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.

  • Letter
  • Published:

Selenium detoxification is required for cancer-cell survival

Abstract

The micronutrient selenium is incorporated via the selenocysteine biosynthesis pathway into the rare amino acid selenocysteine, which is required in selenoproteins such as glutathione peroxidases and thioredoxin reductases1,2. Here, we show that selenophosphate synthetase 2 (SEPHS2), an enzyme in the selenocysteine biosynthesis pathway, is essential for survival of cancer, but not normal, cells. SEPHS2 is required in cancer cells to detoxify selenide, an intermediate that is formed during selenocysteine biosynthesis. Breast and other cancer cells are selenophilic, owing to a secondary function of the cystine/glutamate antiporter SLC7A11 that promotes selenium uptake and selenocysteine biosynthesis, which, by allowing production of selenoproteins such as GPX4, protects cells against ferroptosis. However, this activity also becomes a liability for cancer cells because selenide is poisonous and must be processed by SEPHS2. Accordingly, we find that SEPHS2 protein levels are elevated in samples from people with breast cancer, and that loss of SEPHS2 impairs growth of orthotopic mammary-tumour xenografts in mice. Collectively, our results identify a vulnerability of cancer cells and define the role of selenium metabolism in cancer.

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: Identification of SEPHS2 as an enzyme that is selectively essential to cancer cells and is a potential therapeutic target.
Fig. 2: SEPHS2 is dispensable to non-transformed cells despite being required to produce selenoproteins.
Fig. 3: SLC7A11-mediated reduction of selenite is the initial step of the selenocysteine biosynthesis pathway in cancer cells, and protects against ferroptosis.
Fig. 4: Essentiality of SEPHS2 in cancer cells is due to its role as a selenide-detoxifying enzyme.

Similar content being viewed by others

Data availability

All raw data that support the findings of this study are available from the corresponding author upon request. Source data are provided with this paper.

References

  1. Labunskyy, V. M., Hatfield, D. L. & Gladyshev, V. N. Selenoproteins: molecular pathways and physiological roles. Physiol. Rev. 94, 739–777 (2014).

    Article  CAS  Google Scholar 

  2. Burk, R. F. & Hill, K. E. Regulation of selenium metabolism and transport. Annu. Rev. Nutr. 35, 109–134 (2015).

    Article  CAS  Google Scholar 

  3. Hanahan, D. & Weinberg, R. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011).

    Article  CAS  Google Scholar 

  4. Amoedo, N. D., Obre, E. & Rossignol, R. Drug discovery strategies in the field of tumor energy metabolism: limitations by metabolic flexibility and metabolic resistance to chemotherapy. Biochim. Biophys. Acta Bioenerg. 1858, 674–685 (2017).

    Article  CAS  Google Scholar 

  5. Wexler, P. TOXNET: an evolving web resource for toxicology and environmental health information. Toxicology 157, 3–10 (2001).

    Article  CAS  Google Scholar 

  6. Kanehisa, M. & Goto, S. KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 28, 27–30 (2000).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  8. Banerjee, B. D., Dwivedi, S. & Singh, S. Acute hydrogen selenide gas poisoning admissions in one of the hospitals in Delhi, India: case report. Hum. Exp. Toxicol. 16, 276–278 (1997).

    Article  CAS  Google Scholar 

  9. Schecter, A., Shanske, W., Stenzler, A., Quintilian, H. & Steinberg, H. Acute hydrogen selenide inhalation. Chest 77, 554–555 (1980).

    Article  CAS  Google Scholar 

  10. Strickland, L. B., Dawson, P. J., Santner, S. J. & Miller, F. R. Progression of premalignant MCF10AT generates heterogeneous malignant variants with characteristic histologic types and immunohistochemical markers. Breast Cancer Res. Treat 64, 235–240 (2000).

    Article  CAS  Google Scholar 

  11. Xu, X. M. et al. Selenophosphate synthetase 2 is essential for selenoprotein biosynthesis. Biochem. J. 404, 115–120 (2007).

    Article  CAS  Google Scholar 

  12. Noinaj, N. et al. Structural insights into the catalytic mechanism of Escherichia coli selenophosphate synthetase. J. Bacteriol. 194, 499–508 (2012).

    Article  CAS  Google Scholar 

  13. Cavalieri, R. R., Scott, K. G. & Sairenji, E. Selenite (75Se) as a tumor-localizing agent in man. J. Nucl. Med. 7, 197–208 (1966).

    CAS  PubMed  Google Scholar 

  14. Cavalieri, R. R. & Scott, K. G. Sodium selenite Se 75. A more specific agent for scanning tumors. JAMA 206, 591–595 (1968).

    Article  CAS  Google Scholar 

  15. Jiang, C., Wang, Z., Ganther, H. & Lu, J. Distinct effects of methylseleninic acid versus selenite on apoptosis, cell cycle, and protein kinase pathways in DU145 human prostate cancer cells. Mol. Cancer Ther. 1, 1059–1066 (2002).

    CAS  PubMed  Google Scholar 

  16. Sato, H. et al. Redox imbalance in cystine/glutamate transporter-deficient mice. J. Biol. Chem. 280, 37423–37429 (2005).

    Article  CAS  Google Scholar 

  17. Sato, H., Tamba, M., Ishii, T. & Bannai, S. Cloning and expression of a plasma membrane cystine/glutamate exchange transporter composed of two distinct proteins. J. Biol. Chem. 274, 11455–11458 (1999).

    Article  CAS  Google Scholar 

  18. Olm, E. et al. Extracellular thiol-assisted selenium uptake dependent on the x(c)- cystine transporter explains the cancer-specific cytotoxicity of selenite. Proc. Natl Acad. Sci. USA 106, 11400–11405 (2009).

    Article  CAS  Google Scholar 

  19. Kim, J. Y. et al. Human cystine/glutamate transporter: cDNA cloning and upregulation by oxidative stress in glioma cells. Biochim. Biophys. Acta 1512, 335–344 (2001).

    Article  CAS  Google Scholar 

  20. Jiang, L. et al. Ferroptosis as a p53-mediated activity during tumour suppression. Nature 520, 57–62 (2015).

    Article  CAS  Google Scholar 

  21. Hangauer, M. J. et al. Drug-tolerant persister cancer cells are vulnerable to GPX4 inhibition. Nature 551, 247–250 (2017).

    Article  CAS  Google Scholar 

  22. Yang, W. S. et al. Regulation of ferroptotic cancer cell death by GPX4. Cell 156, 317–331 (2014).

    Article  CAS  Google Scholar 

  23. Doll, S. et al. FSP1 is a glutathione-independent ferroptosis suppressor. Nature 575, 693–698 (2019).

    Article  CAS  Google Scholar 

  24. Bersuker, K. et al. The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature 575, 688–692 (2019).

    Article  CAS  Google Scholar 

  25. Spallholz, J. E. On the nature of selenium toxicity and carcinostatic activity. Free Radic. Biol. Med. 17, 45–64 (1994).

    Article  CAS  Google Scholar 

  26. Alim, I. et al. Selenium drives a transcriptional adaptive program to block ferroptosis and treat stroke. Cell 177, 1262–1279 e1225 (2019).

    Article  CAS  Google Scholar 

  27. Ingold, I. et al. Selenium utilization by GPX4 is required to prevent hydroperoxide-induced ferroptosis. Cell 172, 409–422 e421 (2018).

    Article  CAS  Google Scholar 

  28. Delafiori, J., Ring, G. & Furey, A. Clinical applications of HPLC-ICP-MS element speciation: A review. Talanta 153, 306–331 (2016).

    Article  CAS  Google Scholar 

  29. Suzuki, K. T. & Ogra, Y. Metabolic pathway for selenium in the body: speciation by HPLC-ICP MS with enriched Se. Food Addit. Contam. 19, 974–983 (2002).

    Article  CAS  Google Scholar 

  30. Dauplais, M., Lazard, M., Blanquet, S. & Plateau, P. Neutralization by metal ions of the toxicity of sodium selenide. PLoS One 8, e54353 (2013).

    Article  CAS  Google Scholar 

  31. Wang, T., Wei, J. J., Sabatini, D. M. & Lander, E. S. Genetic screens in human cells using the CRISPR–Cas9 system. Science 343, 80–84 (2014).

    Article  CAS  Google Scholar 

  32. Sheehan, T. M. & Gao, M. Simplified fluorometric assay of total selenium in plasma and urine. Clin. Chem. 36, 2124–2126 (1990).

    Article  CAS  Google Scholar 

  33. A Package for Survival Analysis in S. Version 2.38 (2015); http//cran.r-project.org/package=survival

  34. Harrington, D. P. & Fleming, T. R. A class of rank test procedures for censored survival data. Biometrika 69, 553–566 (1982).

  35. Ciriello, G. et al. Comprehensive molecular portraits of invasive lobular breast. Cancer Cell 163, 506–519 (2015).

    CAS  Google Scholar 

  36. Cerami, E. et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2, 401–404 (2012).

    Article  Google Scholar 

Download references

Acknowledgements

We thank M. Green, E. Baehrecke, A. Mercurio, D. Sabatini, C. Haynes, D. Guertin, B. Bible, K. Krupczak, K. Luk, C. Brown, A. Jaferi and S. Deibler for advice, assistance and feedback. We thank D. Sabatini, S. Wolfe, M. Lee, A. Mercurio and K. -Y. Choi for materials including vectors and cell lines. We thank O. Kwon for illustrations, and C. -C. Hsieh for assistance with statistics. This work was supported by the Suh Kyungbae Foundation (SUHF) Young Investigator Award to D.K.; NIH T32 CA130807-8 to M.E.S.; Searle Scholars Program, Rita Allen Foundation, Whitehall Foundation, Smith Family Foundation, and NIH/NIA DP2 AG067490-01 to P.L.G; and R01 CA229910 to L.M.S.

Author information

Authors and Affiliations

Authors

Contributions

A.E.C. and D.K. conceived the project; A.E.C., N.L. and D.K. designed the research. A.E.C. and N.L. performed most of the experiments with assistance from A.N.M.-O., M.E.S., M.B.D., D.Y., A.B.J. and A.P. In situ hybridization and immunohistochemistry experiments were carried out by S.J.P., N.L. and P.L.G. ICP-MS-based experiments for selenite measurements were conducted by M.S. R.L., N.L., M.B.D. and L.J.Z. conducted mining and analyses of TCGA and other large-scale datasets. A.N.M.-O. and L.M.S. assisted with mouse xenograft studies. K.S. assisted with human sample studies. A.E.C., N.L. and D.K. wrote the manuscript with consultation from all of the authors.

Corresponding author

Correspondence to Dohoon Kim.

Ethics declarations

Competing interests

A.E.C., N.L. and D.K. are listed as the authors on a patent application filed by University of Massachusetts Medical School on targeting SEPHS2 in cancer therapy, and on the methodology for hydrogen-selenide detection. All other authors declare no competing interests.

Additional information

Peer review information Primary Handling Editor: Elena Bellafante.

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

Extended data

Extended Data Fig. 1 Panel of toxic metabolites and putative detoxifying enzymes tested.

A panel of metabolites that have documented toxic properties, and the downstream enzymes which use them as substrates, designated as ‘detoxifiers’, are listed. Chemical structures and PMID of references which document the toxicity of each metabolite as a compound are shown.

Extended Data Fig. 2 Further details of effects of SEPHS2 KO and overexpression in cells and in the MDA231 orthotopic xenograft.

a, Viability of cell lines following KO with guides against SEPHS2 for 9 days. Values are relative to the same cell lines expressing nontargeting (CTRL) guide (=1.0), (b) Light microscope images of MDAMB231 and MCF10A cells subjected to CTRL or SEPHS2 KO. Scale bar=25μm. c, Viability of cell lines following KO with guides against SEPHS2 for 11 days. Values are relative to the same cell lines expressing nontargeting guide (=1.0). d, Map of wild type SEPHS2 and CRISPR resistant mutant SEPHS2_U60C. Details of features are provided in Methods. e, Viability of vector and SEPHS2_U60C overexpressing cell lines subjected to CTRL and SEPHS2 g1 KO. f, Weight measurements for ex vivo CTRL and SEPHS2 KO MDAMB231 orthotopic xenograft tumors. g, In vivo tumor volume measurements for CTRL and SEPHS2 KO MDAMB231 orthotopic xenograft tumors. For f and g, if no tumor has formed, tumor weight and volume was designated 0 for CTRL, n = 6 and for SEPHS2 KO n = 2. h, Immunoblot of SEPHS2 in CTRL and SEPHS2 KO MDAMB231 orthotopic xenograft tumors. Number is an animal identifier number. For a,c,e, n = 3 biological replicates; error bars are S.D. Number is an animal identifier number. i, Viabilities of noncancer (immortalized) and cancer lines following KO with guides against SEPHS1 (blue bars) for 9 days. Values are relative to the same cell lines expressing nontargeting guide (black bars), set at 1.0. j, Immunoblots of selenoprotein expression in MCF10A and MDAMB231 cells subjected to (CRISPR/Cas9-induced) KO with guides against SEPHS1, SEPHS2, or control guides for 11 days. For a,c,e and i, n = 3 biological replicates; error bars are S.D. For all panels, the measure of center is mean. *p > 0.05, **p < 0.01 (student’s two-tailed t test).

Source data

Extended Data Fig. 3 Induction of cell death by SEPHS2 KO in cancer cells.

a, Raw flow cytometry of PI and Annexin V double-stained U251 and MDAMB231 cells following KO with guides against SEPHS2 at 10 days after viral infection. Gating strategy is outlined in Extended Data Fig. 10a–e b, Percentages of Annexin V + /PI- and Annexin V + /PI + cells. c, Cleaved caspase 3 (CC3) staining in MDAMB231 and U251cells following KO with guides against SEPHS2 at 10 days after viral transduction. Scale bar=20μm. d, Quantification of CC3 positive cells over total cells in CTRL and SEPHS2 deficient MDAMB231 and U251. Counts are from random fields. e, Viability of control and SEPHS2 deficient U251 and MDAMB231 cells treated with vehicle or the indicated drugs for 4 days. Values are relative to the cell lines expressing nontargeting (CTRL) guide, set at 1.0. For e, n = 3 biological replicates; error bars are S.D. For all panels, the measure of center is mean. For all panels, *p < 0.05, **p < 0.01, and ***p < 0.001 (student’s two-tailed t test).

Source data

Extended Data Fig. 4 Expression of SEPHS2 in various types of tumor and normal tissues.

a, Expression profiles of SEPHS2 in 28 types of normal and tumor tissues. Normalized mRNA-Seq expression profile data from patient tumor tissues and normal tissues was obtained from GEPIA (Tang, Z. et al. (2017) GEPIA: a web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res, 10.1093/nar/gkx247). Tumor subtypes having over 2-fold SEPHS2 expression in tumor than normal and q-value less than 0.01 indicated with red. q values are have been determined by ANOVA and adjusted for FDR (false discovery rate). (b and c) Box plots of SEPHS2 mRNA expression levels in patient tumors and normal breast tissue. SEPHS2 expression was compared in tumor and normal breast samples from TCGA b, and METABRIC (c) datasets. For panel b, minimum is non-tumor (-0.239), tumor (-0.6386), maximum is non-tumor (0.1874), tumor (1.0702) and median is non-tumor (0.0014), tumor (0.1402). For panel c, minimum is non-tumor (1.10097), tumor (1.26305), maximum is non-tumor (3.85158), tumor (5.67272), and median is non-tumor (1.941515), tumor (2.86621).

Extended Data Fig. 5 Candidate transporters tested and xCT functional characterization.

a, A list of selenium transporter candidates examined in the focused screen, rationale for candidacy, and references. b, Viability of control and SLC7A11 KO U251 cells treated with 12 µM of sodium selenite for 48 hr. Values are relative to that of the same cells treated with vehicle (=1.0). c, Viability of control and SLC3A2 KO U251 cells treated with 12 µM of sodium selenite for 48 hr. Values are relative to that of the same cells treated with vehicle (=1.0). d, Total thiol quantification of 24 hr conditioned media from control and SLC7A11 KO U251 cells. e, Total thiol quantification of 24 hr conditioned media from control and SLC3A2 KO U251 cells. Each value was normalized to that of unconditioned medium. f, Viability of U251 cells treated with vehicle, 12 µM sodium selenite, and/or 10 µM Erastin for 72 hr. Values are relative to the cells treated with vehicle (=1.0). g, Viability of control and SLC7A11 KO U251 cells treated with vehicle or 12 µM sodium selenite for 72 hr. Values are relative to the control cells treated with vehicle (=1.0). h, Viability of control and SEPHS2 KO CAL120 cells treated with vehicle or indicated doses of Erastin for 4 days. Values are relative to each untreated cells (=1.0). For b-h, n = 3 biological replicates; error bars are S.D. For all panels, the measure of center is mean. For all panels, *p < 0.05, **p < 0.01, and ***p < 0.001 (student’s two-tailed t test).

Source data

Extended Data Fig. 6 Effects of hypoxia or nutrient-starved stresses on normal and selenophilic cancer cells.

a, Viability of non-transformed (blue bars) and selenophilic cancer (red bars) cells under hypoxia (1% O2) for 48 hr. Values are relative to cells cultured under normoxic condition (20% O2), set at 1.0. b, Viability of non-transformed (blue bars) and selenophilic cancer (red bars) cells under nutrient starved conditions (10% or 20% growth media) for 48 hr. Values are relative to cells cultured in 100% growth media, set at 1.0. c, Viability of control, SEPHS2, GPX4, and SEPSECS deficient MDAMB231 cells after being cultured in hypoxic condition (1% O2) for 48 hr. Values are relative to the cells cultured in normoxic condition (20% O2), set at 1.0. d, Viability of control, SEPHS2, GPX4, and SEPSECS deficient MDAMB231 cells under nutrient starved conditions (10% growth media) for 48 hr. Values are relative to the cells cultured in 100% growth media, set at 1.0. e, Immunoblot of FPS1 and catalase in SEPHS2 KO sensitive and in sensitive cell lines. For a,b, n = 3 biological replicates; error bars are S.D. For c,d n = 3 independent experiments; error bars are S.D. For b, while trends can be seen, statistical comparisons were not carried out due to variability between cell lines. For all panels, the measure of center is mean. For all panels, *p < 0.05 (student’s two-tailed t test).

Source data

Extended Data Fig. 7 SEPHS2 and GPX4 expression in tumor microenvironments.

a, Validation of SEPHS2 RNA probe by RNAscope in situ hybridization in MDA231 cell pellet block samples. Scale bar=100μm. b, Validation of specificity of GPX4 antibody in CTRL and SEPHS2 KO MDAMB231 cell pellet block by immunohistochemistry. c, SEPHS2 immunoblots from the cells used in b. (d and e) Micrographs of serial xenograft tumor sections stained with CA9, GPX4, KI67 antibodies and in situ hybridized with SEPHS2 RNA probe. The right hand panels are magnifications of the regions outlined by the white square. d, shows a hypoxic region as indicated by membrane staining of CA9, and with a low Ki67 index. e, shows a nonhypoxic region (as indicated by only background nonspecific stain in CA9) with a high Ki67 index. Scale bar is 100μm. f, Micrographs of serial human tumor sections stained with CD31 antibody and in situ hybridized with SEPHS2 RNA probe, showing vasculature. Scale bar is 100μm. g, Immunoblots of GPX4, SEPHS2 and Hif1a in MDAMB231 cells subjected to hypoxia (1% O2) for 48hrs.

Source data

Extended Data Fig. 8 Reactive oxygen species production by excess selenium, and implications in SEPHS2 KO toxicity.

a, Selenoprotein immunoblots of MDAMB231 cells at 24 hr recovery after 2 hr sodium selenite treatment or treated with 2-selenocysteine-containing peptide for 24 hr. b, ROS quantification of cells treated with indicated concentrations of sodium selenite or 2xselenocysteine containing peptide, 250 µM TBH, or 500μM H2O2 for 45 mins. c, Viability of U251 cells overexpressing vector or CAT treated with vehicle or H2O2 for 48 hr. d, Viability of U251, and MDAMB231 cells overexpressing blank vector or CAT and subjected to KO with guides against SEPHS2 for 9 days. e, Immunoblots of CAT and SEPHS2 in U251 and MDAMB231 cells overexpressing blank vector or CAT and subjected to KO with guides against SEPHS2 for 9 days. For b, n = 3 independent experiments; error bars are S.D. For c,d n = 3 biological replicates; error bars are S.D. For all panels, the measure of center is mean. For all panels, *p < 0.05 and **p < 0.01 (student’s two-tailed t test).

Source data

Extended Data Fig. 9 Confirmation of gene knockout or overexpression after lentiviral transduction.

a, Immunoblots of SEPHS2 in 5 non-transformed (blue) and 5 transformed (black) cell lines transduced with Cas9/sgRNAs against SEPHS2, and control (non-targeting sgRNA). b, Immunoblots of SEPHS2 in cells transduced with CRISPR resistant SEPHS2_U60C and control (empty vector). c, Immunoblots of SLC7A11 in cells transduced with Cas9/sgRNAs targeting SLC7A11 or non-targeting sgRNA. d, Immunoblots of SLC3A2 in cells transduced as indicated e, Immunoblots of PSTK in cells transduced as indicated. f, Immunoblots of SEPHS1 in 2 non-transformed and 2 cancer cells transduced as indicated. g, Immunoblots of GPX4 in cells transduced as indicated. h, Immunoblots of FSP1 in cancer cells transduced as indicated. All samples were lysed between 7~8 days after viral transduction.

Source data

Extended Data Fig. 10 Supporting data for flow cytometry and gas toxicity system.

ae, Representative flow cytometry data to address gating and quadrant strategy for FACS measurement of dying/dead cells. MDAMB231 cells were stained with Annexin V-FITC and propidium iodide (PI). The X-axis and y-axis of Flow cytometry density plots were designated as Annexin V-FITC and PI, respectively. The left lower quadrant represents Annexin V negative and PI negative live cells. The right lower quadrant represents Annexin V positive and PI negative early dying cells. The right upper quadrant represents Annexin V positive and PI positive late dying/dead cells. Staining is as follows: (a) unlabeled, (b) PI, (c) Annexin V, (d) Annexin V and PI, (e) Annexin V and PI stained cells treated with 50 µM H2O2 for 24 hr. f, Scanned image of a polystyrene microplate cover with PVP spots embedded with silver nitrate or lead acetate. The plate cover was scanned before (upper panel) and after (lower panel) selenide gas exposure for one min. g, Viability of MDAMB231 cells in the numbered spots outlined on the 96 well plate, exposed to selenide gas. For g, n = 4 independent experiments; values are normalized to untreated cells (=1.0). For all panels, the measure of center is mean. For all panels **p < 0.01, and ***p < 0.001 (student’s two-tailed t test).

Source data

Supplementary information

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. 2

Unprocessed Western Blots

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 Extended Data Fig. 2

Statistical Source Data

Source Data Extended Data Fig. 2

Unprocessed Western Blots

Source Data Extended Data Fig. 3

Statistical Source Data

Source Data Extended Data Fig. 5

Statistical Source Data

Source Data Extended Data Fig. 6

Statistical Source Data

Source Data Extended Data Fig. 6

Unprocessed Western Blots

Source Data Extended Data Fig. 7

Unprocessed Western Blots

Source Data Extended Data Fig. 8

Statistical Source Data

Source Data Extended Data Fig. 8

Unprocessed Western Blots

Source Data Extended Data Fig. 9

Unprocessed Western Blots

Source Data Extended Data Fig. 10

Statistical Source Data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Carlisle, A.E., Lee, N., Matthew-Onabanjo, A.N. et al. Selenium detoxification is required for cancer-cell survival. Nat Metab 2, 603–611 (2020). https://doi.org/10.1038/s42255-020-0224-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-020-0224-7

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