4.7 Article

GTPBP4 promotes hepatocellular carcinoma progression and metastasis via the PKM2 dependent glucose metabolism*

Journal

REDOX BIOLOGY
Volume 56, Issue -, Pages -

Publisher

ELSEVIER
DOI: 10.1016/j.redox.2022.102458

Keywords

Hepatocellular carcinoma; Pyruvate kinase M2; Aerobic glycolysis; GTP binding protein 4

Funding

  1. National Natural Science Foundation of China [82073208, 82103521]
  2. Shanghai Shen Kang Hospital Development Center new frontier technology joint project [SHDC12021109]
  3. Shanghai Sailing Program [21YF1407500]
  4. China Postdoctoral Science Foundation [2021M690674]
  5. Science and Technology Plan Project of Xiamen City [3502Z20194036]
  6. Program of Shanghai Academic Research Leader [20XD1400900]

Ask authors/readers for more resources

GTPBP4 plays a crucial role in the development of hepatocellular carcinoma by promoting aerobic glycolysis through inducing the formation and sumoylation modification of PKM2. The SUMO-modified PKM2 may contribute to HCC progression by activating epithelial-mesenchymal transition and STAT3 signaling pathway. Shikonin, a PKM2-specific inhibitor, shows promise as a therapeutic candidate for HCC patients.
Guanosine triphosphate binding protein 4 (GTPBP4) is a key regulator of cell cycle progression and MAPK activation. However, how its biological properties intersect with cellular metabolism in hepatocellular carcinoma (HCC) development remains poorly unexplained. Here, high GTPBP4 expression is found to be significantly associated with worse clinical outcomes in patients with HCC. Moreover, GTPBP4 upregulation is paralleled by DNA promoter hypomethylation and regulated by DNMT3A, a DNA methyltransferase. Additionally, both gain -and loss-of-function studies demonstrate that GTPBP4 promotes HCC growth and metastasis in vitro and in vivo. Mechanically, GTPBP4 can induce dimeric pyruvate kinase M2 (PKM2) formation through protein sumoylation modification to promote aerobic glycolysis in HCC. Notably, active GTPBP4 facilitates SUMO1 protein activation by UBA2, and acts as a linker bridging activated SUMO1 protein and PKM2 protein to induce PKM2 sumoylation. Furthermore, SUMO-modified PKM2 relocates from the cytoplasm to the nucleus may also could contribute to HCC progression through activating epithelial-mesenchymal transition (EMT) and STAT3 signaling pathway. Shikonin, a PKM2-specific inhibitor, can attenuate PKM2 dependent HCC glycolytic reprogramming, growth and metastasis promoted by GTPBP4, which offers a promising therapeutic candidate for HCC patients. Our findings indicate that GTPBP4-PKM2 regulatory axis plays a vital role in promoting HCC proliferation as well as metastasis by aerobic glycolysis and offer a promising therapeutic target for HCC patients.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available