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:

Glutamine synthetase activity fuels nucleotide biosynthesis and supports growth of glutamine-restricted glioblastoma

Abstract

L-Glutamine (Gln) functions physiologically to balance the carbon and nitrogen requirements of tissues. It has been proposed that in cancer cells undergoing aerobic glycolysis, accelerated anabolism is sustained by Gln-derived carbons, which replenish the tricarboxylic acid (TCA) cycle (anaplerosis). However, it is shown here that in glioblastoma (GBM) cells, almost half of the Gln-derived glutamate (Glu) is secreted and does not enter the TCA cycle, and that inhibiting glutaminolysis does not affect cell proliferation. Moreover, Gln-starved cells are not rescued by TCA cycle replenishment. Instead, the conversion of Glu to Gln by glutamine synthetase (GS; cataplerosis) confers Gln prototrophy, and fuels de novo purine biosynthesis. In both orthotopic GBM models and in patients, 13C–glucose tracing showed that GS produces Gln from TCA-cycle-derived carbons. Finally, the Gln required for the growth of GBM tumours is contributed only marginally by the circulation, and is mainly either autonomously synthesized by GS-positive glioma cells, or supplied by astrocytes.

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: Gln starvation reduces GBM cell proliferation.
Figure 2: The effects of Glu secretion and GLS inhibition on GBM cell growth and metabolism.
Figure 3: GS sustains cell growth during Gln starvation.
Figure 4: GS activity regulates cell growth and purine availability under Gln starvation.
Figure 5: Gln metabolism in differentiated and GBM stem-like (GSC) primary human GBM cells.
Figure 6: Gln metabolism in GBM patients and primary orthotopic xenografts.
Figure 7: Glutamine supply for GBM tumours with low GS expression.
Figure 8: Astrocytes provide GBM cells with Gln.

Similar content being viewed by others

References

  1. Moreadith, R. W. & Lehninger, A. L. The pathways of glutamate and glutamine oxidation by tumor cell mitochondria. Role of mitochondrial NAD(P) + -dependent malic enzyme. J. Biol. Chem. 259, 6215–6221 (1984).

    CAS  PubMed  Google Scholar 

  2. Yuneva, M. O. et al. The metabolic profile of tumors depends on both the responsible genetic lesion and tissue type. Cell Metab. 15, 157–170 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Wise, D. R. et al. Myc regulates a transcriptional program that stimulates mitochondrial glutaminolysis and leads to glutamine addiction. Proc. Natl Acad. Sci. USA 105, 18782–18787 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. DeBerardinis, R. J. et al. Beyond aerobic glycolysis: transformed cells can engage in glutamine metabolism that exceeds the requirement for protein and nucleotide synthesis. Proc. Natl Acad. Sci. USA 104, 19345–19350 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Tardito, S. et al. L-Asparaginase and inhibitors of glutamine synthetase disclose glutamine addiction of β-catenin-mutated human hepatocellular carcinoma cells. Curr. Cancer Drug Targets 11, 929–943 (2011).

    Article  CAS  PubMed  Google Scholar 

  6. Son, J. et al. Glutamine supports pancreatic cancer growth through a KRAS-regulated metabolic pathway. Nature 496, 101–105 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Gameiro, P. A. et al. In vivo HIF-mediated reductive carboxylation is regulated by citrate levels and sensitizes VHL-deficient cells to glutamine deprivation. Cell Metab. 17, 372–385 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Chiu, M. et al. Glutamine depletion by crisantaspase hinders the growth of human hepatocellular carcinoma xenografts. Br. J. Cancer 111, 1159–1167 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Wang, J. B. et al. Targeting mitochondrial glutaminase activity inhibits oncogenic transformation. Cancer Cell 18, 207–219 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Suzuki, S. et al. Phosphate-activated glutaminase (GLS2), a p53-inducible regulator of glutamine metabolism and reactive oxygen species. Proc. Natl Acad. Sci. USA 107, 7461–7466 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Dal Bello, B. et al. Glutamine synthetase immunostaining correlates with pathologic features of hepatocellular carcinoma and better survival after radiofrequency thermal ablation. Clin. Cancer Res. 16, 2157–2166 (2010).

    Article  CAS  PubMed  Google Scholar 

  13. Krebs, H. A. Metabolism of amino-acids: the synthesis of glutamine from glutamic acid and ammonia, and the enzymic hydrolysis of glutamine in animal tissues. Biochem. J. 29, 1951–1969 (1935).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. van den Berg, C. J. & Garfinkel, D. A stimulation study of brain compartments. Metabolism of glutamate and related substances in mouse brain. Biochem. J. 123, 211–218 (1971).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Takano, T. et al. Glutamate release promotes growth of malignant gliomas. Nat. Med. 7, 1010–1015 (2001).

    Article  CAS  PubMed  Google Scholar 

  16. Scott, D. A. et al. Comparative metabolic flux profiling of melanoma cell lines: beyond the Warburg effect. J. Biol. Chem. 286, 42626–42634 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Metallo, C. M. et al. Reductive glutamine metabolism by IDH1 mediates lipogenesis under hypoxia. Nature 481, 380–384 (2012).

    Article  CAS  Google Scholar 

  18. Kamphorst, J. J. et al. Hypoxic and Ras-transformed cells support growth by scavenging unsaturated fatty acids from lysophospholipids. Proc. Natl Acad. Sci. USA 110, 8882–8887 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Fan, J., Kamphorst, J. J., Rabinowitz, J. D. & Shlomi, T. Fatty acid labeling from glutamine in hypoxia can be explained by isotope exchange without net reductive isocitrate dehydrogenase (IDH) flux. J. Biol. Chem. 288, 31363–31369 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Mullen, A. R. et al. Reductive carboxylation supports growth in tumour cells with defective mitochondria. Nature 481, 385–388 (2012).

    Article  CAS  Google Scholar 

  21. Lewerenz, J. et al. The cystine/glutamate antiporter system x(c)(-) in health and disease: from molecular mechanisms to novel therapeutic opportunities. Antioxid. Redox Signal. 18, 522–555 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Duarte, N. C. et al. Global reconstruction of the human metabolic network based on genomic and bibliomic data. Proc. Natl Acad. Sci. USA 104, 1777–1782 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Gao, P. et al. c-Myc suppression of miR-23a/b enhances mitochondrial glutaminase expression and glutamine metabolism. Nature 458, 762–765 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Yuneva, M., Zamboni, N., Oefner, P., Sachidanandam, R. & Lazebnik, Y. Deficiency in glutamine but not glucose induces MYC-dependent apoptosis in human cells. J. Cell Biol. 178, 93–105 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Bax, D. A. et al. Molecular and phenotypic characterisation of paediatric glioma cell lines as models for preclinical drug development. PLoS ONE 4, e5209 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Filonov, G. S. et al. Bright and stable near-infrared fluorescent protein for in vivo imaging. Nat. Biotechnol. 29, 757–761 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Hock, A. K. et al. iRFP is a sensitive marker for cell number and tumor growth in high-throughput systems. Cell Cycle 13, 220–226 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Yin, Y. et al. Glutamine synthetase functions as a negative growth regulator in glioma. J. Neurooncol. 114, 59–69 (2013).

    Article  CAS  PubMed  Google Scholar 

  29. Reznik, E., Mehta, P. & Segre, D. Flux imbalance analysis and the sensitivity of cellular growth to changes in metabolite pools. PLoS Comput. Biol. 9, e1003195 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Calise, S. J. et al. Glutamine deprivation initiates reversible assembly of mammalian rods and rings. Cell. Mol. Life Sci. 71, 2963–2973 (2014).

    Article  CAS  PubMed  Google Scholar 

  31. Carruthers, R. et al. Abrogation of radioresistance in glioblastoma stem-like cells by inhibition of ATM kinase. Mol. Oncol. 9, 192–203 (2015).

    Article  CAS  PubMed  Google Scholar 

  32. Meyerand, M. E., Pipas, J. M., Mamourian, A., Tosteson, T. D. & Dunn, J. F. Classification of biopsy-confirmed brain tumors using single-voxel MR spectroscopy. AJNR Am. J. Neuroradiol. 20, 117–123 (1999).

    CAS  PubMed  Google Scholar 

  33. Parmentier, J. H. et al. Glutaminase activity determines cytotoxicity of L-asparaginases on most leukemia cell lines. Leuk. Res. 39, 757–762 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Had-Aissouni, L. Toward a new role for plasma membrane sodium-dependent glutamate transporters of astrocytes: maintenance of antioxidant defenses beyond extracellular glutamate clearance. Amino Acids 42, 181–197 (2012).

    Article  CAS  PubMed  Google Scholar 

  35. Rosati, A. et al. Glutamine synthetase expression as a valuable marker of epilepsy and longer survival in newly diagnosed glioblastoma multiforme. Neuro-oncol. 15, 618–625 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Marin-Valencia, I. et al. Analysis of tumor metabolism reveals mitochondrial glucose oxidation in genetically diverse human glioblastomas in the mouse brain in vivo. Cell Metab. 15, 827–837 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Maher, E. A. et al. Metabolism of [U-13 C]glucose in human brain tumors in vivo. NMR Biomed. 25, 1234–1244 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Bagga, P. et al. Characterization of cerebral glutamine uptake from blood in the mouse brain: implications for metabolic modeling of 13C NMR data. J. Cereb. Blood Flow Metab. 34, 1666–1672 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Rosati, A. et al. Epilepsy in glioblastoma multiforme: correlation with glutamine synthetase levels. J. Neurooncol. 93, 319–324 (2009).

    Article  CAS  PubMed  Google Scholar 

  40. Noble, M. & Murray, K. Purified astrocytes promote the in vitro division of a bipotential glial progenitor cell. EMBO J. 3, 2243–2247 (1984).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Keunen, O. et al. Anti-VEGF treatment reduces blood supply and increases tumor cell invasion in glioblastoma. Proc. Natl Acad. Sci. USA 108, 3749–3754 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Sanzey, M. et al. Comprehensive analysis of glycolytic enzymes as therapeutic targets in the treatment of glioblastoma. PLoS ONE 10, e0123544 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This study has been supported by Cancer Research UK. S.T. is a recipient of an AIRC/Marie Curie International Fellowship for Cancer Research. The human and animal metabolomic studies were supported by The Norwegian Cancer Society, The Norwegian Research Council, Helse Vest, Haukeland University Hospital and the K.G-Jebsen Foundation. We acknowledge A. Golebiewska, V. Baus-Talko, N. Van Den Broek, G. MacKay, C. Nixon and E. MacKenzie for excellent technical assistance and A. King for excellent editorial work.

Author information

Authors and Affiliations

Authors

Contributions

S.T. conceived the study, designed and performed most experiments, interpreted the data, and wrote the manuscript, A.O. performed the experiments in orthotopic xenograft models, S.U.A. and A.J.C. provided the differentiated and stem-like primary glioblastoma cells, L.Z. supervised the analysis of LC-MS samples, O.K. performed the MRI analysis, F.F. processed the orthotopic and clinical GBM samples, H.M. provided the tissue microarray, A.K.H. designed and provided the iRFP and iRFP–GS constructs, A.Weinstock, A.Wagner and E.R. generated and employed the metabolic modelling, S.C.B. and S.L.L. provided the primary astrocytes, M.L.-J., S.H.M. and P.Ø.S. provided the surgical specimens from the patients, S.P.N. and R.B. conceived and supervised the experiments in orthotopic models and human patients, and E.G. conceived and supervised the study, interpreted the data, and revised the manuscript.

Corresponding author

Correspondence to Eyal Gottlieb.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 5 Glutamine starvation reduces GBM cell proliferation.

(a) Representative microscopic fields of cells incubated for 6 days with or without Gln. (b) Cell cycle distribution of cells incubated for 3 days with or without Gln. Mean ± S.E.M. n = 3 independent experiments. p value refers to a two-tailed t test for unpaired samples. Raw data of independent repeats are provided in the statistics source data Supplementary Table 5.

Supplementary Figure 6 Exchange rates of metabolites in GBM cell lines.

Cells were incubated for 24 h +/− U-13C5 Gln, secretion rates (positive bars) and consumption rates (negative bars) of the indicated metabolites are shown. The sum of all isotopologues is reported for clarity. Alanine (Ala) exchange rates are magnified in the inset. Mean ± S.E.M. n = 3 independent experiments.

Supplementary Figure 7 Effects of Gln availability and Glutaminase inhibition on GBM cell lines growth and redox state.

(a) Correlation between Gln consumption and growth inhibition caused by Gln starvation is shown in six cell lines. Mean ± S.E.M. n = 3 independent experiments. (b) Citrate isotopologues distribution of 13C5-Gln derived atoms. 13C6-Citrate was not detected. Mean ± S.E.M. n = 3 independent experiments. (c) Oxidized (GSSG) to reduced (GSH) glutathione ratio. LN18 cells were incubated for 24 h +/− Gln and supplemented with Glu, α-ketoglutarate dimethylester (dm-αKG), sulfasalazine (SSZ), or cystine, as indicated. GSSG/GSH ratio was assessed by HPLC-MS, and shown as % of untreated control. Data derive from one experiment performed once. Raw data of independent repeats are provided in the statistics source data Supplementary Table 5. (di) Cells were incubated with 0, 2.5, 5, 10, 15, 30 μM BPTES for 72 h, in the absence (solid line) or presence (dotted line) of 4 mM dm-αKG and counted. In all conditions cells were exposed to 0.3%DMSO. Mean ± S.E.M. n = 3 independent experiments.

Supplementary Figure 8 GS expression and its effects on colony formation capacity, and glucosamine biosynthetic pathway in GBM cells.

(a) Cells were incubated for 24h +/− Gln and GLUL (GS), GLS, and MYC mRNA relative expression was assessed by qPCR. Actin expression was used for normalization. Data derive from one experiment performed once. Raw data of independent repeats are provided in the statistics source data Supplementary Table 5. (b) SF188 and U251 cells stably expressing a non-targeting control shRNA (shNTC) and two sequences targeting GS (shGS-1 and shGS-2) were cultured for 12–17 days +/− Gln in medium supplemented with glutamate (4 mM), and ammonia (0.8 mM) as indicated. The quantification of colonies surface area was obtained as described in the Methods section. Mean ± S.E.M., n = 4 independent experiments. (c) iRFP4 and iRFP-GS5 expressing LN18 cells were incubated +/− Gln for 24 h in medium supplemented with 0.8 mM 15NH4+. The intracellular isotopologues distribution of UDP N-acetylglucosamine is shown as % of control (value obtained for the metabolites in iRFP4 cells + Gln). Data derive from one experiment performed twice. Raw data of independent repeats are provided in the statistics source data Supplementary Table 5.

Supplementary Figure 9 Heterogeneous GS expression in human GBM biopsies.

The whole bioptic tissues of three patients included in the tissue microarray were stained for GS, and Hematoxilin and Eosin [H&E], as indicated. Low magnifications of whole biopsies are reported in panels (a,b,h,i,n,o). For each biopsy, tissue regions framed in blue and green are magnified in dg,jm,qt. Biopsy 1 shows necrotic tissue [nec] surrounded by a solid tumour core [TC] with uniformly low GS staining (d,f). (d,e) Invasive tumour areas [TI] show infiltrative cells with heterogeneous immunoreactivity for GS. (c) Normal astrocytes [black arrows] in the adjacent normal brain [Br] were positive for GS, while neurons [white arrows] were GS-negative. Biopsy 2 shows distinct areas with low/absent (k) and high (j) immunoreactivity for GS. GS-negative inflammatory tissue [i] surrounding a necrotic region [nec] is evident in panels h,i,km. Biopsy 3 shows distinct areas with low/absent (q) and high (r) immunoreactivity for GS. (p) GS-positive reactive astrocytes [arrows] with extensive cellular processes forming a network of GS positive fibers surrounding GS-negative tumour cells.

Supplementary Figure 10 Glucose and glutamine metabolic fates in GBM patients, and orthotopic mouse models.

(a) Serum levels of 13C6 and 13C0 glucose in three GBM patients injected with 13C6 glucose before surgical intervention for tumour removal. The dashed line represents the time at which glucose infusion starts. Black arrow points at the time of surgical resection. Values are reported as % of the basal values obtained before infusion. (b) 13C Lactate and (c) 13C Glutamate enrichment in serum at time of tumour resection, in tumour tissue, and in adjacent edematous tissue, of seven (1-7) GBM patients injected with 13C6-glucose. The % of lactate or glutamate incorporating one or more 13C carbons over the total amount of lactate or glutamate detected is reported; na: not available, nd: not detectable. Values were corrected for the natural abundance of 13C. (d,e) Isotopologue distribution of metabolites obtained in the liver of mice orthotopically xenografted with human P3 GBM and injected with 13C6 Glucose (d) or 13C5-Gln (e). The values are mean ± S.E.M. n = 3 mice. (f,g) Isotopologues of Gln (f) and glucose (g) found in the serum of mice orthotopically xenografted with the human GS-positive T16 GBM, and infused for 4h into the carotid artery with 13C5-Gln. Mean ± S.E.M. n = 3 mice. (hj) Isotopologue distributions of Gln, Glu and α-kg in T16 tumours, contralateral brain, and liver tissues of mice infused as in (f,g). Mean ± S.E.M. n = 3 mice. (k) Astrocytes were incubated for 24 hours +/− Gln, and asparagine consumption is reported. Data derive from one experiment performed twice. Raw data of independent repeats are provided in the statistics source data Supplementary Table 5.

Supplementary Figure 11 A toy network used for metabolic modelling.

A toy network (a) and its optimal flux distribution (b).

Supplementary Figure 12 Unprocessed scans of western blots accompanied by size markers.

Red dotted lines delineate the region presented in the respective Figures as indicated. Images were obtained using a Licor Odyssey scanner and acquired using Image Studio 2.0.

Supplementary Table 1 Formulation of DMEM and Serum-like Modified Eagles Medium (SMEM).
Supplementary Table 2 Exchange rates of metabolites in GBM cell lines.
Supplementary Table 3 Representation of SMEM growth medium in the metabolic model Recon1.
Supplementary Table 4 Weighted costs of growth limiting metabolites.

Supplementary information

Supplementary Information

Supplementary Information (PDF 2806 kb)

Supplementary Table 5

Supplementary Information (XLSX 130 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tardito, S., Oudin, A., Ahmed, S. et al. Glutamine synthetase activity fuels nucleotide biosynthesis and supports growth of glutamine-restricted glioblastoma. Nat Cell Biol 17, 1556–1568 (2015). https://doi.org/10.1038/ncb3272

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3272

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