13 research outputs found

    Activation of huTNF promoter by HDACi.

    No full text
    <p>TNF gene expression observed in the monocytoid U937 cells transiently transfected with a plasmid expressing the luciferase gene under the control of the huTNF promoter and treated without (control) or with histone deacetylase inhibitors: trichostatin A (TSA) or sirtinol (Sirt).</p

    Effect of HDACi on cellular apoptosis and cell viability.

    No full text
    <p><b>A</b>. Apoptosis of PBMCs, PBLs, and monocytes from healthy controls after treatment with TSA and Sirt. Apoptosis was measured by annexin-V assay as reported in the Materials and Methods chapter. Flow cytometric data at 72 h treatment are shown from two independent experiments. Mean fluorescence intensity (MFI) is indicated in brackets. BC, Buffy-coats of two healthy donors; PBLs: peripheral blood lymphocytes. <b>B</b>. Measurement of PBMC viability from healthy controls after one and three days of treatment with TSA and Sirt. Cell viability of PBMCs was determined by MTT assay after 24 h and 72 h treatment with TSA (1 microM) and sirtinol (10 microM). Data shown represent mean ± SD of triplicate wells.</p

    Proteomic Profiling of the TRAF3 Interactome Network Reveals a New Role for the ER-to-Golgi Transport Compartments in Innate Immunity

    Get PDF
    <div><p>Tumor Necrosis Factor receptor-associated factor-3 (TRAF3) is a central mediator important for inducing type I interferon (IFN) production in response to intracellular double-stranded RNA (dsRNA). Here, we report the identification of Sec16A and p115, two proteins of the ER-to-Golgi vesicular transport system, as novel components of the TRAF3 interactome network. Notably, in non-infected cells, TRAF3 was found associated with markers of the ER-Exit-Sites (ERES), ER-to-Golgi intermediate compartment (ERGIC) and the cis-Golgi apparatus. Upon dsRNA and dsDNA sensing however, the Golgi apparatus fragmented into cytoplasmic punctated structures containing TRAF3 allowing its colocalization and interaction with Mitochondrial AntiViral Signaling (MAVS), the essential mitochondria-bound RIG-I-like Helicase (RLH) adaptor. In contrast, retention of TRAF3 at the ER-to-Golgi vesicular transport system blunted the ability of TRAF3 to interact with MAVS upon viral infection and consequently decreased type I IFN response. Moreover, depletion of Sec16A and p115 led to a drastic disorganization of the Golgi paralleled by the relocalization of TRAF3, which under these conditions was unable to associate with MAVS. Consequently, upon dsRNA and dsDNA sensing, ablation of Sec16A and p115 was found to inhibit IRF3 activation and anti-viral gene expression. Reciprocally, mild overexpression of Sec16A or p115 in Hec1B cells increased the activation of IFNβ, ISG56 and NF-κB -dependent promoters following viral infection and ectopic expression of MAVS and Tank-binding kinase-1 (TBK1). In line with these results, TRAF3 was found enriched in immunocomplexes composed of p115, Sec16A and TBK1 upon infection. Hence, we propose a model where dsDNA and dsRNA sensing induces the formation of membrane-bound compartments originating from the Golgi, which mediate the dynamic association of TRAF3 with MAVS leading to an optimal induction of innate immune responses.</p> </div

    Enforced retention of TRAF3 at the ER-to-Golgi compartment negatively regulates type I IFN response.

    No full text
    <p>(<b>A</b>) Confocal microscopy analysis of FLAG-tag and GM130 performed in HeLa cells expressing FLAG-TRAF3 (panels 1 and 2) or FLAG-TRAF3-AKKFF (panels 3 and 4) upon no infection (panels 1 and 3) or SeV infection (200 HAU/ml) (panels 2 and 4) for 4 h. The nuclei were stained with DAPI. Arrows indicate TRAF3 aggregates. One of two independent experiments with similar results is shown. Bars represent 10 µm. (<b>B</b>) Hec1B cells were co-transfected with luciferase reporter plasmid pGL3-IFNβ (250 ng) and indicated plasmids (250 ng) for 24 h and infected with SeV (200 HAU/ml) for 16 h. Hec1B cells were also co-transfected with luciferase reporter plasmid pGL3-IFNβ (250 ng), empty vector or MAVS or TRIF (25 ng) along with indicated plasmids (250 ng) for 24 h. Relative luciferase activity was measured as described in <a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1002747#s4" target="_blank">Materials and Methods</a>. Mean values +/− S.D. of triplicate determinations are shown (*** P<0.001). One of four independent experiments with similar results is shown. Cellular extracts from transfected Hec1B cells were also prepared and subjected to immunoblot analysis using indicated antibodies (right lower panel).</p

    Through its ability to interact and colocalize with components of the ERES (Sec16A, depicted as thick green lines), ERGIC (ERGIC53 and p115) and the cis-Golgi apparatus (p115 and GM130), a subpopulation of TRAF3 (red circles) resides in the ER-to-Golgi vesicular compartment in non-infected cells.

    No full text
    <p>Upon dsRNA and dsDNA sensing, the cis-Golgi disorganizes into punctate structures, giving rise to membrane-bound compartments composed of at least GM130 and TRAF3 (dashed line). We propose that these membrane-bound compartments allow the proper positioning of TRAF3 with MAVS at Mitochondrial-Associated endoplasmic reticulum Membranes (MAM) <a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1002747#ppat.1002747-Horner1" target="_blank">[86]</a>. There, being in close proximity with a component of the exocyst (sec5) and the translocon (Sec61β), TRAF3 allows the activation of TBK1 and IRF3 leading to activation of the type I IFN response. A similar scenario was recently proposed for STING (yellow circles) where in response to DNA virus infection, it traffics from the ER to the cis-Golgi apparatus and finally to a distinct perinuclear region for the activation of TBK1 <a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1002747#ppat.1002747-Ishikawa2" target="_blank">[61]</a>, <a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1002747#ppat.1002747-Saitoh1" target="_blank">[62]</a>. MTOC: microtubule-organizing center.</p

    Sec16A and p115 are required for the proper positioning of TRAF3 along the mitochondrial network.

    No full text
    <p>(<b>A</b>) Confocal microscopy of HeLa cells transfected with 40 nM nonsilencing RNA duplexes (panels 1 and 2) or 40 nM siRNA duplexes that specifically target Sec16A (panels 3 and 4) or p115 (panels 5 and 6) and stained for MAVS and endogenous TRAF3 upon no treatment (panels 1, 3 and 5) or SeV infection (200 HAU/ml) for 4 h (panels 2, 4 and 6). Arrows indicate the colocalization of TRAF3 with MAVS. Bars represent 5 µm. One of three independent experiments with similar results is shown. (<b>B</b>) p115 and Sec16A were silenced in HeLa cells as described in (<b>A</b>) and infected with SeV for indicated periods of time. Whole-cell lysates were subjected to immunoprecipitation using an anti-TRAF3 (H-20) antibody followed by immunoblotting for the presence of MAVS and TRAF3. Immunoblot analysis against p115, Sec16A, TRAF3 and SeV proteins are also shown (Input). One of two independent experiments with similar results is shown. (<b>C</b>) Densitometric analysis of the binding activity of MAVS to TRAF3 presented in <a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1002747#ppat-1002747-g006" target="_blank">Figures 6B</a>. Data represent the ratio of immunoprecipitated MAVS over immunoprecipitated TRAF3 and are means +/− S.D. of two experiments.</p

    TRAF3 localizes to the ER-to-Golgi transport compartments and behaves like a cis-Golgi protein.

    No full text
    <p>(<b>A</b>) Confocal microscopy performed in HeLa cells on FLAG-TRAF3 and Myc-p115 (panel 1), FLAG-p115 and GM130 (panel 2), FLAG-TRAF3 and GM130 (panel 3), FLAG-p115 and ERGIC53 (panel 4), FLAG-TRAF3 and ERGIC53 (panel 5), FLAG-TRAF3 and Calnexin (panel 6), FLAG-TRAF3 and lysotracker (panel 7) or mitotracker (panel 8). The nuclei were stained utilizing DAPI. One of three independent experiments with similar results is shown. Bars represent 10 µm. (<b>B</b>) Confocal microscopy performed in HeLa cells on EGFP-Sec16A and calnexin (panel 1), EGFP-Sec16A and FLAG-p115 (panel 2), EGFP-Sec16A and FLAG-TRAF3 (panel 3) and FLAG-TRAF3 and endogenous Sec16A (panel 4). (<b>C</b>) HeLa cells were stained for endogenous TRAF3 and GM130 (panel 1) or endogenous TRAF3 and ERGIC53 (panel 2) before analysis via confocal microscopy. The nuclei were stained utilizing DAPI. Bars represent 5 µm. One of three independent experiments with similar results is shown.</p

    p115 and Sec16A associate with TRAF3 following cytosolic RNA and DNA sensor activation.

    No full text
    <p>(<b>A</b>) Whole-cell lysates (HeLa cells) were prepared and subjected to immunoprecipitation assays using TRAF3 (H-20) or isotype control antibodies followed by immunoblotting for the presence of p115, Sec16A, and TRAF3. (<b>B</b>) HeLa cells were treated as indicated for different periods of time. Whole-cell lysates were prepared and subjected to immunoprecipitation assays using TRAF3 (H-20) antibody followed by immunoblotting for the presence of p115, Sec16A, TBK1 and TRAF3. The Native-PAGE assay was conducted on the same cellular extracts to demonstrate the dimerization and activation of IRF-3 upon indicated treatments. One of three independent experiments with similar results is shown.</p

    Sec16A and p115 influence the type I IFN antiviral response at the transcriptional level.

    No full text
    <p>(<b>A–C</b>) Hec1B cells were co-transfected with the indicated luciferase reporter genes together with 125 ng of empty vector, Myc-p115 or EGFP-Sec16A. Data are expressed as fold-induction following SeV infection (16 h) over the corresponding non-infected condition. (<b>D–F</b>) Hec1B cells were co-transfected with pGL3-IFNβ-luciferase reporter gene together with 125 ng of empty vector, Myc-p115 or Sec16A and 15 ng of FLAG-MAVS (<b>D</b>), 100 ng of FLAG-TBK1 (<b>E</b>), or 15 ng of His-TRIF (<b>F</b>). Data represent the fold-activation over the corresponding vector control. Each value represents the mean +/− S.D. of triplicate determinations. The data are representative of at least four different experiments with similar results. (<b>G</b>) TRAF3 knockout MEF cells were co-transfected with 250 ng of luciferase reporter plasmid pGL3-IFNβ, 500 ng of pcDNA3 or FLAG-TRAF3 plasmid and 375 ng of indicated plasmids. At 24 h post-transfection, cells were left uninfected or infected with SeV (200 HAU/ml) for 16 h and relative luciferase activity was measured as described in <a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1002747#s4" target="_blank">Materials and Methods</a>. Mean values +/− S.D. of triplicate determinations are shown (** P<0.01). One of three independent experiments with similar results is shown.</p

    Requirement of Sec16A and p115 for optimal type I IFN innate immune response in cells exposed to cytosolic DNA and RNA sensor ligands.

    No full text
    <p>(<b>A–D</b>) HeLa cells were transfected with nonsilencing (Ns) RNA duplexes or two different sets of siRNA duplexes that specifically target p115 or Sec16A as indicated. 72 h post-transfection, cells were left untreated (Ctl) or stimulated with poly I:C (2.5 µg/ml), poly dA:dT (1 µg/ml) or SeV (200 HAU/ml) for 6 h to 8 h. RNA was extracted and analyzed by RT-qPCR using primers for <i>ifnβ</i>, <i>ifit1</i>, <i>oas1</i>. Data are means +/− S.D. (n = 3). * Significantly below the induction response; * P<0.05, ** P<0.01, *** P<0.001. (<b>B and D</b>) Cellular extracts were also prepared and subjected to immunoblot analysis using indicated antibodies. One of three independent experiments with similar results is shown.</p
    corecore