1. Academic Validation
  2. Low glucose metabolite 3-phosphoglycerate switches PHGDH from serine synthesis to p53 activation to control cell fate

Low glucose metabolite 3-phosphoglycerate switches PHGDH from serine synthesis to p53 activation to control cell fate

  • Cell Res. 2023 Sep 19. doi: 10.1038/s41422-023-00874-4.
Yu-Qing Wu # 1 Chen-Song Zhang # 1 Jinye Xiong 1 Dong-Qi Cai 1 Chen-Zhe Wang 1 Yu Wang 1 Yan-Hui Liu 1 Yu Wang 2 Yiming Li 2 Jian Wu 2 Jianfeng Wu 3 Bin Lan 4 Xuefeng Wang 4 Siwei Chen 1 Xianglei Cao 1 Xiaoyan Wei 1 Hui-Hui Hu 1 Huiling Guo 1 Yaxin Yu 1 Abdul Ghafoor 1 Changchuan Xie 1 Yaying Wu 1 Zheni Xu 1 Cixiong Zhang 1 Mingxia Zhu 1 Xi Huang 1 Xiufeng Sun 1 Shu-Yong Lin 1 Hai-Long Piao 5 Jianyin Zhou 2 Sheng-Cai Lin 6
Affiliations

Affiliations

  • 1 State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China.
  • 2 Department of Hepatobiliary and Pancreatic Surgery, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, China.
  • 3 Laboratory Animal Research Center, Xiamen University, Xiamen, Fujian, China.
  • 4 Fujian Provincial Key Laboratory of Tumor Biotherapy, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Xiamen, Fujian, China.
  • 5 CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning, China.
  • 6 State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China. [email protected].
  • # Contributed equally.
Abstract

Glycolytic intermediary metabolites such as fructose-1,6-bisphosphate can serve as signals, controlling metabolic states beyond energy metabolism. However, whether glycolytic metabolites also play a role in controlling cell fate remains unexplored. Here, we find that low levels of glycolytic metabolite 3-phosphoglycerate (3-PGA) can switch phosphoglycerate dehydrogenase (PHGDH) from cataplerosis serine synthesis to pro-apoptotic activation of p53. PHGDH is a p53-binding protein, and when unoccupied by 3-PGA interacts with the scaffold protein AXIN in complex with the kinase HIPK2, both of which are also p53-binding proteins. This leads to the formation of a multivalent p53-binding complex that allows HIPK2 to specifically phosphorylate p53-Ser46 and thereby promote Apoptosis. Furthermore, we show that PHGDH mutants (R135W and V261M) that are constitutively bound to 3-PGA abolish p53 activation even under low glucose conditions, while the mutants (T57A and T78A) unable to bind 3-PGA cause constitutive p53 activation and Apoptosis in hepatocellular carcinoma (HCC) cells, even in the presence of high glucose. In vivo, PHGDH-T57A induces Apoptosis and inhibits the growth of diethylnitrosamine-induced mouse HCC, whereas PHGDH-R135W prevents Apoptosis and promotes HCC growth, and knockout of Trp53 abolishes these effects above. Importantly, caloric restriction that lowers whole-body glucose levels can impede HCC growth dependent on PHGDH. Together, these results unveil a mechanism by which glucose availability autonomously controls p53 activity, providing a new paradigm of cell fate control by metabolic substrate availability.

Figures
Products