Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 351
Filter
1.
Virol Sin ; 39(2): 205-217, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38346538

ABSTRACT

Swine are regarded as "intermediate hosts" or "mixing vessels" of influenza viruses, capable of generating strains with pandemic potential. From 2020 to 2021, we conducted surveillance on swine H1N2 influenza (swH1N2) viruses in swine farms located in Guangdong, Yunnan, and Guizhou provinces in southern China, as well as Henan and Shandong provinces in northern China. We systematically analyzed the evolution and pathogenicity of swH1N2 isolates, and characterized their replication and transmission abilities. The isolated viruses are quadruple reassortant H1N2 viruses containing genes from pdm/09 H1N1 (PB2, PB1, PA and NP genes), triple-reassortant swine (NS gene), Eurasian Avian-like (HA and M genes), and recent human H3N2 (NA gene) lineages. The NA, PB2, and NP of SW/188/20 and SW/198/20 show high gene similarities to A/Guangdong/Yue Fang277/2017 (H3N2). The HA gene of swH1N2 exhibits a high evolutionary rate. The five swH1N2 isolates replicate efficiently in human, canine, and swine cells, as well as in the turbinate, trachea, and lungs of mice. A/swine/Shandong/198/2020 strain efficiently replicates in the respiratory tract of pigs and effectively transmitted among them. Collectively, these current swH1N2 viruses possess zoonotic potential, highlighting the need for strengthened surveillance of swH1N2 viruses.


Subject(s)
Evolution, Molecular , Influenza A Virus, H1N2 Subtype , Orthomyxoviridae Infections , Reassortant Viruses , Swine Diseases , Animals , Swine , Reassortant Viruses/genetics , Reassortant Viruses/pathogenicity , Reassortant Viruses/isolation & purification , China/epidemiology , Orthomyxoviridae Infections/virology , Orthomyxoviridae Infections/transmission , Orthomyxoviridae Infections/veterinary , Swine Diseases/virology , Swine Diseases/transmission , Influenza A Virus, H1N2 Subtype/genetics , Influenza A Virus, H1N2 Subtype/pathogenicity , Influenza A Virus, H1N2 Subtype/isolation & purification , Humans , Mice , Dogs , Phylogeny , Virus Replication , Public Health , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza, Human/virology , Influenza, Human/transmission , Mice, Inbred BALB C , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/pathogenicity , Influenza A Virus, H3N2 Subtype/isolation & purification , Virulence , Female
2.
J Virol ; 96(11): e0051922, 2022 06 08.
Article in English | MEDLINE | ID: mdl-35546120

ABSTRACT

Virus strains in the live attenuated influenza vaccine (LAIV) for swine in the United States that was on the market until 2020 encode a truncated nonstructural protein 1 of 126 amino acids (NS1del126). Their attenuation is believed to be due to an impaired ability to counteract the type I interferon (IFN)-mediated antiviral host response. However, this mechanism has been documented only in vitro for H3N2 strain A/swine/Texas/4199-2/98 NS1del126 (lvTX98), and several cases of clinical respiratory disease in the field were associated with the LAIV strains. We therefore further examined the pathobiology, including type I IFN induction, of lvTX98 in pigs and compared it with IFN induction in pig kidney-15 (PK-15) cells. lvTX98 induced up to 3-fold-higher type I IFN titers than wild-type TX98 (wtTX98) after inoculation of PK-15 cells at a high multiplicity of infection, while virus replication kinetics were similar. Mean nasal lvTX98 excretion by intranasally inoculated pigs was on average 50 times lower than that for wtTX98 but still reached titers of up to 4.3 log10 50% tissue culture infective doses/mL. After intratracheal inoculation, mean lvTX98 titers in the lower respiratory tract were significantly reduced at 18 to 48 h postinoculation (hpi) but similar to wtTX98 titers at 72 hpi. lvTX98 caused milder clinical signs than wtTX98 but induced comparable levels of microscopic and macroscopic lung lesions, peak neutrophil infiltration, and peak type I IFN. Thus, lvTX98 was partly attenuated in pigs, but this could not be associated with higher type I IFN levels. IMPORTANCE Swine influenza A viruses (swIAVs) with a truncated NS1del126 protein were strongly attenuated in previous laboratory-based safety studies and therefore approved for use as LAIVs for swine in the United States. In the field, however, the LAIV strains were detected in diagnostic samples and could regain a wild-type NS1 via reassortment with endemic swIAVs. This suggests a significant degree of LAIV replication and urges further investigation of the level and mechanism of attenuation of these LAIV strains in vivo. Here, we show that H3N2 LAIV strain lvTX98 is only partly attenuated in pigs and is excreted at significant titers after intranasal vaccination. Attenuation and restricted replication of lvTX98 in vivo seemed to be associated with the loss of NS1 functions other than type I IFN antagonism. Our findings can help to explain the occurrence of clinical respiratory disease and reassortment events associated with NS1del126-based LAIV strains in the field.


Subject(s)
Influenza A Virus, H3N2 Subtype , Influenza Vaccines , Interferon Type I/immunology , Orthomyxoviridae Infections , Swine Diseases , Animals , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/pathogenicity , Orthomyxoviridae Infections/virology , Swine/virology , Swine Diseases/virology , Vaccines, Attenuated , Viral Nonstructural Proteins/genetics
3.
Int J Mol Sci ; 23(3)2022 Feb 07.
Article in English | MEDLINE | ID: mdl-35163797

ABSTRACT

H1N1 and H3N2 are the two most common subtypes of swine influenza virus (SIV). They not only endanger the pig industry, but are also a huge risk of zoonotic diseases. However, the molecular mechanism and regulatory network of pigs (hosts) against influenza virus infection are still unclear. In this study, porcine alveolar macrophage cell (3D4/21) models infected by swine influenza virus (H1N1 and H3N2) were constructed. The expression profiles of miRNAs, mRNAs, lncRNAs and circRNAs after H1N1 and H3N2 infected 3D4/21 cells were revealed in this study. Then, two ceRNAs (TCONS_00166432-miR10391-MAN2A1 and novel_circ_0004733-miR10391-MAN2A1) that regulated H1N1 and H3N2 infection in 3D4/21 cells were verified by the methods of bioinformatics analysis, gene overexpression, gene interference, real-time quantitative PCR (qPCR), dual luciferase activity assay and RNA immunoprecipitation (RIP). In addition, the important candidate molecules (miR-10391, TCONS_00166432, and novel_circ_0004733) were identified by qPCR and enzyme linked immunosorbent assay (ELISA). Finally, the regulatory effect and possible molecular mechanism of the target gene MAN2A1 were identified by the methods of gene interference, qPCR, Western blot and ELISA. The results of this study suggested that TCONS_00166432 and novel_circ_0004733 could competitively bind miR-10391 to target the MAN2A1 gene to regulate swine influenza virus infecting 3D4/21 cells. This study reported for the first time the ceRNA networks involved in the regulation of the swine influenza virus infecting 3D4/21 cells, which provided a new insight into the molecular mechanism of 3D4/21 cells against swine influenza virus infection.


Subject(s)
Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H3N2 Subtype/pathogenicity , Macrophages, Alveolar/virology , MicroRNAs/genetics , RNA, Circular/genetics , alpha-Mannosidase/genetics , Animals , Cell Line , Computational Biology , Dogs , Gene Expression Profiling , Gene Expression Regulation , Macrophages, Alveolar/chemistry , Macrophages, Alveolar/cytology , Madin Darby Canine Kidney Cells , Models, Biological , Swine
5.
Virology ; 566: 143-152, 2022 01.
Article in English | MEDLINE | ID: mdl-34929590

ABSTRACT

Annual repeat influenza vaccination raises concerns about protective efficacy against mismatched viruses. We investigated the impact of heterologous prime-boost vaccination on inducing cross protection by designing recombinant influenza viruses with chimeric hemagglutinin (HA) carrying M2 extracellular domains (M2e-HA). Heterologous prime-boost vaccination of C57BL/6 mice with M2e-HA chimeric virus more effectively induced M2e and HA stalk specific IgG antibodies correlating with cross protection than homologous prime-boost vaccination. Induction of M2e and HA stalk specific IgG antibodies was compromised in 1-year old mice, indicating significant aging effects on priming subdominant M2e and HA stalk IgG antibody responses. This study demonstrates that a heterologous prime-boost strategy with recombinant influenza virus expressing extra M2e epitopes provides more effective cross protection than homologous vaccination.


Subject(s)
Aging/immunology , Antibodies, Viral/biosynthesis , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Immunoglobulin G/biosynthesis , Influenza Vaccines/genetics , Influenza, Human/prevention & control , Orthomyxoviridae Infections/prevention & control , Age Factors , Aging/genetics , Animals , Antigens, Viral/genetics , Antigens, Viral/immunology , Cross Protection , Female , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Immunization, Secondary/methods , Immunogenicity, Vaccine , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H3N2 Subtype/pathogenicity , Influenza Vaccines/administration & dosage , Influenza Vaccines/biosynthesis , Influenza, Human/immunology , Influenza, Human/virology , Mice , Mice, Inbred C57BL , Models, Molecular , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Vaccination/methods , Vaccines, Synthetic , Viral Matrix Proteins/genetics , Viral Matrix Proteins/immunology
6.
mBio ; 12(6): e0151221, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34872354

ABSTRACT

Human influenza viruses evade host immune responses by accumulating mutations around the receptor-binding region of the hemagglutinin (HA) protein, which is composed of three key elements, the 130-loop, the 190-helix, and the 220-loop. Here, we characterized two human H3N2 influenza viruses with 12- and 16-amino acid deletions around the HA receptor-binding site that were isolated after antigenic selection of mutated H3N2 viruses. Structural modeling suggested that the 12-amino acid deletion eliminated the 190-helix. The 16-amino acid deletion comprises two stretches of 11- and 5-amino acid deletions. As the result of a frameshift, "novel" amino acids (not found in wild-type HA at these positions) are encoded between the deleted regions. Interestingly, structural modeling predicted that the novel sequence forms a structure resembling the 190-helix. However, compared to wild-type HA, the 16-amino acid deletion mutant lacks two antiparallel beta-sheets that connect the 190-helix and the 220-loop in wild-type HA. Nonetheless, both HA deletion mutants replicated in mammalian cells, and the 16-amino acid deletion mutant (with a remodeled 190-helix) also replicated in Syrian hamsters, albeit at low titers. Wild-type virus bound preferentially to α2,6-linked sialic acids, whereas both mutants gained affinity for α2,3-linked sialic acids. Moreover, the 12- and 16-amino acid deletions may affect the antigenic properties of the viruses. Thus, viruses with sizeable deletions around the HA receptor-binding site are viable but may display altered sialic acid preferences, altered antigenic properties, and attenuated replicative ability in cultured cells and virulence in Syrian hamsters. IMPORTANCE The hemagglutinin (HA) protein of influenza viruses serves as the receptor-binding protein and is the principal target of the host immune system. The antigenic epitopes in the receptor-binding region are known to tolerate mutations, but here, we show that even deletions of 12 or 16 amino acids in this region can be accommodated. In cultured cells, 12- and 16-amino acid deletion mutants were attenuated, and the 16-amino acid deletion mutant replicated in Syrian hamsters. Compared with wild-type virus, both mutants showed changes in their reactivity to some of the sera tested and changes in their binding affinity to sialic acids, which serve as influenza virus receptors. Collectively, our findings highlight the plasticity of HA.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza A Virus, H3N2 Subtype/genetics , Influenza, Human/virology , Receptors, Virus/metabolism , Amino Acid Motifs , Animals , Cricetinae , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Humans , Influenza A Virus, H3N2 Subtype/metabolism , Influenza A Virus, H3N2 Subtype/pathogenicity , Influenza, Human/genetics , Influenza, Human/metabolism , Mesocricetus , Protein Binding , Protein Conformation, alpha-Helical , Receptors, Virus/genetics , Sequence Deletion , Virulence , Virus Replication
7.
Int J Mol Sci ; 22(22)2021 Nov 09.
Article in English | MEDLINE | ID: mdl-34829979

ABSTRACT

Influenza is a respiratory virus that alone or in combination with secondary bacterial pathogens can contribute to the development of acute pneumonia in persons >65 years of age. Host innate immune antiviral signaling early in response to influenza is essential to inhibit early viral replication and guide the initiation of adaptive immune responses. Using young adult (3 months) and aged adult mice infected with mouse adapted H1N1 or H3N2, the results of our study illustrate dysregulated and/or diminished activation of key signaling pathways in aged lung contribute to increased lung inflammation and morbidity. Specifically, within the first seven days of infection, there were significant changes in genes associated with TLR and RIG-I signaling detected in aged murine lung in response to H1N1 or H3N2. Taken together, the results of our study expand our current understanding of age-associated changes in antiviral signaling in the lung.


Subject(s)
Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H3N2 Subtype/genetics , Influenza, Human/genetics , Pneumonia/genetics , A549 Cells , Animals , DEAD Box Protein 58/genetics , Disease Models, Animal , Gene Expression Regulation, Viral/genetics , Humans , Immunity, Innate/genetics , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H3N2 Subtype/pathogenicity , Influenza, Human/microbiology , Influenza, Human/virology , Lung/metabolism , Lung/microbiology , Lung/pathology , Mice , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/microbiology , Orthomyxoviridae Infections/virology , Pneumonia/microbiology , Pneumonia/virology , Toll-Like Receptors/genetics , Virus Replication/genetics
8.
Front Immunol ; 12: 732029, 2021.
Article in English | MEDLINE | ID: mdl-34804016

ABSTRACT

Transient receptor potential (TRP) channels, neuronal stimulations widely known to be associated with thermal responses, pain induction, and osmoregulation, have been shown in recent studies to have underlying mechanisms associated with inflammatory responses. The role of TRP channels on inflammatory milieu during bacterial infections has been widely demonstrated. It may vary among types of channels/pathogens, however, and it is not known how TRP channels function during pneumococcal infections. Streptococcus pneumoniae can cause severe infections such as pneumonia, bacteremia, and meningitis, with systemic inflammatory responses. This study examines the role of TRP channels (TRPV1 and TRPV4) for pneumococcal nasal colonization and subsequent development of invasive pneumococcal disease in a mouse model. Both TRPV1 and TRPV4 channels were shown to be related to regulation of the development of pneumococcal diseases. In particular, the influx of neutrophils (polymorphonuclear cells) in the nasal cavity and the bactericidal activity were significantly suppressed among TRPV4 knockout mice. This may lead to severe pneumococcal pneumonia, resulting in dissemination of the bacteria to various organs and causing high mortality during influenza virus coinfection. Regulating host immune responses by TRP channels could be a novel strategy against pathogenic microorganisms causing strong local/systemic inflammation.


Subject(s)
Nasal Mucosa/metabolism , Pneumococcal Infections/metabolism , Streptococcus pneumoniae/pathogenicity , TRPV Cation Channels/metabolism , Animals , Coinfection , Cytokines/metabolism , Disease Models, Animal , Host-Pathogen Interactions , Inflammation Mediators/metabolism , Influenza A Virus, H3N2 Subtype/pathogenicity , Mice, Inbred C57BL , Mice, Knockout , Nasal Mucosa/immunology , Nasal Mucosa/microbiology , Nasal Mucosa/virology , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/microbiology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/microbiology , Phagocytosis , Pneumococcal Infections/genetics , Pneumococcal Infections/immunology , Pneumococcal Infections/microbiology , Signal Transduction , Streptococcus pneumoniae/immunology , TRPV Cation Channels/genetics , Virulence
9.
J Gen Virol ; 102(10)2021 10.
Article in English | MEDLINE | ID: mdl-34661516

ABSTRACT

The polymerase acidic (PA) I38T substitution is a dominant marker of resistance to baloxavir. We evaluated the impact of I38T on the fitness of a contemporary influenza A(H3N2) virus. Influenza A/Switzerland/9715293/2013 (H3N2) wild-type (WT) virus and its I38T mutant were rescued by reverse genetics. Replication kinetics were compared using ST6GalI-MDCK and A549 cells and infectivity/contact transmissibility were evaluated in guinea pigs. Nasal wash (NW) viral titres were determined by TCID50 ml-1 in ST6GalI-MDCK cells. Competition experiments were performed and the evolution of viral population was assessed by droplet digital RT-PCR. I38T did not alter in vitro replication. I38T induced comparable titres vs the WT in guinea pigs NWs and the two viruses transmitted equally by direct contact. However, a 50 %:50 % mixture inoculum evolved to mean WT/I38T ratios of 71 %:29 % and 66.4 %:33.6 % on days 4 and 6 p.i., respectively. Contemporary influenza A(H3N2)-I38T PA variants may conserve a significant level of viral fitness.


Subject(s)
Influenza A Virus, H3N2 Subtype/physiology , Orthomyxoviridae Infections/virology , RNA-Dependent RNA Polymerase/genetics , Viral Proteins/genetics , A549 Cells , Amino Acid Substitution , Animals , Antiviral Agents/pharmacology , Dibenzothiepins/pharmacology , Dogs , Drug Resistance, Viral , Guinea Pigs , Humans , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/pathogenicity , Madin Darby Canine Kidney Cells , Morpholines/pharmacology , Nose/virology , Orthomyxoviridae Infections/transmission , Pyridones/pharmacology , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/metabolism , Reverse Genetics , Triazines/pharmacology , Viral Load , Viral Proteins/chemistry , Viral Proteins/metabolism , Virus Replication
10.
Front Immunol ; 12: 738490, 2021.
Article in English | MEDLINE | ID: mdl-34691044

ABSTRACT

Tumor progression locus 2 (Tpl2) is a serine-threonine kinase known to promote inflammation in response to various pathogen-associated molecular patterns (PAMPs), inflammatory cytokines and G-protein-coupled receptors and consequently aids in host resistance to pathogens. We have recently shown that Tpl2-/- mice succumb to infection with a low-pathogenicity strain of influenza (x31, H3N2) by an unknown mechanism. In this study, we sought to characterize the cytokine and immune cell profile of influenza-infected Tpl2-/- mice to gain insight into its host protective effects. Although Tpl2-/- mice display modestly impaired viral control, no virus was observed in the lungs of Tpl2-/- mice on the day of peak morbidity and mortality suggesting that morbidity is not due to virus cytopathic effects but rather to an overactive antiviral immune response. Indeed, increased levels of interferon-ß (IFN-ß), the IFN-inducible monocyte chemoattractant protein-1 (MCP-1, CCL2), Macrophage inflammatory protein 1 alpha (MIP-1α; CCL3), MIP-1ß (CCL4), RANTES (CCL5), IP-10 (CXCL10) and Interferon-γ (IFN-γ) was observed in the lungs of influenza-infected Tpl2-/- mice at 7 days post infection (dpi). Elevated cytokine and chemokines were accompanied by increased infiltration of the lungs with inflammatory monocytes and neutrophils. Additionally, we noted that increased IFN-ß correlated with increased CCL2, CXCL1 and nitric oxide synthase (NOS2) expression in the lungs, which has been associated with severe influenza infections. Bone marrow chimeras with Tpl2 ablation localized to radioresistant cells confirmed that Tpl2 functions, at least in part, within radioresistant cells to limit pro-inflammatory response to viral infection. Collectively, this study suggests that Tpl2 tempers inflammation during influenza infection by constraining the production of interferons and chemokines which are known to promote the recruitment of detrimental inflammatory monocytes and neutrophils.


Subject(s)
Cytokine Release Syndrome/metabolism , Cytokines/blood , Influenza A Virus, H3N2 Subtype/pathogenicity , Lung/metabolism , MAP Kinase Kinase Kinases/deficiency , Monocytes/metabolism , Neutrophils/metabolism , Orthomyxoviridae Infections/metabolism , Proto-Oncogene Proteins/deficiency , Animals , Biomarkers/blood , Cytokine Release Syndrome/genetics , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/virology , Cytokines/genetics , Disease Models, Animal , Female , Host-Pathogen Interactions , Influenza A Virus, H3N2 Subtype/immunology , Lung/immunology , Lung/virology , MAP Kinase Kinase Kinases/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Monocytes/virology , Neutrophil Infiltration , Neutrophils/immunology , Neutrophils/virology , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Proto-Oncogene Proteins/genetics , Suppressor of Cytokine Signaling 1 Protein/genetics , Suppressor of Cytokine Signaling 1 Protein/metabolism , Time Factors
11.
Bull Exp Biol Med ; 171(6): 736-740, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34705177

ABSTRACT

We studied the effect of tilorone on the dynamics of IFNα, IFNγ, and IL-1ß levels in the lung tissue and blood serum in relation to viral load in the lungs of BALB/c mice with pneumonia caused by influenza virus A/Aichi/2/68 (H3N2). Tilorone was administered per os in doses of 40, 150, and 540 µg per mouse 6, 30, and 78 h postinfection, which simulated the drug regimen used in the clinic for the treatment of influenza and acute respiratory viral infections in Russia and post-Soviet countries. Tilorone reduced viral load with the maximum amplitude (2-3 lg) after 1-2 administrations. The results of studying the dynamics of the cytokine levels in the infected animals in general support the previous hypothesis that, in repeated dosing, tilorone enhances the IFN response (compensates for its deficiency) at the early stages of acute respiratory viral infections and suppresses (damps) excessive production of IFN and proinflammatory cytokines at the later stages.


Subject(s)
Antiviral Agents/pharmacology , Influenza A Virus, H3N2 Subtype/drug effects , Interferon Inducers/pharmacology , Lung/drug effects , Orthomyxoviridae Infections/drug therapy , Tilorone/pharmacology , Animals , Drug Administration Schedule , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/immunology , Influenza A Virus, H3N2 Subtype/growth & development , Influenza A Virus, H3N2 Subtype/pathogenicity , Interferon-alpha/blood , Interferon-alpha/immunology , Interferon-gamma/blood , Interferon-gamma/immunology , Interleukin-1beta/blood , Interleukin-1beta/immunology , Lung/immunology , Lung/virology , Male , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/blood , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Viral Load/drug effects
12.
mBio ; 12(5): e0148421, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34517760

ABSTRACT

Membrane-associated RING-CH8 (MARCH8) impairs the cell surface expression of envelope glycoproteins from different viruses, reducing their incorporation into virions. Using stable cell lines with inducible MARCH8 expression, we show that MARCH8 did not alter susceptibility to influenza A virus (IAV) infection, but virions released from infected cells were markedly less infectious. Knockdown of endogenous MARCH8 confirmed its effect on IAV infectivity. The expression of MARCH8 impaired the infectivity of both H3N2 and H1N1 strains and was dependent on its E3 ligase activity. Although virions released in the presence of MARCH8 expressed smaller amounts of viral hemagglutinin (HA) and neuraminidase (NA) proteins, there was no impact on levels of the viral HA, NA, or matrix 2 (M2) proteins detected on the surface of infected cells. Moreover, mutation of lysine residues in the cytoplasmic tails of HA, NA, and/or M2, or in the viral M1 protein, did not abrogate MARCH8-mediated restriction. While MARCH1 and -8 target similar immunological ligands and both restrict HIV-1, only MARCH8 inhibited IAV infectivity. Deletion of the N-terminal cytoplasmic (N-CT) domain of MARCH8 confirmed it to be a critical determinant of IAV inhibition. Of interest, deletion of the MARCH1 N-CT or its replacement with the MARCH8 N-CT resulted in acquisition of IAV restriction. Together, these data demonstrate that MARCH8 restricts a late stage in IAV replication by a mechanism distinct to its reported activity against other viruses. Moreover, we show that the N-CT of MARCH8 is essential for anti-IAV activity, whereas the MARCH1 N-CT inhibits its ability to restrict IAV. IMPORTANCE The antiviral activity of MARCH8 has been associated with the downregulation of envelope glycoproteins from a range of different viruses, resulting in reduced incorporation into nascent virions. Here, we show that MARCH8 restricts IAV at a late stage in virus replication, but this was not associated with reduced expression of IAV envelope glycoproteins on the surface of infected cells, pointing to a distinct mechanism of antiviral activity. Our studies also demonstrate the differential ability of MARCH1 and -8 to restrict IAV infectivity, highlighting the critical role of the N-CT domain of each protein in modulating IAV restriction. Overall, these studies provide novel insights regarding the mechanisms by which MARCH proteins contribute to cell-intrinsic immunity against IAV.


Subject(s)
Gene Expression , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H3N2 Subtype/genetics , Ubiquitin-Protein Ligases/genetics , Virus Replication/genetics , Animals , Dogs , Down-Regulation , HEK293 Cells , Humans , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H3N2 Subtype/pathogenicity , Madin Darby Canine Kidney Cells
13.
PLoS One ; 16(9): e0257827, 2021.
Article in English | MEDLINE | ID: mdl-34591926

ABSTRACT

Airborne transmission of infectious respiratory pathogens is a significant health hazard for the general public as well as healthcare professionals. Face masks have been frequently utilized as safety measures to limit the transmission of these infectious aerosolized particles. However, the efficacy of face masks in reducing respiratory virus infectivity and pathogenicity is unknown. Improving the effectiveness of masks in blocking viruses is urgently needed. In this study, surgical mask filters were modified by coating the filters with 1, 3, or 5 M of sodium dihydrogen phosphate, and subsequently exposed to the aerosolized respiratory influenza viruses (A/H3N2, A/H5N1) generated by a nebulizer set. Mask filter modification significantly reduced the size and counts of filter pores, which enabled entrapment of 40-60% of aerosolized viruses (captured viruses) with more than 90% of the captured viruses losing their infectivity. Upon contact with the coated mask filters, both the captured viruses and the viruses that managed to bypass the filter pore (passed viruses) were found to be inactivated. Passed viruses demonstrated significantly reduced pathogenicity in mice as indicated by significantly reduced lung virus titers, bodyweight loss, and prolonged survival compared to bare control. These findings highlight the potential of modified mask filters for reducing viral activity and pathogenicity, which contributes to improving facial mask efficacy as well as limiting airborne pathogen transmission.


Subject(s)
Influenza A Virus, H3N2 Subtype/pathogenicity , Influenza A Virus, H5N1 Subtype/pathogenicity , Masks/virology , Orthomyxoviridae Infections/transmission , Animals , Female , Humans , Lung/virology , Mice , Orthomyxoviridae Infections/virology , Viral Load
14.
PLoS One ; 16(8): e0256165, 2021.
Article in English | MEDLINE | ID: mdl-34450617

ABSTRACT

A test-negative case-control study was conducted to assess inactivated influenza vaccine effectiveness (VE) in children aged 6 months-17 years. The database was developed from the US Department of Defense Global Respiratory Pathogen Surveillance Program over four consecutive influenza seasons from 2016 to 2020. A total of 9,385 children including 4,063 medically attended, laboratory-confirmed influenza-positive cases were identified for VE analysis. A generalized linear mixed model with logit link and binomial distribution was used to estimate the VE. The adjusted VE for children was 42% [95% confidence interval (CI): 37-47%] overall, including 55% (95% CI: 47-61%) for influenza A(H1N1)pdm09, 37% (95% CI: 28-45%) for influenza A(H3N2), and 49% (95% CI: 41-55%) for influenza B. The analysis by age groups indicated that the adjusted VE in children aged 6 months-4 years was higher against influenza A(H1N1)pdm09 and influenza B, and comparable against influenza A(H3N2), compared to those in children aged 5-17 years. Further age-stratified analysis showed that the VE against any types of influenza was low and non-significant for children aged 6-11 months (33%; 95% CI:-2-56%), but it was high (54%; 95% CI: 34-67%) in children aged 12-23 months, and then declined linearly with increasing age. In conclusion, the inactivated influenza vaccination was moderately effective against influenza infection, based on the analysis from a large number of children aged 6 months-17 years over multiple influenza seasons.


Subject(s)
Influenza Vaccines/immunology , Influenza, Human/epidemiology , Influenza, Human/prevention & control , Vaccine Efficacy , Adolescent , Child , Child, Preschool , Female , Humans , Infant , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H3N2 Subtype/pathogenicity , Influenza Vaccines/therapeutic use , Influenza, Human/immunology , Influenza, Human/virology , Male , Seasons , Vaccination
15.
Virus Genes ; 57(5): 401-412, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34156583

ABSTRACT

Influenza viruses have a high potential for genetic changes. The objectives of this study were to analyse influenza virus circulation in Bulgaria during the 2019/2020 season, to perform a phylogenetic and molecular analyses of the haemagglutinin (HA) and neuraminidase (NA) sequences of representative influenza strains, and to identify amino acid substitutions compared to the current vaccine strains. Seasonal influenza viruses A(H3N2), A(H1N1)pdm09 and B/Victoria-lineage were detected using a real-time RT-PCR in 323 (23.3%), 149 (10.7%) and 138 (9.9%) out of 1387 patient samples studied, respectively. The HA genes of A(H3N2) viruses analysed belonged to clades 3C.3a (21 strains) and 3C.2a (5 strains): subclades 3C.2a1b + T131K, 3C.2a1b + T135K-B and 3C.2a1b + T135K-A. The clade 3C.3a and subclade 3C.2a1b viruses carried 5 and 14-17 substitutions in HA, as well as 3 and 9 substitutions in NA, respectively, in comparison with the A/Kansas/14/2017 vaccine virus, including some substitutions in the HA antigenic sites A, B, C and E. All 21 A(H1N1)pdm09 viruses sequenced fell into 6B.1A5A subclade. Amino acid sequence analysis revealed the presence of 7-11 substitutions in HA, compared to the A/Brisbane/02/2018 vaccine virus, three of which occurred in antigenic site Sb, along with 6-9 changes at positions in NA. All 10 B/Victoria-lineage viruses sequenced belonged to clade 1A with a triple deletion in HA1 (genetic group 1A(Δ3)B) and carried 7 and 3 substitutions in HA and NA, respectively, with respect to the B/Colorado/06/2017 vaccine virus. The results of this study confirm the rapid evolution of influenza viruses and the need for continuous antigenic and genetic surveillance.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza, Human/genetics , Neuraminidase/genetics , Orthomyxoviridae/genetics , Amino Acid Substitution/genetics , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/pathogenicity , Influenza Vaccines/genetics , Influenza Vaccines/therapeutic use , Influenza, Human/virology , Orthomyxoviridae/classification , Orthomyxoviridae/pathogenicity , Phylogeny , Seasons
16.
Viruses ; 13(6)2021 05 23.
Article in English | MEDLINE | ID: mdl-34071096

ABSTRACT

MicroRNAs (miRNAs) are essential regulators of gene expression in humans and can control pathogenesis and host-virus interactions. Notably, the role of specific host miRNAs during influenza virus infections are still ill-defined. The central goal of this study was to identify novel miRNAs and their target genes in response to influenza virus infections in airway epithelium. Human airway epithelial cells exposed to influenza A virus (IAV) induced several novel miRNAs that were identified using next-generation sequencing (NGS) and their target genes by biochemical methods. NGS analysis predicted forty-two RNA sequences as possible miRNAs based on computational algorithms. The expression patterns of these putative miRNAs were further confirmed using RT-PCR in human bronchial epithelial cells exposed to H1N1, H9N1(1P10), and H9N1 (1WF10) strains of influenza virus. A time-course study showed significant downregulation of put-miR-34 in H1N1 and put-miR-35 in H9N1(1P10)-infected cells, which is consistent with the NGS data. Additionally, put-miR-34 and put-miR-35 showed a high fold enrichment in an argonaute-immunoprecipitation assay compared to the controls, indicating their ability to form a complex with argonaute protein and RNA-induced silencing complex (RISC), which is a typical mode of action found with miRNAs. Our earlier studies have shown that the replication and survival of influenza virus is modulated by certain transcription factors such as NF-ĸB. To identify the target(s) of these putative miRNAs, we screened 84 transcription factors that have a role in viral pathogenesis. Cells transfected with mimic of the put-miR-34 showed a significant decrease in the expression of Signal Transducers and Activators of Transcription 3 (STAT3), whereas the inhibitor of put-miR-34 showed a significant increase in STAT3 expression and its phosphorylation. In addition, put-miR-34 had 76% homology to the untranslated region of STAT3. NGS and PCR array data submitted to the Gene Ontology project also predicted the role of transcription factors modulated by put-miR-34. Our data suggest that put-miR-34 may be a good target for antiviral therapy.


Subject(s)
Host-Pathogen Interactions/genetics , Influenza A virus/genetics , MicroRNAs/genetics , STAT Transcription Factors/genetics , Signal Transduction/genetics , A549 Cells , Bronchi/cytology , Cells, Cultured , Epithelial Cells/virology , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/pathogenicity , Influenza A virus/classification , Influenza A virus/pathogenicity , MicroRNAs/classification , MicroRNAs/isolation & purification , Virus Replication
18.
Emerg Microbes Infect ; 10(1): 472-480, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33657971

ABSTRACT

Pandemic influenza, typically caused by the reassortment of human and avian influenza viruses, can result in severe or fatal infections in humans. Timely identification of potential pandemic viruses must be a priority in influenza virus surveillance. However, the range of host species responsible for the generation of novel pandemic influenza viruses remains unclear. In this study, we conducted serological surveys for avian and human influenza virus infections in farmed mink and determined the susceptibility of mink to prevailing avian and human virus subtypes. The results showed that farmed mink were commonly infected with human (H3N2 and H1N1/pdm) and avian (H7N9, H5N6, and H9N2) influenza A viruses. Correlational analysis indicated that transmission of human influenza viruses occurred from humans to mink, and that feed source was a probable route of avian influenza virus transmission to farmed mink. Animal experiments showed that mink were susceptible and permissive to circulating avian and human influenza viruses, and that human influenza viruses (H3N2 and H1N1/pdm), but not avian viruses, were capable of aerosol transmission among mink. These results indicate that farmed mink could be highly permissive "mixing vessels" for the reassortment of circulating human and avian influenza viruses. Therefore, to reduce the risk of emergence of novel pandemic viruses, feeding mink with raw poultry by-products should not be permitted, and epidemiological surveillance of influenza viruses in mink farms should be urgently implemented.


Subject(s)
Influenza A virus/pathogenicity , Mink/virology , Orthomyxoviridae Infections/transmission , Animals , Disease Models, Animal , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H3N2 Subtype/pathogenicity , Influenza A Virus, H7N9 Subtype/immunology , Influenza A Virus, H7N9 Subtype/pathogenicity , Influenza A Virus, H9N2 Subtype/immunology , Influenza A Virus, H9N2 Subtype/pathogenicity , Influenza A virus/immunology , Mink/immunology , Neutralization Tests , Orthomyxoviridae Infections/immunology , Reassortant Viruses/immunology , Reassortant Viruses/pathogenicity
19.
Arch Environ Occup Health ; 76(8): 526-538, 2021.
Article in English | MEDLINE | ID: mdl-33750267

ABSTRACT

Hydrogen sulfide (H2S) is common in concentrated pig feed operations from the decomposition of manure. Ambient H2S is a respiratory tract irritant and an environmental stressor for caretakers and pigs. Influenza A virus (IAV), a zoonotic pathogen, has caused prior pandemics. The effects of H2S or IAV alone on the respiratory system have been investigated, but their interaction has not. We hypothesized that exposure to environmentally-relevant H2S concentrations increases the pathogenicity of IAV infection in swine. Thirty-five, three-week old pigs of mixed sex were exposed to breathing air or H2S via inhalation 6 hours daily for 12 days. After 7 days, pigs were inoculated with H3N2 IAV (or a placebo). Results showed that ambient H2S increased the severity of respiratory distress and lung pathology. H2S also suppressed IL-IL-1ß, IL-6 and IL-8 cytokine response in BALF and increased viral loads and nasal shedding.


Subject(s)
Hydrogen Sulfide/adverse effects , Influenza A Virus, H3N2 Subtype/pathogenicity , Inhalation Exposure/adverse effects , Orthomyxoviridae Infections/pathology , Animals , Antigens, Viral/metabolism , Cytokines/metabolism , Disease Models, Animal , Female , Lung/metabolism , Lung/pathology , Male , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology , Reactive Nitrogen Species/metabolism , Severity of Illness Index , Swine , Viral Load
20.
Genetics ; 217(2)2021 02 09.
Article in English | MEDLINE | ID: mdl-33724414

ABSTRACT

Basic summary statistics that quantify the population genetic structure of influenza virus are important for understanding and inferring the evolutionary and epidemiological processes. However, the sampling dates of global virus sequences in the last several decades are scattered nonuniformly throughout the calendar. Such temporal structure of samples and the small effective size of viral population hampers the use of conventional methods to calculate summary statistics. Here, we define statistics that overcome this problem by correcting for the sampling-time difference in quantifying a pairwise sequence difference. A simple linear regression method jointly estimates the mutation rate and the level of sequence polymorphism, thus providing an estimate of the effective population size. It also leads to the definition of Wright's FST for arbitrary time-series data. Furthermore, as an alternative to Tajima's D statistic or the site-frequency spectrum, a mismatch distribution corrected for sampling-time differences can be obtained and compared between actual and simulated data. Application of these methods to seasonal influenza A/H3N2 viruses sampled between 1980 and 2017 and sequences simulated under the model of recurrent positive selection with metapopulation dynamics allowed us to estimate the synonymous mutation rate and find parameter values for selection and demographic structure that fit the observation. We found that the mutation rates of HA and PB1 segments before 2007 were particularly high and that including recurrent positive selection in our model was essential for the genealogical structure of the HA segment. Methods developed here can be generally applied to population genetic inferences using serially sampled genetic data.


Subject(s)
Influenza A Virus, H3N2 Subtype/genetics , Influenza, Human/virology , Mutation Rate , Seasons , Humans , Influenza A Virus, H3N2 Subtype/isolation & purification , Influenza A Virus, H3N2 Subtype/pathogenicity , Influenza, Human/epidemiology , Models, Genetic , Sampling Studies , Selection, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...