Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
2.
PLoS One ; 16(7): e0254632, 2021.
Article in English | MEDLINE | ID: mdl-34280220

ABSTRACT

Superoxide radicals and other reactive oxygen species (ROS) are implicated in influenza A virus-induced inflammation. In this in vitro study, we evaluated the effects of TG6-44, a novel quinazolin-derived myeloperoxidase-specific ROS inhibitor, on influenza A virus (A/X31) infection using THP-1 lung monocytic cells and freshly isolated peripheral blood mononuclear cells (PBMC). TG6-44 significantly decreased A/X31-induced ROS and virus-induced inflammatory mediators in THP-1 cells (IL-6, IFN-γ, MCP-1, TNF-α, MIP-1ß) and in human PBMC (IL-6, IL-8, TNF-α, MCP-1). Interestingly, TG6-44-treated THP-1 cells showed a decrease in percent cells expressing viral nucleoprotein, as well as a delay in translocation of viral nucleoprotein into the nucleus. Furthermore, in influenza A virus-infected cells, TG6-44 treatment led to suppression of virus-induced cell death as evidenced by decreased caspase-3 activation, decreased proportion of Annexin V+PI+ cells, and increased Bcl-2 phosphorylation. Taken together, our results demonstrate the anti-inflammatory and anti-infective effects of TG6-44.


Subject(s)
Inflammation Mediators/pharmacology , Inflammation/drug therapy , Influenza A virus/drug effects , Peroxidase/antagonists & inhibitors , Reactive Oxygen Species/antagonists & inhibitors , Cell Survival/drug effects , Chemokine CCL2/genetics , Gene Expression Regulation/drug effects , Humans , Inflammation/pathology , Inflammation/virology , Influenza A virus/pathogenicity , Interleukin-6/genetics , Interleukin-8/genetics , Leukocytes, Mononuclear/drug effects , Lung/drug effects , Lung/pathology , Peroxidase/genetics , Quinazolines/pharmacology , Reactive Oxygen Species/metabolism , Superoxides/metabolism , Tumor Necrosis Factor-alpha/genetics
3.
Emerg Infect Dis ; 27(7): 1953-1957, 2021 07.
Article in English | MEDLINE | ID: mdl-34152954

ABSTRACT

Four cases of oseltamivir-resistant influenza A(H1N1)pdm09 virus infection were detected among inhabitants of a border detention center in Texas, USA. Hemagglutinin of these viruses belongs to 6B.1A5A-156K subclade, which may enable viral escape from preexisting immunity. Our finding highlights the necessity to monitor both drug resistance and antigenic drift of circulating viruses.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza, Human , Antiviral Agents/therapeutic use , Drug Resistance, Viral , Hemagglutinins , Humans , Influenza, Human/drug therapy , Neuraminidase , Oseltamivir/therapeutic use , Texas , Viral Proteins
4.
Antiviral Res ; 188: 105035, 2021 04.
Article in English | MEDLINE | ID: mdl-33581212

ABSTRACT

Pimodivir exerts an antiviral effect on the early stages of influenza A virus replication by inhibiting the cap-binding function of polymerase basic protein 2 (PB2). In this study, we used a combination of sequence analysis and phenotypic methods to evaluate pimodivir susceptibility of influenza A viruses collected from humans and other hosts. Screening PB2 sequences for substitutions previously associated with reduced pimodivir susceptibility revealed a very low frequency among seasonal viruses circulating in the U.S. during 2015-2020 (<0.03%; 3/11,934) and among non-seasonal viruses collected in various countries during the same period (0.2%; 18/8971). Pimodivir potently inhibited virus replication in two assays, a single-cycle HINT and a multi-cycle FRA, with IC50 values in a nanomolar range. Median IC50 values determined by HINT were similar for both subtypes of seasonal viruses, A(H1N1)pdm09 and A(H3N2), across three seasons. Human seasonal viruses with PB2 substitutions S324C, S324R, or N510K displayed a 27-317-fold reduced pimodivir susceptibility by HINT. In addition, pimodivir was effective at inhibiting replication of a diverse group of animal-origin viruses that have pandemic potential, including avian viruses of A(H5N6) and A(H7N9) subtypes. A rare PB2 substitution H357N was identified in an A(H4N2) subtype poultry virus that displayed >100-fold reduced pimodivir susceptibility. Our findings demonstrate a broad inhibitory activity of pimodivir and expand the existing knowledge of amino acid substitutions that can reduce susceptibility to this investigational antiviral.


Subject(s)
Antiviral Agents/pharmacology , Influenza A virus/drug effects , Pyridines/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Viral Proteins/antagonists & inhibitors , Animals , Drug Resistance, Viral , Enzyme Inhibitors/pharmacology , Humans , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H7N9 Subtype/drug effects , Influenza A Virus, H7N9 Subtype/genetics , Influenza A virus/genetics , Influenza, Human/virology , Microbial Sensitivity Tests , Orthomyxoviridae Infections/virology , RNA-Dependent RNA Polymerase/genetics , Viral Proteins/genetics , Virus Replication/drug effects
5.
Antiviral Res ; 182: 104906, 2020 10.
Article in English | MEDLINE | ID: mdl-32798601

ABSTRACT

Baloxavir, a new antiviral drug targeting cap-dependent endonuclease activity of polymerase acidic (PA) protein of influenza viruses, is now approved in multiple countries. Several substitutions at isoleucine 38 in PA protein (e.g., PA-I38T) have been associated with decreased baloxavir susceptibility in vitro and in vivo. In recent years, next generation sequencing (NGS) analysis and pyrosequencing have been used by CDC and U.S. Public Health Laboratories to monitor drug susceptibility of influenza viruses. Here we described an improved pyrosequencing assay for detecting influenza A viruses carrying substitutions at PA-38. Cyclic and customized orders of nucleotide dispensation were evaluated, and pyrosequencing results were compared to those generated using NGS. Our data showed that the customized nucleotide dispensation has improved the pyrosequencing assay performance in identification of double mixtures (e.g., PA-38I/T); however, identification of PA-38 variants in triple mixtures remains a challenge. While NGS analysis indicated the presence of PA-I38K in one clinical specimen and isolate, our attempts to detect this mutation by pyrosequencing or recover the virus carrying PA-I38K in cell culture were unsuccessful, raising a possibility of a rarely occurring sequencing error. Overall, pyrosequencing provides a convenient means to detect baloxavir resistant influenza viruses when NGS is unavailable or a faster turnaround time is required.


Subject(s)
Antiviral Agents/pharmacology , Dibenzothiepins/pharmacology , Drug Resistance, Viral/genetics , Influenza A virus/drug effects , Influenza A virus/genetics , Morpholines/pharmacology , Pyridones/pharmacology , Triazines/pharmacology , Amino Acid Substitution , Animals , Dogs , Genome, Viral , High-Throughput Nucleotide Sequencing , Influenza A virus/classification , Madin Darby Canine Kidney Cells , Virus Replication/drug effects
6.
J Infect Dis ; 221(3): 367-371, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31541547

ABSTRACT

Susceptibility of influenza A viruses to baloxavir can be affected by changes at amino acid residue 38 in the polymerase acidic (PA) protein. Information on replicative fitness of PA-I38-substituted viruses remains sparse. We demonstrated that substitutions I38L/M/S/T not only had a differential effect on baloxavir susceptibility (9- to 116-fold) but also on in vitro replicative fitness. Although I38L conferred undiminished growth, other substitutions led to mild attenuation. In a ferret model, control viruses outcompeted those carrying I38M or I38T substitutions, although their advantage was limited. These findings offer insights into the attributes of baloxavir-resistant viruses needed for informed risk assessment.


Subject(s)
Antiviral Agents/therapeutic use , Drug Resistance, Viral/genetics , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H3N2 Subtype/genetics , Orthomyxoviridae Infections/drug therapy , Oxazines/therapeutic use , Pyridines/therapeutic use , Thiepins/therapeutic use , Triazines/therapeutic use , Virus Replication/genetics , Amino Acid Substitution , Animals , Dibenzothiepins , Disease Models, Animal , Dogs , Ferrets , High-Throughput Nucleotide Sequencing , Madin Darby Canine Kidney Cells , Male , Microbial Sensitivity Tests , Morpholines , Orthomyxoviridae Infections/virology , Pyridones , RNA-Dependent RNA Polymerase/genetics , Seasons , Treatment Outcome , Viral Proteins/genetics
7.
Emerg Infect Dis ; 23(12)2017 12.
Article in English | MEDLINE | ID: mdl-29148400
8.
Nanomedicine ; 12(7): 1909-1917, 2016 10.
Article in English | MEDLINE | ID: mdl-27112307

ABSTRACT

To enhance the immunogenicity of the Influenza H5N1 vaccine, we developed an oil-in-water nanoemulsion (NE) adjuvant. NE displayed good temperature stability and maintained particle size. More importantly, it significantly enhanced IL-6 and MCP-1 production to recruit innate cells, including neutrophils, monocytes/macrophages and dendritic cells to the local environment. Furthermore, NE enhanced dendritic cell function to induce robust antigen-specific T and B cell immune responses. NE-adjuvanted H5N1 vaccine not only elicited significantly higher and long-lasting antibody responses, but also conferred enhanced protection against homologous clade 1 as well as heterologous clade 2 H5N1 virus challenge in young as well as in aged mice. The pre-existing immunity to seasonal influenza did not affect the immunogenicity of NE-adjuvanted H5N1 vaccine.


Subject(s)
Influenza A Virus, H5N1 Subtype , Influenza Vaccines/administration & dosage , Nanoparticles/chemistry , Adjuvants, Immunologic , Animals , Antibodies, Viral , Emulsions , Humans , Influenza, Human/prevention & control , Mice
9.
PLoS One ; 11(2): e0149864, 2016.
Article in English | MEDLINE | ID: mdl-26910342

ABSTRACT

The role of the reactive oxygen species-producing NADPH oxidase family of enzymes in the pathology of influenza A virus infection remains enigmatic. Previous reports implicated NADPH oxidase 2 in influenza A virus-induced inflammation. In contrast, NADPH oxidase 1 (Nox1) was reported to decrease inflammation in mice within 7 days post-influenza A virus infection. However, the effect of NADPH oxidase 1 on lethality and adaptive immunity after influenza A virus challenge has not been explored. Here we report improved survival and decreased morbidity in mice with catalytically inactive NADPH oxidase 1 (Nox1*/Y) compared with controls after challenge with A/PR/8/34 influenza A virus. While changes in lung inflammation were not obvious between Nox1*/Y and control mice, we observed alterations in the T cell response to influenza A virus by day 15 post-infection, including increased interleukin-7 receptor-expressing virus-specific CD8+ T cells in lungs and draining lymph nodes of Nox1*/Y, and increased cytokine-producing T cells in lungs and spleen. Furthermore, a greater percentage of conventional and interstitial dendritic cells from Nox1*/Y draining lymph nodes expressed the co-stimulatory ligand CD40 within 6 days post-infection. Results indicate that NADPH oxidase 1 modulates the innate and adaptive cellular immune response to influenza virus infection, while also playing a role in host survival. Results suggest that NADPH oxidase 1 inhibitors may be beneficial as adjunct therapeutics during acute influenza infection.


Subject(s)
Adaptive Immunity , CD8-Positive T-Lymphocytes/immunology , Immunity, Innate , Influenza A virus/immunology , NADH, NADPH Oxidoreductases/immunology , Orthomyxoviridae Infections/immunology , Animals , CD40 Ligand/genetics , CD40 Ligand/immunology , Dendritic Cells/immunology , Male , Mice , Mice, Transgenic , NADH, NADPH Oxidoreductases/genetics , NADPH Oxidase 1 , Orthomyxoviridae Infections/genetics
10.
J Infect Dis ; 212(12): 1923-9, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26068782

ABSTRACT

We compared the innate immune response to a newly emerged swine-origin influenza A(H3N2) variant containing the M gene from 2009 pandemic influenza A(H1N1), termed "A(H3N2)vpM," to the immune responses to the 2010 swine-origin influenza A(H3N2) variant and seasonal influenza A(H3N2). Our results demonstrated that A(H3N2)vpM-induced myeloid dendritic cells secreted significantly lower levels of type I interferon (IFN) but produced significantly higher levels of proinflammatory cytokines and induced potent inflammasome activation. The reduction in antiviral immunity with increased inflammatory responses upon A(H3N2)vpM infection suggest that these viruses have the potential for increased disease severity in susceptible hosts.


Subject(s)
Inflammasomes/metabolism , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Leukocytes, Mononuclear/immunology , Animals , Cell Line , Cytokines/metabolism , Dendritic Cells/immunology , Humans , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza A Virus, H3N2 Subtype/isolation & purification , Orthomyxoviridae Infections/virology , Swine , Swine Diseases/virology
11.
J Virol ; 89(10): 5419-26, 2015 May.
Article in English | MEDLINE | ID: mdl-25740997

ABSTRACT

UNLABELLED: Human infections by avian influenza A(H7N9) virus entail substantial morbidity and mortality. Treatment of infected patients with the neuraminidase (NA) inhibitor oseltamivir was associated with emergence of viruses carrying NA substitutions. In the NA inhibition (NI) assay, R292K conferred highly reduced inhibition by oseltamivir, while E119V and I222K each caused reduced inhibition. To facilitate establishment of laboratory correlates of clinically relevant resistance, experiments were conducted in ferrets infected with virus carrying wild-type or variant NA genes recovered from the A/Taiwan/1/2013 isolate. Oseltamivir treatment (5 or 25 mg/kg of body weight/dose) was given 4 h postinfection, followed by twice-daily treatment for 5 days. Treatment of ferrets infected with wild-type virus resulted in a modest dose-dependent reduction (0.7 to 1.5 log10 50% tissue culture infectious dose [TCID50]) in nasal wash viral titers and inflammation response. Conversely, treatment failed to significantly inhibit the replication of R292K or E119V virus. A small reduction of viral titers was detected on day 5 in ferrets infected with the I222K virus. The propensity for oseltamivir resistance emergence was assessed in oseltamivir-treated animals infected with wild-type virus; emergence of R292K virus was detected in 3 of 6 ferrets within 5 to 7 days postinfection. Collectively, we demonstrate that R292K, E119V, and I222K reduced the inhibitory activity of oseltamivir, not only in the NI assay, but also in infected ferrets, judged particularly by viral loads in nasal washes, and may signal the need for alternative therapeutics. Thus, these clinical outcomes measured in the ferret model may correlate with clinically relevant oseltamivir resistance in humans. IMPORTANCE: This report provides more evidence for using the ferret model to assess the susceptibility of influenza A(H7N9) viruses to oseltamivir, the most prescribed anti-influenza virus drug. The information gained can be used to assist in the establishment of laboratory correlates of human disease and drug therapy. The rapid emergence of viruses with R292K in treated ferrets correlates well with the multiple reports on this NA variant in treated human patients. Our findings highlight the importance of the discovery and characterization of new antiviral drugs with different mechanisms of action and the use of combination treatment strategies against emerging viruses with pandemic potential, such as avian H7N9 virus, particularly against those carrying drug resistance markers.


Subject(s)
Antiviral Agents/pharmacology , Influenza A Virus, H7N9 Subtype/drug effects , Influenza A Virus, H7N9 Subtype/genetics , Influenza, Human/drug therapy , Influenza, Human/virology , Neuraminidase/genetics , Oseltamivir/pharmacology , Animals , Disease Models, Animal , Drug Resistance, Viral/genetics , Enzyme Inhibitors/pharmacology , Ferrets , Genes, Viral , Humans , Influenza A Virus, H7N9 Subtype/physiology , Male , Mutation , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/virology , Respiratory Tract Infections/drug therapy , Respiratory Tract Infections/pathology , Respiratory Tract Infections/virology , Virus Replication/drug effects
12.
J Infect Dis ; 211(2): 249-57, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25124927

ABSTRACT

BACKGROUND: Patients contracting influenza A(H7N9) infection often developed severe disease causing respiratory failure. Neuraminidase (NA) inhibitors (NAIs) are the primary option for treatment, but information on drug-resistance markers for influenza A(H7N9) is limited. METHODS: Four NA variants of A/Taiwan/1/2013(H7N9) virus containing a single substitution (NA-E119V, NA-I222K, NA-I222R, or NA-R292K) recovered from an oseltamivir-treated patient were tested for NAI susceptibility in vitro; their replicative fitness was evaluated in cell culture, mice, and ferrets. RESULTS: NA-R292K led to highly reduced inhibition by oseltamivir and peramivir, while NA-E119V, NA-I222K, and NA-I222R caused reduced inhibition by oseltamivir. Mice infected with any virus showed severe clinical signs with high mortality rates. NA-I222K virus was the most virulent in mice, whereas virus lacking NA change (NA-WT) and NA-R292K virus seemed the least virulent. Sequence analysis suggests that PB2-S714N increased virulence of NA-I222K virus in mice; NS1-K126R, alone or in combination with PB2-V227M, produced contrasting effects in NA-WT and NA-R292K viruses. In ferrets, all viruses replicated to high titers in the upper respiratory tract but produced only mild illness. NA-R292K virus, showed reduced replicative fitness in this animal model. CONCLUSIONS: Our data highlight challenges in assessment of the replicative fitness of H7N9 NA variants that emerged in NAI-treated patients.


Subject(s)
Antiviral Agents/therapeutic use , Drug Resistance, Viral , Influenza A Virus, H7N9 Subtype/drug effects , Influenza, Human/drug therapy , Influenza, Human/virology , Oseltamivir/therapeutic use , Animals , Disease Models, Animal , Ferrets , Humans , Influenza A Virus, H7N9 Subtype/enzymology , Influenza A Virus, H7N9 Subtype/genetics , Influenza A Virus, H7N9 Subtype/isolation & purification , Mice, Inbred BALB C , Microbial Sensitivity Tests , Mutant Proteins/genetics , Mutation, Missense , Neuraminidase/genetics , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Viral Proteins/genetics , Virus Cultivation , Virus Replication
13.
J Infect Dis ; 210(3): 435-40, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24569063

ABSTRACT

Human infections caused by avian influenza A virus type subtype H7N9 have been associated with substantial morbidity and mortality. Emergence of virus variants carrying markers of decreased susceptibility to neuraminidase inhibitors was reported. Here we show that DAS181 (Fludase), an antiviral drug with sialidase activity, potently inhibited replication of wild-type influenza A(H7N9) and its oseltamivir-resistant R292K variants in mice. A once-daily administration initiated early after lethal infection hampered body weight loss and completely protected mice from lethality. We observed a time-dependent effect for 24-72-hour delayed DAS181 treatments on morbidity and mortality. The results warrant further investigation of DAS181 for influenza treatment.


Subject(s)
Influenza A Virus, H7N9 Subtype/drug effects , Orthomyxoviridae Infections/drug therapy , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Virus Replication/physiology , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Drug Resistance, Viral , Genetic Variation , Influenza A Virus, H7N9 Subtype/physiology , Mice , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/virology , Zoonoses
14.
J Immunol ; 189(5): 2257-65, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22855715

ABSTRACT

Myeloid dendritic cells (mDCs) have long been thought to function as classical APCs for T cell responses. However, we demonstrate that influenza viruses induce rapid differentiation of human monocytes into mDCs. Unlike the classic mDCs, the virus-induced mDCs failed to upregulate DC maturation markers and were unable to induce allogeneic lymphoproliferation. Virus-induced mDCs secreted little, if any, proinflammatory cytokines; however, they secreted a substantial amount of chemoattractants for monocytes (MCP-1 and IP-10). Interestingly, the differentiated mDCs secreted type I IFN and upregulated the expression of IFN-stimulated genes (tetherin, IFITM3, and viperin), as well as cytosolic viral RNA sensors (RIG-I and MDA5). Additionally, culture supernatants from virus-induced mDCs suppressed the replication of virus in vitro. Furthermore, depletion of monocytes in a mouse model of influenza infection caused significant reduction of lung mDC numbers, as well as type I IFN production in the lung. Consequently, increased lung virus titer and higher mortality were observed. Taken together, our results demonstrate that the host responds to influenza virus infection by initiating rapid differentiation of circulating monocytes into IFN-producing mDCs, which contribute to innate antiviral immune responses.


Subject(s)
Cell Differentiation/immunology , Dendritic Cells/immunology , Interferon Type I/biosynthesis , Monocytes/immunology , Myeloid Cells/immunology , Orthomyxoviridae Infections/prevention & control , Animals , Cells, Cultured , Dendritic Cells/pathology , Dendritic Cells/virology , Female , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza, Human/immunology , Influenza, Human/pathology , Influenza, Human/prevention & control , Interferon Type I/physiology , Mice , Mice, Inbred C57BL , Monocytes/metabolism , Monocytes/pathology , Myeloid Cells/pathology , Myeloid Cells/virology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/pathology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...