Oncotarget

Research Papers:

TP53 mutation hits energy metabolism and increases glycolysis in breast cancer

Hajnalka Harami-Papp _, Lőrinc S. Pongor, Gyöngyi Munkácsy, Gergő Horváth, Ádám M. Nagy, Attila Ambrus, Péter Hauser, András Szabó, László Tretter and Balázs Győrffy

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2016; 7:67183-67195. https://doi.org/10.18632/oncotarget.11594

Metrics: PDF 3318 views  |   HTML 5060 views  |   ?  


Abstract

Hajnalka Harami-Papp1,*, Lőrinc S. Pongor1,3,*, Gyöngyi Munkácsy1, Gergő Horváth2, Ádám M. Nagy2, Attila Ambrus2, Péter Hauser3, András Szabó3, László Tretter2, Balázs Győrffy1

1MTA TTK Lendület Cancer Biomarker Research Group, H-1117, Budapest, Hungary

2Semmelweis University, Department of Medical Biochemistry, H-1094, Budapest, Hungary

3Semmelweis University, Second Department of Pediatrics, H-1094, Budapest, Hungary

*These authors have contributed equally to this work

Correspondence to:

Balázs Győrffy, email: [email protected]

László Tretter, email: [email protected]

Keywords: glycolytic efficiency, breast cancer, Warburg effect, next generation sequencing

Received: May 24, 2016     Accepted: August 13, 2016     Published: August 25, 2016

ABSTRACT

Promising new hallmarks of cancer is alteration of energy metabolism that involves molecular mechanisms shifting cancer cells to aerobe glycolysis. Our goal was to evaluate the correlation between mutation in the commonly mutated tumor suppressor gene TP53 and metabolism. We established a database comprising mutation and RNA-seq expression data of the TCGA repository and performed receiver operating characteristics (ROC) analysis to compare expression of each gene between TP53 mutated and wild type samples. All together 762 breast cancer samples were evaluated of which 215 had TP53 mutation. Top up-regulated metabolic genes include glycolytic enzymes (e.g. HK3, GPI, GAPDH, PGK1, ENO1), glycolysis regulator (PDK1) and pentose phosphate pathway enzymes (PGD, TKT, RPIA). Gluconeogenesis enzymes (G6PC3, FBP1) were down-regulated. Oxygen consumption and extracellular acidification rates were measured in TP53 wild type and mutant breast cell lines with a microfluorimetric analyzer. Applying metabolic inhibitors in the presence and absence of D-glucose and L-glutamine in cell culture experiments resulted in higher glycolytic and mitochondrial activity in TP53 mutant breast cancer cell lines. In summary, TP53 mutation influences energy metabolism at multiple levels. Our results provide evidence for the synergistic activation of multiple hallmarks linking to these the mutation status of a key driver gene.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 11594