Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Cell Microbiol ; 12(5): 626-39, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20039881

ABSTRACT

The cytosolic innate immune molecule, NOD1, recognizes peptidoglycan (PG) delivered to epithelial cells via the Helicobacter pylori cag pathogenicity island (cagPAI), and has been implicated in host defence against cagPAI(+)H. pylori bacteria. To further clarify the role of NOD1 in host defence, we investigated NOD1-dependent regulation of human beta-defensins (DEFBs) in two epithelial cell lines. Our findings identify that NOD1 activation, via either cagPAI(+) bacteria or internalized PG, was required for DEFB4 and DEFB103 expression in HEK293 cells. To investigate cell type-specific induction of DEFB4 and DEFB103, we generated stable NOD1'knockdown' (KD) and control AGS cells. Reporter gene assay and RT-PCR analyses revealed that only DEFB4 was induced in an NOD1-/cagPAI-dependent fashion in AGS cells. Moreover, culture supernatants from AGS control, but not AGS NOD1 KD cells, stimulated with cagPAI(+)H. pylori, significantly reduced H. pylori bacterial numbers. siRNA studies confirmed that human beta-defensin 2 (hBD-2), but not hBD-3, contributes to the antimicrobial activity of AGS cell supernatants against H. pylori. This study demonstrates, for the first time, the involvement of NOD1 and hBD-2 in direct killing of H. pylori bacteria by epithelial cells and confirms the importance of NOD1 in host defence mechanisms against cagPAI(+)H. pylori infection.


Subject(s)
Epithelial Cells/immunology , Helicobacter pylori/immunology , Nod1 Signaling Adaptor Protein/immunology , beta-Defensins/immunology , Cell Line , Gene Knockdown Techniques , Humans , Microbial Viability , Nod1 Signaling Adaptor Protein/genetics , Peptidoglycan/immunology , beta-Defensins/biosynthesis
2.
Cell Microbiol ; 12(3): 372-85, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19888989

ABSTRACT

Gram-negative bacterial peptidoglycan is specifically recognized by the host intracellular sensor NOD1, resulting in the generation of innate immune responses. Although epithelial cells are normally refractory to external stimulation with peptidoglycan, these cells have been shown to respond in a NOD1-dependent manner to Gram-negative pathogens that can either invade or secrete factors into host cells. In the present work, we report that Gram-negative bacteria can deliver peptidoglycan to cytosolic NOD1 in host cells via a novel mechanism involving outer membrane vesicles (OMVs). We purified OMVs from the Gram-negative mucosal pathogens: Helicobacter pylori, Pseudomonas aeruginosa and Neisseria gonorrhoea and demonstrated that these peptidoglycan containing OMVs upregulated NF-kappaB and NOD1-dependent responses in vitro. These OMVs entered epithelial cells through lipid rafts thereby inducing NOD1-dependent responses in vitro. Moreover, OMVs delivered intragastrically to mice-induced innate and adaptive immune responses via a NOD1-dependent but TLR-independent mechanism. Collectively, our findings identify OMVs as a generalized mechanism whereby Gram-negative bacteria deliver peptidoglycan to cytosolic NOD1. We propose that OMVs released by bacteria in vivo may promote inflammation and pathology in infected hosts.


Subject(s)
Epithelial Cells/immunology , Gram-Negative Bacteria/immunology , NF-kappa B/immunology , Nod1 Signaling Adaptor Protein/immunology , Peptidoglycan/metabolism , Secretory Vesicles/metabolism , Animals , Female , HeLa Cells , Helicobacter pylori/immunology , Humans , Male , Mice , Neisseria/immunology , Peptidoglycan/immunology , Pseudomonas aeruginosa/immunology , Secretory Vesicles/immunology
3.
J Immunol ; 183(12): 8099-109, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-20007577

ABSTRACT

Helicobacter pylori rapidly activates MAPKs and transcription factors, NF-kappaB and AP-1, in gastric epithelial cells following host attachment. Activation of these signal transducers is largely dependent on the cag pathogenicity island (cagPAI)-encoded Type IV Secretion System. H. pylori was shown to translocate peptidoglycan through the Type IV Secretion System, which is recognized by the pathogen recognition molecule, NOD1, thus resulting in NF-kappaB activation. The mechanisms of H. pylori-induced MAPK and AP-1 activation, however, are less well defined and therefore, we assessed the contribution of NOD1 to their activation. For this, we used gastric epithelial cell lines, stably expressing siRNA to either NOD1 or a control gene. In siNOD1-expressing cells stimulated with cagPAI(+) H. pylori, we observed significant reductions in p38 and ERK phosphorylation (p < 0.05), whereas the levels of Jnk phosphorylation remained unchanged. Consistent with a previous report, however, we were able to demonstrate NOD1-dependent Jnk phosphorylation by the invasive pathogen Shigella flexneri, highlighting pathogen-specific host responses to infection. We also show that NOD1 was essential for H. pylori induction of not only NF-kappaB, but also AP-1 activation, implying that NOD1 induces robust proinflammatory responses, in an attempt to rapidly control infection. Pharmacological inhibition of p38 and ERK activity significantly reduced IL-8 production in response to H. pylori, further emphasizing the importance of MAPKs in innate immune responses to the pathogen. Thus, for the first time we have shown the important role for NOD1 in MAPK and AP-1 activation in response to cagPAI(+) H. pylori.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Helicobacter pylori/immunology , Nod1 Signaling Adaptor Protein/physiology , Transcription Factor AP-1/metabolism , Antigens, Bacterial/physiology , Bacterial Outer Membrane Proteins/metabolism , Bacterial Outer Membrane Proteins/physiology , Bacterial Proteins/physiology , Cell Line , Cell Line, Tumor , Enzyme Activation/immunology , Extracellular Signal-Regulated MAP Kinases/physiology , Gene Targeting , Humans , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , MAP Kinase Kinase 4/metabolism , Nod1 Signaling Adaptor Protein/antagonists & inhibitors , Nod1 Signaling Adaptor Protein/genetics , Phosphorylation/immunology , Secretory Vesicles/metabolism , Signal Transduction/immunology
4.
BMC Microbiol ; 9: 30, 2009 Feb 06.
Article in English | MEDLINE | ID: mdl-19200386

ABSTRACT

BACKGROUND: Enteropathogenic Escherichia coli (EPEC) is an attaching and effacing (A/E) pathogen that possesses a type III secretion system (T3SS) encoded within the locus of enterocyte effacement (LEE). The LEE is essential for A/E lesion formation and directs the secretion and translocation of multiple LEE-encoded and non-LEE encoded effector proteins into the cytosol of infected cells. In this study we used proteomics to compare proteins exported to the culture supernatant by wild type EPEC E2348/69, a DeltaespADB mutant and a DeltaescF T3SS mutant. RESULTS: We observed that flagellin was consistently and strongly present in the secretome of wild type EPEC and the DeltaespADB mutant but present only weakly in the secretome of the DeltaescF mutant. Given the ancestral relationship between the flagella export apparatus and virulence associated T3SSs, we investigated whether FliC could utilise the LEE-encoded T3SS for export. In the absence of a functional flagella export apparatus, we showed that FliC could be secreted by the LEE-encoded T3SS and stimulate (Toll-like receptor 5) TLR5 signalling but could not confer motility. CONCLUSION: Since the secretion of FliC during A/E lesion formation would presumably be disadvantageous for the pathogen, we propose that virulence associated T3SSs and flagella T3SSs have evolved through a system of chaperones and complex regulatory pathways to be functional at different times to ensure that FliC secretion does not occur during T3SS effector translocation.


Subject(s)
Enteropathogenic Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Flagellin/metabolism , Phosphoproteins/metabolism , DNA, Bacterial/metabolism , Electrophoresis, Gel, Two-Dimensional , Enteropathogenic Escherichia coli/genetics , Flagella/metabolism , Gene Expression Regulation, Bacterial , Mutation , Proteome/metabolism , Proteomics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Toll-Like Receptor 5/metabolism
5.
Infect Immun ; 76(5): 2235-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18332213

ABSTRACT

Helicobacter pylori is the etiological agent of human chronic gastritis, a condition seen as a precursor to the development of gastrointestinal ulcers or gastric cancer. This study utilized the murine model of chronic H. pylori infection to characterize the role of macrophages in the induction of specific immune responses and gastritis and in the control of the bacterial burden following H. pylori infection and vaccination. Drug-loaded liposomes were injected intravenously to deplete macrophages from C57BL/6 mice, and effective removal of CD11b+ cells from the spleens and stomachs of mice was confirmed by immunofluorescence microscopy. Transient elimination of macrophages from C57BL/6 mice during the early period of infection reduced the gastric pathology induced by H. pylori SS1 but did not affect the bacterial load in the stomach. These data suggest that macrophages are important to the severity of gastric inflammation during H. pylori infection.


Subject(s)
Gastritis/microbiology , Helicobacter Infections/immunology , Helicobacter Infections/microbiology , Helicobacter pylori/immunology , Macrophages/immunology , Macrophages/microbiology , Animals , CD11b Antigen/analysis , Colony Count, Microbial , Female , Gastritis/immunology , Helicobacter pylori/isolation & purification , Leukocyte Reduction Procedures , Macrophages/chemistry , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Spleen/cytology , Stomach/cytology , Stomach/microbiology , Stomach/pathology
6.
PLoS One ; 4(4): e5396, 2009.
Article in English | MEDLINE | ID: mdl-19401779

ABSTRACT

Helicobacter spp. represent a proportionately small but significant component of the normal intestinal microflora of animal hosts. Several of these intestinal Helicobacter spp. are known to induce colitis in mouse models, yet the mechanisms by which these bacteria induce intestinal inflammation are poorly understood. To address this question, we performed in vitro co-culture experiments with mouse and human epithelial cell lines stimulated with a selection of Helicobacter spp., including known pathogenic species as well as ones for which the pathogenic potential is less clear. Strikingly, a member of the normal microflora of rodents, Helicobacter muridarum, was found to be a particularly strong inducer of CXC chemokine (Cxcl1/KC, Cxcl2/MIP-2) responses in a murine intestinal epithelial cell line. Time-course studies revealed a biphasic pattern of chemokine responses in these cells, with H. muridarum lipopolysaccharide (LPS) mediating early (24-48 h) responses and live bacteria seeming to provoke later (48-72 h) responses. H. muridarum LPS per se was shown to induce CXC chemokine production in HEK293 cells stably expressing Toll-like receptor 2 (TLR2), but not in those expressing TLR4. In contrast, live H. muridarum bacteria were able to induce NF-kappaB reporter activity and CXC chemokine responses in TLR2-deficient HEK293 and in AGS epithelial cells. These responses were attenuated by transient transfection with a dominant negative construct to NOD1, and by stable expression of NOD1 siRNA, respectively. Thus, the data suggest that both TLR2 and NOD1 may be involved in innate immune sensing of H. muridarum by epithelial cells. This work identifies H. muridarum as a commensal bacterium with pathogenic potential and underscores the potential roles of ill-defined members of the normal flora in the initiation of inflammation in animal hosts. We suggest that H. muridarum may act as a confounding factor in colitis model studies in rodents.


Subject(s)
Helicobacter/pathogenicity , Intestines/microbiology , Nod1 Signaling Adaptor Protein/metabolism , Toll-Like Receptor 2/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Chemokine CXCL1/biosynthesis , Chemokine CXCL2/biosynthesis , Chemokines, CXC/biosynthesis , Coculture Techniques , Colitis/etiology , Colitis/immunology , Colitis/metabolism , Colitis/microbiology , DNA Primers/genetics , Disease Models, Animal , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Flagellin/genetics , Helicobacter/genetics , Helicobacter/immunology , Helicobacter/isolation & purification , Helicobacter Infections/etiology , Helicobacter Infections/metabolism , Helicobacter Infections/microbiology , Humans , Immunity, Innate , Intestines/immunology , Lipopolysaccharides/toxicity , Mice , Molecular Sequence Data , NF-kappa B/metabolism , Nod1 Signaling Adaptor Protein/antagonists & inhibitors , Nod1 Signaling Adaptor Protein/genetics , Sequence Homology, Amino Acid , Species Specificity , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
7.
Immunol Cell Biol ; 85(6): 495-502, 2007.
Article in English | MEDLINE | ID: mdl-17680011

ABSTRACT

Eukaryotic organisms of the plant and animal kingdoms have developed evolutionarily conserved systems of defence against microbial pathogens. These systems depend on the specific recognition of microbial products or structures by molecules of the host innate immune system. The first mammalian molecules shown to be involved in innate immune recognition of, and defence against, microbial pathogens were the Toll-like receptors (TLRs). These proteins are predominantly but not exclusively located in the transmembrane region of host cells. Interestingly, mammalian hosts were subsequently found to also harbour cytosolic proteins with analogous structures and functions to plant defence molecules. The members of this protein family exhibit a tripartite domain structure and are characterized by a central nucleotide-binding oligomerization domain (NOD). Moreover, in common with TLRs, most NOD proteins possess a C-terminal leucine-rich repeat (LRR) domain, which is required for the sensing of microbial products and structures. Recently, the name 'nucleotide-binding domain and LRR' (NLR) was coined to describe this family of proteins. It is now clear that NLR proteins play key roles in the cytoplasmic recognition of whole bacteria or their products. Moreover, it has been demonstrated in animal studies that NLRs are important for host defence against bacterial infection. This review will particularly focus on two subfamilies of NLR proteins, the NODs and 'NALPs', which specifically recognize bacterial products, including cell wall peptidoglycan and flagellin. We will discuss the downstream signalling events and host cell responses to NLR recognition of such products, as well as the strategies that bacterial pathogens employ to trigger NLR signalling in host cells. Cytosolic recognition of microbial factors by NLR proteins appears to be one mechanism whereby the innate immune system is able to discriminate between pathogenic bacteria ('foe') and commensal ('friendly') members of the host microflora.


Subject(s)
Nod Signaling Adaptor Proteins/immunology , Animals , Bacterial Infections/immunology , Caspase 1/immunology , Evolution, Molecular , Humans , Peptidoglycan/immunology , Signal Transduction
8.
Appl Environ Microbiol ; 73(3): 1010-3, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17142378

ABSTRACT

We examined the impact of Helicobacter pylori infection on the murine gastric microbiota by culture and terminal-restriction fragment length polymorphism and found that neither acute nor chronic H. pylori infection substantially affected the gastric microbial composition. Interestingly, the total H. pylori burden detected by real-time PCR was significantly higher than that revealed by viable counts, suggesting that the antigenic load sustaining H. pylori-induced gastritis could be considerably higher than previously believed.


Subject(s)
Bacteria/growth & development , Helicobacter pylori/pathogenicity , Stomach/microbiology , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Chronic Disease , Female , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter pylori/genetics , Helicobacter pylori/isolation & purification , Lactobacillus/classification , Lactobacillus/genetics , Lactobacillus/isolation & purification , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Polymorphism, Restriction Fragment Length
9.
Gastroenterology ; 133(4): 1210-8, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17919495

ABSTRACT

BACKGROUND & AIMS: The MUC1 mucin is expressed on the cell surface of epithelial cells lining the gastric mucosa. Epidemiologic studies suggest that functional allelic variations in the MUC1 gene may play a role in human susceptibility to Helicobacter pylori-associated pathologies, including gastric adenocarcinoma. We have evaluated the impact of Muc1 expression on the colonization and pathogenesis of gastric Helicobacter infections. METHODS: Wild-type and Muc1-deficient mice were infected with H pylori and colonization and gastritis levels determined. Primary gastric cells were used to examine the impact of Muc1 expression on bacterial adherence. RESULTS: Mice lacking Muc1 were colonized by 5-fold more H pylori within 1 day of infection, and this difference was maintained for at least 2 months postinfection. Mice heterozygous for the null Muc1 allele developed intermediate bacterial colonization. Although wild-type mice developed only a mild gastritis when infected for 2 months with H pylori, Muc1(-/-) mice developed an atrophic gastritis marked by loss of parietal cells. We demonstrate H pylori adhesion to purified MUC1 and significantly increased adhesion to cultured murine Muc1 null gastric epithelial cells, suggesting that Muc1 acts as a decoy limiting binding to the cell surface. CONCLUSIONS: Muc1 provides a protective barrier, which limits both acute and chronic colonization by H pylori, as well as playing a major role in limiting the inflammation induced by Helicobacter infection. We propose that Muc1 restricts access of H pylori to the epithelial surface, hence reducing exposure of the host to proinflammatory bacterial products.


Subject(s)
Gastric Mucosa/microbiology , Gastritis/microbiology , Helicobacter Infections/microbiology , Helicobacter pylori/pathogenicity , Mucin-1/metabolism , Animals , Antibody Formation , Bacterial Adhesion , Cell Line, Tumor , Cells, Cultured , Colony Count, Microbial , Disease Models, Animal , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Gastritis/metabolism , Gastritis/pathology , Gastritis/prevention & control , Helicobacter Infections/complications , Helicobacter Infections/immunology , Helicobacter Infections/metabolism , Helicobacter Infections/pathology , Helicobacter pylori/isolation & purification , Humans , Immunity, Cellular , Mice , Mice, Knockout , Mucin-1/genetics , Parietal Cells, Gastric/metabolism , Parietal Cells, Gastric/microbiology , Protein Binding , Severity of Illness Index , Time Factors
10.
Infect Immun ; 74(6): 3519-29, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16714584

ABSTRACT

Gastric Helicobacter spp. induce chronic gastritis that may lead to ulceration and dysplasia. The host elicits a T helper 1 (Th1) response that is fundamental to the pathogenesis of these bacteria. We analyzed immune responses in Helicobacter-infected, normal mice depleted of CD4+ CD25+ T cells to investigate the in vivo role of regulatory T cells (Tregs) in the modulation of Helicobacter immunopathology. BALB/c and transgenic mice were depleted of CD4+ CD25+ T cells by administration of an anti-CD25 antibody either at the time of infection with Helicobacter or during chronic infection and gastritis. Depletion of CD25+ Tregs prior to and during infection of mice with Helicobacter spp. did not affect either bacterial colonization or severity of gastritis. Depletion of CD25+ Tregs was associated with increased Helicobacter-specific antibody levels and an altered isotype distribution. Paragastric lymph node cells from CD25+ Treg-depleted and control infected mice showed similar proliferation to Helicobacter antigens, but only cells from anti-CD25-treated animals secreted Th2 cytokines. CD25+ Tregs do not control the level of gastritis induced by gastric Helicobacter spp. in normal, thymus-intact BALB/c mice. However, CD25+ Tregs influence the cytokine and antibody responses induced by infection. Autoimmune gastritis is not induced in Helicobacter-infected mice depleted of CD25+ Tregs but is induced in CD25+ Treg-depleted mice, which have a higher frequency of autoreactive T cells.


Subject(s)
Antibodies, Bacterial/blood , Helicobacter Infections/immunology , T-Lymphocytes, Regulatory/physiology , T-Lymphocytes/immunology , Animals , Antigens, CD , Antigens, Differentiation/physiology , Autoimmunity , CTLA-4 Antigen , Cytokines/biosynthesis , Female , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Gastritis/etiology , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Immunoglobulin Isotypes/blood , Mice , Mice, Inbred BALB C
SELECTION OF CITATIONS
SEARCH DETAIL