4.5 Article

PolG Inhibits Gastric Cancer Glycolysis and Viability by Suppressing PKM2 Phosphorylation

Journal

CANCER MANAGEMENT AND RESEARCH
Volume 13, Issue -, Pages 1559-1570

Publisher

DOVE MEDICAL PRESS LTD
DOI: 10.2147/CMAR.S292306

Keywords

DNA polymerase gamma; energy metabolism; tumor suppressor; PKM protein; stomach neoplasms

Categories

Funding

  1. National Natural Science Foundation of China [51872332]
  2. Natural Science Foundation of Liaoning Province [20170541040]
  3. China Medical University Youth Support Program [111/1210519020]

Ask authors/readers for more resources

PolG plays a role in the prognosis of GC patients by affecting the glycolysis process and proliferation of GC cells. Silencing PolG enhances the proliferation ability of GC cells, as confirmed in both in vivo and in vitro experiments. This study identifies PolG as a potential novel therapeutic target for the treatment of GC, providing new insights into clinical GC therapy.
Purpose: Gastric cancer (GC) is the fifth most frequently diagnosed cancer and the third leading cause of cancer-related death. There is a critical need for the development of novel therapies in GC. DNA polymerase gamma (PolG) has been implicated in mitochondrial homeostasis and affects the development of numerous types of cancer, however, its effects on GC and molecular mechanisms remain to be fully determined. The aim of the present research was to clarify the effects of PolG on GC and its possible molecular mechanism of action. Methods: The GSE62254 dataset was used to predict the effect of PolG on prognostic value in GC patients. Lentivirus-mediated transduction was used to silence PolG expression. Western blot analysis evinced the silencing effect. Co-immunoprecipitation (Co-IP) analysis was performed to explore the potential molecular mechanism of action. Analysis of the glycolysis process in GC cells was also undertaken. Cell proliferation was determined using a CCK-8 (Cell Counting Kit-8) proliferation assay. Cell migration was detected using the Transwell device. Animal experiments were used to measure in vivo xenograft tumor growth. Results: GC patients with low PolG expression have worse overall survival (OS) and progression-free survival (PFS). PolG binds to PKM2 and affects the activation of Tyr105-site phosphorylation, thus interfering with the glycolysis of GC cells. In vitro tumor formation experiments in mice also confirmed that PolG silencing of GC has a stronger proliferation ability. PolG can suppress GC cell growth both in vivo and in vitro. Conclusion: Our study reveals a potential molecular mechanism between PolG and the energy metabolic process of GC tumor cells for the first time, suggesting PolG as an independent novel potential therapeutic target for tumor therapy, and providing new ideas for clinical GC treatment.

Authors

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

Reviews

Primary Rating

4.5
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available