Login / Signup

OGT suppresses S6K1-mediated macrophage inflammation and metabolic disturbance.

Yunfan YangXiruo LiHarding H LuanBichen ZhangKaisi ZhangJin Hyun NamZongyu LiMinnie FuAlexander MunkDongyan ZhangSimeng WangYuyang LiuJoão Paulo AlbuquerqueQunxiang OngRui LiQi WangMarie E RobertRachel J PerryDongjun ChungGerald I ShulmanXiaoyong Yang
Published in: Proceedings of the National Academy of Sciences of the United States of America (2020)
Enhanced inflammation is believed to contribute to overnutrition-induced metabolic disturbance. Nutrient flux has also been shown to be essential for immune cell activation. Here, we report an unexpected role of nutrient-sensing O-linked β-N-acetylglucosamine (O-GlcNAc) signaling in suppressing macrophage proinflammatory activation and preventing diet-induced metabolic dysfunction. Overnutrition stimulates an increase in O-GlcNAc signaling in macrophages. O-GlcNAc signaling is down-regulated during macrophage proinflammatory activation. Suppressing O-GlcNAc signaling by O-GlcNAc transferase (OGT) knockout enhances macrophage proinflammatory polarization, promotes adipose tissue inflammation and lipolysis, increases lipid accumulation in peripheral tissues, and exacerbates tissue-specific and whole-body insulin resistance in high-fat-diet-induced obese mice. OGT inhibits macrophage proinflammatory activation by catalyzing ribosomal protein S6 kinase beta-1 (S6K1) O-GlcNAcylation and suppressing S6K1 phosphorylation and mTORC1 signaling. These findings thus identify macrophage O-GlcNAc signaling as a homeostatic mechanism maintaining whole-body metabolism under overnutrition.
Keyphrases
  • adipose tissue
  • insulin resistance
  • signaling pathway
  • gene expression
  • high fat diet induced
  • high fat diet
  • type diabetes
  • transcription factor
  • small molecule
  • tyrosine kinase
  • binding protein