Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Crit Care Med ; 49(2): 311-323, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33332817

ABSTRACT

OBJECTIVES: In many jurisdictions, ethical concerns require surrogate humane endpoints to replace death in small animal models of acute lung injury. Heterogenous selection and reporting of surrogate endpoints render interpretation and generalizability of findings between studies difficult. We aimed to establish expert-guided consensus among preclinical scientists and laboratory animal veterinarians on selection and reporting of surrogate endpoints, monitoring of these models, and the use of analgesia. DESIGN: A three-round consensus process, using modified Delphi methodology, with researchers who use small animal models of acute lung injury and laboratory animal veterinarians who provide care for these animals. Statements on the selection and reporting of surrogate endpoints, monitoring, and analgesia were generated through a systematic search of MEDLINE and Embase. Participants were asked to suggest any additional potential statements for evaluation. SETTING: A web-based survey of participants representing the two stakeholder groups (researchers, laboratory animal veterinarians). Statements were rated on level of evidence and strength of support by participants. A final face-to-face meeting was then held to discuss results. SUBJECTS: None. INTERVENTIONS: None. MEASUREMENTS AND MAIN RESULTS: Forty-two statements were evaluated, and 29 were rated as important, with varying strength of evidence. The majority of evidence was based on rodent models of acute lung injury. Endpoints with strong support and evidence included temperature changes and body weight loss. Behavioral signs and respiratory distress also received support but were associated with lower levels of evidence. Participants strongly agreed that analgesia affects outcomes in these models and that none may be necessary following nonsurgical induction of acute lung injury. Finally, participants strongly supported transparent reporting of surrogate endpoints. A prototype composite score was also developed based on participant feedback. CONCLUSIONS: We provide a preliminary framework that researchers and animal welfare committees may adapt for their needs. We have identified knowledge gaps that future research should address.


Subject(s)
Acute Lung Injury/physiopathology , Animal Care Committees/organization & administration , Animal Welfare/standards , Animals, Laboratory , Consensus , Animals , Biomarkers , Humans , Models, Animal , Veterinarians/standards
2.
Sci Transl Med ; 11(511)2019 09 25.
Article in English | MEDLINE | ID: mdl-31554740

ABSTRACT

Variants in the leucine-rich repeat kinase-2 (LRRK2) gene are associated with Parkinson's disease, leprosy, and Crohn's disease, three disorders with inflammation as an important component. Because of its high expression in granulocytes and CD68-positive cells, LRRK2 may have a function in innate immunity. We tested this hypothesis in two ways. First, adult mice were intravenously inoculated with Salmonella typhimurium, resulting in sepsis. Second, newborn mouse pups were intranasally infected with reovirus (serotype 3 Dearing), which induced encephalitis. In both mouse models, wild-type Lrrk2 expression was protective and showed a sex effect, with female Lrrk2-deficient animals not controlling infection as well as males. Mice expressing Lrrk2 carrying the Parkinson's disease-linked p.G2019S mutation controlled infection better, with reduced bacterial growth and longer animal survival during sepsis. This gain-of-function effect conferred by the p.G2019S mutation was mediated by myeloid cells and was abolished in animals expressing a kinase-dead Lrrk2 variant, p.D1994S. Mouse pups with reovirus-induced encephalitis that expressed the p.G2019S Lrrk2 mutation showed increased mortality despite lower viral titers. The p.G2019S mutant Lrrk2 augmented immune cell chemotaxis and generated more reactive oxygen species during virulent infection. Reovirus-infected brains from mice expressing the p.G2019S mutant Lrrk2 contained higher concentrations of α-synuclein. Animals expressing one or two p.D1994S Lrrk2 alleles showed lower mortality from reovirus-induced encephalitis. Thus, Lrrk2 alleles may alter the course of microbial infections by modulating inflammation, and this may be dependent on the sex and genotype of the host as well as the type of pathogen.


Subject(s)
Alleles , Infections/enzymology , Infections/genetics , Inflammation/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Sex Characteristics , Animals , Brain/pathology , Brain/virology , Chemotaxis , Encephalitis/virology , Female , Humans , Infections/immunology , Infections/pathology , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/deficiency , Leukocytes/enzymology , Male , Mice, Inbred C57BL , Mutation/genetics , Reactive Oxygen Species/metabolism , Reoviridae/physiology , Salmonella typhimurium/growth & development , Sepsis/microbiology , Survival Analysis , alpha-Synuclein/metabolism
3.
Virology ; 509: 167-177, 2017 09.
Article in English | MEDLINE | ID: mdl-28646652

ABSTRACT

The influenza A virus RNA polymerase cleaves the 5' ends of host RNAs and uses these RNA fragments as primers for viral mRNA synthesis. We performed deep sequencing of the 5' host-derived ends of the eight viral mRNAs of influenza A/Puerto Rico/8/1934 (H1N1) virus in infected A549 cells, and compared the population to those of A/Hong Kong/1/1968 (H3N2) and A/WSN/1933 (H1N1). In the three strains, the viral RNAs target different populations of host RNAs. Host RNAs are cap-snatched based on their abundance, and we found that RNAs encoding proteins involved in metabolism are overrepresented in the cap-snatched populations. Because this overrepresentation could be a reflection of the host response early after infection, and thus of the increased availability of these transcripts, our results suggest that host RNAs are cap-snatched mainly based on their abundance without preferential targeting.


Subject(s)
Epithelial Cells/virology , Host-Pathogen Interactions , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H3N2 Subtype/genetics , RNA Caps/genetics , RNA Caps/metabolism , Virus Replication , Cell Line , Genetic Variation , Humans , Sequence Analysis, DNA
4.
J Neural Transm (Vienna) ; 124(6): 721-738, 2017 06.
Article in English | MEDLINE | ID: mdl-28477284

ABSTRACT

Braak and Del Tredici have proposed that typical Parkinson disease (PD) has its origins in the olfactory bulb and gastrointestinal tract. However, the role of the olfactory system has insufficiently been explored in the pathogeneses of PD and Alzheimer disease (AD) in laboratory models. Here, we demonstrate applications of a new method to process mouse heads for microscopy by sectioning, mounting, and staining whole skulls ('holocranohistochemistry'). This technique permits the visualization of the olfactory system from the nasal cavity to mitral cells and dopamine-producing interneurons of glomeruli in the olfactory bulb. We applied this method to two specific goals: first, to visualize PD- and AD-linked gene expression in the olfactory system, where we detected abundant, endogenous α-synuclein and tau expression in the olfactory epithelium. Furthermore, we observed amyloid-ß plaques and proteinase-K-resistant α-synuclein species, respectively, in cranial nerve-I of APP- and human SNCA-over-expressing mice. The second application of the technique was to the modeling of gene-environment interactions in the nasal cavity of mice. We tracked the infection of a neurotropic respiratory-enteric-orphan virus from the nose pad into cranial nerves-I (and -V) and monitored the ensuing brain infection. Given its abundance in the olfactory epithelia, we questioned whether α-synuclein played a role in innate host defenses to modify the outcome of infections. Indeed, Snca-null mice were more likely to succumb to viral encephalitis versus their wild-type littermates. Moreover, using a bacterial sepsis model, Snca-null mice were less able to control infection after intravenous inoculation with Salmonella typhimurium. Together, holocranohistochemistry enabled new discoveries related to α-synuclein expression and its function in mice. Future studies will address: the role of Mapt and mutant SNCA alleles in infection paradigms; the contribution of xenobiotics in the initiation of idiopathic PD; and the safety to the host when systemically targeting α-synuclein by immunotherapy.


Subject(s)
Brain/metabolism , Brain/virology , Encephalitis, Viral/virology , Olfactory Mucosa/anatomy & histology , Olfactory Mucosa/metabolism , Reoviridae Infections/virology , alpha-Synuclein/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/diagnostic imaging , Brain/pathology , Disease Models, Animal , Encephalitis, Viral/immunology , Encephalitis, Viral/mortality , Encephalitis, Viral/pathology , Female , Head , Humans , Immunohistochemistry , Male , Mammalian orthoreovirus 3 , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Neural Pathways/anatomy & histology , Neural Pathways/diagnostic imaging , Neural Pathways/metabolism , Neural Pathways/pathology , Olfactory Mucosa/pathology , Olfactory Receptor Neurons/metabolism , Olfactory Receptor Neurons/virology , Reoviridae Infections/immunology , Salmonella Infections/immunology , Salmonella Infections/pathology , Salmonella typhimurium , Tissue Preservation/methods , alpha-Synuclein/genetics
5.
Viruses ; 9(5)2017 05 12.
Article in English | MEDLINE | ID: mdl-28498306

ABSTRACT

The non-structural protein, NS1, is a virulence factor encoded by influenza A viruses (IAVs). In this report, we provide evidence that the conserved residue, tyrosine (Y) 84, in a conserved putative SH2-binding domain in A/Duck/Hubei/2004/L-1 [H5N1] NS1 is critical for limiting an interferon (IFN) response to infection. A phenylalanine (F) substitution of this Y84 residue abolishes NS1-mediated downregulation of IFN-inducible STAT phosphorylation, and surface IFNAR1 expression. Recombinant IAV (rIAV) [H1N1] expressing A/Grey Heron/Hong Kong/837/2004 [H5N1] NS1-Y84F (rWSN-GH-NS1-Y84F) replicates to lower titers in human lung epithelial cells and is more susceptible to the antiviral effects of IFN-ß treatment compared with rIAV expressing the intact H5N1 NS1 (rWSN-GH-NS1-wt). Cells infected with rWSN-GH-NS1-Y84F express higher levels of IFN stimulated genes (ISGs) associated with an antiviral response compared with cells infected with rWSN-GH-NS1-wt. In mice, intranasal infection with rWSN-GH-NS1-Y84F resulted in a delay in onset of weight loss, reduced lung pathology, lower lung viral titers and higher ISG expression, compared with mice infected with rWSN-GH-NS1-wt. IFN-ß treatment of mice infected with rWSN-GH-NS1-Y84F reduced lung viral titers and increased lung ISG expression, but did not alter viral titers and ISG expression in mice infected with rWSN-GH-NS1-wt. Viewed altogether, these data suggest that the virulence associated with this conserved Y84 residue in NS1 is, in part, due to its role in regulating the host IFN response.


Subject(s)
Influenza A Virus, H5N1 Subtype/metabolism , Influenza, Human/virology , Interferons/drug effects , Signal Transduction/drug effects , Viral Nonstructural Proteins/antagonists & inhibitors , Virus Diseases/metabolism , A549 Cells , Animals , Antiviral Agents/pharmacology , Disease Models, Animal , Dogs , Epithelial Cells/virology , Fibroblasts , HEK293 Cells , HeLa Cells , Host-Pathogen Interactions/drug effects , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/metabolism , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza A Virus, H5N1 Subtype/physiology , Influenza A virus/genetics , Influenza A virus/metabolism , Influenza A virus/physiology , Interferon-beta , Lung/pathology , Lung/virology , Madin Darby Canine Kidney Cells , Male , Mice , Mice, Inbred C57BL , Mutagenesis, Site-Directed , Neutrophils/pathology , Neutrophils/virology , Proto-Oncogene Proteins c-akt/metabolism , Reverse Genetics , Transfection , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/physiology , Virulence , Virulence Factors
6.
Virology ; 508: 170-179, 2017 08.
Article in English | MEDLINE | ID: mdl-28554059

ABSTRACT

Influenza A virus (IAV) non-structural protein 1 (NS1) suppresses host innate immune responses by inhibiting type I interferon (IFN) production. We provide evidence that residues F103 and M106 in the CPSF4-binding domain of A/HK/1/68 [H3N2] NS1 contribute to post-transcriptional inhibition of antiviral IFN-stimulated genes (ISGs), thereby suppressing an antiviral type I IFN response. Recombinant (r) IAVs encoding F103L and M106I mutations in NS1 replicate to significantly lower viral titers in human A549 lung epithelial cells and primary type II alveolar cells. In A549 cells, rIAVs encoding these mutant NS1s induce higher levels of IFN-ß production and are more sensitive to the antiviral effects of IFN-ß treatment. qPCR characterization of polysomal mRNA, in the presence or absence of IFN-ß treatment, identified a greater proportion of heavy polysome-associated ISGs including EIF2AK2, OAS1, and MxA in A549 cells infected with rIAVs encoding these CPSF4-binding mutant NS1s, in contrast to rIAV encoding wildtype NS1.


Subject(s)
Cleavage And Polyadenylation Specificity Factor/metabolism , Influenza A Virus, H3N2 Subtype/metabolism , Influenza, Human/metabolism , Interferons/genetics , Peptide Chain Initiation, Translational , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , Amino Acid Motifs , Cleavage And Polyadenylation Specificity Factor/genetics , Host-Pathogen Interactions , Humans , Influenza A Virus, H3N2 Subtype/chemistry , Influenza A Virus, H3N2 Subtype/genetics , Influenza, Human/genetics , Influenza, Human/virology , Interferons/metabolism , Protein Binding , Viral Nonstructural Proteins/genetics
7.
Eur J Neurosci ; 45(1): 175-191, 2017 01.
Article in English | MEDLINE | ID: mdl-27859866

ABSTRACT

Fifty-five years after the concept of dopamine replacement therapy was introduced, Parkinson disease (PD) remains an incurable neurological disorder. To date, no disease-modifying therapeutic has been approved. The inability to predict PD incidence risk in healthy adults is seen as a limitation in drug development, because by the time of clinical diagnosis ≥ 60% of dopamine neurons have been lost. We have designed an incidence prediction model founded on the concept that the pathogenesis of PD is similar to that of many disorders observed in ageing humans, i.e. a complex, multifactorial disease. Our model considers five factors to determine cumulative incidence rates for PD in healthy adults: (i) DNA variants that alter susceptibility (D), e.g. carrying a LRRK2 or GBA risk allele; (ii) Exposure history to select environmental factors including xenobiotics (E); (iii) Gene-environment interactions that initiate pathological tissue responses (I), e.g. a rise in ROS levels, misprocessing of amyloidogenic proteins (foremost, α-synuclein) and dysregulated inflammation; (iv) sex (or gender; G); and importantly, (v) time (T) encompassing ageing-related changes, latency of illness and propagation of disease. We propose that cumulative incidence rates for PD (PR ) can be calculated in healthy adults, using the formula: PR (%) = (E + D + I) × G × T. Here, we demonstrate six case scenarios leading to young-onset parkinsonism (n = 3) and late-onset PD (n = 3). Further development and validation of this prediction model and its scoring system promise to improve subject recruitment in future intervention trials. Such efforts will be aimed at disease prevention through targeted selection of healthy individuals with a higher prediction score for developing PD in the future and at disease modification in subjects that already manifest prodromal signs.


Subject(s)
Gene-Environment Interaction , Parkinson Disease/epidemiology , Parkinson Disease/genetics , Dopamine/metabolism , Humans , Incidence , Mutation/genetics , Parkinson Disease/metabolism , Protein Serine-Threonine Kinases/genetics , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
8.
PLoS Pathog ; 12(11): e1006021, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27814389

ABSTRACT

[This corrects the article DOI: 10.1371/journal.ppat.1005446.].

9.
PLoS Pathog ; 12(2): e1005446, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26928844

ABSTRACT

The immune response to influenza virus infection comprises both innate and adaptive defenses. NK cells play an early role in the destruction of tumors and virally-infected cells. NK cells express a variety of inhibitory receptors, including those of the Ly49 family, which are functional homologs of human killer-cell immunoglobulin-like receptors (KIR). Like human KIR, Ly49 receptors inhibit NK cell-mediated lysis by binding to major histocompatibility complex class I (MHC-I) molecules that are expressed on normal cells. During NK cell maturation, the interaction of NK cell inhibitory Ly49 receptors with their MHC-I ligands results in two types of NK cells: licensed ("functional"), or unlicensed ("hypofunctional"). Despite being completely dysfunctional with regard to rejecting MHC-I-deficient cells, unlicensed NK cells represent up to half of the mature NK cell pool in rodents and humans, suggesting an alternative role for these cells in host defense. Here, we demonstrate that after influenza infection, MHC-I expression on lung epithelial cells is upregulated, and mice bearing unlicensed NK cells (Ly49-deficient NKCKD and MHC-I-deficient B2m-/- mice) survive the infection better than WT mice. Importantly, transgenic expression of an inhibitory self-MHC-I-specific Ly49 receptor in NKCKD mice restores WT influenza susceptibility, confirming a direct role for Ly49. Conversely, F(ab')2-mediated blockade of self-MHC-I-specific Ly49 inhibitory receptors protects WT mice from influenza virus infection. Mechanistically, perforin-deficient NKCKD mice succumb to influenza infection rapidly, indicating that direct cytotoxicity is necessary for unlicensed NK cell-mediated protection. Our findings demonstrate that Ly49:MHC-I interactions play a critical role in influenza virus pathogenesis. We suggest a similar role may be conserved in human KIR, and their blockade may be protective in humans.


Subject(s)
Antigens, Ly/metabolism , Immune Evasion , Influenza A virus/immunology , Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily A/metabolism , Orthomyxoviridae Infections/immunology , Receptors, KIR/metabolism , Respiratory Mucosa/immunology , Animals , Antigens, Ly/genetics , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , Crosses, Genetic , Immunity, Innate , Influenza A virus/physiology , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Killer Cells, Natural/virology , Lung/immunology , Lung/metabolism , Lung/pathology , Lung/virology , Mice, Knockout , Mice, Transgenic , NK Cell Lectin-Like Receptor Subfamily A/agonists , NK Cell Lectin-Like Receptor Subfamily A/antagonists & inhibitors , NK Cell Lectin-Like Receptor Subfamily A/genetics , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/metabolism , Receptors, KIR/agonists , Receptors, KIR/antagonists & inhibitors , Receptors, KIR/genetics , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Respiratory Mucosa/virology , Specific Pathogen-Free Organisms , Survival Analysis , beta 2-Microglobulin/genetics , beta 2-Microglobulin/metabolism
10.
Sci Rep ; 4: 6181, 2014 Aug 26.
Article in English | MEDLINE | ID: mdl-25154590

ABSTRACT

The influenza A virus RNA polymerase cleaves the 5' end of host pre-mRNAs and uses the capped RNA fragments as primers for viral mRNA synthesis. We performed deep sequencing of the 5' ends of viral mRNAs from all genome segments transcribed in both human (A549) and mouse (M-1) cells infected with the influenza A/HongKong/1/1968 (H3N2) virus. In addition to information on RNA motifs present, our results indicate that the host primers are divergent between the viral transcripts. We observed differences in length distributions, nucleotide motifs and the identity of the host primers between the viral mRNAs. Mapping the reads to known transcription start sites indicates that the virus targets the most abundant host mRNAs, which is likely caused by the higher expression of these genes. Our findings suggest negligible competition amongst RdRp:vRNA complexes for individual host mRNA templates during cap-snatching and provide a better understanding of the molecular mechanism governing the first step of transcription of this influenza strain.


Subject(s)
Influenza A Virus, H3N2 Subtype/genetics , Transcription, Genetic , 5' Untranslated Regions , Animals , Base Sequence , Cell Line, Tumor , Chromosome Mapping , Consensus Sequence , DNA Primers/genetics , Gene Expression Regulation, Viral , Gene Ontology , Genes, Viral , High-Throughput Nucleotide Sequencing , Humans , Mice , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Viral/genetics , Sequence Analysis, DNA , Transcription Initiation Site
11.
Biochem Biophys Res Commun ; 441(1): 226-9, 2013 Nov 08.
Article in English | MEDLINE | ID: mdl-24140051

ABSTRACT

All influenza viral neuraminidases (NA) of both type A and B viruses have only one universally conserved sequence located between amino acids 222-230. A monoclonal antibody against this region has been previously reported to provide broad inhibition against all nine subtypes of influenza A NA; yet its inhibitory effect against influenza B viral NA remained unknown. Here, we report that the monoclonal antibody provides a broad inhibition against various strains of influenza B viruses of both Victoria and Yamagata genetic lineage. Moreover, the growth and NA enzymatic activity of two drug resistant influenza B strains (E117D and D197E) are also inhibited by the antibody even though these two mutations are conformationally proximal to the universal epitope. Collectively, these data suggest that this unique, highly-conserved linear sequence in viral NA is exposed sufficiently to allow access by inhibitory antibody during the course of infection; it could represent a potential target for antiviral agents and vaccine-induced immune responses against diverse strains of type B influenza virus.


Subject(s)
Antibodies, Monoclonal/immunology , Conserved Sequence , Drug Resistance, Viral/immunology , Epitopes/immunology , Influenza B virus/enzymology , Influenza, Human/prevention & control , Neuraminidase/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Viral/immunology , Dogs , Drug Resistance, Viral/drug effects , Drug Resistance, Viral/genetics , Epitopes/chemistry , Humans , Influenza B virus/drug effects , Influenza B virus/growth & development , Influenza B virus/immunology , Influenza, Human/immunology , Influenza, Human/virology , Madin Darby Canine Kidney Cells , Models, Molecular , Molecular Sequence Data , Mutation/genetics , Neuraminidase/antagonists & inhibitors , Neuraminidase/chemistry
12.
Antiviral Res ; 100(2): 567-74, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24091204

ABSTRACT

The only universally conserved sequence amongst all influenza A viral neuraminidase (NA) is located between amino acids 222-230 and plays crucial roles in viral replication. However, it remained unclear as to whether this universal epitope is exposed during the course of infection to allow binding and inhibition by antibodies. Using a monoclonal antibody (MAb) targeting this specific epitope, we demonstrated that all nine subtypes of NA were inhibited in vitro by the MAb. Moreover, the antibody also provided heterosubtypic protection in mice challenged with lethal doses of mouse-adapted H1N1 and H3N2, which represent group I and II viruses, respectively. Furthermore, we report amino acid residues I222 and E227, located in close proximity to the active site, are indispensable for inhibition by this antibody. This unique, highly-conserved linear sequence in viral NA could be an attractive immunological target for protection against diverse strains of influenza viruses.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Cross Protection , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Neuraminidase/immunology , Orthomyxoviridae Infections/prevention & control , Viral Proteins/immunology , Animals , Antibodies, Monoclonal/isolation & purification , Antibodies, Viral/isolation & purification , Disease Models, Animal , Epitopes, B-Lymphocyte/immunology , Female , Mice , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology
13.
Virol J ; 10: 243, 2013 Jul 25.
Article in English | MEDLINE | ID: mdl-23886034

ABSTRACT

BACKGROUND: The genetic basis for avian to mammalian host switching in influenza A virus is largely unknown. The human A/HK/156/1997 (H5N1) virus that transmitted from poultry possesses NS1 gene mutations F103L + M106I that are virulence determinants in the mouse model of pneumonia; however their individual roles have not been determined. The emergent A/Shanghai/patient1/2013(H7N9)-like viruses also possess these mutations which may contribute to their virulence and ability to switch species. METHODS: NS1 mutant viruses were constructed by reverse genetics and site directed mutagenesis on human and mouse-adapted backbones. Mouse infections assessed virulence, virus yield, tissue infection, and IFN induction. NS1 protein properties were assessed for subcellular distribution, IFN antagonism (mouse and human), CPSF30 and RIG-I domain binding, host transcription (microarray); and the natural prevalence of 103L and 106I mutants was assessed. RESULTS: Each of the F103L and M106I mutations contributes additively to virulence to reduce the lethal dose by >800 and >3,200 fold respectively by mediating alveolar tissue infection with >100 fold increased infectious yields. The 106I NS1 mutant lost CPSF binding but the 103L mutant maintained binding that correlated with an increased general decrease in host gene expression in human but not mouse cells. Each mutation positively modulated the inhibition of IFN induction in mouse cells and activation of the IFN-ß promoter in human cells but not in combination in human cells indicating negative epistasis. Each of the F103L and M106I mutations restored a defect in cytoplasmic localization of H5N1 NS1 in mouse cells. Human H1N1 and H3N2 NS1 proteins bound to the CARD, helicase and RD RIG-I domains, whereas the H5N1 NS1 with the same consensus 103F and 106M mutations did not bind these domains, which was totally or partially restored by the M106I or F103L mutations respectively. CONCLUSIONS: The F103L and M106I mutations in the H5N1 NS1 protein each increased IFN antagonism and mediated interstitial pneumonia in mice that was associated with increased cytoplasmic localization and altered host factor binding. These mutations may contribute to the ability of previous HPAI H5N1 and recent LPAI H7N9 and H6N1 (NS1-103L+106M) viruses to switch hosts and cause disease in humans.


Subject(s)
Cleavage And Polyadenylation Specificity Factor/metabolism , DEAD-box RNA Helicases/metabolism , Influenza A Virus, H5N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/pathogenicity , Interferons/antagonists & inhibitors , Mutation, Missense , Viral Nonstructural Proteins/metabolism , Amino Acid Substitution , Animals , DEAD Box Protein 58 , Female , Host-Pathogen Interactions , Humans , Influenza A Virus, H5N1 Subtype/isolation & purification , Lung/pathology , Lung/virology , Lung Diseases, Interstitial/pathology , Lung Diseases, Interstitial/virology , Mice , Mutagenesis, Site-Directed , Mutant Proteins/genetics , Mutant Proteins/metabolism , Reverse Genetics , Viral Nonstructural Proteins/genetics , Virulence , Virulence Factors/genetics , Virulence Factors/metabolism
14.
J Biol Chem ; 288(25): 18283-9, 2013 Jun 21.
Article in English | MEDLINE | ID: mdl-23645684

ABSTRACT

The only universally conserved sequence among all influenza A viral neuraminidases is located between amino acids 222 and 230. However, the potential roles of these amino acids remain largely unknown. Through an array of experimental approaches including mutagenesis, reverse genetics, and growth kinetics, we found that this sequence could markedly affect viral replication. Additional experiments revealed that enzymes with mutations in this region demonstrated substantially decreased catalytic activity, substrate binding, and thermostability. Consistent with viral replication analyses and enzymatic studies, protein modeling suggests that these amino acids could either directly bind to the substrate or contribute to the formation of the active site in the enzyme. Collectively, these findings reveal the essential role of this unique region in enzyme function and viral growth, which provides the basis for evaluating the validity of this sequence as a potential target for antiviral intervention and vaccine development.


Subject(s)
Epitopes/metabolism , Influenza A virus/enzymology , Neuraminidase/metabolism , Viral Proteins/metabolism , Virus Replication , Amino Acid Substitution , Animals , Binding Sites/genetics , Biocatalysis , Catalytic Domain , Cell Line , Chick Embryo , Enzyme Stability/genetics , Epitopes/chemistry , Epitopes/genetics , HEK293 Cells , Humans , Influenza A virus/genetics , Kinetics , Models, Molecular , Mutation , Neuraminidase/chemistry , Neuraminidase/genetics , Protein Structure, Tertiary , Substrate Specificity , Temperature , Viral Proteins/chemistry , Viral Proteins/genetics
15.
Virol J ; 10: 67, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23453057

ABSTRACT

BACKGROUND: Because mammalian reoviruses are isolated from the respiratory tract we modeled the natural history of respiratory infection of adult and suckling mice with T1 Lang (T1L) and T3 Dearing (T3D) reoviruses. METHODS: Adult and suckling Balb/c mice were infected by the intranasal route and were assessed for dose response of disease as well as viral replication in the lung and other organs. Viral antigen was assessed by immunofluorescence and HRP staining of tissue sections and histopathology was assessed on formalin fixed, H + E stained tissue sections. RESULTS: Intranasal infection of adult mice resulted in fatal respiratory distress for high doses (10(7) pfu) of T1L but not T3D. In contrast both T1L and T3D killed suckling mice at moderate viral dosages (10(5) pfu) but differed in clinical symptoms where T1L induced respiratory failure and T3D caused encephalitis. Infections caused transient viremia that resulted in spread to peripheral tissues where disease correlated with virus replication, and pathology. Immunofluorescent staining of viral antigens in the lung showed reovirus infection was primarily associated with alveoli with lesser involvement of bronchiolar epithelium. Immunofluorescent and HRP staining of viral antigens in brain showed infection of neurons by T3D and glial cells by T1L. CONCLUSIONS: These mouse models of reovirus respiratory infection demonstrated age and strain dependent disease that are expected to be relevant to understanding and modulating natural and therapeutic reovirus infections in humans.


Subject(s)
Antigens, Viral/immunology , Encephalitis, Viral/virology , Orthoreovirus, Mammalian/physiology , Pneumonia, Viral/virology , Reoviridae Infections/virology , Respiratory Tract Infections/virology , Age Factors , Animals , Animals, Suckling , Brain/pathology , Brain/virology , Cell Line , Disease Models, Animal , Encephalitis, Viral/pathology , Female , Humans , Liver/pathology , Liver/virology , Lung/pathology , Lung/virology , Mice , Mice, Inbred BALB C , Orthoreovirus, Mammalian/growth & development , Orthoreovirus, Mammalian/immunology , Pneumonia, Viral/pathology , Reoviridae Infections/pathology , Respiratory Tract Infections/pathology , Species Specificity , Time Factors , Viremia , Virus Replication
16.
J Gen Virol ; 94(Pt 3): 593-605, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23223621

ABSTRACT

Influenza A virus (IAV) non-structural protein 1 (NS1) has multiple functions, is essential for virus replication and may be a good target for IAV diagnosis. To generate broadly cross-reactive NS1-specific mAbs, mice were immunized with A/Hong Kong/1/1968 (H3N2) 6×His-tagged NS1 and hybridomas were screened with glutathione S-transferase-conjugated NS1 of A/Puerto Rico/8/1934 (H1N1). mAbs were isotyped and numerous IgG-type clones were characterized further. Most clones specifically recognized NS1 from various H1N1 and H3N2 IAV types by both immunoblot and immunofluorescence microscopy in mouse M1, canine Madin-Darby canine kidney and human A549 cells. mAb epitopes were mapped by overlapping peptides and selective reactivity to the newly described viral NS3 protein. These mAbs detected NS1 in both the cytoplasm and nucleus by immunostaining, and some detected NS1 as early as 5 h post-infection, suggesting their potential diagnostic use for tracking productive IAV replication and characterizing NS1 structure and function. It was also demonstrated that the newly identified NS3 protein is localized in the cytoplasm to high levels.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Influenza A virus/classification , Influenza A virus/immunology , Viral Nonstructural Proteins/immunology , Amino Acid Sequence , Animals , Antibody Specificity , Cell Line , Dogs , Epitope Mapping , Humans , Mice , Models, Molecular , Molecular Sequence Data , Phylogeny , Protein Conformation
17.
PLoS One ; 8(12): e84673, 2013.
Article in English | MEDLINE | ID: mdl-24391972

ABSTRACT

The NS1 protein of influenza A virus (IAV) is a multifunctional virulence factor. We have previously characterized gain-of-function mutations in the NS1 protein arising from the experimental adaptation of the human isolate A/Hong Kong/1/1968(H3N2) (HK) to the mouse. The majority of these mouse adapted NS1 mutations were demonstrated to increase virulence, viral fitness, and interferon antagonism, but differ in binding to the post-transcriptional processing factor cleavage and polyadenylation specificity factor 30 (CPSF30). Because nuclear trafficking is a major genetic determinant of influenza virus host adaptation, we assessed subcellular localization and host gene expression of NS1 adaptive mutations. Recombinant HK viruses with adaptive mutations in the NS1 gene were assessed for NS1 protein subcellular localization in mouse and human cells using confocal microscopy and cellular fractionation. In human cells the HK wild-type (HK-wt) virus NS1 protein partitioned equivalently between the cytoplasm and nucleus but was defective in cytoplasmic localization in mouse cells. Several adaptive mutations increased the proportion of NS1 in the cytoplasm of mouse cells with the greatest effects for mutations M106I and D125G. The host gene expression profile of the adaptive mutants was determined by microarray analysis of infected mouse cells to show either high or low extents of host-gene regulation (HGR or LGR) phenotypes. While host genes were predominantly down regulated for the HGR group of mutants (D2N, V23A, F103L, M106I+L98S, L98S, M106V, and M106V+M124I), the LGR phenotype mutants (D125G, M106I, V180A, V226I, and R227K) were characterized by a predominant up regulation of host genes. CPSF30 binding affinity of NS1 mutants did not predict effects on host gene expression. To our knowledge this is the first report of roles of adaptive NS1 mutations that impact intracellular localization and regulation of host gene expression.


Subject(s)
Adaptation, Biological/genetics , Cytoplasm/metabolism , Gene Expression Regulation/genetics , Host-Pathogen Interactions/genetics , Influenza A Virus, H3N2 Subtype/genetics , Mutation/genetics , Viral Nonstructural Proteins/genetics , Animals , Cell Fractionation , Cell Line , Densitometry , Dogs , Gene Expression Profiling , Humans , Mice , Microarray Analysis , Microscopy, Confocal , Viral Nonstructural Proteins/metabolism
18.
Avian Dis ; 56(3): 597-600, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23050481

ABSTRACT

Influenza viruses from domestic aquatic birds can be transmitted to chickens, resulting in continued prevalence of the disease. H3 viruses are one of the most frequently identified subtypes in domestic ducks. Results from our previous serologic study suggested that H3 virus infections potentially exist in chickens with a wide geographical distribution in China. To better understand their pathogenic potential, two H3N8 influenza viruses isolated from domestic ducks were selected for experimental infections in chickens. We found that viral shedding lasted for at least 14 days postinfection for both viruses; however, one virus caused mortality in the chickens when coinfected with Escherichia coli. Sequencing of the viral HA gene isolated from the inoculated chickens revealed two amino acid mutations within the gene. These findings demonstrate the pathogenicity of the H3N8 domestic duck influenza viruses to chickens, highlighting the need for routine epidemiologic investigations of H3 subtype influenza viruses in chicken populations.


Subject(s)
Chickens , Ducks , Escherichia coli Infections/veterinary , Influenza A Virus, H3N8 Subtype/pathogenicity , Influenza in Birds/virology , Animals , Diarrhea/microbiology , Diarrhea/veterinary , Diarrhea/virology , Escherichia coli Infections/complications , Feces/virology , Influenza A Virus, H3N8 Subtype/classification , Influenza in Birds/complications , Virus Shedding
19.
J Med Virol ; 84(10): 1571-85, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22930505

ABSTRACT

Chronic hepatitis C virus (HCV) infection is one of the major causes of chronic liver disease worldwide. In order for HCV to persist, the virus must escape immune recognition or inhibit the host immune response. The NS5A protein contains the interferon sensitivity-determining region (ISDR) and is able to repress dsRNA-dependent protein kinase (PKR) thus influencing the response to interferon (IFN) therapy. Patients who respond to IFN therapy have stronger antibody reactivity against the NS5A compared to IFN non-responders. Therefore, given the possible role for the ISDR in IFN resistance and differential antibody reactivity, it is possible that variation in ISDR may be involved in viral immune escape and development of persistent HCV infection employing aspects of host mimicry. In this study, pre-treatment samples obtained from HCV infected patients were used to investigate the effect of different NS5A ISDR variants on the IFN antiviral response and their involvement in immune evasion. The NS5A was identified as a homologue of the variable region of immunoglobulins (Ig). The IFN resistant genotypes had higher levels of similarity to Ig compared to IFN sensitive genotypes. Expression of NS5A-6003 (HCV genotype 1b) and NS5A-6074 (HCV genotype 2a) was able to rescue vesicular stomatitis virus (VSV) from IFN inhibition and restore luciferase activity. A correlation between Ig-like NS5A structure and also antibody response with the outcome of IFN treatment was observed.


Subject(s)
Antiviral Agents/administration & dosage , Hepacivirus/drug effects , Hepacivirus/immunology , Hepatitis C, Chronic/drug therapy , Immune Evasion , Interferons/administration & dosage , Molecular Mimicry , Cell Line , Genes, Reporter , Hepatitis C Antibodies/blood , Hepatitis C, Chronic/immunology , Hepatitis C, Chronic/virology , Humans , Immunoglobulin G/genetics , Interferons/immunology , Luciferases/analysis , Sequence Homology, Amino Acid , Treatment Outcome , Vesiculovirus/genetics , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/immunology , Viral Plaque Assay
20.
PLoS One ; 7(7): e40752, 2012.
Article in English | MEDLINE | ID: mdl-22808250

ABSTRACT

H9N2 influenza viruses have been circulating worldwide in multiple avian species and have repeatedly infected humans to cause typical disease. The continued avian-to-human interspecies transmission of H9N2 viruses raises concerns about the possibility of viral adaption with increased virulence for humans. To investigate the genetic basis of H9N2 influenza virus host range and pathogenicity in mammals, we generated a mouse-adapted H9N2 virus (SD16-MA) that possessed significantly higher virulence than wide-type virus (SD16). Increased virulence was detectable after 8 sequential lung passages in mice. Five amino acid substitutions were found in the genome of SD16-MA compared with SD16 virus: PB2 (M147L, V250G and E627K), HA (L226Q) and M1 (R210K). Assessments of replication in mice showed that all of the SD16-MA PB2, HA and M1 genome segments increased virus replication; however, only the mouse-adapted PB2 significantly increased virulence. Although the PB2 E627K amino acid substitution enhanced viral polymerase activity and replication, none of the single mutations of mouse adapted PB2 could confer increased virulence on the SD16 backbone. The combination of M147L and E627K significantly enhanced viral replication ability and virulence in mice. Thus, our results show that the combination of PB2 amino acids at position 147 and 627 is critical for the increased pathogenicity of H9N2 influenza virus in mammalian host.


Subject(s)
Adaptation, Biological/genetics , Amino Acid Substitution/genetics , Influenza A Virus, H9N2 Subtype/genetics , Influenza A Virus, H9N2 Subtype/pathogenicity , Mutation/genetics , Viral Proteins/genetics , Animals , Cytokines/metabolism , DNA-Directed RNA Polymerases/metabolism , Female , Humans , Influenza A Virus, H9N2 Subtype/enzymology , Influenza A Virus, H9N2 Subtype/growth & development , Kinetics , Lung/immunology , Lung/virology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/virology , Reassortant Viruses/genetics , Survival Analysis , Viral Matrix Proteins/genetics , Virulence/genetics , Virus Replication/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...