Original Article

< Previous         Next >  
Metabolic regulation of T cell development by Sin1–mTORC2 is mediated by pyruvate kinase M2
Xinxing Ouyang1 , Yuheng Han1 , Guojun Qu1 , Man Li1 , Ningbo Wu1 , Hongzhi Liu1 , Omotooke Arojo2 , Hongxiang Sun1 , Xiaobo Liu1 , Dou Liu2 , Lei Chen1 , Qiang Zou 1,* , and Bing Su 1,2,*
1 Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
2 Department of Immunobiology and the Vascular Biology and Therapeutics Program, Yale University School of Medicine, 333 Cedar Street, New Haven, CT06520, USA
*Correspondence to:Qiang Zou, E-mail: qzou1984@sjtu.edu.cn; Bing Su, E-mail: bingsu@sjtu.edu.cn
J Mol Cell Biol, Volume 11, Issue 2, February 2019, 93-106,  https://doi.org/10.1093/jmcb/mjy065
Keyword: mTORC2, Sin1, thymocyte development, PPAR-γ, PKM2, metabolism
Glucose metabolism plays a key role in thymocyte development. The mammalian target of rapamycin complex 2 (mTORC2) is a critical regulator of cell growth and metabolism, but its role in early thymocyte development and metabolism has not been fully studied. We show here that genetic ablation of Sin1, an essential component of mTORC2, in T lineage cells results in severely impaired thymocyte development at the CD4−CD8− double negative (DN) stages but not at the CD4+CD8+ double positive (DP) or later stages. Notably, Sin1-deficient DN thymocytes show markedly reduced proliferation and glycolysis. Importantly, we discover that the M2 isoform of pyruvate kinase (PKM2) is a novel and crucial Sin1 effector in promoting DN thymocyte development and metabolism. At the molecular level, we show that Sin1–mTORC2 controls PKM2 expression through an AKT-dependent PPAR-γ nuclear translocation. Together, our study unravels a novel mTORC2−PPAR-γ−PKM2 pathway in immune-metabolic regulation of early thymocyte development.