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:

A widespread role for SLC transmembrane transporters in resistance to cytotoxic drugs

Abstract

Solute carriers (SLCs) are the largest family of transmembrane transporters in humans and are major determinants of cellular metabolism. Several SLCs have been shown to be required for the uptake of chemical compounds into cellular systems, but systematic surveys of transporter–drug relationships in human cells are currently lacking. We performed a series of genetic screens in a haploid human cell line against 60 cytotoxic compounds representative of the chemical space populated by approved drugs. By using an SLC-focused CRISPR–Cas9 library, we identified transporters whose absence induced resistance to the drugs tested. This included dependencies involving the transporters SLC11A2/SLC16A1 for artemisinin derivatives and SLC35A2/SLC38A5 for cisplatin. The functional dependence on SLCs observed for a significant proportion of the screened compounds suggests a widespread role for SLCs in the uptake and cellular activity of cytotoxic drugs and provides an experimentally validated set of SLC–drug associations for a number of clinically relevant compounds.

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: Experimental setup and overview of the screened compound set.
Fig. 2: Identification of enriched SLCs upon compound treatment.
Fig. 3: Validation of a set of SLC/compound associations by multicolor competition assay (MCA).
Fig. 4: Characterization of selected SLC/drug associations.
Fig. 5: Chemoinformatic analysis of the compound subsets employed in this study.

Similar content being viewed by others

Data availability

The data and code that support the findings of this study are available on request from the corresponding author. Data used to generate Figs. 2c and 3b and Supplementary Fig. 3b are available in Supplementary Datasets 1 and 2.

References

  1. Gonzalez, F. J., Coughtrie, M. & Tukey, R. H. in Goodman & Gilman’s: The Pharmacological Basis of Therapeutics (eds Brunton, L. L., Chabner, B. A. & Knollmann, B. C.) Ch. 6 (McGraw-Hill Education, 2015).

  2. Nigam, S. K. What do drug transporters really do? Nat. Rev. Drug Discov. 14, 29–44 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. César-Razquin, A. et al. A call for systematic research on solute carriers. Cell 162, 478–487 (2015).

    Article  CAS  PubMed  Google Scholar 

  4. Fletcher, J. I., Haber, M., Henderson, M. J. & Norris, M. D. ABC transporters in cancer: more than just drug efflux pumps. Nat. Rev. Cancer 10, 147–156 (2010).

    Article  CAS  PubMed  Google Scholar 

  5. Hediger, M. A., Clémençon, B., Burrier, R. E. & Bruford, E. A. The ABCs of membrane transporters in health and disease (SLC series): introduction. Mol. Aspects Med. 34, 95–107 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Motohashi, H. & Inui, K. Multidrug and toxin extrusion family SLC47: physiological, pharmacokinetic and toxicokinetic importance of MATE1 and MATE2-K. Mol. Aspects Med. 34, 661–668 (2013).

    Article  CAS  PubMed  Google Scholar 

  7. Estudante, M., Soveral, G., Morais, J. G. & Benet, L. Z. Insights into solute carriers: physiological functions and implications in disease and pharmacokinetics. MedChemComm 7, 1462–1478 (2016).

    Article  CAS  Google Scholar 

  8. Geier, E. G. et al. Profiling solute carrier transporters in the human blood–brain barrier. Clin. Pharmacol. Ther. 94, 636–639 (2013).

    Article  CAS  PubMed  Google Scholar 

  9. Giacomini, K. M. et al. International transporter consortium commentary on clinically important transporter polymorphisms. Clin. Pharmacol. Ther. 94, 23–26 (2013).

    Article  CAS  PubMed  Google Scholar 

  10. US Department of Health and Human Services Food and Drug Administration Center for Drug Evaluation and Research (CDER). Clinical Drug Interaction Studies—Study Design, Data Analysis, and Clinical Implications (2017).

  11. Matsson, P. et al. Quantifying the impact of transporters on cellular drug permeability. Trends Pharmacol. Sci. 36, 255–262 (2015).

    Article  CAS  PubMed  Google Scholar 

  12. Dobson, P. D. & Kell, D. B. Carrier-mediated cellular uptake of pharmaceutical drugs: an exception or the rule? Nat. Rev. Drug Discov. 7, 205–220 (2008).

    Article  CAS  PubMed  Google Scholar 

  13. Sugano, K. et al. Coexistence of passive and carrier-mediated processes in drug transport. Nat. Rev. Drug Discov. 9, 597–614 (2010).

    Article  CAS  PubMed  Google Scholar 

  14. Young, J. D., Yao, S. Y. M., Baldwin, J. M., Cass, C. E. & Baldwin, S. A. The human concentrative and equilibrative nucleoside transporter families, SLC28 and SLC29. Mol. Aspects Med. 34, 529–547 (2013).

    Article  CAS  PubMed  Google Scholar 

  15. Icard, P. et al. How the Warburg effect supports aggressiveness and drug resistance of cancer cells? Drug Resist. Updat. 38, 1–11 (2018).

    Article  PubMed  Google Scholar 

  16. Reczek, C. R. et al. A CRISPR screen identifies a pathway required for paraquat-induced cell death. Nat. Chem. Biol. 13, 1274–1279 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Winter, G. E. et al. The solute carrier SLC35F2 enables YM155-mediated DNA damage toxicity. Nat. Chem. Biol. 10, 768–773 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Birsoy, K. et al. MCT1-mediated transport of a toxic molecule is an effective strategy for targeting glycolytic tumors. Nat. Genet. 45, 104–108 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Reiling, J. H. et al. A haploid genetic screen identifies the major facilitator domain containing 2A (MFSD2A) transporter as a key mediator in the response to tunicamycin. Proc. Natl Acad. Sci. USA 108, 11756–11765 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Blomen, V. A. et al. Gene essentiality and synthetic lethality in haploid human cells. Science 350, 1092–1096 (2015).

    Article  CAS  PubMed  Google Scholar 

  21. Klein, M.-C. et al. AXER is an ATP/ADP exchanger in the membrane of the endoplasmic reticulum. Nat. Commun. 9, 3489 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Robinson, A. J., Kunji, E. R. S. & Gross, A. Mitochondrial carrier homolog 2 (MTCH2): the recruitment and evolution of a mitochondrial carrier protein to a critical player in apoptosis. Exp. Cell Res. 318, 1316–1323 (2012).

    Article  CAS  PubMed  Google Scholar 

  23. O’Hagan, S., Wright Muelas, M., Day, P. J., Lundberg, E. & Kell, D. B. GeneGini: assessment via the Gini coefficient of reference ‘housekeeping’ genes and diverse human transporter expression profiles. Cell Syst. 6, 230–244 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. César-Razquin, A. et al. In silico prioritization of transporter–drug relationships from drug sensitivity screens. Front. Pharmacol. 9, 1011 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Licciardello, M. P. et al. A combinatorial screen of the CLOUD uncovers a synergy targeting the androgen receptor. Nat. Chem. Biol. 13, 771–778 (2017).

    Article  CAS  PubMed  Google Scholar 

  26. Zhao, R., Diop-Bove, N., Visentin, M. & Goldman, I. D. Mechanisms of membrane transport of folates into cells and across epithelia. Annu. Rev. Nutr. 31, 177–201 (2011).

    Article  CAS  PubMed  Google Scholar 

  27. Sirotnak, F. M., DeGraw, J. I., Moccio, D. M., Samuels, L. L. & Goutas, L. J. New folate analogs of the 10-deaza-aminopterin series Basis for structural design and biochemical and pharmacologic properties. Cancer Chemother. Pharmacol. 12, 18–25 (1984).

    CAS  PubMed  Google Scholar 

  28. Waalkes, T. P. & Makulu, D. R. Pharmacologic aspects of pentamidine. Natl Cancer Inst. Monogr. 43, 171–177 (1976).

    CAS  PubMed  Google Scholar 

  29. Grottker, J., Rosenberger, A., Burckhardt, G. & Hagos, Y. Interaction of human multidrug and toxin extrusion 1 (MATE1) transporter with antineoplastic agents. Drug Metabol. Drug Interact. 26, 181–189 (2011).

    Article  CAS  PubMed  Google Scholar 

  30. Yang, N.-D. et al. Artesunate induces cell death in human cancer cells via enhancing lysosomal function and lysosomal degradation of ferritin. J. Biol. Chem. 289, 33425–33441 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Cui, L. & Su, X. Discovery, mechanisms of action and combination therapy of artemisinin. Expert Rev. Anti Infect. Ther. 7, 999–1013 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Montalbetti, N., Simonin, A., Kovacs, G. & Hediger, M. A. Mammalian iron transporters: families SLC11 and SLC40. Mol. Aspects Med. 34, 270–287 (2013).

    Article  CAS  PubMed  Google Scholar 

  33. Kell, D. B. Iron behaving badly: inappropriate iron chelation as a major contributor to the aetiology of vascular and other progressive inflammatory and degenerative diseases. BMC Med. Genomics 2, 2 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Pérez-Escuredo, J. et al. Monocarboxylate transporters in the brain and in cancer. Biochim. Biophys. Acta Mol. Cell Res. 1863, 2481–2497 (2016).

    Article  CAS  Google Scholar 

  35. Song, Z. Roles of the nucleotide sugar transporters (SLC35 family) in health and disease. Mol. Aspects Med. 34, 590–600 (2013).

    Article  CAS  PubMed  Google Scholar 

  36. Schiöth, H. B., Roshanbin, S., Hägglund, M. G. A. & Fredriksson, R. Evolutionary origin of amino acid transporter families SLC32, SLC36 and SLC38 and physiological, pathological and therapeutic aspects. Mol. Aspects Med. 34, 571–585 (2013).

    Article  CAS  PubMed  Google Scholar 

  37. Våtsveen, T. K. et al. Artesunate shows potent anti-tumor activity in B-cell lymphoma. J. Hematol. Oncol. 11, 23 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Wang, N., Zeng, G.-Z., Yin, J.-L. & Bian, Z.-X. Artesunate activates the ATF4-CHOP-CHAC1 pathway and affects ferroptosis in Burkitt’s lymphoma. Biochem. Biophys. Res. Commun. 519, 533–539 (2019).

    Article  CAS  PubMed  Google Scholar 

  39. St Germain, C. et al. Cisplatin induces cytotoxicity through the mitogen-activated protein kinase pathways and activating transcription factor 3. Neoplasia 12, 527–538 (2010).

    Article  CAS  Google Scholar 

  40. Wishart, D. S. et al. DrugBank 5.0: a major update to the DrugBank database for 2018. Nucleic Acids Res. 46, D1074–D1082 (2017).

    Article  CAS  PubMed Central  Google Scholar 

  41. Yu, Z. et al. Identification of a transporter complex responsible for the cytosolic entry of nitrogen-containing bisphosphonates. eLife 7, e36620 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Yee, S. W. et al. SLC19A1 pharmacogenomics summary. Pharmacogenet. Genomics 20, 708–715 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Pastor-Anglada, M. & Pérez-Torras, S. Nucleoside transporter proteins as biomarkers of drug responsiveness and drug targets. Front. Pharmacol. 6, 13 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Lai, H. & Singh, N. P. Selective cancer cell cytotoxicity from exposure to dihydroartemisinin and holotransferrin. Cancer Lett. 91, 41–46 (1995).

    Article  CAS  PubMed  Google Scholar 

  45. Duan, G. et al. Increased glutamine consumption in cisplatin-resistant cells has a negative impact on cell growth. Sci. Rep. 8, 4067 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Guidi, N. & Longo, V. D. Periodic fasting starves cisplatin‐resistant cancers to death. EMBO J. 37, e99815 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Yamada, D. et al. Inhibition of the glutamine transporter SNAT1 confers neuroprotection in mice by modulating the mTOR-autophagy system. Commun. Biol. 2, 346 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Farge, T. et al. Chemotherapy-resistant human acute myeloid leukemia cells are not enriched for leukemic stem cells but require oxidative metabolism. Cancer Discov. 7, 716–735 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Bhattacharya, B., Mohd Omar, M. F. & Soong, R. The Warburg effect and drug resistance: the Warburg effect and drug resistance. Br. J. Pharmacol. 173, 970–979 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Lanthaler, K. et al. Genome-wide assessment of the carriers involved in the cellular uptake of drugs: a model system in yeast. BMC Biol. 9, 70 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Doench, J. G. et al. Rational design of highly active sgRNAs for CRISPR-Cas9-mediated gene inactivation. Nat. Biotechnol. 32, 1262–1267 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Uhlén, M. et al. Tissue-based map of the human proteome. Science 347, 1260419 (2015).

    Article  CAS  PubMed  Google Scholar 

  53. Heigwer, F., Kerr, G. & Boutros, M. E-CRISP: fast CRISPR target site identification. Nat. Methods 11, 122–123 (2014).

    Article  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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Konermann, S. et al. Genome-scale transcriptional activation by an engineered CRISPR-Cas9 complex. Nature 517, 583–588 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Montague, T. G., Cruz, J. M., Gagnon, J. A., Church, G. M. & Valen, E. CHOPCHOP: a CRISPR/Cas9 and TALEN web tool for genome editing. Nucleic Acids Res. 42, W401–W407 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Bigenzahn, J. W. et al. LZTR1 is a regulator of RAS ubiquitination and signaling. Science 362, 1171–1177 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Brinkman, E. K., Chen, T., Amendola, M. & van Steensel, B. Easy quantitative assessment of genome editing by sequence trace decomposition. Nucleic Acids Res. 42, e168 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Moskovskich, A. et al. The transporters SLC35A1 and SLC30A1 play opposite roles in cell survival upon VSV virus infection. Sci. Rep. 9, 10471 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank all members of the Superti-Furga laboratory for discussions and feedback. We are also grateful to the Biomedical Sequencing facility for advice on Illumina sequencing and to the Flow Cytometry Core Facility of the Vienna Medical University for help with FACS sorting. We also thank B. Vilagos for graphical input and advice and S. Sdelci for scientific discussions and insights. We acknowledge support by the Austrian Academy of Sciences, the European Research Council (ERC AdG 695214 GameofGates, to E.G. and G.F.), the Austrian Science Fund (FWF I2192-B22 ERASE, to A.C.-R.; FWF P29250-B30 VITRA, to E.G., J.K. and G.F.) and also a Marie Sklodowska-Curie fellowship to E.G. (MSCA-IF-2014-661491). Research in the Kubicek laboratory is supported by the Austrian Federal Ministry for Digital and Economic Affairs and the National Foundation for Research, Technology and Development, the Austrian Science Fund (FWF) F4701 and the European Research Council (ERC) under the European Union’s Horizon 2020 research and innovation programme (ERC-CoG-772437). The Pharmacoinformatics Research Group (Ecker lab) acknowledges funding provided by the Austrian Science Fund FWF AW012321 MolTag.

Author information

Authors and Affiliations

Authors

Contributions

E.G., G.S.-F. conceived and designed the study. E.G., K.P., S.L., J.K., B.G., K.K., G.F., A.I.-P., F.K., A.K. and C.-H.L. performed experiments and analyzed data. A.C.-R. analyzed screening and validation data. V.S. analyzed the transcriptomics data. J.H., S. Kickinger and G.F.E. performed the chemoinformatic analysis. R.K.K. contributed to library design. S. Kubicek, G.F.E. and G.S.-F. provided supervision.

Corresponding author

Correspondence to Giulio Superti-Furga.

Ethics declarations

Competing interests

C.-H.L. is an employee of AstraZeneca Limited (UK). The 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.

Supplementary information

Supplementary Information

Supplementary Tables 1–5 and Figs. 1–6.

Reporting Summary

Supplementary Dataset 1

P and FDR values used to generate Fig. 2c.

Supplementary Dataset 2

Ratios and P values used to generate Fig. 3b and Supplementary Fig. 3b.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Girardi, E., César-Razquin, A., Lindinger, S. et al. A widespread role for SLC transmembrane transporters in resistance to cytotoxic drugs. Nat Chem Biol 16, 469–478 (2020). https://doi.org/10.1038/s41589-020-0483-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-020-0483-3

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research