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.
Nature ; 478(7370): 515-8, 2011 Sep 25.
Article in English | MEDLINE | ID: mdl-21947006

ABSTRACT

The innate immune system detects infection by using germline-encoded receptors that are specific for conserved microbial molecules. The recognition of microbial ligands leads to the production of cytokines, such as type I interferons (IFNs), that are essential for successful pathogen elimination. Cytosolic detection of pathogen-derived DNA is one major mechanism of inducing IFN production, and this process requires signalling through TANK binding kinase 1 (TBK1) and its downstream transcription factor, IFN-regulatory factor 3 (IRF3). In addition, a transmembrane protein called STING (stimulator of IFN genes; also known as MITA, ERIS, MPYS and TMEM173) functions as an essential signalling adaptor, linking the cytosolic detection of DNA to the TBK1-IRF3 signalling axis. Recently, unique nucleic acids called cyclic dinucleotides, which function as conserved signalling molecules in bacteria, have also been shown to induce a STING-dependent type I IFN response. However, a mammalian sensor of cyclic dinucleotides has not been identified. Here we report evidence that STING itself is an innate immune sensor of cyclic dinucleotides. We demonstrate that STING binds directly to radiolabelled cyclic diguanylate monophosphate (c-di-GMP), and we show that unlabelled cyclic dinucleotides, but not other nucleotides or nucleic acids, compete with c-di-GMP for binding to STING. Furthermore, we identify mutations in STING that selectively affect the response to cyclic dinucleotides without affecting the response to DNA. Thus, STING seems to function as a direct sensor of cyclic dinucleotides, in addition to its established role as a signalling adaptor in the IFN response to cytosolic DNA. Cyclic dinucleotides have shown promise as novel vaccine adjuvants and immunotherapeutics, and our results provide insight into the mechanism by which cyclic dinucleotides are sensed by the innate immune system.


Subject(s)
Cyclic GMP/analogs & derivatives , Immunity, Innate/immunology , Membrane Proteins/immunology , Membrane Proteins/metabolism , Adjuvants, Immunologic , Amino Acid Sequence , Animals , Cyclic GMP/immunology , DNA/immunology , HEK293 Cells , Humans , Interferons/immunology , Macrophages/immunology , Macrophages/metabolism , Membrane Proteins/genetics , Mice , Molecular Sequence Data
2.
Infect Immun ; 79(2): 688-94, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21098106

ABSTRACT

Type I interferons (IFNs) are central regulators of the innate and adaptive immune responses to viral and bacterial infections. Type I IFNs are induced upon cytosolic detection of microbial nucleic acids, including DNA, RNA, and the bacterial second messenger cyclic-di-GMP (c-di-GMP). In addition, a recent study demonstrated that the intracellular bacterial pathogen Listeria monocytogenes stimulates a type I IFN response due to cytosolic detection of bacterially secreted c-di-AMP. The transmembrane signaling adaptor Sting (Tmem173, Mita, Mpys, Eris) has recently been implicated in the induction of type I IFNs in response to cytosolic DNA and/or RNA. However, the role of Sting in response to purified cyclic dinucleotides or during in vivo L. monocytogenes infection has not been addressed. In order to identify genes important in the innate immune response, we have been conducting a forward genetic mutagenesis screen in C57BL/6 mice using the mutagen N-ethyl-N-nitrosourea (ENU). Here we describe a novel mutant mouse strain, Goldenticket (Gt), that fails to produce type I IFNs upon L. monocytogenes infection. By genetic mapping and complementation experiments, we found that Gt mice harbor a single nucleotide variant (T596A) of Sting that functions as a null allele and fails to produce detectable protein. Analysis of macrophages isolated from Gt mice revealed that Sting is absolutely required for the type I interferon response to both c-di-GMP and c-di-AMP. Additionally, Sting is required for the response to c-di-GMP and L. monocytogenes in vivo. Our results provide new functions for Sting in the innate interferon response to pathogens.


Subject(s)
Cyclic GMP/analogs & derivatives , Dinucleoside Phosphates/metabolism , Interferon Type I/metabolism , Listeria monocytogenes/immunology , Listeriosis/immunology , Membrane Proteins/physiology , Alleles , Animals , Cell Line , Cyclic GMP/metabolism , Ethylnitrosourea/toxicity , Female , Gene Expression Regulation/physiology , Genetic Complementation Test , Humans , Interferon Type I/genetics , Listeriosis/metabolism , Macrophages/microbiology , Male , Membrane Proteins/genetics , Mice , Mice, Mutant Strains , Mutation , Polymorphism, Single Nucleotide
SELECTION OF CITATIONS
SEARCH DETAIL
...