20 research outputs found

    Correction: Non-Canonical NF-κB Activation and Abnormal B Cell Accumulation in Mice Expressing Ubiquitin Protein Ligase-Inactive c-IAP2

    No full text
    <p>Correction: Non-Canonical NF-κB Activation and Abnormal B Cell Accumulation in Mice Expressing Ubiquitin Protein Ligase-Inactive c-IAP2</p

    Generation of c-IAP1<sup>H582A</sup> mice.

    No full text
    <p>(A) Schematic representation of the c-IAP1 targeting construct and the recombination strategy. (B) c-IAP1<sup>H582A</sup> mice were distinguished from WT littermates by LR-PCR and Spe I digestion. (C) c-IAP1 and c-IAP2 expression in splenocytes from WT, c-IAP1<sup>H582A</sup> heterozygous and homozygous mice were determined by immunoblotting. (D) <i>Ciap1</i> and <i>Ciap2</i> mRNA expression was determined in WT and c-IAP1<sup>H582A</sup> splenocytes by real-time PCR. Bars represent the fold increase over WT expression. (E) WT or c-IAP1<sup>H582A</sup> splenocytes were lysed in sample buffer after 8 hr of incubation with 25 mM chloroquine (CHQ), 1∶200 of the Sigma protease inhibitor cocktail (PIs), 10 µM MG-132, or complete medium (−) and c-IAP1 expression was analyzed by immunoblotting.</p

    c-IAP1 and c-IAP2 are only partially redundant in T cells.

    No full text
    <p>(A) Purified T cells were cultured in vitro with the indicated amount of anti-CD3 in the absence (left panel) or presence (right panel) of 2 µg/ml of anti-CD28. After 48 hr, cells were pulsed with <sup>3</sup>H-thymidine and harvested 18 hr later. The panel represents the average of four independent experiments and the error bars are the SEM. ***<i>p</i><0.005. (B) Expression of p100 and p52 in purified T cells was detected by immunoblotting. For each sample densitometry of p100 and p52 was performed and the results expressed as the ratio between each protein and β-actin. Expression of NIK (C) and c-IAP1/2 (D) in purified T cells was detected by immunoblotting. Values in C represent the ratio between NIK and β-actin. (E) p100/52 immunoblot of T cells freshly purified or stimulated for 24 hr with 1 µg/ml anti-CD3±2 µg/ml anti-CD28. (F) T cells were stimulated as in (E) and ELISA was performed on supernatants collected after 24 hr. One of two independent experiments is shown and the error bars are the SD of the duplicates. *<i>p</i><0.05.</p

    c-IAP2 compensates for c-IAP1<sup>H582A</sup> in non-canonical NF-κB inhibitory- and TNFR1 complexes.

    No full text
    <p>(A) Expression of p100 and p52 in MEFs was detected by immunoblotting. (B and F) MEFs were untreated (−) or transfected with non-targeting control (ctr) or c-IAP2 siRNA (c2) for 24 hr and lysed. p52 and c-IAP1/2 levels were determined by immunoblotting. IκBα expression was analyzed by immunoblotting of lysates of MEFs treated for the indicated times with 1 ng/ml of TNFα (C) or 15′ with the indicated doses of TNFα (D). (F) MEFs were untreated (-) or transfected with non-targeting control (ctr) or c-IAP2 siRNA for 24 hr, then treated with 1 ng/ml TNFα for 15′ and lysed. IκBα expression was analyzed by immunoblotting. Lanes were rearranged for clarity.</p

    Suppression of Dendritic Cell-Derived IL-12 by Endogenous Glucocorticoids Is Protective in LPS-Induced Sepsis

    No full text
    <div><p>Sepsis, an exaggerated systemic inflammatory response, remains a major medical challenge. Both hyperinflammation and immunosuppression are implicated as causes of morbidity and mortality. Dendritic cell (DC) loss has been observed in septic patients and in experimental sepsis models, but the role of DCs in sepsis, and the mechanisms and significance of DC loss, are poorly understood. Here, we report that mice with selective deletion of the glucocorticoid receptor (GR) in DCs (GR<sup>CD11c-cre</sup>) were highly susceptible to LPS-induced septic shock, evidenced by elevated inflammatory cytokine production, hypothermia, and mortality. Neutralizing anti-IL-12 antibodies prevented hypothermia and death, demonstrating that endogenous GC-mediated suppression of IL-12 is protective. In LPS-challenged GR<sup>CD11c-cre</sup> mice, CD8<sup>+</sup> DCs were identified as the major source of prolonged IL-12 production, which correlated with elevations of NK cell-derived IFN-γ. In addition, the loss of GR in CD11c<sup>+</sup> cells rescued LPS-induced loss of CD8<sup>+</sup> DCs but not other DC subsets. Unlike wild-type animals, exposure of GR<sup>CD11c-cre</sup> mice to low-dose LPS did not induce CD8<sup>+</sup> DC loss or tolerance to subsequent challenge with high dose, but neutralization of IL-12 restored the ability of low-dose LPS to tolerize. Therefore, endogenous glucocorticoids blunt LPS-induced inflammation and promote tolerance by suppressing DC IL-12 production.</p></div

    Loss of endotoxin tolerance in GR<sup>CD11c-cre</sup> mice.

    No full text
    <p>(A) Tolerizing dose of LPS-induced loss of CD8<sup>+</sup> DCs in WT but not GR<sup>CD11c-cre</sup> mice. Mice were treated with 0.5 μg/g body weight of LPS for 24 hr and rechallenged with 3 μg/g body weight of LPS for 3 hr before splenic DCs were analyzed by flow cytometry. The experiment was repeated three times, with three mice in each group per experiment. (B) High-dose LPS induced IL-12 production by DCs in GR<sup>CD11c-cre</sup> mice despite prior exposure to low-dose LPS. Ten to twelve wk female GR<sup>CD11c-cre</sup> mice (<i>n</i> = 4) and control mice (<i>n</i> = 5) were treated with 0.5 μg/g body weight of LPS for 24 hr and rechallenged with 10 μg/g body weight of LPS for 6 hr. Percentage of IL-12<sup>+</sup> DCs in the spleen of a representative mouse is shown (left panel). Average numbers of total IL-12<sup>+</sup> DCs were calculated based on the total number of splenocytes in each mouse (right panel). (C) Body temperature of mice rechallenged with 10 μg/g body weight of LPS 24 hr after tolerization. Results were pooled from two independent experiments, <i>n</i> = 8 for WT and <i>n</i> = 5 for GR<sup>CD11c-cre</sup>. (D) Serum levels of TNF-α, IL-1β, IFN-α, and IFN-γ from mice in (B) were measured 6 hr after rechallenge. (E) Body temperature of mice rechallenged with 10 μg/g body weight LPS 24 hr after tolerization and 1 hr after injection with 250 μg anti-IL-12 or isotype control. Results were pooled from two independent experiments with one to two mice per condition, respectively, <i>n</i> = 3 per condition. The asterisk indicates the time by which all GR<sup>CD11c-cre</sup> mice had died. All WT and the GR<sup>CD11c-cre</sup> animals treated with anti-IL-12 survived. The data used to make this figure can be found in <a href="http://www.plosbiology.org/article/info:doi/10.1371/journal.pbio.1002269#pbio.1002269.s001" target="_blank">S1 Data</a>.</p

    Elevated proinflammatory cytokines in GR<sup>CD11c-cre</sup> mice.

    No full text
    <p>(A–C, E, and F) Kinetics of LPS-induced cytokine levels in WT and GR<sup>CD11c-cre</sup> mice. The sera used in <a href="http://www.plosbiology.org/article/info:doi/10.1371/journal.pbio.1002269#pbio.1002269.g001" target="_blank">Fig 1C</a> were also used to determine cytokine concentrations at each time point. Data shown are from four to eight animals per strain per time point pooled from multiple independent experiments. (D) Semiquantitative RT-PCR analysis of IFN-α4 transcription in DC subsets from WT and GR<sup>CD11c-cre</sup> mice 1 hr after LPS challenge. β-actin was used as internal control. Results shown are one representative of two independent experiments with pooled splenocytes from two WT and two GR<sup>CD11c-cre</sup> mice. (G) Splenocytes were prepared from mice 12 hr after LPS injection, and cultured in vitro for 4 hr with Brefeldin A. Intracellular IFN-γ was measured for gated NK and T cells by flow cytometry. One mouse from each genotype is shown out of two independent experiments, each using three mice per group. (H) Mean fluorescence intensity (MFI) and percentage of IFN-γ<sup>+</sup> cells in WT and GR<sup>CD11c-cre</sup> mice 12 hr post LPS challenge (<i>n</i> = 3). (I) GR<sup>CD11c-cre</sup> or WT mice, each represented by an individual symbol, were injected with the indicated doses of neutralizing anti-IL-12 antibody 1 hr before challenge with a lethal dose of LPS (10 μg/g body weight). Surface body temperature at 30 hr is plotted. Results shown are pooled from two independent experiments, each with three to four mice per group. The data used to make this figure can be found in <a href="http://www.plosbiology.org/article/info:doi/10.1371/journal.pbio.1002269#pbio.1002269.s001" target="_blank">S1 Data</a>.</p

    GC suppresses in vivo IL-12 production by CD8<sup>+</sup> DCs.

    No full text
    <p>(A) Six hr after LPS challenge, WT and GR<sup>CD11c-cre</sup> splenocytes were analyzed for intracellular IL-12 levels. The right panels are histograms of CD8 staining of CD11c<sup>+</sup> IL-12<sup>+</sup> cells. (B) Intracellular IL-12 levels in CD8<sup>−</sup> and CD8<sup>+</sup> DCs between WT (shaded) and GR<sup>CD11c-cre</sup> (solid line). One representative pair of a group of three is shown. (C) GC suppression of IL-12 production by CD8<sup>+</sup> DCs ex vivo. Splenocytes were prepared from mice 3 hr after LPS injection and cultured with Brefeldin A in the presence or absence of corticosterone for 4 hr. Intracellular IL-12 levels on gated CD8<sup>+</sup> DCs from one experiment are shown. The data are representative of three independent pairs of mice.</p

    LPS-induced loss of CD8<sup>+</sup> DCs is GR-dependent.

    No full text
    <p>(A–C) Splenic DCs from WT and GR<sup>CD11c-cre</sup> B10.A mice were analyzed by flow cytometry at indicated times after injection with LPS (3 μg/g body weight). Only B220<sup>−</sup>TCRβ<sup>−</sup> cells are shown. Results shown are from one representative experiment of three independent ones, each with three to five mice per group (D) Percentage of CD8<sup>+</sup> DCs in WT and GR<sup>CD11c-cre</sup> mice at time 0 (steady state). Results shown are from three pooled independent experiments, with 3–4 mice per group. (E) Total number of CD8<sup>+</sup> and CD8<sup>−</sup> DCs at time 0, 6 hr, and 24 hr post LPS injection. Results shown are from one representative experiment of three independent ones, each with three to five mice per group. (F) Sensitivity of DC subsets to GC-induced death in vitro. Total splenocytes from WT and GR<sup>CD11c-cre</sup> mice were incubated with 100 nM corticosterone in vitro for 6 hr, and the percentage of dead cells in indicated populations are shown. The data shown are representative of four pairs of mice in two independent experiments. The data used to make this figure can be found in <a href="http://www.plosbiology.org/article/info:doi/10.1371/journal.pbio.1002269#pbio.1002269.s001" target="_blank">S1 Data</a>.</p

    GR<sup>CD11c-cre</sup> mice are more susceptible to LPS-induced septic shock than WT.

    No full text
    <p>(A) and (B) Immunoblot for GR in sorted DCs and other immune subsets from WT and GR<sup>CD11c-cre</sup> mice. β-Actin was blotted as loading control. Some lanes were reordered for clarity, which is indicated by vertical white lines. (C) Plasma corticosterone concentrations after LPS challenge. Age- and sex-matched WT and GR<sup>CD11c-cre</sup> mice were injected with LPS (3 μg/g body weight), and blood was drawn at indicated time points. Data shown are from four to eight animals per strain per time point pooled from multiple independent experiments. (D) GR<sup>CD11c-cre</sup> mice (<i>n</i> = 4–6 per strain for each dose of LPS) were injected with the indicated amount of LPS, surface body temperature was recorded at the indicated times. * Four out of five GR<sup>CD11c-cre</sup> mice died by 24 hr after LPS challenge. The data used to make this figure can be found in <a href="http://www.plosbiology.org/article/info:doi/10.1371/journal.pbio.1002269#pbio.1002269.s001" target="_blank">S1 Data</a>.</p
    corecore