Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
PLoS Biol ; 17(11): e3000558, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31693658

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pbio.1002580.].

2.
J Immunol ; 202(12): 3370-3380, 2019 06 15.
Article in English | MEDLINE | ID: mdl-31092636

ABSTRACT

The importance of antiviral CD8+ T cell recognition of alternative reading frame (ARF)-derived peptides is uncertain. In this study, we describe an epitope (NS1-ARF21-8) present in a predicted 14-residue peptide encoded by the +1 register of NS1 mRNA in the influenza A virus (IAV). NS1-ARF21-8 elicits a robust, highly functional CD8+ T cell response in IAV-infected BALB/c mice. NS1-ARF21-8 is presented from unspliced NS mRNA, likely from downstream initiation on a Met residue that comprises the P1 position of NS1-ARF21-8 Derived from a 14-residue peptide with no apparent biological function and negligible impacts on IAV infection, infectivity, and pathogenicity, NS1-ARF21-8 provides a clear demonstration of how immunosurveillance exploits natural errors in protein translation to provide antiviral immunity. We further show that IAV infection enhances a model cellular ARF translation, which potentially has important implications for virus-induced autoimmunity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/metabolism , Influenza A virus/physiology , Influenza, Human/immunology , Orthomyxoviridae Infections/immunology , Viral Nonstructural Proteins/metabolism , Alternative Splicing , Animals , Disease Models, Animal , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , HEK293 Cells , Host-Pathogen Interactions , Humans , Immunologic Surveillance , Mice , Mice, Inbred BALB C , Open Reading Frames/genetics , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/immunology
3.
PLoS Biol ; 14(11): e1002580, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27855162

ABSTRACT

The extracellular matrix (ECM) provides physical scaffolding for cellular constituents and initiates biochemical and biomechanical cues that are required for physiological activity of living tissues. The ECM enzyme ADAMTS5, a member of the ADAMTS (A Disintegrin-like and Metalloproteinase with Thrombospondin-1 motifs) protein family, cleaves large proteoglycans such as aggrecan, leading to the destruction of cartilage and osteoarthritis. However, its contribution to viral pathogenesis and immunity is currently undefined. Here, we use a combination of in vitro and in vivo models to show that ADAMTS5 enzymatic activity plays a key role in the development of influenza-specific immunity. Influenza virus infection of Adamts5-/- mice resulted in delayed virus clearance, compromised T cell migration and immunity and accumulation of versican, an ADAMTS5 proteoglycan substrate. Our research emphasises the importance of ADAMTS5 expression in the control of influenza virus infection and highlights the potential for development of ADAMTS5-based therapeutic strategies to reduce morbidity and mortality.


Subject(s)
ADAMTS5 Protein/physiology , Immunity, Cellular/physiology , Orthomyxoviridae/immunology , T-Lymphocytes/immunology , ADAMTS5 Protein/genetics , Animals , Immunophenotyping , Mice , Mice, Inbred C57BL , Mice, Knockout , Versicans/metabolism , Weight Loss
4.
Eur J Immunol ; 45(3): 780-93, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25430701

ABSTRACT

Tuberculosis remains a global health problem, in part due to failure of the currently available vaccine, BCG, to protect adults against pulmonary forms of the disease. We explored the impact of pulmonary delivery of recombinant influenza A viruses (rIAVs) on the induction of Mycobacterium tuberculosis (M. tuberculosis)-specific CD4(+) and CD8(+) T-cell responses and the resultant protection against M. tuberculosis infection in C57BL/6 mice. Intranasal infection with rIAVs expressing a CD4(+) T-cell epitope from the Ag85B protein (PR8.p25) or CD8(+) T-cell epitope from the TB10.4 protein (PR8.TB10.4) generated strong T-cell responses to the M. tuberculosis-specific epitopes in the lung that persisted long after the rIAVs were cleared. Infection with PR8.p25 conferred protection against subsequent M. tuberculosis challenge in the lung, and this was associated with increased levels of poly-functional CD4(+) T cells at the time of challenge. By contrast, infection with PR8.TB10.4 did not induce protection despite the presence of IFN-γ-producing M. tuberculosis-specific CD8(+) T cells in the lung at the time of challenge and during infection. Therefore, the induction of pulmonary M. tuberculosis epitope-specific CD4(+), but not CD8(+) T cells, is essential for protection against acute M. tuberculosis infection in the lung.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Epitopes/immunology , Influenza A virus , Mycobacterium tuberculosis/immunology , Tuberculosis Vaccines/immunology , Tuberculosis, Pulmonary/prevention & control , Animals , CD8-Positive T-Lymphocytes/immunology , Epitopes/genetics , Female , Mice , Mycobacterium tuberculosis/genetics , Tuberculosis Vaccines/genetics , Tuberculosis, Pulmonary/immunology
5.
J Virol ; 89(5): 2672-83, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25520513

ABSTRACT

UNLABELLED: Highly pathogenic avian influenza virus infection is associated with severe mortality in both humans and poultry. The mechanisms of disease pathogenesis and immunity are poorly understood although recent evidence suggests that cytokine/chemokine dysregulation contributes to disease severity following H5N1 infection. Influenza A virus infection causes a rapid influx of inflammatory cells, resulting in increased reactive oxygen species production, cytokine expression, and acute lung injury. Proinflammatory stimuli are known to induce intracellular reactive oxygen species by activating NADPH oxidase activity. We therefore hypothesized that inhibition of this activity would restore host cytokine homeostasis following avian influenza virus infection. A panel of airway epithelial and immune cells from mammalian and avian species were infected with A/Puerto Rico/8/1934 H1N1 virus, low-pathogenicity avian influenza H5N3 virus (A/duck/Victoria/0305-2/2012), highly pathogenic avian influenza H5N1 virus (A/chicken/Vietnam/0008/2004), or low-pathogenicity avian influenza H7N9 virus (A/Anhui/1/2013). Quantitative real-time reverse transcriptase PCR showed that H5N1 and H7N9 viruses significantly stimulated cytokine (interleukin-6, beta interferon, CXCL10, and CCL5) production. Among the influenza-induced cytokines, CCL5 was identified as a potential marker for overactive immunity. Apocynin, a Nox2 inhibitor, inhibited influenza-induced cytokines and reactive oxygen species production, although viral replication was not significantly altered in vitro. Interestingly, apocynin treatment significantly increased influenza virus-induced mRNA and protein expression of SOCS1 and SOCS3, enhancing negative regulation of cytokine signaling. These findings suggest that apocynin or its derivatives (targeting host responses) could be used in combination with antiviral strategies (targeting viruses) as therapeutic agents to ameliorate disease severity in susceptible species. IMPORTANCE: Highly pathogenic avian influenza virus infection causes severe morbidity and mortality in both humans and poultry. Wide-spread antiviral resistance necessitates the need for the development of additional novel therapeutic measures to modulate overactive host immune responses after infection. Disease severity following avian influenza virus infection can be attributed in part to hyperinduction of inflammatory mediators such as cytokines, chemokines, and reactive oxygen species. Our study shows that highly pathogenic avian influenza H5N1 virus and low-pathogenicity avian influenza H7N9 virus (both associated with human fatalities) promote inactivation of FoxO3 and downregulation of the TAM receptor tyrosine kinase, Tyro3, leading to augmentation of the inflammatory cytokine response. Inhibition of influenza-induced reactive oxygen species with apocynin activated FoxO3 and stimulated SOCS1 and SOCS3 proteins, restoring cytokine homeostasis. We conclude that modulation of host immune responses with antioxidant and/or anti-inflammatory agents in combination with antiviral therapy may have important therapeutic benefits.


Subject(s)
Influenza A virus/immunology , Reactive Oxygen Species/toxicity , Suppressor of Cytokine Signaling Proteins/metabolism , Acetophenones/metabolism , Animals , Antioxidants/metabolism , Cell Line , Chickens , Cytokines/biosynthesis , Ducks , Gene Expression Profiling , Humans , Reactive Oxygen Species/antagonists & inhibitors , Real-Time Polymerase Chain Reaction , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling 3 Protein
6.
Rev Med Virol ; 25(6): 406-30, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26467906

ABSTRACT

The emergence of H5N1 highly pathogenic avian influenza has caused a heavy socio-economic burden through culling of poultry to minimise human and livestock infection. Although human infections with H5N1 have to date been limited, concerns for the pandemic potential of this zoonotic virus have been greatly intensified following experimental evidence of aerosol transmission of H5N1 viruses in a mammalian infection model. In this review, we discuss the dominance of the haemagglutinin cleavage site motif as a pathogenicity determinant, the host-pathogen molecular interactions driving cleavage activation, reverse genetics manipulations and identification of residues key to haemagglutinin cleavage site functionality and the mechanisms of cell and tissue damage during H5N1 infection. We specifically focus on the disease in chickens, as it is in this species that high pathogenicity frequently evolves and from which transmission to the human population occurs. With >75% of emerging infectious diseases being of zoonotic origin, it is necessary to understand pathogenesis in the primary host to explain spillover events into the human population.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Host-Pathogen Interactions , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza in Birds/pathology , Influenza in Birds/virology , Proteolysis , Virulence Factors/metabolism , Animals , Chickens , Humans , Influenza in Birds/transmission , Zoonoses/pathology , Zoonoses/transmission , Zoonoses/virology
7.
J Immunol ; 191(1): 302-11, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23698750

ABSTRACT

Individuals infected with mycobacteria are likely to experience episodes of concurrent infections with unrelated respiratory pathogens, including the seasonal or pandemic circulating influenza A virus strains. We analyzed the impact of influenza A virus and mycobacterial respiratory coinfection on the development of CD8 T cell responses to each pathogen. Coinfected mice exhibited reduced frequency and numbers of CD8 T cells specific to Mycobacterium bovis bacille Calmette-Guérin (BCG) in the lungs, and the IFN-γ CD8 T cell response to BCG-encoded OVA was decreased in the lungs of coinfected mice, when compared with mice infected with BCG alone. Moreover, after 2 wk of infection, mice coinfected with both pathogens showed a significant increase in the number of mycobacteria present in the lung compared with mice infected with BCG only. Following adoptive transfer into coinfected mice, transgenic CD8 T cells specific for OVA(257-264) failed to proliferate as extensively in the mediastinal lymph nodes as in mice infected only with BCG-OVA. Also noted was a reduction in the proliferation of BCG-specific CD4 transgenic T cells in mice coinfected with influenza compared with mice infected with BCG alone. Furthermore, phenotypic analysis of CD11c(+) dendritic cells from mediastinal lymph nodes of the infected mice showed that coinfection was associated with decreased surface expression of MHC class II and class I. Thus, concurrent pulmonary infection with influenza A virus is associated with decreased MHC expression on dendritic cells, reduced activation of BCG-specific CD4 and CD8 T cells, and impaired clearance of mycobacteria.


Subject(s)
Influenza A virus/immunology , Mycobacterium bovis/immunology , Orthomyxoviridae Infections/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/microbiology , Tuberculosis, Pulmonary/immunology , Animals , Cells, Cultured , Coculture Techniques , Female , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Orthomyxoviridae Infections/complications , Orthomyxoviridae Infections/microbiology , Ovalbumin/immunology , T-Lymphocyte Subsets/pathology , Tuberculosis, Pulmonary/complications , Tuberculosis, Pulmonary/virology
8.
J Gen Virol ; 95(Pt 9): 1880-1885, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24876306

ABSTRACT

Difficulties associated with efficient delivery and targeting of miRNAs to cells is hampering the real world application of miRNA technology. This study utilized an influenza A-based delivery system to express miR-155 in order to knockdown SOCS1 mRNA. Using qPCR and dual luciferase technology we show that miR-155 delivery resulted in a significant increase in cellular miR-155 which facilitated a downregulation of SOCS1 gene expression and a functional increase in IL-6 and IFN-ß cytokines.


Subject(s)
Gene Transfer Techniques , Influenza A virus/genetics , MicroRNAs/genetics , Suppressor of Cytokine Signaling Proteins/genetics , Animals , Cell Line , Chlorocebus aethiops , Dogs , Gene Knockdown Techniques , Genetic Vectors , Interferon-beta/biosynthesis , Interleukin-6/biosynthesis , Madin Darby Canine Kidney Cells , Mice , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins/biosynthesis , Vero Cells
9.
Expert Rev Mol Med ; 16: e2, 2014 Feb 14.
Article in English | MEDLINE | ID: mdl-24528628

ABSTRACT

Reverse genetics systems allow artificial generation of non-segmented and segmented negative-sense RNA viruses, like influenza viruses, entirely from cloned cDNA. Since the introduction of reverse genetics systems over a decade ago, the ability to generate 'designer' influenza viruses in the laboratory has advanced both basic and applied research, providing a powerful tool to investigate and characterise host-pathogen interactions and advance the development of novel therapeutic strategies. The list of applications for reverse genetics has expanded vastly in recent years. In this review, we discuss the development and implications of this technique, including the recent controversy surrounding the generation of a transmissible H5N1 influenza virus. We will focus on research involving the identification of viral protein function, development of live-attenuated influenza virus vaccines, host-pathogen interactions, immunity and the generation of recombinant influenza virus vaccine vectors for the prevention and treatment of infectious diseases and cancer.


Subject(s)
Influenza A virus/genetics , Influenza, Human/virology , Antiviral Agents/pharmacology , Drug Resistance, Viral , Host Specificity , Humans , Immunity, Cellular , Immunity, Innate , Influenza A virus/immunology , Influenza Vaccines/genetics , Influenza Vaccines/immunology , Influenza, Human/immunology , Influenza, Human/prevention & control , Lymphocytes/immunology , Lymphocytes/virology , Reverse Genetics
10.
J Virol ; 87(8): 4146-60, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23345519

ABSTRACT

There is an urgent need for a human immunodeficiency virus (HIV) vaccine that induces robust mucosal immunity. CD8(+) cytotoxic T lymphocytes (CTLs) apply substantial antiviral pressure, but CTLs to individual epitopes select for immune escape variants in both HIV in humans and SIV in macaques. Inducing multiple simian immunodeficiency virus (SIV)-specific CTLs may assist in controlling viremia. We vaccinated 10 Mane-A1*08401(+) female pigtail macaques with recombinant influenza viruses expressing three Mane-A1*08401-restricted SIV-specific CTL epitopes and subsequently challenged the animals, along with five controls, intravaginally with SIV(mac251). Seroconversion to the influenza virus vector resulted and small, but detectable, SIV-specific CTL responses were induced. There was a boost in CTL responses after challenge but no protection from high-level viremia or CD4 depletion was observed. All three CTL epitopes underwent a coordinated pattern of immune escape during early SIV infection. CTL escape was more rapid in the vaccinees than in the controls at the more dominant CTL epitopes. Although CTL escape can incur a "fitness" cost to the virus, a putative compensatory mutation 20 amino acids upstream from an immunodominant Gag CTL epitope also evolved soon after the primary CTL escape mutation. We conclude that vaccines based only on CTL epitopes will likely be undermined by rapid evolution of both CTL escape and compensatory mutations. More potent and possibly broader immune responses may be required to protect pigtail macaques from SIV.


Subject(s)
Immune Evasion , SAIDS Vaccines/administration & dosage , SAIDS Vaccines/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Epitopes/genetics , Epitopes/immunology , Female , Gene Products, gag/genetics , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Macaca nemestrina , Male , Mutation, Missense , Orthomyxoviridae/genetics , Orthomyxoviridae/immunology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/immunology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
11.
J Virol ; 87(7): 3782-91, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23345523

ABSTRACT

Hendra virus is a highly pathogenic zoonotic paramyxovirus in the genus Henipavirus. Thirty-nine outbreaks of Hendra virus have been reported since its initial identification in Queensland, Australia, resulting in seven human infections and four fatalities. Little is known about cellular host factors impacting Hendra virus replication. In this work, we demonstrate that Hendra virus makes use of a microRNA (miRNA) designated miR-146a, an NF-κB-responsive miRNA upregulated by several innate immune ligands, to favor its replication. miR-146a is elevated in the blood of ferrets and horses infected with Hendra virus and is upregulated by Hendra virus in human cells in vitro. Blocking miR-146a reduces Hendra virus replication in vitro, suggesting a role for this miRNA in Hendra virus replication. In silico analysis of miR-146a targets identified ring finger protein (RNF)11, a member of the A20 ubiquitin editing complex that negatively regulates NF-κB activity, as a novel component of Hendra virus replication. RNA interference-mediated silencing of RNF11 promotes Hendra virus replication in vitro, suggesting that increased NF-κB activity aids Hendra virus replication. Furthermore, overexpression of the IκB superrepressor inhibits Hendra virus replication. These studies are the first to demonstrate a host miRNA response to Hendra virus infection and suggest an important role for host miRNAs in Hendra virus disease.


Subject(s)
Hendra Virus/physiology , MicroRNAs/metabolism , Virus Replication/physiology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cloning, Molecular , DNA-Binding Proteins , HeLa Cells , Humans , I-kappa B Proteins/metabolism , NF-kappa B/metabolism , RNA Interference , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
12.
Am J Respir Cell Mol Biol ; 48(1): 17-26, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23002098

ABSTRACT

Oxidative stress caused by excessive reactive oxygen species production is implicated in influenza A virus-induced lung disease. Glutathione peroxidase (GPx)-1 is an antioxidant enzyme that may protect lungs from such damage. The objective of this study was to determine if GPx-1 protects the lung against influenza A virus-induced lung inflammation in vivo. Male wild-type (WT) or GPx-1(-/-) mice were inoculated with HKx31 (H3N2, 1 × 10(4) plaque-forming units), and bronchoalveolar lavage fluid (BALF)/lung compartments were analyzed on Days 3 and 7 after infection for inflammatory marker expression, histology, and viral titer. WT mice infected with HKx31 had significantly more BALF total cells, macrophages, neutrophils, and lymphocytes at Days 3 and 7 compared with naive WT animals (n = 5-8; P < 0.05). However, infected GPx-1(-/-) mice had significantly more BALF inflammation, which included more total cells, macrophages, and neutrophils, compared with WT mice, and this was abolished by treatment with the GPx mimetic ebselen. BALF inflammation persisted in GPx-1(-/-) mice on Day 10 after infection, and GPx-1(-/-) mice had significantly more influenza-specific CD8(+) T cells in spleen compared with WT mice (n = 3-4; P < 0.05). Infected GPx-1(-/-) mice had greater peribronchial and parenchymal inflammation than WT mice, and viral titer was significantly reduced in GPx-1(-/-) mice at Day 3 (n = 5; P < 0.05). Gene expression analysis revealed that infected GPx-1(-/-) mice had higher whole lung TNF-α, macrophage inflammatory protein (MIP)-1α, MIP-2, KC, and matrix metalloproteinase (MMP)-12 mRNA compared with infected WT mice. GPx-1(-/-) mice had more MIP-2 protein in BALF at Day 3 and more active MMP-9 protease in BALF at Days 3 and 7 than WT mice. These data indicate that GPx-1 reduces influenza A virus-induced lung inflammation.


Subject(s)
Glutathione Peroxidase/physiology , Influenza A Virus, H3N2 Subtype , Orthomyxoviridae Infections/enzymology , Pneumonia/enzymology , Pneumonia/prevention & control , Adaptive Immunity , Animals , Azoles/pharmacology , Bronchoalveolar Lavage Fluid/cytology , CD8-Positive T-Lymphocytes/immunology , Chemokines/genetics , Cytokines/genetics , Glutathione Peroxidase/deficiency , Glutathione Peroxidase/genetics , Influenza A Virus, H3N2 Subtype/immunology , Isoindoles , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organoselenium Compounds/pharmacology , Orthomyxoviridae Infections/etiology , Orthomyxoviridae Infections/pathology , Peptide Hydrolases/genetics , Pneumonia/etiology , Pneumonia/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species , Viral Load , Glutathione Peroxidase GPX1
13.
PLoS Pathog ; 7(2): e1001271, 2011 Feb 03.
Article in English | MEDLINE | ID: mdl-21304882

ABSTRACT

Influenza A virus pandemics and emerging anti-viral resistance highlight the urgent need for novel generic pharmacological strategies that reduce both viral replication and lung inflammation. We investigated whether the primary enzymatic source of inflammatory cell ROS (reactive oxygen species), Nox2-containing NADPH oxidase, is a novel pharmacological target against the lung inflammation caused by influenza A viruses. Male WT (C57BL/6) and Nox2(-/y) mice were infected intranasally with low pathogenicity (X-31, H3N2) or higher pathogenicity (PR8, H1N1) influenza A virus. Viral titer, airways inflammation, superoxide and peroxynitrite production, lung histopathology, pro-inflammatory (MCP-1) and antiviral (IL-1ß) cytokines/chemokines, CD8(+) T cell effector function and alveolar epithelial cell apoptosis were assessed. Infection of Nox2(-/y) mice with X-31 virus resulted in a significant reduction in viral titers, BALF macrophages, peri-bronchial inflammation, BALF inflammatory cell superoxide and lung tissue peroxynitrite production, MCP-1 levels and alveolar epithelial cell apoptosis when compared to WT control mice. Lung levels of IL-1ß were ∼3-fold higher in Nox2(-/y) mice. The numbers of influenza-specific CD8+D(b)NP(366)+ and D(b)PA(224)+ T cells in the BALF and spleen were comparable in WT and Nox2(-/y) mice. In vivo administration of the Nox2 inhibitor apocynin significantly suppressed viral titer, airways inflammation and inflammatory cell superoxide production following infection with X-31 or PR8. In conclusion, these findings indicate that Nox2 inhibitors have therapeutic potential for control of lung inflammation and damage in an influenza strain-independent manner.


Subject(s)
Enzyme Inhibitors/therapeutic use , Influenza A virus/physiology , Membrane Glycoproteins/antagonists & inhibitors , NADPH Oxidases/antagonists & inhibitors , Orthomyxoviridae Infections/complications , Pneumonia/etiology , Pneumonia/prevention & control , Animals , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/virology , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Influenza A virus/pathogenicity , Lung/enzymology , Lung/immunology , Lung/metabolism , Lung/pathology , Macrophages/drug effects , Macrophages/pathology , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , NADPH Oxidase 2 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , NADPH Oxidases/physiology , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/pathology , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/genetics , Oxidoreductases/metabolism , Oxidoreductases/physiology , Pneumonia/genetics , Pneumonia/pathology , Reactive Oxygen Species/metabolism
14.
FASEB J ; 26(4): 1372-86, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22247330

ABSTRACT

Influenza virus encodes only 11 viral proteins but replicates in a broad range of avian and mammalian species by exploiting host cell functions. Genome-wide RNA interference (RNAi) has proven to be a powerful tool for identifying the host molecules that participate in each step of virus replication. Meta-analysis of findings from genome-wide RNAi screens has shown influenza virus to be dependent on functional nodes in host cell pathways, requiring a wide variety of molecules and cellular proteins for replication. Because rapid evolution of the influenza A viruses persistently complicates the effectiveness of vaccines and therapeutics, a further understanding of the complex host cell pathways coopted by influenza virus for replication may provide new targets and strategies for antiviral therapy. RNAi genome screening technologies together with bioinformatics can provide the ability to rapidly identify specific host factors involved in resistance and susceptibility to influenza virus, allowing for novel disease intervention strategies.


Subject(s)
High-Throughput Screening Assays/methods , Influenza A virus/genetics , Influenza, Human/therapy , RNA Interference , Viral Proteins/genetics , Animals , Humans , Meta-Analysis as Topic , MicroRNAs/genetics , MicroRNAs/metabolism , NF-kappa B/metabolism , Protein Kinase C/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction/physiology
15.
Biomolecules ; 13(10)2023 10 12.
Article in English | MEDLINE | ID: mdl-37892192

ABSTRACT

The cytokine-inducible SH2 domain-containing (CISH) protein was the first member of the suppressor of cytokine signaling (SOCS) family of negative feedback regulators discovered, being identified in vitro as an inducible inhibitor of erythropoietin (EPO) signaling. However, understanding of the physiological role played by CISH in erythropoiesis has remained limited. To directly assess the function of CISH in this context, mice deficient in CISH were characterized with respect to developmental, steady-state, and EPO-induced erythropoiesis. CISH was strongly expressed in the fetal liver, but CISH knockout (KO) mice showed only minor disruption of primitive erythropoiesis. However, adults exhibited mild macrocytic anemia coincident with subtle perturbation particularly of bone marrow erythropoiesis, with EPO-induced erythropoiesis blunted in the bone marrow of KO mice but enhanced in the spleen. Cish was expressed basally in the bone marrow with induction following EPO stimulation in bone marrow and spleen. Overall, this study indicates that CISH participates in the control of both basal and EPO-induced erythropoiesis in vivo.


Subject(s)
Erythropoiesis , Suppressor of Cytokine Signaling Proteins , Animals , Mice , Anemia/genetics , Cytokines , Erythropoiesis/physiology , Signal Transduction/physiology , src Homology Domains , Suppressor of Cytokine Signaling Proteins/metabolism
16.
Eur J Immunol ; 40(3): 849-58, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19950188

ABSTRACT

TCR repertoire diversity is important for the protective efficacy of CD8(+) T cells, limiting viral escape and cross-reactivity between unrelated epitopes. The exact mechanism for selection of restricted versus diverse TCR repertoires is far from clear, although one thought is that the epitopes resembling self-peptides might select a limited array of TCR due to the deletion of autoreactive TCR. The molecule Aire promotes the expression of tissue-specific Ag on thymic medullary epithelial cells and the deletion of autoreactive cells, and in the absence of Aire autoreactive cells persist. However, the contribution of Aire-dependent peptides to the selection of the Ag-specific TCR repertoire remains unknown. In this study, we dissect restricted (D(b)NP(366)%(+)CD8(+)) and diverse (D(b)PA(224)%(+)CD8(+), K(d)NP(147)%(+)CD8(+)) TCR repertoires responding to three influenza-derived peptides in Aire-deficient mice on both B6 and BALB/c backgrounds. Our study shows that the number, qualitative characteristics and TCR repertoires of all influenza-specific, D(b)NP(366)%(+)CD8(+), D(b)PA(224)%(+)CD8(+) and K(d)NP(147)%(+)CD8(+) T cells are not significantly altered in the absence of Aire. This provides the first demonstration that the selection of an Ag-specific T-cell repertoire is not significantly perturbed in the absence of Aire.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Orthomyxoviridae Infections/immunology , Receptors, Antigen, T-Cell/immunology , Transcription Factors/immunology , Animals , Epitopes, T-Lymphocyte/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae/immunology , Receptors, Antigen, T-Cell/genetics , Reverse Transcriptase Polymerase Chain Reaction , Self Tolerance/immunology , Transcription Factors/genetics , AIRE Protein
17.
Front Mol Biosci ; 8: 703456, 2021.
Article in English | MEDLINE | ID: mdl-34291090

ABSTRACT

Each year, hundreds of thousands of individuals succumb to influenza virus infection and its associated complications. Several preventative and therapeutic options may be applied in order to preserve life. These traditional approaches include administration of seasonal influenza vaccines, pharmacological interventions in the form of antiviral drug therapy and supportive clinical approaches including mechanical ventilation and extracorporeal membrane oxygenation. While these measures have shown varying degrees of success, antiviral therapies and vaccination are constrained due to ongoing antigenic drift. Moreover, clinical approaches can also be associated with complications and drawbacks. These factors have led to the exploration and development of more sophisticated and nuanced therapeutic approaches involving host proteins. Advances in immunotherapy in the cancer field or administration of steroids following virus infection have highlighted the therapeutic potential of targeting host immune responses. We have now reached a point where we can consider the contribution of other "non-traditional" host components such as the extracellular matrix in immunity. Herein, we will review current, established therapeutic interventions and consider novel therapeutic approaches involving the extracellular matrix.

18.
Viruses ; 13(4)2021 04 06.
Article in English | MEDLINE | ID: mdl-33917411

ABSTRACT

Vaccines and therapeutics targeting viral surface glycoproteins are a major component of disease prevention for respiratory viral diseases. Over the years, vaccines have proven to be the most successful intervention for preventing disease. Technological advances in vaccine platforms that focus on viral surface glycoproteins have provided solutions for current and emerging pathogens like SARS-CoV-2, and our understanding of the structural basis for antibody neutralization is guiding the selection of other vaccine targets for respiratory viruses like RSV. This review discusses the role of viral surface glycoproteins in disease intervention approaches.


Subject(s)
Antibodies, Viral/immunology , COVID-19/immunology , Membrane Glycoproteins/immunology , SARS-CoV-2/immunology , Animals , Antibodies, Neutralizing , COVID-19 Vaccines , Humans , Membrane Glycoproteins/drug effects , Respiratory Syncytial Viruses , Seasons , Vaccines, Attenuated/immunology , Viral Vaccines
19.
Front Mol Biosci ; 8: 703868, 2021.
Article in English | MEDLINE | ID: mdl-34527702

ABSTRACT

Remodelling of the extracellular matrix (ECM) by ECM metalloproteinases is increasingly being associated with regulation of immune cell function. ECM metalloproteinases, including Matrix Metalloproteinases (MMPs), A Disintegrin and Metalloproteinases (ADAMs) and ADAMs with Thombospondin-1 motifs (ADAMTS) play a vital role in pathogen defence and have been shown to influence migration of immune cells. This review provides a current summary of the role of ECM enzymes in immune cell migration and function and discusses opportunities and limitations for development of diagnostic and therapeutic strategies targeting metalloproteinase expression and activity in the context of infectious disease.

20.
PLoS One ; 16(11): e0259829, 2021.
Article in English | MEDLINE | ID: mdl-34793507

ABSTRACT

The current Mycobacterium bovis BCG vaccine provides inconsistent protection against pulmonary infection with Mycobacterium tuberculosis. Immunity induced by subcutaneous immunization with BCG wanes and does not promote early recruitment of T cell to the lungs after M. tuberculosis infection. Delivery of Tuberculosis (TB) vaccines to the lungs may increase and prolong immunity at the primary site of M. tuberculosis infection. Pulmonary immunization with recombinant influenza A viruses (rIAVs) expressing an immune-dominant M. tuberculosis CD4+ T cell epitope (PR8-p25 and X31-p25) stimulates protective immunity against lung TB infection. Here, we investigated the potential use of rIAVs to improve the efficacy of BCG using simultaneous immunization (SIM) and prime-boost strategies. SIM with parenteral BCG and intranasal PR8-p25 resulted in equivalent protection to BCG alone against early, acute and chronic M. tuberculosis infection. Boosting BCG with rIAVs increased the frequency of IFN-γ-secreting specific T cells (p<0.001) and polyfunctional CD4+ T cells (p<0.05) in the lungs compared to the BCG alone, however, this did not result in a significant increase in protection against M. tuberculosis compared to BCG alone. Therefore, sequential pulmonary immunization with these rIAVs after BCG increased M. tuberculosis-specific memory T cell responses in the lung, but not protection against M. tuberculosis infection.


Subject(s)
BCG Vaccine/immunology , Influenza A virus/immunology , Mycobacterium tuberculosis/immunology , T-Lymphocytes/immunology , Tuberculosis, Pulmonary/prevention & control , Animals , Epitopes , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Female , Immunization, Secondary , Immunogenicity, Vaccine , Lung/immunology , Memory T Cells/immunology , Mice , Mice, Inbred C57BL , Tuberculosis, Pulmonary/immunology , Vaccines, Synthetic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL