Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Virus Res ; 304: 198545, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34391827

ABSTRACT

The influenza A virus genome contains 8 gene segments encoding 10 commonly recognized proteins. Additional protein products have been identified, including PB1-F2 and PA-X. We report the in-silico identification of novel isoforms of PB1-F2 and PA-X in influenza virus genomes sequenced from avian samples. The isoform observed in PA-X includes a mutated stop codon that should extend the protein product by 8 amino acids. The isoform observed in PB1-F2 includes two nonsense mutations that should truncate the N-terminal region of the protein product and remove the entire mitochondrial targeting domain. Both isoforms were uncovered during automatic annotation of CEIRS sequence data. Nominally termed PA-X8 and PB1-F2-Cterm, both predicted isoforms were subsequently found in other annotated influenza genomes previously deposited in GenBank. Both isoforms were noticed due to discrepant annotations output by two annotation engines, indicating a benefit of incorporating multiple algorithms during gene annotation.


Subject(s)
Influenza A virus , Influenza, Human , Base Sequence , Humans , Influenza A virus/genetics , Influenza A virus/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Viral Proteins/metabolism
2.
Sci Rep ; 6: 36216, 2016 10 31.
Article in English | MEDLINE | ID: mdl-27796371

ABSTRACT

Since the emergence of human H3N2 influenza A viruses in the pandemic of 1968, these viruses have become established as strains of moderate severity. A decline in virulence has been accompanied by glycan accumulation on the hemagglutinin globular head, and hemagglutinin receptor binding has changed from recognition of a broad spectrum of glycan receptors to a narrower spectrum. The relationship between increased glycosylation, binding changes, and reduction in H3N2 virulence is not clear. We evaluated the effect of hemagglutinin glycosylation on receptor binding and virulence of engineered H3N2 viruses. We demonstrate that low-binding virus is as virulent as higher binding counterparts, suggesting that H3N2 infection does not require either recognition of a wide variety of, or high avidity binding to, receptors. Among the few glycans recognized with low-binding virus, there were two structures that were bound by the vast majority of H3N2 viruses isolated between 1968 and 2012. We suggest that these two structures support physiologically relevant binding of H3N2 hemagglutinin and that this physiologically relevant binding has not changed since the 1968 pandemic. Therefore binding changes did not contribute to reduced severity of seasonal H3N2 viruses. This work will help direct the search for factors enhancing influenza virulence.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus , Influenza A Virus, H3N2 Subtype , Virus Attachment , A549 Cells , Animals , Chlorocebus aethiops , Dogs , Glycosylation , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Humans , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/metabolism , Influenza A Virus, H3N2 Subtype/pathogenicity , Madin Darby Canine Kidney Cells , Vero Cells
3.
Emerg Microbes Infect ; 4(7): e40, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26251829

ABSTRACT

Influenza A viruses of the H1N1 subtype have emerged from the avian influenza gene pool in aquatic birds and caused human pandemics at least twice during the past century. Despite this fact, surprisingly little is known about the H1N1 gene pool in the aquatic bird reservoir. A preliminary study showed that an H1N1 virus from a shorebird of the Charadriiformes order was transmitted between animals through the airborne route of infection, whereas an H1N1 virus from a bird of the Anseriformes order was not. Here we show that two of the three H1N1 viruses isolated from Charadriiformes species in 2009 were transmitted between animals through the airborne route of infection, and five H1N1 isolates from Anseriformes species were not. The one H1N1 virus from a Charadriiformes species that failed to transmit through the airborne route was a reassortant possessing multiple internal gene segments from Anseriformes species. The molecular differences between the airborne-transmissible and non-airborne-transmissible H1N1 viruses were multigenic, involving the selection of virus with human-like receptor-binding specificity (α2-6 sialic acid) and multiple differences in the polymerase complex, mainly in the PB2, PB1-F2, and nonstructural genes.


Subject(s)
Air Microbiology , Anseriformes , Charadriiformes , Influenza A Virus, H1N1 Subtype , Influenza in Birds/transmission , Influenza in Birds/virology , Alberta/epidemiology , Animal Migration , Animals , Disease Models, Animal , Disease Reservoirs , Ferrets , Gene Rearrangement , Genome, Viral , Influenza A Virus, H1N1 Subtype/classification , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza in Birds/epidemiology , Male , Nasal Cavity/virology , New Jersey/epidemiology , Phylogeny , Polysaccharides/metabolism , Virus Replication
4.
Curr Opin Virol ; 11: 113-21, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25846699

ABSTRACT

A vaccine formulation that would be effective against all strains of influenza virus has long been a goal of vaccine developers, but antibodies after infection or vaccination were seen to be strain specific and there was little evidence of cross-reactive antibodies that neutralized across subtypes. Recently a number of broadly neutralizing monoclonal antibodies have been characterized. This review describes the different classes of broadly neutralizing antibodies and discusses the potential of their therapeutic use or for design of immunogens that induce a high proportion of broadly neutralizing antibodies.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antigens, Viral/immunology , Cross Reactions , Epitopes/immunology , Immunity, Heterologous , Orthomyxoviridae/immunology , Animals , Humans
6.
Curr Opin Virol ; 7: 128-33, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25061947

ABSTRACT

It has been known for many years that influenza viruses bind by their hemagglutinin surface glycoprotein to sialic acid (N-acetylneuraminic acid) on the surface of the host cell, and that avian viruses most commonly bind to sialic acid linked α2-3 to galactose while most human viruses bind to sialic acid in the α2-6 configuration. Over the past few years there has been a large increase in data on this binding due to technological advances in glycan binding assays, reverse genetic systems for influenza and in X-ray crystallography. The results show some surprising changes in binding specificity that do not appear to affect the ability of the virus to infect host cells.


Subject(s)
Glucans/metabolism , Influenza A virus/metabolism , Influenza in Birds/metabolism , Influenza, Human/metabolism , Receptors, Virus/metabolism , Animals , Birds , Humans , Influenza A virus/genetics , Influenza in Birds/virology , Influenza, Human/virology
7.
Cancer Biomark ; 14(1): 43-53, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24643041

ABSTRACT

Influenza viruses initiate infection by attaching to sialic acid receptors on the surface of host cells. It has been recognized for some time that avian influenza viruses usually bind to terminal sialic acid that is linked in the α2-3 configuration to the next sugar while human viruses show preference for α2-6 linked sialic acid. With developments in synthetic chemistry and chemo-enzymatic methods of synthesizing quite complex glycans, it has become clear that the binding specificity extends beyond the sialic acid, and this has led to considerable interest in developing glycan reagents that could be used either as a diagnostic tool for particular influenza viruses, or to identify cells that are susceptible to infection by certain influenza viruses. Here we describe the use of the Consortium for Functional Glycomics Glycan Array to investigate binding specificity of influenza hemagglutinin and cleavage by neuraminidase, using seasonal and pandemic H1N1 influenza viruses as examples, and compare the results with published data using other array methods.


Subject(s)
Hemagglutinins/metabolism , Influenza A Virus, H1N1 Subtype/metabolism , Neuraminidase/metabolism , Polysaccharides/metabolism , Animals , Chick Embryo , Dogs , Humans , Hydrazines/chemistry , Influenza, Human/virology , Madin Darby Canine Kidney Cells , Polysaccharides/analysis , Receptors, Cell Surface/metabolism , Virus Attachment
8.
J Virol ; 87(16): 8962-70, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23740997

ABSTRACT

The human parainfluenza virus (hPIV) hemagglutinin-neuraminidase (HN) protein binds (H) oligosaccharide receptors that contain N-acetylneuraminic acid (Neu5Ac) and cleaves (N) Neu5Ac from these oligosaccharides. In order to determine if one of HN's two functions is predominant, we measured the affinity of H for its ligands by a solid-phase binding assay with two glycoprotein substrates and by surface plasmon resonance with three monovalent glycans. We compared the dissociation constant (Kd) values from these experiments with previously determined Michaelis-Menten constants (Kms) for the enzyme activity. We found that glycoprotein substrates and monovalent glycans containing Neu5Acα2-3Galß1-4GlcNAc bind HN with Kd values in the 10 to 100 µM range. Km values for HN were previously determined to be on the order of 1 mM (M. M. Tappert, D. F. Smith, and G. M. Air, J. Virol. 85:12146-12159, 2011). A Km value greater than the Kd value indicates that cleavage occurs faster than the dissociation of binding and will dominate under N-permissive conditions. We propose, therefore, that HN is a neuraminidase that can hold its substrate long enough to act as a binding protein. The N activity can therefore regulate binding by reducing virus-receptor interactions when the concentration of receptor is high.


Subject(s)
HN Protein/metabolism , Parainfluenza Virus 1, Human/enzymology , Receptors, Virus/metabolism , Humans , Hydrolysis , Kinetics , Protein Binding , Surface Plasmon Resonance
9.
PLoS One ; 8(6): e66325, 2013.
Article in English | MEDLINE | ID: mdl-23805213

ABSTRACT

It is generally accepted that human influenza viruses bind glycans containing sialic acid linked α2-6 to the next sugar, that avian influenza viruses bind glycans containing the α2-3 linkage, and that mutations that change the binding specificity might change the host tropism. We noted that human H3N2 viruses showed dramatic differences in their binding specificity, and so we embarked on a study of representative human H3N2 influenza viruses, isolated from 1968 to 2012, that had been isolated and minimally passaged only in mammalian cells, never in eggs. The 45 viruses were grown in MDCK cells, purified, fluorescently labeled and screened on the Consortium for Functional Glycomics Glycan Array. Viruses isolated in the same season have similar binding specificity profiles but the profiles show marked year-to-year variation. None of the 610 glycans on the array (166 sialylated glycans) bound to all viruses; the closest was Neu5Acα2-6(Galß1-4GlcNAc)3 in either a linear or biantennary form, that bound 42 of the 45 viruses. The earliest human H3N2 viruses preferentially bound short, branched sialylated glycans while recent viruses bind better to long polylactosamine chains terminating in sialic acid. Viruses isolated in 1996, 2006, 2010 and 2012 bind glycans with α2-3 linked sialic acid; for 2006, 2010 and 2012 viruses this binding was inhibited by oseltamivir, indicating binding of α2-3 sialylated glycans by neuraminidase. More significantly, oseltamivir inhibited virus entry of 2010 and 2012 viruses into MDCK cells. All of these viruses were representative of epidemic strains that spread around the world, so all could infect and transmit between humans with high efficiency. We conclude that the year-to-year variation in receptor binding specificity is a consequence of amino acid sequence changes driven by antigenic drift, and that viruses with quite different binding specificity and avidity are equally fit to infect and transmit in the human population.


Subject(s)
Influenza A Virus, H3N2 Subtype , Influenza, Human , Polysaccharides , Receptors, Virus , Sialic Acids , Animals , Dogs , Female , Humans , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/isolation & purification , Influenza A Virus, H3N2 Subtype/metabolism , Influenza, Human/genetics , Influenza, Human/metabolism , Madin Darby Canine Kidney Cells , Male , Polysaccharides/genetics , Polysaccharides/metabolism , Receptors, Virus/genetics , Sialic Acids/genetics , Sialic Acids/metabolism
10.
PLoS One ; 7(12): e50830, 2012.
Article in English | MEDLINE | ID: mdl-23239987

ABSTRACT

BACKGROUND: Serum antibody responses in humans to inactivated influenza A (H5N1), (H9N2) and A (H7) vaccines have been varied but frequently low, particularly for subunit vaccines without adjuvant despite hemagglutinin (HA) concentrations expected to induce good responses. DESIGN: To help understand the low responses to subunit vaccines, we evaluated influenza A (H5N1), (H9N2), (H7N7) vaccines and 2009 pandemic (H1N1) vaccines for antigen uptake, processing and presentation by dendritic cells to T cells, conformation of vaccine HA in antibody binding assays and gel analyses, HA titers with different red blood cells, and vaccine morphology in electron micrographs (EM). RESULTS: Antigen uptake, processing and presentation of H5, H7, H9 and H1 vaccine preparations evaluated in humans appeared normal. No differences were detected in antibody interactions with vaccine and matched virus; although H7 trimer was not detected in western blots, no abnormalities in the conformation of the HA antigens were identified. The lowest HA titers for the vaccines were <1:4 for the H7 vaccine and 1:661 for an H9 vaccine; these vaccines induced the fewest antibody responses. A (H1N1) vaccines were the most immunogenic in humans; intact virus and virus pieces were prominent in EM. A good immunogenic A (H9N2) vaccine contained primarily particles of viral membrane with external HA and NA. A (H5N1) vaccines intermediate in immunogenicity were mostly indistinct structural units with stellates; the least immunogenic A (H7N7) vaccine contained mostly small 5 to 20 nm structures. SUMMARY: Antigen uptake, processing and presentation to human T cells and conformation of the HA appeared normal for each inactivated influenza A vaccine. Low HA titer was associated with low immunogenicity and presence of particles or split virus pieces was associated with higher immunogenicity.


Subject(s)
Influenza Vaccines , Influenza in Birds , Influenza, Human , Vaccines, Inactivated , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antigen Presentation/immunology , Birds/immunology , Birds/virology , Hemagglutination Inhibition Tests , Humans , Influenza A Virus, H5N1 Subtype/immunology , Influenza A Virus, H7N7 Subtype/immunology , Influenza A Virus, H9N2 Subtype/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Influenza in Birds/immunology , Influenza in Birds/prevention & control , Influenza, Human/immunology , Influenza, Human/prevention & control , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology
11.
J Biol Chem ; 287(53): 44784-99, 2012 Dec 28.
Article in English | MEDLINE | ID: mdl-23115247

ABSTRACT

Human milk contains a large diversity of free glycans beyond lactose, but their functions are not well understood. To explore their functional recognition, here we describe a shotgun glycan microarray prepared from isolated human milk glycans (HMGs), and our studies on their recognition by viruses, antibodies, and glycan-binding proteins (GBPs), including lectins. The total neutral and sialylated HMGs were derivatized with a bifunctional fluorescent tag, separated by multidimensional HPLC, and archived in a tagged glycan library, which was then used to print a shotgun glycan microarray (SGM). This SGM was first interrogated with well defined GBPs and antibodies. These data demonstrated both the utility of the array and provided preliminary structural information (metadata) about this complex glycome. Anti-TRA-1 antibodies that recognize human pluripotent stem cells specifically recognized several HMGs that were then further structurally defined as novel epitopes for these antibodies. Human influenza viruses and Parvovirus Minute Viruses of Mice also specifically recognized several HMGs. For glycan sequencing, we used a novel approach termed metadata-assisted glycan sequencing (MAGS), in which we combine information from analyses of glycans by mass spectrometry with glycan interactions with defined GBPs and antibodies before and after exoglycosidase treatments on the microarray. Together, these results provide novel insights into diverse recognition functions of HMGs and show the utility of the SGM approach and MAGS as resources for defining novel glycan recognition by GBPs, antibodies, and pathogens.


Subject(s)
Biomarkers/chemistry , Glycomics , Milk, Human/chemistry , Polysaccharides/chemistry , Receptors, Virus/analysis , Animals , Carbohydrate Sequence , Cell Line , Embryonic Stem Cells/metabolism , Humans , Mice , Milk, Human/metabolism , Molecular Sequence Data , Polysaccharides/metabolism , Receptors, Virus/genetics , Receptors, Virus/metabolism
12.
PLoS One ; 7(7): e41895, 2012.
Article in English | MEDLINE | ID: mdl-22848649

ABSTRACT

H3N2 influenza viruses have now circulated in the human population for 43 years since the pandemic of 1968, accumulating sequence changes in the hemagglutinin (HA) and neuraminidase (NA) that are believed to be predominantly due to selection for escape from antibodies. Examination of mutations that persist and accumulate led to identification of antigenically significant mutations that are contained in five antigenic sites (A-E) mapped on to the H3 HA. In early H3N2 isolates, antigenic site A appeared to be dominant while in the 1990s site B seemed more important. To obtain experimental evidence for dominance of antigenic sites on modern H3 HAs, we have measured antibodies in plasma of human subjects who received the 2006-07 trivalent subunit influenza vaccine (H3 component A/Wisconsin/67/05) or the 2008-09 formulation (H3 component A/Uruguay/716/07). Plasmas were tested against expressed HA of Wisconsin-like influenza A/Oklahoma/309/06 and site-directed mutants in antigenic site A (NNES121-124ITEG, N126T, N133D, TSSS135-138GSNA, K140I, RSNNS142-146PGSG), and antigenic site B (HL156-157KS, KFK158-160GST, NDQI189-192QEQT, A196V). "Native ELISA" analysis and escape mutant selection with two human monoclonal antibodies demonstrated that antibody E05 binds to antigenic site A and 1_C02 binds to site B. We find that most individuals, after vaccination in seasons 2006-07 and/or 2008-09, showed dominance of antigenic site B recognition over antigenic site A. A minority showed dominance of site A in 2006 but these were reduced in 2008 when the vaccine virus had a site A mutation. A better understanding of immunodominance may allow prediction of future antigenic drift and assist in vaccine strain selection.


Subject(s)
Antigens, Viral/immunology , Epitopes/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H3N2 Subtype/immunology , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antigens, Viral/genetics , Epitopes/genetics , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Influenza A Virus, H3N2 Subtype/genetics , Models, Molecular , Mutation , Protein Conformation , Seasons , Species Specificity , Vaccination
13.
Bioorg Med Chem ; 20(14): 4582-9, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22677529

ABSTRACT

Neuraminidase (NA) plays a critical role in the life cycle of influenza virus and is a target for new therapeutic agents. A series of influenza neuraminidase inhibitors with the pyrrolidinobenzoic acid scaffold containing lipophilic side chains at the C3 position have been synthesized and evaluated for influenza neuraminidase inhibitory activity. The size and geometry of the C3 side chains have been modified in order to investigate structure-activity relationships. The results indicated that size and geometry of the C3-side chain are important for selectivity of inhibition against N1 versus N2 NA, important type A influenza variants that infect man, including the highly lethal avian influenza.


Subject(s)
Antiviral Agents/chemistry , Benzoic Acid/chemistry , Influenza A virus/enzymology , Neuraminidase/antagonists & inhibitors , Pyrrolidinones/chemistry , Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacology , Benzoic Acid/chemical synthesis , Benzoic Acid/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Hydrophobic and Hydrophilic Interactions , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A virus/drug effects , Neuraminidase/metabolism , Structure-Activity Relationship
14.
BMC Struct Biol ; 12: 7, 2012 May 06.
Article in English | MEDLINE | ID: mdl-22559154

ABSTRACT

BACKGROUND: Influenza neuraminidase (NA) is an important target for antiviral inhibitors since its active site is highly conserved such that inhibitors can be cross-reactive against multiple types and subtypes of influenza. Here, we discuss the crystal structure of neuraminidase subtype N9 complexed with a new benzoic acid based inhibitor (2) that was designed to add contacts by overpacking one side of the active site pocket. Inhibitor 2 uses benzoic acid to mimic the pyranose ring, a bis-(hydroxymethyl)-substituted 2-pyrrolidinone ring in place of the N-acetyl group of the sialic acid, and a branched aliphatic structure to fill the sialic acid C6 subsite. RESULTS: Inhibitor 2 {4-[2,2-bis(hydroxymethyl)-5-oxo-pyrrolidin-1-yl]-3-[(dipropylamino)methyl)]benzoic acid} was soaked into crystals of neuraminidase of A/tern/Australia/G70c/75 (N9), and the structure refined with 1.55 Å X-ray data. The benzene ring of the inhibitor tilted 8.9° compared to the previous compound (1), and the number of contacts, including hydrogen bonds, increased. However, the IC50 for compound 2 remained in the low micromolar range, likely because one propyl group was disordered. In this high-resolution structure of NA isolated from virus grown in chicken eggs, we found electron density for additional sugar units on the N-linked glycans compared to previous neuraminidase structures. In particular, seven mannoses and two N-acetylglucosamines are visible in the glycan attached to Asn200. This long, branched high-mannose glycan makes significant contacts with the neighboring subunit. CONCLUSIONS: We designed inhibitor 2 with an extended substituent at C4-corresponding to C6 of sialic acid-to increase the contact surface in the C6-subsite and to force the benzene ring to tilt to maximize these interactions while retaining the interactions of the carboxylate and the pyrolidinone substituents. The crystal structure at 1.55 Å showed that we partially succeeded in that the ring in 2 is tilted relative to 1 and the number of contacts increased, but one hydrophobic branch makes no contacts, perhaps explaining why the IC50 did not decrease. Future design efforts will include branches of unequal length so that both branches may be accommodated in the C6-subsite without conformational disorder. The high-mannose glycan attached to Asn200 makes several inter-subunit contacts and appears to stabilize the tetramer.


Subject(s)
Benzoic Acid/chemistry , Benzoic Acid/pharmacology , Catalytic Domain , Influenza A virus/enzymology , Neuraminidase/antagonists & inhibitors , Neuraminidase/chemistry , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Crystallography, X-Ray , Glucose/chemistry , Inhibitory Concentration 50 , Models, Molecular , Neuraminidase/metabolism , Polysaccharides/chemistry , Protein Binding/drug effects , X-Ray Diffraction
15.
J Virol ; 86(10): 5857-66, 2012 May.
Article in English | MEDLINE | ID: mdl-22398283

ABSTRACT

Genetic drift of influenza virus genomic sequences occurs through the combined effects of sequence alterations introduced by a low-fidelity polymerase and the varying selective pressures experienced as the virus migrates through different host environments. While traditional phylogenetic analysis is useful in tracking the evolutionary heritage of these viruses, the specific genetic determinants that dictate important phenotypic characteristics are often difficult to discern within the complex genetic background arising through evolution. Here we describe a novel influenza virus sequence feature variant type (Flu-SFVT) approach, made available through the public Influenza Research Database resource (www.fludb.org), in which variant types (VTs) identified in defined influenza virus protein sequence features (SFs) are used for genotype-phenotype association studies. Since SFs have been defined for all influenza virus proteins based on known structural, functional, and immune epitope recognition properties, the Flu-SFVT approach allows the rapid identification of the molecular genetic determinants of important influenza virus characteristics and their connection to underlying biological functions. We demonstrate the use of the SFVT approach to obtain statistical evidence for effects of NS1 protein sequence variations in dictating influenza virus host range restriction.


Subject(s)
Host Specificity , Influenza A virus/genetics , Influenza, Human/virology , Viral Nonstructural Proteins/genetics , Amino Acid Sequence , Genetic Variation , Humans , Influenza A virus/chemistry , Influenza A virus/classification , Influenza A virus/physiology , Molecular Sequence Data , Phylogeny , Protein Structure, Secondary , Sequence Alignment , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism
16.
Influenza Other Respir Viruses ; 6(4): 245-56, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22085243

ABSTRACT

Influenza neuraminidase is the target of two licensed antivirals that have been very successful, with several more in development. However, neuraminidase has been largely ignored as a vaccine target despite evidence that inclusion of neuraminidase in the subunit vaccine gives increased protection. This article describes current knowledge on the structure, enzyme activity, and antigenic significance of neuraminidase.


Subject(s)
Neuraminidase/chemistry , Neuraminidase/metabolism , Orthomyxoviridae/enzymology , Viral Proteins/chemistry , Viral Proteins/metabolism , Antiviral Agents/pharmacology , Enzyme Inhibitors/pharmacology , Influenza Vaccines/immunology , Models, Molecular , Neuraminidase/antagonists & inhibitors , Neuraminidase/immunology , Orthomyxoviridae/drug effects , Protein Conformation , Viral Proteins/antagonists & inhibitors , Viral Proteins/immunology
17.
Retrovirology ; 8: 101, 2011 Dec 07.
Article in English | MEDLINE | ID: mdl-22151792

ABSTRACT

BACKGROUND: Bevirimat, the prototype Human Immunodeficiency Virus type 1 (HIV-1) maturation inhibitor, is highly potent in cell culture and efficacious in HIV-1 infected patients. In contrast to inhibitors that target the active site of the viral protease, bevirimat specifically inhibits a single cleavage event, the final processing step for the Gag precursor where p25 (CA-SP1) is cleaved to p24 (CA) and SP1. RESULTS: In this study, photoaffinity analogs of bevirimat and mass spectrometry were employed to map the binding site of bevirimat to Gag within immature virus-like particles. Bevirimat analogs were found to crosslink to sequences overlapping, or proximal to, the CA-SP1 cleavage site, consistent with previous biochemical data on the effect of bevirimat on Gag processing and with genetic data from resistance mutations, in a region predicted by NMR and mutational studies to have α-helical character. Unexpectedly, a second region of interaction was found within the Major Homology Region (MHR). Extensive prior genetic evidence suggests that the MHR is critical for virus assembly. CONCLUSIONS: This is the first demonstration of a direct interaction between the maturation inhibitor, bevirimat, and its target, Gag. Information gained from this study sheds light on the mechanisms by which the virus develops resistance to this class of drug and may aid in the design of next-generation maturation inhibitors.


Subject(s)
Anti-HIV Agents/pharmacology , HIV-1/drug effects , Succinates/pharmacology , Triterpenes/pharmacology , gag Gene Products, Human Immunodeficiency Virus , Binding Sites , Cell Line , HEK293 Cells , HIV Infections/drug therapy , HIV-1/physiology , Humans , Mass Spectrometry , Molecular Sequence Data , Protein Binding , Virus Assembly/drug effects , Virus Replication/drug effects , gag Gene Products, Human Immunodeficiency Virus/antagonists & inhibitors , gag Gene Products, Human Immunodeficiency Virus/metabolism
18.
J Virol ; 85(23): 12146-59, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21917945

ABSTRACT

The hemagglutinin-neuraminidase (HN) protein of human parainfluenza viruses (hPIVs) both binds (H) and cleaves (N) oligosaccharides that contain N-acetylneuraminic acid (Neu5Ac). H is thought to correspond to receptor binding and N to receptor-destroying activity. At present, N's role in infection remains unclear: does it destroy only receptors, or are there other targets? We previously demonstrated that hPIV1 and 3 HNs bind to oligosaccharides containing the motif Neu5Acα2-3Galß1-4GlcNAc (M. Amonsen, D. F. Smith, R. D. Cummings, and G. M. Air, J. Virol. 81:8341-8345, 2007). In the present study, we tested the binding specificity of hPIV2 on the Consortium for Functional Glycomics' glycan array and found that hPIV2 binds to oligosaccharides containing the same motif. We determined the specificities of N on red blood cells, soluble small-molecule and glycoprotein substrates, and the glycan array and compared them to the specificities of H. hPIV2 and -3, but not hPIV1, cleaved their ligands on red blood cells. hPIV1, -2, and -3 cleaved their NeuAcα2-3 ligands on the glycan array; hPIV2 and -3 also cleaved NeuAcα2-6 ligands bound by influenza A virus. While all three HNs exhibited similar affinities for all cleavable soluble substrates, their activities were 5- to 10-fold higher on small molecules than on glycoproteins. In addition, some soluble glycoproteins were not cleaved, despite containing oligosaccharides that were cleaved on the glycan array. We conclude that the susceptibility of an oligosaccharide substrate to N increases when the substrate is fixed to a surface. These findings suggest that HN may undergo a conformational change that activates N upon receptor binding at a cell surface.


Subject(s)
Hemagglutinins/metabolism , Neuraminidase/metabolism , Oligosaccharides/metabolism , Parainfluenza Virus 1, Human/metabolism , Parainfluenza Virus 2, Human/metabolism , Parainfluenza Virus 3, Human/metabolism , Receptors, Cell Surface/metabolism , Amino Acid Sequence , Animals , Cell Membrane/metabolism , Chickens , Erythrocytes/metabolism , Glycosylation , Hemagglutination Tests , Humans , Microarray Analysis , Molecular Sequence Data , Parainfluenza Virus 1, Human/isolation & purification , Parainfluenza Virus 2, Human/isolation & purification , Parainfluenza Virus 3, Human/isolation & purification , Phylogeny , Polysaccharides/metabolism , Receptors, Virus/metabolism , Respirovirus Infections/genetics , Respirovirus Infections/metabolism , Respirovirus Infections/virology , Sequence Homology, Amino Acid , Turkey
19.
J Clin Immunol ; 31(5): 900-12, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21732013

ABSTRACT

INTRODUCTION: The 2009 swine-origin H1N1 influenza virus (swH1N1) provided an opportunity to study immune responses to a new influenza strain in the context of seasonal influenza vaccination. Our goals were: to assess whether analyzing multiple parameters of immune responsiveness to influenza has an advantage over evaluating hemagglutination inhibition (HAI) titer alone, to determine whether vaccination with the seasonal vaccine induced cross-reactive immunity to swH1N1 in some individuals, and to determine whether the immune response against swH1N1 is higher after infection than vaccination. METHODS: Antibody and T cell responses were studied in ten subjects who were first immunized with the 2009-2010 seasonal influenza subunit vaccine, then 6 weeks later with the swH1N1 monovalent subunit vaccine. The amount of antibody against native virus glycoproteins, overall avidity of these antibodies, and HAI titer were measured. T cells were evaluated for proliferation and IFNγ secretion in response to the vaccine in vitro. Individuals with influenza-like illness were also evaluated, adding a microplate neuraminidase inhibition (NAI) test. RESULTS: The immune response to influenza was highly variable and immune parameters did not increase in parallel. The seasonal vaccine induced antibodies recognizing the pandemic virus in 50% of subjects. Antibody affinity and NAI activity to swH1N1 were higher after natural infection than vaccination. CONCLUSIONS: The evaluation of several immune parameters gives a more complete measure of immune responsiveness to influenza infection or vaccination than the HAI test alone.


Subject(s)
Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines , Influenza, Human/epidemiology , Influenza, Human/immunology , Pandemics , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antigens, Viral/immunology , Cells, Cultured , Cross Reactions , Hemagglutination Inhibition Tests/methods , Humans , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza, Human/diagnosis , Influenza, Human/physiopathology , Monitoring, Immunologic/methods , Swine , T-Lymphocytes/immunology , T-Lymphocytes/virology , United States , Vaccination , Zoonoses
20.
J Biol Chem ; 286(36): 31610-22, 2011 Sep 09.
Article in English | MEDLINE | ID: mdl-21757734

ABSTRACT

Many glycan-binding proteins in animals and pathogens recognize sialic acid or its modified forms, but their molecular recognition is poorly understood. Here we describe studies on sialic acid recognition using a novel sialylated glycan microarray containing modified sialic acids presented on different glycan backbones. Glycans terminating in ß-linked galactose at the non-reducing end and with an alkylamine-containing fluorophore at the reducing end were sialylated by a one-pot three-enzyme system to generate α2-3- and α2-6-linked sialyl glycans with 16 modified sialic acids. The resulting 77 sialyl glycans were purified and quantified, characterized by mass spectrometry, covalently printed on activated slides, and interrogated with a number of key sialic acid-binding proteins and viruses. Sialic acid recognition by the sialic acid-binding lectins Sambucus nigra agglutinin and Maackia amurensis lectin-I, which are routinely used for detecting α2-6- and α2-3-linked sialic acids, are affected by sialic acid modifications, and both lectins bind glycans terminating with 2-keto-3-deoxy-D-glycero-D-galactonononic acid (Kdn) and Kdn derivatives stronger than the derivatives of more common N-acetylneuraminic acid (Neu5Ac) and N-glycolylneuraminic acid (Neu5Gc). Three human parainfluenza viruses bind to glycans terminating with Neu5Ac or Neu5Gc and some of their derivatives but not to Kdn and its derivatives. Influenza A virus also does not bind glycans terminating in Kdn or Kdn derivatives. An especially novel aspect of human influenza A virus binding is its ability to equivalently recognize glycans terminated with either α2-6-linked Neu5Ac9Lt or α2-6-linked Neu5Ac. Our results demonstrate the utility of this sialylated glycan microarray to investigate the biological importance of modified sialic acids in protein-glycan interactions.


Subject(s)
Polysaccharides/metabolism , Protein Array Analysis , Proteins/metabolism , Sialic Acids/metabolism , Viruses/metabolism , Binding Sites , Humans , Influenza A virus , Lectins/metabolism , Respirovirus , Sialic Acids/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...