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:

GLS2 is protumorigenic in breast cancers

Abstract

Many types of cancers have a well-established dependence on glutamine metabolism to support survival and growth, a process linked to glutaminase 1 (GLS) isoforms. Conversely, GLS2 variants often have tumor-suppressing activity. Triple-negative (TN) breast cancer (testing negative for estrogen, progesterone, and Her2 receptors) has elevated GLS protein levels and reportedly depends on exogenous glutamine and GLS activity for survival. Despite having high GLS levels, we verified that several breast cancer cells (including TN cells) express endogenous GLS2, defying its role as a bona fide tumor suppressor. Moreover, ectopic GLS2 expression rescued cell proliferation, TCA anaplerosis, redox balance, and mitochondrial function after GLS inhibition by the small molecule currently in clinical trials CB-839 or GLS knockdown of GLS-dependent cell lines. In several cell lines, GLS2 knockdown decreased cell proliferation and glutamine-linked metabolic phenotypes. Strikingly, long-term treatment of TN cells with another GLS-exclusive inhibitor bis-2′-(5-phenylacetamide-1,3,4-thiadiazol-2-yl)ethyl sulfide (BPTES) selected for a drug-resistant population with increased endogenous GLS2 and restored proliferative capacity. GLS2 was linked to enhanced in vitro cell migration and invasion, mesenchymal markers (through the ERK-ZEB1-vimentin axis under certain conditions) and in vivo lung metastasis. Of concern, GLS2 amplification or overexpression is linked to an overall, disease-free and distant metastasis-free worse survival prognosis in breast cancer. Altogether, these data establish an unforeseen role of GLS2 in sustaining tumor proliferation and underlying metastasis in breast cancer and provide an initial framework for exploring GLS2 as a novel therapeutic target.

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
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8

Similar content being viewed by others

References

  1. Gao P, Tchernyshyov I, Chang T-C, Lee Y-S, Kita K, Ochi T, et al. c-Myc suppression of miR-23a/b enhances mitochondrial glutaminase expression and glutamine metabolism. Nature. 2009;458:762–5.

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Elgadi KM, Meguid Ra, Qian M, Souba WW, Abcouwer SF. Cloning and analysis of unique human glutaminase isoforms generated by tissue-specific alternative splicing. Physiol Genom. 1999;1:51–62.

    CAS  Google Scholar 

  3. Wang J-B Bin, Erickson JW, Fuji R, Ramachandran S, Gao P, Dinavahi R, et al. Targeting mitochondrial glutaminase activity inhibits oncogenic transformation. Cancer Cell. 2010;18:207–19.

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Seltzer MJ, Bennett BD, Joshi AD, Gao P, Thomas AG, Ferraris DV, et al. Inhibition of glutaminase preferentially slows growth of glioma cells with mutant IDH1. Cancer Res. 2010;70:8981–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Liu W, Le A, Hancock C, Lane AN, Dang CV, Fan TW-M, et al. Reprogramming of proline and glutamine metabolism contributes to the proliferative and metabolic responses regulated by oncogenic transcription factor c-MYC. Proc Natl Acad Sci USA. 2012;109:8983–8.

    CAS  PubMed  Google Scholar 

  6. van den Heuvel APJ, Jing J, Wooster RF, Bachman KE. Analysis of glutamine dependency in non-small cell lung cancer. Cancer Biol Ther. 2012;13:1185–94.

    PubMed  PubMed Central  Google Scholar 

  7. Yuneva MO, Fan TWM, Allen TD, Higashi RM, Ferraris DV, Tsukamoto T, et al. The metabolic profile of tumors depends on both the responsible genetic lesion and tissue type. Cell Metab. 2012;15:157–70.

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Son J, Lyssiotis Ca, Ying H, Wang X, Hua S, Ligorio M, et al. Glutamine supports pancreatic cancer growth through a KRAS-regulated metabolic pathway. Nature. 2013;496:101–5.

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Gameiro PA, Yang J, Metelo AM, Pérez-Carro R, Baker R, Wang Z, et al. In vivo HIF-mediated reductive carboxylation is regulated by citrate levels and sensitizes VHL-deficient cells to glutamine deprivation. Cell Metab. 2013;17:372–85.

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Timmerman LA, Holton T, Yuneva M, Louie RJ, Padró M, Daemen A, et al. Glutamine sensitivity analysis identifies the xCT antiporter as a common triple-negative breast tumor therapeutic target. Cancer Cell. 2013;24:450–65.

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Cassago A, Ferreira APS, Ferreira IM, Fornezari C, Gomes ERM, Greene KS, et al. Mitochondrial localization and structure-based phosphate activation mechanism of Glutaminase C with implications for cancer metabolism. Proc Natl Acad Sci. 2012;109:1092–7.

    CAS  PubMed  Google Scholar 

  12. Kung HN, Marks JR, Chi JT. Glutamine synthetase is a genetic determinant of cell type-specific glutamine independence in breast epithelia. PLoS Genet. 2011;7:e1002229.

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Gross MI, Demo SD, Dennison JB, Chen L, Chernov-Rogan T, Goyal B, et al. Antitumor activity of the glutaminase inhibitor CB-839 in triple-negative breast cancer. Mol Cancer Ther. 2014;13:890–901.

    CAS  PubMed  Google Scholar 

  14. Hu W, Zhang C, Wu R, Sun Y, Levine A, Feng Z. Glutaminase 2, a novel p53 target gene regulating energy metabolism and antioxidant function. Proc Natl Acad Sci USA. 2010;107:7455–60.

    CAS  PubMed  Google Scholar 

  15. Suzuki S, Tanaka T, Poyurovsky MV, Nagano H, Mayama T, Ohkubo S, et al. Phosphate-activated glutaminase (GLS2), a p53-inducible regulator of glutamine metabolism and reactive oxygen species. Proc Natl Acad Sci USA. 2010;107:7461–6.

    CAS  PubMed  Google Scholar 

  16. Martín-Rufián M, Tosina M, Campos-Sandoval Ja, Manzanares E, Lobo C.Segura Ja,et al. Mammalian glutaminase Gls2 gene encodes two functional alternative transcriptcanism. PLoS ONE. 2012;7:e38380.

    PubMed  PubMed Central  Google Scholar 

  17. Szeliga M, Albrecht J. Opposing roles of glutaminase isoforms in determining glioblastoma cell phenotype. Neurochem Int. 2015;88:6–9.

    CAS  PubMed  Google Scholar 

  18. Liu J, Zhang C, Lin M, Zhu W, Liang Y, Hong X. Glutaminase 2 negatively regulates the PI3K / AKT signaling and shows tumor suppression activity in human hepatocellular carcinoma. Oncotarget. 2014;5:2635–47.

    PubMed  PubMed Central  Google Scholar 

  19. Lee SY, Jeon HM, Ju MK, Jeong EK, Kim CH, Park HG, et al. Dlx-2 and glutaminase upregulate epithelial-mesenchymal transition and glycolytic switch. Oncotarget. 2016;7:7925–39.

    PubMed  PubMed Central  Google Scholar 

  20. Zhang C, Liu J, Zhao Y, Yue X, Zhu Y, Wang X, et al. Glutaminase 2 is a novel negative regulator of small GTPase Rac1 and mediates p53 function in suppressing metastasis. Elife. 2016;5:1–20.

    Google Scholar 

  21. Kuo T, Chen C, Hua K, Yu P, Lee W. Glutaminase 2 stabilizes Dicer to repress Snail and metastasis in hepatocellular carcinoma cells. Cancer Lett. 2016;383:1–13.

    Google Scholar 

  22. Xiao D, Ren P, Su H, Yue M, Xiu R, Hu Y, et al. Myc promotes glutaminolysis in human neuroblastoma through direct activation of glutaminase 2. Oncotarget. 2015;6:40655–66.

    PubMed  PubMed Central  Google Scholar 

  23. Lehmann BD, Bauer JA, Chen X, Sanders ME, Chakravarthy AB, Shyr Y, et al. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J Clin Investig. 2011;121:2750–67.

    CAS  PubMed  Google Scholar 

  24. Lampa M, Arlt H, He T, Ospina B, Reeves J, Zhang B, et al. Glutaminase is essential for the growth of triple-negative breast cancer cells with a deregulated glutamine metabolism pathway and its suppression synergizes with mTOR inhibition. PLoS ONE. 2017;12. https://doi.org/10.1371/journal.pone.0185092.

    PubMed  PubMed Central  Google Scholar 

  25. Ubuka T, Meister A. Studies on the utilization of asparagine by mouse leukemia cells. J Natl Cancer Inst. 1971;46:291–8.

    CAS  PubMed  Google Scholar 

  26. Lorenzi PL, Llamas J, Gunsior M, Ozbun L, Reinhold WC, Varma S, et al. Asparagine synthetase is a predictive biomarker of L-asparaginase activity in ovarian cancer cell lines. Mol Cancer Ther. 2008;7:3123–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Panosyan EH, Wang Y, Xia P, Lee W-NP, Pak Y, Laks DR, et al. Asparagine depletion potentiates the cytotoxic effect of chemotherapy against brain tumors. Mol Cancer Res. 2014;12:694–702.

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Sircar K, Huang H, Hu L, Cogdell D, Dhillon J, Tzelepi V, et al. Integrative molecular profiling reveals asparagine synthetase is a target in castration-resistant prostate cancer. Am J Pathol. 2012;180:895–903.

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Katt WP, Lukey MJ, Cerione RA. A tale of two glutaminases: homologous enzymes with distinct roles in tumorigenesis. Future Med Chem. 2017;9:223–43.

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2:401–4.

    Google Scholar 

  31. Jemal A, Siegel R, Xu J, Ward E. Cancer statistics, 2010. CA Cancer J Clin. 2010;60:277–300.

    PubMed  Google Scholar 

  32. Liu C-Y, Lin H-H, Tang M-J, Wang Y-K. Vimentin contributes to epithelial-mesenchymal transition cancer cell mechanics by mediating cytoskeletal organization and focal adhesion maturation. Oncotarget. 2015;6:15966–83.

    PubMed  PubMed Central  Google Scholar 

  33. Shin S, Dimitri CA, Yoon S, Dowdle W, Blenis J. ERK2 but not ERK1, induces epithelial to mesenchymal transformation via DEF motif dependent signaling events. Mol Cell. 2010;38:114–27.

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Jang MH, Kim HJ, Kim EJ, Chung YR, Park SY. Expression of epithelial-mesenchymal transition–related markers in triple-negative breast cancer: ZEB1 as a potential biomarker for poor clinical outcome. Hum Pathol. 2015;46:1267–74.

    CAS  PubMed  Google Scholar 

  35. Shirakihara T, Saitoh M, Miyazono K. Differential regulation of epithelial and mesenchymal markers by δEF1 proteins in epithelial–mesenchymal transition induced by TGF-β. Mol Biol Cell. 2007;18:3533–44.

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Hollestelle A, Elstrodt F, Nagel JHA, Kallemeijn WW, Schutte M. Phosphatidylinositol-3-OH Kinase or RAS Pathway Mutations in Human Breast Cancer Cell Lines. Mol Cancer Res. 2007;5:195–201.

    CAS  PubMed  Google Scholar 

  37. Sommers CL, Byers SW, Thompson EW, Torri JA, Gelmann ER. Differentiation state and invasiveness of human breast cancer cell lines. Breast Cancer Res Treat. 1994;31:325–35.

    CAS  PubMed  Google Scholar 

  38. Davis FM, Parsonage MT, Cabot PJ, Parat M, Thompson EW. Assessment of gene expression of intracellular calcium channels, pumps and exchangers with epidermal growth factor-induced epithelial-mesenchymal transition in a breast cancer cell line. Cancer Cell Int. 2013;13:1.

    Google Scholar 

  39. Xu Q, Zhang Q, Ishida Y, Hajjar S, Tang X, Shi H, et al. EGF induces epithelial-mesenchymal transition and cancer stem-like cell properties in human oral cancer cells via promoting warburg effect. Oncotarget. 2016;8:9557–71.

    PubMed Central  Google Scholar 

  40. Kim J, Kong J, Chang H, Kim H, Kim A. EGF induces epithelial-mesenchymal transition through phospho-Smad2/3-Snail signaling pathway in breast cancer cells. Oncotarget. 2016;7:85021–32.

    PubMed  PubMed Central  Google Scholar 

  41. Sánchez-Tilló E, Siles L, de Barrios O, Cuatrecasas M, Vaquero EC, Castells A, et al. Expanding roles of ZEB factors in tumorigenesis and tumor progression. Am J Cancer Res. 2011;1:897–912.

    PubMed  PubMed Central  Google Scholar 

  42. McCubrey JA, Steelman LS, Chappell WH, Abrams SL, Wong EWT, Chang F, et al. Roles of the Raf/MEK/ERK pathway in cell growth, malignant transformation and drug resistance. Biochim Biophys Acta. 2007;1773:1263–84.

    CAS  PubMed  Google Scholar 

  43. Zacharias NM, McCullough C, Shanmugavelandy S, Lee J, Lee Y, Dutta P, et al. Metabolic differences in glutamine utilization lead to metabolic vulnerabilities in prostate cancer. Sci Rep. 2017;7:1–11.

    CAS  Google Scholar 

  44. Wellen KE, Lu C, Mancuso A, Lemons JMS, Ryczko M, Dennis JW, et al. The hexosamine biosynthetic pathway couples growth factor-induced glutamine uptake to glucose metabolism. Genes Dev. 2010;24:2784–99.

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Billin aN, Eilers aL, Coulter KL, Logan JS, Ayer DE. MondoA, a novel basic helix-loop-helix-leucine zipper transcriptional activator that constitutes a positive branch of a max-like network. Mol Cell Biol. 2000;20:8845–54.

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Pavlova NN, Hui S, Ghergurovich JM, Fan J, Intlekofer AM, White RM, et al. As extracellular glutamine levels decline, asparagine becomes an essential amino acid. Cell Metab. 2018;27:428–38.

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Koh LW-H, Koh GR-H, Ng FS-L, Toh TB, Sandanaraj E, Chong YK, et al. A distinct reactive oxygen species profile confers chemoresistance in glioma-propagating cells and associates with patient survival outcome. Antioxid Redox Signal. 2013;19:2261–79.

    CAS  PubMed  Google Scholar 

  48. Nitta M, Kozono D, Kennedy R, Stommel J, Ng K, Zinn PO, et al. Targeting EGFR induced oxidative stress by PARP1 inhibition in glioblastoma therapy. PLoS ONE. 2010;5:1–9.

    Google Scholar 

  49. de Guzzi Cassago CA, Dias MM, Pinheiro MP, Pasquali CC, Bastos ACS, Islam Z, et al. Glutaminase affects the transcriptional activity of peroxisome proliferator-activated receptor gamma (PPARγ) via direct interaction. Biochemistry. 2018;acs.biochem.8b00773.

  50. Bastide C, Bagnis C, Mannoni P, Hassoun J, Bladou F. A Nod Scid mouse model to study human prostate cancer. Prostate Cancer Prostatic Dis. 2002;5:311–5.

    CAS  PubMed  Google Scholar 

  51. Kim RK, Suh Y, Yoo KC, Cui YH, Kim H, Kim MJ, et al. Activation of KRAS promotes the mesenchymal features of basal-type breast cancer. Exp Mol Med. 2015;47:e137–9.

    PubMed  PubMed Central  Google Scholar 

  52. Lee Y-Z, Yang C, Chang HH-S, Hsu H, Qiu Y, Chao Y-S, et al. Discovery of selective inhibitors of Glutaminase-2, which inhibit mTORC1, activate autophagy and inhibit proliferation in cancer cells. Oncotarget. 2014;5:6087–101.

    PubMed  PubMed Central  Google Scholar 

  53. Xu D, Takeshita F, Hino Y, Fukunaga S, Kudo Y, Tamaki A, et al. miR-22 represses cancer progression by inducing cellular senescence. J Cell Biol. 2011;193:409–24.

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by the São Paulo State Research Foundation, FAPESP, under grants 2012/14298-9 (ALBA) and 2014/20673-2 (ALBA), 2014/15968-3 (SMGD), 2015/25832-4 (SMGD) and fellowships 2013/05668-0 (IMF) 2013/23510-4 (CFRA), 2012/11577-4 (MMD), 2014/18061-9 (LMR), 2014/17820-3 (DAM), 2016/06625-0 (ACPM), 2014/06512-6 (KRSO), 2011/10127-2 (CAGC), and 2012/09452-9 (MQE). We thank LNBio for financial support and access to all facilities (PBQT, LCCMI, LPP, LEC, LVV, and LIB). Data used in this publication were generated by the National Cancer Institute Clinical Proteomic Tumor Analysis Consortium (CPTAC). The results published here are in whole or part based upon data generated by the TCGA Research Network: http://cancergenome.nih.gov/. We thank Dr Alessandra Girasole for expert technical support. GAC is the Felix L. Haas Endowed Professor in Basic Science. Work in GAC’s laboratory is supported by National Institutes of Health (NIH/NCATS) grant UH3TR00943-01 through the NIH Common Fund, Office of Strategic Coordination (OSC), the NCI grants 1R01 CA182905-01 and 1R01CA222007-01A1, an NIGMS 1R01GM122775-01 grant, a U54 grant #CA096297/CA096300 – UPR/MDACC Partnership for Excellence in Cancer Research 2016 Pilot Project, a Team DOD (CA160445P1) grant, a Chronic Lymphocytic Leukemia Moonshot Flagship project, a Sister Institution Network Fund (SINF) 2017 grant, and the Estate of C. G. Johnson, Jr.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Andre Luis Berteli Ambrosio or Sandra Martha Gomes Dias.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Additional information

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

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dias, M.M., Adamoski, D., dos Reis, L.M. et al. GLS2 is protumorigenic in breast cancers. Oncogene 39, 690–702 (2020). https://doi.org/10.1038/s41388-019-1007-z

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41388-019-1007-z

Search

Quick links