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:

ALOX12 is required for p53-mediated tumour suppression through a distinct ferroptosis pathway

Abstract

It is well established that ferroptosis is primarily controlled by glutathione peroxidase 4 (GPX4). Surprisingly, we observed that p53 activation modulates ferroptotic responses without apparent effects on GPX4 function. Instead, ALOX12 inactivation diminishes p53-mediated ferroptosis induced by reactive oxygen species stress and abrogates p53-dependent inhibition of tumour growth in xenograft models, suggesting that ALOX12 is critical for p53-mediated ferroptosis. The ALOX12 gene resides on human chromosome 17p13.1, a hotspot of monoallelic deletion in human cancers. Loss of one Alox12 allele is sufficient to accelerate tumorigenesis in Eμ-Myc lymphoma models. Moreover, ALOX12 missense mutations from human cancers abrogate its ability to oxygenate polyunsaturated fatty acids and to induce p53-mediated ferroptosis. Notably, ALOX12 is dispensable for ferroptosis induced by erastin or GPX4 inhibitors; conversely, ACSL4 is required for ferroptosis upon GPX4 inhibition but dispensable for p53-mediated ferroptosis. Thus, our study identifies an ALOX12-mediated, ACSL4-independent ferroptosis pathway that is critical for p53-dependent tumour suppression.

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: ALOX12 is essential for p53-mediated ferroptosis upon ROS stress.
Fig. 2: ALOX12 is critical for p53-mediated tumour growth suppression.
Fig. 3: Loss of one Alox12 allele is sufficient to accelerate tumorigenesis in Eμ-Myc lymphoma models.
Fig. 4: ALOX12 missense mutations from human cancers abrogate ALOX12 enzymatic activity and p53-mediated ferroptosis.
Fig. 5: Mechanistic insight into p53-mediated activation of ALOX12.
Fig. 6: p53-mediated ferroptosis is ALOX12 dependent but ACSL4 independent.
Fig. 7: Mechanistic insight into p53-mediated ferroptosis.
Fig. 8: ALOX12 in regulating p53-mediated ferroptosis in human cancer lines.

Similar content being viewed by others

Data availability

The ALOX12 gene expression data for normal versus cancer human tissues were derived from the Oncomine database (https://www.oncomine.org/resource/). The association of ALOX12 with overall survival in patients with pancreatic adenocarcinoma were derived from publicly available clinical information provided by cBioportal for Cancer Genomics databases (http://www.cbioportal.org/). Source data for Figs. 1a,c,f, 2a,b,d,e,g, 3c,d,h, 4b,c,f, 5d–f,h,i, 6b–d,f–h, 7c–e and 8a–g and Supplementary Figs. 1b,d–e, 2b, 3b–d, 5c,d, 6j, 7a–c and 8d have been provided as Supplementary Table 1. All other data supporting the findings of this study are available from the corresponding author on reasonable request.

Code availability

No computational code was used in this study.

References

  1. Kastenhuber, E. R. & Lowe, S. W. Putting p53 in context. Cell 170, 1062–1078 (2017).

    Article  CAS  Google Scholar 

  2. Jennis, M. et al. An African-specific polymorphism in the TP53 gene impairs p53 tumor suppressor function in a mouse model. Genes Dev. 30, 918–930 (2016).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  4. Gao, M., Monian, P., Quadri, N., Ramasamy, R. & Jiang, X. Glutaminolysis and transferrin regulate ferroptosis. Mol. Cell 59, 298–308 (2015).

    Article  CAS  Google Scholar 

  5. Wang, S. J. et al. Acetylation is crucial for p53-mediated ferroptosis and tumor suppression. Cell Rep. 17, 366–373 (2016).

    Article  CAS  Google Scholar 

  6. Saint-Germain, E. et al. SOCS1 regulates senescence and ferroptosis by modulating the expression of p53 target genes. Aging 9, 2137–2162 (2017).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  10. Angeli, J. P. F., Shah, R., Pratt, D. A. & Conrad, M. Ferroptosis inhibition: mechanisms and opportunities. Trends Pharmacol. Sci. 38, 489–498 (2017).

    Article  CAS  Google Scholar 

  11. Viswanathan, V. S. et al. Dependency of a therapy-resistant state of cancer cells on a lipid peroxidase pathway. Nature 547, 453–457 (2017).

    Article  CAS  Google Scholar 

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

    Article  Google Scholar 

  13. Matsushita, M. et al. T cell lipid peroxidation induces ferroptosis and prevents immunity to infection. J. Exp. Med. 212, 555–568 (2015).

    Article  CAS  Google Scholar 

  14. Imai, H. et al. Early embryonic lethality caused by targeted disruption of the mouse PHGPx gene. Biochem. Biophys. Res. Commun. 305, 278–286 (2003).

    Article  CAS  Google Scholar 

  15. Ingold, I. et al. Expression of a catalytically inactive mutant form of glutathione peroxidase 4 (Gpx4) confers a dominant-negative effect in male fertility. J. Biol. Chem. 290, 14668–14678 (2015).

    Article  CAS  Google Scholar 

  16. Wortmann, M. et al. Combined deficiency in glutathione peroxidase 4 and vitamin E causes multiorgan thrombus formation and early death in mice. Circ. Res. 113, 408–417 (2013).

    Article  CAS  Google Scholar 

  17. Schriever, S. C. et al. Alterations in neuronal control of body weight and anxiety behavior by glutathione peroxidase 4 deficiency. Neuroscience 357, 241–254 (2017).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  19. Imai, H., Matsuoka, M., Kumagai, T., Sakamoto, T. & Koumura, T. Lipid peroxidation-dependent cell death regulated by GPx4 and ferroptosis. Curr. Top. Microbiol. Immunol. 403, 143–170 (2017).

    CAS  PubMed  Google Scholar 

  20. 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 

  21. Eischen, C. M., Weber, J. D., Roussel, M. F., Sherr, C. J. & Cleveland, J. L. Disruption of the ARF–Mdm2–p53 tumor suppressor pathway in Myc-induced lymphomagenesis. Genes Dev. 13, 2658–2669 (1999).

    Article  CAS  Google Scholar 

  22. Post, S. M. et al. p53-dependent senescence delays Eµ-myc-induced B-cell lymphomagenesis. Oncogene 29, 1260–1269 (2010).

    Article  CAS  Google Scholar 

  23. Valente, L. J., Grabow, S., Vandenberg, C. J., Strasser, A. & Janic, A. Combined loss of PUMA and p21 accelerates c-MYC-driven lymphoma development considerably less than loss of one allele of p53. Oncogene 35, 3866–3871 (2016).

    Article  CAS  Google Scholar 

  24. Liu, Y. et al. Deletions linked to TP53 loss drive cancer through p53-independent mechanisms. Nature 531, 471–475 (2016).

    Article  CAS  Google Scholar 

  25. Luci, D. et al. in Probe Reports from the NIH Molecular Libraries Program (National Center for Biotechnology Information, 2013).

  26. Gasol, E., Jimenez-Vidal, M., Chillaron, J., Zorzano, A. & Palacin, M. Membrane topology of system xc light subunit reveals a re-entrant loop with substrate-restricted accessibility. J. Biol. Chem. 279, 31228–31236 (2004).

    Article  CAS  Google Scholar 

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

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

  29. Kagan, V. E. et al. Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis. Nat. Chem. Biol. 13, 81–90 (2017).

    Article  CAS  Google Scholar 

  30. Tarangelo, A. et al. p53 suppresses metabolic stress-induced ferroptosis in cancer cells. Cell Rep. 22, 569–575 (2018).

    Article  CAS  Google Scholar 

  31. Xie, Y. C. et al. The tumor suppressor p53 limits ferroptosis by blocking DPP4 activity. Cell Rep. 20, 1692–1704 (2017).

    Article  CAS  Google Scholar 

  32. Ferretti, E., De Smaele, E., Di Marcotullio, L., Screpanti, I. & Gulino, A. Hedgehog checkpoints in medulloblastoma: the chromosome 17p deletion paradigm. Trends Mol. Med. 11, 537–545 (2005).

    Article  CAS  Google Scholar 

  33. Schnaiter, A. & Stilgenbauer, S. 17p deletion in chronic lymphocytic leukemia risk stratification and therapeutic approach. Hematol. Oncol. Clin. North Am. 27, 289–301 (2013).

    Article  Google Scholar 

  34. Tam, C. S. & Stilgenbauer, S. How best to manage patients with chronic lymphocytic leuekmia with 17p deletion and/or TP53 mutation? Leuk. Lymphoma 56, 587–593 (2015).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  36. Johnson, E. N., Brass, L. F. & Funk, C. D. Increased platelet sensitivity to ADP in mice lacking platelet-type 12-lipoxygenase. Proc. Natl Acad. Sci. USA 95, 3100–3105 (1998).

    Article  CAS  Google Scholar 

  37. Bensaad, K. et al. TIGAR, a p53-inducible regulator of glycolysis and apoptosis. Cell 126, 107–120 (2006).

    Article  CAS  Google Scholar 

  38. Kruiswijk, F., Labuschagne, C. F. & Vousden, K. H. p53 in survival, death and metabolic health: a lifeguard with a licence to kill. Nat. Rev. Mol. Cell Biol. 7, 393–405 (2015).

    Article  Google Scholar 

  39. Davies, M. J. Detection of peroxyl and alkoxyl radicals produced by reaction of hydroperoxides with rat liver microsomal fractions. Biochem. J. 257, 603–606 (1989).

    Article  CAS  Google Scholar 

  40. Hughes, R. H., Silva, V. A., Ahmed, I., Shreiber, D. I. & Morrison, B.3rd. Neuroprotection by genipin against reactive oxygen and reactive nitrogen species-mediated injury in organotypic hippocampal slice cultures. Brain Res. 1543, 308–314 (2014).

    Article  CAS  Google Scholar 

  41. Wang, Z., Jiang, H., Chen, S., Du, F. & Wang, X. The mitochondrial phosphatase PGAM5 functions at the convergence point of multiple necrotic death pathways. Cell 148, 228–243 (2012).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank B. Stockwell, R. Baer, X. Jiang, M. Bordyuh and R. Rabadan for critical suggestions in this study. We also appreciate C. D. Funk for critical reagents. This work was supported by the National Cancer Institute of the US National Institutes of Health under Awards 5R01CA216884, 5R01CA190477, 5R01CA085533 and 5RO1CA224272 to W.G. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

Author information

Authors and Affiliations

Authors

Contributions

B.C. and W.G. conceived the study and experimental design. B.C., N.K., D.C., T. Liu, S.S., T. Li and L.J. conducted the experiments and acquired the data. B.C., O.T. and W.G. analysed and interpreted the data. B.C. and W.G. wrote the manuscript.

Corresponding author

Correspondence to Wei Gu.

Ethics declarations

Competing interests

The 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.

Integrated supplementary information

Supplementary Figure 1 ALOX12 is critical for p53-mediated ferroptosis.

(a) Schematic diagram of human ALOX family proteins. (b) Q-PCR of ALOX family members from H1299 Tet-on p533KR cells transfected with control siRNA (ctrl) or a pool of ALOX family specific siRNAs as indicated corresponding to Fig. 1a; Error bars are mean ± s.d., n = 3 independent experiments. (c) Western blot analysis of U2OS control or ALOX12 crispr clones #4 and #5. The experiments were repeated twice, independently, with similar results. (d) U2OS control or ALOX12 crispr clone #5, from c, were pre-incubated with Nutlin (10uM) for 12h, then treated with Nutlin (10 uM) and TBH (300 uM) as indicated for 8h. Error bars are mean ± s.d., n = 3 independent experiments. (e) U2OS control or ALOX12 crispr clone #5, from c, were pre-incubated with Nutlin (10 uM) for 12h, then treated with Nutlin (10uM) and TBH (300uM) for indicated times. Error bars are mean ± s.d., n = 3 independent experiments. All P values (b,d,e) were calculated using two-tailed unpaired Student’s t-test. Detailed statistical tests are described in the Methods. Scanned images of unprocessed blots are shown in Supplementary Fig. 9. Raw data are provided in Supplementary Table 1.

Supplementary Figure 2 Regulation of lipid peroxidation levels by p53.

(a) GPX4 activity is measured by glutathione peroxidase assay kit in U2OS cells with or without Nutlin (10uM) treatment. Error bars are mean ± s.d., n = 3 independent experiments. (b) Lipid peroxidation levels in U2OS cells treated with Nutlin (10uM) and TBH (300uM) were assessed by flow cytometry using C11-BODIPY dye (c) Quantification of lipid peroxidation levels (b) is shown. Error bars are mean ± s.d., n = 3 independent experiments. (d) Representative phase-contrast images of H1299 Tet-on p533KR GPX4 crispr cells transfected with GPX4 vectors. Ferr-1 (1uM) was then withdrew from the cells (Scale bars, 100µm). The experiments were repeated three times, independently, with similar results. (e) Lipid peroxidation levels in H1299 Tet-on p533KR GPX4 crispr cells transfected with GPX4 were assessed by flow cytometry using C11-BODIPY dye. The experiments were repeated three times, independently, with similar results. All P values (a,c) were calculated using two-tailed unpaired Student’s t-test. Raw data are provided in Supplementary Table 1.

Supplementary Figure 3 ALOX12 is critical for p53-mediated ferroptosis in H1299 Tet-on p533KR cells.

(a) Western blot analysis of H1299 Tet-on p533KR control or ALOX12 crispr clones #3 and #4. The experiments were repeated twice, independently, with similar results. (b) H1299 Tet-on p533KR control or ALOX12 crispr clones #4, from a, were pre-incubated with doxycycline (0.5ug/ml) for 12h, then treated with doxycycline (0.5 ug/ml), and TBH (60uM) as indicated for 8h. Quantification of cell death is shown. Error bars are mean ± s.d., n = 3 independent experiments. (c) H1299 Tet-on p533KR control or ALOX12 crispr clones #4, from a, were pre-incubated with doxycycline (0.5ug/ml) for 12h, then treated with doxycycline (0.5 ug/ml), and TBH (60uM) for indicated times. Quantification of cell death is shown. Error bars are mean ± s.d., n = 3 independent experiments. (d)Tumor weight (mg) (each group has n = 3 independent samples) was determined shown in Fig. 2f. Error bars are mean ± s.d. (e) Western blot analysis of xenograft tumors from H1299 Tet-on p533KR control and ALOX12 crispr shown in Fig. 2f. The experiments were repeated twice, independently, with similar results. All P values were calculated using two-tailed unpaired Student’s t-test. Scanned images of unprocessed blots are shown in Supplementary Fig. 9. Raw data are provided in Supplementary Table 1.

Supplementary Figure 4 Characterization of Eµ-myc; ALOX12+/- tumors.

(a) Semi-quantitative PCR of full-length p53 of tumor samples from Eµ-myc; ALOX12+/- and Eµ-myc; p53+/- mice, corresponding to Fig. 3d–g; WT, wild-type; LOH, loss of heterozygosity. The experiments were repeated twice, independently, with similar results. (b) PCR from genomic DNA to genotype ALOX12 of tumor samples from Eµ-myc; ALOX12+/− and control samples, corresponding to Fig. 3d–g; WT, wild-type allele; NULL, null allele. The experiments were repeated twice, independently, with similar results. (c) Western blot analysis of the spleen tissues from WT and ALOX12-/- mice with or without irradiation treatment. The experiments were repeated twice, independently, with similar results. (d) Representative immunohistochemistry staining of caspase-3 on spleen from WT and ALOX12-/- mice. Scale bars, 50 µm. The experiments were repeated three times, independently, with similar results. (e) The effects on apoptosis by loss of ALOX12. Representative immunohistochemistry staining of TUNEL on tumors from Eµ-Myc mice vs. Eµ-Myc/ALOX12+/− mice. Scale bars, 20 µm. The experiments were repeated three times, independently, with similar results. Scanned images of unprocessed blots and gels are shown in Supplementary Fig. 9.

Supplementary Figure 5 ALOX12 is downregulated in human cancers.

(a) Analysis of ALOX12 mRNA underexpression (blue) in different cancer types derived from Oncomine. (b) Box plots derived from gene expression data in Oncoming comparing the expression of ALOX12 mRNA in different cancer types. For Cervical normal tissues n = 10 independent samples, tumor tissues n = 21 independent samples; Head and Neck normal tissues n = 13 independent samples, tumor tissues n = 41 independent samples; Esophageal normal tissues n = 17 independent samples, tumor tissues n = 17 independent samples; Acute Myeloid Leukemia, normal tissues n = 8 independent samples, tumor tissues n = 285 independent samples. Box plots with centre line at median, box limits at 25th/75th centiles. (c) Kaplan-Meier survival curves were generated for overall survival (months) by stratifying patient samples with Pancreatic Adenocarcinoma (TCGA, Provisional) from cBioPortal (n = 186) based on ALOX12 expression levels. The patients were quartiled for ALOX12 expression [ALOX12 low (n = 45; black) and ALOX12 high (n = 45; red)], Log-rank Mantel-Cox test was used (p = 0.016). (d) Sequence alignment of Alox12 proteins from different species downloaded from HomoloGene and aligned using Clustal Omega Multiple Sequence Alignment. Cancer-associated residues (aa 372, 381, and 562) bolded and shown in red. (e) Representative phase-contrast images of H1299 Tet-on p533KR cells treated with doxycycline (0.5 ug/ml), TBH (60 uM), Ferr-1 (2 uM), and ML-355 (4 uM) as indicated for 8h (Scale bars, 100 µm). The experiments were repeated three times, independently, with similar results. (f) H1299 Tet-on p533KR cells were pre-incubated with doxycycline (0.5 ug/ml) for 12h, then treated with doxycycline (0.5 ug/ml), TBH (60 uM), Ferr-1 (2 uM), and ML-355 (4 uM) as indicated for 8 h. Quantification of cell death is shown. Error bars are mean ± s.d., n = 3 independent experiments. (g) U2OS ALOX12 crispr cells transfected with control, ALOX12, or mutant ALOX12 G381R vectors were pre-incubated with Nutlin (10 uM) for 12 h, then treated with TBH (300uM) and Nutlin (10uM) as indicated for 8h. Quantification of cell death is shown. Error bars are mean ± s.d., n = 3 independent experiments. All P values were calculated using two-tailed unpaired Student’s t-test. Raw data are provided in Supplementary Table 1.

Supplementary Figure 6 Interaction of SLC7A11 with ALOX family proteins.

(a, b, c, d) Western blot analysis of the interaction between SLC7A11 and ALOX family proteins (a, b, c, d). 293T cells were co-transfected with indicated constructs, and the extract was analysed by Co-IP assays. The western blot experiments were repeated twice, independently, with similar results. (e, f) Western blot analysis of the protein levels of ALOX12, ALOX15 and SLC7A11 (e, f). H1299 cells were transfected with indicated constructs. The western blot experiments were repeated twice, independently, with similar results. (g) Schematic diagram of WT and mutant SLC7A11. (h) Western blot analysis for the interactions of ALOX12 with WT, SLC7A11∆N, SLC7A11∆C. The western blot experiments were repeated twice, independently, with similar results. (i) Western blot analysis of the interaction of ALOX12 with SLC7A11 ∆91-140. The western blot experiments were repeated twice, independently, with similar results. (j) The effects of ALOX12 activity by SLC7A11-wt and SLC7A11∆91-140. Quantification of cell death is shown. Error bars are mean ± s.d., n = 3 independent experiments. P values were calculated using two-tailed unpaired Student’s t-test. Scanned images of unprocessed blots are shown in Supplementary Fig. 9. Raw data are provided in Supplementary Table 1.

Supplementary Figure 7 Loss of ALOX12 has no effect on Erastin- or RSL-3 -induced ferroptosis.

(a) WT, ALOX12+/−, ALOX12−/− MEFs were treated with different concentrations of Erastin (1, 2.5, 5 and 10 uM) for 12 h. Quantification of cell death is shown. Error bars are mean ± s.d., n = 3 independent experiments. (b) U2OS control or ALOX12 crispr cells, were treated with Erastin (20 uM) and Ferr-1 (2 uM) as indicated for 12 h. Quantification of cell death is shown. Error bars are mean ± s.d., n = 3 independent experiments. (c) Representative phase-contrast images of U2OS control or ALOX12 crispr cells treated with RSL-3 (4 uM) and Ferr-1 (2 uM) as indicated for 12h (Scale bars, 100 µm). The experiments were repeated three times, independently, with similar results. (d) U2OS control or ALOX12 crispr cells treated with RSL-3 (4 uM) and Ferr-1 (2 uM as indicated for 12 h. Quantification of cell death is shown. Error bars are mean ± s.d., n = 3 independent experiments. All P values (a,b,d) were calculated using two-tailed unpaired Student’s t-test. Raw data are provided in Supplementary Table 1

Supplementary Figure 8 Further characterization of ALOX12-mediated ferroptosis.

(a) Schematic diagram of the strategy to generate ACSL4 conditional knockout mice. (b) Validation of ACSL4 knockout by southern blot. The experiments were repeated twice, independently, with similar results. (c) Validation of ACSL4 knockout by genotyping. The experiments were repeated twice, independently, with similar results. (d) U2OS cells were pre-incubated with Nutlin (10 uM) for 12 h, then treated with Nutlin (10 uM), Paraquat (1 mM), Ferr-1 (2 uM), and ML-355 (4 uM) as indicated for 8 h. Quantification of cell death is shown. Error bars are mean ± s.d., n = 3 independent experiments . P values were calculated using two-tailed unpaired Student’s t-test. (e) Schematic diagram of human chromosome 17p13.1 (upper panel) and mouse chromosome 11 (lower panel). ALOX12 and p53 locations indicated with distance between the two genes; MB, megabase. Scanned images of unprocessed blots are shown in Supplementary Fig. 9. Raw data are provided in Supplementary Table 1.

Supplementary Figure 9 Unprocessed images of all gels and blots.

Unprocessed images for Fig. 1b,d,g and 2c. Unprocessed images for Fig. 3a,g and 4c-d. Unprocessed images for Fig. 5a-c,f-g and 6a,e. Unprocessed images for Fig. 7a-b,d-e. Unprocessed images for Supplementary Fig. 1c, 3a,e and 4a-b. Unprocessed images for Supplementary Fig. 4c. Unprocessed images for Supplementary Fig. 6a-f. Unprocessed images for Supplementary Fig. 6h-i and 8b-c.

Supplementary information

Supplementary Information

Supplementary Figures 1–9 and Supplementary Table titles and legends.

Reporting Summary

Supplementary Table 1

Statistics source data.

Supplementary Table 2

Somatic Mutations in Cancer (COSMIC) cancer database identified somatic mutations in individuals with cancer mapping to the lipoxygenase domain of ALOX12.

Supplementary Table 3

The information of primary and secondary antibodies.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chu, B., Kon, N., Chen, D. et al. ALOX12 is required for p53-mediated tumour suppression through a distinct ferroptosis pathway. Nat Cell Biol 21, 579–591 (2019). https://doi.org/10.1038/s41556-019-0305-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-019-0305-6

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