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:

Metabolic adaptation of acute lymphoblastic leukemia to the central nervous system microenvironment depends on stearoyl-CoA desaturase

Abstract

Metabolic reprogramming is a key hallmark of cancer, but less is known about metabolic plasticity of the same tumor at different sites. Here, we investigated the metabolic adaptation of leukemia in two different microenvironments, the bone marrow and the central nervous system (CNS). We identified a metabolic signature of fatty acid synthesis in CNS leukemia, highlighting stearoyl-CoA desaturase (SCD) as a key player. In vivo SCD overexpression increases CNS disease, whereas genetic or pharmacological inhibition of SCD decreases CNS load. Overall, we demonstrated that leukemic cells dynamically rewire metabolic pathways to suit local conditions and that targeting these adaptations can be exploited therapeutically.

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: Fatty acid synthesis-related genes are upregulated in CNS-derived ALL cells in xenograft models.
Fig. 2: SCD is upregulated in primary patient samples in the CNS.
Fig. 3: SCD overexpression confirms a competitive advantage for SCD-high cells in the CNS microenvironment.
Fig. 4: SCD ablation decreases 018z cells engraftment in the CNS.
Fig. 5: SCD pharmacological inhibition decreases 018z cells engraftment in the CNS.
Fig. 6: SCD pharmacological inhibition decreases patient-derived xenograft engraftment in the CNS.

Similar content being viewed by others

Data availability

RNA-seq data supporting this study’s findings have been deposited in GEO (accession number: GSE135115). The GSE135115 SuperSeries is entitled “Gene expression profiles of MLL-AF4 and TEL-AML1 acute lymphoblastic leukemia blasts retrieved from central nervous system and spleen”. This SuperSeries contains two series related to SEM and REH experiments as follows: GSE135113 “Gene expression profiles of MLL-AF4 acute lymphoblastic leukemia blasts retrieved from central nervous system and spleen” and GSE135114 “Gene expression profiles of TEL-AML1 acute lymphoblastic leukemia blasts retrieved from central nervous system and spleen”. Previously published human and primograft data re-analyzed here are available under accession codes GSE60926 and GSE89710. The source data associated with each figure are provided with the manuscript. All other data supporting the findings of this study are available from the corresponding author on reasonable request. Source data are provided with this paper.

References

  1. Cairns, R. A., Harris, I. S. & Mak, T. W. Regulation of cancer cell metabolism. Nat. Rev. Cancer 11, 85–95 (2011).

    CAS  PubMed  Google Scholar 

  2. DeBerardinis, R. J. & Chandel, N. S. Fundamentals of cancer metabolism. Sci. Adv. 2, 1–18 (2016).

    Google Scholar 

  3. Cha, J.-Y. & Lee, H.-J. Targeting lipid metabolic reprogramming as anticancer therapeutics. J. Cancer Prev. 21, 209–215 (2017).

    Google Scholar 

  4. Gisselbrecht, C. Positron emission tomography – Guided therapy of aggressive non-Hodgkin lymphoma: Standard of care after the PETAL study? J. Clin. Oncol. 36, 3272–3273 (2018).

    CAS  Google Scholar 

  5. Caro, P. et al. Metabolic signatures uncover distinct targets in molecular subsets of diffuse large B-cell lymphoma. Cancer Cell 22, 547–560 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Kuntz, E. M. et al. Targeting mitochondrial oxidative phosphorylation eradicates therapy-resistant chronic myeloid leukemia stem cells. Nat. Med. 23, 1234–1240 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Nachmias, B. & Schimmer, A. D. Metabolic flexibility in Leukemia—adapt or die. Cancer Cell 34, 695–696 (2018).

    CAS  PubMed  Google Scholar 

  8. Olivares, O., Däbritz, J. H. M., King, A., Gottlieb, E. & Halsey, C. Research into cancer metabolomics: towards a clinical metamorphosis. Semin. Cell Dev. Biol. 43, 52–64 (2015).

    PubMed  Google Scholar 

  9. Frishman-Levy, L. & Izraeli, S. Advances in understanding the pathogenesis of CNS acute lymphoblastic leukaemia and potential for therapy. Br. J. Haematol. 176, 157–167 (2017).

    PubMed  Google Scholar 

  10. Pui, C. H. & Howard, S. C. Current management and challenges of malignant disease in the CNS in paediatric leukaemia. Lancet Oncol. 9, 257–268 (2008).

    PubMed  Google Scholar 

  11. Halsey, C. et al. The impact of therapy for childhood acute lymphoblastic leukaemia on intelligence quotients; Results of the risk-stratified randomized central nervous system treatment trial MRC UKALL XI. J. Hematol. Oncol. 4, 1–12 (2011).

    Google Scholar 

  12. Iyer, N. S., Balsamo, L. M., Bracken, M. B. & Kadan-Lottick, N. S. Chemotherapy-only treatment effects on long-term neurocognitive functioning in childhood ALL survivors: a review and meta-analysis. Blood 126, 346–353 (2015).

    CAS  PubMed  Google Scholar 

  13. Price, R. A. & Johnson, W. W. The central nervous system in childhood leukemia: I. The arachnoid. Cancer 31, 520–533 (1973).

    CAS  PubMed  Google Scholar 

  14. Williams, M. T. S. et al. The ability to cross the blood-cerebrospinal fluid barrier is a generic property of acute lymphoblastic leukemia blasts. Blood 127, 1998–2006 (2016).

    CAS  PubMed  Google Scholar 

  15. Bartram, J. et al. High throughput sequencing in acute lymphoblastic leukemia reveals clonal architecture of central nervous system and bone marrow compartments. Haematologica 103, e110–e114 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Kato, I. et al. Hypoxic adaptation of leukemic cells infiltrating the CNS affords a therapeutic strategy targeting VEGFA. Blood 129, 3126–3129 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Buonamici, S. et al. CCR7 signalling as an essential regulator of CNS infiltration in T-cell leukaemia. Nature 459, 1000–1004 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Krause, S. et al. Mer tyrosine kinase promotes the survival of t(1;19)-positive acute lymphoblastic leukemia (ALL) in the central nervous system (CNS). Blood 125, 820–830 (2015).

    CAS  PubMed  Google Scholar 

  19. Cario, G. et al. High interleukin-15 expression characterizes childhood acute lymphoblastic leukemia with involvement of the CNS. J. Clin. Oncol. 25, 4813–4820 (2007).

    CAS  PubMed  Google Scholar 

  20. Williams, M. T. S. et al. Interleukin-15 enhances cellular proliferation and upregulates CNS homing molecules in pre-B acute lymphoblastic leukemia. Blood 123, 3116–3127 (2014).

    CAS  PubMed  Google Scholar 

  21. Frishman-Levy, L. et al. Central nervous system acute lymphoblastic leukemia: role of natural killer cells. Blood 125, 3420–3431 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Münch, V. et al. Central nervous system involvement in acute lymphoblastic leukemia is mediated by vascular endothelial growth factor. Blood 130, 643–654 (2017).

    PubMed  Google Scholar 

  23. Spector, R., Robert Snodgrass, S. & Johanson, C. E. A balanced view of the cerebrospinal fluid composition and functions: focus on adult humans. Exp. Neurol. 273, 57–68 (2015).

    CAS  PubMed  Google Scholar 

  24. Hühmer, A. F., Biringer, R. G., Amato, H., Fonteh, A. N. & Harrington, M. G. Protein analysis in human cerebrospinal fluid: physiological aspects, current progress and future challenges. Dis. Markers 22, 3–26 (2006).

    PubMed  Google Scholar 

  25. Damkier, H. H., Brown, P. D. & Praetorius, J. Cerebrospinal fluid secretion by the choroid plexus. Physiol. Rev. 93, 1847–1892 (2013).

    CAS  PubMed  Google Scholar 

  26. Méndez-Ferrer, S. et al. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature 466, 829–834 (2010).

    PubMed  PubMed Central  Google Scholar 

  27. Morrison, S. J. & Scadden, D. T. The bone marrow niche for haematopoietic stem cells. Nature 505, 327–334 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Olechnowicz, S. W. Z. & Edwards, C. M. Contributions of the host microenvironment to cancer-induced bone disease. Cancer Res. 74, 1625–1631 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Eckhoff, E. M., Queudeville, M., Debatin, K.-M. & Meyer, L. H. A novel B cell precursor ALL cell line (018Z) with prominent neurotropism and isolated CNS leukemia in a NOD/SCID/huALL xenotransplantation model. Blood 114, 1630–1630 (2009).

    Google Scholar 

  30. van der Velden, V. H. J. et al. New cellular markers at diagnosis are associated with isolated central nervous system relapse in paediatric B-cell precursor acute lymphoblastic leukaemia. Br. J. Haematol. 172, 769–781 (2016).

    PubMed  Google Scholar 

  31. Theodoropoulos, P. C. et al. Discovery of tumor-specific irreversible inhibitors of stearoyl-CoA desaturase. Nat. Chem. Biol. 12, 218–225 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  Google Scholar 

  33. Angelucci, C. et al. Pivotal role of human stearoyl-CoA desaturases (SCD1 and 5) in breast cancer progression: oleic acid-based effect of SCD1 on cell migration and a novel pro-cell survival role for SCD5. Oncotarget 9, 24364–24380 (2018).

    PubMed  PubMed Central  Google Scholar 

  34. Hess, D., Chisholm, J. W. & Igal, R. A. Inhibition of stearoyl-CoA desaturase activity blocks cell cycle progression and induces programmed cell death in lung cancer cells. PLoS ONE 5, e11394 (2010).

    PubMed  PubMed Central  Google Scholar 

  35. Wang, J. et al. High expression of stearoyl-CoA desaturase 1 predicts poor prognosis in patients with clear-cell renal cell carcinoma. PLoS ONE 11, e0166231 (2016).

    PubMed  PubMed Central  Google Scholar 

  36. Chen, L. et al. Stearoyl-CoA desaturase-1-mediated cell apoptosis in colorectal cancer by promoting ceramide synthesis. Sci. Rep. 6, 1–11 (2016).

    Google Scholar 

  37. Kim, S. J., Choi, H., Park, S. S., Chang, C. & Kim, E. Stearoyl-CoA desaturase (SCD) facilitates proliferation of prostate cancer cells through enhancement of androgen receptor transactivation. Mol. Cells 31, 371–377 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Zhang, H., Li, H., Ho, N., Li, D. & Li, S. Scd1 plays a tumor-suppressive role in survival of leukemia stem cells and the development of chronic myeloid leukemia. Mol. Cell Biol. 32, 1776–1787 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Southam, A. D. et al. Drug redeployment to kill leukemia and lymphoma cells by disrupting SCD1-mediated synthesis of monounsaturated fatty acids. Cancer Res. 75, 2530–2540 (2015).

    CAS  PubMed  Google Scholar 

  40. Imamura, K. et al. Discovery of novel and potent stearoyl coenzyme a desaturase 1 (SCD1) inhibitors as anticancer agents. Bioorganic Med. Chem. 25, 3768–3779 (2017).

    CAS  Google Scholar 

  41. Folger, O. et al. Predicting selective drug targets in cancer through metabolic networks. Mol. Syst. Biol. 7, 1–10 (2011).

    Google Scholar 

  42. Miyazaki, M., Man, W. C. & Ntambi, J. M. Targeted disruption of stearoyl-CoA desaturase1 gene in mice causes atrophy of sebaceous and meibomian glands and depletion of wax esters in the eyelid. J. Nutr. 131, 2260–2268 (2001).

    CAS  PubMed  Google Scholar 

  43. Brown, J. M. & Rudel, L. L. Stearoyl-coenzyme A desaturase 1 inhibition and the metabolic syndrome: considerations for future drug discovery. Curr. Opin. Lipidol. 21, 192–197 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Prieto, C. et al. NG2 antigen is involved in leukemia invasiveness and central nervous system infiltration in MLL-rearranged infant B-ALL. Leukemia 32, 633–644 (2018).

    CAS  PubMed  Google Scholar 

  45. Pieters, R. et al. Outcome of infants younger than 1 year with acute lymphoblastic leukemia treated with the interfant-06 protocol: results from an international phase III randomized study. J. Clin. Oncol. 37, 2246–2256 (2019).

    CAS  PubMed  Google Scholar 

  46. Vriens, K. et al. Evidence for an alternative fatty acid desaturation pathway increasing cancer plasticity. Nature 566, 403–406 (2019).

    PubMed  PubMed Central  Google Scholar 

  47. Ono, A. et al. Feedback activation of AMPK-mediated autophagy acceleration is a key resistance mechanism against SCD1 inhibitor-induced cell growth inhibition. PLoS ONE 12, e0181243 (2017).

    PubMed  PubMed Central  Google Scholar 

  48. Hagedorn, N. et al. Submicroscopic bone marrow involvement in isolated extramedullary relapses in childhood acute lymphoblastic leukemia: a more precise definition of “isolated” and its possible clinical implications, a collaborative study of the Resistant Disease Committee. Blood 110, 4022–4029 (2007).

    CAS  PubMed  Google Scholar 

  49. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Hensley, C. T. et al. Metabolic heterogeneity in human lung tumors. Cell 164, 681–694 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Kerr, E. M., Gaude, E., Turrell, F. K., Frezza, C. & Martins, C. P. Mutant Kras copy number defines metabolic reprogramming and therapeutic susceptibilities. Nature 531, 110–113 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Sciacovelli, M. & Frezza, C. Metabolic reprogramming and epithelial-to-mesenchymal transition in cancer. FEBS J. 284, 3132–3144 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Burrell, R. A., McGranahan, N., Bartek, J. & Swanton, C. The causes and consequences of genetic heterogeneity in cancer evolution. Nature 501, 338–345 (2013).

    CAS  PubMed  Google Scholar 

  54. Sanjana, N. E., Shalem, O. & Zhang, F. Improved vectors and genome-wide libraries for CRISPR screening. Nat. Methods 11, 783–784 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Ibrahimi, A. et al. Highly efficient multicistronic lentiviral vectors with peptide 2A sequences. Hum. Gene Ther. 20, 845–860 (2009).

    CAS  PubMed  Google Scholar 

  56. Agnese, S. T., Spierto, F. W. & Hannon, W. H. Evaluation of four reagents for delipidation of serum. Clin. Biochem. 16, 98–100 (1983).

    CAS  PubMed  Google Scholar 

  57. Tumanov, S. et al. Calibration curve-free GC–MS method for quantitation of amino and non-amino organic acids in biological samples. Metabolomics 12, 1–13 (2016).

    CAS  Google Scholar 

  58. Mackay, G. M., Niels, L. Z., Broek, J. F. van den & Gottlieb, E. in Metabolic Analysis Using Stable Isotopes (ed., Metallo, C. M.) 171–196 (Elsevier, 2015).

  59. Schmittgen, T. D. & Livak, K. J. Analyzing real-time PCR data by the comparative CT method. Nat. Protoc. 3, 1101–1108 (2008).

    CAS  PubMed  Google Scholar 

  60. Martin, M. & N, T. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet.journal 17, 10–12 (2011).

  61. Bray, N. L., Pimentel, H., Melsted, P. & Pachter, L. Near-optimal probabilistic RNA-seq quantification. Nat. Biotechnol. 34, 525–527 (2016).

    CAS  PubMed  Google Scholar 

  62. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 1–21 (2014).

    Google Scholar 

  63. Warnes, G. R. et al. gplots: Various R programming tools for plotting data. R package v.3.0-1. http://CRAN.R-project.org/package=gplots (2015).

Download references

Acknowledgements

We thank the patients and their families who generously donated the samples used in this study to the NHS Greater Glasgow and Clyde Biorepository, Laboratory Medicine Building, Queen Elizabeth University Hospital, the Bloodwise Childhood Leukemia Cell Bank, the Glasgow Neuroimmunology Biobank and the West of Scotland CSF Biobank. In addition, we thank J. Goodfellow, H. Willison, S. Bhatti and Y. Yousafzai for assistance with obtaining primary samples and C. Orange and L. Stevenson for help with histology. Histology slides were scanned by the University of Glasgow slide scanning and image analysis service at the Queen Elizabeth University Hospital. RNA-seq was performed by the Glasgow Polyomics research facility at the University of Glasgow. We also thank K. Keeshan and the Biological Services Unit, Cancer Research UK Beatson Institute for animal assistance. We thank G. Cazzaniga for supplying PDXs, V. Saha for providing reporter plasmids and L. Akimov, I. Muler, H. Fishman, A. Rein and E. Vax for technical assistance. This work was supported by the Chief Scientist Office (O.O. and C.H., grant ETM/374), Fondazione Italiana per la Ricerca sul Cancro *FIRC (A.M.S.), the William and Elizabeth Davies Foundation (A.C., Clinical Research Fellowship), the Laura and Ike Perlmutter Fund (E.G. and I.A.), the German Israel Foundation (S.I. and C.E.), the Norman and Sadie Lee Foundation (S.I.), the Israel Science Foundation 1775/12 (E.G. and I.M.), European Union ERA NET TRASCALL program (S.I.), Israel Cancer Research Foundation City of Hope collaborative program (S.I.) and Cancer Research UK (J.J.K. and G.M.). This project has received funding from the European Union’s Horizon 2020 Research and Innovation Programme under the Marie Skłodowska-Curie grant agreement META-CAN No 766214 (S.I.F., J.F-G., I.M. and E.G.).

Author information

Authors and Affiliations

Authors

Contributions

A.M.S., S.I.F., O.O., E.G., C.H., P.H. and S.I. designed the study. A.M.S., S.I.F., O.O., A.C., A.Z., S.B., I.G., L.F., Y.B., C.E., M.B., E.B. and S.J.R. provided the samples and performed most of the experiments. A.M.S., S.I.F., O.O., A.C., P.H., E.K.M., J.G.-F., C.H., I.M., J.R.C., M.G.K. and E.G. analyzed and interpreted the data. S.T., I.A., J.J.K. and G.M. performed and analyzed the MS experiments. A.M.S., S.I.F., O.O., C.H., E.G., I.M. and S.I. wrote the manuscript.

Corresponding authors

Correspondence to Eyal Gottlieb, Shai Izraeli or Christina Halsey.

Ethics declarations

Competing interests

E.G. is a board member and a shareholder of Metabomed Ltd., Israel, J.J.K. is an employee and shareholder of Rheos Medicines Inc. M.G.K. is a consultant for Accent Therapeutics and M.G.K.’s laboratory receives some financial support from 28-7. These disclosures are not directly related to these studies. All other 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.

Extended data

Extended Data Fig. 1 Leukemic infiltrates in murine CNS and bone marrow.

a, H&E staining of paraffin-embedded leukemic (ALL) murine skull and brain. ** indicates leukemic infiltrate in calvarial bone marrow, dashed line indicates leptomeningeal space filled with leukemic infiltrate b, H&E staining of paraffin embedded murine femur confirming widespread dense leukemic infiltrate throughout the marrow cavity. Images are representative of 8 mice.

Extended Data Fig. 2 Fatty acid synthesis-related genes are upregulated in ALL cells derived from the CNS of xenograft models.

a, Schematic of the RNA Sequencing workflow. Two batches of five NSG mice were xenografted with human ALL cell lines SEM [t(4;11) MLL-AFF1 (MLL-AF4)]. Post engraftment, cells were collected from central nervous system (CNS) and spleens. Before RNA extraction, CNS or spleen ALL cells from each batch were pooled to reach the quantities required for polyA-tailed RNA sequencing. After extraction, RNA was sequenced and analyzed as described in material and methods. b, Top 20 differentially expressed coding genes in CNS compared to spleen from RNA sequencing, excluding genes with a base mean <10, ranked according to their adjusted p-value in SEM (I) and REH (II) cells. Gene function was assigned using NCBI Gene and linked resources. The reported p-value(s) result from a two-sided DESeq2’s Wald test and were FDR-adjusted by the Benjamini-Hochberg procedure. c, I-II Enrichment plots of metabolism of lipids and lipoproteins (REACTOME) and oxidative phosphorylation (KEGG) for REH cell line extracted from the CNS and spleen of engrafted mice (n=2 groups of 5 mice each). Profile of the running ES score & Positions of the Gene Set Members on the Rank Ordered List. III Statistically significant biological functions in REH cells isolated from CNS and spleen of xenografted mice. p-values for positive association with a signature (enrichment) were calculated by permutation test. Plotted are signatures with significant fold-changes in enrichment between the CNS versus spleen groups (log2 scale). Red bars indicate signatures with positive log fold-change (gain) in CNS versus spleen, blue bars indicate negative log fold-change (loss) in CNS versus spleen samples.

Source data

Extended Data Fig. 3 Schematic of the fatty acid metabolism.

Glucose or glutamine-derived citrate or free Acetyl-CoA serve as precursor for saturated fatty acid, further un-saturated to provide either triglycerides or phospholipids. Saturated fatty acids can also enter cycle of degradation within the mitochondrion through beta-oxidation. ACLY: ATP- Citrate Lyase; FASN: Fatty Acid Synthase; SCD: Stearoyl-CoA Desaturase; ACC: Acetyl CoA Carboxylase; CPT: Carnitine Palmitoyltransferase; HMGCR: 3-Hydroxy-3-Methylglutaryl-CoA Reductase; SQLE: Squalene epoxidase; TCA: Tricarboxylic acid cycle; FA: Fatty acid; FAO: Fatty acid oxidation.

Extended Data Fig. 4 Quantitative PCR validation of top ranked genes differentially expressed in CNS blasts compared to spleen in SEM and REH cells.

a, and 018z cells (b) p (two tailed) = one sample T and Wilcoxon test. Results are normalized to 36B4 human housekeeping genes and presented as LogFold2 change enrichment of comparing CNS to spleen for the SEM-REH samples; human HPRT was used as housekeeping gene and enrichment of comparing CNS to BM was calculated for 018z. n=7 for SCD, FASN, ACLY, CPT1a; n=6 for CPT1b; n=5 for CPT2 in (a). n=12 for LDLR, HMGCR, FASN, ACLY, CPT1a, CPT1b; n=11 for SQLE; n=7 for SCD, ABCA1 in b, For box-and-whisker plots, boxes represent 25th and 75th percentiles, center lines indicate median values and whiskers represent minimum and maximum values. c, Western blot of SCD and FASN proteins in SEM cells retrieved from the CNS and spleen of mice (n=4 mice).

Source data

Extended Data Fig. 5 Analysis of available public human databases.

Left side of each panel: Boxplots showing single genes differentially regulated in samples of BM from patients at diagnosis (n=22) and relapse (n=20) and CNS samples at relapse (n=8) from public database GSE60926, unpaired analysis. Right side of each panel: Patient-derived xenograft samples established by transplantation of patient ALL cells onto NSG mice, single dots indicate paired bone marrow and CNS. Public database GSE89710. a, ABCA1: ATP-binding cassette transporter subfamily A member 1; b, ACC: Acetyl-CoA carboxylase; c, ACLY: ATP Citrate Lyase; d, CPT1A: Carnitine Palmitoyltransferase 1A; e, CPT1B: Carnitine Palmitoyltransferase 1B; f, CPT2: Carnitine Palmitoyltransferase 2; g, FASN: Fatty acid synthase; h, HMGCR: 3-Hydroxy-3-Methylglutaryl-CoA Reductase; i, LDLR: Low density lipoprotein receptor; j, SQLE: Squalene. FDR – false discovery rate. For box-and-whisker plots, boxes represent 25th and 75th percentiles, center lines indicate median values and whiskers represent minimum and maximum values.

Source data

Extended Data Fig. 6 Ratios of monounsaturated fatty acids to their saturated precursors.

Ratios of total levels of oleic/stearic acids in total fatty acids extracts in (a) SCD-high and (c) SCD-low 018z cells, in comparison to respective controls (CTL) (n=4 independent experiments for each condition). Ratios of relative levels of oleic/stearic acids in free fatty acids extracts in (b) 018z overexpressing or (d) downregulating SCD, comparatively to corresponding controls (CTL). n=5 independent experiments for each condition, p(two-tailed)=unpaired parametric Student’s t-test. Error bars represent mean ± s.d.

Source data

Extended Data Fig. 7 Increased SCD activity and expression upon genetic modification.

a, Ratio of relative level of oleic/stearic acids in free fatty acids extracts from cells isolated from CNS of mice engrafted with SCD overexpressing (SCD-high, n=3 mice) and control (CTL, n=4 mice) 018z cells. p(two-tailed)=unpaired parametric Student’s t-test. b, Gene expression level of SCD after overexpression in REH cells (n=2 independent experiments). Error bars represent mean ± s.d.

Source data

Extended Data Fig. 8 Effect of FBS delipidation on relative concentrations of metabolites, using fumed silica.

Relative levels of listed metabolites in lipidated (Lip) or delipidated (Delip) FBS – (a) stearic, (b) oleic, (c) palmitic, (d) palmitoleic, (e) arachidonic and (f) linoleic acids, (g) glucose, (h) lactate, (i) glutamine and (j) glutamate. Total fatty acids were extracted by saponification and the polar metabolites were extracted in 50% methanol, 30% acetonitrile, 20% water (“Reg extraction”).

Source data

Extended Data Fig. 9 Body weight of mice treated with SCD1 inhibitor.

Body weight variation of NSG mice transplanted with PDX (a) 1, (b) 2, (c) 3, or (d) 4, treated with SW203668 (Treated, n=5 mice) or vehicle (Control, n=5 mice). Error bars represent mean ± s.d.

Source data

Extended Data Fig. 10 Example of the gating strategy used for flow cytometry analysis.

(a) Gating for live cells. (b) Gating to exclude doublets and cell aggregates. (c) Identification of human and mouse CD45+ specific populations.

Supplementary information

Supplementary Tables 1–5

Reporting Summary

Supplementary Video

Phenotypic representation of SCD overexpression in ALL cells.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 3

Unprocessed western blots.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 4

Unprocessed western blots.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 4

Unprocessed western blots.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Savino, A.M., Fernandes, S.I., Olivares, O. et al. Metabolic adaptation of acute lymphoblastic leukemia to the central nervous system microenvironment depends on stearoyl-CoA desaturase. Nat Cancer 1, 998–1009 (2020). https://doi.org/10.1038/s43018-020-00115-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-020-00115-2

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