Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
MMWR Morb Mortal Wkly Rep ; 71(6): 206-211, 2022 02 11.
Article in English | MEDLINE | ID: mdl-35143464

ABSTRACT

Genomic surveillance is a critical tool for tracking emerging variants of SARS-CoV-2 (the virus that causes COVID-19), which can exhibit characteristics that potentially affect public health and clinical interventions, including increased transmissibility, illness severity, and capacity for immune escape. During June 2021-January 2022, CDC expanded genomic surveillance data sources to incorporate sequence data from public repositories to produce weighted estimates of variant proportions at the jurisdiction level and refined analytic methods to enhance the timeliness and accuracy of national and regional variant proportion estimates. These changes also allowed for more comprehensive variant proportion estimation at the jurisdictional level (i.e., U.S. state, district, territory, and freely associated state). The data in this report are a summary of findings of recent proportions of circulating variants that are updated weekly on CDC's COVID Data Tracker website to enable timely public health action.† The SARS-CoV-2 Delta (B.1.617.2 and AY sublineages) variant rose from 1% to >50% of viral lineages circulating nationally during 8 weeks, from May 1-June 26, 2021. Delta-associated infections remained predominant until being rapidly overtaken by infections associated with the Omicron (B.1.1.529 and BA sublineages) variant in December 2021, when Omicron increased from 1% to >50% of circulating viral lineages during a 2-week period. As of the week ending January 22, 2022, Omicron was estimated to account for 99.2% (95% CI = 99.0%-99.5%) of SARS-CoV-2 infections nationwide, and Delta for 0.7% (95% CI = 0.5%-1.0%). The dynamic landscape of SARS-CoV-2 variants in 2021, including Delta- and Omicron-driven resurgences of SARS-CoV-2 transmission across the United States, underscores the importance of robust genomic surveillance efforts to inform public health planning and practice.


Subject(s)
COVID-19/epidemiology , COVID-19/virology , SARS-CoV-2/genetics , Centers for Disease Control and Prevention, U.S. , Genomics , Humans , Prevalence , Public Health Surveillance/methods , United States/epidemiology
2.
MMWR Morb Mortal Wkly Rep ; 70(3): 95-99, 2021 Jan 22.
Article in English | MEDLINE | ID: mdl-33476315

ABSTRACT

On December 14, 2020, the United Kingdom reported a SARS-CoV-2 variant of concern (VOC), lineage B.1.1.7, also referred to as VOC 202012/01 or 20I/501Y.V1.* The B.1.1.7 variant is estimated to have emerged in September 2020 and has quickly become the dominant circulating SARS-CoV-2 variant in England (1). B.1.1.7 has been detected in over 30 countries, including the United States. As of January 13, 2021, approximately 76 cases of B.1.1.7 have been detected in 12 U.S. states.† Multiple lines of evidence indicate that B.1.1.7 is more efficiently transmitted than are other SARS-CoV-2 variants (1-3). The modeled trajectory of this variant in the U.S. exhibits rapid growth in early 2021, becoming the predominant variant in March. Increased SARS-CoV-2 transmission might threaten strained health care resources, require extended and more rigorous implementation of public health strategies (4), and increase the percentage of population immunity required for pandemic control. Taking measures to reduce transmission now can lessen the potential impact of B.1.1.7 and allow critical time to increase vaccination coverage. Collectively, enhanced genomic surveillance combined with continued compliance with effective public health measures, including vaccination, physical distancing, use of masks, hand hygiene, and isolation and quarantine, will be essential to limiting the spread of SARS-CoV-2, the virus that causes coronavirus disease 2019 (COVID-19). Strategic testing of persons without symptoms but at higher risk of infection, such as those exposed to SARS-CoV-2 or who have frequent unavoidable contact with the public, provides another opportunity to limit ongoing spread.


Subject(s)
COVID-19/epidemiology , COVID-19/virology , SARS-CoV-2/genetics , COVID-19/transmission , Genome, Viral , Humans , Mutation , United States/epidemiology
3.
MMWR Morb Mortal Wkly Rep ; 70(23): 846-850, 2021 Jun 11.
Article in English | MEDLINE | ID: mdl-34111060

ABSTRACT

SARS-CoV-2, the virus that causes COVID-19, is constantly mutating, leading to new variants (1). Variants have the potential to affect transmission, disease severity, diagnostics, therapeutics, and natural and vaccine-induced immunity. In November 2020, CDC established national surveillance for SARS-CoV-2 variants using genomic sequencing. As of May 6, 2021, sequences from 177,044 SARS-CoV-2-positive specimens collected during December 20, 2020-May 6, 2021, from 55 U.S. jurisdictions had been generated by or reported to CDC. These included 3,275 sequences for the 2-week period ending January 2, 2021, compared with 25,000 sequences for the 2-week period ending April 24, 2021 (0.1% and 3.1% of reported positive SARS-CoV-2 tests, respectively). Because sequences might be generated by multiple laboratories and sequence availability varies both geographically and over time, CDC developed statistical weighting and variance estimation methods to generate population-based estimates of the proportions of identified variants among SARS-CoV-2 infections circulating nationwide and in each of the 10 U.S. Department of Health and Human Services (HHS) geographic regions.* During the 2-week period ending April 24, 2021, the B.1.1.7 and P.1 variants represented an estimated 66.0% and 5.0% of U.S. SARS-CoV-2 infections, respectively, demonstrating the rise to predominance of the B.1.1.7 variant of concern† (VOC) and emergence of the P.1 VOC in the United States. Using SARS-CoV-2 genomic surveillance methods to analyze surveillance data produces timely population-based estimates of the proportions of variants circulating nationally and regionally. Surveillance findings demonstrate the potential for new variants to emerge and become predominant, and the importance of robust genomic surveillance. Along with efforts to characterize the clinical and public health impact of SARS-CoV-2 variants, surveillance can help guide interventions to control the COVID-19 pandemic in the United States.


Subject(s)
COVID-19/virology , SARS-CoV-2/genetics , COVID-19/epidemiology , Epidemiological Monitoring , Humans , SARS-CoV-2/isolation & purification , United States/epidemiology
4.
Proc Natl Acad Sci U S A ; 111(22): E2241-50, 2014 Jun 03.
Article in English | MEDLINE | ID: mdl-24843157

ABSTRACT

Influenza viruses bind to host cell surface glycans containing terminal sialic acids, but as studies on influenza binding become more sophisticated, it is becoming evident that although sialic acid may be necessary, it is not sufficient for productive binding. To better define endogenous glycans that serve as viral receptors, we have explored glycan recognition in the pig lung, because influenza is broadly disseminated in swine, and swine have been postulated as an intermediary host for the emergence of pandemic strains. For these studies, we used the technology of "shotgun glycomics" to identify natural receptor glycans. The total released N- and O-glycans from pig lung glycoproteins and glycolipid-derived glycans were fluorescently tagged and separated by multidimensional HPLC, and individual glycans were covalently printed to generate pig lung shotgun glycan microarrays. All viruses tested interacted with one or more sialylated N-glycans but not O-glycans or glycolipid-derived glycans, and each virus demonstrated novel and unexpected differences in endogenous N-glycan recognition. The results illustrate the repertoire of specific, endogenous N-glycans of pig lung glycoproteins for virus recognition and offer a new direction for studying endogenous glycan functions in viral pathogenesis.


Subject(s)
Glycomics/methods , Influenza in Birds/metabolism , Influenza, Human/metabolism , Lung/virology , Orthomyxoviridae/metabolism , Receptors, Virus/metabolism , Agglutination Tests , Animals , Birds , Chickens , Erythrocytes/virology , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Humans , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza A Virus, H1N1 Subtype/metabolism , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H1N2 Subtype/isolation & purification , Influenza A Virus, H1N2 Subtype/metabolism , Influenza A Virus, H1N2 Subtype/pathogenicity , Influenza A Virus, H3N2 Subtype/isolation & purification , Influenza A Virus, H3N2 Subtype/metabolism , Influenza A Virus, H3N2 Subtype/pathogenicity , Influenza in Birds/virology , Influenza, Human/virology , Lectins/metabolism , Lung/metabolism , Orthomyxoviridae/isolation & purification , Orthomyxoviridae/pathogenicity , Polysaccharides/metabolism , Species Specificity , Swine , Virulence
5.
J Virol ; 89(8): 4504-16, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25653452

ABSTRACT

UNLABELLED: Influenza A viruses enter host cells through endosomes, where acidification induces irreversible conformational changes of the viral hemagglutinin (HA) that drive the membrane fusion process. The prefusion conformation of the HA is metastable, and the pH of fusion can vary significantly among HA strains and subtypes. Furthermore, an accumulating body of evidence implicates HA stability properties as partial determinants of influenza host range, transmission phenotype, and pathogenic potential. Although previous studies have identified HA mutations that can affect HA stability, these have been limited to a small selection of HA strains and subtypes. Here we report a mutational analysis of HA stability utilizing a panel of expressed HAs representing a broad range of HA subtypes and strains, including avian representatives across the phylogenetic spectrum and several human strains. We focused on two highly conserved residues in the HA stem region: HA2 position 58, located at the membrane distal tip of the short helix of the hairpin loop structure, and HA2 position 112, located in the long helix in proximity to the fusion peptide. We demonstrate that a K58I mutation confers an acid-stable phenotype for nearly all HAs examined, whereas a D112G mutation consistently leads to elevated fusion pH. The results enhance our understanding of HA stability across multiple subtypes and provide an additional tool for risk assessment for circulating strains that may have other hallmarks of human adaptation. Furthermore, the K58I mutants, in particular, may be of interest for potential use in the development of vaccines with improved stability profiles. IMPORTANCE: The influenza A hemagglutinin glycoprotein (HA) mediates the receptor binding and membrane fusion functions that are essential for virus entry into host cells. While receptor binding has long been recognized for its role in host species specificity and transmission, membrane fusion and associated properties of HA stability have only recently been appreciated as potential determinants. We show here that mutations can be introduced at highly conserved positions to stabilize or destabilize the HA structure of multiple HA subtypes, expanding our knowledge base for this important phenotype. The practical implications of these findings extend to the field of vaccine design, since the HA mutations characterized here could potentially be utilized across a broad spectrum of influenza virus subtypes to improve the stability of vaccine strains or components.


Subject(s)
Genetic Variation , Hemagglutinins/genetics , Influenza A virus/genetics , Models, Molecular , Phenotype , Animals , Chlorocebus aethiops , Cloning, Molecular , Cricetinae , DNA Mutational Analysis , Hemagglutinins/chemistry , Hydrogen-Ion Concentration , Immunoprecipitation , Luciferases , Mutagenesis , Mutation/genetics , Protein Conformation , Protein Stability , Species Specificity , Vero Cells
6.
J Virol ; 88(3): 1502-12, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24227867

ABSTRACT

The zoonotic outbreak of H7N9 subtype avian influenza virus that occurred in eastern China in the spring of 2013 resulted in 135 confirmed human cases, 44 of which were lethal. Sequencing of the viral genome revealed a number of molecular signatures associated with virulence or transmission in mammals. We report here that, in the guinea pig model, a human isolate of novel H7N9 influenza virus, A/Anhui/1/2013 (An/13), is highly dissimilar to an H7N1 avian isolate and instead behaves similarly to a human seasonal strain in several respects. An/13 was found to have a low 50% infectious dose, grow to high titers in the upper respiratory tract, and transmit efficiently among cocaged guinea pigs. The pH of fusion of the hemagglutinin (HA) and the binding of virus to fixed guinea pig tissues were also examined. The An/13 HA displayed a relatively elevated pH of fusion characteristic of many avian strains, and An/13 resembled avian viruses in terms of attachment to tissues. One important difference was seen between An/13 and both the H3N2 human and the H7N1 avian viruses: when inoculated intranasally at a high dose, only the An/13 virus led to productive infection of the lower respiratory tract of guinea pigs. In sum, An/13 was found to retain fusion and attachment properties of an avian influenza virus but displayed robust growth and contact transmission in the guinea pig model atypical of avian strains and indicative of mammalian adaptation.


Subject(s)
Disease Models, Animal , Guinea Pigs , Influenza A Virus, H7N9 Subtype/growth & development , Influenza A Virus, H7N9 Subtype/pathogenicity , Influenza, Human/virology , Animals , Female , Humans , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/physiology , Influenza A Virus, H7N1 Subtype/genetics , Influenza A Virus, H7N1 Subtype/physiology , Influenza A Virus, H7N9 Subtype/genetics , Influenza A Virus, H7N9 Subtype/physiology , Influenza, Human/transmission , Virulence
7.
PLoS Pathog ; 9(2): e1003151, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23459660

ABSTRACT

The influenza A virus (IAV) HA protein must be activated by host cells proteases in order to prime the molecule for fusion. Consequently, the availability of activating proteases and the susceptibility of HA to protease activity represents key factors in facilitating virus infection. As such, understanding the intricacies of HA cleavage by various proteases is necessary to derive insights into the emergence of pandemic viruses. To examine these properties, we generated a panel of HAs that are representative of the 16 HA subtypes that circulate in aquatic birds, as well as HAs representative of the subtypes that have infected the human population over the last century. We examined the susceptibility of the panel of HA proteins to trypsin, as well as human airway trypsin-like protease (HAT) and transmembrane protease, serine 2 (TMPRSS2). Additionally, we examined the pH at which these HAs mediated membrane fusion, as this property is related to the stability of the HA molecule and influences the capacity of influenza viruses to remain infectious in natural environments. Our results show that cleavage efficiency can vary significantly for individual HAs, depending on the protease, and that some HA subtypes display stringent selectivity for specific proteases as activators of fusion function. Additionally, we found that the pH of fusion varies by 0.7 pH units among the subtypes, and notably, we observed that the pH of fusion for most HAs from human isolates was lower than that observed from avian isolates of the same subtype. Overall, these data provide the first broad-spectrum analysis of cleavage-activation and membrane fusion characteristics for all of the IAV HA subtypes, and also show that there are substantial differences between the subtypes that may influence transmission among hosts and establishment in new species.


Subject(s)
Adaptation, Physiological , Hemagglutinin Glycoproteins, Influenza Virus/classification , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Influenza A virus/pathogenicity , Membrane Fusion/physiology , Virus Activation/physiology , Animals , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Host Specificity , Humans , Hydrogen-Ion Concentration , Immunoprecipitation , Influenza A virus/genetics , Influenza, Human/metabolism , Influenza, Human/pathology , Influenza, Human/virology , Phenotype , Protein Conformation , Serine Endopeptidases/metabolism , Trypsin/metabolism , Vero Cells , Virus Replication
8.
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
9.
J Virol ; 85(23): 12387-98, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21917953

ABSTRACT

To examine the range of selective processes that potentially operate when poorly binding influenza viruses adapt to replicate more efficiently in alternative environments, we passaged a virus containing an attenuating mutation in the hemagglutinin (HA) receptor binding site in mice and characterized the resulting mutants with respect to the structural locations of mutations selected, the replication phenotypes of the viruses, and their binding properties on glycan microarrays. The initial attenuated virus had a tyrosine-to-phenylalanine mutation at HA1 position 98 (Y98F), located in the receptor binding pocket, but viruses that were selected contained second-site pseudoreversion mutations in various structural locations that revealed a range of molecular mechanisms for modulating receptor binding that go beyond the scope that is generally mapped using receptor specificity mutants. A comparison of virus titers in the mouse respiratory tract versus MDCK cells in culture showed that the mutants displayed distinctive replication properties depending on the system, but all were less attenuated in mice than the Y98F virus. An analysis of receptor binding properties confirmed that the initial Y98F virus bound poorly to several different species of erythrocytes, while all mutants reacquired various degrees of hemagglutination activity. Interestingly, both the Y98F virus and pseudoreversion mutants were shown to bind very inefficiently to standard glycan microarrays containing an abundance of binding substrates for most influenza viruses that have been characterized to date, provided by the Consortium for Functional Glycomics. The viruses were also examined on a recently developed microarray containing glycans terminating in sialic acid derivatives, and limited binding to a potentially interesting subset of glycans was revealed. The results are discussed with respect to mechanisms for HA-mediated receptor binding, as well as regarding the species of molecules that may act as receptors for influenza virus on host cell surfaces.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Mutation/genetics , Orthomyxoviridae Infections/virology , Receptors, Virus/metabolism , Virus Replication , Animals , Binding Sites , Cattle , Cells, Cultured , Chickens , Dogs , Erythrocytes/metabolism , Erythrocytes/virology , Genetic Vectors , Guinea Pigs , Hemagglutination Tests , Horses , Kidney/cytology , Kidney/metabolism , Kidney/virology , Mice , Microarray Analysis , Models, Molecular , Mutagenesis, Site-Directed , Orthomyxoviridae/pathogenicity , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/metabolism , Polysaccharides/metabolism , Protein Binding , Protein Conformation , Sheep , Turkey
10.
J Virol ; 82(24): 12280-90, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18829753

ABSTRACT

There are many unique aspects of vesicular stomatitis virus (VSV) transcription. In addition to its unusual mRNA capping and methyltransferase mechanisms, the addition of S-adenosyl homocysteine (SAH), which is the by-product and competitive inhibitor of S-adenosyl methionine (SAM)-mediated methyltransferase reactions, leads to synthesis of poly(A) tails on the 3' end of VSV mRNAs that are 10- or 20-fold longer than normal. The mechanism by which this occurs is not understood, since it has been shown that productive transcription is not dependent on 5' cap methylation and full-length VSV mRNAs can be synthesized in the absence of SAM. To investigate this unusual phenotype, we assayed the effects of SAH on transcription using a panel of recombinant viruses that contained mutations in domain VI of the VSV L protein. The L proteins we investigated displayed a range of 5' cap methyltransferase activities. In the present study, we show that the ability of the VSV L protein to catalyze methyl transfer correlates with its sensitivity to SAH with respect to polyadenylation, thereby indicating an intriguing connection between 5' and 3' end mRNA modifications. We also identified an L protein mutant that hyperpolyadenylates mRNA irrespective of the presence or absence of exogenous SAH. Further, the data presented here show that the wild-type L protein hyperpolyadenylates a percentage of VSV mRNAs in infected cells as well as in vitro.


Subject(s)
Adenosine/genetics , Adenosine/metabolism , Polymers/metabolism , Protein Methyltransferases/metabolism , RNA-Dependent RNA Polymerase/metabolism , S-Adenosylhomocysteine/pharmacology , Vesiculovirus/drug effects , Vesiculovirus/metabolism , Viral Proteins/metabolism , Animals , Cell Line , Chlorocebus aethiops , Cricetinae , Methylation , Models, Molecular , Protein Structure, Tertiary , RNA, Messenger/genetics , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/genetics , Substrate Specificity , Transcription, Genetic/genetics , Vesiculovirus/genetics , Viral Proteins/chemistry , Viral Proteins/genetics
11.
Virology ; 536: 78-90, 2019 10.
Article in English | MEDLINE | ID: mdl-31401467

ABSTRACT

Influenza A viruses enter host cells through the endocytic pathway, where acidification triggers conformational changes of the viral hemagglutinin (HA) to drive membrane fusion. During this process, the HA fusion peptide is extruded from its buried position in the neutral pH structure and targeted to the endosomal membrane. Conserved ionizable residues near the fusion peptide may play a role in initiating these structural rearrangements. We targeted highly conserved histidine residues in this region, at HA1 position 17 of Group-2 HA subtypes and HA2 position 111 of Group-1 HA subtypes, to determine their role in fusion activity. WT and mutant HA proteins representing several subtypes were expressed and characterized, revealing that His 111 is essential for HA functional activity of Group-1 subtypes, supporting continued efforts to target this region of the HA structure for vaccination strategies and the design of antiviral compounds.


Subject(s)
Epithelial Cells/virology , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Histidine/chemistry , Influenza A Virus, H2N2 Subtype/chemistry , Influenza A Virus, H5N1 Subtype/chemistry , Membrane Fusion , Animals , Cell Line , Chlorocebus aethiops , Conserved Sequence , Cricetulus , Endosomes/chemistry , Endosomes/metabolism , Endosomes/virology , Epithelial Cells/ultrastructure , Gene Expression , Hemagglutinin Glycoproteins, Influenza Virus/classification , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Histidine/metabolism , Hydrogen-Ion Concentration , Influenza A Virus, H2N2 Subtype/genetics , Influenza A Virus, H2N2 Subtype/metabolism , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/metabolism , Models, Molecular , Phylogeny , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Vero Cells , Virus Internalization
12.
Front Public Health ; 3: 251, 2015.
Article in English | MEDLINE | ID: mdl-26649289

ABSTRACT

Multiple factors ranging from globalization to ecosystem disruption are presenting the global community with evolving biological threats to local, national, and global security that reach beyond the realm of traditional bioweapon threats. As a result, mitigation strategies have adapted necessarily to the increased diversity of biological threats. In general, response and preparedness strategies have largely shifted from being primarily reactive to traditional biological weapons to more proactive in nature. In this review, we briefly explore biological threats through a wider aperture, to embrace a greater appreciation of viral pathogens, antimicrobial resistance, and agricultural pathogens, and their potential to cause civil, economic, and political devastation. In addition, we discuss current mitigation strategies codified by the Global Health Security Agenda and the One Health paradigm as well as some of the available tools to assist with their sustainable implementation.

13.
Vaccine ; 28(38): 6305-13, 2010 Aug 31.
Article in English | MEDLINE | ID: mdl-20656032

ABSTRACT

The use of viral vectors as vaccine candidates has shown promise against a number of pathogens. However, preexisting immunity to these vectors is a concern that must be addressed when deciding which viruses are suitable for use. A number of properties, including the existence of antigenically distinct subtypes, make influenza viruses attractive candidates for use as viral vectors. Here, we evaluate the ability of influenza viral vectors containing inserts of foreign pathogens to elicit antibody and CD8(+) T cell responses against these foreign antigens in the presence of preexisting immunity to influenza virus in mice. Specifically, responses to an H3N1-based vector expressing a 90 amino acid polypeptide derived from the protective antigen (PA) of Bacillus anthracis or an H1N1-based vector containing a CD8(+) T cell epitope from the glycoprotein (GP) of lymphocytic choriomeningitis virus were evaluated following infections with either homosubtypic or heterosubtypic influenza viruses. We found that mice previously infected with influenza viruses, even those expressing HA and NA proteins of completely different subtypes, were severely compromised in their ability to mount an immune response against the inserted epitopes. This inhibition was demonstrated to be mediated by CD8(+) T cells, which recognize multiple strains of influenza viruses. These CD8(+) T cells were further shown to protect mice from a lethal challenge by a heterologous influenza subtype. The implication of these data for the use of influenza virus vectors and influenza vaccination in general are discussed.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Genetic Vectors , Influenza A virus/immunology , Orthomyxoviridae Infections/immunology , Animals , Anthrax Vaccines/immunology , Antibody Formation , Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Cross Reactions , Female , Hemagglutination Inhibition Tests , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Viral Vaccines/immunology
14.
Virology ; 388(2): 286-93, 2009 Jun 05.
Article in English | MEDLINE | ID: mdl-19395055

ABSTRACT

To investigate the polymerase components selectively involved in transcription versus replication of vesicular stomatitis virus (VSV), we sequenced the polymerase gene of a conditionally RNA defective, temperature sensitive VSV: ts(G)114, which has a phenotype upon shift from permissive to non-permissive temperature of shut-down of mRNA transcription and unaffected genome replication. Sequence analysis of the ts(G)114 L gene identified three altered amino acid residues in the L protein. These three changes were specifically engineered individually and in combinations into a functional cDNA clone encoding the VSV genome and tested for association with the temperature sensitive and RNA defective phenotypes in the background of recovered engineered viruses. The data presented in this study show a specific amino acid substitution in domain II of the VSV L protein that significantly affects total RNA synthesis, but when in combination with two additional amino acid substitutions identified in the ts(G)114 L protein, leads to a specific reduction in mRNA transcription, but not replication.


Subject(s)
Gene Expression Regulation, Viral , RNA, Messenger/metabolism , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/metabolism , Temperature , Transcription, Genetic , Vesiculovirus/physiology , Viral Proteins/metabolism , Animals , Cell Line , Chlorocebus aethiops , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/genetics , Vero Cells , Vesiculovirus/genetics , Vesiculovirus/metabolism , Viral Proteins/chemistry , Viral Proteins/genetics , Virion/metabolism , Virus Replication/genetics
15.
Virology ; 394(2): 321-30, 2009 Nov 25.
Article in English | MEDLINE | ID: mdl-19755201

ABSTRACT

A panel of eight single amino acid deletion mutants was generated within the first 24 residues of the fusion peptide domain of the of the hemagglutinin (HA) of A/Aichi/2/68 influenza A virus (H3N2 subtype). The mutant HAs were analyzed for folding, cell surface transport, cleavage activation, capacity to undergo acid-induced conformational changes, and membrane fusion activity. We found that the mutant DeltaF24, at the C-terminal end of the fusion peptide, was expressed in a non-native conformation, whereas all other deletion mutants were transported to the cell surface and could be cleaved into HA1 and HA2 to activate membrane fusion potential. Furthermore, upon acidification these cleaved HAs were able to undergo the characteristic structural rearrangements that are required for fusion. Despite this, all mutants were inhibited for fusion activity based on two separate assays. The results indicate that the mutant fusion peptide domains associate with target membranes in a non-functional fashion, and suggest that structural features along the length of the fusion peptide are likely to be relevant for optimal membrane fusion activity.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/physiology , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/physiology , Sequence Deletion , Viral Fusion Proteins/genetics , Viral Fusion Proteins/physiology , Virus Internalization , Amino Acid Sequence , Animals , Cell Line , Cricetinae , Genes, Viral , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Humans , Hydrogen-Ion Concentration , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Structure, Quaternary , Sequence Homology, Amino Acid , Viral Fusion Proteins/chemistry
16.
Virology ; 382(1): 69-82, 2008 Dec 05.
Article in English | MEDLINE | ID: mdl-18848710

ABSTRACT

The large (L) proteins of non-segmented negative stranded (NNS) RNA viruses contain the core RNA dependent RNA polymerase activity for RNA replication and transcription as well as the activities for polyadenylating and capping the mRNA transcripts and for methylating the cap structures. There is currently no structural information available for these large multi-functional proteins. Phylogenetic analyses have led to the division of the L protein primary structure into six functional domains of high conservation that are linked by variable regions. The studies in this report investigate the role of specific amino acids within domain VI of the VSV L protein, which contains a 2'-O-ribose methyltransferase (MTase) domain. We generated a structural homology model of residues 1644-1842 within domain VI based on the crystal structure determined for the known 2'-O-ribose MTase of E. coli, RrmJ. The information generated by this homology model directed us to residues structurally important for MTase activity and SAM binding. Selected residues were analyzed by site-specific mutagenesis and the mutant L proteins were assayed for their effects on RNA synthesis and cap methylation. The goal of this study was to functionally test the model in order to gain insight into the structural constraints of this region of the L protein. The data presented here revealed specific mutations that affect transcription, replication, and 5' cap methylation, many of which resulted in polymerases temperature sensitive for RNA synthesis.


Subject(s)
Archaeal Proteins/chemistry , Archaeal Proteins/genetics , Methyltransferases/chemistry , Methyltransferases/genetics , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/genetics , Viral Proteins/chemistry , Viral Proteins/genetics , Amino Acid Substitution/genetics , Archaeal Proteins/metabolism , Cell Cycle Proteins/chemistry , Methylation , Methyltransferases/metabolism , Models, Molecular , Mutagenesis, Site-Directed , Protein Structure, Tertiary , RNA Caps/metabolism , RNA, Viral/biosynthesis , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL