Epigenetic conversion of conventional T cells into regulatory T cells by CD28 signal deprivation

N Mikami, R Kawakami, KY Chen… - Proceedings of the …, 2020 - National Acad Sciences
N Mikami, R Kawakami, KY Chen, A Sugimoto, N Ohkura, S Sakaguchi
Proceedings of the National Academy of Sciences, 2020National Acad Sciences
Foxp3-expressing regulatory T cells (Tregs) can be generated in vitro by antigenic
stimulation of conventional T cells (Tconvs) in the presence of TGF-β and IL-2. However,
unlike Foxp3+ naturally occurring Tregs, such in vitro induced Tregs (iTregs) are functionally
unstable mainly because of incomplete Treg-type epigenetic changes at Treg signature
genes such as Foxp3. Here we show that deprivation of CD28 costimulatory signal at an
early stage of iTreg generation is able to establish Treg-specific DNA hypomethylation at …
Foxp3-expressing regulatory T cells (Tregs) can be generated in vitro by antigenic stimulation of conventional T cells (Tconvs) in the presence of TGF-β and IL-2. However, unlike Foxp3+ naturally occurring Tregs, such in vitro induced Tregs (iTregs) are functionally unstable mainly because of incomplete Treg-type epigenetic changes at Treg signature genes such as Foxp3. Here we show that deprivation of CD28 costimulatory signal at an early stage of iTreg generation is able to establish Treg-specific DNA hypomethylation at Treg signature genes. It was achieved, for example, by TCR/TGF-β/IL-2 stimulation of CD28-deficient Tconvs or CD28-intact Tconvs without anti-CD28 agonistic mAb or with CD80/CD86-blocked or -deficient antigen-presenting cells. The signal abrogation could induce Treg-type hypomethylation in memory/effector as well as naive Tconvs, while hindering Tconv differentiation into effector T cells. Among various cytokines and signal activators/inhibitors, TNF-α and PKC agonists inhibited the hypomethylation. Furthermore, CD28 signal deprivation significantly reduced c-Rel expression in iTregs; and the specific genomic perturbation of a NF-κB binding motif at the Foxp3 CNS2 locus enhanced the locus-specific DNA hypomethylation even in CD28 signaling-intact iTregs. In addition, in vitro maintenance of such epigenome-installed iTregs with IL-2 alone, without additional TGF-β or antigenic stimulation, enabled their expansion and stabilization of Treg-specific DNA hypomethylation. These iTregs indeed stably expressed Foxp3 after in vivo transfer and effectively suppressed antigen-specific immune responses. Taken together, inhibition of the CD28-PKC-NF-κB signaling pathway in iTreg generation enables de novo acquisition of Treg-specific DNA hypomethylation at Treg signature genes and abundant production of functionally stable antigen-specific iTregs for therapeutic purposes.
National Acad Sciences