MiR-125a targets effector programs to stabilize Treg-mediated immune homeostasis

  • Wen Pan
  • Shu Zhu
  • Dai Dai
  • Zheng Liu
  • Dan Li
  • Bin Li
  • Nicola Gagliani
  • Yunjiang Zheng
  • Yuanjia Tang
  • Matthew T Weirauch
  • Xiaoting Chen
  • Wei Zhu
  • Yue Wang
  • Bo Chen
  • Youcun Qian
  • Yingxuan Chen
  • Jingyuan Fang
  • Ronald Herbst
  • Laura Richman
  • Bahija Jallal
  • John B Harley
  • Richard A Flavell
  • Yihong Yao
  • Nan Shen

Abstract

Although different autoimmune diseases show discrete clinical features, there are common molecular pathways intimately involved. Here we show that miR-125a is downregulated in peripheral CD4(+) T cells of human autoimmune diseases including systemic lupus erythematosus and Crohn's disease, and relevant autoimmune mouse models. miR-125a stabilizes both the commitment and immunoregulatory capacity of Treg cells. In miR-125a-deficient mice, the balance appears to shift from immune suppression to inflammation, and results in more severe pathogenesis of colitis and experimental autoimmune encephalomyelitis (EAE). The genome-wide target analysis reveals that miR-125a suppresses several effector T-cell factors including Stat3, Ifng and Il13. Using a chemically synthesized miR-125a analogue, we show potential to re-programme the immune homeostasis in EAE models. These findings point to miR-125a as a critical factor that controls autoimmune diseases by stabilizing Treg-mediated immune homeostasis.

Bibliografische Daten

OriginalspracheEnglisch
ISSN2041-1723
DOIs
StatusVeröffentlicht - 12.05.2015
Extern publiziertJa
PubMed 25963922