Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
J Exp Med ; 213(11): 2365-2382, 2016 10 17.
Article in English | MEDLINE | ID: mdl-27697835

ABSTRACT

Cell death and release of proinflammatory mediators contribute to mortality during sepsis. Specifically, caspase-11-dependent cell death contributes to pathology and decreases in survival time in sepsis models. Priming of the host cell, through TLR4 and interferon receptors, induces caspase-11 expression, and cytosolic LPS directly stimulates caspase-11 activation, promoting the release of proinflammatory cytokines through pyroptosis and caspase-1 activation. Using a CRISPR-Cas9-mediated genome-wide screen, we identified novel mediators of caspase-11-dependent cell death. We found a complement-related peptidase, carboxypeptidase B1 (Cpb1), to be required for caspase-11 gene expression and subsequent caspase-11-dependent cell death. Cpb1 modifies a cleavage product of C3, which binds to and activates C3aR, and then modulates innate immune signaling. We find the Cpb1-C3-C3aR pathway induces caspase-11 expression through amplification of MAPK activity downstream of TLR4 and Ifnar activation, and mediates severity of LPS-induced sepsis (endotoxemia) and disease outcome in mice. We show C3aR is required for up-regulation of caspase-11 orthologues, caspase-4 and -5, in primary human macrophages during inflammation and that c3aR1 and caspase-5 transcripts are highly expressed in patients with severe sepsis; thus, suggesting that these pathways are important in human sepsis. Our results highlight a novel role for complement and the Cpb1-C3-C3aR pathway in proinflammatory signaling, caspase-11 cell death, and sepsis severity.


Subject(s)
Caspases/metabolism , Complement System Proteins/metabolism , Sepsis/enzymology , Sepsis/pathology , Animals , CRISPR-Cas Systems , Carboxypeptidase B/metabolism , Caspases, Initiator , Cell Death , Complement C3/metabolism , Endotoxemia/blood , Endotoxemia/pathology , Endotoxins , Enzyme Activation , Gene Expression Regulation , Humans , Inflammation/genetics , Inflammation/pathology , Inflammation Mediators/metabolism , MAP Kinase Signaling System , Macrophages/enzymology , Macrophages/metabolism , Macrophages/pathology , Mice , Models, Biological , Phosphorylation , RAW 264.7 Cells , Receptors, Complement/metabolism , Receptors, Interferon/metabolism , Salmonella/physiology , Shigella/physiology , Toll-Like Receptor 4/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
2.
J Immunol ; 194(7): 3236-45, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25710914

ABSTRACT

Type I IFN production is an important host immune response against viral and bacterial infections. However, little is known about the ligands and corresponding host receptors that trigger type I IFN production during bacterial infections. We used a model intracellular pathogen, Francisella novicida, to begin characterizing the type I IFN response to bacterial pathogens. F. novicida replicates in the cytosol of host cells and elicits a robust type I IFN response that is largely TLR independent, but is dependent on the adapter molecule STING, suggesting that the type I IFN stimulus during F. novicida infection is cytosolic. In this study, we report that the cytosolic DNA sensors, cyclic GMP-AMP synthase (cGAS) and Ifi204, are both required for the STING-dependent type I IFN response to F. novicida infection in both primary and immortalized murine macrophages. We created cGAS, Ifi204, and Sting functional knockouts in RAW264.7 macrophages and demonstrated that cGAS and Ifi204 cooperate to sense dsDNA and activate the STING-dependent type I IFN pathway. In addition, we show that dsDNA from F. novicida is an important type I IFN stimulating ligand. One outcome of cGAS-STING signaling is the activation of the absent in melanoma 2 inflammasome in response to F. novicida infection. Whereas the absent in melanoma 2 inflammasome is beneficial to the host during F. novicida infection, type I IFN signaling by STING and IFN regulatory factor 3 is detrimental to the host during F. novicida infection. Collectively, our studies indicate that cGAS and Ifi204 cooperate to sense cytosolic dsDNA and F. novicida infection to produce a strong type I IFN response.


Subject(s)
Francisella/immunology , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/metabolism , Interferon Type I/metabolism , Nuclear Proteins/metabolism , Nucleotidyltransferases/metabolism , Phosphoproteins/metabolism , Animals , Bone Marrow Cells/metabolism , Cell Line , Cytosol/immunology , Cytosol/metabolism , DNA/immunology , Disease Models, Animal , Gene Expression , Gene Expression Regulation , Gram-Negative Bacterial Infections/genetics , Gram-Negative Bacterial Infections/mortality , Inflammasomes/metabolism , Interferon Regulatory Factor-3/deficiency , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Macrophages/immunology , Macrophages/metabolism , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Nuclear Proteins/genetics , Nucleotidyltransferases/genetics , Phosphoproteins/genetics , Protein Binding , RNA Interference , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...