Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 13(6): 543-550, 2012 Apr 29.
Article in English | MEDLINE | ID: mdl-22544393

ABSTRACT

Type I interferon is an integral component of the antiviral response, and its production is tightly controlled at the levels of transcription and translation. The eukaryotic translation-initiation factor eIF4E is a rate-limiting factor whose activity is regulated by phosphorylation of Ser209. Here we found that mice and fibroblasts in which eIF4E cannot be phosphorylated were less susceptible to virus infection. More production of type I interferon, resulting from less translation of Nfkbia mRNA (which encodes the inhibitor IκBα), largely explained this phenotype. The lower abundance of IκBα resulted in enhanced activity of the transcription factor NF-κB, which promoted the production of interferon-Ɵ (IFN-Ɵ). Thus, regulated phosphorylation of eIF4E has a key role in antiviral host defense by selectively controlling the translation of an mRNA that encodes a critical suppressor of the innate antiviral response.


Subject(s)
Eukaryotic Initiation Factor-4E/metabolism , Interferon Type I/biosynthesis , NF-kappa B/metabolism , Vesicular Stomatitis/immunology , Vesicular stomatitis Indiana virus/physiology , Animals , Electrophoretic Mobility Shift Assay , Eukaryotic Initiation Factor-4E/immunology , Female , I-kappa B Proteins/biosynthesis , I-kappa B Proteins/genetics , I-kappa B Proteins/immunology , Immunity, Innate/immunology , Immunoblotting , Interferon Type I/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-KappaB Inhibitor alpha , NF-kappa B/immunology , Phosphorylation , Protein Biosynthesis , RNA, Messenger/chemistry , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Specific Pathogen-Free Organisms , Vesicular Stomatitis/genetics , Vesicular Stomatitis/metabolism , Vesicular Stomatitis/virology , Vesicular stomatitis Indiana virus/immunology , Virus Replication
2.
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
3.
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
4.
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.].

5.
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 ≥Ā 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 (%)Ā =Ā (EĀ +Ā DĀ +Ā I)Ā Ć—Ā GĀ Ć—Ā 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
6.
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-Ɵ 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
7.
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
8.
J Immunol ; 188(8): 3949-60, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22427645

ABSTRACT

Seasonal influenza outbreaks and recurrent influenza pandemics present major challenges to public health. By studying immunological responses to influenza in different host species, it may be possible to discover common mechanisms of susceptibility in response to various influenza strains. This could lead to novel therapeutic targets with wide clinical application. Using a mouse-adapted strain of influenza (A/HK/1/68-MA20 [H3N2]), we produced a mouse model of severe influenza that reproduces the hallmark high viral load and overexpression of cytokines associated with susceptibility to severe influenza in humans. We mapped genetic determinants of the host response using a panel of 29 closely related mouse strains (AcB/BcA panel of recombinant congenic strains) created from influenza-susceptible A/J and influenza-resistant C57BL/6J (B6) mice. Combined clinical quantitative trait loci (QTL) and lung expression QTL mapping identified candidate genes for two sex-specific QTL on chromosomes 2 and 17. The former includes the previously described Hc gene, a deficit of which is associated with the susceptibility phenotype in females. The latter includes the phospholipase gene Pla2g7 and Tnfrsf21, a member of the TNFR superfamily. Confirmation of the gene underlying the chromosome 17 QTL may reveal new strategies for influenza treatment.


Subject(s)
Chromosomes, Mammalian/genetics , Influenza A Virus, H3N2 Subtype , Influenza, Human/genetics , Quantitative Trait Loci , 1-Alkyl-2-acetylglycerophosphocholine Esterase , Alleles , Animals , Chromosome Mapping , Chromosomes, Mammalian/immunology , Disease Models, Animal , Disease Susceptibility , Female , Genotype , Host Specificity , Humans , Influenza, Human/immunology , Influenza, Human/virology , Lung/immunology , Lung/virology , Male , Mice , Mice, Congenic , Mice, Inbred C57BL , Phenotype , Phospholipases A2/genetics , Phospholipases A2/immunology , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/immunology , Sex Factors
9.
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) 6ƗHis-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
10.
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
11.
J Virol ; 86(4): 1942-54, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22171255

ABSTRACT

Influenza A virus NS1 protein has multiple functions in the infected cell during the virus life cycle. Identification of novel cellular factors that interact with NS1 and understanding their functions in virus infection are of great interest. Recombinant viruses carrying a tagged NS1 are valuable for investigation of interactions between NS1 and cellular factors in the context of virus infection. Here, we report the generation of replication-competent recombinant influenza A viruses bearing a Strep tag in the NS1 protein. Purification of a protein complex associated with Strep-tagged NS1 from virus-infected cells followed by mass spectrometry revealed a number of attractive host factors. Among them, we focused our study on RNA helicase A (RHA) in this report. Through biomedical and functional analyses, we demonstrated that RHA interacts with NS1 in an RNA-dependent manner. Knockdown of RHA resulted in a significant reduction on virus yield and polymerase activity in a minigenome assay. Our cell-free viral genome replication assay showed that viral RNA replication and transcription can be enhanced by addition of RHA, and the enhanced effect of RHA required its ATP-dependent helicase activity. In summary, we established a system to identify cellular factors that interact with NS1 protein during virus infection and furthermore demonstrated that RHA interacts with NS1 and enhances viral replication and transcription.


Subject(s)
DEAD-box RNA Helicases/metabolism , Influenza A Virus, H1N1 Subtype/physiology , Influenza, Human/enzymology , Neoplasm Proteins/metabolism , Viral Nonstructural Proteins/metabolism , Virus Replication , Animals , Cell Line , Chick Embryo , DEAD-box RNA Helicases/genetics , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A virus/genetics , Influenza A virus/physiology , Influenza, Human/genetics , Influenza, Human/virology , Neoplasm Proteins/genetics , Protein Binding , Viral Nonstructural Proteins/genetics
12.
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
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-Ɵ 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 Immunol ; 187(8): 4280-92, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21908731

ABSTRACT

Immunopathology is a major cause of influenza-associated morbidity and mortality worldwide. However, the role and regulatory mechanisms of CD4 T cells in severe lung immunopathology following acute influenza infection are poorly understood. In this paper, we report that the emergence of immunopathogenic CD4 T cells is under the control of a transmembrane immunoadaptor DAP12 pathway during influenza infection. We find that the mice lacking DAP12 have unaltered viral clearance but easily succumb to influenza infection as a result of uncontrolled immunopathology. Such immunopathology is associated with markedly increased CD4 T cells displaying markedly increased cytotoxicity and Fas ligand expression. Furthermore, the immunopathogenic property of these CD4 T cells is transferrable. Thus, depletion of CD4 T cells or abrogation of Fas/Fas ligand signaling pathway improves survival and immunopathology. We further find that DAP12 expressed by dendritic cells plays an important role in controlling the immunopathogenic CD4 T cells during influenza infection. Our findings identify a novel pathway that controls the level of immune-pathogenic CD4 T cells during acute influenza infection.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , CD4-Positive T-Lymphocytes/immunology , Influenza A Virus, H1N1 Subtype , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/pathology , Respiratory Tract Infections/immunology , Respiratory Tract Infections/pathology , Adoptive Transfer , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/immunology , Cell Separation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumonia/immunology , Pneumonia/pathology , Pneumonia/virology , Respiratory Tract Infections/virology , Signal Transduction/immunology
15.
J Gen Virol ; 93(Pt 8): 1649-1657, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22592261

ABSTRACT

The first confirmed outbreak of highly pathogenic avian influenza (HPAI) virus infections in North America was caused by A/turkey/Ontario/7732/1966 (H5N9); however, the phylogeny of this virus is largely unknown. This study performed genomic sequence analysis of 11 avian influenza isolates from 1956 to 1979 for comparison with A/turkey/Ontario/7732/1966 (H5N9). Phylogenetic and genetic analyses included these viruses in combination with all known full-genome sequences of avian viruses isolated before 1981. It was shown that a low-pathogenic avian influenza virus, A/turkey/Ontario/6213/1966 (H5N1), that had been isolated 3 months previously, was the closest known genetic relative with six genome segments of common lineage encoding the polymerase subunits PB2, PB1 and PA, nucleoprotein (NP), haemagglutinin (HA) and non-structural (NS) proteins. The lineages of these genome segments included reassortment with other North American turkey viruses that were all rooted in North American wild waterfowl with the HA gene originating from the H5N2 serotype. The phylogenies demonstrated adaptation from North American wild birds to turkeys with the possible involvement of domestic waterfowl. The turkey isolate, A/turkey/Wisconsin/1968 (H5N9), was the second most closely related poultry isolate to A/turkey/Ontario/7732/1966 (H5N9), possessing five common lineage genome segments (PB2, PB1, PA, HA and neuraminidase). The A/turkey/Ontario/6213/1966 (H5N1) virus was more virulent than A/turkey/Wisconsin/68 (H5N9) for chicken embryos and mice, indicating a greater biological similarity to A/turkey/Ontario/7732/1966 (H5N9). Thus, A/turkey/Ontario/6213/1966 (H5N1) was identified as the closest known ancestral relative of HPAI A/turkey/Ontario/7732/1966 (H5N9), which will serve as a useful reference virus for characterizing the early genetic and biological properties associated with the emergence of pathogenic avian influenza strains.


Subject(s)
Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza A virus/pathogenicity , Influenza in Birds/virology , Reassortant Viruses/pathogenicity , Amino Acid Sequence , Animals , Chick Embryo , Evolution, Molecular , Gene Expression Regulation, Viral , Genome, Viral , Hemagglutinins/chemistry , Hemagglutinins/genetics , Hemagglutinins/metabolism , Influenza A Virus, H5N1 Subtype/genetics , Influenza A virus/genetics , Influenza in Birds/epidemiology , Mice , Molecular Sequence Data , North America/epidemiology , Phylogeny , Poultry , Reassortant Viruses/genetics , Virulence
16.
Am J Pathol ; 179(6): 2963-76, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22001698

ABSTRACT

Lung immunopathology is the main cause of influenza-mediated morbidity and death, and much of its molecular mechanisms remain unclear. Whereas tumor necrosis factor-α (TNF-α) is traditionally considered a proinflammatory cytokine, its role in influenza immunopathology is unresolved. We have investigated this issue by using a model of acute H1N1 influenza infection established in wild-type and TNF-α-deficient mice and evaluated lung viral clearance, inflammatory responses, and immunopathology. Whereas TNF-α was up-regulated in the lung after influenza infection, it was not required for normal influenza viral clearance. However, TNF-α deficiency led not only to a greater extent of illness but also to heightened lung immunopathology and tissue remodeling. The severe lung immunopathology was associated with increased inflammatory cell infiltration, anti-influenza adaptive immune responses, and expression of cytokines such as monocyte chemoattractant protein-1 (MCP-1) and fibrotic growth factor, TGF-Ɵ1. Thus, in vivo neutralization of MCP-1 markedly attenuated lung immunopathology and blunted TGF-Ɵ1 production following influenza infection in these hosts. On the other hand, in vivo transgenic expression of MCP-1 worsened lung immunopathology following influenza infection in wild-type hosts. Thus, TNF-α is dispensable for influenza clearance; however, different from the traditional belief, this cytokine is critically required for negatively regulating the extent of lung immunopathology during acute influenza infection.


Subject(s)
Influenza A Virus, H1N1 Subtype/immunology , Orthomyxoviridae Infections/immunology , Pneumonia, Viral/immunology , Tumor Necrosis Factor-alpha/physiology , Adaptive Immunity , Animals , Body Weight , Bronchoalveolar Lavage Fluid , Chemokine CCL2/deficiency , Chemokine CCL2/metabolism , Chemokines/metabolism , Cytokines/metabolism , Immunity, Cellular , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/metabolism
17.
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
18.
J Immunol ; 184(4): 2048-56, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20083661

ABSTRACT

Influenza viral infection is well-known to predispose to subsequent bacterial superinfection in the lung but the mechanisms have remained poorly defined. We have established a murine model of heterologous infections by an H1N1 influenza virus and Staphylococcus aureus. We found that indeed prior influenza infection markedly increased the susceptibility of mice to secondary S. aureus superinfection. Severe sickness and heightened bacterial infection in flu and S. aureus dual-infected animals were associated with severe immunopathology in the lung. We further found that flu-experienced lungs had an impaired NK cell response in the airway to subsequent S. aureus bacterial infection. Thus, adoptive transfer of naive NK cells to the airway of prior flu-infected mice restored flu-impaired antibacterial host defense. We identified that TNF-alpha production of NK cells played an important role in NK cell-mediated antibacterial host defense as NK cells in flu-experienced lungs had reduced TNF-alpha expression and adoptive transfer of TNF-alpha-deficient NK cells to the airway of flu-infected mice failed to restore flu-impaired antibacterial host defense. Defected NK cell function was found to be an upstream mechanism of depressed antibacterial activities by alveolar macrophages as contrast to naive wild-type NK cells, the NK cells from flu-infected or TNF-alpha-deficient mice failed to enhance S. aureus phagocytosis by alveolar macrophages. Together, our study identifies the weakened NK cell response in the lung to be a novel critical mechanism for flu-mediated susceptibility to bacterial superinfection.


Subject(s)
Influenza A Virus, H1N1 Subtype/immunology , Killer Cells, Natural/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/microbiology , Pneumonia, Bacterial/immunology , Pneumonia, Viral/immunology , Staphylococcal Infections/immunology , Superinfection/immunology , Animals , Disease Susceptibility/immunology , Disease Susceptibility/pathology , Female , Killer Cells, Natural/microbiology , Killer Cells, Natural/virology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/microbiology , Macrophages, Alveolar/pathology , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/pathology , Pneumonia, Bacterial/pathology , Pneumonia, Bacterial/virology , Pneumonia, Viral/microbiology , Pneumonia, Viral/pathology , Staphylococcal Infections/pathology , Staphylococcal Infections/virology , Superinfection/microbiology , Superinfection/pathology , Superinfection/virology
19.
Cancer Cell ; 4(4): 263-75, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14585354

ABSTRACT

Ideally, an oncolytic virus will replicate preferentially in malignant cells, have the ability to treat disseminated metastases, and ultimately be cleared by the patient. Here we present evidence that the attenuated vesicular stomatitis strains, AV1 and AV2, embody all of these traits. We uncover the mechanism by which these mutants are selectively attenuated in interferon-responsive cells while remaining highly lytic in 80% of human tumor cell lines tested. AV1 and AV2 were tested in a xenograft model of human ovarian cancer and in an immune competent mouse model of metastatic colon cancer. While highly attenuated for growth in normal mice, both AV1 and AV2 effected complete and durable cures in the majority of treated animals when delivered systemically.


Subject(s)
Immunity, Innate/physiology , Interferon-beta/metabolism , Vesicular stomatitis Indiana virus/metabolism , Active Transport, Cell Nucleus , Animals , Colonic Neoplasms/therapy , Colonic Neoplasms/virology , Female , Humans , Immunity, Innate/immunology , Interferon-beta/immunology , Lung Neoplasms/therapy , Lung Neoplasms/virology , Mice , Mice, Knockout , Models, Biological , Mutation , Neoplasms, Experimental/virology , Oligonucleotide Array Sequence Analysis , Ovarian Neoplasms/therapy , Ovarian Neoplasms/virology , Signal Transduction , Vesicular stomatitis Indiana virus/genetics , Viral Matrix Proteins/metabolism , Virus Replication/genetics , Virus Replication/physiology
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL