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.

  • Article
  • Published:

Critical role of ASCT2-mediated amino acid metabolism in promoting leukaemia development and progression

Abstract

Amino acid metabolism is involved in diverse cellular functions, including cell survival and growth; however, it remains unclear how it regulates normal haematopoiesis versus leukaemogenesis. Here, we report that knockout of solute carrier family 1 member 5 (Slc1a5/ASCT2), a transporter of neutral amino acids, especially glutamine, results in mild-to-moderate defects in bone marrow and mature blood cell development under steady-state conditions. In contrast, constitutive or induced deletion of Slc1a5 decreases leukaemia initiation and maintenance driven by oncogene MLL-AF9 or phosphatase and tensin homologue (Pten) deficiency. Survival of leukaemic mice is prolonged following Slc1a5 deletion, and pharmacological inhibition of ASCT2 also decreases leukaemia development and progression in xenograft models of human acute myeloid leukaemia. Mechanistically, loss of ASCT2 generates a global effect on cellular metabolism, disrupts leucine (Leu) influx and mechanistic target of rapamycin (mTOR) signalling, and induces apoptosis in leukaemic cells. Given the substantial difference in reliance on ASCT2-mediated amino acid metabolism between normal and malignant blood cells, this in vivo study suggests ASCT2 as a promising therapeutic target for the treatment of leukaemia.

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

Fig. 1: Deletion of ASCT2 moderately and severely decreases blood cell development under steady-state and stress conditions, respectively.
Fig. 2: Constitutive deletion of ASCT2 inhibits MLL-AF9-induced leukaemogenesis.
Fig. 3: Induced deletion of ASCT2 suppresses established leukaemia driven by MLL-AF9.
Fig. 4: Constitutive deletion of ASCT2 inhibits Pten-loss-evoked leukaemogenesis.
Fig. 5: Deletion of ASCT2 decreases mitochondrial metabolism, and induces cell cycle arrest and apoptosis in Pten-deficient leukaemic cells.
Fig. 6: Loss of ASCT2 generates a global effect on cellular metabolism in Pten-deficient leukaemic cells.
Fig. 7: Cell-permeable Leu analogue largely reverses the effect of ASCT2 deletion on mTOR signalling and cell survival in leukaemic cells.
Fig. 8: Pharmacological inhibition of ASCT2 suppresses leukaemia development in xenograft models of human AML.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding author upon reasonable request. The Reporting Summary for this article is available as a Supplementary Information file.

References

  1. Hay, N. Reprogramming glucose metabolism in cancer: can it be exploited for cancer therapy? Nat. Rev. Cancer 16, 635–649 (2016).

    Article  CAS  Google Scholar 

  2. Vander Heiden, M. G. & DeBerardinis, R. J. Understanding the intersections between metabolism and cancer biology. Cell 168, 657–669 (2017).

    Article  CAS  Google Scholar 

  3. Pavlova, N. N. & Thompson, C. B. The emerging hallmarks of cancer metabolism. Cell Metab. 23, 27–47 (2016).

    Article  CAS  Google Scholar 

  4. DeNicola, G. M. & Cantley, L. C. Cancer’s fuel choice: new flavors for a picky eater. Mol. Cell 60, 514–523 (2015).

    Article  CAS  Google Scholar 

  5. Carracedo, A., Cantley, L. C. & Pandolfi, P. P. Cancer metabolism: fatty acid oxidation in the limelight. Nat. Rev. Cancer 13, 227–232 (2013).

    Article  CAS  Google Scholar 

  6. Altman, B. J., Stine, Z. E. & Dang, C. V. From Krebs to clinic: glutamine metabolism to cancer therapy. Nat. Rev. Cancer 16, 619–634 (2016).

    Article  CAS  Google Scholar 

  7. Palm, W. & Thompson, C. B. Nutrient acquisition strategies of mammalian cells. Nature 546, 234–242 (2017).

    Article  CAS  Google Scholar 

  8. Sassone-Corsi, P. Physiology. When metabolism and epigenetics converge. Science 339, 148–150 (2013).

    Article  CAS  Google Scholar 

  9. Lu, C. & Thompson, C. B. Metabolic regulation of epigenetics. Cell Metab. 16, 9–17 (2012).

    Article  CAS  Google Scholar 

  10. Saxton, R. A. & Sabatini, D. M. mTOR signaling in growth, metabolism, and disease. Cell 168, 960–976 (2017).

    Article  CAS  Google Scholar 

  11. Jewell, J. L., Russell, R. C. & Guan, K. L. Amino acid signalling upstream of mTOR. Nat. Rev. Mol. Cell Biol. 14, 133–139 (2013).

    Article  CAS  Google Scholar 

  12. Kekuda, R. et al. Cloning of the sodium-dependent, broad-scope, neutral amino acid transporter Bo from a human placental choriocarcinoma cell line. J. Biol. Chem. 271, 18657–18661 (1996).

    Article  CAS  Google Scholar 

  13. Fuchs, B. C. & Bode, B. P. Amino acid transporters ASCT2 and LAT1 in cancer: partners in crime? Semin. Cancer Biol. 15, 254–266 (2005).

    Article  CAS  Google Scholar 

  14. Hassanein, M. et al. SLC1A5 mediates glutamine transport required for lung cancer cell growth and survival. Clin. Cancer Res. 19, 560–570 (2013).

    Article  CAS  Google Scholar 

  15. van Geldermalsen, M. et al. ASCT2/SLC1A5 controls glutamine uptake and tumour growth in triple-negative basal-like breast cancer. Oncogene 35, 3201–3208 (2016).

    Article  CAS  Google Scholar 

  16. Wang, Q. et al. Targeting ASCT2-mediated glutamine uptake blocks prostate cancer growth and tumour development. J. Pathol. 236, 278–289 (2015).

    Article  CAS  Google Scholar 

  17. Willems, L. et al. Inhibiting glutamine uptake represents an attractive new strategy for treating acute myeloid leukemia. Blood 122, 3521–3532 (2013).

    Article  CAS  Google Scholar 

  18. Schulte, M. L. et al. Pharmacological blockade of ASCT2-dependent glutamine transport leads to antitumor efficacy in preclinical models. Nat. Med. 24, 194–202 (2018).

    Article  CAS  Google Scholar 

  19. Yu, W. M. et al. Metabolic regulation by the mitochondrial phosphatase PTPMT1 is required for hematopoietic stem cell differentiation. Cell Stem Cell 12, 62–74 (2013).

    Article  CAS  Google Scholar 

  20. Liu, X. et al. Maintenance of mouse hematopoietic stem cells ex vivo by reprogramming cellular metabolism. Blood 125, 1562–1565 (2015).

    Article  CAS  Google Scholar 

  21. Oburoglu, L. et al. Glucose and glutamine metabolism regulate human hematopoietic stem cell lineage specification. Cell Stem Cell 15, 169–184 (2014).

    Article  CAS  Google Scholar 

  22. Kühn, R., Schwenk, F., Aguet, M. & Rajewsky, K. Inducible gene targeting in mice. Science 269, 1427–1429 (1995).

    Article  Google Scholar 

  23. Velasco-Hernandez, T., Säwén, P., Bryder, D. & Cammenga, J. Potential pitfalls of the Mx1-Cre system: implications for experimental modeling of normal and malignant hematopoiesis. Stem Cell Rep. 7, 11–18 (2016).

    Article  CAS  Google Scholar 

  24. Guo, W. et al. Multi-genetic events collaboratively contribute to Pten-null leukaemia stem-cell formation. Nature 453, 529–533 (2008).

    Article  CAS  Google Scholar 

  25. Yilmaz, O. H. et al. Pten dependence distinguishes haematopoietic stem cells from leukaemia-initiating cells. Nature 441, 475–482 (2006).

    Article  CAS  Google Scholar 

  26. Zhang, J., Pavlova, N. N. & Thompson, C. B. Cancer cell metabolism: the essential role of the nonessential amino acid, glutamine. EMBO J. 36, 1302–1315 (2017).

    Article  CAS  Google Scholar 

  27. Jin, L., Alesi, G. N. & Kang, S. Glutaminolysis as a target for cancer therapy. Oncogene 35, 3619–3625 (2016).

    Article  CAS  Google Scholar 

  28. Nicklin, P. et al. Bidirectional transport of amino acids regulates mTOR and autophagy. Cell 136, 521–534 (2009).

    Article  CAS  Google Scholar 

  29. Manifava, M. et al. Dynamics of mTORC1 activation in response to amino acids. eLife 5, e19960 (2016).

    Article  Google Scholar 

  30. Esslinger, C. S., Cybulski, K. A. & Rhoderick, J. F. Ngamma-aryl glutamine analogues as probes of the ASCT2 neutral amino acid transporter binding site. Bioorg. Med Chem. 13, 1111–1118 (2005).

    Article  CAS  Google Scholar 

  31. Nakaya, M. et al. Inflammatory T cell responses rely on amino acid transporter ASCT2 facilitation of glutamine uptake and mTORC1 kinase activation. Immunity 40, 692–705 (2014).

    Article  CAS  Google Scholar 

  32. Masle-Farquhar, E., Bröer, A., Yabas, M., Enders, A. & Bröer, S. ASCT2 (SLC1A5)-deficient mice have normal B-cell development, proliferation, and antibody production. Front. Immunol. 8, 549 (2017).

    Article  Google Scholar 

  33. Locasale, J. W. Serine, glycine and one-carbon units: cancer metabolism in full circle. Nat. Rev. Cancer 13, 572–583 (2013).

    Article  CAS  Google Scholar 

  34. Maddocks, O. D. et al. Serine starvation induces stress and p53-dependent metabolic remodelling in cancer cells. Nature 493, 542–546 (2013).

    Article  CAS  Google Scholar 

  35. Durán, R. V. et al. Glutaminolysis activates Rag-mTORC1 signaling. Mol. Cell 47, 349–358 (2012).

    Article  Google Scholar 

  36. Cormerais, Y. et al. The glutamine transporter ASCT2 (SLC1A5) promotes tumor growth independently of the amino acid transporter LAT1 (SLC7A5). J. Biol. Chem. 293, 2877–2887 (2018).

    Article  CAS  Google Scholar 

  37. Bröer, A., Rahimi, F. & Bröer, S. Deletion of amino acid transporter ASCT2 (SLC1A5) reveals an essential role for transporters SNAT1 (SLC38A1) and SNAT2 (SLC38A2) to sustain glutaminolysis in cancer cells. J. Biol. Chem. 291, 13194–13205 (2016).

    Article  Google Scholar 

  38. Chiu, M. et al. GPNA inhibits the sodium-independent transport system L for neutral amino acids. Amino Acids 49, 1365–1372 (2017).

    Article  CAS  Google Scholar 

  39. Dong, L. et al. Leukaemogenic effects of Ptpn11 activating mutations in the stem cell microenvironment. Nature 539, 304–308 (2016).

    Article  Google Scholar 

  40. Xu, D. et al. Non-lineage/stage-restricted effects of a gain-of-function mutation in tyrosine phosphatase Ptpn11 (Shp2) on malignant transformation of hematopoietic cells. J. Exp. Med. 208, 1977–1988 (2011).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by the National Institutes of Health grant nos. DK092722 and HL130995 (to C.K.Q).

Author information

Authors and Affiliations

Authors

Contributions

F.N. and W.M.Y. generated and characterized the haematopoietic cell development in Slc1a5 global and conditional knockout mice, set up the mouse leukaemia models and xenograft models of human AML and analysed leukaemia development/progression. F.N. also performed metabolic assays and rescue experiments. Z.L. performed the immunoblot analyses. L.J. performed the metabolite analyses. D.K.G. and S.L. provided patient specimens and discussed the work. M.R.R. and L.J. conducted TCGA and TARGET database mining and performed the correlation analyses. S.K. and H.E.B. provided critical advice on experimental design and interpretation of the data, and edited the manuscript. C.K.Q. designed the experiments and directed the entire project. F.N. and C.K.Q. wrote the manuscript with input from all authors.

Corresponding author

Correspondence to Cheng-Kui Qu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Supplementary Information

Supplementary Figures 1–13 and Supplementary Table 2

Reporting Summary

Supplementary Table 1

Metabolomic analysis data of ASCT2-deleted and ASCT2-undeleted Pten-deficient Lin– leukemic cells

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ni, F., Yu, WM., Li, Z. et al. Critical role of ASCT2-mediated amino acid metabolism in promoting leukaemia development and progression. Nat Metab 1, 390–403 (2019). https://doi.org/10.1038/s42255-019-0039-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-019-0039-6

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