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.

  • Review Article
  • Published:

Transcription factors in ferroptotic cell death

Abstract

Ferroptosis, a form of regulated cell death, is characterized by an excessive degree of iron accumulation and lipid peroxidation. Although it was originally identified only in cells expressing a mutant RAS oncogene, ferroptosis has also been found in normal cells following treatment by small molecules (e.g., erastin and RSL3) or drugs (e.g., sulfasalazine, sorafenib, and artesunate), which target antioxidant enzyme systems, especially the amino acid antiporter system xc− and the glutathione peroxidase GPX4. Dysfunctional ferroptosis is implicated in various physiological and pathological processes (e.g., metabolism, differentiation, and immunity). Targeting the ferroptotic network appears to a new treatment option for diseases or pathological conditions (e.g., cancer, neurodegeneration, and ischemia reperfusion injury). While the molecular machinery of ferroptosis remains largely unknown, several transcription factors (e.g., TP53, NFE2L2/NRF2, ATF3, ATF4, YAP1, TAZ, TFAP2C, SP1, HIF1A, EPAS1/HIF2A, BACH1, TFEB, JUN, HIC1, and HNF4A) play multiple roles in shaping ferroptosis sensitivity through either transcription-dependent or transcription-independent mechanisms. In this review, we summarize recent progress in understanding the transcriptional regulation underlying ferroptotic cell death, and discuss how it has provided new insights into cancer therapy.

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: Key regulators of ferroptosis.
Fig. 2: Role of transcription factors in ferroptotic cell death.

Similar content being viewed by others

References

  1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394–424.

    Article  PubMed  Google Scholar 

  2. Zugazagoitia J, Guedes C, Ponce S, Ferrer I, Molina-Pinelo S, Paz-Ares L. Current challenges in cancer treatment. Clin Ther. 2016;38:1551–66.

    PubMed  Google Scholar 

  3. Chabner BA, Roberts TG Jr. Timeline: chemotherapy and the war on cancer. Nat Rev Cancer. 2005;5:65–72.

    CAS  PubMed  Google Scholar 

  4. Wong RS. Apoptosis in cancer: from pathogenesis to treatment. J Exp Clin Cancer Res. 2011;30:87.

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Mohammad RM, Muqbil I, Lowe L, Yedjou C, Hsu HY, Lin LT, et al. Broad targeting of resistance to apoptosis in cancer. Semin Cancer Biol. 2015;35(Suppl):S78–103.

    PubMed  PubMed Central  Google Scholar 

  6. Hassannia B, Vandenabeele P, Vanden Berghe T. Targeting ferroptosis to iron out cancer. Cancer Cell. 2019;35:830–49.

    CAS  PubMed  Google Scholar 

  7. Friedmann Angeli JP, Krysko DV, Conrad M. Ferroptosis at the crossroads of cancer-acquired drug resistance and immune evasion. Nat Rev Cancer. 2019;19:405–14.

    CAS  PubMed  Google Scholar 

  8. Najafov A, Chen H, Yuan J. Necroptosis and cancer. Trends Cancer. 2017;3:294–301.

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Liu J, Kuang F, Kang R, Tang D. Alkaliptosis: a new weapon for cancer therapy. Cancer Gene Ther. 2019. https://doi.org/10.1038/s41417-019-0134-6 [Epub ahead of print].

  10. Song X, Zhu S, Xie Y, Liu J, Sun L, Zeng D. et al. JTC801 Induces pH-dependent. Gastroenterology. 2018;154:1480–93.

    CAS  PubMed  Google Scholar 

  11. Dolma S, Lessnick SL, Hahn WC, Stockwell BR. Identification of genotype-selective antitumor agents using synthetic lethal chemical screening in engineered human tumor cells. Cancer Cell. 2003;3:285–96.

    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.

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Angeli JPF, 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–U120.

    PubMed Central  Google Scholar 

  14. Zhou B, Liu J, Kang R, Klionsky DJ, Kroemer G, Tang D. Ferroptosis is a type of autophagy-dependent cell death. Semin Cancer Biol. 2019. pii: S1044-579X(19)30006-9. https://doi.org/10.1016/j.semcancer.2019.03.002 [Epub ahead of print].

  15. Hou W, Xie Y, Song X, Sun X, Lotze MT, Zeh HJ 3rd, et al. Autophagy promotes ferroptosis by degradation of ferritin. Autophagy. 2016;12:1425–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Yang M, Chen P, Liu J, Zhu S, Kroemer G, Klionsky DJ, et al. Clockophagy is a novel selective autophagy process favoring ferroptosis. Sci Adv. 2019;5:eaaw2238.

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Wu Z, Geng Y, Lu X, Shi Y, Wu G, Zhang M, et al. Chaperone-mediated autophagy is involved in the execution of ferroptosis. Proc Natl Acad Sci USA. 2019;116:2996–3005.

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Bai Y, Meng L, Han L, Jia Y, Zhao Y, Gao H, et al. Lipid storage and lipophagy regulates ferroptosis. Biochem Biophys Res Commun. 2018;508:997–1003.

  19. Gao H, Bai Y, Jia Y, Zhao Y, Kang R, Tang D, et al. Ferroptosis is a lysosomal cell death process. Biochem Biophys Res Commun. 2018;503:1550–6.

    CAS  PubMed  Google Scholar 

  20. Torii S, Shintoku R, Kubota C, Yaegashi M, Torii R, Sasaki M, et al. An essential role for functional lysosomes in ferroptosis of cancer cells. Biochem J. 2016;473:769–77.

    CAS  PubMed  Google Scholar 

  21. Wen QR, Liu J, Kang R, Zhou BR, Tang D. The release and activity of HMGB1 in ferroptosis. Biochem Bioph Res Co. 2019;510:278–83.

    CAS  Google Scholar 

  22. Gao MH, Yi JM, Zhu JJ, Minikes AM, Monian P, Thompson CB, et al. Role of mitochondria in ferroptosis. Mol Cell. 2019;73:354.

    CAS  PubMed  Google Scholar 

  23. 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.

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Dixon SJ, Stockwell BR. The hallmarks of ferroptosis. Annu Rev Cancer Biol. 2019;3:35–54.

    Google Scholar 

  25. Xie Y, Hou W, Song X, Yu Y, Huang J, Sun X, et al. Ferroptosis: process and function. Cell Death Differ. 2016;23:369–79.

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Yuan H, Li X, Zhang X, Kang R, Tang D. Identification of ACSL4 as a biomarker and contributor of ferroptosis. Biochem Biophys Res Commun. 2016;478:1338–43.

    CAS  PubMed  Google Scholar 

  27. Doll S, Proneth B, Tyurina YY, Panzilius E, Kobayashi S, Ingold I, et al. ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat Chem Biol. 2017;13:91–8.

    CAS  PubMed  Google Scholar 

  28. Kagan VE, Mao G, Qu F, Angeli JP, Doll S, Croix CS, et al. Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis. Nat Chem Biol. 2017;13:81–90.

    CAS  PubMed  Google Scholar 

  29. Yang WS, Kim KJ, Gaschler MM, Patel M, Shchepinov MS, Stockwell BR. Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis. Proc Natl Acad Sci USA. 2016;113:E4966–75.

    CAS  PubMed  PubMed Central  Google Scholar 

  30. 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.

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Bersuker K, Hendricks JM, Li Z, Magtanong L, Ford B, Tang PH, et al. The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature. 2019;575:688–92.

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Doll S, Freitas FP, Shah R, Aldrovandi M, da Silva MC, Ingold I, et al. FSP1 is a glutathione-independent ferroptosis suppressor. Nature. 2019;575:693–8.

    CAS  PubMed  Google Scholar 

  33. Dai E, Meng L, Kang R, Wang X, Tang D. ESCRT-III-dependent membrane repair blocks ferroptosis. Biochem Biophys Res Commun. 2020;522:415–21.

    CAS  PubMed  Google Scholar 

  34. Dai E, Zhang W, Cong D, Kang R, Wang J, Tang D. AIFM2 blocks ferroptosis independent of ubiquinol metabolism. Biochem Biophys Res Commun. 2020;523:966–71.

  35. Dai E, Han L, Liu J, Xie Y, Kroemer G, Klionsky DJ, et al. Autophagy-dependent ferroptosis drives tumor-associated macrophage polarization via release and uptake of oncogenic KRAS protein. Autophagy. 2020. https://doi.org/10.1080/15548627.2020.1714209 [Epub ahead of print].

  36. Bieging KT, Mello SS, Attardi LD. Unravelling mechanisms of p53-mediated tumour suppression. Nat Rev Cancer. 2014;14:359–70.

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Speidel D. Transcription-independent p53 apoptosis: an alternative route to death. Trends Cell Biol. 2010;20:14–24.

    CAS  PubMed  Google Scholar 

  38. Kang R, Kroemer G, Tang D. The tumor suppressor protein p53 and the ferroptosis network. Free Radical Biol Med. 2019;133:162–8.

    CAS  Google Scholar 

  39. Wang GX, Tu HC, Dong YY, Skanderup AJ, Wang YF, Takeda S, et al. Delta Np63 inhibits oxidative stress-induced cell death, including ferroptosis, and cooperates with the BCL-2 family to promote clonogenic survival. Cell Rep. 2017;21:2926–39.

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Wang SJ, Li DW, Ou Y, Jiang L, Chen Y, Zhao YM, et al. Acetylation is crucial for p53-mediated ferroptosis and tumor suppression. Cell Rep. 2016;17:366–73.

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Chu B, Kon N, Chen DL, Li TY, 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.

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Ou Y, Wang SJ, Li DW, Chu B, Gu W. Activation of SAT1 engages polyamine metabolism with p53-mediated ferroptotic responses. Proc Natl Acad Sci USA. 2016;113:E6806–12.

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Gao MH, Monian P, Quadri N, Ramasamy R, Jiang XJ. Glutaminolysis and transferrin regulate ferroptosis. Mol Cell. 2015;59:298–308.

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Zhang C, Liu J, Zhao YH, Yue XT, Zhu Y, Wang XL, et al. Glutaminase 2 is a novel negative regulator of small GTPase Rac1 and mediates p53 function in suppressing metastasis. Elife. 2016;5:e10727. https://doi.org/10.7554/eLife.10727.

  46. Hu W, Zhang C, Wu R, Sun Y, Levine A, Feng Z. Glutaminase 2, a novel p53 target gene regulating energy metabolism and antioxidant function. Proc Natl Acad Sci USA. 2010;107:7455–60.

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Abbas T, Dutta A. p21 in cancer: intricate networks and multiple activities. Nat Rev Cancer. 2009;9:400–14.

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Wang YF, Yang L, Zhang XJ, Cui W, Liu YP, Sun QR, et al. Epigenetic regulation of ferroptosis by H2B monoubiquitination and p53. Embo Rep. 2019;20:e47563. https://doi.org/10.15252/embr.201847563.

  50. Xie YC, Zhu S, Song XX, Sun XF, Fan Y, Liu JB, et al. The tumor suppressor p53 limits ferroptosis by blocking DPP4 activity. Cell Rep. 2017;20:1692–704.

    CAS  PubMed  Google Scholar 

  51. Ma Q. Role of nrf2 in oxidative stress and toxicity. Annu Rev Pharm Toxicol. 2013;53:401–26.

    CAS  Google Scholar 

  52. de la Vega MR, Chapman E, Zhang DD. NRF2 and the hallmarks of cancer. Cancer Cell. 2018;34:21–43.

    PubMed Central  Google Scholar 

  53. Cuadrado A, Manda G, Hassan A, Alcaraz MJ, Barbas C, Daiber A, et al. Transcription factor NRF2 as a therapeutic target for chronic diseases: a systems medicine approach. Pharm Rev. 2018;70:348–83.

    CAS  PubMed  Google Scholar 

  54. Dodson M, Castro-Portuguez R, Zhang DD. NRF2 plays a critical role in mitigating lipid peroxidation and ferroptosis. Redox Biol. 2019;23:101107.

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Sun X, Ou Z, Chen R, Niu X, Chen D, Kang R, et al. Activation of the p62-Keap1-NRF2 pathway protects against ferroptosis in hepatocellular carcinoma cells. Hepatology. 2016;63:173–84.

    CAS  PubMed  Google Scholar 

  56. Sun X, Niu X, Chen R, He W, Chen D, Kang R, et al. Metallothionein-1G facilitates sorafenib resistance through inhibition of ferroptosis. Hepatology. 2016;64:488–500.

    CAS  PubMed  Google Scholar 

  57. Shin D, Kim EH, Lee J, Roh JL. Nrf2 inhibition reverses resistance to GPX4 inhibitor-induced ferroptosis in head and neck cancer. Free Radic Biol Med. 2018;129:454–62.

    CAS  PubMed  Google Scholar 

  58. Geng N, Shi BJ, Li SL, Zhong ZY, Li YC, Xua WL, et al. Knockdown of ferroportin accelerates erastin-induced ferroptosis in neuroblastoma cells. Eur Rev Med Pharm Sci. 2018;22:3826–36.

    CAS  Google Scholar 

  59. Adedoyin O, Boddu R, Traylor A, Lever JM, Bolisetty S, George JF, et al. Heme oxygenase-1 mitigates ferroptosis in renal proximal tubule cells. Am J Physiol Ren Physiol. 2018;314:F702–14.

    CAS  Google Scholar 

  60. Kwon MY, Park E, Lee SJ, Chung SW. Heme oxygenase-1 accelerates erastin-induced ferroptotic cell death. Oncotarget. 2015;6:24393–403.

    PubMed  PubMed Central  Google Scholar 

  61. Hassannia B, Wiernicki B, Ingold I, Qu F, Van Herck S, Tyurina YY, et al. Nano-targeted induction of dual ferroptotic mechanisms eradicates high-risk neuroblastoma. J Clin Invest. 2018;128:3341–55.

    PubMed  PubMed Central  Google Scholar 

  62. Chiang SK, Chen SE, Chang LC. A dual role of heme oxygenase-1 in cancer cells. Int J Mol Sci. 2018;20:E39.

  63. Nishizawa S, Araki H, Ishikawa Y, Kitazawa S, Hata A, Soga T, et al. Low tumor glutathione level as a sensitivity marker for glutamate-cysteine ligase inhibitors. Oncol Lett. 2018;15:8735–43.

    PubMed  PubMed Central  Google Scholar 

  64. Marengo B, De Ciucis C, Verzola D, Pistoia V, Raffaghello L, Patriarca S, et al. Mechanisms of BSO (L-buthionine-S,R-sulfoximine)-induced cytotoxic effects in neuroblastoma. Free Radic Biol Med. 2008;44:474–82.

    CAS  PubMed  Google Scholar 

  65. Wu KC, Cui JY, Klaassen CD. Beneficial role of Nrf2 in regulating NADPH generation and consumption. Toxicol Sci. 2011;123:590–600.

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Shimada K, Hayano M, Pagano NC, Stockwell BR. Cell-Line selectivity improves the predictive power of pharmacogenomic analyses and helps identify NADPH as biomarker for ferroptosis sensitivity. Cell Chem Biol. 2016;23:225–35.

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Yang WH, Huang Z, Wu J, Ding CC, Murphy SK, Chi JT. A TAZ-ANGPTL4-NOX2 axis regulates ferroptotic cell death and chemoresistance in epithelial ovarian cancer. Mol Cancer Res. 2020;18:79–90.

    CAS  PubMed  Google Scholar 

  68. Gagliardi M, Cotella D, Santoro C, Cora D, Barlev NA, Piacentini M, et al. Aldo-keto reductases protect metastatic melanoma from ER stress-independent ferroptosis. Cell Death Dis. 2019;10:902.

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Almanza A, Carlesso A, Chintha C, Creedican S, Doultsinos D, Leuzzi B, et al. Endoplasmic reticulum stress signalling—from basic mechanisms to clinical applications. FEBS J. 2019;286:241–78.

    CAS  PubMed  Google Scholar 

  70. Dixon SJ, Patel D, Welsch M, Skouta R, Lee E, Hayano M, et al. Pharmacological inhibition of cystine-glutamate exchange induces endoplasmic reticulum stress and ferroptosis. Elife. 2014;3:e02523.

    PubMed  PubMed Central  Google Scholar 

  71. Sun Y, Zheng Y, Wang C, Liu Y. Glutathione depletion induces ferroptosis, autophagy, and premature cell senescence in retinal pigment epithelial cells. Cell Death Dis. 2018;9:753.

    PubMed  PubMed Central  Google Scholar 

  72. 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.

  73. Stelzl U, Worm U, Lalowski M, Haenig C, Brembeck FH, Goehler H, et al. A human protein–protein interaction network: a resource for annotating the proteome. Cell. 2005;122:957–68.

    CAS  PubMed  Google Scholar 

  74. Kim KH, Jeong JY, Surh YJ, Kim KW. Expression of stress-response ATF3 is mediated by Nrf2 in astrocytes. Nucleic Acids Res. 2010;38:48–59.

    CAS  PubMed  Google Scholar 

  75. Chen D, Fan Z, Rauh M, Buchfelder M, Eyupoglu IY, Savaskan N. ATF4 promotes angiogenesis and neuronal cell death and confers ferroptosis in a xCT-dependent manner. Oncogene. 2017;36:5593–608.

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Zhu S, Zhang Q, Sun X, Zeh HJ 3rd, Lotze MT, Kang R, et al. HSPA5 regulates ferroptotic cell death in cancer cells. Cancer Res. 2017;77:2064–77.

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Wang N, Zeng GZ, Yin JL, Bian ZX. Artesunate activates the ATF4-CHOP-CHAC1 pathway and affects ferroptosis in Burkitt’s lymphoma. Biochem Biophys Res Commun. 2019;519:533–9.

    CAS  PubMed  Google Scholar 

  78. Ye P, Mimura J, Okada T, Sato H, Liu T, Maruyama A. et al. Nrf2- and ATF4-dependent upregulation of xCT modulates the sensitivity of T24 bladder carcinoma cells to proteasome inhibition (vol 34, pg 3421, 2014). Mol Cell Biol. 2015;35:2366

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Koppula P, Zhang YL, Shi JJ, Li W, Gan BY. The glutamate/cystine antiporter SLC7A11/xCT enhances cancer cell dependency on glucose by exporting glutamate. J Biol Chem. 2017;292:14240–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Luo MY, Wu LF, Zhang KX, Wang H, Zhang T, Gutierrez L, et al. miR-137 regulates ferroptosis by targeting glutamine transporter SLC1A5 in melanoma. Cell Death Differ. 2018;25:1457–72.

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Chen MS, Wang SF, Hsu CY, Yin PH, Yeh TS, Lee HC, et al. CHAC1 degradation of glutathione enhances cystine-starvation-induced necroptosis and ferroptosis in human triple negative breast cancer cells via the GCN2-eIF2alpha-ATF4 pathway. Oncotarget. 2017;8:114588–602.

    PubMed  PubMed Central  Google Scholar 

  82. Lim JKM, Delaidelli A, Minaker SW, Zhang HF, Colovic M, Yang H, et al. Cystine/glutamate antiporter xCT (SLC7A11) facilitates oncogenic RAS transformation by preserving intracellular redox balance. Proc Natl Acad Sci USA. 2019;116:9433–42.

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Harvey KF, Zhang X, Thomas DM. The Hippo pathway and human cancer. Nat Rev Cancer. 2013;13:246–57.

    CAS  PubMed  Google Scholar 

  84. Wu J, Minikes AM, Gao M, Bian H, Li Y, Stockwell BR, et al. Intercellular interaction dictates cancer cell ferroptosis via NF2-YAP signalling. Nature. 2019;572:402–6.

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Yang WH, Ding CC, Sun T, Rupprecht G, Lin CC, Hsu D, et al. The hippo pathway effector TAZ regulates ferroptosis in renal cell carcinoma. Cell Rep. 2019;28:2501–8. e4.

    CAS  PubMed  Google Scholar 

  86. Romero FJ, Bosch-Morell F, Romero MJ, Jareno EJ, Romero B, Marin N, et al. Lipid peroxidation products and antioxidants in human disease. Environ Health Perspect. 1998;106(Suppl. 5):1229–34.

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Ran Q, Liang H, Gu M, Qi W, Walter CA, Roberts LJ 2nd, et al. Transgenic mice overexpressing glutathione peroxidase 4 are protected against oxidative stress-induced apoptosis. J Biol Chem. 2004;279:55137–46.

    CAS  PubMed  Google Scholar 

  88. Canli O, Alankus YB, Grootjans S, Vegi N, Hultner L, Hoppe PS, et al. Glutathione peroxidase 4 prevents necroptosis in mouse erythroid precursors. Blood. 2016;127:139–48.

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Kang R, Zeng L, Zhu S, Xie Y, Liu J, Wen Q, et al. Lipid peroxidation drives gasdermin D-mediated pyroptosis in lethal polymicrobial sepsis. Cell Host Microbe. 2018;24:97–108. e4.

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Kulak MV, Cyr AR, Woodfield GW, Bogachek M, Spanheimer PM, Li T, et al. Transcriptional regulation of the GPX1 gene by TFAP2C and aberrant CpG methylation in human breast cancer. Oncogene. 2013;32:4043–51.

    CAS  PubMed  Google Scholar 

  91. Alim I, Caulfield JT, Chen Y, Swarup V, Geschwind DH, Ivanova E, et al. Selenium drives a transcriptional adaptive program to block ferroptosis and treat stroke. Cell. 2019;177:1262–79. e25.

    CAS  PubMed  Google Scholar 

  92. Wang X, Sun D, Tai JD, Chen S, Yu M, Ren D, et al. TFAP2C promotes stemness and chemotherapeutic resistance in colorectal cancer via inactivating hippo signaling pathway. J Exp Clin Canc Res. 2018;37:27.

  93. De Andrade JP, Park JM, Gu VW, Woodfield GW, Kulak MV, Lorenzen AW, et al. EGFR is regulated by TFAP2C in luminal breast cancer and is a target for vandetanib. Mol Cancer Ther. 2016;15:503–11.

    PubMed  PubMed Central  Google Scholar 

  94. Williams CM, Scibetta AG, Friedrich JK, Canosa M, Berlato C, Moss CH, et al. AP-2gamma promotes proliferation in breast tumour cells by direct repression of the CDKN1A gene. EMBO J. 2009;28:3591–601.

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Semenza GL. Hypoxia-inducible factors in physiology and medicine. Cell. 2012;148:399–408.

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Liu J, Yang M, Kang R, Klionsky DJ, Tang D. Autophagic degradation of the circadian clock regulator promotes ferroptosis. Autophagy. 2019;15:2033–5.

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Zou Y, Palte MJ, Deik AA, Li H, Eaton JK, Wang W, et al. A GPX4-dependent cancer cell state underlies the clear-cell morphology and confers sensitivity to ferroptosis. Nat Commun. 2019;10:1617.

    PubMed  PubMed Central  Google Scholar 

  98. Nishizawa H, Matsumoto M, Shindo T, Saigusa D, Kato H, Suzuki K, et al. Ferroptosis is controlled by the coordinated transcriptional regulation of glutathione and labile iron metabolism by the transcription factor BACH1. J Biol Chem. 2020;295:69–82.

  99. Li L, Sun S, Tan L, Wang Y, Wang L, Zhang Z, et al. Polystyrene nanoparticles reduced ROS and inhibited ferroptosis by triggering lysosome stress and TFEB nucleus translocation in a size-dependent manner. Nano Lett. 2019;19:7781–92.

  100. Chen Y, Zhu G, Liu Y, Wu Q, Zhang X, Bian Z, et al. O-GlcNAcylated c-Jun antagonizes ferroptosis via inhibiting GSH synthesis in liver cancer. Cell Signal. 2019;63:109384.

    CAS  PubMed  Google Scholar 

  101. Zhang X, Du LT, Qiao YX, Zhang XB, Zheng WS, Wu Q, et al. Ferroptosis is governed by differential regulation of transcription in liver cancer. Redox Biol. 2019;24:101211. https://doi.org/10.1016/j.redox.2019.101211.

  102. Tang D, Kang R, Berghe TV, Vandenabeele P, Kroemer G. The molecular machinery of regulated cell death. Cell Res. 2019;29:347–64.

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We apologize to those whose work was not referenced either due to space limitations or our oversight. We thank Dave Primm (Department of Surgery, University of Texas Southwestern Medical Center) for his critical reading of the manuscript. D.T. is supported by grants from the US National Institutes of Health (R01CA229275 and R01CA160417). R.K. is supported by a grant from the US National Institutes of Health (R01CA211070).

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Rui Kang or Daolin Tang.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Additional information

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

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dai, C., Chen, X., Li, J. et al. Transcription factors in ferroptotic cell death. Cancer Gene Ther 27, 645–656 (2020). https://doi.org/10.1038/s41417-020-0170-2

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41417-020-0170-2

This article is cited by

Search

Quick links