Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

Phosphoglycerate dehydrogenase diverts glycolytic flux and contributes to oncogenesis

Abstract

Most tumors exhibit increased glucose metabolism to lactate, however, the extent to which glucose-derived metabolic fluxes are used for alternative processes is poorly understood1,2. Using a metabolomics approach with isotope labeling, we found that in some cancer cells a relatively large amount of glycolytic carbon is diverted into serine and glycine metabolism through phosphoglycerate dehydrogenase (PHGDH). An analysis of human cancers showed that PHGDH is recurrently amplified in a genomic region of focal copy number gain most commonly found in melanoma. Decreasing PHGDH expression impaired proliferation in amplified cell lines. Increased expression was also associated with breast cancer subtypes, and ectopic expression of PHGDH in mammary epithelial cells disrupted acinar morphogenesis and induced other phenotypic alterations that may predispose cells to transformation. Our findings show that the diversion of glycolytic flux into a specific alternate pathway can be selected during tumor development and may contribute to the pathogenesis of human cancer.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Observation of glycolytic metabolism being diverted into serine and glycine metabolism.
Figure 2: PHGDH amplification in human cancers and requirement for proliferation.
Figure 3: Growth dependence of PHGDH expression and altered serine metabolism in PHGDH-amplified human melanoma cells.
Figure 4: Ectopic expression of PHGDH in breast ductal morphogenesis.

Similar content being viewed by others

References

  1. Vander Heiden, M.G., Cantley, L.C. & Thompson, C.B. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 324, 1029–1033 (2009).

    Article  CAS  Google Scholar 

  2. DeBerardinis, R.J., Lum, J.J., Hatzivassiliou, G. & Thompson, C.B. The biology of cancer: metabolic reprogramming fuels cell growth and proliferation. Cell Metab. 7, 11–20 (2008).

    Article  CAS  Google Scholar 

  3. Warburg, O., Posener, K. & Negelein, E. Ueber den Stoffwechsel der Tumoren. Biochem. Z. 152, 319–344 (1924).

    Google Scholar 

  4. Bodenhausen, G. & Ruben, D.J. Natural abundance nitrogen-15 NMR by enhanced heteronuclear spectroscopy. Chem. Phys. Lett. 69, 185–189 (1980).

    Article  CAS  Google Scholar 

  5. Bismut, H., Caron, M., Coudray-Lucas, C. & Capeau, J. Glucose contribution to nucleic acid base synthesis in proliferating hepatoma cells: a glycine-biosynthesis-mediated pathway. Biochem. J. 308, 761–767 (1995).

    Article  CAS  Google Scholar 

  6. Snell, K., Natsumeda, Y. & Weber, G. The modulation of serine metabolism in hepatoma 3924A during different phases of cellular proliferation in culture. Biochem. J. 245, 609–612 (1987).

    Article  CAS  Google Scholar 

  7. Kit, S. The biosynthesis of free glycine and serine by tumors. Cancer Res. 15, 715–718 (1955).

    CAS  PubMed  Google Scholar 

  8. de Koning, T.J. et al. L-serine in disease and development. Biochem. J. 371, 653–661 (2003).

    Article  CAS  Google Scholar 

  9. Achouri, Y., Rider, M.H., Van Schaftingen, E. & Robbi, M. Cloning, sequencing and expression of rat liver 3-phosphoglycerate dehydrogenase. Biochem. J. 323, 365–370 (1997).

    Article  CAS  Google Scholar 

  10. Lu, W., Bennett, B.D. & Rabinowitz, J.D. Analytical strategies for LC-MS-based targeted metabolomics. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 871, 236–242 (2008).

    Article  CAS  Google Scholar 

  11. Beroukhim, R. et al. The landscape of somatic copy-number alteration across human cancers. Nature 463, 899–905 (2010).

    Article  CAS  Google Scholar 

  12. Greshock, J. et al. A comparison of DNA copy number profiling platforms. Cancer Res. 67, 10173–10180 (2007).

    Article  CAS  Google Scholar 

  13. Slamon, D.J. et al. Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science 235, 177–182 (1987).

    Article  CAS  Google Scholar 

  14. Luo, J., Solimini, N.L. & Elledge, S.J. Principles of cancer therapy: oncogene and non-oncogene addiction. Cell 136, 823–837 (2009).

    Article  CAS  Google Scholar 

  15. Pollari, S. et al. Enhanced serine production by bone metastatic breast cancer cells stimulates osteoclastogenesis. Breast Cancer Res. Treat. 125, 421–430 (2011).

    Article  CAS  Google Scholar 

  16. Foulkes, W.D., Smith, I.E. & Reis-Filho, J. Triple-negative breast cancer. N. Engl. J. Med. 363, 1938–1948 (2010).

    Article  CAS  Google Scholar 

  17. Debnath, J. & Brugge, J.S. Modelling glandular epithelial cancers in three-dimensional cultures. Nat. Rev. Cancer 5, 675–688 (2005).

    Article  CAS  Google Scholar 

  18. Schafer, Z.T. et al. Antioxidant and oncogene rescue of metabolic defects caused by loss of matrix attachment. Nature 461, 109–113 (2009).

    Article  CAS  Google Scholar 

  19. Tabatabaie, L. et al. Novel mutations in 3-phosphoglycerate dehydrogenase (PHGDH) are distributed throughout the protein and result in altered enzyme kinetics. Hum. Mutat. 30, 749–756 (2009).

    Article  CAS  Google Scholar 

  20. Thompson, C.B. Metabolic enzymes as oncogenes or tumor suppressors. N. Engl. J. Med. 360, 813–815 (2009).

    Article  CAS  Google Scholar 

  21. Teperino, R., Schoonjans, K. & Auwerx, J. Histone methyl transferases and demethylases: can they link metabolism and transcription? Cell Metab. 12, 321–327 (2010).

    Article  CAS  Google Scholar 

  22. Nomura, D.K. et al. Monoacylglycerol lipase regulates a fatty acid network that promotes cancer pathogenesis. Cell 140, 49–61 (2010).

    Article  CAS  Google Scholar 

  23. Hara, K. et al. Amino acid sufficiency and mTOR regulate p70 S6 kinase and eIF-4E BP1 through a common effector mechanism. J. Biol. Chem. 273, 14484–14494 (1998).

    Article  CAS  Google Scholar 

  24. Vander Heiden, M.G. et al. Evidence for an alternative glycolytic pathway in rapidly proliferating cells. Science 329, 1492–1499 (2010).

    Article  CAS  Google Scholar 

  25. Locasale, J.W. & Cantley, L.C. Altered metabolism in cancer. BMC Biol. 8, 88 (2010).

    Article  Google Scholar 

  26. Eng, C.H., Yu, K., Lucas, J., White, E. & Abraham, R.T. Ammonia derived from glutaminolysis is a diffusible regulator of autophagy. Sci. Signal. 3, ra31 (2010).

    PubMed  Google Scholar 

  27. Antoniewicz, M.R., Kelleher, J.K. & Stephanopoulos, G. Accurate assessment of amino acid mass isotopomer distributions for metabolic flux analysis. Anal. Chem. 79, 7554–7559 (2007).

    Article  CAS  Google Scholar 

  28. Fernandez, C.A., Des Rosiers, C., Previs, S.F., David, F. & Brunengraber, H. Correction of 13C mass isotopomer distributions for natural stable isotope abundance. J. Mass Spectrom. 31, 255–262 (1996).

    Article  CAS  Google Scholar 

  29. Richardson, A.L. et al. X chromosomal abnormalities in basal-like human breast cancer. Cancer Cell 9, 121–132 (2006).

    Article  CAS  Google Scholar 

  30. Hoek, K. et al. Expression profiling reveals novel pathways in the transformation of melanocytes to melanomas. Cancer Res. 64, 5270–5282 (2004).

    Article  CAS  Google Scholar 

  31. Rhodes, D.R. et al. ONCOMINE: a cancer microarray database and integrated data-mining platform. Neoplasia 6, 1–6 (2004).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

Microscopy data for this study were acquired and analyzed in the Nikon Imaging Center at Harvard Medical School. J.W.L. was supported by postdoctoral fellowships from the US National Institutes of Health (NIH) and the American Cancer Society. A.R.G. is a recipient of a National Science Foundation (NSF) Graduate Research Fellowship. L.C.C. and J.S.B. were supported by grants from the NIH and the National Cancer Institute (NCI). M.G.V.H. was supported by grants from the NIH, NCI, Smith Family, Damon Runyon Cancer Research Foundation and the Burroughs Wellcome Fund. We thank N. Vena for technical assistance with the FISH analysis and K. Webster and I. Carrecedo for help with immunohistochemistry. We thank J. Rabinowitz, A. Carrecedo and S.-C. Ng for helpful comments on the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

J.W.L., M.G.V.H. and L.C.C. designed the study and wrote the paper. J.W.L., C.A.L., E.M., K.R.M., D.A., H.S., M.G.V.H. and T. Melman carried out experiments. J.W.L. and T. Melman carried out computational analyses. A.J.B., R.B. and M.M. provided help with copy number data. L.C. and A.L.R. provided human cancer samples. N.I.V. and A.H.L. carried out the FISH analysis. J.W.L. and J.M.A. carried out the LC/MS/MS experiments. J.W.L., G.H. and G.W. carried out the NMR experiments. J.W.L., N.I.V., C.M.M. and G.S. carried out the GC/MS experiments. M.S. and A.T.S. generated reagents. J.S.B., T. Muranen and A.R.G. carried out experiments involving acinar morphogenesis and imaging analysis.

Corresponding authors

Correspondence to Jason W Locasale, Lewis C Cantley or Matthew G Vander Heiden.

Ethics declarations

Competing interests

J.W.L., M.V.H. and L.C.C. are consultants, scientific advisors and part owners of Agios Pharmaceuticals and hold patents pertaining to targeting cellular metabolism for cancer treatment. Agios Pharmaceuticals is interested in developing therapeutics that target altered metabolism in cancer.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 and Supplementary Tables 1–4. (PDF 1929 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Locasale, J., Grassian, A., Melman, T. et al. Phosphoglycerate dehydrogenase diverts glycolytic flux and contributes to oncogenesis. Nat Genet 43, 869–874 (2011). https://doi.org/10.1038/ng.890

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.890

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer