35,375
Views
571
CrossRef citations to date
0
Altmetric
Review

Ferroptosis: machinery and regulation

, , , ORCID Icon & ORCID Icon
Pages 2054-2081 | Received 10 Jun 2020, Accepted 06 Aug 2020, Published online: 26 Aug 2020

References

  • Galluzzi L, Vitale I, Aaronson SA, et al. Molecular mechanisms of cell death: recommendations of the nomenclature committee on cell death 2018. Cell Death Differ. 2018;25:486–541.
  • Dixon SJ, Lemberg KM, Lamprecht MR, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell. 2012;149(5):1060–1072.
  • Weinlich R, Oberst A, Beere HM, et al. Necroptosis in development, inflammation and disease. Nat Rev Mol Cell Biol. 2017;18(2):127–136.
  • Bergsbaken T, Fink SL, Cookson BT. Pyroptosis: host cell death and inflammation. Nat Rev Microbiol. 2009;7(2):99–109.
  • Song X, Zhu S, Xie Y, et al. JTC801 Induces pH-dependent Death Specifically in Cancer Cells and Slows Growth of Tumors in Mice. Gastroenterology. 2018;154(5):1480–1493.
  • Liu J, Kuang F, Kang R, et al. Alkaliptosis: a new weapon for cancer therapy. Cancer Gene Ther. 2020;27(5):267–269.
  • Tang D, Kang R, Berghe TV, et al. The molecular machinery of regulated cell death. Cell Res. 2019;29:347–364.
  • Xie Y, Hou W, Song X, et al. Ferroptosis: process and function. Cell Death Differ. 2016;23(3):369–379.
  • Stockwell BR, Friedmann Angeli JP, Bayir H, et al. Ferroptosis: a regulated cell death nexus linking metabolism, redox biology, and disease. Cell. 2017;171(2):273–285.
  • Liu J, Kuang F, Kroemer G, et al. Autophagy-dependent ferroptosis: machinery and regulation. Cell Chem Biol. 2020;27(4):420–435.
  • Zhou B, Liu J, Kang R, et al. Ferroptosis is a type of autophagy-dependent cell death. Semin Cancer Biol. 2019. DOI:https://doi.org/10.1016/j.semcancer.2019.03.002
  • Kang R, Tang D. Autophagy and ferroptosis - what’s the connection? Curr Pathobiol Rep. 2017;5(2):153–159.
  • Dolma S, Lessnick SL, Hahn WC, et al. Identification of genotype-selective antitumor agents using synthetic lethal chemical screening in engineered human tumor cells. Cancer Cell. 2003;3(3):285–296.
  • Yang WS, SriRamaratnam R, Welsch ME, et al. Regulation of ferroptotic cancer cell death by GPX4. Cell. 2014;156(1–2):317–331.
  • Dai E, Han L, Liu J, et al. Autophagy-dependent ferroptosis drives tumor-associated macrophage polarization via release and uptake of oncogenic KRAS protein. Autophagy. 2020;1–15. DOI:https://doi.org/10.1080/15548627.2020.1714209
  • Hassannia B, Vandenabeele P, Vanden Berghe T. Targeting ferroptosis to iron out cancer. Cancer Cell. 2019;35(6):830–849.
  • Tan S, Schubert D, Maher P. Oxytosis: a novel form of programmed cell death. Curr Top Med Chem. 2001;1(6):497–506.
  • Sato H, Shiiya A, Kimata M, et al. Redox imbalance in cystine/glutamate transporter-deficient mice. J Biol Chem. 2005;280(45):37423–37429.
  • Wang H, An P, Xie E, et al. Characterization of ferroptosis in murine models of hemochromatosis. Hepatology. 2017;66(2):449–465.
  • Badgley MA, Kremer DM, Maurer HC, et al. Cysteine depletion induces pancreatic tumor ferroptosis in mice. Science. 2020;368(6486):85–89.
  • Imai H, Hirao F, Sakamoto T, et al. Early embryonic lethality caused by targeted disruption of the mouse PHGPx gene. Biochem Biophys Res Commun. 2003;305(2):278–286.
  • Yant LJ, Ran Q, Rao L, et al. The selenoprotein GPX4 is essential for mouse development and protects from radiation and oxidative damage insults. Free Radic Biol Med. 2003;34(4):496–502.
  • Ingold I, Aichler M, Yefremova E, 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. 2015;290(23):14668–14678.
  • Brutsch SH, Wang CC, Li L, et al. Expression of inactive glutathione peroxidase 4 leads to embryonic lethality, and inactivation of the Alox15 gene does not rescue such knock-in mice. Antioxid Redox Signal. 2015;22(4):281–293.
  • Ran Q, Van Remmen H, Gu M, et al. Embryonic fibroblasts from Gpx4± mice: a novel model for studying the role of membrane peroxidation in biological processes. Free Radic Biol Med. 2003;35(9):1101–1109.
  • Canli O, Alankus YB, Grootjans S, et al. Glutathione peroxidase 4 prevents necroptosis in mouse erythroid precursors. Blood. 2016;127(1):139–148.
  • Kang R, Zeng L, Zhu S, et al. Lipid Peroxidation Drives Gasdermin D-Mediated Pyroptosis in Lethal Polymicrobial Sepsis. Cell Host Microbe. 2018;24(1):97–108 e4.
  • Bosl MR, Takaku K, Oshima M, et al. Early embryonic lethality caused by targeted disruption of the mouse selenocysteine tRNA gene (Trsp). Proc Natl Acad Sci U S A. 1997;94(11):5531–5534.
  • Ingold I, Berndt C, Schmitt S, et al. Selenium Utilization by GPX4 Is Required to Prevent Hydroperoxide-Induced Ferroptosis. Cell. 2018;172(3):409–22 e21.
  • Alim I, Caulfield JT, Chen Y, et al. Selenium drives a transcriptional adaptive program to block ferroptosis and treat stroke. Cell. 2019;177(5):1262–79 e25.
  • Gao J, Yang F, Che J, et al. Selenium-encoded isotopic signature targeted profiling. ACS Cent Sci. 2018;4(8):960–970.
  • Llabani E, Hicklin RW, Lee HY, et al. Diverse compounds from pleuromutilin lead to a thioredoxin inhibitor and inducer of ferroptosis. Nat Chem. 2019;11(6):521–532.
  • Shimada K, Skouta R, Kaplan A, et al. Global survey of cell death mechanisms reveals metabolic regulation of ferroptosis. Nat Chem Biol. 2016;12(7):497–503.
  • Doll S, Freitas FP, Shah R, et al. FSP1 is a glutathione-independent ferroptosis suppressor. Nature. 2019;575(7784):693–698.
  • Bersuker K, Hendricks JM, Li Z, et al. The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature. 2019;575(7784):688–692.
  • Kraft VAN, Bezjian CT, Pfeiffer S, et al. GTP cyclohydrolase 1/tetrahydrobiopterin counteract ferroptosis through lipid remodeling. ACS Cent Sci. 2020;6(1):41–53.
  • Dai E, Zhang W, Cong D, et al. AIFM2 blocks ferroptosis independent of ubiquinol metabolism. Biochem Biophys Res Commun. 2020;523(4):966–971.
  • Wu M, Xu LG, Li X, et al. AMID, an apoptosis-inducing factor-homologous mitochondrion-associated protein, induces caspase-independent apoptosis. J Biol Chem. 2002;277(28):25617–25623.
  • Miriyala S, Thippakorn C, Chaiswing L, et al. Novel role of 4-hydroxy-2-nonenal in AIFm2-mediated mitochondrial stress signaling. Free Radic Biol Med. 2016;91:68–80.
  • Ma S, Henson ES, Chen Y, et al. Ferroptosis is induced following siramesine and lapatinib treatment of breast cancer cells. Cell Death Dis. 2016;7(7):e2307.
  • Gao M, Monian P, Quadri N, et al. Glutaminolysis and transferrin regulate ferroptosis. Mol Cell. 2015;59(2):298–308.
  • Yang WS, Stockwell BR. Synthetic lethal screening identifies compounds activating iron-dependent, nonapoptotic cell death in oncogenic-RAS-harboring cancer cells. Chem Biol. 2008;15(3):234–245.
  • Lane DJ, Merlot AM, Huang ML, et al. Cellular iron uptake, trafficking and metabolism: key molecules and mechanisms and their roles in disease. Biochim Biophys Acta. 2015;1853(5):1130–1144.
  • Knutson MD. Non-transferrin-bound iron transporters. Free Radic Biol Med. 2019;133:101–111.
  • Song X, Zhu S, Chen P, et al. AMPK-mediated BECN1 phosphorylation promotes ferroptosis by directly blocking system Xc(-) activity. Curr Biol. 2018;28(15):2388–99 e5.
  • Yu Y, Jiang L, Wang H, et al. Hepatic transferrin plays a role in systemic iron homeostasis and liver ferroptosis. Blood. 2020;136(6):726–739.
  • Gozzelino R, Soares MP. Coupling heme and iron metabolism via ferritin H chain. Antioxid Redox Signal. 2014;20(11):1754–1769.
  • Imoto S, Kono M, Suzuki T, et al. Haemin-induced cell death in human monocytic cells is consistent with ferroptosis. Transfus Apher Sci. 2018;57(4):524–531.
  • Hassannia B, Wiernicki B, Ingold I, et al. Nano-targeted induction of dual ferroptotic mechanisms eradicates high-risk neuroblastoma. J Clin Invest. 2018;128(8):3341–3355.
  • Sun X, Ou Z, Chen R, et al. Activation of the p62-Keap1-NRF2 pathway protects against ferroptosis in hepatocellular carcinoma cells. Hepatology. 2016;63(1):173–184.
  • Egyed A, Saltman P. Iron is maintained as Fe(II) under aerobic conditions in erythroid cells. Biol Trace Elem Res. 1984;6(4):357–364.
  • Breuer W, Epsztejn S, Cabantchik ZI. Iron acquired from transferrin by K562 cells is delivered into a cytoplasmic pool of chelatable iron(II). J Biol Chem. 1995;270(41):24209–24215.
  • Philpott CC, Jadhav S. The ins and outs of iron: escorting iron through the mammalian cytosol. Free Radic Biol Med. 2019;133:112–117.
  • Nandal A, Ruiz JC, Subramanian P, et al. Activation of the HIF prolyl hydroxylase by the iron chaperones PCBP1 and PCBP2. Cell Metab. 2011;14(5):647–657.
  • Protchenko O, Baratz E, Jadhav S, et al. Iron chaperone PCBP1 protects murine liver from lipid peroxidation and steatosis. Hepatology. 2020. DOI:https://doi.org/10.1002/hep.31328.
  • Alvarez SW, Sviderskiy VO, Terzi EM, et al. NFS1 undergoes positive selection in lung tumours and protects cells from ferroptosis. Nature. 2017;551(7682):639–643.
  • Shaw GC, Cope JJ, Li L, et al. Mitoferrin is essential for erythroid iron assimilation. Nature. 2006;440(7080):96–100.
  • Paradkar PN, Zumbrennen KB, Paw BH, et al. Regulation of mitochondrial iron import through differential turnover of mitoferrin 1 and mitoferrin 2. Mol Cell Biol. 2009;29(4):1007–1016.
  • Li C, Zhang Y, Cheng X, et al. PINK1 and PARK2 suppress pancreatic tumorigenesis through control of mitochondrial iron-mediated immunometabolism. Dev Cell. 2018;46(4):441–55 e8.
  • Kang R, Xie Y, Zeh HJ, et al. Mitochondrial quality control mediated by PINK1 and PRKN: links to iron metabolism and tumor immunity. Autophagy. 2019;15(1):172–173.
  • Chiabrando D, Marro S, Mercurio S, et al. The mitochondrial heme exporter FLVCR1b mediates erythroid differentiation. J Clin Invest. 2012;122(12):4569–4579.
  • Muckenthaler MU, Rivella S, Hentze MW, et al. A red carpet for iron metabolism. Cell. 2017;168(3):344–361.
  • Yuan H, Li X, Zhang X, et al. CISD1 inhibits ferroptosis by protection against mitochondrial lipid peroxidation. Biochem Biophys Res Commun. 2016;478(2):838–844.
  • Mancias JD, Wang X, Gygi SP, et al. Quantitative proteomics identifies NCOA4 as the cargo receptor mediating ferritinophagy. Nature. 2014;509(7498):105–109.
  • De Domenico I, Vaughn MB, Li L, et al. Ferroportin-mediated mobilization of ferritin iron precedes ferritin degradation by the proteasome. Embo J. 2006;25(22):5396–5404.
  • Wang YQ, Chang SY, Wu Q, et al. The protective role of mitochondrial ferritin on erastin-induced ferroptosis. Front Aging Neurosci. 2016;8:308.
  • Hou W, Xie Y, Song X, et al. Autophagy promotes ferroptosis by degradation of ferritin. Autophagy. 2016;12(8):1425–1428.
  • Gao M, Monian P, Pan Q, et al. Ferroptosis is an autophagic cell death process. Cell Res. 2016;26(9):1021–1032.
  • Brown CW, Amante JJ, Chhoy P, et al. Prominin2 drives ferroptosis resistance by stimulating iron export. Dev Cell. 2019;51(5):575–86 e4.
  • Shang Y, Luo M, Yao F, et al. Ceruloplasmin suppresses ferroptosis by regulating iron homeostasis in hepatocellular carcinoma cells. Cell Signal. 2020;72:109633.
  • Tuo QZ, Lei P, Jackman KA, et al. Tau-mediated iron export prevents ferroptotic damage after ischemic stroke. Mol Psychiatry. 2017;22(11):1520–1530.
  • Detivaud L, Island ML, Jouanolle AM, et al. Ferroportin diseases: functional studies, a link between genetic and clinical phenotype. Hum Mutat. 2013;34(11):1529–1536.
  • Bogdan AR, Miyazawa M, Hashimoto K, et al. Regulators of iron homeostasis: new players in metabolism, cell death, and disease. Trends Biochem Sci. 2016;41(3):274–286.
  • Geng N, Shi BJ, Li SL, et al. Knockdown of ferroportin accelerates erastin-induced ferroptosis in neuroblastoma cells. Eur Rev Med Pharmacol Sci. 2018;22(12):3826–3836.
  • Tesfay L, Paul BT, Konstorum A, et al. Stearoyl-CoA desaturase 1 protects ovarian cancer cells from ferroptotic cell death. Cancer Res. 2019;79(20):5355–5366.
  • Jiang Y, Mao C, Yang R, et al. EGLN1/c-Myc induced lymphoid-specific helicase inhibits ferroptosis through lipid metabolic gene expression changes. Theranostics. 2017;7:3293–3305.
  • Magtanong L, Ko PJ, To M, et al. Exogenous monounsaturated fatty acids promote a ferroptosis-resistant cell state. Cell Chem Biol. 2019;26(3):420–32 e9.
  • Lee H, Zandkarimi F, Zhang Y, et al. Energy-stress-mediated AMPK activation inhibits ferroptosis. Nat Cell Biol. 2020;22(2):225–234.
  • Plotz T, Hartmann M, Lenzen S, et al. The role of lipid droplet formation in the protection of unsaturated fatty acids against palmitic acid induced lipotoxicity to rat insulin-producing cells. Nutr Metab (Lond). 2016;13(1):16.
  • Bai Y, Meng L, Han L, et al. Lipid storage and lipophagy regulates ferroptosis. Biochem Biophys Res Commun. 2019;508(4):997–1003.
  • Kagan VE, Mao G, Qu F, et al. Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis. Nat Chem Biol. 2017;13(1):81–90.
  • Miess H, Dankworth B, Gouw AM, et al. The glutathione redox system is essential to prevent ferroptosis caused by impaired lipid metabolism in clear cell renal cell carcinoma. Oncogene. 2018;37(40):5435–5450.
  • Blomme A, Ford CA, Mui E, et al. 2,4-dienoyl-CoA reductase regulates lipid homeostasis in treatment-resistant prostate cancer. Nat Commun. 2020;11(1):2508.
  • Doll S, Proneth B, Tyurina YY, et al. ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat Chem Biol. 2017;13(1):91–98.
  • Yuan H, Li X, Zhang X, et al. Identification of ACSL4 as a biomarker and contributor of ferroptosis. Biochem Biophys Res Commun. 2016;478(3):1338–1343.
  • Chu B, Kon N, Chen D, et al. ALOX12 is required for p53-mediated tumour suppression through a distinct ferroptosis pathway. Nat Cell Biol. 2019;21(5):579–591.
  • Haeggstrom JZ, Funk CD. Lipoxygenase and leukotriene pathways: biochemistry, biology, and roles in disease. Chem Rev. 2011;111(10):5866–5898.
  • Schnurr K, Belkner J, Ursini F, et al. The selenoenzyme phospholipid hydroperoxide glutathione peroxidase controls the activity of the 15-lipoxygenase with complex substrates and preserves the specificity of the oxygenation products. J Biol Chem. 1996;271(9):4653–4658.
  • Conrad M, Sandin A, Forster H, et al. 12/15-lipoxygenase-derived lipid peroxides control receptor tyrosine kinase signaling through oxidation of protein tyrosine phosphatases. Proc Natl Acad Sci U S A. 2010;107(36):15774–15779.
  • Yang WS, Kim KJ, Gaschler MM, et al. Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis. Proc Natl Acad Sci U S A. 2016;113(34):E4966–75.
  • Wenzel SE, Tyurina YY, Zhao J, et al. PEBP1 wardens ferroptosis by enabling lipoxygenase generation of lipid death signals. Cell. 2017;171(3):628–41 e26.
  • Friedmann Angeli JP, Schneider M, Proneth B, et al. Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nat Cell Biol. 2014;16(12):1180–1191.
  • Matsushita M, Freigang S, Schneider C, et al. T cell lipid peroxidation induces ferroptosis and prevents immunity to infection. J Exp Med. 2015;212(4):555–568.
  • Zou Y, Li H, Graham ET, et al. Cytochrome P450 oxidoreductase contributes to phospholipid peroxidation in ferroptosis. Nat Chem Biol. 2020;16(3):302–309.
  • Wang L, Cai H, Hu Y, et al. A pharmacological probe identifies cystathionine beta-synthase as a new negative regulator for ferroptosis. Cell Death Dis. 2018;9(10):1005.
  • Martinez AM, Mirkovic J, Stanisz ZA, et al. NSC-34 motor neuron-like cells are sensitized to ferroptosis upon differentiation. FEBS Open Bio. 2019;9(4):582–593.
  • Wang M, Mao C, Ouyang L, et al. Long noncoding RNA LINC00336 inhibits ferroptosis in lung cancer by functioning as a competing endogenous RNA. Cell Death Differ. 2019;26(11):2329–2343.
  • Hayano M, Yang WS, Corn CK, et al. Loss of cysteinyl-tRNA synthetase (CARS) induces the transsulfuration pathway and inhibits ferroptosis induced by cystine deprivation. Cell Death Differ. 2016;23(2):270–278.
  • Cao J, Chen X, Jiang L, et al. DJ-1 suppresses ferroptosis through preserving the activity of S-adenosyl homocysteine hydrolase. Nat Commun. 2020;11(1):1251.
  • Shin D, Lee J, You JH, et al. Dihydrolipoamide dehydrogenase regulates cystine deprivation-induced ferroptosis in head and neck cancer. Redox Biol. 2020;30:101418.
  • Jennis M, Kung CP, Basu S, et al. An African-specific polymorphism in the TP53 gene impairs p53 tumor suppressor function in a mouse model. Genes Dev. 2016;30(8):918–930.
  • Niu Y, Zhang J, Tong Y, et al. Physcion 8-O-beta-glucopyranoside induced ferroptosis via regulating miR-103a-3p/GLS2 axis in gastric cancer. Life Sci. 2019;237:116893.
  • Luo M, Wu L, Zhang K, et al. miR-137 regulates ferroptosis by targeting glutamine transporter SLC1A5 in melanoma. Cell Death Differ. 2018;25(8):1457–1472.
  • Shimada K, Hayano M, Pagano NC, et al. Cell-line selectivity improves the predictive power of pharmacogenomic analyses and helps identify NADPH as biomarker for ferroptosis sensitivity. Cell Chem Biol. 2016;23(2):225–235.
  • Kim H, Lee JH, Park JW. Down-regulation of IDH2 sensitizes cancer cells to erastin-induced ferroptosis. Biochem Biophys Res Commun. 2020;525(2):366–371.
  • Wang TX, Liang JY, Zhang C, et al. The oncometabolite 2-hydroxyglutarate produced by mutant IDH1 sensitizes cells to ferroptosis. Cell Death Dis. 2019;10(10):755.
  • Ding CC, Rose J, Sun T, et al. MESH1 is a cytosolic NADPH phosphatase that regulates ferroptosis. Nat Metab. 2020;2(3):270–277.
  • Xie Y, Zhu S, Song X, et al. The tumor suppressor p53 limits ferroptosis by blocking DPP4 activity. Cell Rep. 2017;20(7):1692–1704.
  • Yang WH, Ding CC, Sun T, et al. The hippo pathway effector TAZ regulates ferroptosis in renal cell carcinoma. Cell Rep. 2019;28(10):2501–8 e4.
  • Yang WH, Huang Z, Wu J, et al. A TAZ-ANGPTL4-NOX2 axis regulates ferroptotic cell death and chemoresistance in epithelial ovarian cancer. Mol Cancer Res. 2020;18(1):79–90.
  • Gaschler MM, Hu F, Feng H, et al. Determination of the subcellular localization and mechanism of action of ferrostatins in suppressing ferroptosis. ACS Chem Biol. 2018;13(4):1013–1020.
  • Gao M, Yi J, Zhu J, et al. Role of mitochondria in ferroptosis. Mol Cell. 2019;73(2):354–63 e3.
  • Agmon E, Solon J, Bassereau P, et al. Modeling the effects of lipid peroxidation during ferroptosis on membrane properties. Sci Rep. 2018;8(1):5155.
  • Catala A. Lipid peroxidation of membrane phospholipids generates hydroxy-alkenals and oxidized phospholipids active in physiological and/or pathological conditions. Chem Phys Lipids. 2009;157(1):1–11.
  • Dixon SJ, Patel DN, Welsch M, et al. Pharmacological inhibition of cystine-glutamate exchange induces endoplasmic reticulum stress and ferroptosis. Elife. 2014;3:e02523.
  • Jimenez AJ, Maiuri P, Lafaurie-Janvore J, et al. ESCRT machinery is required for plasma membrane repair. Science. 2014;343(6174):1247136.
  • Dai E, Meng L, Kang R, et al. ESCRT-III-dependent membrane repair blocks ferroptosis. Biochem Biophys Res Commun. 2020;522(2):415–421.
  • Gong YN, Guy C, Olauson H, et al. ESCRT-III acts downstream of MLKL to regulate necroptotic cell death and its consequences. Cell. 2017;169(2):286–300 e16.
  • Ruhl S, Shkarina K, Demarco B, et al. ESCRT-dependent membrane repair negatively regulates pyroptosis downstream of GSDMD activation. Science. 2018;362(6417):956–960.
  • Liu J, Kang R, Tang D. ESCRT-III-mediated membrane repair in cell death and tumor resistance. Cancer Gene Ther. 2020.
  • Zhang Y, Tan H, Daniels JD, et al. Imidazole ketone erastin induces ferroptosis and slows tumor growth in a mouse lymphoma model. Cell Chem Biol. 2019;26(5):623–33 e9.
  • Eaton JK, Furst L, Ruberto RA, et al. Selective covalent targeting of GPX4 using masked nitrile-oxide electrophiles. Nat Chem Biol. 2020;16(5):497–506.
  • Eaton JK, Ruberto RA, Kramm A, et al. Diacylfuroxans Are Masked Nitrile Oxides That Inhibit GPX4 Covalently. J Am Chem Soc. 2019;141(51):20407–20415.
  • Wenz C, Faust D, Linz B, et al. t-BuOOH induces ferroptosis in human and murine cell lines. Arch Toxicol. 2018;92(2):759–775.
  • Gaschler MM, Andia AA, Liu H, et al. FINO2 initiates ferroptosis through GPX4 inactivation and iron oxidation. Nat Chem Biol. 2018;14(5):507–515.
  • Li Q, Han X, Lan X, et al. Inhibition of neuronal ferroptosis protects hemorrhagic brain. JCI Insight. 2017;2(7):e90777.
  • Baba Y, Higa JK, Shimada BK, et al. Protective effects of the mechanistic target of rapamycin against excess iron and ferroptosis in cardiomyocytes. Am J Physiol Heart Circ Physiol. 2018;314(3):H659–H68.
  • Yoshida M, Minagawa S, Araya J, et al. Involvement of cigarette smoke-induced epithelial cell ferroptosis in COPD pathogenesis. Nat Commun. 2019;10(1):3145.
  • Mai TT, Hamai A, Hienzsch A, et al. Salinomycin kills cancer stem cells by sequestering iron in lysosomes. Nat Chem. 2017;9(10):1025–1033.
  • Sui S, Zhang J, Xu S, et al. Ferritinophagy is required for the induction of ferroptosis by the bromodomain protein BRD4 inhibitor (+)-JQ1 in cancer cells. Cell Death Dis. 2019;10(5):331.
  • Li C, Zhang Y, Liu J, et al. Mitochondrial DNA stress triggers autophagy-dependent ferroptotic death. Autophagy. 2020 Mar 18;1-13. doi: https://doi.org/10.1080/15548627.2020.1739447.
  • Conrad M, Pratt DA. The chemical basis of ferroptosis. Nat Chem Biol. 2019;15(12):1137–1147.
  • Zilka O, Shah R, Li B, et al. On the mechanism of cytoprotection by ferrostatin-1 and liproxstatin-1 and the role of lipid peroxidation in ferroptotic cell death. ACS Cent Sci. 2017;3(3):232–243.
  • Miotto G, Rossetto M, Di Paolo ML, et al. Insight into the mechanism of ferroptosis inhibition by ferrostatin-1. Redox Biol. 2020;28:101328.
  • Shah R, Margison K, Pratt DA. The Potency of Diarylamine Radical-Trapping Antioxidants as Inhibitors of Ferroptosis Underscores the Role of Autoxidation in the Mechanism of Cell Death. ACS Chem Biol. 2017;12(10):2538–2545.
  • Griesser M, Shah R, Van Kessel AT, et al. The catalytic reaction of nitroxides with peroxyl radicals and its relevance to their cytoprotective properties. J Am Chem Soc. 2018;140(10):3798–3808.
  • Krainz T, Gaschler MM, Lim C, et al. A mitochondrial-targeted nitroxide is a potent inhibitor of ferroptosis. ACS Cent Sci. 2016;2(9):653–659.
  • Shah R, Shchepinov MS, Pratt DA. Resolving the role of lipoxygenases in the initiation and execution of ferroptosis. ACS Cent Sci. 2018;4(3):387–396.
  • Skouta R, Dixon SJ, Wang J, et al. Ferrostatins inhibit oxidative lipid damage and cell death in diverse disease models. J Am Chem Soc. 2014;136(12):4551–4556.
  • Wu Z, Geng Y, Lu X, et al. Chaperone-mediated autophagy is involved in the execution of ferroptosis. Proc Natl Acad Sci U S A. 2019;116(8):2996–3005.
  • Muller T, Dewitz C, Schmitz J, et al. Necroptosis and ferroptosis are alternative cell death pathways that operate in acute kidney failure. Cell Mol Life Sci. 2017;74(19):3631–3645.
  • Zhu S, Zhang Q, Sun X, et al. HSPA5 regulates ferroptotic cell death in cancer cells. Cancer Res. 2017;77(8):2064–2077.
  • Wang D, Peng Y, Xie Y, et al. Antiferroptotic activity of non-oxidative dopamine. Biochem Biophys Res Commun. 2016;480(4):602–607.
  • Ishii T, Bannai S, Sugita Y. Mechanism of growth stimulation of L1210 cells by 2-mercaptoethanol in vitro. Role of the mixed disulfide of 2-mercaptoethanol and cysteine. J Biol Chem. 1981;256(23):12387–12392.
  • Dai C, Chen X, Li J, et al. Transcription factors in ferroptotic cell death. Cancer Gene Ther. 2020. DOI:https://doi.org/10.1038/s41417-020-0170-2
  • Kang R, Kroemer G, Tang D. The tumor suppressor protein p53 and the ferroptosis network. Free Radic Biol Med. 2019;133:162–168.
  • Jiang L, Kon N, Li T, et al. Ferroptosis as a p53-mediated activity during tumour suppression. Nature. 2015;520(7545):57–62.
  • Bensaad K, Tsuruta A, Selak MA, et al. TIGAR, a p53-inducible regulator of glycolysis and apoptosis. Cell. 2006;126(1):107–120.
  • Hu W, Zhang C, Wu R, et al. Glutaminase 2, a novel p53 target gene regulating energy metabolism and antioxidant function. Proc Natl Acad Sci U S A. 2010;107(16):7455–7460.
  • Tarangelo A, Magtanong L, Bieging-Rolett KT, et al. p53 suppresses metabolic stress-induced ferroptosis in cancer cells. Cell Rep. 2018;22(3):569–575.
  • Ou Y, Wang SJ, Li D, et al. Activation of SAT1 engages polyamine metabolism with p53-mediated ferroptotic responses. Proc Natl Acad Sci U S A. 2016;113(44):E6806–E12.
  • 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 U S A. 2019;116(17):8390–8396.
  • Sun X, Niu X, Chen R, et al. Metallothionein-1G facilitates sorafenib resistance through inhibition of ferroptosis. Hepatology. 2016;64(2):488–500.
  • Shin D, Kim EH, Lee J, et al. Nrf2 inhibition reverses resistance to GPX4 inhibitor-induced ferroptosis in head and neck cancer. Free Radic Biol Med. 2018;129:454–462.
  • Roh JL, Kim EH, Jang H, et al. Nrf2 inhibition reverses the resistance of cisplatin-resistant head and neck cancer cells to artesunate-induced ferroptosis. Redox Biol. 2017;11:254–262.
  • Liu N, Lin X, Huang C. Activation of the reverse transsulfuration pathway through NRF2/CBS confers erastin-induced ferroptosis resistance. Br J Cancer. 2020;122(2):279–292.
  • Fan Z, Wirth AK, Chen D, et al. Nrf2-Keap1 pathway promotes cell proliferation and diminishes ferroptosis. Oncogenesis. 2017;6(8):e371.
  • Chen D, Tavana O, Chu B, et al. NRF2 Is a major target of ARF in p53-independent tumor suppression. Mol Cell. 2017;68(1):224–32 e4.
  • Anandhan A, Dodson M, Schmidlin CJ, et al. Breakdown of an ironclad defense system: the critical role of NRF2 in mediating ferroptosis. Cell Chem Biol. 2020;27(4):436–447.
  • Wu J, Minikes AM, Gao M, et al. Intercellular interaction dictates cancer cell ferroptosis via NF2-YAP signalling. Nature. 2019;572(7769):402–406.
  • Wenz C, Faust D, Linz B, et al. Cell-cell contacts protect against t-BuOOH-induced cellular damage and ferroptosis in vitro. Arch Toxicol. 2019;93(5):1265–1279.
  • Mao C, Wang X, Liu Y, et al. A G3BP1-interacting lncRNA promotes ferroptosis and apoptosis in cancer via nuclear sequestration of p53. Cancer Res. 2018;78(13):3484–3496.
  • Zhang Y, Shi J, Liu X, et al. BAP1 links metabolic regulation of ferroptosis to tumour suppression. Nat Cell Biol. 2018;20(10):1181–1192.
  • Wang Y, Yang L, Zhang X, et al. Epigenetic regulation of ferroptosis by H2B monoubiquitination and p53. EMBO Rep. 2019;20(7):e47563.
  • Wang Y, Zhao Y, Wang H, et al. Histone demethylase KDM3B protects against ferroptosis by upregulating SLC7A11. FEBS Open Bio. 2020;10(4):637–643.
  • Sun Z, Chin YE, Zhang DD. Acetylation of Nrf2 by p300/CBP augments promoter-specific DNA binding of Nrf2 during the antioxidant response. Mol Cell Biol. 2009;29(10):2658–2672.
  • Zhang Y, Xiong Y, Yarbrough WG. ARF promotes MDM2 degradation and stabilizes p53: ARF-INK4a locus deletion impairs both the Rb and p53 tumor suppression pathways. Cell. 1998;92(6):725–734.
  • Klionsky DJ. Autophagy as a regulated pathway of cellular degradation. Science. 2000;290(5497):1717–1721.
  • Levine B, Kroemer G. Biological functions of autophagy genes: a disease perspective. Cell. 2019;176(1–2):11–42.
  • Xie Y, Kang R, Sun X, et al. Posttranslational modification of autophagy-related proteins in macroautophagy. Autophagy. 2015;11(1):28–45.
  • Xie Y, Li J, Kang R, et al. Interplay between lipid metabolism and autophagy. Front Cell Dev Biol. 2020;8:431.
  • Liu Y, Wang Y, Liu J, et al. Interplay between MTOR and GPX4 signaling modulates autophagy-dependent ferroptotic cancer cell death. Cancer Gene Ther. 2020. DOI:https://doi.org/10.1038/s41417-020-0182-y
  • Xie Y, Kuang F, Liu J, et al. DUSP1 blocks autophagy-dependent ferroptosis in pancreatic cancer. J Pancreatol. 2020;Publish Ahead of Print. DOI:https://doi.org/10.1097/JP9.0000000000000054
  • Bellelli R, Federico G, Matte A, et al. NCOA4 deficiency impairs systemic iron homeostasis. Cell Rep. 2016;14(3):411–421.
  • Park E, Chung SW. ROS-mediated autophagy increases intracellular iron levels and ferroptosis by ferritin and transferrin receptor regulation. Cell Death Dis. 2019;10(11):822.
  • Singh R, Kaushik S, Wang Y, et al. Autophagy regulates lipid metabolism. Nature. 2009;458(7242):1131–1135.
  • Yang M, Chen P, Liu J, et al. Clockophagy is a novel selective autophagy process favoring ferroptosis. Sci Adv. 2019;5(7):eaaw2238.
  • Liu J, Yang M, Kang R, et al. Autophagic degradation of the circadian clock regulator promotes ferroptosis. Autophagy. 2019;15(11):2033–2035.
  • Liu Y, Wang Y, Liu J, et al. The circadian clock protects against ferroptosis-induced sterile inflammation. Biochem Biophys Res Commun. 2020;525(3):620–625.
  • NaveenKumar SK, Hemshekhar M, Kemparaju K, et al. Hemin-induced platelet activation and ferroptosis is mediated through ROS-driven proteasomal activity and inflammasome activation: protection by Melatonin. Biochim Biophys Acta Mol Basis Dis. 2019;1865(9):2303–2316.
  • Dice JF. Chaperone-mediated autophagy. Autophagy. 2007;3(4):295–299.
  • Liu T, Jiang L, Tavana O, et al. The Deubiquitylase OTUB1 Mediates Ferroptosis via Stabilization of SLC7A11. Cancer Res. 2019;79(8):1913–1924.
  • Kan CF, Singh AB, Stafforini DM, et al. Arachidonic acid downregulates acyl-CoA synthetase 4 expression by promoting its ubiquitination and proteasomal degradation. J Lipid Res. 2014;55(8):1657–1667.
  • Yagoda N, von Rechenberg M, Zaganjor E, et al. RAS-RAF-MEK-dependent oxidative cell death involving voltage-dependent anion channels. Nature. 2007;447(7146):864–868.
  • Yang Y, Luo M, Zhang K, et al. Nedd4 ubiquitylates VDAC2/3 to suppress erastin-induced ferroptosis in melanoma. Nat Commun. 2020;11(1):433.
  • Wang Y, Liua Y, Liua J, et al. NEDD4L-mediated LTF protein degradation limits ferroptosis. Biochem Biophys Res Commun. 2020. DOI:https://doi.org/10.1016/j.bbrc.2020.07.032
  • Hangauer MJ, Viswanathan VS, Ryan MJ, et al. Drug-tolerant persister cancer cells are vulnerable to GPX4 inhibition. Nature. 2017;551(7679):247–250.
  • Tang D, Kang R, Coyne CB, et al. PAMPs and DAMPs: signal 0s that spur autophagy and immunity. Immunol Rev. 2012;249(1):158–175.
  • Wen Q, Liu J, Kang R, et al. The release and activity of HMGB1 in ferroptosis. Biochem Biophys Res Commun. 2019;510(2):278–283.
  • Yu Y, Xie Y, Cao L, et al. The ferroptosis inducer erastin enhances sensitivity of acute myeloid leukemia cells to chemotherapeutic agents. Mol Cell Oncol. 2015;2(4):e1054549.
  • Friedmann Angeli JP, Krysko DV, Conrad M. Ferroptosis at the crossroads of cancer-acquired drug resistance and immune evasion. Nat Rev Cancer. 2019;19(7):405–414.
  • Zhang L, Liu W, Liu F, et al. IMCA Induces Ferroptosis Mediated by SLC7A11 through the AMPK/mTOR Pathway in Colorectal Cancer. Oxid Med Cell Longev. 2020;2020:1675613.
  • Lorincz T, Jemnitz K, Kardon T, et al. Ferroptosis is involved in acetaminophen induced cell death. Pathol Oncol Res. 2015;21(4):1115–1121.
  • Woo JH, Shimoni Y, Yang WS, et al. Elucidating compound mechanism of action by network perturbation analysis. Cell. 2015;162(2):441–451.
  • Meng P, Zhang S, Jiang X, et al. Arsenite induces testicular oxidative stress in vivo and in vitro leading to ferroptosis. Ecotoxicol Environ Saf. 2020;194:110360.
  • Ooko E, Saeed ME, Kadioglu O, et al. Artemisinin derivatives induce iron-dependent cell death (ferroptosis) in tumor cells. Phytomedicine. 2015;22(11):1045–1054.
  • Eling N, Reuter L, Hazin J, et al. Identification of artesunate as a specific activator of ferroptosis in pancreatic cancer cells. Oncoscience. 2015;2(5):517–532.
  • Chang LC, Chiang SK, Chen SE, et al. Heme oxygenase-1 mediates BAY 11-7085 induced ferroptosis. Cancer Lett. 2018;416:124–137.
  • Basit F, van Oppen LM, Schockel L, et al. Mitochondrial complex I inhibition triggers a mitophagy-dependent ROS increase leading to necroptosis and ferroptosis in melanoma cells. Cell Death Dis. 2017;8(3):e2716.
  • Alborzinia H, Ignashkova TI, Dejure FR, et al. Golgi stress mediates redox imbalance and ferroptosis in human cells. Commun Biol. 2018;1(1):210.
  • Deng G, Li Y, Ma S, et al. Caveolin-1 dictates ferroptosis in the execution of acute immune-mediated hepatic damage by attenuating nitrogen stress. Free Radic Biol Med. 2020;148:151–161.
  • Zeng T, Deng G, Zhong W, et al. Indoleamine 2, 3-dioxygenase 1enhanceshepatocytes ferroptosis in acute immune hepatitis associated with excess nitrative stress. Free Radic Biol Med. 2020;152:668–679.
  • Chen GQ, Benthani FA, Wu J, et al. Artemisinin compounds sensitize cancer cells to ferroptosis by regulating iron homeostasis. Cell Death Differ. 2020;27(1):242–254.
  • Du J, Wang T, Li Y, et al. DHA inhibits proliferation and induces ferroptosis of leukemia cells through autophagy dependent degradation of ferritin. Free Radic Biol Med. 2019;131:356–369.
  • Fang X, Wang H, Han D, et al. Ferroptosis as a target for protection against cardiomyopathy. Proc Natl Acad Sci U S A. 2019;116(7):2672–2680.
  • Chen P, Wu Q, Feng J, et al. Erianin, a novel dibenzyl compound in Dendrobium extract, inhibits lung cancer cell growth and migration via calcium/calmodulin-dependent ferroptosis. Signal Transduct Target Ther. 2020;5(1):51.
  • Fang S, Yu X, Ding H, et al. Effects of intracellular iron overload on cell death and identification of potent cell death inhibitors. Biochem Biophys Res Commun. 2018;503(1):297–303.
  • Wu J, Yang JJ, Cao Y, et al. Iron overload contributes to general anaesthesia-induced neurotoxicity and cognitive deficits. J Neuroinflammation. 2020;17(1):110.
  • Jiang T, Chu J, Chen H, et al. Gastrodin inhibits H2O2-induced ferroptosis through its antioxidative effect in rat glioma cell line C6. Biol Pharm Bull. 2020;43(3):480–487.
  • Wang W, Green M, Choi JE, et al. CD8+ T cells regulate tumour ferroptosis during cancer immunotherapy. Nature. 2019;569(7755):270–274.
  • Lang X, Green MD, Wang W, et al. Radiotherapy and Immunotherapy Promote Tumoral Lipid Oxidation and Ferroptosis via Synergistic Repression of SLC7A11. Cancer Discov. 2019;9(12):1673–1685.
  • Lei G, Zhang Y, Koppula P, et al. The role of ferroptosis in ionizing radiation-induced cell death and tumor suppression. Cell Res. 2020;30(2):146–162.
  • Ye LF, Chaudhary KR, Zandkarimi F, et al. Radiation-induced lipid peroxidation triggers ferroptosis and synergizes with ferroptosis inducers. ACS Chem Biol. 2020;15(2):469–484.
  • Liang C, Zhang X, Yang M, et al. Recent progress in ferroptosis inducers for cancer therapy. Adv Mater. 2019;31(51):e1904197.
  • Shen Z, Song J, Yung BC, et al. Emerging strategies of cancer therapy based on ferroptosis. Adv Mater. 2018;30(12):e1704007.
  • Yamaguchi Y, Kasukabe T, Kumakura S. Piperlongumine rapidly induces the death of human pancreatic cancer cells mainly through the induction of ferroptosis. Int J Oncol. 2018;52(3):1011–1022.
  • Wang Y, Tang M. PM2.5 induces ferroptosis in human endothelial cells through iron overload and redox imbalance. Environ Pollut. 2019;254:112937.
  • Wang Z, Ding Y, Wang X, et al. Pseudolaric acid B triggers ferroptosis in glioma cells via activation of Nox4 and inhibition of xCT. Cancer Lett. 2018;428:21–33.
  • Wu C, Zhao W, Yu J, et al. Induction of ferroptosis and mitochondrial dysfunction by oxidative stress in PC12 cells. Sci Rep. 2018;8(1):574.
  • Palmer LD, Jordan AT, Maloney KN, et al. Zinc intoxication induces ferroptosis in A549 human lung cells. Metallomics. 2019;11(5):982–993.
  • Torii S, Shintoku R, Kubota C, et al. An essential role for functional lysosomes in ferroptosis of cancer cells. Biochem J. 2016;473(6):769–777.
  • Gao H, Bai Y, Jia Y, et al. Ferroptosis is a lysosomal cell death process. Biochem Biophys Res Commun. 2018;503(3):1550–1556.
  • Xie Y, Song X, Sun X, et al. Identification of baicalein as a ferroptosis inhibitor by natural product library screening. Biochem Biophys Res Commun. 2016;473(4):775–780.
  • Neitemeier S, Jelinek A, Laino V, et al. BID links ferroptosis to mitochondrial cell death pathways. Redox Biol. 2017;12:558–570.
  • Jelinek A, Heyder L, Daude M, et al. Mitochondrial rescue prevents glutathione peroxidase-dependent ferroptosis. Free Radic Biol Med. 2018;117:45–57.
  • Mishima E, Sato E, Ito J, et al. Drugs repurposed as antiferroptosis agents suppress organ damage, including AKI, by functioning as lipid peroxyl radical scavengers. J Am Soc Nephrol. 2020;31(2):280–296.
  • Do Van B, Gouel F, Jonneaux A, et al. Ferroptosis, a newly characterized form of cell death in Parkinson’s disease that is regulated by PKC. Neurobiol Dis. 2016;94:169–178.
  • Li X, Zou Y, Xing J, et al. Pretreatment with roxadustat (FG-4592) attenuates folic acid-induced kidney injury through antiferroptosis via Akt/GSK-3beta/Nrf2 pathway. Oxid Med Cell Longev. 2020;2020:6286984.
  • Liu B, Zhao C, Li H, et al. Puerarin protects against heart failure induced by pressure overload through mitigation of ferroptosis. Biochem Biophys Res Commun. 2018;497(1):233–240.
  • Wang L, Liu Y, Du T, et al. ATF3 promotes erastin-induced ferroptosis by suppressing system Xc. Cell Death Differ. 2020;27(2):662–675.
  • Chen Y, Mi Y, Zhang X, et al. Dihydroartemisinin-induced unfolded protein response feedback attenuates ferroptosis via PERK/ATF4/HSPA5 pathway in glioma cells. J Exp Clin Cancer Res. 2019;38(1):402.
  • Chen D, Fan Z, Rauh M, et al. ATF4 promotes angiogenesis and neuronal cell death and confers ferroptosis in a xCT-dependent manner. Oncogene. 2017;36(40):5593–5608.
  • Wang N, Zeng GZ, Yin JL, et al. Artesunate activates the ATF4-CHOP-CHAC1 pathway and affects ferroptosis in Burkitt’s Lymphoma. Biochem Biophys Res Commun. 2019;519(3):533–539.
  • Chen MS, Wang SF, Hsu CY, 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(70):114588–114602.
  • Nishizawa H, Matsumoto M, Shindo T, 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(1):69–82.
  • Wang GX, Tu HC, Dong Y, et al. DeltaNp63 inhibits oxidative stress-induced cell death, including ferroptosis, and cooperates with the BCL-2 family to promote clonogenic survival. Cell Rep. 2017;21(10):2926–2939.
  • Zhang X, Du L, Qiao Y, et al. Ferroptosis is governed by differential regulation of transcription in liver cancer. Redox Biol. 2019;24:101211.
  • Zou Y, Palte MJ, Deik AA, et al. A GPX4-dependent cancer cell state underlies the clear-cell morphology and confers sensitivity to ferroptosis. Nat Commun. 2019;10(1):1617.
  • Sun X, Ou Z, Xie M, et al. HSPB1 as a novel regulator of ferroptotic cancer cell death. Oncogene. 2015;34(45):5617–5625.
  • Chen Y, Zhu G, Liu Y, et al. O-GlcNAcylated c-Jun antagonizes ferroptosis via inhibiting GSH synthesis in liver cancer. Cell Signal. 2019;63:109384.
  • Chen PH, Wu J, Ding CC, et al. Kinome screen of ferroptosis reveals a novel role of ATM in regulating iron metabolism. Cell Death Differ. 2020;27(3):1008–1022.
  • Cao JY, Poddar A, Magtanong L, et al. A genome-wide haploid genetic screen identifies regulators of glutathione abundance and ferroptosis sensitivity. Cell Rep. 2019;26(6):1544–56 e8.
  • Gagliardi M, Cotella D, Santoro C, et al. Aldo-keto reductases protect metastatic melanoma from ER stress-independent ferroptosis. Cell Death Dis. 2019;10(12):902.
  • Park SJ, Cho SS, Kim KM, et al. Protective effect of sestrin2 against iron overload and ferroptosis-induced liver injury. Toxicol Appl Pharmacol. 2019;379:114665.
  • Li Y, Feng D, Wang Z, et al. Ischemia-induced ACSL4 activation contributes to ferroptosis-mediated tissue injury in intestinal ischemia/reperfusion. Cell Death Differ. 2019;26(11):2284–2299.
  • Brown CW, Amante JJ, Goel HL, et al. The alpha6beta4 integrin promotes resistance to ferroptosis. J Cell Biol. 2017;216(12):4287–4297.
  • Li L, Sun S, Tan L, 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(11):7781–7792.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.