Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 127
1.
Biomed Res Int ; 2021: 3890681, 2021.
Article En | MEDLINE | ID: mdl-34337007

The entire H5N1 highly pathogenic avian influenza viral genomes were identified in the frozen autopsy specimens: the trachea, lung, colon, and intestinal feces from a patient who died of the disease in 2006. Phylogenetic analysis of the viral genomes showed that these viruses belonged to clade 1 and were the reassortants generated from the reassortment of the viruses within the same clade. The sequencing data from the autopsy specimens revealed at least 8 quasispecies of the H5N1 viruses across all 4 specimen types. These sequences were compared to those derived from the virus isolates grown in Madin Darby canine kidney (MDCK) cells. The virus isolates from the trachea, lung, and fecal specimens showed 27 nucleotide substitutions, leading to the changes of 18 amino acid residues. However, there was no change in the amino acid residues that determined the viral virulence. The changes were more commonly observed in the lung, particularly in the HA and NA genes. Our study suggested that the adaptation changes for the viral fitness to survive in a new host species (MDCK cells) might involve many genes, for example, the amino acid substitution 177G or 177W adjacent to the receptor-binding residues in the HA1 globular head and the substitution M315I in PB2. However, a mutation changes near the receptor binding domain may play an important role in determining the cell tropism and is needed to be further explored.


Adaptation, Physiological , Autopsy , Cell Culture Techniques , Genetic Variation , Genome, Viral , Influenza A Virus, H5N1 Subtype/growth & development , Influenza A Virus, H5N1 Subtype/genetics , Adaptation, Physiological/genetics , Amino Acid Sequence , Animals , Base Sequence , Dogs , Fatal Outcome , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Madin Darby Canine Kidney Cells , Male , Middle Aged , Phylogeny , Virulence/genetics
2.
mBio ; 12(1)2021 02 09.
Article En | MEDLINE | ID: mdl-33563825

Since the emergence of highly pathogenic avian influenza viruses of the H5 subtype, the major viral antigen, hemagglutinin (HA), has undergone constant evolution, resulting in numerous genetic and antigenic (sub)clades. To explore the consequences of amino acid changes at sites that may affect the antigenicity of H5 viruses, we simultaneously mutated 17 amino acid positions of an H5 HA by using a synthetic gene library that, theoretically, encodes all combinations of the 20 amino acids at the 17 positions. All 251 mutant viruses sequenced possessed ≥13 amino acid substitutions in HA, demonstrating that the targeted sites can accommodate a substantial number of mutations. Selection with ferret sera raised against H5 viruses of different clades resulted in the isolation of 39 genotypes. Further analysis of seven variants demonstrated that they were antigenically different from the parental virus and replicated efficiently in mammalian cells. Our data demonstrate the substantial plasticity of the influenza virus H5 HA protein, which may lead to novel antigenic variants.IMPORTANCE The HA protein of influenza A viruses is the major viral antigen. In this study, we simultaneously introduced mutations at 17 amino acid positions of an H5 HA expected to affect antigenicity. Viruses with ≥13 amino acid changes in HA were viable, and some had altered antigenic properties. H5 HA can therefore accommodate many mutations in regions that affect antigenicity. The substantial plasticity of H5 HA may facilitate the emergence of novel antigenic variants.


Amino Acid Substitution/genetics , Antigenic Variation/genetics , Evolution, Molecular , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza A Virus, H5N1 Subtype/genetics , Animals , Antigens, Viral/immunology , COS Cells , Cell Line , Chlorocebus aethiops , Dogs , Female , Ferrets , Gene Library , HEK293 Cells , Hemagglutinin Glycoproteins, Influenza Virus/classification , Humans , Influenza A Virus, H5N1 Subtype/chemistry , Influenza A Virus, H5N1 Subtype/growth & development , Influenza A Virus, H5N1 Subtype/pathogenicity , Madin Darby Canine Kidney Cells , Mutation
3.
Viruses ; 12(10)2020 10 08.
Article En | MEDLINE | ID: mdl-33050000

Janus kinase (JAK) inhibitors have been developed as novel immunomodulatory drugs and primarily used for treating rheumatoid arthritis and other inflammatory diseases. Recent studies have suggested that this category of anti-inflammatory drugs could be potentially useful for the control of inflammation "storms" in respiratory virus infections. In addition to their role in regulating immune cell functions, JAK1 and JAK2 have been recently identified as crucial cellular factors involved in influenza A virus (IAV) replication and could be potentially targeted for antiviral therapy. Gingerenone A (Gin A) is a compound derived from ginger roots and a dual inhibitor of JAK2 and p70 S6 kinase (S6K1). Our present study aimed to determine the antiviral activity of Gin A on influenza A virus (IAV) and to understand its mechanisms of action. Here, we reported that Gin A suppressed the replication of three IAV subtypes (H1N1, H5N1, H9N2) in four cell lines. IAV replication was also inhibited by Ruxolitinib (Rux), a JAK inhibitor, but not by PF-4708671, an S6K1 inhibitor. JAK2 overexpression enhanced H5N1 virus replication and attenuated Gin A-mediated antiviral activity. In vivo experiments revealed that Gin A treatment suppressed IAV replication in the lungs of H5N1 virus-infected mice, alleviated their body weight loss, and prolonged their survival. Our study suggests that Gin A restricts IAV replication by inhibiting JAK2 activity; Gin A could be potentially useful for the control of influenza virus infections.


Antiviral Agents/pharmacology , Diarylheptanoids/pharmacology , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H5N1 Subtype/drug effects , Influenza A Virus, H9N2 Subtype/drug effects , Janus Kinase 2/antagonists & inhibitors , A549 Cells , Animals , Cell Line , Dogs , Female , HEK293 Cells , Humans , Imidazoles/pharmacology , Influenza A Virus, H1N1 Subtype/growth & development , Influenza A Virus, H5N1 Subtype/growth & development , Influenza A Virus, H9N2 Subtype/growth & development , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred C57BL , Nitriles , Piperazines/pharmacology , Pyrazoles/pharmacology , Pyrimidines , Ribosomal Protein S6 Kinases, 70-kDa/antagonists & inhibitors , Virus Replication/drug effects
4.
Biosci Rep ; 40(5)2020 05 29.
Article En | MEDLINE | ID: mdl-32432671

BACKGROUND: Influenza A virus (IAV) has greatly affected public health in recent decades. Accumulating data indicated that host microRNAs (miRNAs) were related to IAV replication. The present study mainly focused on the effects of microRNA-21-3p (miR-21-3p) on H5N1 replication. METHODS: The levels of miR-21-3p, virus structural factors (matrix 1 (M1), nucleoprotein (NP)), type I interferon (IFN) response markers (IFN-ß, IFN-α), IFN-stimulated genes (protein kinase R (PKR), myxovirus resistance A (MxA), 2'-5'-oligoadenylate synthetase 2 (OAS)), and fibroblast growth factor 2 (FGF2) were measured by quantitative real-time polymerase chain reaction (qRT-PCR). The protein levels of M1, NP, and FGF2 were tested by Western blot assay. The virus titer was assessed by tissue culture infective dose 50% (TCID50) assay. The dual-luciferase reporter assay and ribonucleic acid (RNA) immunoprecipitation (RIP) assay were used to verify the interaction between miR-21-3p and FGF2. RESULTS: MiR-21-3p was reduced in H5N1-infected patients and A549 cells. MiR-21-3p overexpression facilitated the levels of M1, NP, TCID50 value, and reduced the levels of IFN-ß, IFN-α, PKR, MxA, and OAS in H5N1-infected A549 cells. FGF2 was verified as a direct target of miR-21-3p. The introduction of FGF2 counteracted miR-21-3p-mediated decrease in the levels of M1, NP, and TCID50 value, as well as reduction in the levels of IFN-ß, IFN-α, PKR, MxA, and OAS in H5N1-infected A549 cells. CONCLUSION: MiR-21-3p down-regulated FGF2 expression to accelerate H5N1 replication and confine IFN response.


Fibroblast Growth Factor 2/metabolism , Influenza A Virus, H5N1 Subtype/growth & development , Influenza, Human/virology , Interferon Type I/metabolism , MicroRNAs/metabolism , Virus Replication , A549 Cells , Adult , Case-Control Studies , Female , Fibroblast Growth Factor 2/genetics , Gene Expression Regulation , Host-Pathogen Interactions , Humans , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza, Human/genetics , Influenza, Human/metabolism , Interferon Type I/genetics , Male , MicroRNAs/blood , MicroRNAs/genetics
5.
J Vet Med Sci ; 82(5): 619-625, 2020 May 20.
Article En | MEDLINE | ID: mdl-32173692

We compared the Mx expression and anti-viral function and the 3D structure of Mx protein in four species: chicken (Gallus gallus), whooper swan (Cygnus cygnus), jungle crow (Corvus macrorhynchos), and rock dove (Columba livia). We observed different mortalities associated with highly pathogenic avian influenza virus (HPAIV) infection to understand the relationship between Mx function as an immune response factor and HPAIV proliferation in bird cells. Different levels of Mx were observed among the different bird species after virus infection. Strong Mx expression was confirmed in the rock dove and whooper swan 6 hr after viral infection. The lowest virus copy numbers were observed in rock dove. The virus infectivity was significantly reduced in the BALB/3T3 cells expressing rock dove and jungle crow Mx. These results suggested that high Mx expression and significant Mx-induced anti-viral effects might result in the rock dove primary cells having the lowest virus copy number. Comparison of the expected 3D structure of Mx protein in all four bird species demonstrated that the structure of loop L4 varied among the investigated species. It was reported that differences in amino acid sequence in loop L4 affect antiviral activity in human and mouse cells, and a significant anti-viral effect was observed in the rock dove Mx. Thus, the amino acid sequence of loop L4 in rock dove might represent relatively high anti-viral activity.


Influenza A Virus, H5N1 Subtype/immunology , Influenza in Birds/immunology , Myxovirus Resistance Proteins/chemistry , Myxovirus Resistance Proteins/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , Birds , Cells, Cultured , Influenza A Virus, H5N1 Subtype/growth & development , Influenza in Birds/virology , Mice , Myxovirus Resistance Proteins/genetics , Protein Conformation , RNA, Viral , Species Specificity
6.
Virology ; 537: 263-271, 2019 11.
Article En | MEDLINE | ID: mdl-31539775

Activation of c-Jun terminal kinase (JNK) by the nonstructural protein 1 (NS1) of the H5N1 subtype of influenza A virus (IAV) plays an important role in inducing autophagy and virus replication. However, the mechanisms of NS1-induced JNK activation remain elusive. Here we first confirmed the ability of H5N1 (A/mallard/Huadong/S/2005) to activate JNK and to induce autophagy in 293T cells, a human embryonic kidney cell line. We further showed that TAK1, MAP kinase kinase 4 (MKK4), and JNK were activated in 293T cells transfected with the NS1 gene of the H5N1 virus. JNK activation by the NS1 protein or by H5N1 virus was blocked by 5Z-7-Oxozeaenol (5Z), a TAK1-specific inhibitor, and by TAK1 siRNA. Further study showed that 5Z and TAK1 siRNA suppressed H5N1 virus-induced autophagy and inhibited virus replication. Our study unveiled a previously unrecognized role of TAK1 in IAV replication, IAV-induced JNK activation, and autophagy.


Adaptor Proteins, Signal Transducing/metabolism , Host-Pathogen Interactions , Influenza A Virus, H5N1 Subtype/growth & development , MAP Kinase Kinase 4/metabolism , Viral Nonstructural Proteins/metabolism , Virus Replication , Autophagy , HEK293 Cells , Humans , Mitogen-Activated Protein Kinases/metabolism , Plant Proteins/metabolism
7.
Virology ; 537: 110-120, 2019 11.
Article En | MEDLINE | ID: mdl-31493649

Influenza A virus infection activates various intracellular signaling pathways, which is mediated by the transcription factors. Here, a quantitative phosphoproteomic analysis of A549 cells after infection with influenza A virus (H5N1) was performed and we found that the transcription factor STAT1 was highly activated. Unexpectedly, upon inhibition of p-STAT1, titers of progeny virus and viral protein synthesis were both reduced. The STAT1 inhibitor Fludarabine (FLUD) inhibited an early progeny step in viral infection and reduced the levels of influenza virus genomic RNA (vRNA). Concomitantly, there was reduced expression of inflammatory cytokines in p-STAT1 inhibited cells. In vivo, suppression of p-STAT1 improved the survival of H5N1 virus-infected mice, reduced the pulmonary inflammatory response and viral burden. Thus, our data demonstrated a critical role for p-STAT1 in influenza virus replication and inflammatory responses. We speculate that STAT1 is an example of a putative antiviral signaling component to support effective replication.


Inflammation/immunology , Influenza A Virus, H5N1 Subtype/growth & development , Influenza A Virus, H5N1 Subtype/immunology , STAT1 Transcription Factor/metabolism , Virus Replication , A549 Cells , Animals , Cytokines/analysis , Disease Models, Animal , Dogs , Humans , Lethal Dose 50 , Lung/pathology , Lung/virology , Madin Darby Canine Kidney Cells , Mice , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Phosphoproteins/analysis , Proteome/analysis , RNA, Viral/analysis , Survival Analysis , Viral Load , Viral Proteins/analysis
8.
PLoS One ; 14(8): e0220803, 2019.
Article En | MEDLINE | ID: mdl-31404117

Since 1997, the highly pathogenic influenza H5N1 virus has spread from Hong Kong. According to the WHO bulletin report, the H5N1 virus is a zoonotic disease threat that has infected more than 850 humans, causing over 450 deaths. In addition, an outbreak of another new and highly pathogenic influenza virus (H7N9) occurred in 2013 in China. These highly pathogenic influenza viruses could potentially cause a worldwide pandemic. it is crucial to develop a rapid production platform to meet this surge demand against any possible influenza pandemic. A potential solution for this problem is the use of cell-based bioreactors for rapid vaccine production. These novel bioreactors, used for cell-based vaccine production, possess various advantages. For example, they enable a short production time, allow for the handling highly pathogenic influenza in closed environments, and can be easily scaled up. In this study, two novel disposable cell-based bioreactors, BelloCell and TideCell, were used to produce H5N1 clade II and H7N9 candidate vaccine viruses (CVVs). Madin-Darby canine kidney (MDCK) cells were used for the production of these influenza CVVs. A novel bench-scale bioreactor named BelloCell bioreactor was used in the study. All culturing conditions were tested and scaled to 10 L industrial-scale bioreactor known as TideCell002. The performances of between BelloCell and TideCell were similar in cell growth, the average MDCK cell doubling time was slightly decreased to 25 hours. The systems yielded approximately 39.2 and 18.0 µg/ml of HA protein with the 10-liter TideCell002 from the H5N1 clade II and H7N9 CVVs, respectively. The results of this study not only highlight the overall effectiveness of these bioreactors but also illustrate the potential of maintaining the same outcome when scaled up to industrial production, which has many implications for faster vaccine production. Although additional studies are required for process optimization, the results of this study are promising and show that oscillating bioreactors may be a suitable platform for pandemic influenza virus production.


Bioreactors , Disposable Equipment , Influenza A Virus, H5N1 Subtype/growth & development , Influenza A Virus, H7N9 Subtype/growth & development , Influenza Vaccines/biosynthesis , Animals , Chlorocebus aethiops , Dogs , Humans , Influenza, Human/epidemiology , Influenza, Human/virology , Madin Darby Canine Kidney Cells/virology , Pandemics , Vero Cells/virology
9.
Article En | MEDLINE | ID: mdl-30972307

This study aimed to detect changes in the complete transcriptome of MDCK cells after infection with the H5N1 and H3N2 canine influenza viruses using high-throughput sequencing, search for differentially expressed RNAs in the transcriptome of MDCK cells infected with H5N1 and H3N2 using comparative analysis, and explain the differences in the pathogenicity of H5N1 and H3N2 at the transcriptome level. Based on the results of our comparative analysis, significantly different levels of expression were found for 2,464 mRNAs, 16 miRNAs, 181 lncRNAs, and 262 circRNAs in the H3N2 infection group and 448 mRNAs, 12 miRNAs, 77 lncRNAs, and 189 circRNAs in the H5N1 infection group. Potential functions were predicted by performing Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses of the target genes of miRNAs, lncRNAs and circRNAs, and the ncRNA-mRNA regulatory network was constructed based on differentially expressed RNAs. A greater number of pathways regulating immune metabolism were altered in the H3N2 infection group than in the H5N1 infection group, which may be one reason why the H3N2 virus is less pathogenic than is the H5N1 virus. This study provides detailed data on the production of ncRNAs during infection of MDCK cells by the canine influenza viruses H3N2 and H5N1, analyzed differences in the total transcriptomes between H3N2- and H5N1-infected MDCK cells, and explained these differences with regard to the pathogenicity of H3N2 and H5N1 at the transcriptional level.


Gene Expression Profiling , Influenza A Virus, H3N2 Subtype/growth & development , Influenza A Virus, H5N1 Subtype/growth & development , Madin Darby Canine Kidney Cells/virology , Animals , Dogs , High-Throughput Nucleotide Sequencing , Molecular Sequence Annotation
10.
Bull Exp Biol Med ; 166(3): 358-363, 2019 Jan.
Article En | MEDLINE | ID: mdl-30627911

Changes in the kidney structure in outbred and inbred male BALB/c mice were analyzed in the acute period after infection with influenza viruses A/H5N1 (10 MLD50; 10 days) and A/H1N1 (1 MLD50; 30 days). Antibodies to influenza viruses of both strains were most often expressed by endothelial cells of the glomeruli and arterioles and were rarely expressed by mesangiocytes and tubule epithelial cells. In the kidney, destructive processes induced by viruses and by ischemia due to massive blood vessel thrombosis. Mesangiocytes expressed factors, indicating that they could be qualified as M1 and M2 macrophages. Kidney destruction was more significant after infection of mice with the A/H5N1 virus, but in both experiments cell infiltrates were actually absent, probably due to blood vessel thrombosis and limited possibility of migration of mononuclear phagocytes and lymphocytes to the kidney.


Endothelial Cells/pathology , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H5N1 Subtype/pathogenicity , Kidney Glomerulus/pathology , Macrophages/pathology , Mesangial Cells/pathology , Orthomyxoviridae Infections/pathology , Animals , Animals, Outbred Strains , Antigens, Viral/genetics , Antigens, Viral/immunology , Endothelial Cells/virology , Gene Expression , Host-Pathogen Interactions , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/growth & development , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/growth & development , Interleukin-16/genetics , Interleukin-16/immunology , Kidney Glomerulus/blood supply , Kidney Glomerulus/virology , Lipopolysaccharide Receptors/genetics , Lipopolysaccharide Receptors/immunology , Macrophages/virology , Male , Mesangial Cells/virology , Mice , Mice, Inbred BALB C , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/immunology , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
11.
Article En | MEDLINE | ID: mdl-30460207

The inflammatory response and apoptosis have been proved to have a crucial role in the pathogenesis of the influenza A virus (IAV). Previous studies indicated that while IAV commonly causes pancreatitis and pancreatic damage in naturally and experimentally infected animals, the molecular mechanisms of the pathogenesis of IAV infection are less reported. In the present study, we showed for the first time that both avian-like (α-2,3-linked) and human-like (α-2,6-linked) sialic acid (SA) receptors were expressed by the mouse pancreatic cancer cell line PAN02 and the human pancreatic cancer cell line PANC-1. Using growth kinetics experiments, we also showed that PAN02 and PANC-1 cells supported the productive replication of the H5N1 highly pathogenic avian influenza while exhibited the limited replication of IAV subtypes H1N1 and H7N2 in vitro. The in vivo infection of H5N1 in pancreatic cells was confirmed by the histopathological and immunohistochemical staining of pancreas tissue from mice. Other than H1N1 and H7N2, severe damage and extensive positive signals were observed in pancreas of H5N1 infected mice. All three virus subtypes induced apoptosis but also triggered the infected PAN02 and PANC-1 cells to release pro-inflammatory cytokines and chemokines including interferon (IFN)-α, IFN-ß, IFN-γ, chemokine (C-C motif) ligand 2 (CCL2), tumor necrosis factor (TNF)-α, and interleukin (IL)-6. Notably, the subtypes of H5N1 could significantly upregulate these cytokines and chemokines in both two cells when compared with H1N1 and H7N2. The present data provide further understanding of the pathogenesis of H5N1 IAV in pancreatic cells derived from humans and mammals and may also benefit the development of new treatment against H5N1 influenza virus infection.


Apoptosis , Cytokines/metabolism , Influenza A Virus, H5N1 Subtype/growth & development , Influenza A Virus, H5N1 Subtype/immunology , Viral Tropism , Virus Replication , Animals , Cell Line, Tumor , Histocytochemistry , Humans , Immunohistochemistry , Influenza A Virus, H1N1 Subtype/growth & development , Influenza A Virus, H7N2 Subtype/growth & development , Mice , Microscopy , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Pancreas/pathology , Pancreas/virology
12.
BMC Microbiol ; 18(1): 138, 2018 10 19.
Article En | MEDLINE | ID: mdl-30340527

BACKGROUND: Endothelial cells play a major role in highly pathogenic avian influenza (HPAI) virus pathogenesis in gallinaceous poultry species (e.g. chicken, turkey and quail). Upon infection of gallinaceous poultry with HPAI viruses, endothelial cells throughout the body become rapidly infected, leading to systemic dissemination of the virus, disseminated intravascular coagulation, oedema and haemorrhaging. In contrast, the pathogenesis of HPAI viruses in most wild bird species (e.g. duck, goose and gull species) is not associated with endothelial tropism. Indeed, viral antigen is not found in the endothelial cells of most wild bird species following infection with HPAI viruses. This differential endothelial cell tropism in avian species is poorly understood, mainly due to the absence of appropriate cell culture systems. RESULTS: Here, we describe the isolation and purification of primary duck endothelial cells from the aorta or bone marrow of Pekin duck embryos. Cells were differentiated in the presence of vascular endothelial growth factor and, if needed, enriched via fluorescent-activated cell sorting based on the uptake of acetylated low-density lipoprotein. The expression of von Willebrand factor, a key marker of endothelial cells, was confirmed by polymerase chain reaction. Monocultures of duck endothelial cells, either derived from the aorta or the bone marrow, were susceptible to infection with an H5N1 HPAI virus but to a much lesser extent than chicken endothelial cells. CONCLUSIONS: The methods described herein to isolate and purify duck endothelial cells from the aorta or bone marrow could also be applied to obtain microvascular endothelial cells from other tissues and organs, such as the lung or the intestine, and represent a valuable tool to study the pathogenesis of avian viruses.


Cell Culture Techniques , Endothelial Cells/virology , Influenza A Virus, H5N1 Subtype/growth & development , Influenza A Virus, H5N1 Subtype/pathogenicity , Animals , Antigens, Viral , Aorta/cytology , Aorta/virology , Bone Marrow Cells/virology , Cells, Cultured , Ducks/virology , Flow Cytometry , Influenza in Birds/virology , Poultry Diseases/virology , Vascular Endothelial Growth Factor A/pharmacology
13.
Sci Rep ; 8(1): 15468, 2018 10 19.
Article En | MEDLINE | ID: mdl-30341336

Current prophylactic and therapeutic strategies targeting human influenza viruses include vaccines and antivirals. Given variable rates of vaccine efficacy and antiviral resistance, alternative strategies are urgently required to improve disease outcomes. Here we describe the use of HiSeq deep sequencing to analyze host gene expression in primary human alveolar epithelial type II cells infected with highly pathogenic avian influenza H5N1 virus. At 24 hours post-infection, 623 host genes were significantly upregulated, including the cell adhesion molecule CEACAM1. H5N1 virus infection stimulated significantly higher CEACAM1 protein expression when compared to influenza A PR8 (H1N1) virus, suggesting a key role for CEACAM1 in influenza virus pathogenicity. Furthermore, silencing of endogenous CEACAM1 resulted in reduced levels of proinflammatory cytokine/chemokine production, as well as reduced levels of virus replication following H5N1 infection. Our study provides evidence for the involvement of CEACAM1 in a clinically relevant model of H5N1 infection and may assist in the development of host-oriented antiviral strategies.


Alveolar Epithelial Cells/virology , Antigens, CD/metabolism , Cell Adhesion Molecules/metabolism , Host-Pathogen Interactions , Influenza A Virus, H5N1 Subtype/growth & development , Cells, Cultured , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Influenza A Virus, H1N1 Subtype/growth & development
14.
Sci Rep ; 8(1): 13066, 2018 08 30.
Article En | MEDLINE | ID: mdl-30166556

The role of the influenza virus polymerase complex in host range restriction has been well-studied and several host range determinants, such as the polymerase PB2-E627K and PB2-D701N mutations, have been identified. However, there may be additional, currently unknown, human adaptation polymerase mutations. Here, we used a database search of influenza virus H5N1 clade 1.1, clade 2.3.2.1 and clade 2.3.4 strains isolated from 2008-2012 in Southern China, Vietnam and Cambodia to identify polymerase adaptation mutations that had been selected in infected patients. Several of these mutations acted either alone or together to increase viral polymerase activity in human airway cells to levels similar to the PB2-D701N and PB2-E627K single mutations and to increase progeny virus yields in infected mouse lungs to levels similar to the PB2-D701N single mutation. In particular, specific mutations acted synergistically with the PB2-D701N mutation and showed synergistic effects on viral replication both in human airway cells and mice compared with the corresponding single mutations. Thus, H5N1 viruses in infected patients were able to acquire multiple polymerase mutations that acted cooperatively for human adaptation. Our findings give new insight into the human adaptation of AI viruses and help in avian influenza virus risk assessment.


Adaptation, Physiological/genetics , DNA-Directed RNA Polymerases/genetics , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/isolation & purification , Mutation/genetics , A549 Cells , Animals , Asia , Birds/virology , DNA-Directed RNA Polymerases/chemistry , Epithelial Cells/virology , HEK293 Cells , Humans , Influenza A Virus, H5N1 Subtype/growth & development , Lung/pathology , Mice , Models, Molecular , Virus Replication
15.
J Interferon Cytokine Res ; 38(7): 290-297, 2018 07.
Article En | MEDLINE | ID: mdl-30016179

The highly pathogenic H5N1 avian influenza virus (AIV) is widespread in waterfowl, causing enormous economic losses and posing a significant threat to public health. An increasing number of reagents have been identified to prevent the spread of influenza; however, there have been no reports on the anti-H5N1 effects of duck interferons, which exhibit antiviral activity against other viruses. Our aim was to investigate the antiviral effects of purified duck interferons. In this study, we successfully cloned and expressed duck interferon gamma (IFN-γ) in Escherichia coli. The antiviral effects of this recombinant duck IFN-γ (rDuIFN-γ) was assessed in vitro and in vivo. rDuIFN-γ displayed antiviral activity against vesicular stomatitis virus and AIV in duck embryo fibroblasts. Pretreating ducks with 3.4 × 104 U rDuIFN-γ also partially decreased mortality from 70% to 30% and delayed onset in 2-day-old Peking ducks. Virus titers in tissues and viral shedding decreased, and the expression of interferon-stimulated genes increased in brain and spleen in rDuIFN-γ-treated ducks. These results indicate that duck IFN-γ has the potential to inhibit viral replication in ducks.


Antiviral Agents/pharmacology , Influenza A Virus, H5N1 Subtype/drug effects , Interferon-gamma/pharmacology , Recombinant Proteins/pharmacology , Virus Replication/drug effects , Animals , Ducks , Influenza A Virus, H5N1 Subtype/growth & development , Microbial Sensitivity Tests
16.
J Microbiol Biotechnol ; 28(6): 997-1006, 2018 Jun 28.
Article En | MEDLINE | ID: mdl-29642288

As shown during the 2009 pandemic H1N1 (A(H1N1)pdm09) outbreak, egg-based influenza vaccine production technology is insufficient to meet global demands during an influenza pandemic. Therefore, there is a need to adapt cell culture-derived vaccine technology using suspended cell lines for more rapid and larger-scale vaccine production. In this study, we attempted to generate a high-growth influenza vaccine strain in MDCK cells using an A/Puerto/8/1934 (H1N1) vaccine seed strain. Following 48 serial passages with four rounds of virus plaque purification in MDCK cells, we were able to select several MDCK-adapted plaques that could grow over 108 PFU/ml. Genetic characterization revealed that these viruses mainly had amino acid substitutions in internal genes and exhibited higher polymerase activities. By using a series of Rg viruses, we demonstrated the essential residues of each gene and identified a set of high-growth strains in MDCK cells (PB1D153N, M1A137T, and NS1N176S). In addition, we confirmed that in the context of the high-growth A/PR/8/34 backbone, A/California/7/2009 (H1N1), A/Perth/16/2009 (H3N2), and A/environment/Korea/deltaW150/2006 (H5N1) also showed significantly enhanced growth properties (more than 107 PFU/ml) in both attached- and suspended-MDCK cells compared with each representative virus and the original PR8 vaccine strain. Taken together, this study demonstrates the feasibility of a cell culture-derived approach to produce seed viruses for influenza vaccines that are cheap and can be grown promptly and vigorously as a substitute for egg-based vaccines. Thus, our results suggest that MDCK cell-based vaccine production is a feasible option for producing large-scale vaccines in case of pandemic outbreaks.


Influenza A Virus, H1N1 Subtype/growth & development , Influenza Vaccines/isolation & purification , Technology, Pharmaceutical/methods , Virus Cultivation/methods , Amino Acid Substitution , Animals , Dogs , Genome, Viral , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/growth & development , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/growth & development , Madin Darby Canine Kidney Cells , Mutation, Missense , Sequence Analysis, DNA , Serial Passage
17.
J Virol ; 92(11)2018 06 01.
Article En | MEDLINE | ID: mdl-29593036

Influenza virus-specific CD8+ T lymphocytes (CTLs) contribute to clearance of influenza virus infections and reduce disease severity. Variation at amino acid residues located in or outside CTL epitopes has been shown to affect viral recognition by virus-specific CTLs. In the present study, we investigated the effect of naturally occurring variation at residues outside the conserved immunodominant and HLA*0201-restricted M158-66 epitope, located in the influenza virus M1 protein, on the extent of virus replication in the presence of CTLs specific for the epitope. To this end, we used isogenic viruses with an M1 gene segment derived from either an avian or a human influenza virus, HLA-transgenic human epithelial cells, human T cell clones specific for the M158-66 epitope or a control epitope, and a novel, purposely developed in vitro system to coculture influenza virus-infected cells with T cells. We found that the M gene segment of a human influenza A/H3N2 virus afforded the virus the capacity to replicate better in the presence of M158-66-specific CTLs than the M gene segment of avian viruses. These findings are in concordance with previously observed differential CTL activation, caused by variation at extra-epitopic residues, and may reflect an immune adaptation strategy of human influenza viruses that allows them to cope with potent CTL immunity to the M158-66 epitope in HLA-A*0201-positive individuals, resulting in increased virus replication and shedding and possibly increasing disease severity.IMPORTANCE Influenza viruses are among the leading causes of acute respiratory tract infections. CD8+ T lymphocytes display a high degree of cross-reactivity with influenza A viruses of various subtypes and are considered an important correlate of protection. Unraveling viral immune evasion strategies and identifying signs of immune adaptation are important for defining the role of CD8+ T lymphocytes in affording protection more accurately. Improving our insight into the interaction between influenza viruses and virus-specific CD8+ T lymphocyte immunity may help to advance our understanding of influenza virus epidemiology, aid in risk assessment of potentially pandemic influenza virus strains, and benefit the design of vaccines that induce more broadly protective immunity.


CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza, Human/immunology , Viral Matrix Proteins/immunology , A549 Cells , Animals , Cell Line, Tumor , Dogs , Epitopes, T-Lymphocyte/genetics , HLA-A2 Antigen/immunology , Humans , Influenza A Virus, H1N1 Subtype/growth & development , Influenza A Virus, H3N2 Subtype/growth & development , Influenza A Virus, H5N1 Subtype/growth & development , Influenza, Human/virology , Madin Darby Canine Kidney Cells , Viral Matrix Proteins/genetics , Virus Replication/immunology
18.
Sci Rep ; 8(1): 2092, 2018 02 01.
Article En | MEDLINE | ID: mdl-29391557

The human interferon (IFN)-induced MxA protein is a key antiviral host restriction factor exhibiting broad antiviral activity against many RNA viruses, including highly pathogenic avian influenza A viruses (IAV) of the H5N1 and H7N7 subtype. To date the mechanism for how MxA exerts its antiviral activity is unclear, however, additional cellular factors are believed to be essential for this activity. To identify MxA cofactors we performed a genome-wide siRNA-based screen in human airway epithelial cells (A549) constitutively expressing MxA using an H5N1 reporter virus. These data were complemented with a proteomic screen to identify MxA-interacting proteins. The combined data identified SMARCA2, the ATPase subunit of the BAF chromatin remodeling complex, as a crucial factor required for the antiviral activity of MxA against IAV. Intriguingly, our data demonstrate that although SMARCA2 is essential for expression of some IFN-stimulated genes (ISGs), and the establishment of an antiviral state, it is not required for expression of MxA, suggesting an indirect effect on MxA activity. Transcriptome analysis of SMARCA2-depleted A549-MxA cells identified a small set of SMARCA2-regulated factors required for activity of MxA, in particular IFITM2 and IGFBP3. These findings reveal that several virus-inducible factors work in concert to enable MxA restriction of IAV.


Influenza A Virus, H5N1 Subtype/growth & development , Influenza A Virus, H7N7 Subtype/growth & development , Influenza, Human/virology , Myxovirus Resistance Proteins/metabolism , Transcription Factors/metabolism , A549 Cells , Antiviral Agents/pharmacology , Gene Expression Profiling , Host-Pathogen Interactions , Humans , Influenza A Virus, H5N1 Subtype/drug effects , Influenza A Virus, H7N7 Subtype/drug effects , Influenza, Human/drug therapy , Influenza, Human/metabolism , Interferons/pharmacology , Myxovirus Resistance Proteins/genetics , Proteome/analysis , Transcription Factors/genetics , Virus Replication
19.
Biochem Biophys Res Commun ; 494(1-2): 298-304, 2017 12 09.
Article En | MEDLINE | ID: mdl-29017920

Infection with the highly pathogenic avian influenza H5N1 virus results in a high incidence of mortality in humans. Severe complications from infection are often associated with hypercytokinemia. However, current neuraminidase inhibitors (NAIs) have several limitations including the appearance of oseltamivir-resistant H5N1 virus and the inability to completely ameliorate hyper-immune responses. To overcome these limitations, we evaluated the anti-viral activity of mycophenolic mofetil (MMF) against A/Vietnam/1194/2004 (H5N1) virus infection using MDCK cells and mice. The IC50 of MMF (0.94 µM) was comparable to that of zanamivir (0.87 µM) in H5N1 virus-infected MDCK cells based on ELISA. Time-course assays demonstrated that MMF completely inhibited H5N1 viral mRNA replication and protein expression for approximately 8 h after the initiation of treatment. In addition, MMF treatment protected 100% of mice, and lung viral titers were substantially reduced. The anti-viral mechanism of MMF against H5N1 virus infection was further confirmed to depend on the inhibition of cellular inosine monophosphate dehydrogenase (IMPDH) by exogenous guanosine, which inhibits viral mRNA and protein expression. Moreover, IL-1ß, IFN-ß, IL-6, and IP-10 mRNA expression levels were significantly downregulated in MDCK cells with MMF treatment. These results indicated that MMF could represent a novel inhibitor of viral replication and a potent immunomodulator for the treatment of H5N1 virus infection.


Antiviral Agents/pharmacology , Immunologic Factors/pharmacology , Influenza A Virus, H5N1 Subtype/drug effects , Mycophenolic Acid/pharmacology , Orthomyxoviridae Infections/drug therapy , Oseltamivir/pharmacology , Animals , Chemokine CXCL10/antagonists & inhibitors , Chemokine CXCL10/genetics , Chemokine CXCL10/immunology , Chick Embryo , Dogs , Female , Gene Expression Regulation , Host-Pathogen Interactions/drug effects , IMP Dehydrogenase/antagonists & inhibitors , IMP Dehydrogenase/genetics , IMP Dehydrogenase/immunology , Influenza A Virus, H5N1 Subtype/growth & development , Influenza A Virus, H5N1 Subtype/pathogenicity , Interferon-beta/antagonists & inhibitors , Interferon-beta/genetics , Interferon-beta/immunology , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-6/antagonists & inhibitors , Interleukin-6/genetics , Interleukin-6/immunology , Lung/drug effects , Lung/immunology , Lung/virology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/pathology , RNA, Viral/antagonists & inhibitors , RNA, Viral/biosynthesis , Survival Analysis , Virus Replication/drug effects , Zanamivir/pharmacology
20.
Mol Biol (Mosk) ; 51(4): 717-723, 2017.
Article Ru | MEDLINE | ID: mdl-28900092

We have previously described nanocomposites containing conjugates or complexes of native oligodeoxyribonucleotides with poly-L-lysine and TiO2 nanoparticles. We have shown that these nanocomposites efficiently suppressed influenza A virus reproduction in MDCK cells. Here, we have synthesized previously undescribed nanocomposites that consist of TiO2 nanoparticles and polylysine conjugates with oligonucleotides that contain phosphoryl guanidine or phosphorothioate internucleotide groups. These nanocomposites have been shown to exhibit antiviral activity in MDCK cells infected with H5N1 influenza A virus. The nanocomposites containing phosphorothioate oligonucleotides inhibited virus replication ~130-fold. More potent inhibition, i.e., ~5000-fold or ~4600-fold, has been demonstrated by nanocomposites that contain phosphoryl guanidine or phosphodiester oligonucleotides, respectively. Free oligonucleotides have been nearly inactive. The antiviral activity of oligonucleotides of all three types, when delivered by Lipofectamine, has been significantly lower compared to the oligonucleotides delivered in the nanocomposites. In the former case, the phosphoryl guanidine oligonucleotide has appeared to be the most efficient; it has inhibited the virus replication by a factor of 400. The results make it possible to consider phosphoryl guanidine oligonucleotides, along with other oligonucleotide derivatives, as potential antiviral agents against H5N1 avian flu virus.


Antiviral Agents/pharmacology , Guanidines/chemistry , Influenza A Virus, H5N1 Subtype/drug effects , Nanocomposites/chemistry , Organophosphates/pharmacology , Phosphorothioate Oligonucleotides/pharmacology , Animals , Antiviral Agents/chemical synthesis , Dogs , Dose-Response Relationship, Drug , Drug Carriers , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/growth & development , Lipids/chemistry , Madin Darby Canine Kidney Cells , Oligonucleotides/chemistry , Oligonucleotides/pharmacology , Organophosphates/chemistry , Phosphorothioate Oligonucleotides/chemistry , Polylysine/chemistry , Titanium/chemistry , Virus Replication/drug effects
...