5A) In contrast, addition of CD4+CD25+ cells had no significant

5A). In contrast, addition of CD4+CD25+ cells had no significant effect on the ability of lpr DC to induce IFN-γ production by hapten-specific WT CD8+ T cells under the same culture conditions (Fig. 5B). Thus, CD4+CD25+ cells inhibited the activation of effector CD8+ T cells indirectly

through effects on Fas-expressing hapten-presenting DC. To test the FasL-dependent regulatory activity of CD4+CD25+ cells in vivo, naïve mice were primed by intradermal injection of DC from sensitized WT or lpr mice. The development of hapten-specific IFN-γ producing CD8+ T cells was markedly increased in mice primed by WT DC and treated with anti-CD25 mAb when compared with control mice treated with rat IgG (Fig. 5C, *p<0.05). In contrast, anti-CD25 mAb treatment of mice primed by Fas-defective Bcr-Abl inhibitor DC did not increase the development of hapten-specific CD8+ T cells when compared with the control group (Fig. 5C). Collectively, these results indicated that the priming activity of hapten-presenting

DC expressing functional Fas is restricted during induction of CHS response by CD4+CD25+ regulatory T cells, while the priming activity of Fas-defective DC is not. The data presented to this point suggest a model in which hapten application to the skin induces the emigration of DC from the skin to the draining LN where the hapten-presenting DC express Fas and subsequently activate and/or engage CD4+CD25+FasL+ T cells that mediate apoptosis of the DC, limiting the duration and magnitude Selleckchem Ruxolitinib of hapten-reactive CD8 T-cell priming. This model predicts that at times when this CD4+CD25+ T regulatory cell activity is in operation to mediate apoptosis of the hapten-presenting DC, the active Osimertinib CD4+CD25+ T cells may also mediate the apoptosis of DC presenting other haptens that enter the skin draining LN. This activity would result in decreased CD8 T-cell responses to these other haptens. Therefore, we tested if CD4+CD25+ regulatory T cells activated to suppress the CHS response to a specific hapten were also capable

of suppressing the response to subsequent sensitization with a different hapten. Mice were first sensitized with FITC to induce a FITC-specific CHS response and then sensitized with DNFB 5 days later to activate DNFB-specific CD8+ T cells. Distinct areas of the skin (on the back and on the abdomen) were sensitized with FITC or with DNFB to exclude the possibility that cutaneous DC from the sensitized skin present both haptens to the two populations of hapten-specific effector CD8+ T cells. Induction of DNFB-specific IFN-γ producing CD8+ T cells was reduced twofold in mice pre-sensitized with FITC when compared with control mice sensitized with DNFB only (Fig. 6A). This non-specific regulation was completely abrogated by treatment with anti-CD25 mAb at the time of pre-sensitization with FITC, as the numbers of DNFB-specific IFN-γ producing CD8+ T cells in anti-CD25 mAb-treated group were similar to the numbers in the control group sensitized with DNFB only (Fig.

Comments are closed.