Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 117(27): 15789-15798, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32581129

ABSTRACT

Patients infected with influenza are at high risk of secondary bacterial infection, which is a major proximate cause of morbidity and mortality. We have shown that in mice, prior infection with influenza results in increased inflammation and mortality upon Staphylococcus aureus infection, recapitulating the human disease. Lipidomic profiling of the lungs of superinfected mice revealed an increase in CYP450 metabolites during lethal superinfection. These lipids are endogenous ligands for the nuclear receptor PPARα, and we demonstrate that Ppara-/- mice are less susceptible to superinfection than wild-type mice. PPARα is an inhibitor of NFκB activation, and transcriptional profiling of cells isolated by bronchoalveolar lavage confirmed that influenza infection inhibits NFκB, thereby dampening proinflammatory and prosurvival signals. Furthermore, network analysis indicated an increase in necrotic cell death in the lungs of superinfected mice compared to mice infected with S. aureus alone. Consistent with this, we observed reduced NFκB-mediated inflammation and cell survival signaling in cells isolated from the lungs of superinfected mice. The kinase RIPK3 is required to induce necrotic cell death and is strongly induced in cells isolated from the lungs of superinfected mice compared to mice infected with S. aureus alone. Genetic and pharmacological perturbations demonstrated that PPARα mediates RIPK3-dependent necroptosis and that this pathway plays a central role in mortality following superinfection. Thus, we have identified a molecular circuit in which infection with influenza induces CYP450 metabolites that activate PPARα, leading to increased necrotic cell death in the lung which correlates with the excess mortality observed in superinfection.


Subject(s)
Inflammation/genetics , Influenza, Human/genetics , PPAR alpha/genetics , Staphylococcal Infections/genetics , Superinfection/genetics , Animals , Bronchoalveolar Lavage/methods , Coinfection/genetics , Coinfection/microbiology , Coinfection/mortality , Cytochrome P-450 Enzyme System/genetics , Disease Models, Animal , Disease Susceptibility , Humans , Inflammation/microbiology , Inflammation/mortality , Influenza, Human/microbiology , Influenza, Human/mortality , Lung/microbiology , Lung/pathology , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Mice , Mice, Knockout , Necroptosis/genetics , Staphylococcal Infections/microbiology , Staphylococcal Infections/mortality , Superinfection/mortality
2.
Cell ; 154(1): 213-27, 2013 Jul 03.
Article in English | MEDLINE | ID: mdl-23827684

ABSTRACT

Bioactive lipid mediators play a crucial role in the induction and resolution of inflammation. To elucidate their involvement during influenza infection, liquid chromatography/mass spectrometry lipidomic profiling of 141 lipid species was performed on a mouse influenza model using two viruses of significantly different pathogenicity. Infection by the low-pathogenicity strain X31/H3N2 induced a proinflammatory response followed by a distinct anti-inflammatory response; infection by the high-pathogenicity strain PR8/H1N1 resulted in overlapping pro- and anti-inflammatory states. Integration of the large-scale lipid measurements with targeted gene expression data demonstrated that 5-lipoxygenase metabolites correlated with the pathogenic phase of the infection, whereas 12/15-lipoxygenase metabolites were associated with the resolution phase. Hydroxylated linoleic acid, specifically the ratio of 13- to 9-hydroxyoctadecadienoic acid, was identified as a potential biomarker for immune status during an active infection. Importantly, some of the findings from the animal model were recapitulated in studies of human nasopharyngeal lavages obtained during the 2009-2011 influenza seasons.


Subject(s)
Eicosanoids/isolation & purification , Fatty Acids, Unsaturated/isolation & purification , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H3N2 Subtype/physiology , Influenza, Human/immunology , Lipids/analysis , Orthomyxoviridae Infections/immunology , Animals , Arachidonate 5-Lipoxygenase/metabolism , Cytokines/immunology , Disease Models, Animal , Eicosanoids/immunology , Fatty Acids, Unsaturated/immunology , Humans , Inflammation Mediators/analysis , Metabolic Networks and Pathways , Mice , Nasal Lavage Fluid/immunology , Transcriptome
3.
J Immunol ; 189(1): 23-7, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22661094

ABSTRACT

Cellular fusion of macrophages into multinucleated giant cells is a distinguishing feature of the granulomatous response to inflammation, infection, and foreign bodies (Kawai and Akira. 2011. Immunity 34: 637-650). We observed a marked increase in fusion of macrophages genetically deficient in Dicer, an enzyme required for canonical microRNA (miRNA) biogenesis. Gene expression profiling of miRNA-deficient macrophages revealed an upregulation of the IL-4-responsive fusion protein Tm7sf4, and analyses identified miR-7a-1 as a negative regulator of macrophage fusion, functioning by directly targeting Tm7sf4 mRNA. miR-7a-1 is itself an IL-4-responsive gene in macrophages, suggesting feedback control of cellular fusion. Collectively, these data indicate that miR-7a-1 functions to regulate IL-4-directed multinucleated giant cell formation.


Subject(s)
Cell Differentiation/immunology , Giant Cells, Langhans/immunology , Macrophages/cytology , Macrophages/immunology , MicroRNAs/physiology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Differentiation/genetics , Cell Fusion/methods , Cells, Cultured , DEAD-box RNA Helicases/deficiency , DEAD-box RNA Helicases/genetics , Giant Cells, Langhans/cytology , Giant Cells, Langhans/metabolism , HEK293 Cells , Humans , Interleukin-4/physiology , Macrophages/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , MicroRNAs/genetics , Ribonuclease III/deficiency , Ribonuclease III/genetics , Transcription, Genetic/immunology
4.
Proc Natl Acad Sci U S A ; 108(28): 11536-41, 2011 Jul 12.
Article in English | MEDLINE | ID: mdl-21709223

ABSTRACT

Precise control of the innate immune response is essential to ensure host defense against infection while avoiding inflammatory disease. Systems-level analyses of Toll-like receptor (TLR)-stimulated macrophages suggested that SHANK-associated RH domain-interacting protein (SHARPIN) might play a role in the TLR pathway. This hypothesis was supported by the observation that macrophages derived from chronic proliferative dermatitis mutation (cpdm) mice, which harbor a spontaneous null mutation in the Sharpin gene, exhibited impaired IL-12 production in response to TLR activation. Systems biology approaches were used to define the SHARPIN-regulated networks. Promoter analysis identified NF-κB and AP-1 as candidate transcription factors downstream of SHARPIN, and network analysis suggested selective attenuation of these pathways. We found that the effects of SHARPIN deficiency on the TLR2-induced transcriptome were strikingly correlated with the effects of the recently described hypomorphic L153P/panr2 point mutation in Ikbkg [NF-κB Essential Modulator (NEMO)], suggesting that SHARPIN and NEMO interact. We confirmed this interaction by co-immunoprecipitation analysis and furthermore found it to be abrogated by panr2. NEMO-dependent signaling was affected by SHARPIN deficiency in a manner similar to the panr2 mutation, including impaired p105 and ERK phosphorylation and p65 nuclear localization. Interestingly, SHARPIN deficiency had no effect on IκBα degradation and on p38 and JNK phosphorylation. Taken together, these results demonstrate that SHARPIN is an essential adaptor downstream of the branch point defined by the panr2 mutation in NEMO.


Subject(s)
Carrier Proteins/immunology , Carrier Proteins/metabolism , Toll-Like Receptor 2/immunology , Toll-Like Receptor 2/metabolism , Animals , Base Sequence , Carrier Proteins/genetics , DNA Primers/genetics , Immunity, Innate/genetics , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , NF-kappa B/metabolism , Protein Interaction Mapping , Signal Transduction , Systems Analysis , Systems Biology , Toll-Like Receptor 2/genetics , Transcription Factor AP-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...