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:

Translational reprogramming marks adaptation to asparagine restriction in cancer

This article has been updated

Abstract

While amino acid restriction remains an attractive strategy for cancer therapy, metabolic adaptations limit its effectiveness. Here we demonstrate a role of translational reprogramming in the survival of asparagine-restricted cancer cells. Asparagine limitation in melanoma and pancreatic cancer cells activates receptor tyrosine kinase–MAPK signalling as part of a feedforward mechanism involving mammalian target of rapamycin complex 1 (mTORC1)-dependent increase in MAPK-interacting kinase 1 (MNK1) and eukaryotic translation initiation factor 4E (eIF4E), resulting in enhanced translation of activating transcription factor 4 (ATF4) mRNA. MAPK inhibition attenuates translational induction of ATF4 and the expression of its target asparagine synthetase (ASNS), sensitizing melanoma and pancreatic tumours to asparagine restriction, reflected in inhibition of their growth. Correspondingly, low ASNS expression is among the top predictors of response to inhibitors of MAPK signalling in patients with melanoma and is associated with favourable prognosis when combined with low MAPK signalling activity. These studies reveal an axis of adaptation to asparagine deprivation and present a rationale for clinical evaluation of MAPK inhibitors in combination with asparagine restriction approaches.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: ATF4 activity impedes growth suppression in response to asparagine limitation.
Fig. 2: In silico pan-tumour analysis predicts RTKs, BRAF, and MNK1 as ASNS SL partners.
Fig. 3: The MAPK pathway is required for ATF4 upregulation on asparagine limitation.
Fig. 4: ATF4 induction following ASNS suppression requires MAPK–mTORC1–eIF4E signalling.
Fig. 5: MNK1 is essential for ATF4 induction in response to asparagine limitation.
Fig. 6: ASNS suppression upregulates RTKs to induce MAPK signalling.
Fig. 7: Combined l-Aase treatment and MEK inhibition suppresses pancreatic and melanoma tumour growth.

Similar content being viewed by others

Data availability

The pancancer data were retrieved from TCGA Research Network: http://cancergenome.nih.gov/. The dataset used to infer the SL interactions in the earlier stage of the study is available in Broad GDAC Firehose (https://gdac.broadinstitute.org). For other TCGA analysis which was performed in the later stage, we used more updated datasets that are available through UCSC Xena Browser (https://xenabrowser.net). In particular, https://xenabrowser.net/datapages/?cohort=TCGA%20Pan-Cancer%20(PANCAN) and https://xenabrowser.net/datapages/?cohort=TCGA%20TARGET%20GTEx. All patients’ data were analyzed from published papers that are referenced and publicly available accordingly. Raw data for the GC-MS figures were deposited in Figshare with the Digital ObjectIdentifier 10.6084/m9.figshare.9887984. All data supporting the findings of this study are available from the corresponding author on reasonable request.

Change history

  • 20 December 2019

    In the version of this article originally published, the source data files for the main figures and Extended Data figures were linked incorrectly. The errors have been corrected.

References

  1. Maddocks, O. D. K. et al. Modulating the therapeutic response of tumours to dietary serine and glycine starvation. Nature 544, 372–376 (2017).

    CAS  PubMed  Google Scholar 

  2. Sun, L. et al. cMyc-mediated activation of serine biosynthesis pathway is critical for cancer progression under nutrient deprivation conditions. Cell Res. 25, 429–444 (2015).

    CAS  PubMed  Google Scholar 

  3. Wise, D. R. & Thompson, C. B. Glutamine addiction: a new therapeutic target in cancer. Trends Biochem. Sci. 35, 427–433 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Altman, B. J., Stine, Z. E. & Dang, C. V. From Krebs to clinic: glutamine metabolism to cancer therapy. Nat. Rev. Cancer 16, 749 (2016).

    CAS  PubMed  Google Scholar 

  5. Pavlova, N. N. et al. As extracellular glutamine levels decline, asparagine becomes an essential amino acid. Cell Metab. 27, 428–438 e425 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Zhang, J. et al. Asparagine plays a critical role in regulating cellular adaptation to glutamine depletion. Mol. Cell. 56, 205–218 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Knott, S. R. V. et al. Asparagine bioavailability governs metastasis in a model of breast cancer. Nature 554, 378–381 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Gwinn, D. M. et al. Oncogenic KRAS regulates amino acid homeostasis and asparagine biosynthesis via ATF4 and alters sensitivity to l-asparaginase. Cancer Cell 33, 91–107 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Pieters, R. et al. l-Asparaginase treatment in acute lymphoblastic leukemia: a focus on Erwinia asparaginase. Cancer 117, 238–249 (2011).

    CAS  PubMed  Google Scholar 

  10. Stams, W. A. et al. Asparagine synthetase expression is linked with l-asparaginase resistance in TEL–AML1-negative but not TEL–AML1-positive pediatric acute lymphoblastic leukemia. Blood 105, 4223–4225 (2005).

    CAS  PubMed  Google Scholar 

  11. Lessner, H. E., Valenstein, S., Kaplan, R., DeSimone, P. & Yunis, A. Phase II study of l-asparaginase in the treatment of pancreatic carcinoma. Cancer Treat. Rep. 64, 1359–1361 (1980).

    CAS  PubMed  Google Scholar 

  12. Bachet, J. B. et al. Asparagine synthetase expression and phase I study with l-asparaginase encapsulated in red blood cells in patients with pancreatic adenocarcinoma. Pancreas 44, 1141–1147 (2015).

    CAS  PubMed  Google Scholar 

  13. Taylor, C. W., Dorr, R. T., Fanta, P., Hersh, E. M. & Salmon, S. E. A phase I and pharmacodynamic evaluation of polyethylene glycol-conjugated l-asparaginase in patients with advanced solid tumors. Cancer Chemother. Pharmacol. 47, 83–88 (2001).

    CAS  PubMed  Google Scholar 

  14. Kilberg, M. S., Shan, J. & Su, N. ATF4-dependent transcription mediates signaling of amino acid limitation. Trends Endocrinol. Metab. 20, 436–443 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Nakamura, A. et al. Inhibition of GCN2 sensitizes ASNS-low cancer cells to asparaginase by disrupting the amino acid response. Proc. Natl Acad. Sci. USA 115, E7776–E7785 (2018).

    PubMed  Google Scholar 

  16. Lee, J. S. et al. Harnessing synthetic lethality to predict the response to cancer treatment. Nat. Commun. 9, 2546 (2018).

    PubMed  PubMed Central  Google Scholar 

  17. Bramham, C. R., Jensen, K. B. & Proud, C. G. Tuning specific translation in cancer metastasis and synaptic memory: control at the MNK–eIF4E axis. Trends Biochem. Sci. 41, 847–858 (2016).

    CAS  PubMed  Google Scholar 

  18. Wu, J., Ivanov, A. I., Fisher, P. B. & Fu, Z. Polo-like kinase 1 induces epithelial-to-mesenchymal transition and promotes epithelial cell motility by activating CRAF/ERK signaling. eLife 5, 10734 (2016).

    Google Scholar 

  19. Eferl, R. & Wagner, E. F. AP-1: a double-edged sword in tumorigenesis. Nat. Rev. Cancer 3, 859–868 (2003).

    CAS  PubMed  Google Scholar 

  20. Sears, R. et al. Multiple Ras-dependent phosphorylation pathways regulate Myc protein stability. Genes Dev. 14, 2501–2514 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Balgi, A. D. et al. Regulation of mTORC1 signaling by pH. PLoS ONE 6, e21549 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Roux, P. P. & Topisirovic, I. Signaling pathways involved in the regulation of mRNA translation. Mol. Cell Biol. 38, e00070-18 (2018).

    PubMed  PubMed Central  Google Scholar 

  23. Gandin, V.et al. Polysome fractionation and analysis of mammalian translatomes on a genome-wide scale. J. Vis. Exp, doi:51455 (2014).

  24. Waskiewicz, A. J. et al. Phosphorylation of the cap-binding protein eukaryotic translation initiation factor 4E by protein kinase Mnk1 in vivo. Mol. Cell Biol. 19, 1871–1880 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Wendel, H. G. et al. Dissecting eIF4E action in tumorigenesis. Genes Dev. 21, 3232–3237 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Topisirovic, I., Ruiz-Gutierrez, M. & Borden, K. L. Phosphorylation of the eukaryotic translation initiation factor eIF4E contributes to its transformation and mRNA transport activities. Cancer Res. 64, 8639–8642 (2004).

    CAS  PubMed  Google Scholar 

  27. Waskiewicz, A. J., Flynn, A., Proud, C. G. & Cooper, J. A. Mitogen-activated protein kinases activate the serine/threonine kinases Mnk1 and Mnk2. EMBO J. 16, 1909–1920 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Furic, L. et al. eIF4E phosphorylation promotes tumorigenesis and is associated with prostate cancer progression. Proc. Natl Acad. Sci. USA 107, 14134–14139 (2010).

    CAS  PubMed  Google Scholar 

  29. Webb, T. E., Hughes, A., Smalley, D. S. & Spriggs, K. A. An internal ribosome entry site in the 5’ untranslated region of epidermal growth factor receptor allows hypoxic expression. Oncogenesis 4, e134 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Guan, B. J. et al. A Unique ISR program determines cellular responses to chronic stress. Mol. Cell 68, 885–900 e886 (2017).

    CAS  PubMed  Google Scholar 

  31. de la Parra, C. et al. A widespread alternate form of cap-dependent mRNA translation initiation. Nat. Commun. 9, 3068 (2018).

    PubMed  PubMed Central  Google Scholar 

  32. Liu, S. et al. METTL13 methylation of eEF1A increases translational output to promote tumorigenesis. Cell 176, 491–504 (2019).

    CAS  PubMed  Google Scholar 

  33. Dorard, C. et al. RAF proteins exert both specific and compensatory functions during tumour progression of NRAS-driven melanoma. Nat. Commun. 8, 15262 (2017).

    CAS  PubMed  Google Scholar 

  34. Bhoumik, A. et al. An ATF2-derived peptide sensitizes melanomas to apoptosis and inhibits their growth and metastasis. J. Clin. Invest. 110, 643–650 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Falletta, P. et al. Translation reprogramming is an evolutionarily conserved driver of phenotypic plasticity and therapeutic resistance in melanoma. Genes Dev. 31, 18–33 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Kakavand, H. et al. PD-L1 expression and immune escape in melanoma resistance to MAPK inhibitors. Clin. Cancer Res. 23, 6054–6061 (2017).

    CAS  PubMed  Google Scholar 

  37. Rizos, H. et al. BRAF inhibitor resistance mechanisms in metastatic melanoma: spectrum and clinical impact. Clin. Cancer Res. 20, 1965–1977 (2014).

    CAS  PubMed  Google Scholar 

  38. Kwong, L. N. et al. Co-clinical assessment identifies patterns of BRAF inhibitor resistance in melanoma. J. Clin. Invest. 125, 1459–1470 (2015).

    PubMed  PubMed Central  Google Scholar 

  39. Zhang, G. et al. Targeting mitochondrial biogenesis to overcome drug resistance to MAPK inhibitors. J. Clin. Invest. 126, 1834–1856 (2016).

    PubMed  Google Scholar 

  40. Wek, R. C. Role of eIF2α kinases in translational control and adaptation to cellular stress. Cold Spring Harb. Perspect. Biol. 10, a032870 (2018).

    PubMed  Google Scholar 

  41. Krall, A. S., Xu, S., Graeber, T. G., Braas, D. & Christofk, H. R. Asparagine promotes cancer cell proliferation through use as an amino acid exchange factor. Nat. Commun. 7, 11457 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Pathria, G. et al. Targeting the Warburg effect via LDHA inhibition engages ATF4 signaling for cancer cell survival. EMBO J. 37, e99735 (2018).

    PubMed  PubMed Central  Google Scholar 

  43. Young, S. K. & Wek, R. C. Upstream open reading frames differentially regulate gene-specific translation in the integrated stress response. J. Biol. Chem. 291, 16927–16935 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Chen, R. et al. The general amino acid control pathway regulates mTOR and autophagy during serum/glutamine starvation. J. Cell Biol. 206, 173–182 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Shin, S. et al. ERK2 mediates metabolic stress response to regulate cell fate. Mol. Cell 59, 382–398 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Thiaville, M. M. et al. MEK signaling is required for phosphorylation of eIF2alpha following amino acid limitation of HepG2 human hepatoma cells. J. Biol. Chem. 283, 10848–10857 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Oakes, S. A. & Papa, F. R. The role of endoplasmic reticulum stress in human pathology. Annu. Rev. Pathol. 10, 173–194 (2015).

    CAS  PubMed  Google Scholar 

  48. Fiorese, C. J. et al. The transcription factor ATF5 mediates a mammalian mitochondrial UPR. Curr. Biol. 26, 2037–2043 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Pereira, E. R., Frudd, K., Awad, W. & Hendershot, L. M. Endoplasmic reticulum (ER) stress and hypoxia response pathways interact to potentiate hypoxia-inducible factor 1 (HIF-1) transcriptional activity on targets like vascular endothelial growth factor (VEGF). J. Biol. Chem. 289, 3352–3364 (2014).

    CAS  PubMed  Google Scholar 

  50. Lu, D., Wolfgang, C. D. & Hai, T. Activating transcription factor 3, a stress-inducible gene, suppresses Ras-stimulated tumorigenesis. J. Biol. Chem. 281, 10473–10481 (2006).

    CAS  PubMed  Google Scholar 

  51. Sang, N. et al. MAPK signaling up-regulates the activity of hypoxia-inducible factors by its effects on p300. J. Biol. Chem. 278, 14013–14019 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Wethmar, K. et al. Comprehensive translational control of tyrosine kinase expression by upstream open reading frames. Oncogene 35, 1736–1742 (2016).

    CAS  PubMed  Google Scholar 

  53. Calvo, S. E., Pagliarini, D. J. & Mootha, V. K. Upstream open reading frames cause widespread reduction of protein expression and are polymorphic among humans. Proc. Natl Acad. Sci. USA 106, 7507–7512 (2009).

    CAS  PubMed  Google Scholar 

  54. Ingolia, N. T., Lareau, L. F. & Weissman, J. S. Ribosome profiling of mouse embryonic stem cells reveals the complexity and dynamics of mammalian proteomes. Cell 147, 789–802 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Sidrauski, C., McGeachy, A. M., Ingolia, N. T. & Walter, P. The small molecule ISRIB reverses the effects of eIF2alpha phosphorylation on translation and stress granule assembly. eLife 4, 05033 (2015).

    Google Scholar 

  56. Reddy, K. B., Nabha, S. M. & Atanaskova, N. Role of MAP kinase in tumor progression and invasion. Cancer Metastasis Rev. 22, 395–403 (2003).

    CAS  PubMed  Google Scholar 

  57. Ackermann, J. et al. Metastasizing melanoma formation caused by expression of activated N-RasQ61K on an INK4a-deficient background. Cancer Res. 65, 4005–4011 (2005).

    CAS  PubMed  Google Scholar 

  58. Valérie Petit, V. et al. C57BL/6 congenic mouse NRAS melanoma cell lines are highly sensitive to the combination of Mek and Akt inhibitors in vitro and in vivo. Pigment Cell Melanoma Res. 32, 829–841 (2019).

    PubMed  Google Scholar 

  59. Ratnikov, B. et al. Glutamate and asparagine cataplerosis underlie glutamine addiction in melanoma. Oncotarget 6, 7379–7389 (2015).

    PubMed  PubMed Central  Google Scholar 

  60. Marcotte, R. et al. Functional genomic landscape of human breast cancer drivers, vulnerabilities, and resistance. Cell 164, 293–309 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Cowley, G. S. et al. Parallel genome-scale loss of function screens in 216 cancer cell lines for the identification of context-specific genetic dependencies. Sci. Data 1, 140035 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Marcotte, R. et al. Essential gene profiles in breast, pancreatic, and ovarian cancer cells. Cancer Discov. 2, 172–189 (2012).

    CAS  PubMed  Google Scholar 

  63. Iorio, F. et al. A landscape of pharmacogenomic interactions in cancer. Cell 166, 740–754 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Sahu, A. D. et al. Genome-wide prediction of synthetic rescue mediators of resistance to targeted and immunotherapy. Mol. Syst. Biol. 15, e8323 (2019).

    PubMed  PubMed Central  Google Scholar 

  65. Bilal, E. et al. Improving breast cancer survival analysis through competition-based multidimensional modeling. PLoS Comput. Biol. 9, e1003047 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Therneau, T. M. & Grambsch, P. M. Modeling Survival Data: Extending the Cox Model (Springer, 2000).

Download references

Acknowledgements

We thank D. Tuveson and members of his laboratory for providing KPC cells, M. Herlyn for providing the melanoma cell lines, members of the L. Larue laboratory for establishing the NRAS mutant melanoma cell line MaNRAS1 (1007), M. Leibovitch for technical assistance, O. Zagnitko for metabolic analyses and members of the Ronai lab for continued discussion. The Cancer Metabolism Core at Sanford Burnham Prebys Medical Discovery Institute is supported by National Cancer Institute Cancer Center Support Grant P30CA030199. I.T. is a scholar of the Fonds de Recherche du Québec-Santé (FRQS; Junior 2). Support from National Cancer Institute grants R35CA197465, P01CA128814 and Hevery Foundation Gift (to Z.A.R.) are gratefully acknowledged.

Author information

Authors and Affiliations

Authors

Contributions

G.P. and Z.A.R. conceived the study; G.P., E.H., I.T., E.R. and Z.A.R. designed experiments; G.P., E.H., K.T., S.V. and Y.F. performed experiments; G.P. and D.A.S. conducted metabolic analyses; L.L. provided valuable reagents; G.P., J.S.L., A.D.S., E.H., K.T., E.R. and Z.A.R. analysed data; and G.P., E.R., I.T. and Z.A.R. wrote the manuscript.

Corresponding authors

Correspondence to Gaurav Pathria or Ze’ev A. Ronai.

Ethics declarations

Competing interests

Z.R. is co-founder and serves as scientific advisor to Pangea Therapeutics. E.R. was co-founder (divested) and serves as a non-paid scientific advisor to Pangea Therapeutics.

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 ASNS Suppression Induces the Amino Acid Response Pathway.

a, GC-MS-based estimation of intracellular asparagine levels in A375 and UACC-903 cells 72 hr after treatment with si-ASNS. b, Relative proliferation of A375 and UACC-903 cells treated with si-ASNS#1 or si-ASNS#2, with or without supplementation with L-asparagine (L-Asn; 0.3 mM), over the indicated time course. c, Immunoblotting of ASNS, ATF4, phosphorylated and total GCN2 and eIF2α in melanoma cells 72 hr after treatment with si-ASNS, L-Asn (0.3 mM), or both. d, Immunoblotting of ATF4 in melanoma lines 72 hr after treatment with si-ASNS, ISRIB, or both (left). Relative proliferation of melanoma cells treated as in (left) for indicated time course (right). Data are representative of three independent experiments and presented as mean ±SEM of n=3 biological replicates in b and d. Data shown as mean ±SEM of n=3 independent experiments in a. Statistical significance was calculated using two-tailed unpaired Student’s t-test for a and two-way Anova for b and d. In b, black and orange P values correspond to the comparison between si-ASNS#1 and si-ASNS#1+L-Asn and si-ASNS#2 and si-ASNS#2+L-Asn respectively.

Source Data

Extended Data Fig. 2 In-Silico Pan-Tumor Analysis Predicts Synthetic Lethal Partners of ASNS.

a, Sensitivity to the PLK1/3 inhibitor GW843682X of pan-tumor cell lines segregated based on high (ASNShi) and low (ASNSlo) ASNS expression. P-value calculated by one-sided Wilcoxon rank sum test. Each dot represents a cell line (ASNShi, n=204; ASNSlo, n=20). Middle line and the whiskers represent the mean and the standard deviation respectively. b, Conditional essentiality of PLK1 (ASNShi, n=24; ASNSlo, n=22), JAK3 (ASNShi, n=69; ASNSlo, n=67) and ATF4 (ASNShi, n=69; ASNSlo, n=67) in ASNSlo and ASNShi cell lines. One-sided Wilcoxon rank sum P values are denoted for each gene knockdown. Data information: In the boxplots, the top and bottom horizontal lines represent the 75th and the 25th percentile, respectively, and the middle horizontal line represents the median. The size of the box represents the interquartile range, and the top and bottom whiskers represent the maximum and the minimum values respectively.

Extended Data Fig. 3 MAPK Signaling is Critical for ATF4 Induction Upon Asparagine Limitation.

a, Proliferation of melanoma cells as measured 72 hr after indicated treatment. Proliferation is shown relative to mock (NT-siRNA and DMSO)-treated cells, set to 1.0. b, Immunoblotting of indicated proteins in melanoma cells 72 hr after indicated treatment. c, qRT-PCR analysis of indicated transcripts in melanoma cells 48 hr after treatment with si-ASNS, PLX-4032, or both. d, Immunoblotting of indicated proteins 72 hr after treatment with si-ASNS, PD-325901 or both. e, Immunoblotting of phosphorylated and total ERK1/2 protein in indicated cancer cell lines 72 hr after combined treatment with si-ASNS and L-Asn, with or without L-A’ase. f, Immunoblotting of phosphorylated ERK1/2 in melanoma cells 72 hr after treatment with si-ASNS#1 or #2, with or without supplementation with L-Asn. g, Proliferation of cancer cell lines measured 72 hr after treatment with si-ASNS, PD-325901, or both relative to mock. h, GC-MS-based estimation of intracellular Ser, Gly, and Ala levels in UACC-903 cells 72 hr after treatment with si-ASNS. i, qRT-PCR analysis of indicated transcripts in UACC-903 cells 48 hr after treatment with si-ASNS, PLX-4032, or both. j, GC-MS-based 13C6-glucose fractional isotope labeling of serine and glycine in UACC-903 cells 72 hr after treatment with si-ASNS, PLX-4032, or both. k, qRT-PCR analysis of LDHA transcript in UACC-903 cells 48 hr after indicated treatment. Data are representative of three independent experiments and presented as the mean ±SEM of n=3 biological replicates in a and g, mean ±SEM of n=3 technical replicates in c and k, mean ±SEM of n=2 technical replicates in i. Data shown as mean ±SEM of n=3 independent experiments in h and j. Statistical significance was calculated using two-tailed unpaired Student’s t-test, except in a, where ordinary one-way Anova was used.

Source Data

Extended Data Fig. 4 The MAPK-mTORC1-eIF4E Axis is Essential for ATF4 Induction Following Asparagine Limitation.

a, Immunoblotting of phosphorylated and total S6 protein in melanoma cells 72 hr after treatment with si-ASNS, si-ATF4, si-BRAF, or indicated combinations. Fig. 3d and Extended Data Fig. 4a show parts of the same experiment and share the internal control (HSP90). b, Immunoblotting of phosphorylated and total S6 protein and 4E-BP1 in melanoma cells 72 hr after treatment with si-ASNS, SCH-772984, or both. c, Immunoblotting of ATF4 and phosphorylated and total S6 protein in Mia-Paca-2 cells 72 hr after treatment with si-ASNS#1 or #2, PD-325901, or a combination of the respective si-ASNS and PD-325901. d, Immunoblotting of ATF4 and phosphorylated and total S6 protein in pancreatic cancer cell lines 72 hr after treatment with si-ASNS, Rapamycin (Rapa), or both. e, Immunoblotting of ATF4 and phosphorylated and total S6 protein in Mia-Paca-2 cells 72 hr after treatment with si-ASNS, Torin 1, or both. f, Proliferation of cancer cell lines measured 72 hr after treatment with si-ASNS, Rapa, or both. Values are shown relative to mock (NT-siRNA and DMSO)-treated cells. Data are representative of three independent experiments and presented as the mean ±SEM of n=3 biological replicates in f. Statistical significance was calculated using two-tailed unpaired Student’s t-test.

Source Data

Extended Data Fig. 5 MNK1 is Critical for ASNS Suppression-Associated ATF4 Induction.

a, Immunoblotting of MNK1 and ATF4 in melanoma cells 72 hr after treatment with si-ASNS, si-MNK1#1, #2 or #3, or indicated combinations of si-ASNS with si-MNK1#1-#3. b, Immunoblotting of ATF4 and MNK1 in Mia-Paca-2 and Panc-1 cells 72 hr after treatment with si-ASNS, si-MNK1, or both. c, Proliferation of cancer cell lines 72 hr after treatment with si-ASNS, si-MNK1, or both relative to NT-siRNA treated cells (set to 1.0). d, qRT-PCR analysis of MNK1 transcript in melanoma cells 48 hr after treatment with indicated si-ASNS. e, Immunoblotting of phospho and total MNK1 and eIF4E proteins in melanoma cells after 72 hr treatment with si-ASNS and L-Asn (0.3 mM), with or without L-A’ase. f, qRT-PCR analysis of eIF4E transcript in melanoma cells 48 hr after treatment with si-ASNS. g, qRT-PCR analysis of H3A and CDH1 mRNA levels in subpolysomal, light, and heavy polysomal fractions of A375 cells treated for 48 hr with si-ASNS relative to mock treatment, set to 1.0. h, Immunoblotting of ATF4 and phospho and total eIF4E protein in UACC-903 cells treated with si-ASNS, eFT508, or both for 72 hr. i and j, Immunoblotting of phospho and total MNK1 and eIF4E proteins in melanoma cells treated 72 hr with si-ASNS, SCH-772984, or both (i), or si-ASNS, Rapa, or both (j). Data are representative of three independent experiments and presented as the mean ±SEM of n=3 biological replicates in c, mean ±SEM of n=4 technical replicates in d and g, and mean ±SEM of n=3 technical replicates in f. Statistical significance was calculated using two-tailed unpaired Student’s t-test.

Source Data

Extended Data Fig. 6 Asparagine Limitation Increases RTK Expression.

a, Immunoblotting of EGFR in melanoma cells 72 hr after treatment with indicated si-ASNS. b, qRT-PCR analysis of EGFR transcript in melanoma cells 48 hr after treatment with si-ASNS. c-e, Immunoblotting of EGFR in melanoma cells 72 hr after treatment either individually or as a combination with si-ASNS and Torin 1 (c), si-ASNS and si-eIF4E (d), or si-ASNS and si-MNK1 (e). f, Immunoblotting of phospho and total ERK1/2 in melanoma cells treated 72 hr with si-ASNS, EGFR inhibitor (Gefitinib), or both. g, qRT-PCR analysis of VEGFR-2 transcript levels in melanoma cells 48 hr after treatment with si-ASNS#2 or #3. h, (left) Immunoblotting of VEGFR-2, PDGFR-B, α-Tubulin, and GAPDH in melanoma cells treated with NT-siRNA, si-ASNS#1 or #2 followed by L-azidohomoalanine (AHA) labeling (see Methods). (right) Ponseau-S staining of streptavidin pull-down fraction from melanoma cells left untreated or treated with AHA. i, Immunoblotting of VEGFR-2 and PDGFR-B in melanoma cells 72 hr after treatment with si-ASNS, ISRIB, or both. j, Immunoblotting of the indicated proteins in melanoma cells 72 hr after treatment with si-ASNS, si-GCN2, or both. Data are representative of three independent experiments and presented as the mean ±SEM of n=3 technical replicates in b and g.

Source Data

Extended Data Fig. 7 L-Asparaginase and MEK Inhibitor Combination Suppresses In Vivo Tumor Growth.

a, Comparison of ASNS expression in cancer vs. healthy tissue samples from TCGA and GTEx dataset using two-sided Wilcoxon rank sum test. b, Proliferation of melanoma and pancreatic cancer cell lines as measured 72 hr after treatment with L-A’ase, PD-325901, or both relative to mock-treated cells. All cell lines were grown in L-Asn-supplemented DMEM. c, Volume of tumours from C3H/HeN mice injected subcutaneously with SW1 mouse melanoma cells and treated as indicated (n=8 mice per group). Statistical analysis: Welch’s t-test (two-tailed). In c, brown, blue, and pink P values correspond to the comparison between L-A’ase+PD-325901 (2.5 mg/Kg) and Vehicle, L-A’ase+PD-325901 (2.5 mg/Kg) and L-A’ase, and L-A’ase+PD-325901(2.5 mg/Kg) and PD-325901 (2.5 mg/Kg) respectively. In c, green, red, and black P values correspond to the comparison between L-A’ase+PD-325901(5.0 mg/Kg) and Vehicle, L-A’ase+PD-325901 (5.0 mg/Kg) and L-A’ase, and L-A’ase+PD-325901(5.0 mg/Kg) and PD-325901 (5.0 mg/Kg) respectively. In a, LUSC, Lung Squamous Cell Carcinoma; DLBC, Diffuse Large B-Cell Lymphoma; COAD, Colon Adenocarcinoma; ESCA, Esophageal Carcinoma; SARC, Sarcoma; UCEC, Uterine Corpus Endometrial Carcinoma; OV, Ovarian Cancer; KICH, Kidney Chromophobe; BLCA, Bladder Urothelial Carcinoma; CESC, Cervical Squamous Cell Carcinoma; KIRP, Kidney Renal Papillary Cell Carcinoma; BRCA, Breast Cancer; HNSC, Head and Neck Squamous Cell Carcinoma; KIRC, Kidney Renal Clear Cell Carcinoma; LUAD, Lung Adenocarcinoma; LIHC, Liver Hepatocellular Carcinoma; TCGT, Tenosynovial Giant Cell Tumor; LAML, Acute Myeloid Leukaemia; PRAD, Prostate Adenocarcinoma; GBM, Glioblastoma Multiforme; STAD, Stomach Adenocarcinoma; THCA, Thyroid Carcinoma; LGG, Low Grade Glioma. Data information: In the boxplots, the top and bottom horizontal lines represent the 75th and the 25th percentile, respectively, and the middle horizontal line represents the median. The size of the box represents the interquartile range, and the top and bottom whiskers represent the maximum and the minimum values respectively. In b, data are representative of three independent experiments and presented as the mean ±SEM of n=4 biological replicates and the statistical significance was calculated using two-tailed unpaired Student’s t-test.

Source Data

Supplementary information

Reporting Summary

Supplementary Table 1

ASNS Synthetic Lethal Partners.

Supplementary Table 2

Gene List- Predictors of Response to MAPK Signaling Inhibition.

Supplementary Table 3

qRT-PCR Primer List.

Source data

Source Data Fig. 1

Statistical Source Data

Source Data Fig. 1

Unprocessed Western Blots

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. 5

Unprocessed Western Blots

Source Data Fig. 6

Statistical Source Data

Source Data Fig. 6

Unprocessed Western Blots

Source Data Fig. 7

Statistical Source Data

Source Data Fig. 7

Unprocessed Western Blots

Source Data Extended Data Fig. 1

Statistical Source Data

Source Data Extended Data Fig. 1

Unprocessed Western Blots

Source Data Extended Data Fig. 3

Statistical Source Data

Source Data Extended Data Fig. 3

Unprocessed Western Blots

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. 5

Unprocessed Western Blots

Source Data Extended Data Fig. 6

Statistical Source Data

Source Data Extended Data Fig. 6

Unprocessed Western Blots

Source Data Extended Data Fig. 7

Statistical Source Data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pathria, G., Lee, J.S., Hasnis, E. et al. Translational reprogramming marks adaptation to asparagine restriction in cancer. Nat Cell Biol 21, 1590–1603 (2019). https://doi.org/10.1038/s41556-019-0415-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-019-0415-1

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