Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Gut Microbes ; 5(1): 28-39, 2014.
Article in English | MEDLINE | ID: mdl-24637807

ABSTRACT

The intestinal microbiota changes dynamically from birth to adulthood. In this study we identified γ-Proteobacteria as a dominant phylum present in newborn mice that is suppressed in normal adult microbiota. The transition from a neonatal to a mature microbiota was in part regulated by induction of a γ-Proteobacteria-specific IgA response. Neocolonization experiments in germ-free mice further revealed a dominant Proteobacteria-specific IgA response triggered by the immature microbiota. Finally, a role for B cells in the regulation of microbiota maturation was confirmed in IgA-deficient mice. Mice lacking IgA had persistent intestinal colonization with γ-Proteobacteria that resulted in sustained intestinal inflammation and increased susceptibility to neonatal and adult models of intestinal injury. Collectively, these results identify an IgA-dependent mechanism responsible for the maturation of the intestinal microbiota.


Subject(s)
Antibodies, Bacterial/immunology , Colitis/immunology , Immunoglobulin A/immunology , Intestines/growth & development , Intestines/immunology , Microbiota , Proteobacteria/immunology , Animals , Colitis/genetics , Colitis/microbiology , Female , Humans , Intestines/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Proteobacteria/classification , Proteobacteria/genetics , Proteobacteria/isolation & purification
2.
Proc Natl Acad Sci U S A ; 110(26): 10711-6, 2013 Jun 25.
Article in English | MEDLINE | ID: mdl-23754402

ABSTRACT

IFN-γ is a major cytokine that is critical for host resistance to a broad range of intracellular pathogens. Production of IFN-γ by natural killer and T cells is initiated by the recognition of pathogens by Toll-like receptors (TLRs). In an experimental model of toxoplasmosis, we have identified the presence of a nonlymphoid source of IFN-γ that was particularly evident in the absence of TLR-mediated recognition of Toxoplasma gondii. Genetically altered mice lacking all lymphoid cells due to deficiencies in Recombination Activating Gene 2 and IL-2Rγc genes also produced IFN-γ in response to the protozoan parasite. Flow-cytometry and morphological examinations of non-NK/non-T IFN-γ(+) cells identified neutrophils as the cell type capable of producing IFN-γ. Selective elimination of neutrophils in TLR11(-/-) mice infected with the parasite resulted in acute susceptibility similar to that observed in IFN-γ-deficient mice. Similarly, Salmonella typhimurium infection of TLR-deficient mice induces the appearance of IFN-γ(+) neutrophils. Thus, neutrophils are a crucial source for IFN-γ that is required for TLR-independent host protection against intracellular pathogens.


Subject(s)
Host-Pathogen Interactions/immunology , Interferon-gamma/physiology , Neutrophils/immunology , Neutrophils/metabolism , Toll-Like Receptors/immunology , Animals , Host-Parasite Interactions/immunology , Immunity, Innate , Interferon-gamma/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Salmonella typhimurium/immunology , Salmonella typhimurium/pathogenicity , T-Lymphocytes/immunology , Toll-Like Receptors/deficiency , Toll-Like Receptors/genetics , Toxoplasma/immunology , Toxoplasma/pathogenicity
3.
Nat Immunol ; 14(2): 136-42, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23263554

ABSTRACT

Activation of Toll-like receptors (TLRs) by pathogens triggers cytokine production and T cell activation, immune defense mechanisms that are linked to immunopathology. Here we show that IFN-γ production by CD4(+) T(H)1 cells during mucosal responses to the protozoan parasite Toxoplasma gondii resulted in dysbiosis and the elimination of Paneth cells. Paneth cell death led to loss of antimicrobial peptides and occurred in conjunction with uncontrolled expansion of the Enterobacteriaceae family of Gram-negative bacteria. The expanded intestinal bacteria were required for the parasite-induced intestinal pathology. The investigation of cell type-specific factors regulating T(H)1 polarization during T. gondii infection identified the T cell-intrinsic TLR pathway as a major regulator of IFN-γ production in CD4(+) T cells responsible for Paneth cell death, dysbiosis and intestinal immunopathology.


Subject(s)
Enterobacteriaceae Infections/pathology , Enterobacteriaceae/growth & development , Paneth Cells/pathology , Signal Transduction/immunology , Th1 Cells/pathology , Toxoplasma/growth & development , Toxoplasmosis, Animal/pathology , Animals , CD4-Positive T-Lymphocytes , Cell Death , Enterobacteriaceae/immunology , Enterobacteriaceae Infections/complications , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/microbiology , Gene Expression Regulation , Host-Parasite Interactions , Host-Pathogen Interactions , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-12/genetics , Interleukin-12/immunology , Lymphocyte Activation , Mice , Mice, Transgenic , Paneth Cells/microbiology , Paneth Cells/parasitology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Th1 Cells/microbiology , Th1 Cells/parasitology , Toxoplasma/immunology , Toxoplasmosis, Animal/complications , Toxoplasmosis, Animal/immunology , Toxoplasmosis, Animal/parasitology , alpha-Defensins/deficiency
4.
Immunity ; 36(2): 228-38, 2012 Feb 24.
Article in English | MEDLINE | ID: mdl-22306056

ABSTRACT

The Toll-like receptor adaptor protein MyD88 is essential for the regulation of intestinal homeostasis in mammals. In this study, we determined that Myd88-deficient mice are susceptible to colonic damage that is induced by dextran sulfate sodium (DSS) administration resulting from uncontrolled dissemination of intestinal commensal bacteria. The DSS-induced mortality of Myd88-deficient mice was completely prevented by antibiotic treatment to deplete commensal bacteria. By using cell type-specific Myd88-deficient mice, we established that B cell-intrinsic MyD88 signaling plays a central role in the resistance to DSS-induced colonic damage via the production of IgM and complement-mediated control of intestinal bacteria. Our results indicate that the lack of intact MyD88 signaling in B cells, coupled with impaired epithelial integrity, enables commensal bacteria to function as highly pathogenic organisms, causing rapid host death.


Subject(s)
B-Lymphocytes/immunology , Colon/immunology , Colon/microbiology , Myeloid Differentiation Factor 88/metabolism , Animals , Anti-Bacterial Agents/pharmacology , B-Lymphocytes/metabolism , Colon/drug effects , Colon/injuries , Complement System Proteins/metabolism , Dextran Sulfate/toxicity , Host-Pathogen Interactions/immunology , Immunoglobulin A/metabolism , Immunoglobulin M/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , Signal Transduction
5.
J Immunol ; 188(2): 800-10, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22147768

ABSTRACT

Foxp3(+) regulatory T (Treg) cells are a critical cell population that suppresses T cell activation in response to microbial and viral pathogens. We identify a cell-intrinsic mechanism by which effector CD4(+) T cells overcome the suppressive effects of Treg cells in the context of three distinct infections: Toxoplasma gondii, Listeria monocytogenes, and vaccinia virus. The acute responses to the parasitic, bacterial, and viral pathogens resulted in a transient reduction in frequency and absolute number of Treg cells. The infection-induced partial loss of Treg cells was essential for the initiation of potent Th1 responses and host protection against the pathogens. The observed disappearance of Treg cells was a result of insufficiency in IL-2 caused by the expansion of pathogen-specific CD4(+) T cells with a limited capacity of IL-2 production. Exogenous IL-2 treatment during the parasitic, bacterial, and viral infections completely prevented the loss of Treg cells, but restoration of Treg cells resulted in a greatly enhanced susceptibility to the pathogens. These results demonstrate that the transient reduction in Treg cells induced by pathogens via IL-2 deprivation is essential for optimal T cell responses and host resistance to microbial and viral pathogens.


Subject(s)
Interleukin-2/deficiency , Listeriosis/immunology , Lymphocyte Activation/immunology , Lymphopenia/immunology , T-Lymphocytes, Regulatory/immunology , Toxoplasmosis, Animal/immunology , Vaccinia/immunology , Acute Disease , Animals , CD4 Lymphocyte Count , Disease Resistance/genetics , Disease Resistance/immunology , Epitopes, T-Lymphocyte/immunology , Interleukin-2/biosynthesis , Interleukin-2/physiology , Listeriosis/microbiology , Listeriosis/pathology , Lymphopenia/pathology , Lymphopenia/prevention & control , Mice , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/pathology , Toxoplasmosis, Animal/parasitology , Toxoplasmosis, Animal/pathology , Vaccinia/pathology , Vaccinia/virology
6.
PLoS Pathog ; 7(12): e1002413, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22174679

ABSTRACT

Small RNA viruses have evolved many mechanisms to increase the capacity of their short genomes. Here we describe the identification and characterization of a novel open reading frame (ORF4) encoded by the murine norovirus (MNV) subgenomic RNA, in an alternative reading frame overlapping the VP1 coding region. ORF4 is translated during virus infection and the resultant protein localizes predominantly to the mitochondria. Using reverse genetics we demonstrated that expression of ORF4 is not required for virus replication in tissue culture but its loss results in a fitness cost since viruses lacking the ability to express ORF4 restore expression upon repeated passage in tissue culture. Functional analysis indicated that the protein produced from ORF4 antagonizes the innate immune response to infection by delaying the upregulation of a number of cellular genes activated by the innate pathway, including IFN-Beta. Apoptosis in the RAW264.7 macrophage cell line was also increased during virus infection in the absence of ORF4 expression. In vivo analysis of the WT and mutant virus lacking the ability to express ORF4 demonstrated an important role for ORF4 expression in infection and virulence. STAT1-/- mice infected with a virus lacking the ability to express ORF4 showed a delay in the onset of clinical signs when compared to mice infected with WT virus. Quantitative PCR and histopathological analysis of samples from these infected mice demonstrated that infection with a virus not expressing ORF4 results in a delayed infection in this system. In light of these findings we propose the name virulence factor 1, VF1 for this protein. The identification of VF1 represents the first characterization of an alternative open reading frame protein for the calicivirus family. The immune regulatory function of the MNV VF1 protein provide important perspectives for future research into norovirus biology and pathogenesis.


Subject(s)
Apoptosis/genetics , Immunity, Innate/genetics , Norovirus/genetics , Open Reading Frames , Viral Proteins/genetics , Virulence Factors/genetics , Amino Acid Sequence , Animals , Apoptosis/immunology , Blotting, Western , Caliciviridae Infections/genetics , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation , Immunoprecipitation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Molecular Sequence Data , Norovirus/immunology , Norovirus/pathogenicity , RNA, Viral/genetics , Real-Time Polymerase Chain Reaction , Reverse Genetics , Reverse Transcriptase Polymerase Chain Reaction , Viral Proteins/immunology , Viral Proteins/metabolism , Virulence/genetics , Virulence Factors/immunology , Virulence Factors/metabolism , Virus Replication/genetics
7.
Proc Natl Acad Sci U S A ; 108(1): 278-83, 2011 Jan 04.
Article in English | MEDLINE | ID: mdl-21173242

ABSTRACT

Toll-like receptors (TLRs) play an important role in host defense against a variety of microbial pathogens. We addressed the mechanism by which TLRs contribute to host defense against the lethal parasite Toxoplasma gondii by using mice with targeted inactivation of the TLR adaptor protein myeloid differentiation primary response gene 88 (MyD88) in different innate cell types. Lack of MyD88 in dendritic cells (DCs), but not in macrophages or neutrophils, resulted in high susceptibility to the T. gondii infection. In the mice deficient in MyD88 in DCs, the early IL-12 response by DCs was ablated, the IFN-γ response by natural killer cells was delayed, and the recruited inflammatory monocytes were incapable of killing the T. gondii parasites. The T-cell response, although attenuated in these mice, was sufficient to eradicate the parasite during the chronic stage, provided that defects in DC activation were compensated by IL-12 treatment early after infection. These results demonstrate a central role of DCs in orchestrating the innate immune response to an intracellular pathogen and establish that defects in pathogen recognition by DCs can predetermine sensitivity to infection.


Subject(s)
Dendritic Cells/immunology , Immunity, Innate/immunology , Toll-Like Receptors/metabolism , Toxoplasmosis/immunology , Animals , Dendritic Cells/metabolism , Flow Cytometry , Interferon-gamma/immunology , Interleukin-12/immunology , Mice , Mice, Knockout , Monocytes/immunology , Myeloid Differentiation Factor 88/genetics , Polymerase Chain Reaction , Statistics, Nonparametric
8.
J Biol Chem ; 286(5): 3307-14, 2011 Feb 04.
Article in English | MEDLINE | ID: mdl-21097503

ABSTRACT

Toll-like receptor (TLR) activation relies on biochemical recognition of microbial molecules and localization of the TLR within specific cellular compartments. Cell surface TLRs largely recognize bacterial membrane components, and intracellular TLRs are exclusively involved in sensing nucleic acids. Here we show that TLR11, an innate sensor for the Toxoplasma protein profilin, is an intracellular receptor that resides in the endoplasmic reticulum. The 12 membrane-spanning endoplasmic reticulum-resident protein UNC93B1 interacts directly with TLR11 and regulates the activation of dendritic cells in response to Toxoplasma gondii profilin and parasitic infection in vivo. A deficiency in functional UNC93B1 protein abolished TLR11-dependent IL-12 secretion by dendritic cells, attenuated Th1 responses against T. gondii, and dramatically enhanced susceptibility to the parasite. Our results reveal that the association with UNC93B1 and the intracellular localization of TLRs are not unique features of nucleic acid-sensing TLRs but is also essential for TLR11-dependent recognition of T. gondii profilin and for host protection against this parasite.


Subject(s)
Interleukin-12/immunology , Membrane Transport Proteins/immunology , Toll-Like Receptors/metabolism , Toxoplasma/immunology , Animals , Dendritic Cells/immunology , Immunity , Mice , Mice, Inbred C57BL , Profilins/metabolism , Th1 Cells/immunology , Toll-Like Receptors/analysis , Toll-Like Receptors/deficiency , Toxoplasmosis, Animal/immunology , Toxoplasmosis, Animal/parasitology
9.
J Virol ; 84(6): 2859-70, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20053745

ABSTRACT

Interactions of host cell factors with RNA sequences and structures in the genomes of positive-strand RNA viruses play various roles in the life cycles of these viruses. Our understanding of the functional RNA elements present in norovirus genomes to date has been limited largely to in vitro analysis. However, we recently used reverse genetics to identify evolutionarily conserved RNA structures and sequences required for norovirus replication. We have now undertaken a more detailed analysis of RNA structures present at the 3' extremity of the murine norovirus (MNV) genome. Biochemical data indicate the presence of three stable stem-loops, including two in the untranslated region, and a single-stranded polypyrimidine tract [p(Y)] of variable length between MNV isolates, within the terminal stem-loop structure. The well-characterized host cell pyrimidine binding proteins PTB and PCBP bound the 3'-untranslated region via an interaction with this variable sequence. Viruses lacking the p(Y) tract were viable both in cell culture and upon mouse infection, demonstrating that this interaction was not essential for virus replication. However, competition analysis with wild-type MNV in cell culture indicated that the loss of the p(Y) tract was associated with a fitness cost. Furthermore, a p(Y)-deleted mutant showed a reduction in virulence in the STAT1(-/-) mouse model, highlighting the role of RNA structures in norovirus pathogenesis. This work highlights how, like with other positive-strand RNA viruses, RNA structures present at the termini of the norovirus genome play important roles in virus replication and virulence.


Subject(s)
3' Untranslated Regions , Base Sequence , Genome, Viral , Norovirus , Nucleic Acid Conformation , RNA, Viral/chemistry , RNA, Viral/genetics , Animals , Cell Line , Evolution, Molecular , Gene Expression Regulation, Viral , Humans , Mice , Mice, Knockout , Molecular Sequence Data , Mutation , Norovirus/genetics , Norovirus/pathogenicity , Open Reading Frames , Pyrimidines , RNA, Viral/metabolism , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Virus Replication/genetics
10.
Cell Host Microbe ; 6(2): 187-96, 2009 Aug 20.
Article in English | MEDLINE | ID: mdl-19683684

ABSTRACT

Toxoplasma gondii is a universally distributed pathogen that infects over one billion people worldwide. Host resistance to this protozoan parasite depends on a Th1 immune response with potent production of the cytokines interleukin-12 and interferon gamma. Although Toll-like receptor 11 (TLR11) plays a major role in controlling Th1 immunity to this pathogen in mice, this innate immune receptor is nonfunctional in humans, and the mechanisms of TLR11-independent sensing of T. gondii remain elusive. Here, we show that oral infection by T. gondii triggers a TLR11-independent but MyD88-dependent Th1 response that is impaired in TLR2xTLR4 double knockout and TLR9 single knockout mice. These mucosal innate and adaptive immune responses to T. gondii rely on the indirect stimulation of dendritic cells by normal gut microflora. Thus, our results reveal that gut commensal bacteria can serve as molecular adjuvants during parasitic infection, providing indirect immunostimulation that protects against T. gondii in the absence of TLR11.


Subject(s)
Bacteria/immunology , Dendritic Cells/immunology , Gastrointestinal Tract/microbiology , Toxoplasma/immunology , Toxoplasmosis, Animal/immunology , Animals , Cytokines/biosynthesis , Humans , Leukocytes, Mononuclear/immunology , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/immunology , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/immunology , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/immunology , Toll-Like Receptor 9/deficiency , Toll-Like Receptor 9/immunology , Toxoplasmosis, Animal/parasitology , Toxoplasmosis, Animal/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...