Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Antimicrob Agents Chemother ; 68(5): e0172723, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38587392

ABSTRACT

Antiviral susceptibility of influenza viruses was assessed using a high-content imaging-based neutralization test. Cap-dependent endonuclease inhibitors, baloxavir and AV5116, were superior to AV5115 against type A viruses, and AV5116 was most effective against PA mutants tested. However, these three inhibitors displayed comparable activity (EC50 8-22 nM) against type C viruses from six lineages. Banana lectin and a monoclonal antibody, YA3, targeting the hemagglutinin-esterase protein effectively neutralized some, but not all, type C viruses.


Subject(s)
Antiviral Agents , Dibenzothiepins , Triazines , Antiviral Agents/pharmacology , Humans , Triazines/pharmacology , Dibenzothiepins/pharmacology , Gammainfluenzavirus/drug effects , Gammainfluenzavirus/genetics , Morpholines/pharmacology , Pyridones/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Madin Darby Canine Kidney Cells , Dogs , Cyclopropanes/pharmacology , Influenza A virus/drug effects , Neutralization Tests , Pyridines/pharmacology
2.
Antiviral Res ; 218: 105719, 2023 10.
Article in English | MEDLINE | ID: mdl-37717821

ABSTRACT

Influenza virus neuraminidase (NA) can act as a receptor-binding protein, a role commonly attributed to hemagglutinin (HA). In influenza A(H3N2) viruses, three NA amino acid residues have previously been associated with NA-mediated hemagglutination: T148, D151, and more recently, H150. These residues are part of the 150-loop of the NA monomer. Substitutions at 148 and 151 arise from virus propagation in laboratory cell cultures, whereas changes at 150 occurred during virus evolution in the human host. In this study, we examined the effect of natural amino acid polymorphism at position 150 on NA-mediated hemagglutination. Using the A/Puerto Rico/8/34 backbone, we generated a comprehensive panel of recombinant A(H3N2) viruses that have different NAs but shared an HA that displays poor binding to red blood cells (RBCs). None of the tested substitutions at 150 (C, H, L, R, and S) promoted NA-binding. However, we identified two new determinants of NA-binding, Q136K and T439R, that emerged during virus culturing. Similar to T148I, both Q136K and T439R reduced NA enzyme activity by 48-86% and inhibition (14- to 173-fold) by the NA inhibitor zanamivir. NA-binding was observed when a virus preparation contained approximately 10% of NA variants with either T148I or T439R, highlighting the benefit of using deep sequencing in virus characterization. Taken together, our findings provide new insights into the molecular mechanisms underlying the ability of NA to function as a binding protein. Information gained may aid in the design of new and improved NA-targeting antivirals.


Subject(s)
Hemagglutination , Influenza A Virus, H3N2 Subtype , Neuraminidase , Humans , Amino Acids/pharmacology , Antiviral Agents/pharmacology , Neuraminidase/genetics , Neuraminidase/metabolism
3.
Materials (Basel) ; 16(17)2023 Aug 31.
Article in English | MEDLINE | ID: mdl-37687669

ABSTRACT

This work was undertaken to evaluate the influence of friction-stir welding (FSW) under a high-heat input condition on microstructural evolution. Given the extreme combination of deformation conditions associated with such an FSW regime (including the highest strain, temperature, and strain rate), it was expected to result in an unusual structural response. For this investigation, a commercial 6013 aluminum alloy was used as a program material, and FSW was conducted at a relatively high spindle rate of 1100 rpm and an extremely low feed rate of 13 mm/min; moreover, a Ti-6Al-4V backing plate was employed to reduce heat loss during welding. It was found that the high-heat-input FSW resulted in the formation of a pronounced fine-grained layer at the upper weld surface. This observation was attributed to the stirring action exerted by the shoulder of the FSW tool. Another important issue was the retardation of continuous recrystallization. This interesting phenomenon was explained in terms of a competition between recrystallization and recovery at high temperatures. Specifically, the activation of recovery should reduce dislocation density and thus retard the development of deformation-induced boundaries.

4.
Antiviral Res ; 217: 105679, 2023 09.
Article in English | MEDLINE | ID: mdl-37494978

ABSTRACT

Clade 2.3.4.4b highly pathogenic avian influenza (HPAI) A(H5N1) viruses that are responsible for devastating outbreaks in birds and mammals pose a potential threat to public health. Here, we evaluated their susceptibility to influenza antivirals. Of 1,015 sequences of HPAI A(H5N1) viruses collected in the United States during 2022, eight viruses (∼0.8%) had a molecular marker of drug resistance to an FDA-approved antiviral: three adamantane-resistant (M2-V27A), four oseltamivir-resistant (NA-H275Y), and one baloxavir-resistant (PA-I38T). Additionally, 31 viruses contained mutations that may reduce susceptibility to inhibitors of neuraminidase (NA) (n = 20) or cap-dependent endonuclease (CEN) (n = 11). A panel of 22 representative viruses was tested phenotypically. Overall, clade 2.3.4.4b A(H5N1) viruses lacking recognized resistance mutations were susceptible to FDA-approved antivirals. Oseltamivir was least potent at inhibiting NA activity, while the investigational NA inhibitor AV5080 was most potent, including against NA mutants. A novel NA substitution T438N conferred 12-fold reduced inhibition by zanamivir, and in combination with the known marker N295S, synergistically affected susceptibility to all five NA inhibitors. In cell culture-based assays HINT and IRINA, the PA-I38T virus displayed 75- to 108-fold and 37- to 78-fold reduced susceptibility to CEN inhibitors, baloxavir and the investigational AV5116, respectively. Viruses with PA-I38M or PA-A37T showed 5- to 10-fold reduced susceptibilities. As HPAI A(H5N1) viruses continue to circulate and evolve, close monitoring of drug susceptibility is needed for risk assessment and to inform decisions regarding antiviral stockpiling.


Subject(s)
Influenza A Virus, H5N1 Subtype , Influenza in Birds , Animals , United States/epidemiology , Antiviral Agents/pharmacology , Oseltamivir/pharmacology , Influenza A Virus, H5N1 Subtype/genetics , Influenza in Birds/epidemiology , Enzyme Inhibitors/pharmacology , Birds , Mammals , Drug Resistance, Viral/genetics , Neuraminidase
5.
Antiviral Res ; 208: 105457, 2022 12.
Article in English | MEDLINE | ID: mdl-36332755

ABSTRACT

Year-round virological characterization of circulating epidemic influenza viruses is conducted worldwide to detect the emergence of viruses that may escape pre-existing immunity or acquire resistance to antivirals. High throughput phenotypic assays are needed to complement the sequence-based analysis of circulating viruses and improve pandemic preparedness. The recent entry of a polymerase inhibitor, baloxavir, into the global market further highlighted this need. Here, we optimized a cell-based assay that considerably streamlines antiviral and antigenic testing by replacing lengthy immunostaining and imaging procedures used in current assay with measuring the enzymatic activity of nascent neuraminidase (NA) molecules expressed on the surface of virus-infected cells. For convenience, this new assay was named IRINA (Influenza Replication Inhibition Neuraminidase-based Assay). IRINA was successfully validated to assess inhibitory activity of baloxavir on virus replication by testing a large set (>150) of influenza A and B viruses, including drug resistant strains and viruses collected during 2017-2022. To test its versatility, IRINA was utilized to evaluate neutralization activity of a broadly reactive human anti-HA monoclonal antibody, FI6, and post-infection ferret antisera, as well as the inhibition of NA enzyme activity by NA inhibitors. Performance of IRINA was tested in parallel using respective conventional assays. IRINA offers an attractive alternative to current phenotypic assays, while maintaining reproducibility and high throughput capacity. Additionally, the improved turnaround time may prove to be advantageous when conducting time sensitive studies, such as investigating a new virus outbreak. This assay can meet the needs of surveillance laboratories by providing a streamlined and cost-effective approach for virus characterization.


Subject(s)
Influenza, Human , Neuraminidase , Animals , Humans , Reproducibility of Results , Drug Resistance, Viral , Ferrets , Virus Replication , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Enzyme Inhibitors/pharmacology , Oseltamivir/pharmacology
6.
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
7.
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
8.
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
9.
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
10.
Emerg Infect Dis ; 25(10): 1969-1972, 2019 10.
Article in English | MEDLINE | ID: mdl-31287050

ABSTRACT

Baloxavir showed broad-spectrum in vitro replication inhibition of 4 types of influenza viruses (90% effective concentration range 1.2-98.3 nmol/L); susceptibility pattern was influenza A ˃ B ˃ C ˃ D. This drug also inhibited influenza A viruses of avian and swine origin, including viruses that have pandemic potential and those resistant to neuraminidase inhibitors.


Subject(s)
Antiviral Agents/pharmacology , Gammainfluenzavirus/drug effects , Influenza A virus/drug effects , Influenza B virus/drug effects , Oxazines/pharmacology , Pyridines/pharmacology , Thiepins/pharmacology , Thogotovirus/drug effects , Triazines/pharmacology , Animals , Chickens/virology , Dibenzothiepins , Dogs , Humans , Influenza in Birds/drug therapy , Influenza in Birds/virology , Influenza, Human/drug therapy , Influenza, Human/virology , Madin Darby Canine Kidney Cells/virology , Microbial Sensitivity Tests , Morpholines , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/veterinary , Orthomyxoviridae Infections/virology , Pyridones , Swine/virology , Swine Diseases/drug therapy , Swine Diseases/virology
11.
Article in English | MEDLINE | ID: mdl-31187572

ABSTRACT

Mutations in the influenza virus neuraminidase (NA) that cause reduced susceptibility to the NA inhibitor (NAI) oseltamivir may occur naturally or following antiviral treatment. Currently, detection uses either a traditional NA inhibition assay or gene sequencing to identify known markers associated with reduced inhibition by oseltamivir. Both methods are laborious and require trained personnel. The influenza antiviral resistance test (iART), a prototype system developed by Becton, Dickinson and Company for research use only, offers a rapid and simple method to identify such viruses. This study investigated application of iART to influenza A viruses isolated from non-human hosts with a variety of NA subtypes (N1-N9).

12.
Sci Rep ; 9(1): 2676, 2019 02 25.
Article in English | MEDLINE | ID: mdl-30804469

ABSTRACT

Influenza A(H3N2) viruses evade human immunity primarily by acquiring antigenic changes in the haemagglutinin (HA). HA receptor-binding features of contemporary A(H3N2) viruses hinder traditional antigenic characterization using haemagglutination inhibition and promote selection of HA mutants. Thus, alternative approaches are needed to reliably assess antigenic relatedness between circulating viruses and vaccines. We developed a high content imaging-based neutralization test (HINT) to reduce antigenic mischaracterization resulting from virus adaptation to cell culture. Ferret reference antisera were raised using clinical specimens containing viruses representing recent vaccine strains. Analysis of viruses circulating during 2011-2018 showed that gain of an N158-linked glycosylation in HA was a molecular determinant of antigenic distancing between A/Hong Kong/4801/2014-like (clade 3C.2a) and A/Texas/50/2012-like viruses (clade 3C.1), while multiple evolutionary HA F193S substitution were linked to antigenic distancing from A/Switzerland/97152963/2013-like (clade 3C.3a) and further antigenic distancing from A/Texas/50/2012-like viruses. Additionally, a few viruses carrying HA T135K and/or I192T showed reduced neutralization by A/Hong Kong/4801/2014-like antiserum. Notably, this technique elucidated the antigenic characteristics of clinical specimens, enabling direct characterization of viruses produced in vivo, and eliminating in vitro culture, which rapidly alters the genotype/phenotype. HINT is a valuable new antigenic analysis tool for vaccine strain selection.


Subject(s)
Antibodies, Viral/immunology , Antigens, Viral/immunology , Hemagglutination Inhibition Tests/methods , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza, Human/immunology , Neutralization Tests/methods , Animals , Ferrets/immunology , Ferrets/virology , Glycosylation , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Humans , Immune Sera/immunology , Influenza A Virus, H3N2 Subtype/classification , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/physiology , Influenza Vaccines/immunology , Influenza, Human/diagnosis , Influenza, Human/virology , Phylogeny
13.
Euro Surveill ; 24(3)2019 Jan.
Article in English | MEDLINE | ID: mdl-30670144

ABSTRACT

The anti-influenza therapeutic baloxavir targets cap-dependent endonuclease activity of polymerase acidic (PA) protein. We monitored baloxavir susceptibility in the United States with next generation sequencing analysis supplemented by phenotypic one-cycle infection assay. Analysis of PA sequences of 6,891 influenza A and B viruses collected during 2016/17 and 2017/18 seasons showed amino acid substitutions: I38L (two A(H1N1)pdm09 viruses), E23G (two A(H1N1)pdm09 viruses) and I38M (one A(H3N2) virus); conferring 4-10-fold reduced susceptibility to baloxavir.


Subject(s)
Amino Acid Substitution/drug effects , Antiviral Agents/pharmacology , Drug Resistance, Viral/genetics , Enzyme Inhibitors/pharmacology , Influenza A Virus, H3N2 Subtype/drug effects , Influenza B virus/drug effects , Influenza, Human/drug therapy , Oxazines/pharmacology , Pyridines/pharmacology , Thiepins/pharmacology , Triazines/pharmacology , Amino Acid Substitution/genetics , Antiviral Agents/therapeutic use , Dibenzothiepins , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/isolation & purification , Influenza B virus/genetics , Influenza B virus/isolation & purification , Influenza, Human/epidemiology , Influenza, Human/virology , Microbial Sensitivity Tests , Morpholines , Pyridones , Seasons , Sentinel Surveillance , United States , Viral Proteins/genetics
14.
Euro Surveill ; 22(18)2017 May 04.
Article in English | MEDLINE | ID: mdl-28494845

ABSTRACT

A new rapid assay for detecting oseltamivir resistance in influenza virus, iART, was used to test 149 clinical specimens. Results were obtained for 132, with iART indicating 41 as 'resistant'. For these, sequence analysis found known and suspected markers of oseltamivir resistance, while no such markers were detected for the remaining 91 samples. Viruses isolated from the 41 specimens showed reduced or highly reduced inhibition by neuraminidase inhibition assay. iART may facilitate broader antiviral resistance testing.


Subject(s)
Antiviral Agents/pharmacology , Drug Resistance, Viral , Influenza A Virus, H1N1 Subtype/drug effects , Neuraminidase/antagonists & inhibitors , Oseltamivir/pharmacology , Antiviral Agents/therapeutic use , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza, Human , Microbial Sensitivity Tests/methods , Neuraminidase/genetics , Neuraminidase/metabolism , Neuraminidase/therapeutic use , Oseltamivir/therapeutic use
15.
Emerg Infect Dis ; 23(4): 686-690, 2017 04.
Article in English | MEDLINE | ID: mdl-28322707

ABSTRACT

In February 2016, three influenza B/Victoria/2/87 lineage viruses exhibiting 4- to 158-fold reduced inhibition by neuraminidase inhibitors were detected in Laos. These viruses had an H134N substitution in the neuraminidase and replicated efficiently in vitro and in ferrets. Current antiviral drugs may be ineffective in controlling infections caused by viruses harboring this mutation.


Subject(s)
Antiviral Agents/pharmacology , Drug Resistance, Viral/genetics , Influenza B virus/drug effects , Influenza, Human/epidemiology , Influenza, Human/virology , Neuraminidase/genetics , Adolescent , Adult , Aged , Amino Acid Substitution , Child , Child, Preschool , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Viral , Humans , Infant , Influenza B virus/genetics , Laos/epidemiology , Male , Middle Aged , Young Adult
16.
Anal Chem ; 89(5): 3130-3137, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28192976

ABSTRACT

Mounting evidence suggests that neuraminidase's functionality extends beyond its classical role in influenza virus infection and that antineuraminidase antibodies offer protective immunity. Therefore, a renewed interest in the development of neuraminidase (NA)-specific methods to characterize the glycoprotein and evaluate potential advantages for NA standardization in influenza vaccines has emerged. NA displays sialidase activity by cleaving off the terminal N-acetylneuraminic acid on α-2,3 or α-2,6 sialic acid containing receptors of host cells. The type and distribution of these sialic acid containing receptors is considered to be an important factor in transmission efficiency of influenza viruses between and among host species. Changes in hemagglutinin (HA) binding and NA specificity in reassortant viruses may be related to the emergence of new and potentially dangerous strains of influenza. Current methods to investigate neuraminidase activity use small derivatized sugars that are poor models for natural glycoprotein receptors and do not provide information on the linkage specificity. Here, a novel approach for rapid and accurate quantification of influenza neuraminidase activity is achieved utilizing ultra-high performance liquid chromatography (UPLC) and isotope dilution mass spectrometry (IDMS). Direct LC-MS/MS quantification of NA-released sialic acid provides precise measurement of influenza neuraminidase activity over a range of substrates. The method provides exceptional sensitivity and specificity with a limit of detection of 0.38 µM for sialic acid and the capacity to obtain accurate measurements of specific enzyme activity preference toward α-2,3-sialyllactose linkages, α-2,6-sialyllactose linkages, or whole glycosylated proteins such as fetuin.


Subject(s)
Chromatography, High Pressure Liquid , Influenza A Virus, H1N1 Subtype/enzymology , Neuraminidase/metabolism , Tandem Mass Spectrometry , Viral Proteins/metabolism , Carbon Isotopes/chemistry , Humans , Influenza Vaccines/analysis , Influenza Vaccines/metabolism , Kinetics , Lactose/analogs & derivatives , Lactose/analysis , Substrate Specificity
17.
J Clin Microbiol ; 55(1): 145-154, 2017 01.
Article in English | MEDLINE | ID: mdl-27795346

ABSTRACT

The rapid evolution of influenza A(H3N2) viruses necessitates close monitoring of their antigenic properties so the emergence and spread of antigenic drift variants can be rapidly identified. Changes in hemagglutinin (HA) acquired by contemporary A(H3N2) viruses hinder antigenic characterization by traditional methods, thus complicating vaccine strain selection. Sequence-based approaches have been used to infer virus antigenicity; however, they are time consuming and mid-throughput. To facilitate virological surveillance and epidemiological studies, we developed and validated a pyrosequencing approach that enables identification of six HA clades of contemporary A(H3N2) viruses. The identification scheme of viruses of the H3 clades 3C.2, 3C.2a, 3C.2b, 3C.3, 3C.3a, and 3C.3b is based on the interrogation of five single nucleotide polymorphisms (SNPs) within three neighboring HA regions, namely 412 to 431, 465 to 481, and 559 to 571. Two bioinformatics tools, IdentiFire (Qiagen) and FireComb (developed in-house), were utilized to expedite pyrosequencing data analysis. The assay's analytical sensitivity was 10 focus forming units, and respiratory specimens with threshold cycle (CT) values of <34 typically produced good quality pyrograms. When applied to 120 A(H3N2) virus isolates and 27 respiratory specimens, the assay displayed 100% agreement with clades determined by HA sequencing coupled with phylogenetics. The multi-SNP analysis described here was readily adopted by another laboratory with pyrosequencing capabilities. The implementation of this approach enhanced the findings from virological surveillance and epidemiological studies between 2013 and 2016, which examined more than 3,000 A(H3N2) viruses.


Subject(s)
Genetic Drift , Genotyping Techniques/methods , High-Throughput Nucleotide Sequencing , Influenza A Virus, H3N2 Subtype/classification , Influenza A Virus, H3N2 Subtype/genetics , Influenza, Human/virology , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Influenza A Virus, H3N2 Subtype/isolation & purification , Polymorphism, Single Nucleotide , Sensitivity and Specificity
18.
J Virol ; 90(23): 10446-10458, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27630240

ABSTRACT

The pandemic threat posed by emerging zoonotic influenza A viruses necessitates development of antiviral agents effective against various antigenic subtypes. Human monoclonal antibody (hMAb) targeting the hemagglutinin (HA) stalk offers a promising approach to control influenza virus infections. Here, we investigated the ability of the hMAb 81.39a to inhibit in vitro replication of human and zoonotic viruses, representing 16 HA subtypes. The majority of viruses were effectively neutralized by 81.39a at a 50% effective concentration (EC50) of <0.01 to 4.9 µg/ml. Among group 2 HA viruses tested, a single A(H7N9) virus was not neutralized at 50 µg/ml; it contained HA2-Asp19Gly, an amino acid position previously associated with resistance to neutralization by the group 2 HA-neutralizing MAb CR8020. Notably, among group 1 HA viruses, H11-H13 and H16 subtypes were not neutralized at 50 µg/ml; they shared the substitution HA2-Asp19Asn/Ala. Conversely, H9 viruses harboring HA2-Asp19Ala were fully susceptible to neutralization. Therefore, amino acid variance at HA2-Asp19 has subtype-specific adverse effects on in vitro neutralization. Mice given a single injection (15 or 45 mg/kg of body weight) at 24 or 48 h after infection with recently emerged A(H5N2), A(H5N8), A(H6N1), or A(H7N9) viruses were protected from mortality and showed drastically reduced lung viral titers. Furthermore, 81.39a protected mice infected with A(H7N9) harboring HA2-Asp19Gly, although the antiviral effect was lessened. A(H1N1)pdm09-infected ferrets receiving a single dose (25 mg/kg) had reduced viral titers and showed less lung tissue injury, despite 24- to 72-h-delayed treatment. Taken together, this study provides experimental evidence for the therapeutic potential of 81.39a against diverse influenza A viruses. IMPORTANCE: Zoonotic influenza viruses, such as A(H5N1) and A(H7N9) subtypes, have caused severe disease and deaths in humans, raising public health concerns. Development of novel anti-influenza therapeutics with a broad spectrum of activity against various subtypes is necessary to mitigate disease severity. Here, we demonstrate that the hemagglutinin (HA) stalk-targeting human monoclonal antibody 81.39a effectively neutralized the majority of influenza A viruses tested, representing 16 HA subtypes. Furthermore, delayed treatment with 81.39a significantly suppressed virus replication in the lungs, prevented dramatic body weight loss, and increased survival rates of mice infected with A(H5Nx), A(H6N1), or A(H7N9) viruses. When tested in ferrets, delayed 81.39a treatment reduced viral titers, particularly in the lower respiratory tract, and substantially alleviated disease symptoms associated with severe A(H1N1)pdm09 influenza. Collectively, our data demonstrated the effectiveness of 81.39a against both seasonal and emerging influenza A viruses.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A virus/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/therapeutic use , Antibodies, Viral/therapeutic use , Antigenic Variation/genetics , Antigenic Variation/immunology , Female , Ferrets , Hemagglutinin Glycoproteins, Influenza Virus/classification , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , In Vitro Techniques , Influenza A virus/classification , Influenza A virus/genetics , Influenza, Human/immunology , Influenza, Human/virology , Lung/pathology , Lung/virology , Male , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/therapy , Orthomyxoviridae Infections/virology , Treatment Outcome
19.
J Infect Dis ; 214(7): 1010-9, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27190176

ABSTRACT

BACKGROUND: During the 2014-2015 US influenza season, expanded genetic characterization of circulating influenza A(H3N2) viruses was used to assess the impact of the genetic variability of influenza A(H3N2) viruses on influenza vaccine effectiveness (VE). METHODS: A novel pyrosequencing assay was used to determine genetic group, based on hemagglutinin (HA) gene sequences, of influenza A(H3N2) viruses from patients enrolled at US Influenza Vaccine Effectiveness Network sites. VE was estimated using a test-negative design comparing vaccination among patients infected with influenza A(H3N2) viruses and uninfected patients. RESULTS: Among 9710 enrollees, 1868 (19%) tested positive for influenza A(H3N2) virus; genetic characterization of 1397 viruses showed that 1134 (81%) belonged to 1 HA genetic group (3C.2a) of antigenically drifted influenza A(H3N2) viruses. Effectiveness of 2014-2015 influenza vaccination varied by influenza A(H3N2) virus genetic group from 1% (95% confidence interval [CI], -14% to 14%) against illness caused by antigenically drifted influenza A(H3N2) virus group 3C.2a viruses versus 44% (95% CI, 16%-63%) against illness caused by vaccine-like influenza A(H3N2) virus group 3C.3b viruses. CONCLUSIONS: Effectiveness of 2014-2015 influenza vaccination varied by genetic group of influenza A(H3N2) virus. Changes in HA genes related to antigenic drift were associated with reduced VE.


Subject(s)
Genetic Variation , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Influenza, Human/virology , Adolescent , Adult , Aged , Aged, 80 and over , Child, Preschool , Female , Genetic Drift , Hemagglutinin Glycoproteins, Influenza Virus/genetics , High-Throughput Nucleotide Sequencing , Humans , Infant , Influenza A Virus, H3N2 Subtype/classification , Influenza Vaccines/administration & dosage , Male , Middle Aged , Treatment Outcome , United States , Young Adult
20.
J Virol ; 90(12): 5770-5784, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27053557

ABSTRACT

UNLABELLED: During 2014, a subclade 2.3.4.4 highly pathogenic avian influenza (HPAI) A(H5N8) virus caused poultry outbreaks around the world. In late 2014/early 2015, the virus was detected in wild birds in Canada and the United States, and these viruses also gave rise to reassortant progeny, composed of viral RNA segments (vRNAs) from both Eurasian and North American lineages. In particular, viruses were found with N1, N2, and N8 neuraminidase vRNAs, and these are collectively referred to as H5Nx viruses. In the United States, more than 48 million domestic birds have been affected. Here we present a detailed structural and biochemical analysis of the surface antigens of H5N1, H5N2, and H5N8 viruses in addition to those of a recent human H5N6 virus. Our results with recombinant hemagglutinin reveal that these viruses have a strict avian receptor binding preference, while recombinantly expressed neuraminidases are sensitive to FDA-approved and investigational antivirals. Although H5Nx viruses currently pose a low risk to humans, it is important to maintain surveillance of these circulating viruses and to continually assess future changes that may increase their pandemic potential. IMPORTANCE: The H5Nx viruses emerging in North America, Europe, and Asia pose a great public health concern. Here we report a molecular and structural study of the major surface proteins of several H5Nx influenza viruses. Our results improve the understanding of these new viruses and provide important information on their receptor preferences and susceptibilities to antivirals, which are central to pandemic risk assessment.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Influenza A Virus, H5N1 Subtype/chemistry , Influenza A Virus, H5N2 Subtype/chemistry , Influenza A Virus, H5N8 Subtype/chemistry , Neuraminidase/chemistry , Neuraminidase/metabolism , Animals , Animals, Wild/virology , Asia/epidemiology , Canada/epidemiology , Disease Outbreaks , Europe/epidemiology , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Influenza A Virus, H5N1 Subtype/enzymology , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N2 Subtype/enzymology , Influenza A Virus, H5N2 Subtype/genetics , Influenza A Virus, H5N8 Subtype/enzymology , Influenza A Virus, H5N8 Subtype/genetics , Influenza in Birds/epidemiology , Influenza in Birds/virology , Influenza, Human/virology , Neuraminidase/genetics , North America/epidemiology , Phylogeny , Poultry , Reassortant Viruses
SELECTION OF CITATIONS
SEARCH DETAIL
...