4.7 Article

Covalent Inhibition of Pyruvate Kinase M2 Reprograms Metabolic and Inflammatory Pathways in Hepatic Macrophages against Non-alcoholic Fatty Liver Disease

期刊

INTERNATIONAL JOURNAL OF BIOLOGICAL SCIENCES
卷 18, 期 14, 页码 5260-5275

出版社

IVYSPRING INT PUBL
DOI: 10.7150/ijbs.73890

关键词

macrophage polarization; pyruvate kinase M2; celastrol; covalent modification; non-alcoholic fatty liver disease

资金

  1. General Research Fund (GRF) [17146216, 17100317, 17119619]
  2. National Natural Science Foundation of China [81701464, 81703726, 21778046]
  3. Health and Medical Research Fund [16171751, 17181231]
  4. Midstream Research Programme for Universities [(MRP) 053/18X (2018)]

向作者/读者索取更多资源

This study demonstrated that celastrol can reprogram metabolic and inflammatory pathways in M1 macrophages by binding to pyruvate kinase M2 (PKM2). Celastrol inhibits the expression of glycolytic enzymes and related signaling proteins, shifts aerobic glycolysis to mitochondrial oxidative phosphorylation, and skews macrophage polarization from inflammatory M1 type to anti-inflammatory M2 type. Animal experiments showed that celastrol can reduce weight loss, serum cholesterol level, lipid accumulation, and hepatic fibrosis in the mouse model of NAFLD.
Warburg effect of aerobic glycolysis in hepatic M1 macrophages is a major cause for metabolic dysfunction and inflammatory stress in non-alcoholic fatty liver disease (NAFLD). Plant-derived triterpene celastrol markedly inhibited macrophage M1 polarization and adipocyte hypertrophy in obesity. The present study was designed to identify the celastrol-bound proteins which reprogrammed metabolic and inflammatory pathways in M1 macrophages. Pyruvate kinase M2 (PKM2) was determined to be a major celastrol-bound protein. Peptide mapping revealed that celastrol bound to the residue Cys(31) while covalent conjugation altered the spatial conformation and inhibited the enzyme activity of PKM2. Mechanistic studies showed that celastrol reduced the expression of glycolytic enzymes (e.g., GLUT1, HK2, LDHA, PKM2) and related signaling proteins (e.g., Akt, HIF-1 alpha, mTOR), shifted aerobic glycolysis to mitochondrial oxidative phosphorylation and skewed macrophage polarization from inflammatory M1 type to anti-inflammatory M2 type. Animal experiments indicated that celastrol promoted weight loss, reduced serum cholesterol level, lipid accumulation and hepatic fibrosis in the mouse model of NAFLD. Collectively, the present study demonstrated that celastrol might alleviate lipid accumulation, inflammation and fibrosis in the liver via covalent modification of PKM2.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.7
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据