4.6 Article

Cancer cells metabolically fertilize the tumor microenvironment with hydrogen peroxide, driving the Warburg effect Implications for PET imaging of human tumors

Journal

CELL CYCLE
Volume 10, Issue 15, Pages 2504-2520

Publisher

TAYLOR & FRANCIS INC
DOI: 10.4161/cc.10.15.16585

Keywords

tumor stroma; microenvironment; hydrogen peroxide; aerobic glycolysis; mitochondrial oxidative phosphorylation; glucose uptake; oxidative stress; reactive oxygen species (ROS); cancer associated fibroblasts; PET imaging; the field effect; caveolin-1

Categories

Funding

  1. NIH/NCI [R01-CA-080250, R01-CA-098779, R01-CA-120876, R01-AR-055660, R01-CA-70896, R01-CA-75503, R01-CA-86072, R01-CA-107382]
  2. Susan G. Komen Breast Cancer Foundation
  3. W.W. Smith Charitable Trust
  4. Breast Cancer Alliance (BCA)
  5. American Cancer Society (ACS)
  6. Margaret Q. Landenberger Research Foundation
  7. Dr. Ralph and Marian C. Falk Medical Research Trust
  8. NIH/NCI Cancer Center [P30-CA-56036]
  9. Pennsylvania Department of Health
  10. Breakthrough Breast Cancer in the UK
  11. European Research Council

Ask authors/readers for more resources

Previously, we proposed that cancer cells behave as metabolic parasites, as they use targeted oxidative stress as a weapon to extract recycled nutrients from adjacent stromal cells. Oxidative stress in cancer-associated fibroblasts triggers autophagy and mitophagy, resulting in compartmentalized cellular catabolism, loss of mitochondrial function, and the onset of aerobic glycolysis, in the tumor stroma. As such, cancer-associated fibroblasts produce high-energy nutrients (such as lactate and ketones) that fuel mitochondrial biogenesis and oxidative metabolism in cancer cells. We have termed this new energy-transfer mechanism the Reverse Warburg Effect. To further test the validity of this hypothesis, here we used an in vitro MCF7-fibroblast co-culture system and quantitatively measured a variety of metabolic parameters by FACS analysis (analogous to laser-capture micro-dissection). Mitochondrial activity, glucose uptake and ROS production were measured with highly-sensitive fluorescent probes (MitoTracker, NBD-2-deoxy-glucose and DCF-DA). Interestingly, using this approach, we directly show that cancer cells initially secrete hydrogen peroxide that then triggers oxidative stress in neighboring fibroblasts. Thus, oxidative stress is contagious (spreads like a virus) and is propagated laterally and vectorially from cancer cells to adjacent fibroblasts. Experimentally, we show that oxidative stress in cancer-associated fibroblasts quantitatively reduces mitochondrial activity and increases glucose uptake, as the fibroblasts become more dependent on aerobic glycolysis. Conversely, co-cultured cancer cells show significant increases in mitochondrial activity and corresponding reductions in both glucose uptake and GLUT1 expression. Pretreatment of co-cultures with extracellular catalase (an anti-oxidant enzyme that detoxifies hydrogen peroxide) blocks the onset of oxidative stress and potently induces the death of cancer cells, likely via starvation. Given that cancer-associated fibroblasts show the largest increases in glucose uptake, we suggest that PET imaging of human tumors, with Fluoro-2-deoxy-D-glucose (F-2-DG), may be specifically detecting the tumor stroma, rather than epithelial cancer cells.

Authors

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

Reviews

Primary Rating

4.6
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available