4.6 Article

Evidence for a stromal-epithelial lactate shuttle in human tumors MCT4 is a marker of oxidative stress in cancer-associated fibroblasts

Journal

CELL CYCLE
Volume 10, Issue 11, Pages 1772-1783

Publisher

TAYLOR & FRANCIS INC
DOI: 10.4161/cc.10.11.15659

Keywords

caveolin-1; oxidative stress; pseudohypoxia; lactate shuttle; MCT1; MCT4; metabolic coupling; tumor stroma; predictive biomarker; SLC16A1; SLC16A3; monocarboxylic acid transporter

Categories

Funding

  1. W.W. Smith Charitable Trust
  2. Breast Cancer Alliance (BCA)
  3. American Cancer Society (ACS)
  4. 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, P30-CA-56036]
  5. Susan G. Komen Breast Cancer Foundation
  6. Breast Cancer Alliance, Inc.
  7. Susan G. Komen Career Catalyst Grant
  8. Dr. Ralph and Marian C. Falk Medical Research Trust
  9. Margaret Q. Landenberger Research Foundation
  10. Breakthrough Breast Cancer in the UK
  11. European Research Council

Ask authors/readers for more resources

Recently, we proposed a new mechanism for understanding the Warburg effect in cancer metabolism. In this new paradigm, cancer-associated fibroblasts undergo aerobic glycolysis, and extrude lactate to feed adjacent cancer cells, which then drives mitochondrial biogenesis and oxidative mitochondrial metabolism in cancer cells. Thus, there is vectorial transport of energy-rich substrates from the fibroblastic tumor stroma to anabolic cancer cells. A prediction of this hypothesis is that cancer-associated fibroblasts should express MCT4, a mono-carboxylate transporter that has been implicated in lactate efflux from glycolytic muscle fibers and astrocytes in the brain. To address this issue, we co-cultured MCF7 breast cancer cells with normal fibroblasts. Interestingly, our results directly show that breast cancer cells specifically induce the expression of MCT4 in cancer-associated fibroblasts; MCF7 cells alone and fibroblasts alone, both failed to express MCT4. We also show that the expression of MCT4 in cancer-associated fibroblasts is due to oxidative stress, and can be prevented by pre-treatment with the anti-oxidant N-acetyl-cysteine. In contrast to our results with MCT4, we see that MCT1, a transporter involved in lactate uptake, is specifically upregulated in MCF7 breast cancer cells when co-cultured with fibroblasts. Virtually identical results were also obtained with primary human breast cancer samples. In human breast cancers, MCT4 selectively labels the tumor stroma, e. g., the cancer-associated fibroblast compartment. Conversely, MCT1 was selectively expressed in the epithelial cancer cells within the same tumors. Functionally, we show that overexpression of MCT4 in fibroblasts protects both MCF7 cancer cells and fibroblasts against cell death, under co-culture conditions. Thus, we provide the first evidence for the existence of a stromal-epithelial lactate shuttle in human tumors, analogous to the lactate shuttles that are essential for the normal physiological function of muscle tissue and brain. These data are consistent with the reverse Warburg effect, which states that cancer-associated fibroblasts undergo aerobic glycolysis, thereby producing lactate, which is utilized as a metabolic substrate by adjacent cancer cells. In this model, energy transfer or metabolic-coupling between the tumor stroma and epithelial cancer cells fuels tumor growth and metastasis, via oxidative mitochondrial metabolism in anabolic cancer cells. Most importantly, our current findings provide a new rationale and novel strategy for anti-cancer therapies, by employing MCT inhibitors.

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