Abstract
Ferroptosis is a non-apoptotic cell death mechanism characterized by iron-dependent membrane lipid peroxidation. Here, we review what is known about the cellular mechanisms mediating the execution and regulation of ferroptosis. We first consider how the accumulation of membrane lipid peroxides leads to the execution of ferroptosis by altering ion transport across the plasma membrane. We then discuss how metabolites and enzymes that are distributed in different compartments and organelles throughout the cell can regulate sensitivity to ferroptosis by impinging upon iron, lipid and redox metabolism. Indeed, metabolic pathways that reside in the mitochondria, endoplasmic reticulum, lipid droplets, peroxisomes and other organelles all contribute to the regulation of ferroptosis sensitivity. We note how the regulation of ferroptosis sensitivity by these different organelles and pathways seems to vary between different cells and death-inducing conditions. We also highlight transcriptional master regulators that integrate the functions of different pathways and organelles to modulate ferroptosis sensitivity globally. Throughout this Review, we highlight open questions and areas in which progress is needed to better understand the cell biology of ferroptosis.
Introduction
Ferroptosis is a non-apoptotic cell death mechanism that requires the redox-active metal iron1,2. Free intracellular iron or iron-containing enzymes react with oxygen and polyunsaturated fatty acid (PUFA)-containing lipids to generate high levels of membrane lipid peroxides — the defining feature of ferroptosis in relation to other forms of cell death3 (Box 1). These membrane lipid peroxides can be lethal to the cell when they accumulate at high levels. Any cell in the biosphere containing iron, oxygen and PUFA-containing lipids may therefore be at risk of undergoing ferroptosis. For this reason, cells have evolved powerful enzyme-catalysed mechanisms to defend against oxidative membrane damage and the onset of ferroptosis4,5 (Fig. 1a). Ferroptosis is a physiological process with a role in homeostasis, namely tumour suppression6,7. Ferroptosis may also be activated in acute and chronic disease conditions8. Accordingly, there is considerable interest in understanding the nature and regulation of this mechanism.
Box 1. How is ferroptosis defined?
There is ongoing debate about whether ferroptosis represents an evolved form of cell death, like apoptosis, or whether it is best understood as a form of ‘cell sabotage’ that is a manifestation of aberrant interactions between different intracellular metabolic networks that only occur under unusual circumstances associated with disease or non-physiological stimuli11. Regardless, ferroptosis is defined as a regulated form of cell death because it is possible to modulate (that is, enhance or suppress) sensitivity by changing the function of specific proteins and pathways in the cell90,202. Note that oxidative glutamate toxicity and oxytosis are likely different names used in the past to describe the same lethal mechanism in certain cells203–205. Mechanistically, high levels of membrane lipid peroxidation are observed in ferroptosis but not other forms of cell death206. This is not a mere correlate of cell death but rather a necessary feature of the ferroptosis mechanism. In fact, ferroptosis can be defined operationally as cell death that is blocked by inhibitors of membrane lipid peroxidation, especially lipophilic radical-trapping antioxidants such as ferrostatin 1 or liproxstatin 1 (refs. 36,207). In practice, lipid peroxide accumulation can be detected in live cells and some tissues using chemical probes such as C11-BODIPY 581/591 (refs. 28,97), Liperfluo30 and STY-BODIPY97,208. Oxidized phospholipid species can also be detected using mass spectrometry methods21,30. It may be hard or impossible to detect lipid peroxides or oxidized phospholipids in fixed tissues using chemical probes. Breakdown products of lipid peroxidation, such as 4-hydroxynonenal, can be detected in fixed tissue sections43. However, since lipid peroxidation and 4-hydroxynonenal accumulation are not entirely specific to ferroptosis, identifying additional markers of this process that can be used to monitor ferroptosis in vivo remains a priority187,191,201,209.
Fig. 1 |. The general mechanism of ferroptosis.

a, Ferroptosis execution can be distinguished from the regulation of ferroptosis sensitivity. Ferroptosis execution involves membrane lipid peroxidation, leading to plasma membrane rupture and cell death. The glutathione (GSH)–glutathione peroxidase 4 (GPX4) and NADPH–ferroptosis suppressor protein 1 (FSP1) systems limit membrane lipid peroxidation and inhibit ferroptosis. Ferroptosis sensitivity is dictated by the propensity of the cell to accumulate high levels of membrane lipid peroxides. Whether a cell will accumulate high levels of lipid peroxides relates to the amount of iron and oxidizable lipids in the cell, and to the status of the cellular oxidant-producing and antioxidant systems. The regulation of ferroptosis sensitivity likely encompasses hundreds of distinct metabolites, enzymes and other biomolecules that impinge upon the membrane lipid peroxidation and membrane-rupturing mechanisms. b, Plasma membrane rupture is the key event that results in cell death during ferroptosis. It involves the accumulation of phospholipid hydroperoxides, which may be generated enzymatically (for example, via lipoxygenases) and non-enzymatically through radical-mediated reactions. Phospholipid peroxidation has several consequences for the cell, including altered ion fluxes (for example, increased Piezo-mediated Ca2+ uptake, increased transient receptor potential (TRP)-mediated Ca2+ and Na+ uptake, and reduced Na+/K+ ATPase-mediated Na+ export and K+ uptake), water ingress and biophysical effects on the membrane. Cell swelling and increased membrane stiffness can further activate ion channels (for example, Piezo and TRP) in a feedforward manner, further enhancing ion fluxes and accelerating plasma membrane rupture. c, Important ferroptosis defence mechanisms localize to the plasma membrane. The system xc− antiporter imports cystine in exchange for glutamate. In the cytosol, cystine is rapidly reduced to cysteine. Cysteine can be used to synthesize (reduced) GSH. GSH is the cofactor for GPX4, an enzyme that can convert toxic phospholipid peroxides (L-OOH) into benign lipid alcohols (L-OH). Parallel to GPX4, the oxidoreductase FSP1 uses NAD(P)H to regenerate the reduced form of radical-trapping antioxidants (for example, coenzyme Q10 (CoQ) or vitamin K), which in turn terminate the lipid peroxidation process by donating electrons to phospholipid peroxyl radicals (LOO•). Receptor-mediated endocytosis influences ferroptosis sensitivity. For example, the uptake of iron in complex with transferrin by the transferrin receptor enhances ferroptosis sensitivity while uptake of the selenium-rich protein SEPP1 by its cognate receptor LDL receptor-related protein 8 (LRP8) suppresses ferroptosis sensitivity. Iron, and presumably selenium in the form of selenocysteine, are subsequently released from the lysosome. Iron can react with soluble and lipid peroxides to generate hydroxyl and lipid alkoxyl radicals that promote lipid peroxidation. Cysteine and selenium released from the lysosome can be used to synthesize GSH and the selenoprotein GPX4, respectively. ACSL4, acyl-CoA synthetase long-chain family member 4; CHMP, charged multivesicular body protein; CoA, coenzyme A; GR, GSH reductase; GSSG, oxidized GSH; PKCβII, protein kinase Cβ; PUFA, polyunsaturated fatty acid.
We can distinguish the execution of ferroptosis from mechanisms that regulate ferroptosis sensitivity. The execution of ferroptosis involves membrane lipid peroxidation, the aberrant movement of ions across the plasma membrane, cell swelling and plasma membrane rupture9. Cellular sensitivity to ferroptosis — the likelihood that a given stimulus will cause plasma membrane rupture through the ferroptosis mechanism — is regulated positively and negatively by molecules and pathways that control lipid metabolism, iron homeostasis, redox regulation and related processes. Accordingly, sensitivity to ferroptosis is governed by hundreds of enzymes, reactions and molecules in the cell10. These species are distributed throughout the plasma membrane and cytosol, as well as in different organelles. Depending on the cell type and the ferroptosis-inducing condition, different metabolites, enzymes and organelles appear more important than others for ferroptosis execution and the regulation of ferroptosis sensitivity10. Understanding this context-specific regulation of ferroptosis sensitivity presents an interesting challenge.
In this Review, we examine ferroptosis mechanisms at the cellular level. We first describe the events that lead to lethal plasma membrane rupture. We then discuss how different enzymes and molecules regulate ferroptosis sensitivity from different locations in the cell. Finally, we point out examples of how these processes may be integrated between different regions of the cell to execute or prevent ferroptosis. Throughout this Review, we highlight unanswered questions that exist in connection with the cell biology of ferroptosis.
The execution and regulation of ferroptosis at the plasma membrane
Plasma membrane rupture is the terminal event in many forms of cell death. For some forms of non-apoptotic cell death, such as necroptosis and pyroptosis, this terminal process involves permeabilization of the plasma membrane by pore-forming proteins such as mixed lineage kinase domain-like pseudokinase (MLKL), gasdermin D and ninjurin 1 (refs. 11,12). Ninjurin 1 may facilitate the execution of ferroptosis in some but not all cells9,13. Otherwise, there is little evidence that the execution of ferroptosis requires a specific pore-forming protein. Rather, ferroptosis execution and regulation involve a distinct set of lipid-centric mechanisms that govern plasma membrane integrity.
Ferroptosis execution at the plasma membrane
The execution of ferroptosis involves the peroxidation of PUFA-containing phospholipids and the formation of lipid hydroperoxides14,15. Once formed, lipid hydroperoxides can react with iron to generate highly reactive lipid radicals and become further modified or truncated in ways that likely contribute to the execution of ferroptosis16. Lipid hydroperoxides may be synthesized by lipoxygenase enzymes17 or formed through chemical reactions between PUFAs, soluble reactive oxygen species (ROS) and iron10. It is controversial which mechanism is most important for ferroptosis and it seems likely that both mechanisms are relevant in one cell type or another17–20. Within the membrane, dozens of different PUFA-containing phospholipid species can be peroxidized to contribute to the execution of ferroptosis, with PUFA-phosphatidylethanolamines being especially important in many cells14,21–26. When cells are exposed to ferroptosis-inducing conditions, lipid peroxides seem to initially accumulate within the endoplasmic reticulum (ER) and other organelles and subsequently at the plasma membrane prior to membrane rupture27–30 (Box 2).
Box 2. Small-molecule probes illuminate the cell biology of ferroptosis.
Ferroptosis was first recognized by studying the effects of lethal small molecules that could induce cancer cell death. Subsequent work then identified potent and specific small-molecule inhibitors of ferroptosis that helped further reveal the nature of this mechanism. The history of ferroptosis is closely entwined with this series of chemical biology screens and analyses. Moreover, these chemicals have provided starting points for the development of drug candidates that may be used to activate or inhibit ferroptosis to treat disease.
Inducing ferroptosis
Erastin, sulfasalazine and other agents can trigger ferroptosis by inhibiting the system xc− cystine–glutamate antiporter at the plasma membrane58. Glutathione peroxidase 4 (GPX4) can be inhibited by RSL3, ML162, ML210 and related covalent binders210. These small molecules presumably inhibit all forms of GPX4 (cytosolic, mitochondrial, nuclear). Inactivation of cytosolic GPX4 is likely responsible for ferroptosis in most cases, although mitochondrial GPX4 may be a relevant target in some contexts37–39. iFSP1, FSEN1 and related molecules inhibit ferroptosis suppressor protein 1 (FSP1)42,211, which blocks ferroptosis from the inner leaflet of the plasma membrane. FSP1 can also be indirectly inhibited by icFSP1, which triggers the formation of intracellular phase-separated condensates of FSP1 (ref. 212). The earliest signs of membrane lipid peroxidation in response to system xc− inhibitors and GPX4 inhibitors are observed on internal membranes, especially the endoplasmic reticulum (ER), before lipid peroxidation is observed at the plasma membrane27,29,30. One interpretation of these results is that the loss of Cys-containing metabolites and/or GPX4 inhibition occurs in some form of gradient within the cytosol, with the ER or regions nearer to the ER being affected sooner than regions closer to the plasma membrane. An alternative model could be that iron and more oxidizable phospholipids are present in greater abundance in or near the ER, thereby favouring initiation at this location. However, it seems that ferroptosis can be initiated at any of several different sites and still eventually lead to lethal lipid peroxidation at the plasma membrane. Analogues of the pro-ferroptotic molecule FINO2 that selectively localize to different organelles — the ER, lysosomes and mitochondria — appear sufficient to initiate ferroptosis29. It also remains possible that there is no requirement for direct communication between these compartments for the induction of ferroptosis, and that peroxidation of lipids at the plasma membrane is an independent process that simply takes longer.
Inhibiting ferroptosis
Ferroptotic plasma membrane rupture can be delayed but not fully inhibited by polyethylene glycol molecules and by non-specific inhibitors of plasma membrane-localized ion channels (for example, ruthenium red)9,31,32. The membrane lipid peroxidation process that drives ferroptosis can be terminated by radical-trapping antioxidants and iron chelators. The potency of radical-trapping antioxidants against ferroptosis correlates with the lipophilicity of these molecules90,213, consistent with membranes being the crucial site of lipid peroxidation that leads to ferroptosis. Imaging studies using the chemical probe C11-BODIPY 581/591 suggest that ferrostatin 1 can block lipid peroxidation throughout the cell28. Detailed imaging studies using diyne-tagged versions of ferrostatin 1 together with stimulated Raman scattering microscopy show that this molecule inhibits ferroptosis when it accumulates in the ER but not when it accumulates in lysosomes or mitochondria81. (In fact, forced lysosomal accumulation reduces ferrostatin potency.) Other evidence indicates that mitochondrially targeted inhibitors (for example, mitoquinone or mitoTEMPO) can suppress ferroptosis214–216, but whether these molecules act in the mitochondria when applied to cells or actually block lipid peroxidation at another site within the cell due to a ‘spillover’ effect is an unresolved question36. In connection with iron, some chelators are freely membrane permeable and may bind iron at many sites within the cell. Other chelators may be more specifically localized. For example, deferoxamine potently inhibits ferroptosis1 despite having low cell membrane permeability. Deferoxamine is taken into cells by endocytosis and binds iron in the endosomal and lysosomal compartments217,218. This localization would bring deferoxamine into contact with lysosomal iron pools and may therefore suggest that the lysosomal iron pool is essential for the initiation or propagation of ferroptosis. Indeed, direct inhibition of ferritin catabolism by silencing the adaptor protein required for lysosomal-mediated ferritinophagy (nuclear receptor coactivator 4 (NCOA4)) suppresses ferroptosis219. In relation to lipids, small-molecule inhibitors of acyl-CoA synthetase long-chain family member 4 (ACSL4) (for example, rosiglitazone)106,220,221 and lysophosphatidylcholine acyltransferase 3 (LPCAT3) (for example, (R)-HTS-3)115 — enzymes that localize primarily to the ER or endomembrane system — can effectively inhibit ferroptosis by reducing the levels of polyunsaturated fatty acids in one or more cellular compartments.
Events downstream of plasma membrane lipid peroxidation that promote ferroptosis execution have recently been clarified (Fig. 1b). Increasing lipid peroxidation raises membrane tension, which in turn activates Piezo1 and transient receptor potential (TRP) mechanosensitive ion channels9. Channel opening leads to Ca2+ and Na+ influx and K+ efflux. These ion fluxes are enhanced by the simultaneous inactivation of the plasma membrane Na+/K+ ATPase, possibly due to altered interactions between this protein and peroxidized plasma membrane phospholipids9. Collectively, these changes lead to a loss of ionic homeostasis and osmotic cell swelling that eventually result in plasma membrane rupture. Plasma membrane rupture can be transiently delayed by polyethylene glycol molecules that block membrane nanopores to prevent the flow of ions and water across the plasma membrane31,32. However, polyethylene glycol molecules only delay the onset of ferroptosis and do not completely prevent plasma membrane rupture over longer periods of time31. It seems likely that, as more lipids become peroxidized, this disrupts the biophysical properties of the plasma membrane33 so severely that plasma membrane rupture becomes inevitable.
Suppression of membrane lipid peroxidation
Ferroptosis is normally inhibited by the continuous activity of coupled enzyme-metabolite systems that prevent the accumulation of membrane lipid peroxides to toxic levels. These systems are, in fact, the targets of small molecules that helped elucidate the ferroptosis mechanism in the first place (see Box 2 for a description of common ferroptosis inducers and inhibitors). The enzyme glutathione peroxidase 4 (GPX4) reduces potentially toxic lipid hydroperoxides to less dangerous lipid alcohols34,35. GPX4 appears to be the most critical enzyme for preventing lipid hydroperoxide accumulation in most cells35,36 (Fig. 1c). Several isoforms of GPX4 exist, including those designated as cytosolic, mitochondrial and nuclear. Cytosolic GPX4 seems the most essential for preventing ferroptosis, although mitochondrial GPX4 may also help inhibit ferroptosis in some cases37–39. Cytosolic GPX4 does not contain an obvious plasma membrane-targeting element. However, modelling studies indicate that a patch of positively charged lysine and arginine residues may facilitate the electrostatic interactions with phospholipids that help keep GPX4 in the vicinity of the plasma membrane40.
Ferroptosis suppressor protein 1 (FSP1; also known as AIFM2) inhibits plasma membrane lipid peroxidation in parallel to GPX4 (Fig. 1c). FSP1 is an NAD(P)H- and FAD-dependent oxidoreductase that reduces coenzyme Q10 (CoQ)41,42 and vitamin K43. The reduced forms of CoQ and vitamin K act as lipophilic radical-trapping antioxidants (RTAs) that can terminate lipid peroxidation chain reactions. FSP1 localizes specifically to the plasma membrane and to lipid droplets41,42. During translation, the N-terminal initiator methionine of FSP1 is removed and the 14-carbon fatty acid myristate is covalently attached to the subsequent glycine residue41,42. The irreversibly conjugated myristate anchors FSP1 to the plasma membrane and is necessary for its anti-ferroptotic activity41,42. Targeting FSP1 selectively to the plasma membrane is sufficient to suppress ferroptosis, implying that this is the key site where endogenous RTAs function to suppress ferroptosis41. Co-immunoprecipitation and biochemistry data support the possibility that human FSP1 exists as a dimer44, but whether this is the functional state of FSP1 at the membrane is unknown. In addition to CoQ and vitamin K, other endogenous metabolites can serve as RTA inhibitors of ferroptosis, including tetrahydrobiopterin23,45, vitamin E and vitamin A46. The relative contribution of these different metabolic ferroptosis suppressors appears to vary between cell types.
Plasma membrane repair as an anti-ferroptotic mechanism
Plasma membrane repair processes oppose the terminal execution of ferroptosis. As noted above, lipid peroxidation can trigger an increase in intracellular Ca2+ levels31. Increased intracellular Ca2+ acts as a signal to recruit charged multivesicular body protein 5 (CHMP5) and CHMP6 — the components of the endosomal sorting complexes required for transport (ESCRT)-III — to the plasma membrane47, where they participate in local membrane repair. Indeed, genetic silencing of CHMP5, CHMP6 or CHMP4B can enhance ferroptosis sensitivity to some degree31,47 (Fig. 1b). However, membrane repair mediated by CHMP4B, CHMP5 and CHMP6 can be overwhelmed by high levels of lipid peroxidation31,47. Of note, the ESCRT-III complex can also limit protein pore-dependent membrane damage during necroptosis and pyroptosis48,49 and is therefore a negative regulator of several forms of non-apoptotic cell death.
Ferroptosis regulation by plasma membrane transporters and enzymes
Integral plasma membrane proteins can contribute to the execution and regulation of ferroptosis in many ways, including through the generation of ROS1,50,51, and to the mediation of iron or metabolite trafficking52–57. Here, we focus on proteins that are involved in the import of cysteine and selenium, two metabolites that contribute essentially to the negative regulation of ferroptosis (see also next section) (Fig. 1c).
Cysteine is required to synthesize sulfur-containing anti-ferroptotic molecules, including the GPX4 cofactor (reduced) glutathione (GSH)58. The cysteine disulfide, cystine, can be imported by the cell surface system xc− cystine–glutamate antiporter. Cysteine can also be acquired indirectly via endocytosis of extracellular cysteine-rich proteins, such as albumin, and their subsequent lysosomal catabolism59. Extracellular GSH can also directly fuel de novo intracellular GSH synthesis via the γ-glutamyl cycle. Plasma membrane-localized γ-glutamyltransferase 1 (GGT1) cleaves the usual γ-glutamyl bond that links glutamate to cysteine in the GSH tripeptide. The resulting cysteinylglycine dipeptide can then be imported and further catabolized by dipeptidases, including carnosine dipeptidase 2 (CNDP2), to release cysteine for use in GSH synthesis60. Some brain cancer cells overcome the need for system xc−-mediated cystine import via GGT1-mediated catabolism of extracellular GSH61. Moreover, in a rare kidney cancer (chromophobe renal cell carcinoma), the absence of GGT1 function renders these cells exquisitely dependent upon extracellular cystine for survival relative to normal tissues62. This helps illustrate how differences in protein expression between cells can result in unique sensitivities to pro-ferroptotic stimuli such as cystine deprivation.
Selenium is found within the active-site selenocysteine of GPX4 and a small number of other enzymes. In cancer cells, high levels of GPX4 translation promote ferroptosis resistance but, under conditions of low selenium, this GPX4 synthesis is inhibited by ribosome stalling and collisions, which causes early GPX4 translation termination (see also next section)63. Selenium can enter the cell by endocytosis of the liver-secreted, selenium-containing carrier protein selenoprotein P, which is bound and endocytosed by cell surface receptors such as the LDL receptor-related protein 8 (LRP8)63–65. In triple-negative breast cancer cells, LRP8 knockout sensitizes cells to ferroptosis inducers63. In MYCN-amplified neuroblastoma cells, loss of LRP8 alone is sufficient to trigger ferroptosis, likely due to coincident low expression of system xc− subunits65. Selenium can also enter the cell via the conversion of inorganic selenium (that is, selenite) to volatile selenide through a process that involves system xc−-dependent cystine uptake, which provides thiols that can reduce extracellular inorganic selenium to selenide, which is then presumed to diffuse across the plasma membrane66. How cystine and selenium uptake and metabolism are coordinated to ensure proper GPX4 function is an interesting open question.
Ferroptosis regulatory activities occurring in the cytosol
Ferroptosis execution centres on the plasma membrane. However, key ferroptosis regulatory activities occur in the cytosol. The translation of selenoproteins such as GPX4 occurs in the cytosol. Selenocysteine is synthesized directly on a dedicated tRNA (tRNA[Ser]Sec) in a multistep process. tRNA[Ser]Sec is first conjugated with serine, which is then phosphorylated, allowing for the subsequent incorporation of selenium from selenophosphate and yielding the selenocysteine-conjugated tRNA. Selenoprotein transcripts are unique for two reasons. First, they contain a UGA codon within the open reading frame. UGA codons are typically stop codons that bind release factors to terminate translation. However, in selenoprotein transcripts, these UGA codons are recoded for selenocysteine insertion. Second, they contain a stem-loop-like structure called the selenocysteine insertion sequence (SECIS) element in the 3′ untranslated region that recruits machinery such as SECIS binding protein 2 to recode the ribosome for insertion of selenocysteine at the UGA codon, rather than the binding of a ribosome release factor. This unique mode of translation makes selenoprotein translation less efficient and prone to early termination, especially under conditions of limiting selenium63,67.
Important anti-ferroptotic metabolites are also synthesized in the cytosol. The GPX4 cofactor GSH is synthesized in a two-step reaction by glutamate cysteine ligase (GCL; comprising a catalytic and a modifier subunit, GCLC and GCLM, respectively) and GSH synthetase (GSS). Cysteine, obtained from imported cystine or other sources, is rate-limiting for GSH synthesis. It is often assumed that GSH alone is sufficient to inhibit ferroptosis. However, this might not be the case. Genetic disruption of GCLC does not seem to induce ferroptosis despite eliminating de novo GSH synthesis68. It seems possible that one or more additional cysteine-derived, sulfur-containing metabolites can suppress ferroptosis, in parallel to GSH. For example, co-depletion of GSH and the metabolic intermediate coenzyme A (CoA) is necessary and sufficient to trigger ferroptosis in some cancer cells69,70. How CoA impacts ferroptosis is unclear but could be related to the requirement for CoA in lipid metabolism or to a direct role for CoA in modulating protein function via post-translational modification (that is, CoAlation). Additionally, hydropersulfides can suppress lipid peroxidation71,72, and the synthesis of these metabolites requires cysteine. A further possibility is that GSH tripeptide (γ-glutamylcysteinylglycine) serves as a sink for glutamate, which otherwise can act in a pro-ferroptotic manner. Under conditions of intracellular cysteine starvation, GCL remains active and synthesizes an array of alternative dipeptides and tripeptides that contain glutamate and different amino acids in place of cysteine (for example, γ-glutamyl-threonine)73. The synthesis of these unusual metabolites removes free glutamate from the cell, which otherwise enhances pro-ferroptotic oxidative stress through a downstream mechanism that requires further elicidation73.
NADPH is a key electron carrier in the cell. NADP can be synthesized in the cytosol from nicotinamide adenine dinucleotide (NAD+) by NAD kinase (NADK) and then reduced to NADPH by malic enzyme 1 (ME1), isocitrate dehydrogenase (IDH1), or enzymes in the oxidative pentose phosphate pathway74. Higher NADPH levels generally correlate with greater ferroptosis resistance75; this is sensible as NADPH is used to synthesize or regenerate endogenous antioxidant metabolites such as GSH from oxidized glutathione (GSSG) by glutathione-disulfide reductase (GSR), reduced CoQ from oxidized CoQ and reduced vitamin K from oxidized vitamin K by FSP1 (refs. 41,42), and reduced tetrahydrobiopterin (BH4) from BH2 by dihydrofolate reductase (DHFR)23,45. Notably, however, NADPH is also an electron donor for the synthesis of ROS by NOX enzymes at the plasma membrane and by the oxidoreductases cytochrome P450 oxidoreductase (POR) and cytochrome b5 reductase 1 (CYB5R1) at the ER membrane76,77. Thus, NADPH likely has both pro-ferroptotic and anti-ferroptotic functions whose relative importance will vary between cells.
Recent studies connect NADPH to ferroptosis in unexpected ways. HD domain-containing 3 (HDDC3; also known as MESH1) is proposed to function as an NADPH phosphatase whose activity enhances ferroptosis sensitivity by decreasing the levels of NADPH78. Experiments manipulating cytosolic NADK and mitochondrial NADK2 establish that cytosolic NADPH is most critical for governing ferroptosis sensitivity78. NADPH may also be directly sensed by an ER-localized enzyme. The ER-localized E3 ubiquitin ligase membrane-associated ring-CH-type finger 6 (MARCHF6) may directly sense cytosolic NADPH levels via a unique C-terminal binding region, increasing the activity of this enzyme and resulting in changes in the levels of key regulators of ferroptosis, including acyl-CoA synthetase long-chain family member 4 (ACSL4) and p53 (ref. 79). Remarkably, embryonic development is impaired in Marchf6−/− animals and this can be partially reverted by a maternal diet enriched in the natural RTA vitamin E. Thus, MARCHF6 may play a role in preventing ferroptosis during development.
The role of specific organelles in modulating ferroptosis
Most organelles have been linked to the regulation of ferroptosis sensitivity in one way or another, and often in several potentially contradictory ways at once. How these different and sometimes opposing activities are coordinated to generate a unified effect on ferroptosis in a given context is an important open question. Key aspects of ferroptosis regulation by different organelles are described below.
Mitochondria
Mitochondria are multifunctional organelles that can regulate ferroptosis sensitivity in several different ways (Fig. 2). In cells treated with the system xc− inhibitor erastin, mitochondria are smaller, with disorganized cristae1,80. This represents a potential morphological marker of ferroptosis. However, it is not clear whether changes in mitochondrial morphology contribute to the execution of ferroptosis or are merely a correlate of this process. For example, mitochondria can be eliminated from some cells with little effect on ferroptosis sensitivity81. In other cells, mitochondria seem to be essential for ferroptosis82. Below, we describe examples of the pro-ferroptotic and anti-ferroptotic effects of mitochondria, touching on specific examples of context-dependent effects and areas of disagreement in the literature.
Fig. 2 |. Ferroptosis regulation in the mitochondria.

Mitochondria play several context-dependent roles in the regulation of ferroptosis sensitivity. a, Metabolic reactions — the breakdown of sugar and amino acids via glycolysis and the tricarboxylic acid (TCA) cycle yields NADH and FADH2, which pass electrons to the mitochondrial electron transport chain (ETC). The ETC can partially reduce oxygen to generate reactive oxygen species (ROS) such as superoxide (O2•−) at complexes I and III. Dismutation of O2•− yields hydrogen peroxide, which can result in the formation of hydroxyl radicals (HO•) via reactions with Fe2+ in the Fenton reaction. Glutamine can be catabolized to glutamate (a substrate for the system xc− antiporter) and then to the TCA cycle intermediate α-ketoglutarate (αKG). αKG synthesis promotes ferroptosis, possibly by enhancing mitochondrial ROS production. b, Iron handling — mitoferrin 1 (MFRN1) and MFRN2 mediate iron transport across the inner mitochondrial membrane. The synthesis of Fe–S clusters and haem prosthetic groups in the mitochondria consumes labile iron that otherwise may accumulate to promote ROS accumulation. c, Mitochondrial integrated stress response — OMA1-dependent proteolytic processing of DAP3 binding cell death enhancer 1 (DELE1) releases a protein fragment that initiates the integrated stress response, culminating in activating transcription factor 4 (ATF4)-dependent transcription of a programme that enhances glutathione metabolism and protects against ferroptosis. d, Coenzyme Q10 (CoQ) synthesis — the final steps in de novo CoQ synthesis occur in the mitochondria. CoQ is employed as a key electron carrier in the ETC. CoQ can be reduced by dihydroorotate dehydrogenase (DHODH) to CoQH2, which can either transfer electrons to complex III in the ETC or function as a local radical-trapping antioxidant to prevent peroxidation of mitochondrial lipids and limit ferroptosis. In addition, CoQ is trafficked by a processed form of StAR-related lipid transfer protein 7 (STARD7) from the mitochondria to the plasma membrane, where it can contribute to ferroptosis protection mediated by ferroptosis suppressor protein 1. CoA, coenzyme A.
Several mitochondrial activities reduce ferroptosis sensitivity. Mitofusin 1 (MFN1)-mediated mitochondrial fusion may reduce sensitivity to RSL3-induced ferroptosis, at least in cultured cancer cells83. Likewise, in cells with dysfunctional mitochondria, activation of the mitochondrial stress response pathway mediated by metalloen-dopeptidase OMA1 and DAP3 binding cell death enhancer 1 (DELE1) can inhibit ferroptosis and delay cardiomyopathy84. Here, DELE1 activates cytosolic activating transcription factor 4 (ATF4), which in turn promotes GSH synthesis and GPX4 protein stability. Mitochondria are also the site of CoQ metabolite synthesis85. CoQ is an electron carrier in the mitochondrial electron transport chain (ETC) but also functions as an important anti-ferroptotic RTA41,42. Key enzymatic machinery necessary for CoQ synthesis (for example, the COQ proteins) localize to the mitochondria. A specific proteolytically processed version of StAR-related lipid transfer protein 7 (STARD7) transports CoQ from the mitochondria to the plasma membrane, where it can be reduced by FSP1 to inhibit lipid peroxidation and ferroptosis86. Whether STARD7 might act at membrane contact sites between the mitochondria and plasma membrane to promote efficient and directional CoQ transfer is unknown. Another important protective role for mitochondria is in the breakdown of PUFAs that could otherwise be incorporated into membrane phospholipids, peroxidized and thereby promote ferroptosis. This catabolic process requires the mitochondrial enzyme 2,4-dienoyl-CoA reductase 1 (DECR1) and, for reasons that remain unclear, appears especially relevant in prostate cancer87,88. Collectively, these mitochondrial functions would tend to reduce ferroptosis sensitivity.
Other mitochondrial mechanisms increase ferroptosis sensitivity. For example, the mitochondrial tricarboxylic acid (TCA) cycle promotes ferroptosis sensitivity, at least in response to cystine deprivation89. During glutaminolysis, glutamine is converted to glutamate and the TCA cycle intermediate α-ketoglutarate within mitochondria; both glutamate and α-ketoglutarate can promote ferroptosis53,73,90. In these cases, the precise effector mechanism linking TCA cycle activity with the induction of ferroptosis remains obscure, although enhanced oxidative stress seems to be involved. Mitochondria are also central hubs for the use and metabolism of iron2. Increased mitochondrial iron uptake mediated by mitoferrin 1 (MFRN1; encoded by SLC25A37) can enhance ferroptosis sensitivity, although the exact mechanism is unclear91. Disruption of mitochondrial biosynthesis of Fe–S clusters can also enhance ferroptosis sensitivity. Here, depletion of NFS1 cysteine desulfurase lowers the synthesis of Fe–S clusters, triggering a feedback iron starvation response that increases iron import and heightens ferroptosis sensitivity92. It might be the case that cystine deprivation caused by system xc− inhibition has unique effects on mitochondrial Fe–S biosynthesis given the need for cysteine-derived sulfur in this process. One model to explain observed differences in the ferroptosis phenotypes caused by cystine deprivation versus direct GPX4 inhibition10 is that cystine deprivation uniquely perturbs mitochondrial Fe–S cluster biosynthesis45. Finally, mitochondrial outer membrane permeabilization, when it occurs in a partial and non-lethal manner throughout the cell, results in both activation of the ATF4 pathway and enhanced sensitivity to ferroptosis induced by GPX4 inhibitors93. This may suggest that the loss of one or more mitochondrial anti-ferroptotic functions can, on balance, overcome the protective effect of ATF4 pathway activation.
In some cancer cells, mitochondrial ETC activity promotes ferroptosis, at least in response to cystine deprivation, as shown using specific ETC poisons89. How the ETC promotes ferroptosis is not clear. One obvious candidate is the production of ROS that might be expected to initiate lipid peroxidation. Indeed, ROS scavengers that target mitochondria can attenuate ferroptosis in some models, implying that mitochondria-derived oxidants may contribute to cell death (Box 2). However, in other cancer cells, depleting the mitochondrial DNA, resulting in complete loss of ETC function, has no effect on ferroptosis sensitivity1,94. Moreover, ETC function is necessary for the activity of two mitochondrially localized enzymes that may be able to generate reduced CoQ: dihydroorotate dehydrogenase (DHODH)37 and glycerol-3-phosphate dehydrogenase 2 (GPD2)95. Accordingly, loss of ETC function might be expected to enhance ferroptosis sensitivity by disrupting the synthesis of reduced mitochondrial CoQ. Overall, it seems likely that the role of the mitochondrial ETC varies by cell type and by the nature of the lethal stimulus. An emerging (and controversial)38 concept is that the mitochondria might assume a greater importance in overall ferroptosis regulation when cytosolic anti-ferroptotic systems are disrupted37,39,96. This model of a conditional role for mitochondria depending on circumstances could provide a reconciliation for some conflicting observations.
Endoplasmic reticulum
The ER plays an important role in ferroptosis regulation. POR and CYB5R1 are enzymes that reside in the ER membrane and use NADPH to produce ROS that can promote membrane lipid peroxidation76,77 (Fig. 3a). Indeed, there is good evidence that the ER is one site where membrane lipid peroxidation is initiated during ferroptosis (Box 2). As noted above, whether lipid peroxidation spreads from the ER to the plasma membrane to execute ferroptosis, for example, through vesicular trafficking or phospholipid transfer at membrane contact sites, is an open question.
Fig. 3 |. Ferroptosis regulation in the ER.

a, Cytochrome P450 oxidoreductase (POR) and cytochrome b5 reductase 1 (CYB5R1) are endoplasmic reticulum (ER) enzymes that promote ferroptosis by generating H2O2, which can further react with iron to produce reactive oxygen species species that initiate lipid peroxidation, leading to ferroptosis. Lipid peroxidation in the ER is an early event in ferroptosis. Whether damage propagates from the ER or accumulates at a slower rate in other membranes is an open question (see also Box 2). b, The ER is the primary site of lipid synthesis, including the synthesis of glycerolipids, namely phospholipids (PLs) and triacylglycerols (TAGs). The incorporation of monounsaturated fatty acids (MUFAs) and polyunsaturated fatty acids (PUFAs) into PLs or TAGs helps establish the overall sensitivity of the cell to lipid peroxidation and ferroptosis. TAGs are stored within lipid droplets that emerge from the outer leaflet of the ER. PLs are trafficked to the plasma membrane via the secretory pathway or transferred at ER–plasma membrane contact sites. Phosphatidylethanolamine (PE) is often oxidized during ferroptosis. The synthesis of PE involves the transfer of phosphatidylserine from the ER to mitochondria, where it is enzymatically converted to PE prior to transport back to the ER. The relative contributions of these PL trafficking pathways to ferroptosis remain to be defined. c, A complex series of enzymatic steps in the ER mediate the synthesis of cholesterol via the mevalonate pathway. Several intermediates in this pathway protect against ferroptosis. Farnesyl pyrophosphate (FPP) is needed to synthesize the endogenous radical-trapping antioxidants coenzyme Q10 (CoQ) and vitamin K in the mitochondria and Golgi, respectively. Isopentenyl pyrophosphate (IPP) is attached to the selenocysteine tRNA as an isopentenyl moiety, a key modification for its function, promoting the synthesis of selenoproteins, including the anti-ferroptotic enzyme glutathione peroxidase 4 (GPX4). 7-Dehydrocholesterol (7DHC) is highly prone to oxidation and can suppress ferroptosis by competing with PL for oxidation. Squalene also suppresses ferroptosis, but the mechanism is not understood. d, Proteolytic processing of the membrane-tethered transcription factors sterol regulatory element-binding protein 1 (SREBP1) and nuclear factor erythroid 2-related factor 1 (NFE2L1) releases soluble, active transcription factors that traffic to the nucleus and initiate transcriptional programmes to control lipid metabolism and the cellular oxidative stress response. The phosphoinositide 3-kinase (PI3K)–mechanistic target of rapamycin complex 1 (mTORC1) pathway promotes ferroptosis resistance by increasing SREBP1 activation and expression of stearoyl-CoA desaturase 1 (SCD1), which generates ferroptosis-suppressive MUFAs. NFE2L1 is dislocated from the ER into the cytoplasm, where it is deglycosylated by N-glycanase 1 (NGLY1) and proteolytically processed by DNA-damage inducible 1 homologue 2 (DDI2). The amount of NFE2L1 that escapes proteasomal clearance is a determinant of NFE2L1 transcriptional signalling. HMG-CoA, 3-hydroxy-3-methylglutaryl coenzyme A; MVA, mevalonate; GGPP, geranylgeranyl pyrophosphate; UBIAD1, UBIA prenyltransferase domain-containing protein 1.
Ferroptosis is a lipid-dependent process. The ER is a major hub for lipid metabolism in the cell. Lipid desaturation, phospholipid synthesis, phospholipid remodelling and biogenesis of lipid droplets are all ER-localized activities (Fig. 3b). Thus, it is no surprise that ER-resident lipid metabolic enzymes contribute importantly to the regulation of ferroptosis sensitivity. Overall, the ferroptotic threshold of the cell can be determined by lipid composition and, more specifically, by the relative levels of different lipid species97. Decreasing the ratio of more oxidizable PUFAs to less oxidizable monounsaturated fatty acids (MUFAs) is sufficient to convert cells from ferroptosis sensitive to ferroptosis resistant28,98–101 whereas increasing the PUFA to MUFA ratio can have the opposite effect22,77. ER-resident enzymes shape the PUFA and MUFA content of cellular lipid pools. For example, fatty acid desaturase 1 (FADS1) and FADS2, which introduce carbon–carbon double bonds into fatty acids, can enhance or reduce ferroptosis sensitivity, respectively, by synthesizing specific PUFAs or MUFAs102,103. The overall activity of these desaturation processes can be governed within the ER by calcium levels, which are regulated by the tetraspanin membrane-spanning 4-domains subfamily A member 15 (MS4A15)104.
Acyl-CoA synthetase long-chain (ACSL) enzymes ‘activate’ both PUFAs and MUFAs to PUFA-CoA and MUFA-CoA species, which can then be incorporated into membrane phospholipids, triacylglycerols (TAGs) or other lipids. ACSL4 and ACSL3 appear generally to function in opposition, with ACSL4-driven PUFA metabolism increasing ferroptosis sensitivity and ACSL3-dependent MUFA metabolism promoting ferroptosis resistance24,28,30,99,105,106. However, exceptions to this general scheme exist, for example, the pro-ferroptotic role of ACSL3 in KRAS-mutant lung cancer cells107. Further work is required to establish where in the cell these enzymes function to regulate ferroptosis. Genetic rescue data suggests that ER-localized ACSL4 may be critical for ferroptosis regulation27. However, ACSL4 can also sometimes be found in lipid droplets, the Golgi apparatus or at the plasma membrane24,108,109. ACSL3 can also localize to both the ER and to lipid droplets110. Which of these localizations is most important for ferroptosis regulation remains somewhat mysterious and could differ by cell type. How the activities of these enzymes at the ER impact plasma membrane lipid composition and peroxidation27 is also not at all clear.
The ER is the site of de novo phospholipid synthesis111. There is evidence from large-scale genetic screens that ER-resident enzymes involved in de novo phospholipid synthesis, including glycerol-3-phosphate acyltransferase 4 (GPAT4), can promote ferroptosis sensitivity27,111. However, the role of de novo phospholipid synthesis is less explored than the role of phospholipid remodelling (that is, the Lands cycle). The Lands cycle involves cleavage of one acyl chain of a phospholipid by a phospholipase, followed by reacylation of the lysophospholipid with an acyl-CoA to reform the phospholipid. Calcium-independent phospholipase A2 (also known as PLA2G6, PNPLA9 or iPLA2β) can cleave oxidized PUFA acyl chains from phospholipids, thereby directly limiting the spread of lipid peroxidation and the onset of ferroptosis112–114. A related enzyme, PLA2G4C, may also negatively regulate ferroptosis in KRAS-mutant lung cancer in a similar manner107. A distinct lysophospholipase enzyme, abhydrolase domain-containing protein 12 (ABHD12), cleaves PUFA-containing lysophosphatidylserine and oxidized phosphatidylserines specifically, and inhibition of this enzyme is sufficient to increase PUFA-phosphatidylserine (that is, 18:0/20:4 phosphatidylserine) levels and boost ferroptosis sensitivity. Lysophospholipids are reacylated (or, in de novo synthesis, acylated) by phospholipid acyltransferases22. For example, reacylation of lysophospholipids with PUFAs is catalysed by ER-resident enzymes, including lysophosphatidylcholine acyltransferase 3 (LPCAT3; also known as MBOAT5), 1-acylglycerol-3-phosphate O-acyltransferase 3 (AGPAT3)25,105,115 and MBOAT7 (ref. 24), while ER-resident MBOAT1 and MBOAT2 acylate lysophospholipids with MUFAs116,117. Presumably, there is competition between phospholipid acyltransferases to reacylate (or acylate) the same substrates, and the relative levels of different enzymes and substrates will help determine the overall composition of the membrane and, in turn, the ferroptosis sensitivity of the cell. Although LPCAT3 is an ER-resident enzyme118, the phospholipase PLA2G6 may be found through the cytosol with the possibility of being recruited to the vicinity of the plasma membrane119. Thus, it is also unclear whether all ferroptosis-relevant lipid synthesis and remodelling events occur in the ER, or whether localized remodelling in other endomembrane compartments and/or at the plasma membrane also helps shape ferroptosis sensitivity.
In addition to enzymes involved in phospholipid metabolism, the ER houses the enzymes of the mevalonate pathway (Fig. 3c). Although this pathway is best known for the synthesis of cholesterol, several intermediates within this pathway are important in ferroptosis suppression. For example, an intermediate in cholesterol synthesis, 7-dehydrocholesterol, is highly prone to oxidation and can act as a sacrificial endogenous RTA to limit membrane lipid peroxidation120,121. A different metabolite, squalene, has also been implicated in ferroptosis suppression122, but the mechanism remains unclear. Another product of the mevalonate pathway is farnesyl pyrophosphate, a key precursor for the synthesis of vitamin K in the Golgi123 and CoQ in the mitochondria85. Finally, another mevalonate pathway product, isopentenyl pyrophosphate is necessary for isopentylation of the Sec-tRNA and it thereby modulates selenoprotein translation. Whether isopentylation impacts GPX4 levels to an extent that would affect ferroptosis sensitivity remains to be directly tested124.
Beyond its roles in lipid synthesis, the ER is a site for the processing of important transcription factors that modulate the cellular lipid and redox environments (Fig. 3d). Sterol regulatory element-binding proteins (SREBPs) are ER-resident transcription factors that regulate the expression of many enzymes related to lipid metabolism. Tethered to the ER by two transmembrane domains, SREBPs undergo regulated trafficking to the Golgi, where they are proteolytically processed to release the active transcription factor, which traffics to the nucleus to initiate transcription. A signalling pathway involving phosphatidylinositol 3-kinase (PI3K), AKT and mechanistic target of rapamycin complex 1 (mTORC1), can activate SREBP1 to protect cancer cells from ferroptosis125. SREBP1-dependent expression of stearoyl-CoA desaturase 1 (SCD1) is required for this effect125. SCD1 catalyses the synthesis of long-chain MUFAs (for example, oleate); thus, increasing MUFA production should reduce membrane susceptibility to peroxidation and ferroptosis sensitivity. A second transcription factor that is processed via the ER is nuclear factor erythroid 2-related factor 1 (NFE2L1). NFE2L1 undergoes a complex series of post-translational modifications, including glycosylation in the ER and subsequent deglycosylation in the cytosol by N-glycanase 1 (NGLY1), as well as proteolytic cleavage by DNA-damage inducible 1 homologue 2 (DDI2), to yield an active transcription factor126–129. The NGLY1–NFE2L1 axis helps inhibit ferroptosis in some cells by promoting GPX4 protein stability indirectly, possibly through regulation of proteasome gene expression or function130,131.
Finally, the overall function of the ER may be impaired in cells undergoing ferroptosis, at least when induced by cystine deprivation. Cystine deprivation can trigger a global integrated stress response and ER stress response, including changes in gene expression and the viscosity and pH of the ER58,132. Whether these changes contribute directly to the execution of ferroptosis is not known. However, certain changes, such as the transcriptional upregulation of SLC7A11 (which encodes the transport subunit of system xc−)58, might be expected to reduce ferroptosis sensitivity, while changes in viscosity or pH could very well modulate the function of one or more ER-resident enzymes or mechanisms noted above. In sum, the ER has a multifaceted role in ferroptosis regulation.
Lipid droplets
Lipid droplets are dynamic ER-derived lipid storage organelles that have a context-specific role in ferroptosis regulation133 (Fig. 4). Lipid droplets consist of a neutral lipid core, composed mostly of TAG and steryl esters, encircled by a phospholipid monolayer containing integral and peripheral regulatory proteins134. Different fatty acids can be stored in or released from lipid droplet-resident lipids with varying effects on ferroptosis sensitivity. For example, cancer cells grown in acidic conditions that mimic the tumour microenvironment accumulate lipid droplets, and inhibition of lipid droplet synthesis channels PUFAs away from TAGs and into phospholipids, sensitizing cells to ferroptosis135. In mice, oral gavage with PUFA-rich oils together with blockade of lipid droplet biosynthesis increases PUFA-containing phospholipids and is sufficient to trigger ferroptosis in tumour xenografts135. Conceptually similar, in patient-derived glioblastoma cells, if PUFAs are not sequestered in lipid droplets away from membrane phospholipids they can promote ferroptosis sensitivity136. Here, storage of PUFAs in lipid droplets versus incorporation into membrane phospholipids is genotype dependent, with CDKN2A-null glioblastoma cells specifically showing reduced PUFA sequestration in lipid droplets and enhanced ferroptosis induction136. Finally, in cell-cycle-arrested cancer cells, PUFAs can be channelled into lipid droplets, promoting ferroptosis resistance137. Thus, in some cells, the preferential sequestration of PUFAs in lipid droplets, away from membrane phospholipids, limits ferroptosis sensitivity.
Fig. 4 |. PUFA flux and sequestration in lipid droplets.

Polyunsaturated fatty acids (PUFAs), which may be derived from dietary sources or released from the phospholipids by phospholipases, are conjugated with coenzyme A (CoA) by acyl-CoA synthetase enzymes such as acyl-CoA synthetase long-chain family member 4 (ACSL4), a process known as fatty acid activation, and are then incorporated into triacylglycerol (TAG) that is stored in lipid droplets. Sequestration of PUFAs within TAGs can reduce the amount of PUFAs available to be incorporated into phospholipids and may also protect PUFAs from oxidation. PUFAs esterified within TAG can be released from lipid droplets via lipolysis, which involves a series of lipid droplet-associated lipases, or via lipophagy. The released PUFAs may be incorporated into newly synthesized phospholipids or existing phospholipids (via the Lands cycle), thereby sensitizing membranes to oxidative damage. Lipid droplets may sequester and/or release oxidized PUFAs or their breakdown products to influence ferroptosis. AGPAT, acyl-CoA:acylglycerol phosphate acyltransferase; ATGL, adipose triglyceride lipase; DAG, diacylglycerol; DGAT, acyl-CoA:diacylglycerol acyltransferase; ER, endoplasmic reticulum; G-3-P, glycerol-3-phosphate; GPAT, glycerol phosphate acyltransferase; HSL, hormone-sensitive lipase; LPA, lysophosphatidic acid; MAG, monoacylglycerol; MAGL, monoacylglycerol lipase; PA, phosphatidic acid.
However, lipid droplets do not always seem to protect against ferroptosis. In clear cell renal cell carcinoma cells, hypoxia-inducible factor 2α (HIF2α; also known as EPAS1) regulates the expression of hypoxia-inducible lipid droplet-associated protein (HILPDA). HILPDA inhibits patatin-like phospholipase domain-containing 2 (PNPLA2; also known as ATGL), the rate-limiting enzyme in TAG lipolysis. HILPDA expression increases lipid droplet numbers and PUFA-TAG levels, concomitant with increased ferroptosis sensitivity138. Conversely, HILPDA depletion reduces ferroptosis sensitivity138. Furthermore, system xc− inhibition together with dehydroascorbic acid (that is, oxidized vitamin C) treatment can trigger ferroptosis that involves the oxidation of lipid droplets139. In this scenario, inhibition of diacylglycerol O-acyltransferases (DGATs; the rate-limiting enzymes in TAG synthesis) prevents ferroptosis139. Thus, lipid droplets do not always serve as a protective enclosure that prevents PUFA oxidation. Oxidized TAG species have been measured by lipidomics140, but whether the oxidation of TAGs is common and whether oxidized PUFAs are released from TAGs to propagate ferroptosis is unclear. The mechanisms that influence TAG oxidation are also unknown. Given the localization of FSP1 to lipid droplets one can speculate that FSP1 recycles RTAs at this site to protect TAGs from oxidation, but this possibility remains untested. Furthermore, the mechanisms that determine PUFA sequestration or release via lipolysis or lipophagy are also outstanding questions.
Finally, in some contexts, lipid droplet formation does not affect ferroptosis sensitivity positively or negatively. For example, in fibrosarcoma cells incubated with extracellular MUFAs, the number of lipid droplets is increased, but disrupting lipid droplet synthesis using small-molecule DGAT1 and DGAT2 inhibitors does not alter ferroptosis sensitivity or the ability of exogenous MUFAs to inhibit ferroptosis28. Inhibition of the DGAT enzymes also has no effect on ferroptosis sensitivity in an osteosarcoma cell line41, further highlighting the context-specific role of lipid droplets in ferroptosis.
Peroxisomes
Peroxisomes have important roles in several metabolic processes, including very-long-chain fatty acid breakdown and the catabolism of hydrogen peroxide by catalase. However, peroxisomes are currently best understood to contribute to the regulation of ferroptosis sensitivity via the synthesis of ether phospholipids. PUFA-containing ether phospholipids are abundant in the plasma membrane, and these lipids contribute to ferroptosis execution in some contexts. Ether lipid synthesis is a multistep process that involves collaboration between several compartments of the cell. A key precursor molecule, 1-O-alkyl-glycerol-3-phosphate (AGP), is synthesized in the peroxisomes by FAR1, GNPAT, and AGPS and then shuttled to the ER, where further acylation, head group addition and other modifications can be introduced. Genetic disruption of peroxisome biogenesis or peroxisomal AGP synthesis suppresses ferroptosis in renal cell carcinoma models, hepatocellular carcinoma models and endometrial carcinoma models, concomitant with a reduction in the levels of PUFA-containing ether lipid species25. However, there are also cell-specific differences. For example, deletion of AGPS in fibrosarcoma cells eliminates PUFA-containing ether lipids without altering ferroptosis sensitivity27. Moreover, in Caenorhabditis elegans, disruption of the worm AGPS orthologue (ads-1) actually enhances ferroptosis sensitivity in germ cells101. Here, the protective effect of ADS-1-dependent ether lipid synthesis may be explained by the fact that, in C. elegans (versus mammalian cells), ether lipids are more likely to be acylated with anti-ferroptotic MUFA species than with pro-ferroptotic PUFA species141. These findings highlight how the same metabolic pathway can have different effects on ferroptosis depending on the nature of the final products that are synthesized.
Golgi apparatus
The Golgi apparatus is a complex stack of membranous compartments with a role in protein and vesicle trafficking and lipid metabolism. Disrupting Golgi structure pharmacologically can enhance ferroptosis sensitivity in HeLa and other cancer cells, possibly by increasing intracellular oxidative stress142. Mechanistically, this may involve the disruption of endogenous RTA metabolism. UBIA prenyltransferase domain-containing protein 1 (UBIAD1) undergoes regulated cycling between the ER and Golgi apparatus143. UBIAD1 utilizes geranylgeranyl pyrophosphate in the synthesis of menaquinone 4 from menadione144. Menaquinone 4 (a vitamin K-family metabolite) acts as an endogenous lipophilic RTA that can be recycled by FSP1 and vitamin K epoxide reductase complex subunit 1 (VKORC1; an ER-localized enzyme145) to suppress ferroptosis43,146,147. UBIAD1 is also proposed to directly mediate CoQ synthesis in the Golgi to prevent lipid peroxidation148, which would be important as this could potentially provide a source of non-mitochondrial CoQ in the secretory pathway and plasma membrane that is accessible by FSP1. The relative contributions of UBIAD1-synthesized CoQ148 and STARD7 transport of mitochondrially synthesized CoQ86 to the pool of plasma membrane CoQ remain unknown. Of note, UBIAD1 binds and inhibits the sterol-accelerated degradation of 3-hydroxy-3-methylglutaryl (HMG)-CoA reductase in the ER149. Binding of geranylgeranyl pyrophosphate releases UBIAD1 from HMG-CoA reductase and promotes UBIAD1 trafficking to the Golgi149. Thus, UBIAD1-dependent regulation of the mevalonate pathway may indirectly influence de novo synthesis of CoQ in the mitochondria by controlling the synthesis of CoQ precursors (for example, farnesyl pyrophosphate).
Lysosomes
Lipid peroxidation may occur early on in this organelle under ferroptosis-inducing conditions150. Moreover, lysosomal de-acidification generally inhibits ferroptosis150. These effects are likely explained by the role of lysosomes in iron metabolism. Iron can be imported into the cell via the transferrin–transferrin receptor (TFRC) system. Here, Fe3+–transferrin complexes are internalized by TFRC and eventually trafficked to and liberated within the acidic environment of the lysosome. TFRC silencing inhibits ferroptosis, presumably by limiting the uptake of iron into the cell52,53. Iron storage proteins are also degraded in the lysosome. The adaptor protein nuclear receptor coactivator 4 (NCOA4) links iron-loaded ferritin to the lysosomal catabolic machinery through a selective ferritin autophagy pathway known as ferritinophagy; NCOA4 disruption reduces ferroptosis sensitivity by preventing iron retrieval from ferritin and thereby limiting the amount of loosely coordinated (labile) intracellular iron available to participate in redox reactions151,152. Some evidence indicates that NCOA4-mediated ferritin catabolism may be more important for ferroptosis in response to system xc− inhibition than direct GPX4 inhibition153, an example of context-dependent ferroptosis regulation. More broadly, lysosomal catabolic processes are regulated in a multilayered manner by the transcription factor nuclear factor erythroid 2-related factor 2 (NRF2; also known as NFE2L2); one way that loss of NRF2 can enhance ferroptosis sensitivity is by altering lysosome function to increase the pool of labile intracellular iron that can promote lipid peroxidation154.
In addition to regulating iron metabolism, the catabolic function of lysosomes can be engaged either to promote or to prevent ferroptosis. Chaperone-mediated autophagy (CMA) is a degradative process that terminates in lysosomal protein degradation. CMA requires heat shock 70 kDa protein 8 (HSPA8) and the lysosome-associated membrane glycoprotein 2 (LAMP2). System xc− inhibitors can increase LAMP2 expression, the association of HSPA8 and LAMP2 with GPX4, and the degradation of GPX4 via CMA, thereby enhancing ferroptosis155. GPX4 degradation via CMA is reduced by creatine kinase B (CKB), which, acting as a protein kinase, phosphorylates GPX4 at Ser104 to limit GPX4 interaction with HSPA8 and LAMP2 (ref. 156). While CMA of GPX4 tends to promote ferroptosis, the catabolism of other proteins can inhibit ferroptosis. Namely, lysosomal cathepsin D-mediated catabolism of cysteine-rich extracellular proteins such as albumin, followed by cystinosin-mediated export of cystine from the lysosome to the cytosol, can enhance GSH synthesis and inhibit ferroptosis when extracellular free cystine is limiting59. Albumin is abundant outside the cell, potentially providing an important alternative cysteine supply. Within the cell, one response to cysteine deprivation is activation of the integrated stress response, including increased expression of the transcription factor ATF4 (ref. 157). Depletion of lysosomal cystine specifically increases ATF4 expression and reduces ferroptosis sensitivity158.
The surface of the lysosome is an important site where the nutrient-sensing mTORC1 can be activated. In some cells, active mTORC1 promotes ferroptosis by favouring mRNA translation and the consumption of limited intracellular cysteine supplies in the synthesis of proteins rather than GSH90. In other cells, active mTORC1 inhibits ferroptosis by stimulating the translation of GPX4 or the activation of SREBP1, which, as noted above, promotes anti-ferroptotic MUFA synthesis125,159. Which of these pro-ferroptotic and anti-ferroptotic effects dominates at a given time seems likely to vary by cell line and context.
The nucleus
Morphological alterations of the nucleus (for example, karyorrhexis) are defining features of apoptosis160. By contrast, during ferroptosis, the nuclei appear to remain largely intact and DNA damage is neither a defining feature nor a necessary mechanistic event in this process161. The nuclear envelope of mammalian cells is continuous with the ER membrane and includes PUFA-containing phospholipids that may be susceptible to lipid peroxidation162. A functional role for nuclear lipid peroxidation in ferroptosis has not been reported. However, increased levels of the DNA-damage marker 8-oxo-2′-deoxyguanosine (8-oxo-dG) can correlate with the induction of ferroptosis in vivo163. While DNA damage may not contribute to the execution of ferroptosis, one possibility is that peroxidation of nuclear membrane phospholipids leads to the formation of lipid fragments that locally damage DNA during ferroptosis.
Within the nucleus, transcription is a major activity and contributes importantly to the regulation of ferroptosis sensitivity. As one might expect from the context-dependent nature of ferroptosis regulation, diverse transcriptional processes occurring in the nucleus help set the ferroptosis threshold of a given cell. A non-exhaustive list of transcription factors and epigenetic regulators that can regulate ferroptosis sensitivity include p53 (refs. 6,147,164), NRF2 (refs. 154,165–168), NFE2L1 (refs. 130,131), ZEB1 (ref. 169), STAT1 (ref. 170), ATF3 (ref. 171), YAP1 and TAZ50,172, HIF2α138, PPARα173, NUPR1 (ref. 174), MYCN65,175,176, and BAP1 (refs. 7,177). These proteins regulate the expression of genes (for example, SLC7A11, ACSL4, HILPDA, FSP1, NCOA4, LCN2, VKORC1L1 and LRP8) that, in turn, govern ferroptosis sensitivity in different ways, typically by regulating the levels of proteins or metabolites that impinge on membrane lipid peroxidation. Again, context-specific regulation appears to be the rule, with some transcription factors having an important role in ferroptosis regulation in one cell type but not in another178.
Dynamic and spatially distributed processes shape ferroptosis sensitivity
Above, we have largely described the roles of different structures and organelles in ferroptosis regulation in isolation. However, activities in the plasma membrane, cytosol and various intracellular organelles interact with one another (for example, synthesis of CoQ in the mitochondria and Golgi followed by transport to the plasma membrane). Moreover, metabolism is more broadly coordinated by signalling and transcriptional networks in ways that can alter ferroptosis sensitivity. Some additional examples of these interconnections and how they can regulate ferroptosis are highlighted below.
Lipid metabolism
Fatty acid synthesis, trafficking and flux help dictate ferroptosis sensitivity by controlling the abundance and localization of more oxidizable versus less oxidizable lipids. These processes involve interactions between plasma membrane transport processes and enzymes and signalling pathways that are distributed throughout the cell. For example, cytosolic acetyl-CoA carboxylase 1 (ACC1) synthesizes malonyl-CoA, which can be used both in the synthesis of long-chain saturated fatty acids by fatty acid synthase (FASN) and in the elongation of imported PUFAs55 into longer-chain-length PUFA species. The desaturation of long-chain saturated fatty acids by SCD1, FADS1 and FADS2 occurs in the ER, which is also the site where fatty acids are activated by ACSL enzymes and incorporated into phospholipids or TAGs by a host of acyltransferases. ACC1, FASN and SCD1 can all regulate ferroptosis sensitivity but seem to do so in different ways in different cells and situations125,179–181. Furthermore, signalling networks impact the function of these enzymes. In one example, under conditions of glucose limitation, cytosolic AMPK is activated and can phosphorylate and inhibit ACC1 and ACC2, which ultimately lowers ferroptosis sensitivity by reducing the abundance of oxidizable PUFA-containing membrane phospholipids (for example, PE 18:0/22:4)30,182, perhaps by limiting the malonyl-CoA-dependent elongation of shorter-chain PUFAs. In another example, oncogenic KRAS activation can increase the expression of FASN, which in turn leads to more incorporation of saturated fatty acids and MUFAs into membrane phosphatidylcholines in place of PUFAs, reducing ferroptosis sensitivity107. Likewise, in FLT3-mutant acute myeloid leukaemia, the expression of FASN and SCD1 is increased, resulting in reduced ferroptosis sensitivity176. Outside the context of cancer, how different signalling networks may dynamically regulate lipid metabolism to influence lipid peroxidation and ferroptosis sensitivity is mostly unclear.
How lipids are channelled into different classes of membrane phospholipids, to either suppress or enhance ferroptosis, is also not well understood. For example, phosphatidylethanolamine is a key phospholipid that is often oxidized during ferroptosis21. The synthesis of phosphatidylethanolamine involves the transport of phosphatidylserine from the ER to mitochondria at membrane contact sites known as mitochondria-associated membranes183 (Fig. 3b). Within the mitochondria, phosphatidylserine is enzymatically converted into phosphatidylethanolamine and then transported back to the ER, at which point it can traffic to other compartments in the cell (for example, plasma membrane) via the secretory pathway or lipid transfer proteins183 (Fig. 3b). How these trafficking events may contribute to ferroptosis is not clear. In particular, while oxidized phosphatidylethanolamine has been implicated in ferroptosis, its trafficking within the cell has not been directly studied in the context of ferroptosis. Likewise, the flux of PUFAs into TAGs, concomitant with the biogenesis of lipid droplets from the ER, can be protective against ferroptosis in some cases by restricting the incorporation of PUFAs into phospholipids. However, how decisions to channel fatty acids into different lipid species and to traffic phospholipids from one compartment to another are made is not well understood. Additionally, it is possible that cellular decisions about lipid fluxes are made for reasons completely independent of cell death regulation (for example, connected to membrane remodelling events needed for cell division)117,184 and that increased sensitivity or resistance to ferroptosis is a by-product of these changes.
Ferroptosis amplification mechanisms
Rising plasma membrane lipid peroxidation can trigger feedforward mechanisms that accelerate this process (Fig. 1b). First, lipid peroxidation leads to water entry and cell swelling, increasing plasma membrane tension, which then further activates mechanosensitive Piezo1 and TRP ion channels, accelerating this loop9. Second, lipid peroxidation can stimulate the activity of protein kinase Cβ (PKCβII), which in turn phosphorylates ACSL4 at Thr328, increasing the activity of this lipid metabolic enzyme and enhancing the synthesis of oxidizable PUFA-containing membrane phospholipids185. Where exactly in the cell this pool of ACSL4 resides is not entirely clear; ACSL4 is thought to localize to the ER but a pool of this enzyme may also be found at the plasma membrane and is therefore possibly able to modulate plasma membrane lipid composition directly. Third, plasma membrane lipid peroxidation can cause the protein CAVIN1 to be released from plasma membrane caveolae, allowing it to migrate into the cytosol, where it can bind to and promote the degradation of NRF2 (ref. 186), thereby limiting the protective response mediated by this transcription factor. Fourth, increasing lipid peroxidation can lead to the hyperoxidation of mitochondrial peroxiredoxin 3 (PRDX3), which then translocates to the plasma membrane and inhibits the function of the system xc− cystine–glutamate antiporter, which will tend to increase ferroptosis sensitivity187. These amplification mechanisms may be engaged to overcome the powerful lipid peroxidation defence and membrane repair mechanisms that operate at the plasma membrane, ensuring that the execution of ferroptosis proceeds to completion once this process has been initiated188.
Feedback suppression of ferroptosis sensitivity
While some mechanisms may serve to accelerate ferroptosis, others operating in parallel will tend to restrain this process. These mechanisms can also be distributed spatially within the cell. For example, in breast cancer cells exposed to GPX4 inhibitors, the pentaspanin protein prominin 2 (PROM2) can be upregulated and stimulate the release of extracellular vesicles containing iron-loaded ferritin from the cell, thereby attenuating lipid peroxidation189. In another example, in tumour-infiltrating neutrophils, signals emanating from cell surface receptors eventually lead to upregulation of aconitate decarboxylase 1 (ACOD1) expression in the mitochondria, which synthesizes the metabolite itaconate. Itaconate can then diffuse out of the mitochondria, bind the NRF2 negative regulator KEAP1 in the cytosol and stabilize NRF2 to suppress ferroptosis190.
Intracellular trafficking
Lipid peroxidation occurring within the cell in response to GPX4 inhibition can have a profound effect on the function of the secretory pathway. As noted above, in some cells, GPX4 inhibition stimulates the secretion of extracellular vesicles189. Additionally, in response to the same stimulus, TFRC is depleted from the Golgi apparatus and the endosomal recycling compartment and accumulates at the plasma membrane191. Why TFRC gets ‘stuck’ at the plasma membrane following GPX4 inhibition is unclear, especially since a different cell surface protein, EGFR, traffics normally in response to GPX4 inhibitor treatment191. This TFRC protein redistribution within the cell is a potential ferroptosis molecular marker. Functionally, reduced trafficking of a key iron metabolism-related protein such as TFRC may also tend to reduce ferroptosis sensitivity by limiting iron uptake. The trafficking of other proteins within the secretory pathway, such as the innate immune regulator stimulator of interferon genes (STING)192, is also inhibited following GPX4 inactivation and increased levels of lipid peroxidation. It is unclear whether the inhibition of STING trafficking contributes to ferroptosis execution. However, this example suggests that a pro-ferroptotic stimulus such as GPX4 inactivation can have diverse effects on protein trafficking that are likely to alter cellular responses to other stimuli. Understanding how intracellular protein and lipid transport networks may change over time in response to ferroptotic stress to either reduce or increase ferroptosis sensitivity is an interesting frontier area.
Transcription programmes
Different transcription factors can induce gene expression programmes that ultimately regulate ferroptosis sensitivity by modulating the function of multiple pathways and organelles in parallel. NRF2 will generally promote ferroptosis resistance by positively regulating the expression of several factors: GSH biosynthetic enzymes that localize to the cytosol130, FSP1 (refs. 165,166), which is required at the plasma membrane, and proteins involved in iron homeostasis found at the lysosome154. However, these mechanisms are not entirely universal. High NRF2 expression can, in some cancer cells, tend to promote ferroptosis sensitivity by upregulating the expression of a plasma membrane-localized transporter (MRP1; encoded by ABCC1) that exports GSH from the cell57. The tumour suppressor protein p53 also seems to be capable of either promoting or inhibiting ferroptosis through various molecular mechanisms, depending on the cell type and even on the nature of the ferroptosis-inducing stimulus6,117,164,193. Likewise, it seems possible that similar competing mechanisms could exist for other transcriptional regulators of ferroptosis. For example, SREBP1 can positively regulate the expression of ER-resident SCD1 (refs. 125,180) but also of cytosolic enzymes involved in malonyl-CoA and NADPH synthesis194. The coordinate expression of these genes may help enable anti-ferroptotic MUFA synthesis but could conceivably also be involved in enhancing ferroptosis in other ways as described above (for example, NADPH-dependent ROS synthesis that initiates ferroptosis). While it is uncomfortable to not have universal models of ferroptosis regulation by individual transcription factors that apply to all cell types and contexts, it does help illustrate the importance and value of detailed investigations of ferroptosis regulation across systems, with careful attention to how ferroptosis is being induced.
Conclusion and perspective
The execution and regulation of ferroptosis are governed by a network of metabolites, enzymes and biochemical pathways that are distributed throughout the cell (Fig. 5). Ferroptosis execution centres on plasma membrane lipid peroxidation and plasma membrane rupture. Major unanswered questions include whether and how lipid peroxidation reactions spread within the cell, how individual lipid species break down in response to peroxidation, and how these events in turn promote (or inhibit) membrane rupture. Intracellular lipid transport is another area where additional investigation could prove fruitful. Beyond STARD7, the role of lipid transport proteins in ferroptosis is largely unexplored. In general, how inter-organelle communication may shape the ferroptosis response remains a frontier area. This communication may take different forms. For example, stable contacts between organelles could provide a means for the direct spread of lipid peroxidation or the sharing of key pro-ferroptotic or anti-ferroptotic metabolites. One specific possibility is that lipid peroxidation could spread between organelles via membrane contact sites, enabling the propagation of ferroptosis within the cell. More broadly, much of the knowledge described here concerning the cell biology of ferroptosis is derived from studies of cultured cancer cells. Normal cells, including more specialized cells such as neurons or microglia195,196, may use distinct mechanisms to execute ferroptosis or to govern sensitivity to this process.
Fig. 5 |. A cellular map of ferroptosis regulation.

Cellular sensitivity to ferroptosis is influenced by spatially segregated processes that occur in many organelles. These include processes that regulate fatty acid availability and the incorporation of oxidizable fatty acids into phospholipids, the synthesis and recycling of endogenous antioxidants, the production and metabolism of reactive oxygen species (ROS), and iron metabolism and sequestration. CoQ, coenzyme Q10; MUFA, monounsaturated fatty acid; NFE2L1, nuclear factor erythroid 2-related factor 1; PUFA, polyunsaturated fatty acid; VLCFA, very-long-chain fatty acid; SREBP, sterol regulatory element-binding protein.
The sensitivity of the cell to lethal membrane lipid peroxidation is governed by a host of molecules — hundreds of different metabolites and enzymes, along with iron, distributed throughout the plasma membrane, cytosol and the various organelles. It seems possible that every protein, metabolite or biomolecule that can alter iron, lipid or redox metabolism may have a quantitative effect on lipid peroxidation and sensitivity to ferroptosis10. The relative importance of these molecules may be tied to relative expression and abundance, explaining why they vary between different cells and systems10 (Box 3). To understand and manipulate ferroptosis most effectively, it will be important to fully enumerate all relevant species and understand where they act within the cell to modulate ferroptosis. At a higher level of organization, understanding how contact and communication between two or more mammalian cells32,197,198 and how molecules released by other organisms in the environment that act on mammalian cells199,200 can regulate ferroptosis sensitivity will also be important for a full understanding of this mechanism.
Box 3. Ferroptosis sensitivity: the role of sex and ethnicity.
Differences in ferroptosis sensitivity are observed between different cell lines and tissues and in response to different inducing conditions. In some cases, these differences can be directly tied to sex and manifest in the context of disease. In the kidney, genetic disruption of Gpx4 and ischaemic stress induce ferroptosis more extensively in males than in females222. Mechanistically, higher expression of nuclear factor erythroid 2-related factor 2 (NRF2) in the female kidney may explain this mechanism of selective protection. In another example, a sex-dependent difference in lipid remodelling pathways impacts ferroptosis sensitivity. In oestrogen receptor-positive breast cancer cells, upregulation of MBOAT1 can drive ferroptosis resistance by increasing the incorporation of protective monounsaturated fatty acids into membrane phospholipids116. In androgen receptor-positive prostate cancer cells, the paralogous protein MBOAT2 mediates the same anti-ferroptotic effect116. Antagonists of the oestrogen receptor and androgen receptor may help reduce ferroptosis resistance in these diseases. By contrast, in prostate cancer, DECR1 expression is negatively regulated by androgen receptor, meaning that androgen receptor antagonists will increase DECR1 expression, reducing the levels of intracellular polyunsaturated fatty acids and thereby reducing ferroptosis sensitivity87,88. Another cancer-related, sex-specific interaction is observed in patients with colorectal cancer with KRAS mutations. Here, male patients exhibit poorer overall survival than female patients, and this is correlated to changes in multiple ferroptosis metabolic and gene expression biomarkers and with increased resistance to ferroptosis223. However, the specific molecular interactions between KRAS mutation and sex in the colon that may give rise to differences in ferroptosis sensitivity remain to be clarified.
Ethnicity may also be associated with differences in ferroptosis sensitivity. A connection of interest relates to the tumour suppressor p53. A significant fraction of individuals of African descent (~1–8%) have a single-nucleotide polymorphism at codon 47 that results in a Pro-to-Ser mutation224. This change eliminates a key Pro residue in the context of a Ser46-Pro47 dipeptide that serves as a recognition site for Ser-directed kinases. This ‘S47’ variant has a reduced ability to suppress tumour formation in vivo, and this is linked to a reduced ability to stimulate or sensitize to ferroptosis224. Mechanistically, the p53 S47 variant is less able to transactivate certain metabolic genes, such as the mitochondrial glutamine metabolic gene Gls2, which other studies have suggested is essential for ferroptosis53. Compared to wild-type cells, S47 cells generate more NADPH via the oxidative pentose phosphate pathway and also have higher intracellular levels of the metabolites glutathione and coenzyme A, all of which tend to reduce ferroptosis sensitivity70,225. These studies suggest that certain individuals of African descent may be more prone to having cancer due to an inability to eliminate incipient tumour cells through activation of p53 and the induction of ferroptosis.
Finally, viewing ferroptosis from a cell biological perspective advances our understanding of how we might best promote or interfere with this process by modulating the function of different structures within the cell. Intriguing connections between ferroptosis and other forms of non-apoptotic cell death are emerging at the level of membrane repair, where ESCRT-III complex proteins have roles in negatively regulating multiple forms of cell death. Speculatively, interventions that augment this repair system could delay or prevent multiple forms of cell death at one time, which could be useful for pathologies where more than one form of non-apoptotic cell death is activated. By contrast, agents that can inactivate these pan-pathway protective mechanisms may be especially useful to treat diseases such as cancer, where activation of any one lethal mechanism may be overcome by compensatory mechanisms. Efforts to target ferroptosis therapeutically will be greatly aided by the discovery of additional markers of this process. Recent studies identifying re-localized transferrin receptor and PRDX3 as ferroptosis markers187,191, as well as computer-aided analyses of ferroptotic cell morphology201, highlight the importance of incorporating cell biology into these efforts.
Acknowledgements
The authors thank L. Magtanong, D. Pratt and members of the Dixon and Olzmann labs for discussion and comments on the manuscript. This work is supported by the National Institutes of Health (R01GM122923 to S.J.D. and R01GM112948 to J.A.O.) and the American Cancer Society (RSG-21-017-01 to S.J.D. and RSG-19-192-01 to J.A.O.). J.A.O. is a Chan Zuckerberg Biohub Investigator and is also supported by a Bakar Fellows Spark Award.
Glossary
- Caveolae
Small invaginations of the plasma membrane enriched for certain lipids and proteins.
- Endocytosis
The process of taking up materials from outside the cells into intracellular vesicles called endosomes. Endosomes can then transport materials within the cell to other compartments such as the lysosome.
- Ether lipid
A class of phospholipid where one of the two fatty acyl chains is bound to the glycerol backbone by an ether bond rather than the more common ester bond. Ether lipids may contribute importantly to the execution of ferroptosis in some cells.
- Ferritinophagy
The catabolism of iron-laden ferritin nanocages in the autophagolysosome to release free iron atoms.
- Fe–S clusters
Iron–sulfur clusters are essential enzyme cofactors. Several different configurations of iron and sulfur atoms yield distinct types of clusters.
- Integrated stress response
A gene expression programme that can help the cell respond to a shortage of individual amino acids by increasing the expression of membrane transporters and other metabolic enzymes that restore homeostasis.
- Lipolysis
The enzymatic breakdown of triacylglycerol to glycerol and free fatty acids mediated by a series of lipases recruited to the lipid droplet surface.
- Lipophagy
The breakdown of the lipid droplet, or a portion of the lipid droplet, through its delivery to the lysosome by a selective autophagic pathway.
- Membrane contact sites
Regions of close proximity between two organelles that are often stabilized by protein tethers. These sites typically function as sites for the exchange of lipids and metabolites.
- Monounsaturated fatty acids
(MUFAs). Fatty acid molecules that contain a single carbon–carbon double bond.
- Necroptosis and pyroptosis
Two forms of non-apoptotic cell death that are biochemically distinct from each other and from ferroptosis.
- Polyunsaturated fatty acid
(PUFA). Fatty acid molecule that contains multiple carbon–carbon double bonds, making it more sensitive to oxidative damage.
- Reactive oxygen species
(ROS). An umbrella term for a number of small oxygen radicals and species that contain oxygen and which can easily form radical-containing species. Oxygen radicals that may initiate lipid peroxidation, leading to ferroptosis, include the hydroperoxyl radical (the conjugate acid of superoxide) and the hydroxyl radical, which itself can be formed from the Fenton reaction between hydrogen peroxide and iron.
- Selenoprotein
One of a small number of proteins in mammalian cells that incorporate the unusual selenium-containing amino acid selenocysteine. Typically, these proteins are involved in some aspect of redox regulation in the cell.
- Stimulator of interferon genes
(STING). A protein that can sense DNA in the cytosol and orchestrate a downstream immune response by binding to other signalling proteins.
Footnotes
Competing interests
S.J.D. is a co-founder of Prothegen and a member of the scientific advisory board for Hillstream BioPharma. S.J.D. holds patents related to ferroptosis. J.A.O. is a member of the scientific advisory board for Vicinitas Therapeutics and holds patents related to ferroptosis.
References
- 1.Dixon SJ et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell 149, 1060–1072 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Galy B, Conrad M & Muckenthaler M Mechanisms controlling cellular and systemic iron homeostasis.Nat. Rev. Mol. Cell Biol 10.1038/s41580-023-00648-1 (2023). [DOI] [PubMed] [Google Scholar]
- 3.Conrad M & Pratt DA The chemical basis of ferroptosis. Nat. Chem. Biol 15, 1137–1147 (2019). [DOI] [PubMed] [Google Scholar]
- 4.Ursini F & Maiorino M Lipid peroxidation and ferroptosis: the role of GSH and GPx4. Free Radic. Biol. Med 152, 175–185 (2020). [DOI] [PubMed] [Google Scholar]
- 5.Conrad M et al. Regulation of lipid peroxidation and ferroptosis in diverse species. Genes Dev 32, 602–619 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Jiang L et al. Ferroptosis as a p53-mediated activity during tumour suppression. Nature 520, 57–62 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Zhang Y et al. BAP1 links metabolic regulation of ferroptosis to tumour suppression. Nat. Cell Biol 20, 1181–1192 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Stockwell BR Ferroptosis turns 10: emerging mechanisms, physiological functions, and therapeutic applications. Cell 185, 2401–2421 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Hirata Y et al. Lipid peroxidation increases membrane tension, Piezo1 gating, and cation permeability to execute ferroptosis.Curr. Biol 33, 1282–1294.e5 (2023). [DOI] [PubMed] [Google Scholar]
- 10.Dixon SJ & Pratt DA Ferroptosis: a flexible constellation of related biochemical mechanisms. Mol. Cell 83, 1030–1042 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Green DR The coming decade of cell death research: five riddles. Cell 177, 1094–1107 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Kayagaki N et al. NINJ1 mediates plasma membrane rupture during lytic cell death. Nature 591, 131–136 (2021). [DOI] [PubMed] [Google Scholar]
- 13.Dondelinger Y et al. NINJ1 is activated by cell swelling to regulate plasma membrane permeabilization during regulated necrosis. Cell Death Dis 14, 755 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Pope LE & Dixon SJ Regulation of ferroptosis by lipid metabolism. Trends Cell Biol 33, 1077–1087 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Liang D, Minikes AM & Jiang X Ferroptosis at the intersection of lipid metabolism and cellular signaling. Mol. Cell 82, 2215–2227 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Li Z, Lange M, Dixon SJ & Olzmann JA Lipid quality control and ferroptosis: from concept to mechanism. Annu. Rev. Biochem 10.1146/annurev-biochem-052521-033527 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Wenzel SE et al. PEBP1 wardens ferroptosis by enabling lipoxygenase generation of lipid death signals. Cell 171, 628–641.e6 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Shah R, Shchepinov MS & Pratt DA Resolving the role of lipoxygenases in the initiation and execution of ferroptosis. ACS Cent. Sci 4, 387–396 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Zilka O 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 3, 232–243 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Anthonymuthu TS et al. Resolving the paradox of ferroptotic cell death: ferrostatin-1 binds to 15LOX/PEBP1 complex, suppresses generation of peroxidized ETE-PE, and protects against ferroptosis. Redox Biol 38, 101744 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Tyurina YY et al. Redox phospholipidomics discovers pro-ferroptotic death signals in A375 melanoma cells in vitro and in vivo. Redox Biol 61, 102650 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Kathman SG, Boshart J, Jing H & Cravatt BF Blockade of the lysophosphatidylserine lipase ABHD12 potentiates ferroptosis in cancer cells. ACS Chem. Biol 15, 871–877 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Kraft VAN et al. GTP cyclohydrolase 1/tetrahydrobiopterin counteract ferroptosis through lipid remodeling. ACS Cent. Sci 6, 41–53 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Phadnis VV et al. MMD collaborates with ACSL4 and MBOAT7 to promote polyunsaturated phosphatidylinositol remodeling and susceptibility to ferroptosis. Cell Rep 42, 113023 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Zou Y et al. Plasticity of ether lipids promotes ferroptosis susceptibility and evasion. Nature 585, 603–608 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Reed A, Ware T, Li H, Fernando Bazan J & Cravatt BF TMEM164 is an acyltransferase that forms ferroptotic C20:4 ether phospholipids. Nat. Chem. Biol 19, 378–388 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Magtanong L et al. Context-dependent regulation of ferroptosis sensitivity. Cell Chem. Biol 29, 1409–1418.e6 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Magtanong L et al. Exogenous monounsaturated fatty acids promote a ferroptosis-resistant cell state. Cell Chem. Biol 26, 420–432.e9 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.von Krusenstiern AN et al. Identification of essential sites of lipid peroxidation in ferroptosis. Nat. Chem. Biol 19, 719–730 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Kagan VE et al. Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis. Nat. Chem. Biol 13, 81–90 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Pedrera L et al. Ferroptotic pores induce Ca2+ fluxes and ESCRT-III activation to modulate cell death kinetics. Cell Death Differ 28, 1644–1657 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Riegman M et al. Ferroptosis occurs through an osmotic mechanism and propagates independently of cell rupture. Nat. Cell Biol 22, 1042–1048 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Agmon E, Solon J, Bassereau P & Stockwell BR Modeling the effects of lipid peroxidation during ferroptosis on membrane properties. Sci. Rep 8, 5155 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Roveri A, Maiorino M, Nisii C & Ursini F Purification and characterization of phospholipid hydroperoxide glutathione peroxidase from rat testis mitochondrial membranes. Biochim. Biophys. Acta 1208, 211–221 (1994). [DOI] [PubMed] [Google Scholar]
- 35.Seiler A et al. Glutathione peroxidase 4 senses and translates oxidative stress into 12/15-lipoxygenase dependent- and AIF-mediated cell death. Cell Metab 8, 237–248 (2008). [DOI] [PubMed] [Google Scholar]
- 36.Friedmann Angeli JP et al. Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nat. Cell Biol 16, 1180–1191 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Mao C et al. DHODH-mediated ferroptosis defence is a targetable vulnerability in cancer. Nature 593, 586–590 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Mishima E et al. DHODH inhibitors sensitize to ferroptosis by FSP1 inhibition. Nature 619, E9–E18 (2023). [DOI] [PubMed] [Google Scholar]
- 39.Mao C, Liu X, Yan Y, Olszewski K & Gan B Reply to: DHODH inhibitors sensitize to ferroptosis by FSP1 inhibition. Nature 619, E19–E23 (2023). [DOI] [PubMed] [Google Scholar]
- 40.Labrecque CL & Fuglestad B Electrostatic drivers of GPx4 interactions with membrane, lipids, and DNA. Biochemistry 60, 2761–2772 (2021). [DOI] [PubMed] [Google Scholar]
- 41.Bersuker K et al. The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature 575, 688–692 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Doll S et al. FSP1 is a glutathione-independent ferroptosis suppressor. Nature 575, 693–698 (2019). [DOI] [PubMed] [Google Scholar]
- 43.Mishima E et al. A non-canonical vitamin K cycle is a potent ferroptosis suppressor. Nature 608, 778–783 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Lv Y et al. Structural insights into FSP1 catalysis and ferroptosis inhibition. Nat. Commun 14, 5933 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Soula M et al. Metabolic determinants of cancer cell sensitivity to canonical ferroptosis inducers. Nat. Chem. Biol 16, 1351–1360 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Jakaria M, Belaidi AA, Bush AI & Ayton S Vitamin A metabolites inhibit ferroptosis. Biomed. Pharmacother 164, 114930 (2023). [DOI] [PubMed] [Google Scholar]
- 47.Dai E, Meng L, Kang R, Wang X & Tang D ESCRT-III-dependent membrane repair blocks ferroptosis. Biochem. Biophys. Res. Commun 522, 415–421 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Gong YN et al. ESCRT-III acts downstream of MLKL to regulate necroptotic cell death and its consequences. Cell 169, 286–300.e16 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Ruhl S et al. ESCRT-dependent membrane repair negatively regulates pyroptosis downstream of GSDMD activation. Science 362, 956–960 (2018). [DOI] [PubMed] [Google Scholar]
- 50.Yang WH et al. The hippo pathway effector TAZ regulates ferroptosis in renal cell carcinoma. Cell Rep 28, 2501–2508.e4 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Poursaitidis I et al. Oncogene-selective sensitivity to synchronous cell death following modulation of the amino acid nutrient cystine. Cell Rep 18, 2547–2556 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Yang WS & Stockwell BR Synthetic lethal screening identifies compounds activating iron-dependent, nonapoptotic cell death in oncogenic-RAS-harboring cancer cells. Chem. Biol 15, 234–245 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Gao M, Monian P, Quadri N, Ramasamy R & Jiang X Glutaminolysis and transferrin regulate ferroptosis. Mol. Cell 59, 298–308 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Guan Y et al. A single genetic locus controls both expression of DPEP1/CHMP1A and kidney disease development via ferroptosis. Nat. Commun 12, 5078 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Ma X et al. CD36-mediated ferroptosis dampens intratumoral CD8+ T cell effector function and impairs their antitumor ability. Cell Metab 33, 1001–1012.e5 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Lin Z et al. The lipid flippase SLC47A1 blocks metabolic vulnerability to ferroptosis. Nat. Commun 13, 7965 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Cao JY et al. A genome-wide haploid genetic screen identifies regulators of glutathione abundance and ferroptosis sensitivity. Cell Rep 26, 1544–1556.e8 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Dixon SJ et al. Pharmacological inhibition of cystine-glutamate exchange induces endoplasmic reticulum stress and ferroptosis. eLife 3, e02523 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Armenta DA et al. Ferroptosis inhibition by lysosome-dependent catabolism of extracellular protein. Cell Chem. Biol 29, 1588–1600.e7 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Kobayashi S et al. Carnosine dipeptidase II (CNDP2) protects cells under cysteine insufficiency by hydrolyzing glutathione-related peptides. Free Radic. Biol. Med 174, 12–27 (2021). [DOI] [PubMed] [Google Scholar]
- 61.Hayashima K & Katoh H Expression of gamma-glutamyltransferase 1 in glioblastoma cells confers resistance to cystine deprivation-induced ferroptosis. J. Biol. Chem 298, 101703 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Zhang L et al. Hypersensitivity to ferroptosis in chromophobe RCC is mediated by a glutathione metabolic dependency and cystine import via solute carrier family 7 member 11. Proc. Natl Acad. Sci. USA 119, e2122840119 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Li Z et al. Ribosome stalling during selenoprotein translation exposes a ferroptosis vulnerability. Nat. Chem. Biol 18, 751–761 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Greenough MA et al. Selective ferroptosis vulnerability due to familial Alzheimer’s disease presenilin mutations. Cell Death Differ 29, 2123–2136 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Alborzinia H et al. LRP8-mediated selenocysteine uptake is a targetable vulnerability in MYCN-amplified neuroblastoma. EMBO Mol. Med 15, e18014 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Carlisle AE et al. Selenium detoxification is required for cancer-cell survival. Nat. Metab 2, 603–611 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Howard MT, Carlson BA, Anderson CB & Hatfield DL Translational redefinition of UGA codons is regulated by selenium availability. J. Biol. Chem 288, 19401–19413 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Harris IS et al. Deubiquitinases maintain protein homeostasis and survival of cancer cells upon glutathione depletion. Cell Metab 29, 1166–1181.e6 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Badgley MA et al. Cysteine depletion induces pancreatic tumor ferroptosis in mice. Science 368, 85–89 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.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 116, 8390–8396 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Barayeu U et al. Hydropersulfides inhibit lipid peroxidation and ferroptosis by scavenging radicals. Nat. Chem. Biol 19, 28–37 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Wu Z et al. Hydropersulfides inhibit lipid peroxidation and protect cells from ferroptosis. J. Am. Chem. Soc 144, 15825–15837 (2022). [DOI] [PubMed] [Google Scholar]
- 73.Kang YP et al. Non-canonical glutamate-cysteine ligase activity protects against ferroptosis. Cell Metab 33, 174–189.e7 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Chen L et al. NADPH production by the oxidative pentose-phosphate pathway supports folate metabolism. Nat. Metab 1, 404–415 (2019). [PMC free article] [PubMed] [Google Scholar]
- 75.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 23, 225–235 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Yan B et al. Membrane damage during ferroptosis is caused by oxidation of phospholipids catalyzed by the oxidoreductases POR and CYB5R1. Mol. Cell 81, 355–369.e10 (2021). [DOI] [PubMed] [Google Scholar]
- 77.Zou Y et al. Cytochrome P450 oxidoreductase contributes to phospholipid peroxidation in ferroptosis. Nat. Chem. Biol 16, 302–309 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Ding CC et al. MESH1 is a cytosolic NADPH phosphatase that regulates ferroptosis. Nat. Metab 2, 270–277 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Nguyen KT et al. The MARCHF6 E3 ubiquitin ligase acts as an NADPH sensor for the regulation of ferroptosis. Nat. Cell Biol 24, 1239–1251 (2022). [DOI] [PubMed] [Google Scholar]
- 80.Yagoda N et al. RAS-RAF-MEK-dependent oxidative cell death involving voltage-dependent anion channels. Nature 447, 864–868 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Gaschler MM et al. Determination of the subcellular localization and mechanism of action of ferrostatins in suppressing ferroptosis. ACS Chem. Biol 13, 1013–1020 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Bogacz M & Krauth-Siegel RL Tryparedoxin peroxidase-deficiency commits trypanosomes to ferroptosis-type cell death. eLife 7, e37503 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Guo Y et al. Small molecule agonist of mitochondrial fusion repairs mitochondrial dysfunction. Nat. Chem. Biol 19, 468–477 (2023). [DOI] [PubMed] [Google Scholar]
- 84.Ahola S et al. OMA1-mediated integrated stress response protects against ferroptosis in mitochondrial cardiomyopathy. Cell Metab 34, 1875–1891.e7 (2022). [DOI] [PubMed] [Google Scholar]
- 85.Guerra RM & Pagliarini DJ Coenzyme Q biochemistry and biosynthesis. Trends Biochem. Sci 48, 463–476 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Deshwal S et al. Mitochondria regulate intracellular coenzyme Q transport and ferroptotic resistance via STARD7. Nat. Cell Biol 25, 246–257 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Blomme A et al. 2,4-Dienoyl-CoA reductase regulates lipid homeostasis in treatment-resistant prostate cancer. Nat. Commun 11, 2508 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Nassar ZD et al. Human DECR1 is an androgen-repressed survival factor that regulates PUFA oxidation to protect prostate tumor cells from ferroptosis. eLife 9, e54166 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Gao M et al. Role of mitochondria in ferroptosis. Mol. Cell 73, 354–363.e3 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Conlon M et al. A compendium of kinetic modulatory profiles identifies ferroptosis regulators. Nat. Chem. Biol 17, 665–674 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Zhang T et al. ENO1 suppresses cancer cell ferroptosis by degrading the mRNA of iron regulatory protein 1. Nat. Cancer 3, 75–89 (2022). [DOI] [PubMed] [Google Scholar]
- 92.Alvarez SW et al. NFS1 undergoes positive selection in lung tumours and protects cells from ferroptosis. Nature 551, 639–643 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Kalkavan H et al. Sublethal cytochrome c release generates drug-tolerant persister cells. Cell 185, 3356–3374.e22 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Tarangelo A et al. Nucleotide biosynthesis links glutathione metabolism to ferroptosis sensitivity. Life Sci. Alliance 5, e202101157 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Wu S et al. A ferroptosis defense mechanism mediated by glycerol-3-phosphate dehydrogenase 2 in mitochondria. Proc. Natl Acad. Sci. USA 119, e2121987119 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Yang C et al. De novo pyrimidine biosynthetic complexes support cancer cell proliferation and ferroptosis defence. Nat. Cell Biol 25, 836–847 (2023). [DOI] [PubMed] [Google Scholar]
- 97.Wang F et al. PALP: a rapid imaging technique for stratifying ferroptosis sensitivity in normal and tumor tissues in situ. Cell Chem. Biol 29, 157–170.e6 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Yang WS et al. Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis. Proc. Natl Acad. Sci. USA 113, E4966–E4975 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Ubellacker JM et al. Lymph protects metastasizing melanoma cells from ferroptosis. Nature 585, 113–118 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Mann J et al. Ferroptosis inhibition by oleic acid mitigates iron-overload-induced injury. Cell Chem. Biol 10.1016/j.chembiol.2023.10.012 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Perez MA, Magtanong L, Dixon SJ & Watts JL Dietary lipids induce ferroptosis in Caenorhabditis elegans and human cancer cells. Dev. Cell 54, 447–454.e4 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Lee JY et al. Polyunsaturated fatty acid biosynthesis pathway determines ferroptosis sensitivity in gastric cancer. Proc. Natl Acad. Sci. USA 117, 32433–32442 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Yamane D et al. FADS2-dependent fatty acid desaturation dictates cellular sensitivity to ferroptosis and permissiveness for hepatitis C virus replication. Cell Chem. Biol 29, 799–810.e4 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Xin S et al. MS4A15 drives ferroptosis resistance through calcium-restricted lipid remodeling. Cell Death Differ 29, 670–686 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Dixon SJ et al. Human haploid cell genetics reveals roles for lipid metabolism genes in nonapoptotic cell death. ACS Chem. Biol 10, 1604–1609 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Doll S et al. ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat. Chem. Biol 13, 91–98 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Bartolacci C et al. Targeting de novo lipogenesis and the Lands cycle induces ferroptosis in KRAS-mutant lung cancer. Nat. Commun 13, 4327 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Radif Y et al. The endogenous subcellular localisations of the long chain fatty acid-activating enzymes ACSL3 and ACSL4 in sarcoma and breast cancer cells. Mol. Cell Biochem 448, 275–286 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Kuch EM et al. Differentially localized acyl-CoA synthetase 4 isoenzymes mediate the metabolic channeling of fatty acids towards phosphatidylinositol. Biochim. Biophys. Acta 1841, 227–239 (2014). [DOI] [PubMed] [Google Scholar]
- 110.Poppelreuther M et al. The N-terminal region of acyl-CoA synthetase 3 is essential for both the localization on lipid droplets and the function in fatty acid uptake. J. Lipid Res 53, 888–900 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Zhu XG et al. CHP1 regulates compartmentalized glycerolipid synthesis by activating GPAT4. Mol. Cell 74, 45–58.e7 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Beharier O et al. PLA2G6 guards placental trophoblasts against ferroptotic injury. Proc. Natl Acad. Sci. USA 117, 27319–27328 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Sun WY et al. Phospholipase iPLA2β averts ferroptosis by eliminating a redox lipid death signal. Nat. Chem. Biol 17, 465–476 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Chen D et al. iPLA2β-mediated lipid detoxification controls p53-driven ferroptosis independent of GPX4. Nat. Commun 12, 3644 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Reed A et al. LPCAT3 inhibitors remodel the polyunsaturated phospholipid content of human cells and protect from ferroptosis. ACS Chem. Biol 17, 1607–1618 (2022). [DOI] [PubMed] [Google Scholar]
- 116.Liang D et al. Ferroptosis surveillance independent of GPX4 and differentially regulated by sex hormones. Cell 186, 2748–2794.e22 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Rodencal J et al. Sensitization of cancer cells to ferroptosis coincident with cell cycle arrest. Cell Chem. Biol 10.1016/j.chembiol.2023.10.011 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Zhao Y et al. Identification and characterization of a major liver lysophosphatidylcholine acyltransferase. J. Biol. Chem 283, 8258–8265 (2008). [DOI] [PubMed] [Google Scholar]
- 119.Mishra RS, Carnevale KA & Cathcart MK iPLA2β: front and center in human monocyte chemotaxis to MCP-1. J. Exp. Med 205, 347–359 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Friedmann Angeli JP et al. 7-Dehydrocholesterol is an endogenous suppressor of ferroptosis. Preprint at Res. Sq 10.21203/rs.3.rs-943221/v1 (2021). [DOI] [Google Scholar]
- 121.Yamada N et al. DHCR7 as a novel regulator of ferroptosis in hepatocytes. Preprint at bioRxiv 10.1101/2022.06.15.496212 (2022). [DOI] [Google Scholar]
- 122.Garcia-Bermudez J et al. Squalene accumulation in cholesterol auxotrophic lymphomas prevents oxidative cell death. Nature 567, 118–122 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Schumacher MM & DeBose-Boyd RA Posttranslational regulation of HMG CoA reductase, the rate-limiting enzyme in synthesis of cholesterol. Annu. Rev. Biochem 90, 659–679 (2021). [DOI] [PubMed] [Google Scholar]
- 124.Conrad M & Proneth B Selenium: tracing another essential element of ferroptotic cell death. Cell Chem. Biol 27, 409–419 (2020). [DOI] [PubMed] [Google Scholar]
- 125.Yi J, Zhu J, Wu J, Thompson CB & Jiang X Oncogenic activation of PI3K-AKT-mTOR signaling suppresses ferroptosis via SREBP-mediated lipogenesis. Proc. Natl Acad. Sci. USA 117, 31189–31197 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Vangala JR, Sotzny F, Kruger E, Deshaies RJ & Radhakrishnan SK Nrf1 can be processed and activated in a proteasome-independent manner. Curr. Biol 26, R834–R835 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Tomlin FM et al. Inhibition of NGLY1 inactivates the transcription factor Nrf1 and potentiates proteasome inhibitor cytotoxicity. ACS Cent. Sci 3, 1143–1155 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Chavarria C et al. ER-trafficking triggers NRF1 ubiquitination to promote its proteolytic activation. iScience 26, 107777 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Dirac-Svejstrup AB et al. DDI2 Is a ubiquitin-directed endoprotease responsible for cleavage of transcription factor NRF1. Mol. Cell 79, 332–341.e7 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Forcina GC et al. Ferroptosis regulation by the NGLY1/NFE2L1 pathway. Proc. Natl Acad. Sci. USA 119, e2118646119 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Kotschi S et al. NFE2L1-mediated proteasome function protects from ferroptosis. Mol. Metab 57, 101436 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Song W, Zhang W, Yue L & Lin W Revealing the effects of endoplasmic reticulum stress on ferroptosis by two-channel real-time imaging of pH and viscosity. Anal. Chem 94, 6557–6565 (2022). [DOI] [PubMed] [Google Scholar]
- 133.Danielli M, Perne L, Jarc Jovicic E & Petan T Lipid droplets and polyunsaturated fatty acid trafficking: balancing life and death. Front. Cell Dev. Biol 11, 1104725 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Olzmann JA & Carvalho P Dynamics and functions of lipid droplets. Nat. Rev. Mol. Cell Biol 20, 137–155 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Dierge E et al. Peroxidation of n-3 and n-6 polyunsaturated fatty acids in the acidic tumor environment leads to ferroptosis-mediated anticancer effects. Cell Metab 33, 1701–1715.e5 (2021). [DOI] [PubMed] [Google Scholar]
- 136.Minami JK et al. CDKN2A deletion remodels lipid metabolism to prime glioblastoma for ferroptosis. Cancer Cell 41, 1048–1060.e9 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Lee H et al. Cell cycle arrest induces lipid droplet formation and confers ferroptosis resistance. Nat. Commun 15, 79 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Zou Y et al. A GPX4-dependent cancer cell state underlies the clear-cell morphology and confers sensitivity to ferroptosis. Nat. Commun 10, 1617 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Ferrada L, Barahona MJ, Vera M, Stockwell BR & Nualart F Dehydroascorbic acid sensitizes cancer cells to system xc− inhibition-induced ferroptosis by promoting lipid droplet peroxidation. Cell Death Dis 14, 637 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Mohammadyani D et al. Molecular speciation and dynamics of oxidized triacylglycerols in lipid droplets: mass spectrometry and coarse-grained simulations. Free Radic. Biol. Med 76, 53–60 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Perez MA et al. Ether lipid deficiency disrupts lipid homeostasis leading to ferroptosis sensitivity. PLoS Genet 18, e1010436 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Alborzinia H et al. Golgi stress mediates redox imbalance and ferroptosis in human cells. Commun. Biol 1, 210 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Huang Y et al. UBIAD1 alleviates ferroptotic neuronal death by enhancing antioxidative capacity by cooperatively restoring impaired mitochondria and Golgi apparatus upon cerebral ischemic/reperfusion insult. Cell Biosci 12, 42 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Nakagawa K et al. Identification of UBIAD1 as a novel human menaquinone-4 biosynthetic enzyme. Nature 468, 117–121 (2010). [DOI] [PubMed] [Google Scholar]
- 145.Rost S et al. Mutations in VKORC1 cause warfarin resistance and multiple coagulation factor deficiency type 2. Nature 427, 537–541 (2004). [DOI] [PubMed] [Google Scholar]
- 146.Jin DY et al. A genome-wide CRISPR-Cas9 knockout screen identifies FSP1 as the warfarin-resistant vitamin K reductase. Nat. Commun 14, 828 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Yang X et al. Regulation of VKORC1L1 is critical for p53-mediated tumor suppression through vitamin K metabolism.Cell Metab 35, 1474–1490.e8 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Mugoni V et al. Ubiad1 is an antioxidant enzyme that regulates eNOS activity by CoQ10 synthesis. Cell 152, 504–518 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Jo Y & DeBose-Boyd RA Post-translational regulation of HMG CoA reductase. Cold Spring Harb. Perspect. Biol 14, a041253 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Torii S et al. An essential role for functional lysosomes in ferroptosis of cancer cells. Biochem. J 473, 769–777 (2016). [DOI] [PubMed] [Google Scholar]
- 151.Hou W et al. Autophagy promotes ferroptosis by degradation of ferritin. Autophagy 12, 1425–1428 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Gao M et al. Ferroptosis is an autophagic cell death process. Cell Res 26, 1021–1032 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Gryzik M, Asperti M, Denardo A, Arosio P & Poli M NCOA4-mediated ferritinophagy promotes ferroptosis induced by erastin, but not by RSL3 in HeLa cells. Biochim. Biophys. Acta Mol. Cell Res 1868, 118913 (2021). [DOI] [PubMed] [Google Scholar]
- 154.Anandhan A et al. NRF2 controls iron homeostasis and ferroptosis through HERC2 and VAMP8. Sci. Adv 9, eade9585 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Wu Z et al. Chaperone-mediated autophagy is involved in the execution of ferroptosis. Proc. Natl Acad. Sci. USA 116, 2996–3005 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Wu K et al. Creatine kinase B suppresses ferroptosis by phosphorylating GPX4 through a moonlighting function. Nat. Cell Biol 25, 714–725 (2023). [DOI] [PubMed] [Google Scholar]
- 157.Dixon SJ & Stockwell BR The role of iron and reactive oxygen species in cell death. Nat. Chem. Biol 10, 9–17 (2014). [DOI] [PubMed] [Google Scholar]
- 158.Swanda RV et al. Lysosomal cystine governs ferroptosis sensitivity in cancer via cysteine stress response. Mol. Cell 10, 3347–3359.e9 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Zhang Y et al. mTORC1 couples cyst(e)ine availability with GPX4 protein synthesis and ferroptosis regulation. Nat. Commun 12, 1589 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Elmore S Apoptosis: a review of programmed cell death. Toxicol. Pathol 35, 495–516 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.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 3, 285–296 (2003). [DOI] [PubMed] [Google Scholar]
- 162.Vaca CE & Harms-Ringdahl M Nuclear membrane lipid peroxidation products bind to nuclear macromolecules. Arch. Biochem. Biophys 269, 548–554 (1989). [DOI] [PubMed] [Google Scholar]
- 163.Zhang Y et al. Imidazole ketone erastin induces ferroptosis and slows tumor growth in a mouse lymphoma model. Cell Chem. Biol 26, 623–633.e9 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Tarangelo A et al. p53 suppresses metabolic stress-induced ferroptosis in cancer cells. Cell Rep 22, 569–575 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Muller F et al. Elevated FSP1 protects KRAS-mutated cells from ferroptosis during tumor initiation. Cell Death Differ 30, 442–456 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Koppula P et al. A targetable CoQ-FSP1 axis drives ferroptosis- and radiation-resistance in KEAP1 inactive lung cancers. Nat. Commun 13, 2206 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Takahashi N et al. 3D culture models with CRISPR screens reveal hyperactive NRF2 as a prerequisite for spheroid formation via regulation of proliferation and ferroptosis. Mol. Cell 80, 828–844.e6 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Kuang F, Liu J, Xie Y, Tang D & Kang R MGST1 is a redox-sensitive repressor of ferroptosis in pancreatic cancer cells. Cell Chem. Biol 28, 765–775.e5 (2021). [DOI] [PubMed] [Google Scholar]
- 169.Viswanathan VS et al. Dependency of a therapy-resistant state of cancer cells on a lipid peroxidase pathway. Nature 547, 453–457 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Wang W et al. CD8+ T cells regulate tumour ferroptosis during cancer immunotherapy. Nature 569, 270–274 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Wang L et al. ATF3 promotes erastin-induced ferroptosis by suppressing system Xc−. Cell Death Differ 27, 662–675 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Wu J et al. Intercellular interaction dictates cancer cell ferroptosis via NF2-YAP signalling. Nature 572, 402–406 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Venkatesh D et al. MDM2 and MDMX promote ferroptosis by PPARα-mediated lipid remodeling. Genes Dev 34, 526–543 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Liu J et al. NUPR1 is a critical repressor of ferroptosis. Nat. Commun 12, 647 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Alborzinia H et al. MYCN mediates cysteine addiction and sensitizes neuroblastoma to ferroptosis. Nat. Cancer 3, 471–485 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Floros KV et al. MYCN-amplified neuroblastoma is addicted to iron and vulnerable to inhibition of the system Xc−/glutathione axis. Cancer Res 81, 1896–1908 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Zhang Y, Koppula P & Gan B Regulation of H2A ubiquitination and SLC7A11 expression by BAP1 and PRC1. Cell Cycle 18, 773–783 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Valente LJ et al. p53 deficiency triggers dysregulation of diverse cellular processes in physiological oxygen. J. Cell Biol 219, e201908212 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Wohlhieter CA et al. Concurrent mutations in STK11 and KEAP1 promote ferroptosis protection and SCD1 dependence in lung cancer. Cell Rep 33, 108444 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Zhao Y et al. HCAR1/MCT1 regulates tumor ferroptosis through the lactate-mediated AMPK-SCD1 activity and its therapeutic implications. Cell Rep 33, 108487 (2020). [DOI] [PubMed] [Google Scholar]
- 181.Tesfay L et al. Stearoyl-CoA desaturase 1 protects ovarian cancer cells from ferroptotic cell death. Cancer Res 79, 5355–5366 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Lee H et al. Energy-stress-mediated AMPK activation inhibits ferroptosis. Nat. Cell Biol 22, 225–234 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Acoba MG, Senoo N & Claypool SM Phospholipid ebb and flow makes mitochondria go. J. Cell Biol 219, e202023131 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Barger SR, Penfield L & Bahmanyar S Coupling lipid synthesis with nuclear envelope remodeling. Trends Biochem. Sci 47, 52–65 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Zhang HL et al. PKCβII phosphorylates ACSL4 to amplify lipid peroxidation to induce ferroptosis. Nat. Cell Biol 24, 88–98 (2022). [DOI] [PubMed] [Google Scholar]
- 186.Wu Y et al. Caveolae sense oxidative stress through membrane lipid peroxidation and cytosolic release of CAVIN1 to regulate NRF2. Dev. Cell 58, 376–397.e4 (2023). [DOI] [PubMed] [Google Scholar]
- 187.Cui S et al. Identification of hyperoxidized PRDX3 as a ferroptosis marker reveals ferroptotic damage in chronic liver diseases. Mol. Cell 83, 3931–3939.e5 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Rodencal J & Dixon SJ Positive feedback amplifies ferroptosis. Nat. Cell Biol 24, 4–5 (2022). [DOI] [PubMed] [Google Scholar]
- 189.Brown CW et al. Prominin2 drives ferroptosis resistance by stimulating iron export. Dev. Cell 51, 575–586.e4 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Zhao Y et al. Neutrophils resist ferroptosis and promote breast cancer metastasis through aconitate decarboxylase 1. Cell Metab 35, 1688–1703.e10 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Feng H et al. Transferrin receptor is a specific ferroptosis marker. Cell Rep 30, 3411–3423.e7 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Jia M et al. Redox homeostasis maintained by GPX4 facilitates STING activation. Nat. Immunol 21, 727–735 (2020). [DOI] [PubMed] [Google Scholar]
- 193.Xie Y et al. The tumor suppressor p53 limits ferroptosis by blocking DPP4 activity. Cell Rep 20, 1692–1704 (2017). [DOI] [PubMed] [Google Scholar]
- 194.Horton JD, Goldstein JL & Brown MS SREBPs: activators of the complete program of cholesterol and fatty acid synthesis in the liver. J. Clin. Invest 109, 1125–1131 (2002). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Tian R et al. Genome-wide CRISPRi/a screens in human neurons link lysosomal failure to ferroptosis. Nat. Neurosci 24, 1020–1034 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Ryan SK et al. Microglia ferroptosis is regulated by SEC24B and contributes to neurodegeneration. Nat. Neurosci 26, 12–26 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Linkermann A et al. Synchronized renal tubular cell death involves ferroptosis. Proc. Natl Acad. Sci. USA 111, 16836–16841 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Brown CW, Amante JJ & Mercurio AM Cell clustering mediated by the adhesion protein PVRL4 is necessary for α6β4 integrin-promoted ferroptosis resistance in matrix-detached cells. J. Biol. Chem 293, 12741–12748 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Dar HH et al. Pseudomonas aeruginosa utilizes host polyunsaturated phosphatidylethanolamines to trigger theft-ferroptosis in bronchial epithelium. J. Clin. Invest 128, 4639–4653 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Belavgeni A et al. vPIF-1 is an insulin-like antiferroptotic viral peptide. Proc. Natl Acad. Sci. USA 120, e2300320120 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Jin J et al. Machine learning classifies ferroptosis and apoptosis cell death modalities with TfR1 immunostaining. ACS Chem. Biol 17, 654–660 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Shimada K et al. Global survey of cell death mechanisms reveals metabolic regulation of ferroptosis. Nat. Chem. Biol 12, 497–503 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Murphy TH, Miyamoto M, Sastre A, Schnaar RL & Coyle JT Glutamate toxicity in a neuronal cell line involves inhibition of cystine transport leading to oxidative stress. Neuron 2, 1547–1558 (1989). [DOI] [PubMed] [Google Scholar]
- 204.Tan S, Schubert D & Maher P Oxytosis: a novel form of programmed cell death. Curr. Top. Med. Chem 1, 497–506 (2001). [DOI] [PubMed] [Google Scholar]
- 205.Ratan RR, Murphy TH & Baraban JM Macromolecular synthesis inhibitors prevent oxidative stress-induced apoptosis in embryonic cortical neurons by shunting cysteine from protein synthesis to glutathione. J. Neurosci 14, 4385–4392 (1994). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Wiernicki B et al. Excessive phospholipid peroxidation distinguishes ferroptosis from other cell death modes including pyroptosis. Cell Death Dis 11, 922 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Skouta R et al. Ferrostatins inhibit oxidative lipid damage and cell death in diverse disease models. J. Am. Chem. Soc 136, 4551–4556 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Poon JF, Zilka O & Pratt DA Potent ferroptosis inhibitors can catalyze the cross-dismutation of phospholipid-derived peroxyl radicals and hydroperoxyl radicals. J. Am. Chem. Soc 142, 14331–14342 (2020). [DOI] [PubMed] [Google Scholar]
- 209.Hadian K & Stockwell BR A roadmap to creating ferroptosis-based medicines. Nat. Chem. Biol 17, 1113–1116 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Yang WS et al. Regulation of ferroptotic cancer cell death by GPX4. Cell 156, 317–331 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Hendricks JM et al. Identification of structurally diverse FSP1 inhibitors that sensitize cancer cells to ferroptosis. Cell Chem. Biol 30, 1090–1103.e7 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Nakamura T et al. Phase separation of FSP1 promotes ferroptosis. Nature 619, 371–377 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Farmer LA et al. Intrinsic and extrinsic limitations to the design and optimization of inhibitors of lipid peroxidation and associated cell death. J. Am. Chem. Soc 144, 14706–14721 (2022). [DOI] [PubMed] [Google Scholar]
- 214.Jelinek A et al. Mitochondrial rescue prevents glutathione peroxidase-dependent ferroptosis. Free Radic. Biol. Med 117, 45–57 (2018). [DOI] [PubMed] [Google Scholar]
- 215.Fang X et al. Ferroptosis as a target for protection against cardiomyopathy. Proc. Natl Acad. Sci. USA 116, 2672–2680 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Merkel M et al. Mitochondrial reactive oxygen species formation determines ACSL4/LPCAT2-mediated ferroptosis. Antioxidants 12, 1590 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Doulias PT, Christoforidis S, Brunk UT & Galaris D Endosomal and lysosomal effects of desferrioxamine: protection of HeLa cells from hydrogen peroxide-induced DNA damage and induction of cell-cycle arrest. Free Radic. Biol. Med 35, 719–728 (2003). [DOI] [PubMed] [Google Scholar]
- 218.Kurz T, Gustafsson B & Brunk UT Intralysosomal iron chelation protects against oxidative stress-induced cellular damage. FEBS J 273, 3106–3117 (2006). [DOI] [PubMed] [Google Scholar]
- 219.Fang Y et al. Inhibiting ferroptosis through disrupting the NCOA4-FTH1 interaction: a new mechanism of action. ACS Cent. Sci 7, 980–989 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Xu Y, Li X, Cheng Y, Yang M & Wang R Inhibition of ACSL4 attenuates ferroptotic damage after pulmonary ischemia-reperfusion. FASEB J 34, 16262–16275 (2020). [DOI] [PubMed] [Google Scholar]
- 221.Li Y et al. Ischemia-induced ACSL4 activation contributes to ferroptosis-mediated tissue injury in intestinal ischemia/reperfusion. Cell Death Differ 26, 2284–2299 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Ide S et al. Sex differences in resilience to ferroptosis underlie sexual dimorphism in kidney injury and repair. Cell Rep 41, 111610 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Yan H et al. Discovery of decreased ferroptosis in male colorectal cancer patients with KRAS mutations. Redox Biol 62, 102699 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Leu JI, Murphy ME & George DL Functional interplay among thiol-based redox signaling, metabolism, and ferroptosis unveiled by a genetic variant of TP53. Proc. Natl Acad. Sci. USA 117, 26804–26811 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
