12 research outputs found

    Suppression by thimerosal of ex-vivo CD4+ T cell response to influenza vaccine and induction of apoptosis in primary memory T cells.

    Get PDF
    International audienceThimerosal is a preservative used widely in vaccine formulations to prevent bacterial and fungal contamination in multidose vials of vaccine. Thimerosal was included in the multidose non-adjuvanted pandemic 2009 H1N1 vaccine Panenza. In the context of the analysis of the ex-vivo T cell responses directed against influenza vaccine, we discovered the in vitro toxicity Panenza, due to its content in thimerosal. Because thimerosal may skew the immune response to vaccines, we investigated in detail the ex-vivo effects of thimerosal on the fate and functions of T cells in response to TCR ligation. We report that ex-vivo exposure of quiescent or TCR-activated primary human T cells to thimerosal induced a dose-dependent apoptotic cell death associated with depolarization of mitochondrial membrane, generation of reactive oxygen species, cytochrome c release from the mitochondria and caspase-3 activation. Moreover, exposure to non-toxic concentrations of thimerosal induced cell cycle arrest in G0/G1 phase of TCR-activated T cells, and inhibition of the release of proinflammatory cytokines such as IFN gamma, IL-1 beta, TNF alpha, IL-2, as well as the chemokine MCP1. No shift towards Th2 or Th17 cells was detected. Overall these results underline the proapoptotic effect of thimerosal on primary human lymphocytes at concentrations 100 times less to those contained in the multidose vaccine, and they reveal the inhibitory effect of this preservative on T-cell proliferation and functions at nanomolar concentrations

    Zika Virus Inhibits IFN-a Response by Human Plasmacytoid Dendritic Cells and Induces NS1-Dependent Triggering of CD303 (BDCA-2) Signaling

    No full text
    International audienceZika virus (ZIKV) dramatically emerged in French Polynesia and subsequently in the Americas where it has been associated with severe neurological complications in adults and newborns, respectively. Although plasmacytoid dendritic cells (pDCs) are a key sensor of viral infection and are critical for initiating an antiviral response, little is known about the impact of ZIKV infection on pDCs. Here, we investigated the susceptibility of human pDCs to infection with multiple strains of ZIKV and further investigated the impact of infection on pDCs functions. We observed that pDCs were refractory to cell-free ZIKV virions but were effectively infected when co-cultured with ZIKV-infected cells. However, exposure of pDCs to ZIKV-infected cells resulted in limited maturation/activation with significant down regulation of CD303 expression, a severe impairment of inflammatory cytokine production, and an inability to mount an IFN-a response. We show that ZIKV developed a strategy to inhibit the IFN-a response in primary human pDCs likely mediated through NS1-dependent CD303 signaling, thus suggesting a new mechanism of immune evasion

    Dose-dependent Toxicity of Panenza on Memory T cells specific for influenza vaccine.

    No full text
    <p>A- PBMC from patients vaccinated with the seasonal flu vaccine (Mutagrip) and the pandemic 2009 H1N1 vaccine (Pandemrix) were collected 21 days after vaccine administration. Cells (5×10<sup>5</sup> per well) were stimulated for 5 days with Mutagrip or the nonadjuvanted pandemic 2009 H1N1 Panenza vaccine at the indicated concentrations (vaccine concentration is expressed as the final concentration of HA). Cells were also stimulated with CMV or EBV peptides at 0.25 μg/ml, tetanus toxoid (TT) or tuberculin PPD at 5 μg/ml. Cultures were pulsed with 1 μCi per well of [3H] thymidine over the final 16 h of culture and cell proliferation expressed in cpm. All tests were done in triplicate and results show the mean values for three patients. The bars indicate the mean and standard deviation. Asterisks indicate significant <i>P-</i>values (p = 0.03) for comparison of cultures stimulated with Panenza at 0.02 to 0.5 μg/ml to cultures stimulated with 0.01 μg/ml of Panenza (ANOVA test). B- PBMC from Pandemrix vaccinated patients were incubated with soluble Panenza at indicated concentrations, or added to plates coated overnight at 4°C with indicated concentrations of Panenza. H3TdR (1μCi/well) was added at day 4 and T-cell proliferation was assessed at day 5. The bars indicate the mean and standard deviation. Asterisks indicate significant <i>P-</i>values (* <0.05, ** <0.01, ***<0.001) for comparison of cultures stimulated with coated Panenza to cultures stimulated with soluble Panenza. C- PBMC from a patient vaccinated with Mutagrip and then with Pandemrix were incubated overnight with culture medium, Mutagrip or Panenza at 0.25 μg/ml and the percentage of apoptotic cells was determined by flow cytometry combining 7-AAD staining with CD3 and CD4 membrane detection. Dot plots represent the co-expression of CD4 and 7-AAD on gated CD3<sup>+</sup> T cells. D- PBMC from a patient vaccinated with Mutagrip and Panenza were incubated overnight with either vaccine at various concentrations and the percentage of apoptotic (7-AAD<sup>+</sup>) cells within indicated subsets was determined combining 7-AAD staining with CD19, CD3, CD8, CD4, CD56, CD16 or CD14. B cells were CD3<sup>−</sup>CD19<sup>+</sup>, T cells were CD3<sup>+</sup>CD8<sup>+</sup> or CD3<sup>+</sup> CD4<sup>+</sup>, NK cells were CD3<sup>−</sup>CD56<sup>+</sup>CD16<sup>+</sup>, monocytes were CD3<sup>−</sup>CD19<sup>−</sup>CD14<sup>+</sup>.</p

    ROS induction by Thimerosal in TCR-activated T cells and anti-apoptotic effect of NAC.

    No full text
    <p>A- PBMC from a HD were stimulated overnight with anti-CD3 mAbs in the presence of thimerosal at 3 μg/ml, and NAC (2.5 μM) was added in half of the cultures. ROS production was detected with HE, ΔY<sub>m</sub> was assessed using the DiOC<sub>6</sub>(3) probe, and apoptosis was measured with 7-AAD. The dot plots show combined analysis of ΔY<sub>m</sub> and apoptosis or ΔY<sub>m</sub> and ROS production on gated CD4<sup>+</sup> T cells, in a representative experiment out of three experiments performed with three different HD. B- PBMC from a HD were either unstimulated (left panel) or stimulated overnight with anti-CD3 mAbs in the presence or absence of thimerosal at 3 μg/ml (right panel). The anti-oxidant NAC (2.5 μM) was added in some cultures. The percentages of CD4<sup>+</sup> and CD4<sup>−</sup> T cells with a concomitant drop in ΔY<sub>m</sub> and increased oxidizing ability (DIOC<sub>6</sub><sup>high</sup> 7-AAD<sup>neg</sup>) under the indicated culture conditions are shown. The bars indicate the mean and standard deviation from 3 experiments with three different donors. Asterisks indicate significant <i>P-</i>values (** <0.001, ***0.0001) for comparison of cultures stimulated with thimerosal and NAC to cultures stimulated with thimerosal only.</p

    Thimerosal induces cytochrome c release and caspase-3 activation.

    No full text
    <p>A- Confocal analysis of PBMC incubated 16 hours either in the presence of thimerosal (0.9 or 3 μg/ml), or with staurosporine (1 μg/ml) as a positive control. Cells were processed for cytochrome c staining (green) and co-stained with DAPI (blue) to detect nuclei modifications. B-C Western blot analysis of active caspase-3 expression in PBMC incubated for 16 h in medium (unstimulated) (B) or stimulated with anti-CD3 mAbs (C). The impact of indicated concentrations of thimerosal or staurosporine on active caspase-3 expression is shown, together with the influence of the broad caspase inhibitor QVD and the anti-oxidant NAC.</p

    Toxicity of thimerosal on TCR-activated T cells through alterations in mitochondrial membrane potential.

    No full text
    <p>A- Left panel: PBMC from a HD were stimulated overnight with anti-CD3/CD28 mAbs in the presence or absence of thimerosal at indicated concentrations. Cell viability was assessed using the MTT assay. Right panel: Kinetics of thimerosal effect on the viability of PBMC from a HD activated overnight with anti-CD3/CD28 mAbs, assessed using the MTT assay. Cultures were done in triplicate and these data are representative of at least three experiments performed with three different donors. The bars indicate the mean and standard deviation. Asterisks indicate significant <i>p-</i>values (** <0.05, ***0.01, ANOVA test) for comparison of cultures stimulated in the presence of thimerosal to cultures stimulated without thimerosal. B- Induction by thimerosal of apoptotic cell death and drop in the mitochondrial transmembrane potential ΔY<sub>m</sub> in unstimulated PBMC incubated overnight with the indicated concentrations of the agent. Staurosporine was used as a positive control. Apoptosis was measured with 7-AAD and variations in ΔY<sub>m</sub> were assessed using the DiOC<sub>6</sub>(3) probe. Dot plots in the upper panel show the dose-dependent apoptotic cell death induced by thimerosal in CD3<sup>+</sup>CD4<sup>+</sup> and CD3<sup>+</sup>CD4<sup>−</sup> T cells, and dot plots in the lower panel show the evolution of ΔY<sub>m</sub> in association with cell death on gated CD3<sup>+</sup>CD4<sup>+</sup> T cells. C- Evolution of ΔY<sub>m</sub> in association with cell death on gated CD4<sup>+</sup> T cells following overnight stimulation of PBMC with anti-CD3/CD28 mAbs in the presence of thimerosal or staurosporine. The four transition states (A to D) from living cells with intact ΔY<sub>m</sub> (DIOC<sub>6</sub><sup>high</sup> 7-AAD<sup>neg</sup>) to apoptotic cells (DIOC<sub>6</sub><sup>low</sup> 7-AAD<sup>high</sup>) are calculated from the dot plots and shown in pie charts. Data of one representative experiment among six are shown.</p

    Thimerosal inhibits T-cell proliferation and induces a G0/G1 cell cycle arrest.

    No full text
    <p>A- PBMC from a HD were labeled with CFSE and stimulated during 4 days with anti-CD3/CD28 mAbs in the presence or not of thimerosal. At the end of the culture, cells were co-stained with anti-CD4 mAbs and 7-AAD, and the rate of proliferation in CD4 T cells was determined (upper panel) combined with their priming for apoptosis (lower panel). B- The experiment of panel A was performed with 7 different HD and the bars indicate the mean and standard deviation. The left panel shows the percentage of total proliferating cells, the middle panel shows the proliferation of living cells and the right panel shows the proliferation of dying cells. Asterisks indicate significant <i>P-</i>values (* <0.05, ANOVA test) for comparison of cultures stimulated in the presence of thimerosal at 180 ng/ml to cultures stimulated in the absence of thimerosal. C: Cell cycle analysis of PBMC stimulated with anti-CD3/CD28 mAbs in the presence of thimerosal at three different concentrations. The pie charts show the repartition of cells in the various phases of the cell cycle. D- Curves show the mean values from 7 HD, demonstrating the accumulation of cells in G0/G1 phase and the disappearance of cells in the S phase after exposure to increasing concentrations of thimerosal. Asterisks indicate significant <i>P-</i>values (** <0.01, ***0.001) for comparison of the proportions of cells in G0/G1 or in S phase with or without thimerosal.</p

    Impact of thimerosal on cytokine and chemokine release upon TCR ligation.

    No full text
    <p>Pattern of cytokines and chemokines simultaneously quantified by multiplex bead assay arrays in supernatants from freshly isolated PBMC following 16-hour stimulation with anti-CD3/CD28 mAbs in the absence or presence of three different concentrations of thimerosal. The bars indicate the mean and standard deviation from experiments with three different donors. Asterisks indicate significant <i>P-</i>values (* <0.05, ANOVA test) for comparison with cultures without thimerosal.</p

    HMGB1/anti-HMGB1 antibodies define a molecular signature of early stages of HIV-Associated Neurocognitive Disorders (HAND)

    No full text
    Background: HIV-associated neurocognitive disorders (HAND) persist in the post-HAART era, characterized by asymptomatic neurocognitive impairment (ANI) and mild neurocognitive disorders (MND). High mobility group box 1 (HMGB1) is a non-histone chromosomal protein widely expressed in the nucleus of all eukaryotic cells, including brain cells, which acts as a potent proinflammatory cytokine when actively secreted from immune cells. Recent reports suggested that HMGB1 acts on microglial cells to promote neuroinflammation. In this study, our aim was to determine whether HMGB1 is involved in HAND, but also to identify early new markers of neurological impairment in HIV-infected patients. Methods: CSF and serum were collected from 103 HIV-1-infected patients enrolled in Neuradapt, a prospective study of the prevalence of HAND in HIV-1 infected patients at Nice University Hospital. Stored fluids were assessed for immunological, virological, and brain metabolite parameters. In addition to HIV RNA and DNA measurements, expression of T-cell surface markers of activation (CD38 and HLA-DR) was analyzed on whole blood. Concentration of 27 cytokines and chemokines was measured using multiplex bead assays on serum and CSF. Concentration of HMGB1 and anti-HMGB1 IgG autoantibodies were also measured on the same samples. Changes in cerebral metabolites N-acetyl aspartate (NAA), Choline (Cho) and creatinine (Cr) were assessed by magnetic resonance microscopy (MRS). Results: Clinical, virological and immunological characteristics were comparable between HAND (n = 30) and no HAND (n = 73) patients, except the absolute numbers of CD8+ T cells, which were higher in patients with HAND. Among the 29 molecules tested, only 4 of them were significantly upregulated in the CSF from HAND patients as compared to healthy donors i.e. HMGB1, anti-HMGB1 IgG antibodies, IP-10 and MCP1. CSF HMGB1 levels were positively correlated with HIV-1 DNA in aviremic HAND patients, suggesting a positive impact of HMGB1 on HIV reservoirs. Moreover, in contrast to NAA/Cr and Cho/NAA ratios, circulating anti-HMGB1 IgG antibody levels could discriminate patients with no HAND from patients with no HAND and a single deficit (average ROC-AUC = 0.744, p = 0.03 for viremic patients), thus enabling the identification of a very early stage of neurocognitive impairment, Conclusion: We report that brain injury in chronically HIV-infected patients on stable HAART is strongly associated with persistent CNS inflammation, which is correlated with increased levels of HMGB1 and anti-HMGB1 IgG in the CSF. Moreover, we identified circulating anti-HMGB1 IgG as a very early biomarker of neurological impairment in patients without HAND. These results might have important implication for the identification of patients who are at high risk of developing neurological disorders
    corecore