Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Elife ; 42015 Apr 21.
Article in English | MEDLINE | ID: mdl-25898005

ABSTRACT

Effective mucosal adjuvants enhance the magnitude and quality of the vaccine response. Cyclic di-GMP (CDG) is a promising mucosal vaccine adjuvant. However, its in vivo mechanisms are unclear. Here, we showed, in mice, that CDG elicits stronger Ab and TH responses than the mammalian 2'3'-cyclic GMP-AMP (cGAMP), and generated better protection against Streptococcus pneumoniae infection than 2'3'-cGAMP adjuvanted vaccine. We identified two in vivo mechanisms of CDG. First, intranasally administered CDG greatly enhances Ag uptake, including pinocytosis and receptor-mediated endocytosis in vivo. The enhancement depends on MPYS (STING, MITA) expression in CD11C(+) cells. Second, we found that CDG selectively activated pinocytosis-efficient-DCs, leading to T(H) polarizing cytokines IL-12p70, IFNγ, IL-5, IL-13, IL-23, and IL-6 production in vivo. Notably, CDG induces IFNλ, but not IFNß, in vivo. Our study revealed previously unrecognized in vivo functions of MPYS and advanced our understanding of CDG as a mucosal vaccine adjuvant.


Subject(s)
Adjuvants, Immunologic/metabolism , Antigens/metabolism , Cytokines/metabolism , Nucleotides, Cyclic/immunology , Animals , Endocytosis/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucous Membrane/immunology , Pinocytosis/immunology
2.
Mol Microbiol ; 93(1): 65-79, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24806618

ABSTRACT

Tuberculosis (TB) remains a major cause of morbidity and mortality worldwide. The pathogenesis by the causative agent, Mycobacterium tuberculosis, is still not fully understood. We have previously reported that M. tuberculosis Rv3586 (disA) encodes a diadenylate cyclase, which converts ATP to cyclic di-AMP (c-di-AMP). In this study, we demonstrated that a protein encoded by Rv2837c (cnpB) possesses c-di-AMP phosphodiesterase activity and cleaves c-di-AMP exclusively to AMP. Our results showed that in M. tuberculosis, deletion of disA abolished bacterial c-di-AMP production, whereas deletion of cnpB significantly enhanced the bacterial c-di-AMP accumulation and secretion. The c-di-AMP levels in both mutants could be corrected by expressing the respective gene. We also found that macrophages infected with ΔcnpB secreted much higher levels of IFN-ß than those infected with the wild type (WT) or the complemented mutant. Interestingly, mice infected with M. tuberculosis ΔcnpB displayed significantly reduced inflammation, less bacterial burden in the lungs and spleens, and extended survival compared with those infected with the WT or the complemented mutant. These results indicate that deletion of cnpB results in attenuated virulence, which is correlated with elevated c-di-AMP levels.


Subject(s)
Bacterial Proteins/genetics , Dinucleoside Phosphates/metabolism , Mycobacterium tuberculosis/pathogenicity , Phosphoric Diester Hydrolases/genetics , Tuberculosis/microbiology , Animals , Bacterial Proteins/metabolism , Disease Models, Animal , Female , Lung/microbiology , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Inbred BALB C , Mycobacterium tuberculosis/enzymology , Phosphoric Diester Hydrolases/metabolism , Spleen/microbiology , Tuberculosis/pathology , Virulence
3.
J Immunol ; 192(1): 492-502, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24307739

ABSTRACT

The bacterial second messenger (3'-5')-cyclic-di-guanosine-monophosphate (CDG) is a promising mucosal adjuvant candidate that activates balanced Th1/Th2/Th17 responses. We showed previously that CDG activates stimulator of IFN genes (STING)-dependent IFN-I production in vitro. However, it is unknown whether STING or IFN-I is required for the CDG adjuvant activity in vivo. In this study, we show that STING(-/-) mice (Tmem173()) do not produce Ag-specific Abs or Th1/Th2/Th17 cytokines during CDG/Ag immunization. Intranasal administration of CDG did not induce TNF-α, IL-1ß, IL-6, IL-12, or MCP-1 production in STING(-/-) mice. Surprisingly, we found that the cytokine and Ab responses were unaltered in CDG/Ag-immunized IFNAR(-/-) mice. Instead, we found that CDG activates STING-dependent, IFN-I-independent TNF-α production in vivo and in vitro. Furthermore, using a TNFR1(-/-) mouse, we demonstrate that TNF-α signaling is critical for CDG-induced Ag-specific Ab and Th1/Th2 cytokine production. This is distinct from STING-mediated DNA adjuvant activity, which requires IFN-I, but not TNF-α, production. Finally, we found that CDG activates STING-dependent, but IRF3 stimulation-independent, NF-κB signaling. Our results established an essential role for STING-mediated TNF-α production in the mucosal adjuvant activity of CDG in vivo and revealed a novel IFN-I stimulation-independent STING-NF-κB-TNF-α pathway.


Subject(s)
Cyclic GMP/analogs & derivatives , Membrane Proteins/metabolism , Mucous Membrane/immunology , Mucous Membrane/metabolism , Tumor Necrosis Factor-alpha/metabolism , Adjuvants, Immunologic , Animals , Chemokines/biosynthesis , Cyclic GMP/immunology , Cytokines/biosynthesis , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Immunoglobulin G/immunology , Interferon Regulatory Factor-3/metabolism , Interferon Type I/metabolism , Mice , NF-kappa B/metabolism , Ovalbumin/immunology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...