20 research outputs found

    Increased autophagic sequestration in adaptor protein-3 deficient dendritic cells limits inflammasome activity and impairs antibacterial immunity

    No full text
    <div><p>Bacterial pathogens that compromise phagosomal membranes stimulate inflammasome assembly in the cytosol, but the molecular mechanisms by which membrane dynamics regulate inflammasome activity are poorly characterized. We show that in murine dendritic cells (DCs), the endosomal adaptor protein AP-3 –which optimizes toll-like receptor signaling from phagosomes–sustains inflammasome activation by particulate stimuli. AP-3 independently regulates inflammasome positioning and autophagy induction, together resulting in delayed inflammasome inactivation by autophagy in response to <i>Salmonella</i> Typhimurium (STm) and other particulate stimuli specifically in DCs. AP-3-deficient DCs, but not macrophages, hyposecrete IL-1β and IL-18 in response to particulate stimuli <i>in vitro</i>, but caspase-1 and IL-1β levels are restored by silencing autophagy. Concomitantly, AP-3-deficient mice exhibit higher mortality and produce less IL-1β, IL-18, and IL-17 than controls upon oral STm infection. Our data identify a novel link between phagocytosis, inflammasome activity and autophagy in DCs, potentially explaining impaired antibacterial immunity in AP-3-deficient patients.</p></div

    AP-3 is required for optimal transcriptional activation of pro-IL-1β and some NLRs after particulate LPS priming.

    No full text
    <p>BMDCs from WT or pearl (pe) mice were untreated or stimulated with LPS or LPS beads for 2 h (<b>A</b>-<b>C</b>) or 3 h (<b>D</b>, <b>E</b>). <b>A-C</b>. cDNA generated from isolated RNA was analyzed by RT-PCR. Shown are mRNA levels of: <b>A</b>, NLRC1, NLRC2, NLRP3; <b>B</b>, pro-IL-1β, pro-IL-18; and <b>C</b>, NLRC4, pro-caspase-1 and ASC. Data from three independent experiments were normalized to the average of five housekeeping genes, and the ΔΔCt values were calculated and represented as mean ± SD fold induction of mRNA in stimulated cells relative to unstimulated cells. <b>D</b>, <b>E.</b> Cell pellets were lysed and fractionated by SDS-PAGE, and NLRP3, NLRC4, pro-caspase-1 (pro-casp. 1), pro-IL-1β and ASC were detected by immunoblotting. <b>D</b>, representative blots. <b>E</b>, quantification of band intensities represented as mean ± SD fold induction in stimulated cells relative to unstimulated cells for pro-IL-1β (<i>top</i>) and NLRP3 (<i>bottom</i>), normalized to tubulin levels. Two or more fold induction was considered significant. *p< 0.05; **p<0.01. See also <b><a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1006785#ppat.1006785.s008" target="_blank">S1 Table</a></b>.</p

    AP-3 promotes survival upon <i>Salmonella</i> Typhimurium infection.

    No full text
    <p>WT CD57BL/6J and congenic pearl (pe) mice were infected orally with 10<sup>8</sup> STm (+ STm) or received PBS as a control (naïve). (<b>A, B).</b> Mouse survival was assessed over 12 days (<b>A</b>; n = 5) or 7 days (<b>B</b>; n = 11 each mouse type; surviving mice were euthanized on day 7). (<b>C-E).</b> Peyer patches, MLN and spleens were harvested 5 days post-infection, homogenized and plated to measure bacterial load. CFUs were normalized to tissue weight (expressed as CFU/ g of tissue). Data are pooled from three independent experiments. Dotted lines, background (threshold value from uninfected mice); solid lines, geometric means of values above background. *p<0.05; **p<0.01; ***p<0.001. See also <b><a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1006785#ppat.1006785.s001" target="_blank">S1</a> and <a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1006785#ppat.1006785.s002" target="_blank">S2</a> Figs</b>.</p

    AP-3 is required for perinuclear inflammasome positioning and limits autophagy induction after <i>Salmonella</i> Typhimurium infection in DCs.

    No full text
    <p><b>(A-C).</b> WT and pearl (pe) BMDCs expressing ASC-GFP were infected with flagellin-expressing mCherry-STm (stimulates NLRC4). Cells were fixed 1 h after infection, labeled with DAPI and analyzed by fluorescence microscopy. <b>A.</b> Representative images showing ASC speck (green) relative to STm (red) and nucleus (blue) in four infected WT and pearl DCs each. <b>B.</b> Quantification of perinuclear (within a radius of one μm from the nucleus) and non-perinuclear ASC specks in 20 cells per cell type in each of four independent experiments. <b>C.</b> Quantification of ASC speck distance to the nucleus in 15 cells per cell type in each of three independent experiments. <b>(D</b>, <b>E)</b>. WT and pearl BMDCs were infected with STm, and endogenous LC3 (and actin as a control) was detected by immunoblotting fractionated cell lysates at the indicated time points. <b>D.</b> Representative blot with positions of molecular weight markers (MW) indicated at left. <b>E</b>. Quantification of LC3-II band intensities from three independent experiments, expressed as fold increase relative to unstimulated cells and normalized to LC3-I and β-actin levels. (<b>F</b>-<b>I).</b> WT and pearl BMDCs expressing ASC-GFP alone or with mCherry-LC3 were infected with STm and analyzed by live fluorescence imaging (for LC3) or fixed immunofluorescence microscopy (for p62) 1 h later. <b>F.</b> Representative images showing ASC speck (green) and either LC3 puncta (red, <i>left panels</i>) or endogenous p62 puncta (red) and nuclei (blue; <i>right panels</i>) in infected cells. Corresponding DIC images show nuclear position. <b>G, H.</b> Quantification of LC3 (<b>G</b>) or p62 (<b>H</b>) puncta within a radius of 0.5 μm from the ASC speck (representative image shown at right) in 15 cells per cell type in each of 3 independent experiments. <b>I</b>, Quantification of total p62 puncta normalized to cell area. Data represent mean ± SD. Scale bar: 10 μm.**p<0.01; ***p<0.001. See also <b><a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1006785#ppat.1006785.s005" target="_blank">S5</a></b>and <b><a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1006785#ppat.1006785.s006" target="_blank">S6</a> Figs</b>.</p

    AP-3 limits inflammasome sequestration and autophagy induction after STm infection or alum stimulation.

    No full text
    <p><b>(A</b>, <b>B)</b>. WT and pearl (pe) BMDCs expressing ASC-GFP were infected with flagellin-expressing STm (stimulates NLRC4) for 30 or 60 min. Cells were then permeabilized for 1 min with 50 μg/ml digitonin or throughout labeling with 0.1% saponin as indicated, washed, and incubated with mouse anti-GFP and allophycocyanin (APC)-conjugated anti-mouse antibodies. Cells were analyzed by flow cytometry, gating on GFP<sup>+</sup> cells (R1). <b>A</b>. Shown are representative dot plots of transduced WT and pe DCs indicating gated region based on GFP fluorescence and side scatter (SSC, <i>left panels</i>), and representative histogram plots indicating GFP (<i>middle panels</i>) or APC (anti-GFP) fluorescence (<i>right panels</i>). <i>Black lines</i>, WT; <i>blue lines</i>, pe; <i>dotted lines</i>, secondary antibody alone. <b>B</b>. The ratio of mean fluorescence intensity (MFI) values for anti-GFP signal in digitonin-treated DCs relative to saponin-treated DCs is shown from 4 independent experiments. (<b>C-L)</b>. WT and pearl (pe) BMDCs that were non-transduced (-) or transduced with lentiviruses encoding non-target (ctrl) or either of two ATG7-specific shRNAs or ATG-5- or LC3b-specific shRNAs were infected with STm (<b>C-E</b>) or primed for 3 h with soluble LPS and stimulated with alum (<b>F-L</b>). (<b>C-E</b>) Cell supernatants collected 2 h after Stm infection were assayed for IL-1β by ELISA. <b>C.</b> Representative experiment. <b>D</b>. Data from 3 independent experiments were normalized to IL-1β values from cells treated with non-target shRNA and presented as fold induction. <b>E</b>. IL-1β values for pearl BMDC treated with non-target or ATG7 shRNAs from 3 independent experiments are shown as percent of values for WT DCs treated with the same shRNAs. (<b>F-L</b>) Cell pellets collected 4 h after alum stimulation were lysed, fractionated by SDS-PAGE and immunoblotted for caspase-1 and tubulin. (<b>F, H</b>). Representative immunoblots, showing pro-caspase-1 (pro-casp.-1) and mature p10 (casp.-1 p10) bands. (<b>G, I</b>) Quantification of band intensities for caspase-1 p10 normalized to pro-caspase-1 and tubulin from three independent experiments are shown as caspase-1 fold change relative to unstimulated (-) WT cells (mean ± SD). (<b>J, K</b>) Data from three independent experiments were normalized to caspase-1 values from untreated cells and presented as fold increase. (<b>L</b>). Caspase-1 values for pearl BMDC treated with non-target, ATG5, ATG7 or LC3b shRNAs from 3 independent experiments are shown as percent of values for WT DCs treated with the same shRNAs. Data in all panels represent mean ± SD. **p<0.01; ***p<0.001. See also <b><a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1006785#ppat.1006785.s007" target="_blank">S7 Fig</a></b>.</p

    Neutrophils and Ly6C<sup>hi</sup> monocytes collaborate in generating an optimal cytokine response that protects against pulmonary <i>Legionella pneumophila</i> infection

    No full text
    <div><p>Early responses mounted by both tissue-resident and recruited innate immune cells are essential for host defense against bacterial pathogens. In particular, both neutrophils and Ly6C<sup>hi</sup> monocytes are rapidly recruited to sites of infection. While neutrophils and monocytes produce bactericidal molecules, such as reactive nitrogen and oxygen species, both cell types are also capable of synthesizing overlapping sets of cytokines important for host defense. Whether neutrophils and monocytes perform redundant or non-redundant functions in the generation of anti-microbial cytokine responses remains elusive. Here, we sought to define the contributions of neutrophils and Ly6C<sup>hi</sup> monocytes to cytokine production and host defense during pulmonary infection with <i>Legionella pneumophila</i>, responsible for the severe pneumonia Legionnaires’ disease. We found that both neutrophils and monocytes are critical for host defense against <i>L</i>. <i>pneumophila</i>. Both monocytes and neutrophils contribute to maximal IL-12 and IFNγ responses, and monocytes are also required for TNF production. Moreover, natural killer (NK) cells, NKT cells, and γδ T cells are sources of IFNγ, and monocytes direct IFNγ production by these cell types. Thus, neutrophils and monocytes cooperate in eliciting an optimal cytokine response that promotes effective control of bacterial infection.</p></div

    Exogenously administered IL-12 partially restores bacterial control and IFNγ production in mice lacking neutrophils and/or monocytes following infection.

    No full text
    <p>B6 mice treated with isotype control (ISO) or anti-Gr-1 (α-Gr-1) antibody (A), Ccr2<sup>-/-</sup> mice (B), or B6 mice treated with isotype control or anti-Ly6G (α-Ly6G) antibody (C) were infected with Δ<i>flaA L</i>. <i>pneumophila</i> and given 500ng of recombinant IL-12 (rIL-12) intranasally. CFUs were enumerated and IFNγ levels in BALF were measured 72 hours post-infection. Data shown are the pooled results of 2–3 independent experiments with 3 or 4 mice per group per experiment. * is p<0.05, ** is p<0.01 and *** is p<0.001 by unpaired t-test. NS is not significant.</p

    Ly6C<sup>hi</sup> monocytes are required for control of pulmonary <i>L</i>. <i>pneumophila</i> infection.

    No full text
    <p>B6 or <i>Ccr2</i><sup>-/-</sup> mice were infected with Δ<i>flaA L</i>. <i>pneumophila</i>. (A) Representative flow cytometry plots of lung cells from B6 or <i>Ccr2</i><sup>-/-</sup> mice 24 hours post-infection, with gates drawn around neutrophils and Ly6C<sup>hi</sup> cells. Ly6C<sup>hi</sup> monocytes (MCs) were further defined as CD11b<sup>+</sup> cells. Total numbers of Ly6C<sup>hi</sup> MCs (B), neutrophils (C), and DCs (D) in the lung were quantified at 24 and 48 hours post-infection. (E) <i>L</i>. <i>pneumophila</i> CFUs in the lung were enumerated at 24, 48, and 96 hours post-infection. Data shown are the pooled results of 3 to 5 independent experiments per time point with 3 or 4 mice per group per experiment. * is p<0.05 and *** is p<0.001 by unpaired t-test. NS is not significant. Dashed line represents the limit of detection.</p

    Gr-1<sup>+</sup> cells are required for TNF and IL-12 production early during pulmonary infection with <i>L</i>. <i>pneumophila</i>.

    No full text
    <p>B6 mice treated with isotype control (ISO) or anti-Gr-1 (α-Gr-1) antibody were infected with Δ<i>flaA L</i>. <i>pneumophila</i>. Levels of IL-1α (A), IL-1β (B), IL-6 (C), TNF (D), IL-18 (E), IL-12p70 (F), IL-12p40 (G), IL-4 (H), and IL-10 (I) in the BALF at 24 and 72 hours post-infection were quantified by ELISA. Data shown are the pooled results of 2 independent experiments with 3 or 4 mice per group per experiment. * is p<0.05, ** is p<0.01, and *** is p<0.001 by unpaired t-test. NS is not significant. Dashed line represents the limit of detection.</p

    Neutrophils are required for maximal IL-12 production early during pulmonary infection with <i>L</i>. <i>pneumophila</i>.

    No full text
    <p>B6 mice treated with either isotype control (ISO) or anti-Ly6G (α-Ly6G) antibody were infected with <i>ΔflaA L</i>. <i>pneumophila</i>. Levels of IL-1α (A), IL-1β (B), IL-6 (C), TNF (D), IL-18 (E), IL-12p70 (F), and IL-12p40 (G) in the BALF at 24 and 72 hours post-infection were quantified by ELISA. Data shown are the pooled results of 4 independent experiments with 3 or 4 mice per group per experiment. * is p<0.05, ** is p<0.01, and *** is p<0.001 by unpaired t-test. NS is not significant. Dashed line represents the limit of detection.</p
    corecore