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:

Ferroptosis as a mechanism to mediate p53 function in tumor radiosensitivity

Abstract

Ferroptosis, a form of regulated cell death triggered by lipid peroxidation, was recently identified as an important mechanism in radiotherapy (RT)-mediated tumor suppression and radioresistance, although the exact genetic contexts in which to target ferroptosis in RT remains to be defined. p53 is the most commonly mutated gene in human cancers and a major effector to RT. Here, we identify ferroptosis as a critical mechanism to mediate p53 function in tumor radiosensitivity. Mechanistically, RT-mediated p53 activation antagonizes RT-induced SLC7A11 expression and represses glutathione synthesis, thereby promoting RT-induced lipid peroxidation and ferroptosis. p53 deficiency promotes radioresistance in cancer cells or tumors at least partly through SLC7A11-mediated ferroptosis inhibition. Ferroptosis inducers (FINs) that inhibit SLC7A11 exert significant radiosensitizing effects in tumor organoids and patient-derived xenografts with p53 mutation or deficiency. Finally, we show that RT-induced ferroptosis correlates with p53 activation and better clinical outcomes to RT in cancer patients. Together, our study uncovers a previously unappreciated role of ferroptosis in p53-mediated radiosensitization and suggest using FINs in combination with RT to treat p53-mutant cancers.

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: Ferroptosis is involved in p53-mediated radiosensitization.
Fig. 2: p53 promotes RT-induced ferroptosis partly via antagonizing SLC7A11 induction.
Fig. 3: p53 deficiency contributes to radioresistance partly via SLC7A11-mediated GSH synthesis and ferroptosis inhibition.
Fig. 4: FINs radiosensitize p53-deficient/-mutant cancer cells and tumor organoids.
Fig. 5: Ferroptosis is involved in p53 deficiency-mediated radioresistance and FINs radiosensitize p53-mutant tumors in vivo.
Fig. 6: Ferroptosis induction correlates with p53 activation and better clinical responses to RT in cancer patients.

Similar content being viewed by others

References

  1. Delaney G, Jacob S, Featherstone C, Barton M. The role of radiotherapy in cancer treatment: estimating optimal utilization from a review of evidence-based clinical guidelines. Cancer. 2005;104:1129–37.

    Article  PubMed  Google Scholar 

  2. Jaffray DA. Image-guided radiotherapy: from current concept to future perspectives. Nat Rev Clin Oncol. 2012;9:688–99.

    Article  CAS  PubMed  Google Scholar 

  3. Baidoo KE, Yong K, Brechbiel MW. Molecular pathways: targeted alpha-particle radiation therapy. Clin Cancer Res. 2013;19:530–7.

    Article  CAS  PubMed  Google Scholar 

  4. Azzam EI, Jay-Gerin JP, Pain D. Ionizing radiation-induced metabolic oxidative stress and prolonged cell injury. Cancer Lett. 2012;327:48–60.

    Article  CAS  PubMed  Google Scholar 

  5. Fei P, El-Deiry WS. P53 and radiation responses. Oncogene. 2003;22:5774–83.

    Article  CAS  PubMed  Google Scholar 

  6. Gudkov AV, Komarova EA. The role of p53 in determining sensitivity to radiotherapy. Nat Rev Cancer. 2003;3:117–29.

    Article  CAS  PubMed  Google Scholar 

  7. Vousden KH, Prives C. Blinded by the light: the growing complexity of p53. Cell. 2009;137:413–31.

    Article  CAS  PubMed  Google Scholar 

  8. Muller PA, Vousden KH. p53 mutations in cancer. Nat Cell Biol. 2013;15:2–8.

    Article  CAS  PubMed  Google Scholar 

  9. Bykov VJN, Eriksson SE, Bianchi J, Wiman KG. Targeting mutant p53 for efficient cancer therapy. Nat Rev Cancer. 2018;18:89–102.

    Article  CAS  PubMed  Google Scholar 

  10. Lee JM, Bernstein A. p53 mutations increase resistance to ionizing radiation. Proc Natl Acad Sci USA. 1993;90:5742–6.

    Article  CAS  PubMed  Google Scholar 

  11. Lowe SW, Bodis S, McClatchey A, Remington L, Ruley HE, Fisher DE, et al. p53 status and the efficacy of cancer therapy in vivo. Science. 1994;266:807–10.

    Article  CAS  PubMed  Google Scholar 

  12. Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell. 2012;149:1060–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Stockwell BR, Friedmann Angeli JP, Bayir H, Bush AI, Conrad M, Dixon SJ, et al. Ferroptosis: a regulated cell death nexus linking metabolism, redox biology, and disease. Cell. 2017;171:273–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Seibt TM, Proneth B, Conrad M. Role of GPX4 in ferroptosis and its pharmacological implication. Free Radic Biol Med. 2019;133:144–52.

    Article  CAS  PubMed  Google Scholar 

  15. Friedmann Angeli JP, Schneider M, Proneth B, Tyurina YY, Tyurin VA, Hammond VJ, et al. Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nat Cell Biol. 2014;16:1180–91.

    Article  CAS  PubMed  Google Scholar 

  16. Yang WS, SriRamaratnam R, Welsch ME, Shimada K, Skouta R, Viswanathan VS, et al. Regulation of ferroptotic cancer cell death by GPX4. Cell. 2014;156:317–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Koppula P, Zhang Y, Zhuang L, Gan B. Amino acid transporter SLC7A11/xCT at the crossroads of regulating redox homeostasis and nutrient dependency of cancer. Cancer Commun. 2018;38:12.

    Article  Google Scholar 

  18. Conrad M, Sato H. The oxidative stress-inducible cystine/glutamate antiporter, system x (c) (−): cystine supplier and beyond. Amino Acids. 2012;42:231–46.

    Article  CAS  PubMed  Google Scholar 

  19. Koppula P, Zhuang L, Gan B. Cystine transporter SLC7A11/xCT in cancer: ferroptosis, nutrient dependency, and cancer therapy. Protein Cell 2020: 1–22. https://doi.org/10.1007/s13238-020-00789-5.

  20. Liu X, Zhang Y, Zhuang L, Olszewski K, Gan B. NADPH debt drives redox bankruptcy: SLC7A11/xCT-mediated cystine uptake as a double-edge sword in cellular redox regulation. Genes Dis. 2020. https://doi.org/10.1016/j.gendis.2020.11.010.

  21. Jiang L, Kon N, Li T, Wang SJ, Su T, Hibshoosh H, et al. Ferroptosis as a p53-mediated activity during tumour suppression. Nature. 2015;520:57–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Chu B, Kon N, Chen D, Li T, Liu T, Jiang L, et al. ALOX12 is required for p53-mediated tumour suppression through a distinct ferroptosis pathway. Nat Cell Biol. 2019;21:579–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Gan B. DUBbing ferroptosis in cancer cells. Cancer Res. 2019;79:1749–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Liu T, Jiang L, Tavana O, Gu W. The deubiquitylase OTUB1 mediates ferroptosis via stabilization of SLC7A11. Cancer Res. 2019;79:1913–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Jennis M, Kung CP, Basu S, Budina-Kolomets A, Leu JI, Khaku S, et al. An African-specific polymorphism in the TP53 gene impairs p53 tumor suppressor function in a mouse model. Genes Dev. 2016;30:918–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Zhang Y, Shi J, Liu X, Feng L, Gong Z, Koppula P, et al. BAP1 links metabolic regulation of ferroptosis to tumour suppression. Nat Cell Biol. 2018;20:1181–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Gao M, Yi J, Zhu J, Minikes AM, Monian P, Thompson CB, et al. Role of mitochondria in ferroptosis. Mol Cell. 2019;73:354–63 e353.

    Article  CAS  PubMed  Google Scholar 

  28. Zhang Y, Zhuang L, Gan B. BAP1 suppresses tumor development by inducing ferroptosis upon SLC7A11 repression. Mol Cell Oncol. 2019;6:1536845.

    Article  PubMed  Google Scholar 

  29. Lei G, Zhang Y, Koppula P, Liu X, Zhang J, Lin SH, et al. The role of ferroptosis in ionizing radiation-induced cell death and tumor suppression. Cell Res. 2020;30:146–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Lang X, Green MD, Wang W, Yu J, Choi JE, Jiang L, et al. Radiotherapy and immunotherapy promote tumoral lipid oxidation and ferroptosis via synergistic repression of SLC7A11. Cancer Discov. 2019;9:1673–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Ye LF, Chaudhary KR, Zandkarimi F, Harken AD, Kinslow CJ, Upadhyayula PS, et al. Radiation-induced lipid peroxidation triggers ferroptosis and synergizes with ferroptosis inducers. ACS Chem Biol. 2020;15:469–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Tarangelo A, Magtanong L, Bieging-Rolett KT, Li Y, Ye J, Attardi LD, et al. p53 Suppresses metabolic stress-induced ferroptosis in cancer cells. Cell Rep. 2018;22:569–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Xie Y, Zhu S, Song X, Sun X, Fan Y, Liu J, et al. The tumor suppressor p53 limits ferroptosis by blocking DPP4 activity. Cell Rep. 2017;20:1692–704.

    Article  CAS  PubMed  Google Scholar 

  34. Ou Y, Wang SJ, Li D, Chu B, Gu W. Activation of SAT1 engages polyamine metabolism with p53-mediated ferroptotic responses. Proc Natl Acad Sci USA. 2016;113:E6806–E6812.

    Article  CAS  PubMed  Google Scholar 

  35. Leu JI, Murphy ME, George DL. Mechanistic basis for impaired ferroptosis in cells expressing the African-centric S47 variant of p53. Proc Natl Acad Sci USA. 2019;116:8390–6.

    Article  CAS  PubMed  Google Scholar 

  36. Feng H, Schorpp K, Jin J, Yozwiak CE, Hoffstrom BG, Decker AM, et al. Transferrin receptor is a specific ferroptosis marker. Cell Rep. 2020;30:3411–23. e3417.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Zheng H, Chen L, Pledger WJ, Fang J, Chen JJO. p53 promotes repair of heterochromatin DNA by regulating JMJD2b and SUV39H1 expression. Oncogene. 2014;33:734–44.

    Article  CAS  PubMed  Google Scholar 

  38. Wang L, Liu Y, Du T, Yang H, Lei L, Guo M, et al. ATF3 promotes erastin-induced ferroptosis by suppressing system Xc–. Cell Death Differ. 2020;27:662–75.

    Article  CAS  PubMed  Google Scholar 

  39. 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. 1999;274:11455–8.

    Article  CAS  PubMed  Google Scholar 

  40. Drost J, Clevers H. Organoids in cancer research. Nat Rev Cancer. 2018;18:407–18.

    Article  CAS  PubMed  Google Scholar 

  41. Li M, Izpisua Belmonte JC. Organoids—preclinical models of human disease. N Engl J Med. 2019;380:569–79.

    Article  PubMed  Google Scholar 

  42. Cancer Genome Atlas Research Network. Integrated genomic analyses of ovarian carcinoma. Nature. 2011;474:609–15.

    Article  Google Scholar 

  43. Zhang Y, Tan H, Daniels JD, Zandkarimi F, Liu H, Brown LM, et al. Imidazole ketone erastin induces ferroptosis and slows tumor growth in a mouse lymphoma model. Cell Chem Biol. 2019;26:623–.e9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Gout PW, Buckley AR, Simms CR, Bruchovsky N. Sulfasalazine, a potent suppressor of lymphoma growth by inhibition of the x(c)- cystine transporter: a new action for an old drug. Leukemia. 2001;15:1633–40.

    Article  CAS  PubMed  Google Scholar 

  45. Xie L, Song X, Yu J, Guo W, Wei L, Liu Y, et al. Solute carrier protein family may involve in radiation-induced radioresistance of non-small cell lung cancer. J Cancer Res Clin Oncol. 2011;137:1739–47.

    Article  CAS  PubMed  Google Scholar 

  46. Lin A, Piao HL, Zhuang L, Sarbassov DD, Ma L, Gan B. FoxO transcription factors promote AKT Ser473 phosphorylation and renal tumor growth in response to pharmacological inhibition of the PI3K-AKT pathway. Cancer Res. 2014;74:1682–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Xiao ZD, Han L, Lee H, Zhuang L, Zhang Y, Baddour J, et al. Energy stress-induced lncRNA FILNC1 represses c-Myc-mediated energy metabolism and inhibits renal tumor development. Nat Commun. 2017;8:783.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Liu X, Olszewski K, Zhang Y, Lim EW, Shi J, Zhang X, et al. Cystine transporter regulation of pentose phosphate pathway dependency and disulfide stress exposes a targetable metabolic vulnerability in cancer. Nat Cell Biol. 2020;22:476–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Kopper O, de Witte CJ, Lohmussaar K, Valle-Inclan JE, Hami N, Kester L, et al. An organoid platform for ovarian cancer captures intra- and interpatient heterogeneity. Nat Med. 2019;25:838–49.

    Article  CAS  PubMed  Google Scholar 

  50. Zhang Y, Shi J, Liu X, Xiao Z, Lei G, Lee H, et al. H2A monoubiquitination links glucose availability to epigenetic regulation of the endoplasmic reticulum stress response and cancer cell death. Cancer Res. 2020;80:2243–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This research was supported by Institutional Research Fund and RO Strategic Initiatives (ROSI) Boot Walk Seed Award from The University of Texas MD Anderson Cancer Center, KC180131 from Department of Defense Kidney Cancer Research Program, and R01CA181196, R01CA244144, and R01CA247992 from the National Institutes of Health (to BG); P50 CA217685, American Cancer Society, and MD Anderson Moon Shot Program in Ovarian Cancer (to AKS). BG was an Andrew Sabin Family Fellow. PK is supported by CPRIT Research Training Grant (RP170067) and Dr. John J. Kopchick Research Award from The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences. This research has also been 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

GL performed most of the experiments with assistance from YZ, TH, XL, CM, WC, PK, and YY; JL, AKS, TH, and XD provided ovarian cancer organoids; BG designed and supervised the experiments; BG and GL wrote most of the manuscript with assistance from other co-authors; all authors commented on the manuscript.

Corresponding author

Correspondence to Boyi Gan.

Ethics declarations

Conflict of interest

AKS declares the following competing of interests: consulting (Merck, Kiyatec); shareholder (BioPath); and research support (M-Trap). 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

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lei, G., Zhang, Y., Hong, T. et al. Ferroptosis as a mechanism to mediate p53 function in tumor radiosensitivity. Oncogene 40, 3533–3547 (2021). https://doi.org/10.1038/s41388-021-01790-w

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41388-021-01790-w

This article is cited by

Search

Quick links