Your browser doesn't support javascript.
loading
OGT suppresses S6K1-mediated macrophage inflammation and metabolic disturbance.
Yang, Yunfan; Li, Xiruo; Luan, Harding H; Zhang, Bichen; Zhang, Kaisi; Nam, Jin Hyun; Li, Zongyu; Fu, Minnie; Munk, Alexander; Zhang, Dongyan; Wang, Simeng; Liu, Yuyang; Albuquerque, João Paulo; Ong, Qunxiang; Li, Rui; Wang, Qi; Robert, Marie E; Perry, Rachel J; Chung, Dongjun; Shulman, Gerald I; Yang, Xiaoyong.
Affiliation
  • Yang Y; Program in Integrative Cell Signaling and Neurobiology of Metabolism, Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520.
  • Li X; Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520.
  • Luan HH; Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520.
  • Zhang B; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520.
  • Zhang K; Program in Integrative Cell Signaling and Neurobiology of Metabolism, Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520.
  • Nam JH; Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520.
  • Li Z; Program in Integrative Cell Signaling and Neurobiology of Metabolism, Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520.
  • Fu M; Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520.
  • Munk A; Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC 29425.
  • Zhang D; Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520.
  • Wang S; Program in Integrative Cell Signaling and Neurobiology of Metabolism, Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520.
  • Liu Y; Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520.
  • Albuquerque JP; Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520.
  • Ong Q; Program in Integrative Cell Signaling and Neurobiology of Metabolism, Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520.
  • Li R; Program in Integrative Cell Signaling and Neurobiology of Metabolism, Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520.
  • Wang Q; Program in Integrative Cell Signaling and Neurobiology of Metabolism, Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520.
  • Robert ME; Program in Integrative Cell Signaling and Neurobiology of Metabolism, Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520.
  • Perry RJ; Program in Integrative Cell Signaling and Neurobiology of Metabolism, Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520.
  • Chung D; Program in Integrative Cell Signaling and Neurobiology of Metabolism, Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520.
  • Shulman GI; Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520.
  • Yang X; Department of Pathology, Yale University School of Medicine, New Haven, CT 06520.
Proc Natl Acad Sci U S A ; 117(28): 16616-16625, 2020 07 14.
Article in En | MEDLINE | ID: mdl-32601203
ABSTRACT
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 ß-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.
Subject(s)
Key words

Full text: 1 Database: MEDLINE Main subject: N-Acetylglucosaminyltransferases / Ribosomal Protein S6 Kinases, 90-kDa / Macrophages / Obesity Type of study: Prognostic_studies Limits: Animals / Humans Language: En Year: 2020 Type: Article

Full text: 1 Database: MEDLINE Main subject: N-Acetylglucosaminyltransferases / Ribosomal Protein S6 Kinases, 90-kDa / Macrophages / Obesity Type of study: Prognostic_studies Limits: Animals / Humans Language: En Year: 2020 Type: Article