As shown in Fig  1a, there was a clear increase in the percentage

As shown in Fig. 1a, there was a clear increase in the percentage of IFN-γ-producing

CD4+ T cells (Th1 cells) and IL-17-producing CD4+ T cells (Th17 cells) after CII restimulation compared with controls (both P < 0·05). No significant difference was observed in the percentage of Treg in SMNCs with or without CII restimulation (P > 0·05). Figure 1b shows the typical flow cytometric results of three T subsets in dot-plots. These results indicate that CII-specific reactivation tends to Th1- and Th17-type expansion. As recent evidence suggests that Notch signalling is an important modulator of T cell-mediated immune Selleckchem Adriamycin responses, we next wanted to know whether Notch signalling could be activated in the collagen-specific T cell response. To explore this, SMNCs from immunized mice MK-2206 molecular weight were restimulated by CII for 3 days and then CD4+ T cells were purified by magnetic sorting kits and assessed for increased transcript levels of Hes1 and four Notch receptors, including Notch1, Notch2, Notch3 and Notch4. Hes1 is a downstream target of Notch signalling, and an increase in transcripts of this gene indicates

active Notch signalling in cells. As shown in Fig. 1c, CII restimulation induced up-regulated transcript levels of Hes1 in CII-reactive CD4+ T cells. The mRNA level of Notch3 was also up-regulated significantly, while the levels of the other three Notch receptors were not increased. These data indicate the participation of Notch signalling and the potential role of Notch3 receptor in CII-specific Th1- and Th17-type expansion. Based on the above data, we next used the γ-secretase inhibitor DAPT, which prevents

activation of all Notch receptors by Rucaparib molecular weight inhibiting the final enzymatic cleavage and specific neutralizing antibody to Notch3 to determine the effect of Notch signalling inhibition on collagen-specific T cell responses. Data in Fig. 2a indicate that both DAPT (5 µM) and α-Notch3 (10 µg/ml) could induce suppression for CII-initiated lymphoproliferation well, as expected. As shown in Fig. 2b, addition of DAPT reduced the percentage of Th1 and Th17 cells in SMNCs co-cultured with CII. Similar results were obtained when SMNCs were incubated with CII and α-Notch3. Neither DAPT nor α-Notch3 changed the percentage of Treg cells. No significant difference of suppression effect between DAPT and α-Notch3 was observed. These results demonstrate that activation of Notch signalling through Notch3 receptor mediates collagen-specific Th1- and Th17-type expansion. Notch signalling is initiated by ligand–receptor interaction between neighbouring cells. We next used Delta-like 1-Fc and Jagged1-Fc fusion proteins that bind to Notch receptors and activate the Notch pathway to explore the effect of Notch ligands on this expansion. As depicted in Fig. 3a and b, the addition of Delta-like 1-Fc fusion protein increased the percentage of Th1 and Th17 cells while Jagged1-Fc fusion protein did not change the percentage of Th1 and Th17 cells significantly.

Comments are closed.