4.8 Article

Modeling the Genetic Regulation of Cancer Metabolism: Interplay between Glycolysis and Oxidative Phosphorylation

Journal

CANCER RESEARCH
Volume 77, Issue 7, Pages 1564-1574

Publisher

AMER ASSOC CANCER RESEARCH
DOI: 10.1158/0008-5472.CAN-16-2074

Keywords

-

Categories

Funding

  1. Frontiers Center NSF grant [PHY-1427654]
  2. NSF [DMS-1361411]
  3. Cancer Prevention and Research Institute of Texas (CPRIT) grants [R1110, R1111]
  4. Keck Center for Interdisciplinary Bioscience Training of the Gulf Coast Consortia (CPRIT Grant) [RP140113]
  5. National Institutes of Health [R01-GM067801, R01-GM116280]
  6. Welch Foundation [Q-1512]
  7. Tauber Family Funds
  8. Maguy-Glass Chair in Physics of Complex Systems
  9. National Cancer Institute (NCI) grants [R21CA173150, R21CA179720]
  10. Direct For Mathematical & Physical Scien
  11. Division Of Physics [1427654] Funding Source: National Science Foundation
  12. Division Of Mathematical Sciences
  13. Direct For Mathematical & Physical Scien [1361411] Funding Source: National Science Foundation

Ask authors/readers for more resources

Abnormal metabolism is a hallmark of cancer, yet its regulation remains poorly understood. Cancer cells were considered to utilize primarily glycolysis for ATP production, referred to as the Warburg effect. However, recent evidence suggests that oxidative phosphorylation (OXPHOS) plays a crucial role during cancer progression. Here we utilized a systems biology approach to decipher the regulatory principle of glycolysis and OXPHOS. Integrating information from literature, we constructed a regulatory network of genes and metabolites, from which we extracted a core circuit containing HIF-1, AMPK, and ROS. Our circuit analysis showed that while normal cells have an oxidative state and a glycolytic state, cancer cells can access a hybrid state with both metabolic modes coexisting. This was due to higher ROS production and/or oncogene activation, such as RAS, MYC, and c-SRC. Guided by the model, we developed two signatures consisting of AMPK and HIF-1 downstream genes, respectively, to quantify the activity of glycolysis and OXPHOS. By applying the AMPK and HIF-1 signatures to The Cancer Genome Atlas patient transcriptomics data of multiple cancer types and single-cell RNA-seq data of lung adenocarcinoma, we confirmed an anticorrelation between AMPK and HIF-1 activities and the association of metabolic states with oncogenes. We propose that the hybrid phenotype contributes to metabolic plasticity, allowing cancer cells to adapt to various microenvironments. Using model simulations, our theoretical framework of metabolism can serve as a platform to decode cancer metabolic plasticity and design cancer therapies targeting metabolism.

Authors

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

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available