S2) In contrast,

levels of IP-10 correlated well with ea

S2). In contrast,

levels of IP-10 correlated well with ear swelling, because AZD6244 chemical structure only CTLA-4-Ig treatment in the sensitization phase – but not the challenge phase – could reduce the levels of IP-10. These data suggest that the release of IL-4, IL-1β, MIP-2 and IP-10 locally in the inflamed ear is regulated differently by CTLA-4-Ig; whereas IL-4, MIP-2 and IP-10 are suppressed when CTLA-4-Ig is present only in the sensitization phase, our data show that MIP-2 and IL-4 can also be suppressed when CTLA-4-Ig is present during the challenge phase alone. In this study we show that CTLA-4-Ig treatment suppresses hapten-induced inflammation in two skin inflammation models. The effect of CTLA-4-Ig has been shown previously in the DNFB-induced find more CHS model but not in the oxazolone-induced CHS

model. The short-term effect on ear swelling was detected in both the DNFB-induced model, where 25 mg/kg was sufficient to suppress the response completely, while in the oxazolone model 125 mg/kg was necessary to obtain the same degree of suppression. DNFB has been described previously to induce a T helper type 1 (Th1)-mediated response [16, 19], whereas oxazolone is shown to mediate a mixed phenotype characterized by both Th1 and Th2 cells [20]. The different efficacy of CTLA-4-Ig in the two models may be attributed to the notion that CTLA-4-Ig may suppress Th1 responses more efficiently than Th2 responses [16]. Furthermore, in our hands oxazolone-induced inflammation is dominated more by neutrophils than T cells (compared to the DNFB-model); thus, it is formally possible that the effect of CTLA-4-Ig is less efficient in the oxazolone model because of the considerable involvement of neutrophils. Alternatively, the need for a higher dose to suppress oxazolone-induced inflammation could also reflect the stronger overall response by oxazolone Ergoloid compared to DNFB (see Fig. 1). In addition to the short-term effect, we found that CTLA-4-Ig induces a long-lasting suppression

of inflammation in both models even in the absence of any detectable, circulating CTLA-4-Ig during the secondary response. The lack of circulating CTLA-4-Ig 3 weeks after administration is expected, as the half-life of human CTLA-4-Ig has been estimated to be 30 h in mice [13, 21]. Interestingly, sustained immune modulation by CTLA-4-Ig has also been shown in other settings, including transplantation, where short-term CTLA-4-Ig therapy led to long-term tissue- and organ-graft survival and induction of tolerance [14, 22-24]. Furthermore, it has been shown in vitro that CTLA-4-Ig induces a long-lasting hypo-responsiveness in human mixed leucocyte cultures (MLC) [12]. The precise mechanisms by which CTLA-4-Ig mediates the sustained suppression are not entirely clear.

Comments are closed.