An intrinsic mechanism predisposes Foxp3-expressing regulatory T cells to Th2 conversion in vivo

Y Wang, A Souabni, RA Flavell… - The Journal of …, 2010 - journals.aai.org
Y Wang, A Souabni, RA Flavell, YY Wan
The Journal of Immunology, 2010journals.aai.org
Naturally occurring regulatory T (nTreg) cells express Foxp3 and were originally discovered
as immune suppressors critical for self-tolerance and immune homeostasis. Through yet-to-
be-defined mechanisms, nTreg cells were recently shown to convert into proinflammatory
cells. Particularly, attenuation of Foxp3 expression led to Th2 conversion of nTreg cells in
vivo. In this paper, we demonstrated an nTreg-specific mechanism controlling their Th2
conversion. We found that wild-type nTreg cells expressing reduced levels of Foxp3 but not …
Abstract
Naturally occurring regulatory T (nTreg) cells express Foxp3 and were originally discovered as immune suppressors critical for self-tolerance and immune homeostasis. Through yet-to-be-defined mechanisms, nTreg cells were recently shown to convert into proinflammatory cells. Particularly, attenuation of Foxp3 expression led to Th2 conversion of nTreg cells in vivo. In this paper, we demonstrated an nTreg-specific mechanism controlling their Th2 conversion. We found that wild-type nTreg cells expressing reduced levels of Foxp3 but not those expressing no Foxp3 produced the Th2 cytokine IL-4. Intriguingly, IL-4 production by converted nTreg cells is required for Th2 differentiation of coexisting naive CD4 T cells in vivo, suggesting that Th2 conversion of nTreg cells might be critical for directing Th2 immune responses. Th2 conversion of nTreg cells was not due to their inability to become Th1 cells, because IFN-γ was produced by Foxp3-low–expressing cells when IL-4/STAT-6 signaling was abrogated. Surprisingly, however, unlike naive CD4 T cells whose IL-4 production is dependent on STAT-6, Foxp3-low–expressing cells generated IL-4 independent of STAT-6, indicating an intrinsic mechanism that favors nTreg-to-Th2 differentiation. Indeed, compared with naive CD4 T cells, nTreg expressed elevated levels of GATA-3 independent of STAT-6. And GATA-3 was required for nTreg-to-Th2 conversion. Foxp3 may account for this GATA-3 upregulation in nTreg cells, because ectopic expression of Foxp3 preferentially promoted GATA-3 but not T-bet expression. Thus, we have identified an intrinsic mechanism that imposes a Th2/Th1 imbalance and predisposes Foxp3-expressing cells to IL-4 production independent of STAT-6 signaling.
journals.aai.org