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:

DHODH-mediated ferroptosis defence is a targetable vulnerability in cancer

Matters Arising to this article was published on 05 July 2023

An Author Correction to this article was published on 02 August 2021

This article has been updated

Abstract

Ferroptosis, a form of regulated cell death that is induced by excessive lipid peroxidation, is a key tumour suppression mechanism1,2,3,4. Glutathione peroxidase 4 (GPX4)5,6 and ferroptosis suppressor protein 1 (FSP1)7,8 constitute two major ferroptosis defence systems. Here we show that treatment of cancer cells with GPX4 inhibitors results in acute depletion of N-carbamoyl-l-aspartate, a pyrimidine biosynthesis intermediate, with concomitant accumulation of uridine. Supplementation with dihydroorotate or orotate—the substrate and product of dihydroorotate dehydrogenase (DHODH)—attenuates or potentiates ferroptosis induced by inhibition of GPX4, respectively, and these effects are particularly pronounced in cancer cells with low expression of GPX4 (GPX4low). Inactivation of DHODH induces extensive mitochondrial lipid peroxidation and ferroptosis in GPX4low cancer cells, and synergizes with ferroptosis inducers to induce these effects in GPX4high cancer cells. Mechanistically, DHODH operates in parallel to mitochondrial GPX4 (but independently of cytosolic GPX4 or FSP1) to inhibit ferroptosis in the mitochondrial inner membrane by reducing ubiquinone to ubiquinol (a radical-trapping antioxidant with anti-ferroptosis activity). The DHODH inhibitor brequinar selectively suppresses GPX4low tumour growth by inducing ferroptosis, whereas combined treatment with brequinar and sulfasalazine, an FDA-approved drug with ferroptosis-inducing activity, synergistically induces ferroptosis and suppresses GPX4high tumour growth. Our results identify a DHODH-mediated ferroptosis defence mechanism in mitochondria and suggest a therapeutic strategy of targeting ferroptosis in cancer treatment.

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: Metabolomics link DHODH to ferroptosis.
Fig. 2: DHODH deletion promotes ferroptosis.
Fig. 3: DHODH suppresses mitochondrial lipid peroxidation.
Fig. 4: Inhibition of DHODH suppresses tumour growth by inducing ferroptosis.

Similar content being viewed by others

Data availability

All data that support the conclusions in this manuscript are available from the corresponding author upon reasonable request. The source data of immunoblots are provided. The raw data used for generating Figs. 14 and Extended Data Figs. 19 are included in the Source Data. Source data are provided with this paper.

Change history

References

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Stockwell, B. R. et al. Ferroptosis: a regulated cell death nexus linking metabolism, redox biology, and disease. Cell 171, 273–285 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  4. Zhang, Y. et al. BAP1 links metabolic regulation of ferroptosis to tumour suppression. Nat. Cell Biol. 20, 1181–1192 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Friedmann Angeli, J. P. et al. Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nat. Cell Biol. 16, 1180–1191 (2014).

    Article  CAS  PubMed  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  Google Scholar 

  9. Koppula, P., Zhuang, L. & Gan, B. Cystine transporter SLC7A11/xCT in cancer: ferroptosis, nutrient dependency, and cancer therapy. Protein Cell https://doi.org/10.1007/s13238-020-00789-5 (2020).

  10. Doll, S. et al. ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat. Chem. Biol. 13, 91–98 (2017).

    Article  CAS  PubMed  Google Scholar 

  11. Basu, A. et al. An interactive resource to identify cancer genetic and lineage dependencies targeted by small molecules. Cell 154, 1151–1161 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Vasan, K., Werner, M. & Chandel, N. S. Mitochondrial metabolism as a target for cancer therapy. Cell Metab. 32, 341–352 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Arai, M. et al. Import into mitochondria of phospholipid hydroperoxide glutathione peroxidase requires a leader sequence. Biochem. Biophys. Res. Commun. 227, 433–439 (1996).

    Article  CAS  PubMed  Google Scholar 

  14. Trnka, J., Blaikie, F. H., Smith, R. A. & Murphy, M. P. A mitochondria-targeted nitroxide is reduced to its hydroxylamine by ubiquinol in mitochondria. Free Radic. Biol. Med. 44, 1406–1419 (2008).

    Article  CAS  PubMed  Google Scholar 

  15. Prime, T. A. et al. A ratiometric fluorescent probe for assessing mitochondrial phospholipid peroxidation within living cells. Free Radic. Biol. Med. 53, 544–553 (2012).

    Article  CAS  PubMed  Google Scholar 

  16. Shimada, K. et al. Global survey of cell death mechanisms reveals metabolic regulation of ferroptosis. Nat. Chem. Biol. 12, 497–503 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Hakkaart, G. A., Dassa, E. P., Jacobs, H. T. & Rustin, P. Allotopic expression of a mitochondrial alternative oxidase confers cyanide resistance to human cell respiration. EMBO Rep. 7, 341–345 (2006).

    Article  CAS  PubMed  Google Scholar 

  18. Peters, G. J. et al. In vivo inhibition of the pyrimidine de novo enzyme dihydroorotic acid dehydrogenase by brequinar sodium (DUP-785; NSC 368390) in mice and patients. Cancer Res. 50, 4644–4649 (1990).

    CAS  PubMed  Google Scholar 

  19. Natale, R. et al. Multicenter phase II trial of brequinar sodium in patients with advanced melanoma. Ann. Oncol. 3, 659–660 (1992).

    Article  CAS  PubMed  Google Scholar 

  20. Urba, S. et al. Multicenter phase II trial of brequinar sodium in patients with advanced squamous-cell carcinoma of the head and neck. Cancer Chemother. Pharmacol. 31, 167–169 (1992).

    Article  CAS  PubMed  Google Scholar 

  21. Maroun, J. et al. Multicenter phase II study of brequinar sodium in patients with advanced lung cancer. Cancer Chemother. Pharmacol. 32, 64–66 (1993).

    Article  CAS  PubMed  Google Scholar 

  22. Lei, G. et al. The role of ferroptosis in ionizing radiation-induced cell death and tumor suppression. Cell Res. 30, 146–162 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Gout, P. W., Buckley, A. R., Simms, C. R. & Bruchovsky, N. Sulfasalazine, a potent suppressor of lymphoma growth by inhibition of the xc cystine transporter: a new action for an old drug. Leukemia 15, 1633–1640 (2001).

    Article  CAS  PubMed  Google Scholar 

  24. Zou, Y. et al. A GPX4-dependent cancer cell state underlies the clear-cell morphology and confers sensitivity to ferroptosis. Nat. Commun. 10, 1617 (2019).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  25. Soula, M. et al. Metabolic determinants of cancer cell sensitivity to canonical ferroptosis inducers. Nat. Chem. Biol. 16, 1351–1360 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Gao, M. et al. Role of mitochondria in ferroptosis. Mol. Cell 73, 354–363.e3 (2019).

    Article  CAS  PubMed  Google Scholar 

  27. Lee, H. et al. Energy-stress-mediated AMPK activation inhibits ferroptosis. Nat. Cell Biol. 22, 225–234 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Wang, W. et al. CD8+ T cells regulate tumour ferroptosis during cancer immunotherapy. Nature 569, 270–274 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  29. Lang, X. et al. Radiotherapy and immunotherapy promote tumoral lipid oxidation and ferroptosis via synergistic repression of SLC7A11. Cancer Discov. 9, 1673–1685 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Ye, L. F. et al. Radiation-induced lipid peroxidation triggers ferroptosis and synergizes with ferroptosis inducers. ACS Chem. Biol. 15, 469–484 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Zhang, Y., Koppula, P. & Gan, B. Regulation of H2A ubiquitination and SLC7A11 expression by BAP1 and PRC1. Cell Cycle 18, 773–783 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Chauhan, A. S. et al. STIM2 interacts with AMPK and regulates calcium-induced AMPK activation. FASEB J. 33, 2957–2970 (2019).

    Article  CAS  PubMed  Google Scholar 

  33. Liu, X. et al. Cystine transporter regulation of pentose phosphate pathway dependency and disulfide stress exposes a targetable metabolic vulnerability in cancer. Nat. Cell Biol. 22, 476–486 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Heinrich, P. et al. Correcting for natural isotope abundance and tracer impurity in MS-, MS/MS- and high-resolution-multiple-tracer-data from stable isotope labeling experiments with IsoCorrectoR. Sci. Rep. 8, 17910 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  35. Koppula, P., Zhang, Y., Shi, J., Li, W. & Gan, B. The glutamate/cystine antiporter SLC7A11/xCT enhances cancer cell dependency on glucose by exporting glutamate. J. Biol. Chem. 292, 14240–14249 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Liu, X. & Gan, B. lncRNA NBR2 modulates cancer cell sensitivity to phenformin through GLUT1. Cell Cycle 15, 3471–3481 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Dai, F. et al. BAP1 inhibits the ER stress gene regulatory network and modulates metabolic stress response. Proc. Natl Acad. Sci. USA 114, 3192–3197 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  38. Zhang, Y. et al. H2A monoubiquitination links glucose availability to epigenetic regulation of the endoplasmic reticulum stress response and cancer cell death. Cancer Res. 80, 2243–2256 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Fang, J. et al. Dihydro-orotate dehydrogenase is physically associated with the respiratory complex and its loss leads to mitochondrial dysfunction. Biosci. Rep. 33, e00021 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Nagase, M., Yamamoto, Y., Mitsui, J. & Tsuji, S. Simultaneous detection of reduced and oxidized forms of coenzyme Q10 in human cerebral spinal fluid as a potential marker of oxidative stress. J. Clin. Biochem. Nutr. 63, 205–210 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Zhang, Y. et al. Imidazole ketone erastin induces ferroptosis and slows tumor growth in a mouse lymphoma model. Cell Chem. Biol. 26, 623–633.e9 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Liu, X. et al. LncRNA NBR2 engages a metabolic checkpoint by regulating AMPK under energy stress. Nat. Cell Biol. 18, 431–442 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Lee, H. et al. BAF180 regulates cellular senescence and hematopoietic stem cell homeostasis through p21. Oncotarget 7, 19134–19146 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Lin, A. et al. The FoxO-BNIP3 axis exerts a unique regulation of mTORC1 and cell survival under energy stress. Oncogene 33, 3183–3194 (2014).

    Article  CAS  PubMed  Google Scholar 

  45. Lin, A. et al. FoxO transcription factors promote AKT Ser473 phosphorylation and renal tumor growth in response to pharmacologic inhibition of the PI3K-AKT pathway. Cancer Res. 74, 1682–1693 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Gan, B. et al. FoxOs enforce a progression checkpoint to constrain mTORC1-activated renal tumorigenesis. Cancer Cell 18, 472–484 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Gan, B. et al. Role of FIP200 in cardiac and liver development and its regulation of TNFalpha and TSC-mTOR signaling pathways. J. Cell Biol. 175, 121–133 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We apologize to the colleagues whose relevant work cannot be cited here owing to space limitations. This research was supported by Institutional Research Fund from The University of Texas MD Anderson Cancer Center, and grants R01CA181196, R01CA190370, R01CA244144, R01CA247992 from the National Institutes of Health (to B.G.). PDX generation and annotation were supported by the University of Texas MD Anderson Cancer Center Moon Shots Program, Specialized Program of Research Excellence (SPORE) grant CA070907 and University of Texas PDX Development and Trial Center grant U54CA224065. This research was also supported by the National Institutes of Health Cancer Center Support Grant P30CA016672 to The University of Texas MD Anderson Cancer Center.

Author information

Authors and Affiliations

Authors

Contributions

C.M. performed most of the experiments with assistance from X.L., Y.Z., G.L., Y.Y., H.L., P.K., S.W. and L.Z.; K.O. conducted all metabolomic analyses; B.F. provided PDXs used in this study; M.V.P. provided resources for the project; B.G., C.M., and K.O. designed the experiments; B.G. supervised the study, established collaborations, allocated funding for this study, and wrote most of the manuscript with assistance from K.O. and C.M.; and all authors commented on the manuscript.

Corresponding authors

Correspondence to Kellen Olszewski or Boyi Gan.

Ethics declarations

Competing interests

K.O. and M.V.P. are full-time employees of Kadmon Corporation, LLC. B.G., K.O., and M.C. have filed a patent application relating to the use of DHODH inhibitors to target ferroptosis in cancer therapy. Other authors declare no competing financial interests.

Additional information

Peer review information Nature thanks Kivanç Birsoy and Navdeep Chandel for their contribution to the peer review of this work. Peer reviewer reports are available.

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

Extended data figures and tables

Extended Data Fig. 1 Pharmacological inhibition of GPX4 affects intermediate levels in the de novo pyrimidine biosynthesis pathway.

ac, Volcano plots comparing metabolomic profiles from HT-1080 (a), A-498 (b) or RCC4 (c) cells treated with vehicle and the same cells treated with RSL3 (10 μM) or ML162 (10 μM) for 2 h. d, e, Fold change in C-Asp and uridine induced by RSL3 (10 μM) or ML162 (10 μM) treatment for 2 h compared with vehicle treatment in A-498 (d) or RCC4 (e) cells. f, Simplified schematic of de novo pyrimidine biosynthesis pathway. g, Fold change in intracellular DHO and OA levels upon treatment with vehicle, DHO (100 μM) or OA (100 μM), respectively, for 48 h in NCI-H226 cells. h, Fold change in intracellular C-Asp levels upon treatment with vehicle or C-Asp (100 μM) for 48 h in NCI-H226 cells. i, DHO activity in HT-1080 cells treated with RSL3 (10 μM) for 2 h, following pretreatment with vehicle, OA (100 μM) for 24 h, or Lip-1 (10 μM) for 48 h. j, GPX4 protein levels in different cell lines determined by western blotting. k, Cell viability in TK-10, UMRC2, A-498 and RCC4 cells treated with different doses of RSL3 for 4 h, following pretreatment with vehicle, C-Asp (100 μM), DHO (100 μM), OA (100 μM), or uridine (50 μM) for 48 h. l, Cell viability in SW620, U-87 MG, A549, NCI-H1437, MDA-MB-436 and MDA-MB-231 cells treated with different doses of RSL3 for 4 h, following pretreatment with vehicle, DHO (100 μM) or OA (100 μM) for 48 h. m, GPX4, DHODH, and FSP1 protein levels in different cancer cell lines determined by western blotting. n, Cell viability in GPX4high (HT-1080, A-498, RCC4, 786-O, and 769-P) and GPX4low (HCT-8, UMRC6, TK-10, UMRC2, and NCI-H226) cells treated with different doses of the DHODH inhibitors BQR, leflunomide (LFM), or teriflunomide (TF) for 4 h. Data are presented as mean ± s.d., n = 3 independent repeats; unpaired, two-tailed t-test. Western blots are representative of two biological replicates. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. ns, not significant. Asp, aspartate; C-P, carbamoyl phosphate; P, phosphate; FMN, flavin mononucleotide; FMNH2, reduced flavin mononucleotide; PRPP, phosphoribosyl pyrophosphate; PPi, inorganic pyrophosphate; OMP, orotidine 5′-monophosphate; UMP, uridine 5′-monophosphate.

Source data

Extended Data Fig. 2 The effect of DHODH inhibitors on inducing ferroptosis in different cancer cells with differential expression of GPX4.

a, b, Cell survival fraction and PTGS2 mRNA levels in NCI-H226 (a) and HT-1080 (b) cells upon treatment with BQR (500 μM for NCI-H226 cells; 5 mM for HT-1080 cells), following pretreatment with vehicle, ZVF (10 μM), and/or Lip-1 (10 μM) for 24 h. c, Cell viability in HT-1080 cells treated with different doses of RSL3 and co-treated with LFM (100 μM) or TF (500 μM) for 4 h, following pretreatment with vehicle or Lip-1 (10 μM) for 24 h. d, Cell viability in HT-1080 cells treated with different doses of ML162 and co-treated with BQR (500 μM), LFM (100 μM), or TF (500 μM) for 4 h, following pretreatment with vehicle or Lip-1 (10 μM) for 24 h. e, Cell survival fraction and PTGS2 mRNA levels in HT-1080 cells upon treatment with RSL3 (1 μM) and/or BQR (500 μM) for 4 h, following pretreatment with vehicle or Lip-1 (10 μM) for 24 h. f, Cell viability in HT-1080 cells treated with different doses of sulfasalazine (SAS) and co-treated with BQR (500 μM), LFM (100 μM) or TF (500 μM) for 4 h, following pretreatment with vehicle or Lip-1 (10 μM) for 24 h. g, Cell viability in HT-1080 cells treated with different doses of erastin and co-treated with BQR (500 μM), LFM (100 μM) or TF (500 μM) for 4 h, following pretreatment with vehicle or Lip-1 (10 μM) for 24 h. h, mRNA levels of SLC7A11, GPX4, or ACSL4 (bar charts) and their protein expression (western blot), were measured in HT-1080 cells treated with BQR (500 μM), LFM (100 μM), or TF (500 μM) for 4 h. i, GSH level measurement in HT-1080 cells upon treatment with BQR (500 μM), LFM (100 μM), or TF (500 μM) for 2 h. Data are presented as mean ± s.d., n = 3 independent repeats; unpaired, two-tailed t-test. Western blot is representative of two biological replicates. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. ns, not significant.

Source data

Extended Data Fig. 3 DHODH deletion sensitizes GPX4high cancer cells to ferroptosis or induces ferroptosis in GPX4low cancer cells.

a, DHODH protein levels in Cas9 control and DHODH KO GPX4high cancer cell lines. b, DHO activity in Cas9 control and DHODH KO HT-1080 cells. c, Cell survival fraction in Cas9 control and DHODH KO HT-1080 cells upon treatment with vehicle or uridine (50 μM). d, PTGS2 mRNA levels in Cas9 control and DHODH KO HT-1080 cells. e, Lipid peroxidation in Cas9 control and DHODH KO GPX4high cell lines as indicated. f, Cell viability in Cas9 control and DHODH KO HT-1080 cells treated with different doses of ML162 for 4 h. g, Cell survival fraction and PTGS2 mRNA levels in Cas9 control and DHODH KO HT-1080 cells upon treatment with RSL3 (1 μM) for 4 h. h, Western blot analysis of DHODH and ACSL4 protein levels in HT-1080 cells with indicated genotypes. i, Cell viability measurement in HT-1080 cells with indicated genotypes treated with different doses of RSL3 for 4 h. j, Measurement of SLC7A11, GPX4, and ACSL4 mRNA (bar charts) and protein levels (western blot) in Cas9 control and DHODH KO HT-1080 cells. k, GSH levels in Cas9 control and DHODH KO HT-1080 cells. l, DHODH protein levels in Cas9 control and DHODH KO GPX4low cell lines. m, DHO activity in Cas9 control and DHODH KO NCI-H226 cells. n, Cell proliferation of Cas9 control and DHODH KO NCI-H226 cells. o, PTGS2 mRNA levels in Cas9 control and DHODH KO NCI-H226 cells. p, Lipid peroxidation in Cas9 control and DHODH KO GPX4low cells. Cells were grown in medium supplemented with Lip-1 (10 μM) (l, m) and/or uridine (50 μM) (a, b, dp). Data are presented as mean ± s.d., n = 3 independent repeats; unpaired, two-tailed t-test. Western blots are representative of two biological replicates. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. ns, not significant.

Source data

Extended Data Fig. 4 Analyses of genetic interactions between DHODH and GPX4 (or FSP1).

a, Western blotting analysis of GPX4 and DHODH protein levels in shControl and shGPX4 HT-1080 cells. b, Cell proliferation of shControl and shGPX4 HT-1080 cells. c, Cell viability of shControl and shGPX4 HT-1080 cells treated with different doses of LFM or TF for 4 h. d, Cell survival fraction and PTGS2 mRNA levels in shControl and shGPX4 HT-1080 cells upon treatment with BQR (500 μM) for 4 h. e, Western blot analysis of GPX4 and DHODH protein levels in HT-1080 cells with indicated genotypes. f, PTGS2 mRNA levels in HT-1080 cells with indicated genotypes. g, Cell proliferation of HT-1080 cells with DHODH KO and shControl or shGPX4. h, Western blot analysis of DHODH and FSP1 protein levels in HT-1080 cells with indicated genotypes. i, Cell viability in Cas9 control or DHODH KO HT-1080 cells with indicated genotypes treated with different doses of RSL3 for 4 h, following pretreatment with vehicle or Lip-1 (10 μM) for 24 h. j, Western blot analysis of DHODH and FSP1 protein levels in HT-1080 cells with indicated genotypes. k, Cell viability in Cas9 control or DHODH KO HT-1080 cells with indicated genotypes treated with different doses of RSL3 for 4 h, following pretreatment with vehicle or Lip-1 (10 μM) for 24 h. l, Cell viability in Cas9 control or FSP1 KO HT-1080 cells treated with vehicle or BQR (500 μM), and different doses of RSL3 for 4 h. m, Simplified schematic of DHODH protein and its mutants. n, Western blotting showing DHODH protein levels in cytosolic and mitochondrial fractions from DHODH KO HT-1080 cells that express the indicated DHODH constructs. o, DHO activity in DHODH KO HT-1080 cells that express the indicated DHODH constructs. p, Cell viability in DHODH KO HT-1080 cells that express the indicated DHODH constructs treated with different doses of ML162 for 4 h. q, Cell survival fraction, lipid peroxidation and PTGS2 mRNA levels in DHODH KO HT-1080 cells that express the indicated DHODH constructs upon treatment with RSL3 (1 μM). Cells were grown in medium supplemented with uridine (50 μM) (el, nq). Data are presented as mean ± s.d., n = 3 independent repeats (bd, f, g, i, k, l, oq); unpaired, two-tailed t-test. Western blots are representative of two biological replicates. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. ns, not significant. MTS, mitochondrial targeting sequence; DHOD domain, dihydroorotate dehydrogenase domain.

Source data

Extended Data Fig. 5 DHODH cooperates with mitochondrial GPX4 to suppress ferroptosis.

a, Western blotting analysis of GPX4 levels in cytosolic and mitochondrial fractions in a panel of cancer cell lines. b, Simplified schematic of cytosolic and mitochondrial GPX4 protein constructs. c, Western blotting showing GPX4 protein levels in cytosolic and mitochondrial fractions from shGPX4 HT-1080 cells that express the indicated GPX4 constructs. d, Cell viability in shGPX4 HT-1080 cells that express the indicated GPX4 constructs treated with different doses of LFM or TF for 4 h. e, Cell survival fraction, lipid peroxidation and PTGS2 mRNA levels in shGPX4 HT-1080 cells that express the indicated GPX4 constructs upon treatment with BQR (500 μM). f, Western blotting showing GPX4 protein levels in shGPX4 cells that express the indicated GPX4 constructs in a variety of cell lines. g, Cell viability measurement in various shGPX4 cells that express the indicated GPX4 constructs treated with different doses of BQR for 4 h. Data are presented as mean ± s.d., n = 3 independent repeats (d, e, g); unpaired, two-tailed t-test. Western blots are representative of two biological replicates. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. ns, not significant.

Source data

Extended Data Fig. 6 Inactivation of DHODH and GPX4 induces mitochondrial lipid peroxidation.

a, Western blot showing GPX4 protein levels in cytosolic and mitochondrial fractions from NCI-H226 cells that express the indicated GPX4 constructs. b, Cell proliferation of NCI-H226 cells that express the indicated GPX4 constructs. c, Cell viability in NCI-H226 cells that express the indicated GPX4 constructs treated with different doses of BQR, LFM or TF for 4 h. d, Cell survival fraction, lipid peroxidation and PTGS2 mRNA levels in NCI-H226 cells that express the indicated GPX4 constructs upon treatment with BQR (500 μM). e, Cell viability in Cas9 control and DHODH KO HT-1080 cells treated with different doses of ML162 for 4 h, following pretreatment with vehicle, TEMPO (10 μM), MitoTEMPO (10 μM), or Lip-1 (10 μM) for 24 h. f, Cas9 control and DHODH KO HT-1080 cells were treated with RSL3 (1 μM) for 2 h, then stained with mito-BODIPY. Oxidized mito-BODIPY (green) indicates mitochondrial lipid peroxidation (scale bar, 5 μM). g, Mitochondrial lipid peroxidation in Cas9 control and DHODH KO HT-1080 cells upon treatment with RSL3 (1 μM) for 2 h. h, Mitochondrial lipid peroxidation in shControl and shGPX4 HT-1080 cells upon treatment with BQR (500 μM) for 2 h. i, Mitochondrial lipid peroxidation in HT-1080 cells upon treatment with RSL3 (1 μM) and/or BQR (500 μM), LFM (100 μM), or TF (500 μM) for 2 h, following pretreatment with vehicle or Lip-1 (10 μM) for 24 h. j, Mitochondrial lipid peroxidation in HT-1080 cells upon treatment with ML162 (1 μM) and/or BQR (500 μM), LFM (100 μM), or TF (500 μM) for 2 h, following pretreatment with vehicle or Lip-1 (10 μM) for 24 h. k, Mitochondrial lipid peroxidation in DHODH KO HT-1080 cells that express the indicated DHODH constructs upon treatment with RSL3 (1 μM) for 2 h. l, m, Mitochondrial lipid peroxidation in Cas9 control and DHODH KO HT-1080 cells with indicated genotypes upon treatment with RSL3 (1 μM) for 2 h, following pretreatment with vehicle or Lip-1 (10 μM) for 24 h. n, Mitochondrial lipid peroxidation in Cas9 control and FSP1 KO HT-1080 cells upon treatment with RSL3 (1 μM) and/or BQR (500 μM) for 2 h. o, Western blot analysis of DHODH and FSP1 protein levels in cytosolic and mitochondrial fractions of HT-1080 cells with indicated genotypes. p, Cell viability in Cas9 control and DHODH KO HT-1080 cells with indicated genotypes treated with different doses of RSL3 for 4 h, following pretreatment with vehicle or Lip-1 (10 μM) for 24 h. q, Mitochondrial lipid peroxidation in Cas9 control and DHODH KO HT-1080 cells with indicated genotypes upon treatment with RSL3 (1 μM) for 2 h, following pretreatment with vehicle or Lip-1 (10 μM) for 24 h. r, Mitochondrial lipid peroxidation in shGPX4 HT-1080 cells that express the indicated GPX4 constructs upon treatment with BQR (500 μM) for 2 h. s, Mitochondrial lipid peroxidation in NCI-H226 cells that express the indicated GPX4 constructs upon treatment with BQR (500 μM) for 2 h. Cells were grown in medium supplemented with uridine (50 μM) (eg, kq). Data are presented as mean ± s.d., n = 3 independent repeats (be, gn, ps); unpaired, two-tailed t-test. Western blots are representative of two biological replicates. Images are representative of at least n = 5 imaged cells (f). *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. ns. Not significant. Mito-C11, fluorescent mitochondria-targeted lipid peroxidation probe.

Source data

Extended Data Fig. 7 DHODH regulation of ferroptosis relates to its function to reduce CoQ to CoQH2 in mitochondria.

a, Cell viability in HT-1080 cells treated with different doses of FIN56 and co-treated with BQR (500 μM), LFM (100 μM) or TF (500 μM) for 4 h, following pretreatment with vehicle or Lip-1 (10 μM) for 24 h. b, Cell survival fraction, mitochondrial lipid peroxidation and PTGS2 mRNA levels in HT-1080 cells upon treatment with vehicle, FIN56 (50 μM) and/or BQR (500 μM), following pretreatment with vehicle or Lip-1 (10 μM) for 24 h. c, Western blot analysis of COQ2 and DHODH protein levels in HT-1080 cells with indicated genotypes. d, Total CoQ in Cas9 control and COQ2 KO HT-1080 cells. e, Total CoQ in HT-1080 cells that were treated with vehicle or 4-CBA (5 mM) for 24 h. f, Cell viability measurement in Cas9 control and DHODH KO HT-1080 cells with indicated genotypes treated with different doses of RSL3 for 4 h, following pretreatment with vehicle or Lip-1 (10 μM) for 24 h. g, Cell viability in Cas9 control and DHODH KO HT-1080 cells with indicated genotypes treated with different doses of ML162 for 4 h, following pretreatment with vehicle or Lip-1 (10 μM) for 24 h. h, Cell viability in Cas9 control and DHODH KO HT-1080 cells treated with different doses of RSL3 for 4 h, following pretreatment with vehicle, 4-CBA (5 mM), or 4-CBA (5 mM) + Lip-1 (10 μM) for 24 h. i, Cell viability in Cas9 control and DHODH KO HT-1080 cells treated with different doses of ML162 for 4 h, following pretreatment with vehicle, 4-CBA (5 mM) or Lip-1 (10 μM) for 24 h. j, Mitochondrial lipid peroxidation in Cas9 control and DHODH KO HT-1080 cells upon treatment with RSL3 (1 μM), following pretreatment with vehicle, 4-CBA (5 mM), or 4-CBA (5 mM) + Lip-1 (10 μM) for 24 h. k, Simplified schematic showing how DHODH couples the oxidation of DHO to OA to the reduction of CoQ to CoQH2 in the mitochondrial inner membrane. l, CoQ/CoQH2 ratio in NCI-H226 cells that were treated with BQR (1 mM) for 2 h. m, Cell viability in Cas9 control and DHODH KO HT-1080 cells treated with different doses of ML162 for 4 h, following pretreatment with vehicle, MitoQ (10 μM), MitoQH2 (10 μM), or Lip-1 (10 μM) for 24 h. n, Mitochondrial lipid peroxidation in Cas9 control and DHODH KO HT-1080 cells upon treatment with RSL3 (1 μM) for 2 h, following pretreatment with vehicle, MitoQ (10 μM), MitoQH2 (10 μM), or Lip-1 (10 μM) for 24 h. o, Lipid peroxidation in Cas9 control and DHODH KO HT-1080 cells upon treatment with RSL3 (1 μM) for 2 h, following pretreatment with vehicle, MitoQ (10 μM), MitoQH2 (10 μM), or Lip-1 (10 μM) for 24 h. Cells were grown in medium supplemented with uridine (50 μM) (c, d, fj, mo). Data are presented as mean ± s.d., n = 3 independent repeats (a, b, dj, lo); unpaired, two-tailed t-test. Western blot is representative of two biological replicates. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. ns, not significant. OCR, oxygen consumption rate; MitoQ, [10-(4,5-dimethoxy-2-methyl-3,6-dioxo-1,4-cyclohexadien-1-yl)decyl] triphenyl-phosphonium, monomethanesulfonate; MitoQH2, [10-(2,5-dihydroxy-3,4-dimethoxy-6-methylphenyl)decyl] triphenyl-phosphonium, monomethanesulfonate.

Source data

Extended Data Fig. 8 The effects of mitoQ and mitoQH2 on RSL3- and BQR-induced ferroptosis in a variety of cell lines.

a, GPX4, DHODH and FSP1 protein levels in indicated cell lines determined by western blotting. bj, Cell viability in 293T (b), Hela (c), Jurkat (d), SW620 (e), U-87 MG (f), A549 (g), NCI-H1437 (h), MDA-MB-436 (i), and MDA-MB-231 (j) cells treated with different doses of RSL3 with vehicle or BQR (500 μM) for 4 h, following pretreatment with vehicle, MitoQ (10 μM), MitoQH2 (10 μM), or Lip-1 (10 μM) for 24 h. k, CoQ/CoQH2 ratio in HT-1080 cells that were treated with myxothiazol (10 μM) for 2 h. l, Cell viability in Cas9 control and DHODH KO HT-1080 cells treated with different doses of RSL3 for 4 h, following pretreatment with vehicle or myxothiazol (1 μM) for 24 h. m, CoQ/CoQH2 ratio in A549 cells that were treated with myxothiazol (10 μM) for 2 h. n, Cell viability in A549 cells treated with different doses of RSL3 with or without BQR (500 μM) for 4 h, following pretreatment with vehicle or myxothiazol (1 μM) for 24 h. o, Western blot analysis of DHODH and CiAOX protein levels in HT-1080 cells with indicated genotypes. p, Mitochondrial lipid peroxidation in HT-1080 cells with indicated genotypes upon treatment with RSL3 (1 μM) for 2 h. Cells were grown in medium supplemented with uridine (50 μM) (l, o, p). Data are presented as mean ± s.d., n = 3 independent repeats (bn, p); unpaired, two-tailed t-test. Western blots are representative of two biological replicates. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. ns, not significant.

Source data

Extended Data Fig. 9 DHODH inhibitor selectively suppresses GPX4low tumour growth.

a, Weights of shControl and shGPX4 HT-1080 xenograft tumours with the indicated treatments. bd, Representative immunochemical images from shControl and shGPX4 HT-1080 xenograft tumours with the indicated treatments (b; scale bars, 20 μM), and staining scores of cleaved-caspase 3 (c) and ki67 (d). e, Weight measurements of NCI-H226 xenograft tumours with the indicated treatments. f, Weight measurements of TC632, TC629, or TC494 PDX tumours with the indicated treatments. g, Volumes of Cas9 control and DHODH KO NCI-H226 xenograft tumours with the indicated treatments at different time points (days). h, Weights of Cas9 control and DHODH KO NCI-H226 xenograft tumours with the indicated treatments. i, Weight measurements of HT-1080 xenograft tumours with the indicated treatments. jl, Representative immunochemistry images of HT-1080 xenograft tumours with the indicated treatments (j; scale bars, 20 μM) and staining scores of cleaved-caspase 3 (k) and ki67 (l). m, Volumes of TC629 PDX tumours with the indicated treatments at different time points (days). n, Weights of TC632 and TC629 PDX tumours with the indicated treatments. o, Weights of mice for all cell line xenografts or PDXs with different treatments at different time points (days). Box plots indicate median, minima and maxima of the distributions, and with whiskers from minimum to maximum. Data are presented as mean ± s.d., n = 8 (a, e, gi), n = 5 (c, d, k, l) or n = 6 independent tumours (f, m, n). n = 4 for nude mouse weights and n = 8 for NSG mouse weights (o). Unpaired, two-tailed t-test. Images are representative of n = 5 images. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. ns, not significant. 4-HNE, 4-hydroxynonenal.

Source data

Extended Data Fig. 10 Working model depicting how GPX4, FSP1, and DHODH suppress ferroptosis in different subcellular compartments.

See main text for a detailed description. PLOOH, phospholipid hydroperoxide; PLOO·, phospholipid hydroperoxyl radical; GSSH, oxidized glutathione; NAD(P)H, reduced nicotinamide adenine dinucleotide (phosphate); NAD(P)+, oxidized nicotinamide adenine dinucleotide (phosphate).

Supplementary information

Supplementary Figure

This file contains a figure exemplifying the gating strategy.

Reporting Summary

Supplementary Table 1

A list of sequences and primers used in the study.

Peer Review File

Source data

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mao, C., Liu, X., Zhang, Y. et al. DHODH-mediated ferroptosis defence is a targetable vulnerability in cancer. Nature 593, 586–590 (2021). https://doi.org/10.1038/s41586-021-03539-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-021-03539-7

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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