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:

Global survey of cell death mechanisms reveals metabolic regulation of ferroptosis

Abstract

Apoptosis is one type of programmed cell death. Increasingly, non-apoptotic cell death is recognized as being genetically controlled, or 'regulated'. However, the full extent and diversity of alternative cell death mechanisms remain uncharted. Here we surveyed the landscape of pharmacologically accessible cell death mechanisms. In an examination of 56 caspase-independent lethal compounds, modulatory profiling showed that 10 compounds induced three different types of regulated non-apoptotic cell death. Optimization of one of those ten resulted in the discovery of FIN56, a specific inducer of ferroptosis. Ferroptosis has been found to occur when the lipid-repair enzyme GPX4 is inhibited. FIN56 promoted degradation of GPX4. FIN56 also bound to and activated squalene synthase, an enzyme involved in isoprenoid biosynthesis, independent of GPX4 degradation. These discoveries show that dysregulation of lipid metabolism is associated with ferroptosis. This systematic approach is a means to discover and characterize novel cell death phenotypes.

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

Access options

Buy this article

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

Figure 1: Modulatory profiling revealed three types of regulated non-apoptotic cell death.
Figure 2: Optimization of CIL56 revealed a potent and selective ferroptosis inducer.
Figure 3: FIN56-induced ferroptosis decreases GPX4 expression.
Figure 4: SQS encoded by FDFT1 as FIN56's target protein.
Figure 5: Validating SQS as the functionally relevant target for FIN56's lethality.
Figure 6: ACC inhibitor prevents GPX4 protein degradation.

Similar content being viewed by others

References

  1. Fuchs, Y. & Steller, H. Programmed cell death in animal development and disease. Cell 147, 742–758 (2011).

    Article  CAS  Google Scholar 

  2. Aravind, L., Dixit, V.M. & Koonin, E.V. The domains of death: evolution of the apoptosis machinery. Trends Biochem. Sci. 24, 47–53 (1999).

    Article  CAS  Google Scholar 

  3. Degterev, A. et al. Identification of RIP1 kinase as a specific cellular target of necrostatins. Nat. Chem. Biol. 4, 313–321 (2008).

    Article  CAS  Google Scholar 

  4. Vanden Berghe, T., Linkermann, A., Jouan-Lanhouet, S., Walczak, H. & Vandenabeele, P. Regulated necrosis: the expanding network of non-apoptotic cell death pathways. Nat. Rev. Mol. Cell Biol. 15, 135–147 (2014).

    Article  CAS  Google Scholar 

  5. Kaczmarek, A., Vandenabeele, P. & Krysko, D.V. Necroptosis: the release of damage-associated molecular patterns and its physiological relevance. Immunity 38, 209–223 (2013).

    Article  CAS  Google Scholar 

  6. Kono, H. & Rock, K.L. How dying cells alert the immune system to danger. Nat. Rev. Immunol. 8, 279–289 (2008).

    Article  CAS  Google Scholar 

  7. Skouta, R. et al. Ferrostatins inhibit oxidative lipid damage and cell death in diverse disease models. J. Am. Chem. Soc. 136, 4551–4556 (2014).

    Article  CAS  Google Scholar 

  8. Linkermann, A. et al. Synchronized renal tubular cell death involves ferroptosis. Proc. Natl. Acad. Sci. USA 111, 16836–16841 (2014).

    Article  CAS  Google Scholar 

  9. Dixon, S.J. et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell 149, 1060–1072 (2012).

    Article  CAS  Google Scholar 

  10. Dixon, S.J. et al. Pharmacological inhibition of cystine-glutamate exchange induces endoplasmic reticulum stress and ferroptosis. eLife 3, e02523 (2014).

    Article  Google Scholar 

  11. Hayano, M., Yang, W.S., Corn, C.K., Pagano, N.C. & Stockwell, B.R. Loss of cysteinyl-tRNA synthetase (CARS) induces the transsulfuration pathway and inhibits ferroptosis induced by cystine deprivation. Cell Death Differ. 23, 270–278 (2016).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  13. Imai, H. & Nakagawa, Y. Biological significance of phospholipid hydroperoxide glutathione peroxidase (PHGPx, GPx4) in mammalian cells. Free Radic. Biol. Med. 34, 145–169 (2003).

    Article  CAS  Google Scholar 

  14. Linkermann, A., Stockwell, B.R., Krautwald, S. & Anders, H.-J. Regulated cell death and inflammation: an auto-amplification loop causes organ failure. Nat. Rev. Immunol. 14, 759–767 (2014).

    Article  CAS  Google Scholar 

  15. Hitomi, J. et al. Identification of a molecular signaling network that regulates a cellular necrotic cell death pathway. Cell 135, 1311–1323 (2008).

    Article  CAS  Google Scholar 

  16. Hahn, W.C. et al. Creation of human tumour cells with defined genetic elements. Nature 400, 464–468 (1999).

    Article  CAS  Google Scholar 

  17. Wolpaw, A.J. et al. Modulatory profiling identifies mechanisms of small molecule-induced cell death. Proc. Natl. Acad. Sci. USA 108, E771–E780 (2011).

    Article  CAS  Google Scholar 

  18. Degterev, A. et al. Chemical inhibitor of nonapoptotic cell death with therapeutic potential for ischemic brain injury. Nat. Chem. Biol. 1, 112–119 (2005).

    Article  CAS  Google Scholar 

  19. Takahashi, N. et al. Necrostatin-1 analogues: critical issues on the specificity, activity and in vivo use in experimental disease models. Cell Death Dis. 3, e437 (2012).

    Article  CAS  Google Scholar 

  20. Yang, W.S. & Stockwell, B.R. Synthetic lethal screening identifies compounds activating iron-dependent, nonapoptotic cell death in oncogenic-RAS-harboring cancer cells. Chem. Biol. 15, 234–245 (2008).

    Article  CAS  Google Scholar 

  21. Shoemaker, R.H. The NCI60 human tumour cell line anticancer drug screen. Nat. Rev. Cancer 6, 813–823 (2006).

    Article  CAS  Google Scholar 

  22. Shimada, K., Hayano, M., Pagano, N.C. & Stockwell, B.R. Cell-line selectivity improves the predictive power of pharmacogenomic analyses and helps identify NADPH as biomarker for ferroptosis sensitivity. Cell Chem. Biol. 23, 225–235 (2016).

    Article  CAS  Google Scholar 

  23. Romanowska, M. et al. Effects of selenium supplementation on expression of glutathione peroxidase isoforms in cultured human lung adenocarcinoma cell lines. Lung Cancer 55, 35–42 (2007).

    Article  Google Scholar 

  24. Fradejas, N. et al. Mammalian Trit1 is a tRNA([Ser]Sec)-isopentenyl transferase required for full selenoprotein expression. Biochem. J. 450, 427–432 (2013).

    Article  CAS  Google Scholar 

  25. Song, J.H. et al. Deletion of Pim kinases elevates the cellular levels of reactive oxygen species and sensitizes to K-Ras-induced cell killing. Oncogene 34, 3728–3736 (2015).

    Article  CAS  Google Scholar 

  26. Tansey, T.R. & Shechter, I. Structure and regulation of mammalian squalene synthase. Biochim. Biophys. Acta 1529, 49–62 (2000).

    Article  CAS  Google Scholar 

  27. Liu, C.-I. et al. Acta Crystallogr. D Biol. Crystallogr. 70, 231–241 (2014).

    Article  CAS  Google Scholar 

  28. Chugh, A., Ray, A. & Gupta, J.B. Squalene epoxidase as hypocholesterolemic drug target revisited. Prog. Lipid Res. 42, 37–50 (2003).

    Article  CAS  Google Scholar 

  29. Gueven, N., Woolley, K. & Smith, J. Border between natural product and drug: comparison of the related benzoquinones idebenone and coenzyme Q10 . Redox Biol. 4, 289–295 (2015).

    Article  CAS  Google Scholar 

  30. Dixon, S.J. et al. Human haploid cell genetics reveals roles for lipid metabolism genes in nonapoptotic cell death. ACS Chem. Biol. 10, 1604–1609 (2015).

    Article  CAS  Google Scholar 

  31. Hirsch, H.A. et al. A transcriptional signature and common gene networks link cancer with lipid metabolism and diverse human diseases. Cancer Cell 17, 348–361 (2010).

    Article  CAS  Google Scholar 

  32. Santos, C.R. & Schulze, A. Lipid metabolism in cancer. FEBS J. 279, 2610–2623 (2012).

    Article  CAS  Google Scholar 

  33. Kamphorst, J.J., Fan, J., Lu, W., White, E. & Rabinowitz, J.D. Liquid chromatography-high resolution mass spectrometry analysis of fatty acid metabolism. Anal. Chem. 83, 9114–9122 (2011).

    Article  CAS  Google Scholar 

  34. Yagoda, N. et al. RAS-RAF-MEK-dependent oxidative cell death involving voltage-dependent anion channels. Nature 447, 864–868 (2007).

    Article  Google Scholar 

  35. Cholody, W.M. et al. Derivatives of fluorene, anthracene, xanthene, dibenzosuberone and acridine and uses thereof. US patent application PCT/US2008/006015 (2008).

  36. Backman, T.W.H., Cao, Y. & Girke, T. ChemMine tools: an online service for analyzing and clustering small molecules. Nucleic Acids Res. 39, W486–W491 (2011).

    Article  CAS  Google Scholar 

  37. Alegre-Aguarón, E. et al. Growth factor priming differentially modulates components of the extracellular matrix proteome in chondrocytes and synovium-derived stem cells. PLoS One 9, e88053 (2014).

    Article  Google Scholar 

Download references

Acknowledgements

We thank H. Li (Columbia University, New York, New York, USA) and C. Henderson (Biogen Idec, Cambridge, Massachusetts, USA) for providing mevalonic acid and farnesyl pyrophosphate, C.-I. Liu and A.H.J. Wang (Academia Sinica, Taipei, Taiwan) for the truncated human squalene synthase construct, R. Weinberg (Whitehead Institute, Cambridge, Massachusetts, USA) for engineered BJ cell lines (BJeLR, DRD, BJeHLT, and BJeH), E. Schon (Columbia University, New York, New York, USA) for 143B cells, E. Lee for assistance with ACC experiments, and V. Viswanathan for helpful discussions. This research was funded by the Howard Hughes Medical Institute, the US National Institutes of Health (grants 5R01CA097061, 5R01GM085081, and R01CA161061 to B.R.S.), New York Stem Cell Science (grant C026715 to B.R.S.) and the US National Cancer Institute (K99 Pathway to Independence Award 1K99CA166517-01 to S.J.D.).

Author information

Authors and Affiliations

Authors

Contributions

K.S. and B.R.S. conceived of the project, designed the experiments, analyzed the data, and wrote the manuscript. K.S. performed all experiments and analyses except the following: R.S. and C.A.V. synthesized CIL56 analogs; A.K. expressed and purified SQS protein and performed confirmatory SQS pulldown experiments and competition assays; W.S.Y. performed the GPX4 enzymatic assay; and M.H. performed confirmatory siRNA experiments. L.M.B. performed proteomic analysis for target identification. S.J.D. participated in characterization of TOFA's effect on FIN56. A.J.W. assisted in modulatory profiling experiments. S.J.D. assisted in writing the manuscript.

Corresponding author

Correspondence to Brent R Stockwell.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Results, Supplementary Figures 1–14 and Supplementary Tables 1–3. (PDF 5029 kb)

Supplementary Data Set 1

Lists of lethal compounds, chemical death modulators, cell lines, and modulatory profiles of characterized and uncharacterized lethal compounds. (XLSX 104 kb)

Supplementary Note 1

Supplementary Note 1 (PDF 2402 kb)

Supplementary Note 2

Synthetic Procedures (PDF 1066 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shimada, K., Skouta, R., Kaplan, A. et al. Global survey of cell death mechanisms reveals metabolic regulation of ferroptosis. Nat Chem Biol 12, 497–503 (2016). https://doi.org/10.1038/nchembio.2079

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nchembio.2079

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