral Agents/pharmacology , Ferric Compounds/chemistry , Micropore Filters , Nanoparticles/chemistry , Amyloid/pharmacology , Antiviral Agents/chemistry , Ferric Compounds/pharmacology , Humans , Lactoglobulins/chemistry , Micropore Filters/virology , Virus Inactivation/drug effects , Viruses/classification , Viruses/drug effects , Viruses/isolation & purification , Water Purification
12.
Curr Opin Virol ; 48: 23-29, 2021 06.
Article in English | MEDLINE | ID: mdl-33838498

ABSTRACT

Influenza A viruses (IAV) are a major burden for human health and thus the topic of intense research efforts. The entry of IAV into host cells is of particular interest as early infection steps are the ideal target for intervention strategies. Here, we review recent key findings in the field of IAV entry. Specifically, we discuss the identification of novel entry receptors, the emerging role of the viral neuraminidase in entry, as well as recent progress from structural studies on the viral hemagglutinin during the fusion process and the viral matrix protein involved in virus uncoating. We also highlight remaining gaps in our understanding of IAV entry and point out important questions for ongoing research efforts.


Subject(s)
Host Microbial Interactions/physiology , Host-Pathogen Interactions/physiology , Influenza A virus/physiology , Influenza, Human/virology , Virus Internalization , Hemagglutinin Glycoproteins, Influenza Virus , Humans , Hydrogen-Ion Concentration , Neuraminidase , Viral Matrix Proteins/metabolism , Virus Attachment , Virus Replication
13.
J Exp Med ; 218(6)2021 06 07.
Article in English | MEDLINE | ID: mdl-33882122

ABSTRACT

The disease severity of influenza is highly variable in humans, and one genetic determinant behind these differences is the IFITM3 gene. As an effector of the interferon response, IFITM3 potently blocks cytosolic entry of influenza A virus (IAV). Here, we reveal a novel level of inhibition by IFITM3 in vivo: We show that incorporation of IFITM3 into IAV particles competes with incorporation of viral hemagglutinin (HA). Decreased virion HA levels did not reduce infectivity, suggesting that high HA density on IAV virions may be an antagonistic strategy used by the virus to prevent direct inhibition. However, we found that IFITM3-mediated reduction in HA content sensitizes IAV to antibody-mediated neutralization. Mathematical modeling predicted that this effect decreases and delays peak IAV titers, and we show that, indeed, IFITM3-mediated sensitization of IAV to antibody-mediated neutralization impacts infection outcome in an in vivo mouse model. Overall, our data describe a previously unappreciated interplay between the innate effector IFITM3 and the adaptive immune response.


Subject(s)
Antibodies, Neutralizing/immunology , Influenza A virus/immunology , Membrane Proteins/genetics , Membrane Proteins/immunology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , A549 Cells , Adaptive Immunity/immunology , Animals , Cell Line , Cell Line, Tumor , Dogs , Female , HEK293 Cells , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Influenza, Human/immunology , Influenza, Human/virology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred C57BL , Mice, Knockout , Proteolysis
14.
Trends Microbiol ; 29(11): 973-982, 2021 11.
Article in English | MEDLINE | ID: mdl-33757684

ABSTRACT

Pandemics are caused by novel pathogens to which pre-existing antibody immunity is lacking. Under these circumstances, the body must rely on innate interferon-mediated defenses to limit pathogen replication and allow development of critical humoral protection. Here, we highlight studies on disease susceptibility during H1N1 influenza and COVID-19 (SARS-CoV-2) pandemics. An emerging concept is that genetic and non-genetic deficiencies in interferon system components lead to uncontrolled virus replication and severe illness in a subset of people. Intriguingly, new findings suggest that individuals with autoantibodies neutralizing the antiviral function of interferon are at increased risk of severe COVID-19. We discuss key questions surrounding how such autoantibodies develop and function, as well as the general implications of diagnosing interferon deficiencies for personalized therapies.


Subject(s)
Disease Resistance , Host-Pathogen Interactions , Interferons/metabolism , Virus Diseases/etiology , Virus Diseases/metabolism , Alleles , Animals , Antibodies, Neutralizing/immunology , Autoantibodies/immunology , Autoimmunity , Disease Progression , Disease Resistance/immunology , Disease Susceptibility , Genetic Predisposition to Disease , Host-Pathogen Interactions/immunology , Humans , Interferons/antagonists & inhibitors , Interferons/immunology , Loss of Function Mutation , Polymorphism, Single Nucleotide , Severity of Illness Index , Virus Diseases/diagnosis , Virus Diseases/epidemiology
15.
PLoS Biol ; 19(3): e3001006, 2021 03.
Article in English | MEDLINE | ID: mdl-33760807

ABSTRACT

Since entering the human population, Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2; the causative agent of Coronavirus Disease 2019 [COVID-19]) has spread worldwide, causing >100 million infections and >2 million deaths. While large-scale sequencing efforts have identified numerous genetic variants in SARS-CoV-2 during its circulation, it remains largely unclear whether many of these changes impact adaptation, replication, or transmission of the virus. Here, we characterized 14 different low-passage replication-competent human SARS-CoV-2 isolates representing all major European clades observed during the first pandemic wave in early 2020. By integrating viral sequencing data from patient material, virus stocks, and passaging experiments, together with kinetic virus replication data from nonhuman Vero-CCL81 cells and primary differentiated human bronchial epithelial cells (BEpCs), we observed several SARS-CoV-2 features that associate with distinct phenotypes. Notably, naturally occurring variants in Orf3a (Q57H) and nsp2 (T85I) were associated with poor replication in Vero-CCL81 cells but not in BEpCs, while SARS-CoV-2 isolates expressing the Spike D614G variant generally exhibited enhanced replication abilities in BEpCs. Strikingly, low-passage Vero-derived stock preparation of 3 SARS-CoV-2 isolates selected for substitutions at positions 5/6 of E and were highly attenuated in BEpCs, revealing a key cell-specific function to this region. Rare isolate-specific deletions were also observed in the Spike furin cleavage site during Vero-CCL81 passage, but these were rapidly selected against in BEpCs, underscoring the importance of this site for SARS-CoV-2 replication in primary human cells. Overall, our study uncovers sequence features in SARS-CoV-2 variants that determine cell-specific replication and highlights the need to monitor SARS-CoV-2 stocks carefully when phenotyping newly emerging variants or potential variants of concern.


Subject(s)
SARS-CoV-2/physiology , Virus Replication/physiology , Amino Acid Substitution , Animals , Base Sequence , Bronchi/pathology , COVID-19/diagnosis , COVID-19/virology , Cells, Cultured , Chlorocebus aethiops , Epithelial Cells/pathology , Epithelial Cells/virology , Furin/metabolism , Host-Pathogen Interactions , Humans , SARS-CoV-2/isolation & purification , Vero Cells
16.
mBio ; 11(5)2020 09 10.
Article in English | MEDLINE | ID: mdl-32913009

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the cause of coronavirus disease 2019 (COVID-19), is a recently emerged respiratory coronavirus that has infected >23 million people worldwide with >800,000 deaths. Few COVID-19 therapeutics are available, and the basis for severe infections is poorly understood. Here, we investigated properties of type I (ß), II (γ), and III (λ1) interferons (IFNs), potent immune cytokines that are normally produced during infection and that upregulate IFN-stimulated gene (ISG) effectors to limit virus replication. IFNs are already in clinical trials to treat COVID-19. However, recent studies highlight the potential for IFNs to enhance expression of host angiotensin-converting enzyme 2 (ACE2), suggesting that IFN therapy or natural coinfections could exacerbate COVID-19 by upregulating this critical virus entry receptor. Using a cell line model, we found that beta interferon (IFN-ß) strongly upregulated expression of canonical antiviral ISGs, as well as ACE2 at the mRNA and cell surface protein levels. Strikingly, IFN-λ1 upregulated antiviral ISGs, but ACE2 mRNA was only marginally elevated and did not lead to detectably increased ACE2 protein at the cell surface. IFN-γ induced the weakest ISG response but clearly enhanced surface expression of ACE2. Importantly, all IFN types inhibited SARS-CoV-2 replication in a dose-dependent manner, and IFN-ß and IFN-λ1 exhibited potent antiviral activity in primary human bronchial epithelial cells. Our data imply that type-specific mechanisms or kinetics shape IFN-enhanced ACE2 transcript and cell surface levels but that the antiviral action of IFNs against SARS-CoV-2 counterbalances any proviral effects of ACE2 induction. These insights should aid in evaluating the benefits of specific IFNs, particularly IFN-λ, as repurposed therapeutics.IMPORTANCE Repurposing existing, clinically approved, antiviral drugs as COVID-19 therapeutics is a rapid way to help combat the SARS-CoV-2 pandemic. Interferons (IFNs) usually form part of the body's natural innate immune defenses against viruses, and they have been used with partial success to treat previous new viral threats, such as HIV, hepatitis C virus, and Ebola virus. Nevertheless, IFNs can have undesirable side effects, and recent reports indicate that IFNs upregulate the expression of host ACE2 (a critical entry receptor for SARS-CoV-2), raising the possibility that IFN treatments could exacerbate COVID-19. Here, we studied the antiviral- and ACE2-inducing properties of different IFN types in both a human lung cell line model and primary human bronchial epithelial cells. We observed differences between IFNs with respect to their induction of antiviral genes and abilities to enhance the cell surface expression of ACE2. Nevertheless, all the IFNs limited SARS-CoV-2 replication, suggesting that their antiviral actions can counterbalance increased ACE2.


Subject(s)
Antiviral Agents/pharmacology , Coronavirus Infections/drug therapy , Interferon Type I/pharmacology , Interferon-gamma/pharmacology , Interferons/pharmacology , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/drug therapy , Aged , Angiotensin-Converting Enzyme 2 , Animals , Betacoronavirus/immunology , COVID-19 , Cell Line , Chlorocebus aethiops , Female , Humans , Immunotherapy/methods , Interferon Type I/adverse effects , Interferon-gamma/adverse effects , Interferons/adverse effects , Pandemics , Peptidyl-Dipeptidase A/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Virus/metabolism , Respiratory Mucosa/cytology , Respiratory Mucosa/virology , SARS-CoV-2 , Up-Regulation/drug effects , Vero Cells , Virus Replication/drug effects , Interferon Lambda
17.
mSphere ; 5(4)2020 08 26.
Article in English | MEDLINE | ID: mdl-32848003

ABSTRACT

Natural adaptation of an antigenically novel avian influenza A virus (IAV) to be transmitted efficiently in humans has the potential to trigger a devastating pandemic. Understanding viral genetic determinants underlying adaptation is therefore critical for pandemic preparedness, as the knowledge gained enhances surveillance and eradication efforts, prepandemic vaccine design, and efficacy assessment of antivirals. However, this work has risks, as making gain-of-function substitutions in fully infectious IAVs may create a pathogen with pandemic potential. Thus, such experiments must be tightly controlled through physical and biological risk mitigation strategies. Here, we applied a previously described biological containment system for IAVs to a 2009 pandemic H1N1 strain and a highly pathogenic H5N1 strain. The system relies on deletion of the essential viral hemagglutinin (HA) gene, which is instead provided in trans, thereby restricting multicycle virus replication to genetically modified HA-complementing cells. In place of HA, a Renilla luciferase gene is inserted within the viral genome, and a live-cell luciferase substrate allows real-time quantitative monitoring of viral replication kinetics with a high dynamic range. We demonstrate that biologically contained IAV-like particles exhibit wild-type sensitivities to approved antivirals, including oseltamivir, zanamivir, and baloxavir. Furthermore, the inability of these IAV-like particles to genetically acquire the host-encoded HA allowed us to introduce gain-of-function substitutions in the H5 HA gene that promote mammalian transmissibility. Biologically contained "transmissible" H5N1 IAV-like particles exhibited wild-type sensitivities to approved antivirals, to the fusion inhibitor S20, and to neutralization by existing H5 monoclonal and polyclonal sera. This work represents a proof of principle that biologically contained IAV systems can be used to safely conduct selected gain-of-function experiments.IMPORTANCE Understanding how animal influenza viruses can adapt to spread in humans is critical to prepare for, and prevent, new pandemics. However, working safely with pathogens that have pandemic potential requires tight regulation and the use of high-level physical and biological risk mitigation strategies to stop accidental loss of containment. Here, we used a biological containment system for influenza viruses to study strains with pandemic potential. The system relies on deletion of the essential HA gene from the viral genome and its provision by a genetically modified cell line, to which virus propagation is therefore restricted. We show that this method permits safe handling of these pathogens, including gain-of-function variants, without the risk of generating fully infectious viruses. Furthermore, we demonstrate that this system can be used to assess virus sensitivity to both approved and experimental drugs, as well as the antigenic profile of viruses, important considerations for evaluating prepandemic vaccine and antiviral strategies.


Subject(s)
Adaptation, Physiological/genetics , Gain of Function Mutation , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/pathogenicity , Pandemics/prevention & control , Animals , Antiviral Agents/pharmacology , Cell Line , Dogs , Gene Deletion , Genes, Reporter , HEK293 Cells , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/drug effects , Influenza, Human/virology , Madin Darby Canine Kidney Cells , Proof of Concept Study , Virus Replication
18.
Nat Commun ; 11(1): 1288, 2020 03 09.
Article in English | MEDLINE | ID: mdl-32152316

ABSTRACT

Co-inhibitory pathways have a fundamental function in regulating T cell responses and control the balance between promoting efficient effector functions and restricting immune pathology. The TIGIT pathway has been implicated in promoting T cell dysfunction in chronic viral infection. Importantly, TIGIT signaling is functionally linked to IL-10 expression, which has an effect on both virus control and maintenance of tissue homeostasis. However, whether TIGIT has a function in viral persistence or limiting tissue pathology is unclear. Here we report that TIGIT modulation effectively alters the phenotype and cytokine profile of T cells during influenza and chronic LCMV infection, but does not affect virus control in vivo. Instead, TIGIT has an important effect in limiting immune pathology in peripheral organs by inducing IL-10. Our data therefore identify a function of TIGIT in limiting immune pathology that is independent of viral clearance.


Subject(s)
Receptors, Immunologic/metabolism , Virus Diseases/immunology , Virus Diseases/pathology , Acute Disease , Animals , CD8-Positive T-Lymphocytes/immunology , Chronic Disease , Cytokines/metabolism , Inflammation Mediators/metabolism , Interleukin-10/biosynthesis , Liver/pathology , Liver/virology , Lung/blood supply , Lung/pathology , Lymphocytic choriomeningitis virus/physiology , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/pathology , Spleen/immunology
19.
Sci Rep ; 10(1): 768, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31964943

ABSTRACT

Current treatment options for influenza virus infections in humans are limited and therefore the development of novel antivirals is of high priority. Inhibiting influenza virus attachment to host cells would provide an early and efficient block of the infection and thus, receptor analogs have been considered as options for antiviral treatment. Here, we describe the rapid and efficient synthesis of PAMAM dendrimers conjugated with either 3'-sialyllactose (3SL) or 6'-sialyllactose (6SL) and their potential to inhibit a diverse range of human and avian influenza virus strains. We show in a hemagglutination inhibition (HAI) assay that human IAV strains can be inhibited by (6SL)- and to a lesser extent also by (3SL)-conjugated PAMAM dendrimers. In contrast, avian strains could only be inhibited by (3SL)-conjugated dendrimers. Importantly, the differential sensitivities of human and avian IAV to the two types of sialyllactose-conjugated dendrimers could be confirmed in cell-based neutralization assays. Based on our findings, we suggest to further develop both, (3SL)- and (6SL)-conjugated PAMAM dendrimers, as influenza virus inhibitors.


Subject(s)
Antiviral Agents/chemical synthesis , Dendrimers/chemistry , Influenza A virus/drug effects , Lactose/analogs & derivatives , Oligosaccharides/chemical synthesis , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Birds , Chick Embryo , Dogs , Hemagglutination Inhibition Tests , Humans , Influenza A virus/immunology , Lactose/chemical synthesis , Lactose/chemistry , Lactose/pharmacology , Madin Darby Canine Kidney Cells , Oligosaccharides/chemistry , Oligosaccharides/pharmacology , Species Specificity
20.
J Virol ; 94(4)2020 01 31.
Article in English | MEDLINE | ID: mdl-31776280

ABSTRACT

The influenza A virus (IAV) envelope protein hemagglutinin binds α2,6- or α2,3-linked sialic acid as a host cell receptor. Bat IAV subtypes H17N10 and H18N11 form an exception to this rule and do not bind sialic acid but enter cells via major histocompatibility complex (MHC) class II. Here, we review current knowledge on IAV receptors with a focus on sialoglycan variants, protein coreceptors, and alternative receptors that impact IAV attachment and internalization beyond the well-described sialic acid binding.


Subject(s)
Influenza A virus/metabolism , Receptors, Virus/immunology , Receptors, Virus/metabolism , Animals , Chiroptera/virology , HEK293 Cells , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Humans , Influenza A virus/immunology , Influenza A virus/pathogenicity , N-Acetylneuraminic Acid/metabolism , Neuraminidase/metabolism , Virus Attachment , Virus Internalization
SELECTION OF CITATIONS
SEARCH DETAIL
...