[HTML][HTML] Conserved human effector Treg cell transcriptomic and epigenetic signature in arthritic joint inflammation

G Mijnheer, L Lutter, M Mokry, M van der Wal… - Nature …, 2021 - nature.com
G Mijnheer, L Lutter, M Mokry, M van der Wal, R Scholman, V Fleskens, A Pandit, W Tao
Nature communications, 2021nature.com
Treg cells are critical regulators of immune homeostasis, and environment-driven Treg cell
differentiation into effector (e) Treg cells is crucial for optimal functioning. However, human
Treg cell programming in inflammation is unclear. Here, we combine transcriptional and
epigenetic profiling to identify a human eTreg cell signature. Inflammation-derived functional
Treg cells have a transcriptional profile characterized by upregulation of both a core Treg
cell (FOXP3, CTLA4, TIGIT) and effector program (GITR, BLIMP-1, BATF). We identify a …
Abstract
Treg cells are critical regulators of immune homeostasis, and environment-driven Treg cell differentiation into effector (e)Treg cells is crucial for optimal functioning. However, human Treg cell programming in inflammation is unclear. Here, we combine transcriptional and epigenetic profiling to identify a human eTreg cell signature. Inflammation-derived functional Treg cells have a transcriptional profile characterized by upregulation of both a core Treg cell (FOXP3, CTLA4, TIGIT) and effector program (GITR, BLIMP-1, BATF). We identify a specific human eTreg cell signature that includes the vitamin D receptor (VDR) as a predicted regulator in eTreg cell differentiation. H3K27ac/H3K4me1 occupancy indicates an altered (super-)enhancer landscape, including enrichment of the VDR and BATF binding motifs. The Treg cell profile has striking overlap with tumor-infiltrating Treg cells. Our data demonstrate that human inflammation-derived Treg cells acquire a conserved and specific eTreg cell profile guided by epigenetic changes, and fine-tuned by environment-specific adaptations.
nature.com