26 research outputs found

    Kinase suppressor of Ras 1 is not required for the generation of regulatory and memory T cells

    Get PDF
    The mammalian target of rapamycin (mTOR) kinase is a critical regulator of the differentiation of helper and regulatory CD4+ T cells, as well as memory CD8+ T cells. In this study, we investigated the role of the ERK signaling pathway in regulating mTOR activation in T cells. We showed that activation of ERK following TCR engagement is required for sustained mTOR complex 1 (mTORC1) activation. Absence of kinase suppressor of Ras 1 (KSR1), a scaffold protein of the ERK signaling pathway, or inhibition of ERK resulted in decreased mTORC1 activity following T cell activation. However, KSR1-deficient mice displayed normal regulatory CD4+ T cell development, as well as normal memory CD8+ T cell responses to LCMV and Listeria monocytogenes infection. These data indicate that despite its role in mTORC1 activation, KSR1 is not required in vivo for mTOR-dependent T cell differentiation

    Normal effector and memory CD8+ T cell response to Listeria in KSR1-deficient mice.

    No full text
    <p>WT and KSR1-/- mice were infected with 2×10<sup>3</sup> cfu of Lm-OVA, and presence of OVA epitope specific CD8+ T cells in the spleen was analyzed by flow cytometry 7 (A, B) and 30 (C, D, G, H) days after infection. 30 days after Lm-OVA infection, mice were challenged with 10<sup>5</sup> cfu of Lm-OVA and presence of OVA epitope specific CD8+ T cells in the spleen was assessed 5 days later (E, F). A, C, E: Percentage of Db-GP33 Tetramer positive cells in CD8+ T cells. Plots are representative of two experiments with 3–8 mice per group. B, D, F: Number of Db-GP33 Tetramer+ CD8+ T cells per spleen. Dashed line represents average numbers in naive mice and mice infected with Lm. G, H: CD62L and CD127 expression in Db-GP33 Tetramer+ CD8+ T splenocytes 30 days after Lm-OVA infection. Data is presented as mean + SEM and is representative of two experiments with 3–8 mice per group. There were no significant differences between WT and KO mice (t-test).</p

    Normal effector and memory CD8+ T cell response to LCMV in KSR1-deficient mice.

    No full text
    <p>WT, KSR1-/-, and WT mice treated with rapamycin were infected with 10<sup>5</sup> pfu of LCMV-Armstrong, and presence of GP33 epitope specific CD8+ T cells was analyzed by flow cytometry 7 (A, B) and 30 (C–H) days after infection in the blood (A–D) and spleen (E–H). A, C, E: Percentage of Db-GP33 Tetramer positive cells in CD8+ T cells. Plots are representative of two experiments with 5 mice per group. B, D: Percentage of Db-GP33 Tetramer+ CD8+ T cells in peripheral blood mononuclear cells. F: Number of Db-GP33 Tetramer+ CD8+ T cells per spleen 30 days after infection. G, H: CD62L and CD127 expression in Db-GP33 Tetramer+ CD8+ T splenocytes 30 days after infection. Data is presented as mean + SEM and is representative of two experiments with 5 mice per group. *: p<0.05, t-test.</p

    MEK inhibition decreases mTOR activity in T cells.

    No full text
    <p>Splenocytes from B6 mice were pretreated or not for one hour with 10 µM UO126 and cultured for 2 to 48 hrs with or without anti-CD3 + anti-CD28 stimulation (5 µg/mL each). Phosphorylation of ERK (A, B), T421/S424-S6K (C, D) and T389-S6K (E, F) in CD8+ T cells was measured by flow cytometry. A, C, E: Representative histograms of unstimulated cells (filled histogram), cells stimulated with CD3+CD28 and treated (red line) or not (black line) with UO126, gated on CD8+ T cells. Graphs are representative of 3 to 5 experiments. C, D, F: Fold increase phosphorylation of ERK, T421/S424-S6K and T389-S6K in anti-CD3 + anti-CD28 stimulated CD8+ T cells treated (red) or not (black) with UO126, compared to unstimulated cells. Each dot represents an independent experiment. p-ERK, p-T421/S424-S6K and p-T389-S6K levels were normalized within each experiment to unstimulated control T cells. *: p<0.05, paired t-test.</p

    Regulatory CD4+ T cell development is normal in KSR1-deficient T cells.

    No full text
    <p>Thymocytes (A, B) and splenocytes (C, D) of WT (white bars) and KSR1-/- (black bars) mice were analyzed for the presence of regulatory CD4+ T cells by flow cytometry. A, C: Percentage of FoxP3+ CD25+ cells in CD4+ CD8- single positive (SP) thymocytes (A) and CD4+ CD3+ T cells (B). B, D: Numbers of FoxP3+ CD25+ CD4 SP by thymuses (B) and CD4+ T cells by spleens (D). Data is presented as mean + SEM and represent 10-12 mice per group. There were no significant differences between WT and KO mice (t-test).</p

    mTOR activity is decreased in KSR1-deficient T cells.

    No full text
    <p>Splenocytes from WT or KSR1-/- mice were cultured with or without anti-CD3 + anti-CD28 (5 µg/mL each) stimulation for 2 to 48 hrs. Phosphorylation of ERK (A, B), T421/S424-S6K (C, D) and T389-S6K (E, F), and expression of CD127 (G,H) in CD8+ T cells was measured by flow cytometry. A, C, E: Representative histograms of unstimulated WT cells (filled grey histogram), unstimulated KO cells (blue line), and WT (black line) or KO (red line) cells stimulated with CD3+CD28, gated on CD8+ T cells. Graphs are representative of 3 to 4 experiments. C, D, F: Fold increase phosphorylation of ERK, T421/S424-S6K and T389-S6K in stimulated WT (black) and KO (red) CD8+ T cells compared to non-stimulated cells. Each dot represents an independent experiment. p-ERK, p-T421/S424-S6K and p-T389-S6K levels were normalized within each experiment to unstimulated control T cells. *: p<0.05, paired t-test. G-H. Representative histograms (G) and fold increase in comparison to non-stimulated control T cells (H) of CD127 expression in WT (black) and KO (red) cells stimulated with CD3+CD28, gated on CD8+ T cells. Grey line: isotype control. H. Data is presented as mean + SEM and is representative of two independent experiments. *: p<0.05, unpaired t-test.</p

    Growth hormone enhances thymic function in HIV-1–infected adults

    No full text
    Growth hormone (GH) is an underappreciated but important regulator of T cell development that can reverse age-related declines in thymopoiesis in rodents. Here, we report findings of a prospective randomized study examining the effects of GH on the immune system of HIV-1–infected adults. GH treatment was associated with increased thymic mass. In addition, GH treatment enhanced thymic output, as measured by both the frequency of T cell receptor rearrangement excision circles in circulating T cells and the numbers of circulating naive and total CD4+ T cells. These findings provide compelling evidence that GH induces de novo T cell production and may, accordingly, facilitate CD4+ T cell recovery in HIV-1–infected adults. Further, these randomized, prospective data have shown that thymic involution can be pharmacologically reversed in humans, suggesting that immune-based therapies could be used to enhance thymopoiesis in immunodeficient individuals
    corecore