Summary
Ferroptosis is a type of regulated cell death that drives the pathophysiology of many diseases. Oxidative stress is detectable in many types of regulated cell death, but only ferroptosis involves lipid peroxidation and iron dependency. Ferroptosis originates and propagates from several organelles, including the mitochondria, endoplasmic reticulum (ER), the Golgi, and lysosomes. Recent data has revealed that immune cells can both induce and undergo ferroptosis. A mechanistic understanding of how ferroptosis regulates immunity is critical to understand how ferroptosis controls immune responses, and how this is dysregulated in disease. Translationally, more work is needed to produce ferroptosis-modulating immunotherapeutics. This review focuses on the role of ferroptosis in immune-related diseases, including infection, autoimmune diseases, and cancer. We discuss how ferroptosis is regulated in immunity, how this regulation contributes to disease pathogenesis, and how targeting ferroptosis may lead to novel therapies.
Keywords: Ferroptosis, apoptosis, pyroptosis, T cell, interferon, glutathione, system xc-, SLC7A11, ACSL4, FSP1, immune checkpoint blockade, cancer therapy
eTOC/In Brief:
This review focuses on the role of ferroptosis in modulating immune-related diseases. It describes how ferroptosis functions in autoimmune disease, infection, and tumor, and discusses opportunities to translate basic discoveries in ferroptosis to clinical treatments.
Ferroptosis is a distinct form of regulated cell death highlighted by the contributions of lipid peroxidation and iron. In this review, Bell and colleagues summarize and interpret the literature on mechanism and role of ferroptosis in the context of immune homeostasis and immune-related diseases.
Introduction
Cell death is a fundamental biological process essential for survival that regulates development, tissue homeostasis, and disease. Regulated cell death eliminates unneeded cells and cells damaged by infection, mutation, or old age1. Spatial, temporal, and mechanistic regulation of cell death are critical checkpoints for preventing human illness. Regulated cell death can be either immunostimulatory or immunoinhibitory, but the overall goal is the systematic removal of cells targeted to die without harming nearby neighbors2,3. Immunogenic cell death can be mediated by changes in membrane proteins on the cell surface alongside the release of soluble molecules. Together, these signals stimulate antigen presenting cells (APCs), such as macrophages and dendritic cells (DCs), to present antigens to T cells and induce adaptive immunity3. How different cell death stimuli contribute to divergent cell surface markers and secreted proteomes is an active area of exploration4,5. Regardless of immunogenicity, failure of the balance between pro-survival and death-inducing signals is a hallmark of infection, autoimmune disease, cancer, and organismal development. This dynamic and responsive balance is also critical in the physiological regulation of the immune system. Regulated cell death of non-immune cells modulates the immune response to pathogens, controls damage inflicted by autoimmunity, and regulates anti-tumor immunity2,6–11. Regulated cell death of immune cells regulates the enrichment and specificity of T cell responses, stimulates antiviral defense, regulates the inflammatory response, mediates immune tolerance, and generates protective immunity to tumor and pathogens2,6–8,11.
Ferroptosis is a form of regulated cell death driven by iron, reactive oxygen species, and lipid peroxidation of cell membrane lipids12. Although many forms of cell death have oxidative features, ferroptosis is morphologically and molecularly distinct. Ferroptotic cells have shrunken mitochondria with fewer cristae, reduced cellular volume with normal nuclear volume, and an accumulation of membrane lipid peroxides13,14. The lipid peroxidation of the plasma membrane is a final event in ferroptosis15. In contrast to other forms of regulated cell death, there are no discovered overarching ferroptotic “execution genes.” Ferroptosis is instead regulated by a complex interplay between death-promoting signals, which are often metabolic, and defense systems, some of which rely on metabolites, such as cystine and glutathione (GSH). Although ferroptosis still needs additional specific “smoking gun” markers, molecular in vitro studies and in vivo use of inhibitors and inducers have begun to reveal its biological role. A missing piece of ferroptosis research, endogenous ferroptosis inducers that do not require synthetic chemical compounds, has recently been identified. Ferroptosis can be induced by pro-inflammatory cytokines, such as interferon-γ (IFNγ), and some fatty acids, such as arachidonic acid, in the tumor microenvironment (TME); in addition, polyunsaturated fatty acids (PUFAs) or iron can trigger ferroptosis in some contexts16. Endogenous glutamate accumulation after system xc- inhibition can determine ferroptosis sensitivity by suppressing the yes-associated protein (YAP) pathway17. The search for other endogenous mediators of ferroptosis is ongoing.
Regulated cell death is essential for organ and tissue development. In development, regulated cell death has been viewed as predominantly executed through apoptosis. Nonetheless, the emerging understanding of ferroptosis highlights that although ferroptosis lacks the characteristic biochemical and genetic executioners of apoptosis, it plays a role in development. The emerging roles of ferroptosis in tissue homeostasis and the increasing evidence supporting a role for ferroptosis in many pathologies has driven a flurry of recent research. Compelling evidence demonstrates that ferroptosis is a regulated form of cell death—a cell death process mediated by a cell’s microenvironment and may be regulated in some contexts.
Ferroptosis is regulated by the accumulation of iron, reactive oxygen species, and lipids with susceptibility to peroxidation. Defense against ferroptosis is primarily controlled by the activity and protein level of the glutathione peroxidase (GPX)418. GPX4 is a selenoprotein that is the only GPX family member that can detoxify lipid hydroperoxides embedded into the cellular membrane19,20. Inactivation of GPX4 leads to lipid peroxidation and cell death in many cell types21,22. GPX4 itself is essential in preventing lipid oxidation induced renal failure and death in non-transformed murine models23. GPX4 is the primary protein responsible for preventing the final execution of ferroptotic cell death18–20. Lipid peroxidation can originate from damage to mitochondria, the lysosome, or the endoplasmic reticulum (ER)24.
We highlight here the mitochondria as an essential hub for the terminal execution of ferroptosis. The mitochondria plays an essential role in many forms of regulated cell death, including ferroptosis, apoptosis, and pyroptosis. In pyroptosis, gasdermin D permeabilizes the mitochondria25. In apoptosis, mitochondrial outer membrane permeabilization releases cytochrome c and initiates caspase activation26. In ferroptosis, cystine deprivation results in mitochondrial lipid peroxide accumulation, while inhibiting the electron transport chain prevents lipid peroxidation and ferroptosis27. Potentially, the mitochondria are an underappreciated node for modulation and crosstalk between different types of regulated cell death15.
Ferroptosis has been identified in cultured cell lines and in mice as death that is inhibited by iron chelation, lipophilic antioxidants, lipid peroxidation inhibitors, by GPX4, or by the depletion of PUFAs12,14,21,28. Ferroptosis was initially defined through the study of synthetic small molecule cell death inducers, such as erastin and Ras-selective-lethal 3 (RSL3). Erastin, sulfasalazine, quisqualic acid, and (S)-4-carboxyphenylglycine selectively inhibit the cystine importer system xc-, while RSL3 inhibits GPX418,29,30. It has also been suggested that RSL3 acts through inhibition of another selenoprotein thioredoxin reductase 1 (TXNRD1) or that it requires 14-3-3ε activity31,32. Other work suggests that the activity of RSL3 is not specific, but that RSL3 can target a variety of selenoproteins33. This screen reveals that. RSL3 may function through many mediators, including voltage dependent anion selective channel protein 2 (VDAC2)33. Ferroptosis plays a role in several diseases9,16,23,34–41. In this Review, we focus on the interplay between ferroptosis and immunity to better understand the implications of ferroptosis in infectious disease, autoimmunity, and cancer.
Ferroptotic Mechanisms of Action
The mechanisms and regulators of ferroptosis have been discussed extensively in previous reviews24,42. We present an abbreviated description of its basic biology and mechanisms of regulation. Ferroptosis is defined as an oxidative and iron-dependent form of cell death12,28,43. Genetic inactivation of GPX4 is similarly found to induce a non-apoptotic form of oxidative cell death21. GPX4 and system xc− are the primary ferroptosis defense proteins42. As the only enzyme that can detoxify lipid peroxides embedded in cellular membranes, GPX4 is critical for cellular defense against lipid peroxidation and cell death20,44. Genetic deletion or chemical inhibition of GPX4 induces ferroptosis (Figure 1a).21,28. System xc− is the predominant amino acid antiporter that imports L-cystine, through the transporter subunit solute carrier family 7 member 11 (SLC7a11) in exchange for glutamate (Figure 1a)45,46. Cystine is converted to cysteine intracellularly through a nicotinamide-adenine-dinucleotide-phosphate-(NADPH)-consuming reduction reaction and can then be used to synthesize GSH, an important cellular cofactor for antioxidant systems47. Depleting cystine availability or blocking SLC7a11 through erastin induces ferroptosis21,28,48. Ferroptosis is executed by the peroxidation of PUFA moieties in phospholipids and ether lipids, which results in membrane permeabilization and ferroptotic death. Acyl-CoA synthase long-chain family member 4 (ACSL4) and lysophosphatidylcholine acyltransferase 3 (LPCAT3) enrich cellular membranes with PUFAs49. Genetic loss of Acsl4 or pharmacological inhibition of ACSL4 induces resistance to ferroptosis (Figure 1a)49. Treatment of cells or animals with PUFAs containing deuterium in place of hydrogen at reactive bisallylic sites can also protect against ferroptosis by preventing lipid peroxidation50,51.
Figure 1: Ferroptosis mechanisms and sites.

a) An overview of ferroptosis. b) Schematic depicting the predominant endogenous lipid peroxidation defense systems. c) Schematic illustrating the function of various organelles in ferroptosis.
High iron concentrations or low concentrations of cystine or GSH tip the cells to a pro-ferroptotic state and can then lead to multiorgan failure24,52. Increased ACSL4, or decreased expression of GPX4 or SLC7a11 favor ferroptosis24. Selenium drives GPX4 translation and is protective against ferroptosis, as is iron chelation through deferoxamine53,54. Selenium augments the transcription of GPX4 and other genes in a transcriptional program, the selenome, through the activation of transcription factors TFAP2c and Sp153. Selenome regulation is critical in neurons during hemorrhagic stroke, but the role of the selenome in immunity is currently underexplored53.
Nuclear receptor coactivator 4 (NCOA4) regulates ferroptosis through ferritinophagy, the delivery of ferritin to lysosomes for iron recycling55. NCOA4-deficient cells cannot degrade ferritin, and therefore have decreased intracellular iron55. Modulating NCOA4-mediated ferritonophagic flux regulates ferroptosis56. Other aspects of cancer metabolism also control sensitivity to ferroptosis. For example, glutamate oxaloacetate transaminase 1 (GOT1) inhibition promotes pancreatic cancer cell death via ferroptosis, highlighting a potential targetable vulnerability of tumor cells57. Inactivation of dihydroorotate dehydrogenase (DHODH) was suggested to induce mitochondrial lipid peroxidation and ferroptosis in GPX4-low cancer cells and to synergize with small molecule ferroptosis inducers in GPX4-high cancer cells (Figure 1a)58. The role of DHODH has recently been questioned59. DHODH likely operates alongside mitochondrial GPX4 to inhibit ferroptosis in the mitochondrial inner membrane by reducing ubiquinone to ubiquinol, a powerful antioxidant58. Ferroptosis suppressor protein 1 (FSP1) protects against ferroptosis induced by GPX4 deletion60,61. FSP1 catalyzes the regeneration of ubiquinone using NADPH. FSP1 inhibition synergizes with GPX4 inhibitors, suggesting that the FSP1-Coenzyme-Q10-NADPH axis is an independent system to prevent ferroptosis61. This pathway provides a potential explanation for why pharmacological targeting of GPX4 may not achieve major antitumor effects, due to FSP1-mediated protection.
The mitochondrial role of ferroptosis was recently strengthened by the identification of hyperoxidized peroxiredoxin 3 (PRDX3) as a ferroptotic marker in vivo and in vitro62. PRDX3 hyperoxidation is triggered by the accumulation of lipid peroxides in the mitochondria62. Once hyperoxidized, PRDX3 translocates from the mitochondria to the plasma membrane, where it inhibits cystine uptake and promotes ferroptosis62. Perhaps hyperoxidized PRDX3 facilitates the execution of ferroptosis, supporting the idea that the mitochondria are a key hub for ferroptosis. However, how hyperoxidized PRDX3 is translocated to the plasma membrane, and whether hyperoxidized PRDX3 is present and functionally critical in endogenous ferroptosis in different tissue types remains poorly understood.
We highlight several synthetic inducers and inhibitors of ferroptosis that are commonly used in research (Figure 1a), and endogenous ferroptosis defense systems (highlighted in Figure 1b). Ferroptosis inhibitors, such as ferrostatin-1 and liproxstatin-1 and related compounds, prevent ferroptosis by acting as radical trapping antioxidants and, in some cases, as inhibitors of lipoxygenase53,54,63. Ferroptosis can be defined as cell death that is inhibited by ferrostatin-1 or liproxstatin-1, as well as by iron chelators. This dependence on synthetic chemical inhibition and induction of ferroptosis has led to questions of whether these aspects of ferroptosis are non-physiological, despite the similarity to pan-caspase inhibitors being used to identify apoptotic cell death. This debate may be partly eased by the identification of endogenous ferroptosis inducers, such as activated T cell derived IFNγ plus fatty acids, in the TME16,39. As early as the mid- 1990s, it had been observed that cystine transport activity in murine macrophages was repressed by IFNγ, but only recently has the biological impact of this regulation in tumor immunology been explored16,39,64. Understanding how ferroptosis is regulated in vivo is critical for characterizing the physiological and pathological functions of this cell death process. Ferroptosis seems to be a natural consequence of iron metabolism and oxidation that is prevented by a network of overlapping defense proteins and factors.
Ferroptosis can be regulated through cell-cell contacts. E-cadherin induced by cell-cell contact suppresses ferroptosis by activating intracellular neurofibromatosis type 2 (NF2) and the Hippo signaling pathway65. Removal of the downstream protein, tafazzin, confers ferroptotic resistance, suggesting that this is the end-mediator of cellular-density-regulated ferroptosis66. Genetically inactivating NF2 increases ferroptosis sensitivity, revealing another potential intervention point to translationally drive ferroptosis, particularly in the case of cancer65.
The final stage of ferroptosis is the peroxidation of membrane PUFA-containing lipids, which leads to membrane permeabilization (Figure 1c) and generates a wealth of lipid-derived electrophiles (LDEs). Ferroptosis may also transfer between cells through the interaction of oxidized lipids with adjacent cell plasma membranes, creating wave-like propagation of cell death15,67. According to this model, ferroptotic cell rupture is mediated by plasma membrane pores, and intercellular propagation of ferroptotic death is induced by erastin, but not by direct inhibition of GPX415,67. Propagation of ferroptotic death between cells is upstream of cell rupture and is spread through cellular swelling dependent on lipid peroxide and iron15. However, this mechanism of cell killing has only been demonstrated in cell culture and limited ex vivo settings, so the relevance to in vivo ferroptosis is uncertain. Potentially, a small fraction of tumor cell ferroptotic death induced by immunotherapy could initiate a wave of killing that might induce tumor regression, but this has yet to be experimentally demonstrated. Another possible mechanism for the spread of ferroptosis cell-to-cell is through metabolic adaptation to counteract phospholipid oxidation. Accumulation of 7-dehydrocholesterol shields phospholipids from peroxidation and prevents membrane fragmentation by preferentially reacting with peroxyl radicals68. How the intercellular propagation of cell death and eventual rupture impacts immunity in response to different pathogenic insults is not clear.
A variety of cellular organelles regulate ferroptotic cell death (Figure 1c). The ER is a central site of lipid peroxidation50. The ER membranes have abundant PUFA-PLs, likely explaining their centrality in driving lipid peroxidation during ferroptosis (Figure 1c)50. This suggests an ordered progression of membrane peroxidation from initial peroxidation in the ER and later accumulation in the plasma membrane50. The mitochondria house a likely ferroptosis-inhibiting pathway discussed above mediated by DHODH58. Mitochondria also drive ferroptosis by providing oxidation through the electron transport chain (Figure 1c)58. In fact, inhibiting the electron transport chain or depletion of mitochondria prevents ferroptosis but not ferroptosis induced by inhibiting GPX469,70.
The peroxisome has also been implicated in ferroptosis regulation. A genome wide CRISPR suppressor screen has determined that peroxisomes contribute to ferroptosis by synthesizing polyunsaturated ether phospholipids, a ferroptosis substrate71. Ferroptosis resistance in vivo is driven by the downregulation of polyunsaturated ether phospholipid metabolism71. Targeting peroxisomal lipid biosynthesis may therefore induce or inhibit ferroptosis.
The Golgi’s role in mediating ferroptosis has been considerably less studied. One report demonstrates that Golgi-dispersing compounds, such as brefeldin A and golgicide A, induce ferroptosis72. Inhibitors of ferroptosis prevent cell death induced by these compounds and maintain Golgi function. However, it is unclear how Golgi damage triggers ferroptosis or if ferroptosis affects the integrity of the Golgi. The endosomal recycling compartment has also been implicated in ferroptosis, as transferrin receptor 1 (also known as CD71) localizes to this Golgi-adjacent compartment and is mobilized to the plasma membrane during ferroptosis in many cell types, and thus serves as a marker of ferroptosis73,74.
The lysosome can also mediate ferroptosis in response to specific compounds, such as salinomycin, ironomycin, and related compounds75,76. Inhibition of the lysosome by bafilomycin A1 prevents erastin-induced ferroptosis, although lysosomes do not seem to be needed for ferrostatin-1 rescue of ferroptosis (Figure 1c)77,78. Thus, several cellular organelle membranes are implicated in the initiation, execution, and completion of ferroptosis.
The organellar initiator of ferroptosis is up for debate, but the conclusion that lipid peroxidation is essential for ferroptotic death remains clear. Lipidomics, especially of peroxidized lipids, is technically challenging compared to proteomics or genomics79. This is due to the almost limitless plethora of peroxidation products, the lack of off-the-shelf reagents, the necessity of expensive and technically demanding equipment to quantify and analyze these lipid changes, and the lack of availability of robust, non-chemical ferroptosis models. Only recently have commercial kits to detect lipid peroxidation become available, and these have limited utility due to an inability to identify many peroxidized lipid species. Only a few groups can measure the oxidized lipidome80,81. Lipidomics is just reaching the single cell level but remains challenging due to the impossibility of amplifying lipids82,83.
PUFAs are highly vulnerable to peroxidation and are critical for ferroptotic death49,84,85. In contrast, monounsaturated fatty acids are protective against ferroptosis18,49,84–86. Recent data suggests that ferroptosis involves increased oxidation on a myriad of phospholipid species87. The relative contribution of individual phospholipids is undetermined, providing novel opportunities to counteract ferroptosis, such as through treatment with Vitamin K87. Therefore, fatty acid, ether lipid biosynthesis, and phospholipid remodeling are essential components to fully understand the steps involved in ferroptosis. Understanding lipid metabolism could also help reveal ferroptotic biomarkers. Phosphatidylethanolamine-(18:0/20:4-OOH) and phosphatidylethanolamine-(18:0/22:4-OOH) are possible ferroptosis markers81. These can be oxidatively truncated to PE-(18:0/hydroxy-8-oxo-oct-6-enoic acid), another molecule enriched in cells treated with RSL381. Integrating biological manipulation with lipidomics would be required to understand the biochemical underpinnings of ferroptosis.
Ferroptosis is regulated by lipid composition, iron and cystine availability, and the activity and expression of ferroptosis defense proteins. Ferroptosis requires endogenous metabolites, such as cystine, GSH, and iron. It is not fully understood what in vivo factors determine whether a cell dies through ferroptosis or a different type of regulated cell death. In addition, the stage at which ferroptosis is irreversible and how ferroptotic priming impacts other regulated cell death types through crosstalk is underexplored. Furthermore, cell type differences in ferroptosis sensitivity need more exploration, as does how to modulate microenvironmental factors to therapeutically induce or inhibit ferroptosis88. The specific species of PUFAs that drive ferroptosis are also not fully characterized. This area of research is challenged by the difficulty of characterizing ferroptosis mechanisms at the single-cell level. Despite these technical challenges, there are important advances in the immune aspect of this dynamic and growing research field.
The Roles of Ferroptosis During Homeostasis
How ferroptosis contributes to the development and homeostasis of the immune system is understudied. A recent report shows that human hematopoietic stem cells (HSCs) are vulnerable to ferroptosis89. This observation began through the study of a rare, genetic cause of bone marrow failure caused by the loss of the histone deubiquitinase MYSM1 (myb-like SWIRM and MPN domains 1). Loss of MYSM1 results in reduced global protein translation and increased labile iron through an unknown mechanism, thereby inducing lipid peroxidation, and ferroptosis in the HSC population89. Inhibiting ferroptosis rescues the HSC depletion seen in Mysm1−/− mice89. However, it is unknown whether increased ferroptosis is evident in other models of stem cell failure, or how MYSM1 loss mechanistically sensitizes cells to ferroptosis. Nonetheless, this work suggests that hematopoietic homeostasis may be regulated through ferroptosis. In line with this, a previous report demonstrates that iron overload impairs the proliferation of HSCs mediated by increased reactive oxygen species (ROS), while iron chelation or ferroptosis inhibition may help to promote HSC expansion90. Hence, ferroptosis inhibition may be valuable in the treatment of bone marrow failure caused by radiation exposure, somatic mutations, or acquired diseases, such as aplastic anemia and myelodysplastic syndrome89. In addition, ferroptosis, as detected by a marker of lipid peroxidation, is present in nucleated erythrocytes at embryonic day 13.5 in a Fischer-344 rats, but ferroptosis disappears in enucleated erythrocytes at embryonic day 18.591. Preventing ferroptosis using liproxstatin 1 significantly delays erythrocyte developmental enucleation91. This data suggests that ferroptosis may control the development of red blood cells.
In addition to HSC development, the role of ferroptosis in modulating HSC function is not yet clear. The genetic ablation of Gpx4 in HSCs does not inhibit HSC function in healthy adult mice. HSCs are only dependent on GPX4 when their diet is depleted of vitamin E, a lipophilic antioxidant92. This suggests that sensitivity of HSCs to ferroptosis is context dependent, or at least that vitamin E can replace GPX4 in protecting HSCs from ferroptosis. Monitoring HSCs may be beneficial when using ferroptosis inducers as a therapy. How ferroptosis regulates the stem cells that produce immune cells is unknown.
Ferroptosis also contributes to aging. There is an increase in ferroptosis and labile iron in most major organs as rats age91. A high fat diet increases ferroptosis in aged tissues, potentially contributing to aging. Ferrous iron also accumulates over time in C. elegans93. This, coupled with age-related decreases in GSH, primes older cells for ferroptosis in nematodes. Blocking ferroptosis in C. elegans prevents age-related cell death and increases both lifespan and health span93. Perhaps, inhibiting ferroptosis may support healthy aging. This provides the first hint that ferroptosis may play a role in physiological processes, such as development and aging, although the precise mechanisms remain unclear.
Ferroptosis impacts macrophage physiology. Erythrophagocytosis results in ferroptosis in splenic red pulp macrophages, accompanied by an increase in ROS, lipid peroxidation, and monocyte migration from the bone marrow to spleen94. As the lifespan of erythrocytes is controlled by macrophage-mediated erythrophagocytosis, it is plausible that this process regulates ferroptosis in macrophages physiologically.
Genetic or pharmacologic depletion of inducible nitric oxide synthase (iNOS) increases the sensitivity of classically activated phagocytes to ferroptosis95. Classically activated macrophages are more resistant to ferroptosis in vivo than alternatively activated macrophages, but this resistance is reduced in iNOS-deficient cells in the TME. This suggests the involvement of iNOS in macrophage ferroptosis in the TME. How ferroptosis physiologically regulates macrophage subset development, function, and survival during homeostasis is unknown95.
Evidence is emerging that ferroptosis regulates aging, hematopoiesis, and macrophage function. However, these results remain an isolated island in a sea of ferroptosis papers (more than 10,000 as of the end of 2023) using a disease-based model focused on cancer or infection. In both physiology and pathology, ferroptosis is microenvironmentally and metabolically modulated. Understanding how ferroptosis contributes to immune homeostasis may reveal new ways to intervene in disease or even improve organismal lifespan.
The Roles of Ferroptosis in Immunity to Infection
Pathogens commandeer regulated cell death for their own propagation and survival. For instance, hepatitis B virus (HBV) induces ferroptosis in hepatocytes by suppressing SLC7a11 expression through H3K27Me3 modification, while overexpression of SLC7a11 or treatment with ferrostatin-1 rescues hepatocytes from ferroptosis and prevents acute liver failure96. Thus, inducing ferroptosis may be a major mechanism of hepatocyte death in HBV infection (Figure 2a)96. Interestingly, hepatitis C virus (HCV) itself is sensitive to lipid peroxidation (Figure 2a)97. As the viral protease is directly regulated by lipid peroxides, lipid peroxidation restricts HCV replication, thereby potentially preventing viral replication in oxidative tissues and allowing for viral persistence97. In line with this observation, inducing ferroptosis chemically prevents HCV replication and increases sensitivity to antiviral agents97. Whether preventing or inducing ferroptosis alters the therapeutic course of either HBV or HCV in humans remains unexplored. In addition, SARS-CoV-2 infection causes ferroptosis and pyroptosis gene signatures in COVID-19 patients98. Whether genetic or epigenetic reduction in SLC7a11 expression or cystine transport capability mediates susceptibility to viral induced tissue damage or sensitivity to viral infection has not been investigated.
Figure 2: The role of ferroptosis in viral and bacterial immunity.

a) Hepatitis B and C virus use different approaches to manipulate ferroptosis to promote viral infection. b) Bacteria induce ferroptosis in mammalian cells to facilitate bacterial replication and dissemination throughout the host. AA-PE, arachidonic acid phosphatidylethanolamine.
Bacterial pathogens can also exploit ferroptosis to induce host cell damage and facilitate bacterial replication (Figure 2b). Intracellular pathogens, such as Mycobacterium tuberculosis (Mtb), reduce the expression of GPX4, and increase free iron, mitochondrial superoxide, and lipid peroxidation in infected host cells, causing ferroptotic cell death (Figure 2b)99. Moreover, gram-negative bacteria, such as Pseudomonas aeruginosa, express lipoxygenase that directly oxidizes host arachidonic-acid-phosphatidylethanolamines and triggers ferroptosis in human bronchial epithelia (Figure 2b)100. This bacterium often causes deadly infection in cystic fibrosis patients. Fungi can also promote ferroptosis: The rice (Oryza sativa) fungal pathogen Magnaporthe oryzae promotes ferroptosis in host plants but leads to protection from infection, ferroptosis functions as a defense mechanism in this case101. Given that pathogens induce ferroptosis in host cells, combining ferroptosis inhibitors with antibiotics could allow the immune system to effectively restrain this infection102.
Ferroptosis also regulates the immune response to pathogens. For example, selenium supplementation increases GPX4 expression in follicular helper T (Tfh) cells and reduces their ferroptosis potential, thereby improving antibody responses in influenza immunized mice (Figure 2b)54. In line with this, T-cell-specific GPX4-deficient mice cannot fight against infections with acute lymphocytic choriomeningitis virus or Leishmania parasite54. This immune deficiency can be prevented by high dose supplementation with Vitamin E, a lipophilic antioxidant, which protects GPX4−/− T cells from ferroptosis103. Nonetheless, when, and how T cell ferroptosis occurs in vivo during the immune response to different pathogen infections is largely unknown.
How pathogens coopt ferroptosis to facilitate their own survival, and how host organisms use ferroptosis to prevent infection needs further investigation. How the different forms of cell death regulate each other and communicate between execution pathways in infection is unknown. Understanding what pathogen factors direct a target cell towards a certain cell death fate could lead to the development of novel classes of antibiotics or antivirals. Ferroptosis may be a double-edged sword in infectious immunity, as different pathogens can either be limited or promoted by ferroptotic death. How both the pathogens themselves and the host immune system react to ferroptosis-modulating agents must be carefully studied for effective clinical translation.
The Roles of Ferroptosis in Autoimmunity
Ferroptosis can affect the pathology of several autoimmune diseases, including inflammatory bowel disease (IBD), rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), and multiple sclerosis (MS) (Figure 3).
Figure 3: Ferroptosis induces damage in autoimmune diseases.

Autoimmune diseases frequently have reduced GPX4 expression and increased lipid peroxidation, but the mechanisms of ferroptosis in this sphere are poorly understood.
Ferroptosis contributes to the pathogenesis of IBD (Figure 3). As early as the 1980s, far before the conceptual establishment of ferroptosis, it was reported that patients with ulcerative colitis (UC) have increased PUFAs, suggesting that inflammation alters the lipid composition in the colon104. We now appreciate that this lipid alteration may increase the sensitivity of intestinal epithelial cells to ferroptosis105. In addition to UC, intestinal epithelial cells in Crohn’s Disease (CD) have decreased GPX4 activity and increased lipid peroxidation106. Treatment with a diet enriched in PUFAs results in neutrophilic granulomas and enteritis in mice lacking one allele of GPX4106. Potentially, a high PUFA diet interacts with genetic factors to modulate ferroptosis in patients with autoimmune disease.
RA consists of progressive deterioration of joints mediated by the predominant cellular components of the synovial fluid, such as activated neutrophils and fibroblasts. Neutrophils in the synovial fluid in RA patients exhibit increased oxidative stress, manifested by high ROS, hydroxyl radicals, and lipid peroxidation (Figure 3)107. Treatment with imidazole ketone erastin, a ferroptosis inducer, reduces the fibroblast number in the synovial fluid in an RA mouse model and its combinatory therapy with a tumor necrosis factor (TNF) antagonist, etanercept, slows down arthritis progression108. Nonetheless, research is lacking in how to target specific cell types with ferroptosis-modulating drugs. This becomes even more important in a disease like RA, where one cell type is damaging another. The published literature does not yet elucidate the mechanism of how oxidation alters RA progression.
The dysregulation of ferroptosis in autoimmune diseases is not limited to IBD and RA. In a mouse model of experimental autoimmune encephalomyelitis (EAE), a murine model of multiple sclerosis, the amounts of GPX4 and SLC7a11 are reduced, along with increased lipid peroxidation, in the gray matter and spinal cord109. EAE mice have an abnormal neuronal mitochondrial morphology characteristic of ferroptosis109. Inhibiting ferroptosis or reducing ACSL4 expression in EAE mice improves the phenotype, reduced neuronal death, and reduced neuroinflammation110. Potentially, ferroptosis occurs early in EAE pathology, and this may promote T cell activation and autoimmunity110. This suggests a role for ferroptosis in mediating central nervous system damage in multiple sclerosis111.
Few studies have investigated how ferroptosis is induced in SLE. Kidneys isolated from patients or mice with lupus nephritis demonstrate increased lipid peroxidation and increased expression of ACSL4112. Serum from patients with lupus nephritis increases the sensitivity of human proximal tubular cells to ferroptosis, suggesting that a serum factor may propagate lupus nephritis112. SLE damage may also be mediated through ferroptosis of neutrophils. Neutrophils isolated from an SLE mouse model or patients with SLE undergo increased ferroptosis (Figure 3)113. Autoantibodies and IFNα enhance binding of a transcriptional repressor to the GPX4 promoter, which leads to reduced expression of GPX4, and eventual elevation of lipid peroxidation. In addition, mice with neutrophil deletion of one allele of GPX4 exhibit autoantibodies, neutropenia, skin lesions, and proteinuria, which are symptoms of human SLE113. Treating SLE-prone mice with a ferroptosis inhibitor reduces disease severity113. Thus, neutrophil ferroptosis may play a role in SLE pathology. In psoriasis, an autoimmune skin disease, ACSL4 is upregulated in psoriatic lesions in the epithelium, driving the production of lipid substrates for peroxidation114. This promotes an inflammatory response by activating ferroptosis114. Inhibiting ferroptosis with ferrostatin suppresses ferroptosis related changes in erastin-treated keratinocytes and prevented dermatitis in murine models. Together, these data suggest that ferroptosis plays a role in psoriasis.
Overall, autoimmunity is associated with increased lipid peroxidation and ferroptotic cell death in different cell types. Targeting ferroptosis to treat autoimmune diseases may be a therapeutic option.
The Roles of Ferroptosis in Tumor Immunity
Recent research has shed light on the role of ferroptosis in anti-tumor immunity. T cells use several mechanisms to induce apoptosis in cancer cells, including the Fas and Fas-ligand pathway and the perforin-granzyme pathway. Both pathways activate caspases, inducing apoptosis (Figure 4a). T cells can also induce a regulated cell death known as pyroptosis (Figure 4b)115,116. Granzyme A, secreted by natural killer (NK) cells and cytotoxic T lymphocytes (CTLs) can kill through pyroptosis116. Granzyme A cleaves Gasdermin B, which unleashes its capability of forming pores116. Gasdermin B is highly expressed in some tumors derived from the digestive tract116. This suggests that Granzyme-A-induced pyroptosis may be an important anti-tumor immunity mechanism in some tumors. Notably, killer cells can activate caspase-independent pyroptosis via Granzyme B by directly cleaving Gasdermin E115.
Figure 4: T cells kill tumors through apoptosis, pyroptosis, and ferroptosis.

a) T cells can kill tumors through apoptosis mediated through the perforin-granzyme pathway and the FAS/FAS ligand pathway that both activate caspases. b) T cells can kill tumor cells using pyroptosis mediated by the cleavage of gasdermins (GSDM). c) T-cell-secreted IFNγ targets system xc and ACSL4 and collaborates with arachidonic acid to induce lipid peroxidation and ferroptotic cell death.
Interestingly, activated T-cell-derived IFNγ combined with specific fatty acids in the TME can induce tumor cell ferroptosis (Figure 4c)16,39. IFNγ can be released from several immune cell subsets, such as T helper-1 (Th1) cells, NK cells, and natural killer T (NKT) cells. Hence, IFNγ-producing immune cells can trigger cell ferroptosis in a specific lipid-enriched microenvironment. Given that most ferroptosis studies use synthetic chemical inducers, such as RSL3 and Erastin, the identification of IFNγ and fatty acids as endogenous ferroptosis inducers provides information critical to understanding ferroptosis in vivo16. Other potential endogenous inducers remain to be discovered in future studies.
Tumor ferroptosis contributes to cancer immunity and immunotherapy. Immunotherapy stimulates CD8+ T cells to produce IFNγ. On the one hand, IFNγ reduces the expression of system xc-(SLC3A2 and SLC7A11), thereby impairing the uptake of cystine by tumor cells, abolishing tumor cell GSH, and consequently sensitizing tumors to ferroptosis39. On the other hand, IFNγ stimulates ACSL4 expression and alters tumor cell lipid profiles in the presence of arachidonic acid and other fatty acids, thereby increasing incorporations of arachidonic acid into C16 and C18 acyl chain-containing phospholipids and inducing tumor cell ferroptosis16. Thus, tumor-infiltrating T cells can induce tumor cell ferroptosis through multiple mechanisms16,39. High expression of the cystine import machinery and low amounts of ACSL4 in tumors are associated with poor cancer patient outcome. Thus, inducing ferroptosis alongside immune checkpoint blockade (ICB) is a potential therapeutic approach16,39. Similarly, CD8+ T cells can sensitize cancer cells to cisplatin-based chemotherapy via IFNγ117.Cisplatin can be chelated by GSH, and the GSH-platinum complex subsequently effluxes from the tumor cells, causing tumor chemoresistance117. IFNγ inhibits system xc-, diminishing cystine and GSH in tumor-associated fibroblasts117. This results in reduced cellular GSH in tumor cells, decreased GSH-platinum complex formation, increased intracellular platinum accumulation, and increased tumor killing117. Ferroptosis also mediates tumor cell death induced by tumor radiotherapy36. Radiotherapy-activated ataxia telangiectasia mutated (ATM) synergistically interacts with IFNγ to suppress SLC7A11 expression and cystine uptake in tumor cells36. This leads to increased tumor cell ferroptosis and tumor control in radiation therapy36. Activation of the endoplasmic reticulum stress response factor X-box binding protein (XBP1) in DCs suppresses anti-tumor immunity by driving abnormal lipid accumulation118. This lipid accumulation inhibits DC and T cell interaction118. Silencing XBP1 in DCs improved their anti-tumor functions, and immunostimulatory dendritic cell function118. Although this paper did not demonstrate directly that XBP1 activation induced ferroptosis in DCs, it suggests that targeting the ER stress response pathway may enhance anti-cancer immunity as well as directly induce tumor death. Altogether, ferroptosis is an emerging mechanism of immune killing of tumor cells.
The role of granulocyte ferroptosis in the tumor microenvironment is a developing area of research. In a murine glioblastoma model, neutrophils isolated from brain tumors kill tumor cells through ferroptosis119. Neutrophils induce lipid peroxides in tumor cells by transferring myeloperoxidase into tumor cells through granules119. In human glioblastoma, ferroptosis is associated with necrosis and predict poor survival119. Potentially, ferroptosis drives necrosis in brain tumors and is pro-tumorigenic. However, the crosstalk between ferroptotic, necrotic, and apoptotic pathways, and how this impacts immune response to tumors is unknown.
How phagocytes clear ferroptotic cells is also poorly understood. One key phagocytic signal on the ferroptotic cell surface is oxidized phospholipid, 1-steaoryl-2–15-HpETE-sn-glycero-3-phosphatidylethanolamine (SAPE-OOH)120. Enriching with SAPE-OOH improves macrophage phagocytosis of ferroptotic cells mediated through toll like receptor 2120. TLR2 is critical for the clearance of ferroptotic cells in an in vivo mammary tumor mouse model as well120. How SAPE-OOH is regulated, and whether this TLR pathway is immunogenic or immunosuppressive must still be investigated.
Ferroptotic tumor cells may regulate immune cell infiltration and activity in the TME. DCs are professional APCs. How ferroptotic cancer cells impact DC function and tumor immune responses remains uncertain121,122. Wiernicki and colleagues report that co-culturing of early ferroptotic cancer cells with DCs reduces DC maturation and antigen cross-presentation122. Wiernicki et al found that DCs loaded with ferroptotic, in contrast to necroptotic cancer cells, cannot protect against tumor growth, and that ferroptotic cancer cells reduce the vaccination potential of apoptotic cancer cells122. In contrast, Efimova et al indicate that ferroptosis is immunogenic in vitro and in vivo, and that ferroptotic tumor cells promote DC maturation and elicit a vaccination like effect in immune-competent mice121. However, in the Efimova et al paper, tumor growth at the vaccination site was not considered, making it difficult to distinguish between vaccination with living tumors cells and cells undergoing the earliest stages of ferroptosis121. . For a viable translatable vaccine, no tumor growth at the vaccination site is critical, limiting the potential of using early ferroptotic cells as an immunogen. These conflicting reports have not fully revealed the causal mechanisms linking ferroptosis and DCs. The discrepancy may be related to different experimental conditions, such as different chemical inducers in vitro, different types of tumor cells, different exposure times to tumor cells with these ferroptosis-inducing chemicals, and different degrees of tumor cell ferroptosis. The latter is critical, as the remaining live tumor cells post-ferroptosis inducers are ferroptosis-resistant cells, which likely possess an aggressive tumor phenotype, potentially generating conflicting results.
Ferroptosis of immune cell subsets may alter tumor immune responses in the TME. A recent study shows that granular myeloid-derived suppressor cells (G-MDSCs), which are neutrophils, are sensitive to ferroptosis in the TME123. These ferroptotic neutrophils release oxidized lipids and limit the activity of T cells123. In contrast, a different report demonstrates that tumor-infiltrating neutrophils are resistant to ferroptosis124. Mechanistically, tumor-infiltrating neutrophils produce itaconate, which prevents ferroptosis and allows neutrophils to remain live in the TME124. Notably, tumor-associated G-MDSCs, namely neutrophils, are well-established as immunosuppressive entities irrespective of their involvement in ferroptosis. A direct comparison regarding whether apoptotic, ferroptotic, or live neutrophils in the TME are differentially immunosuppressive is currently lacking in the literature. Hence, it is misleading to conclude that because ferroptotic neutrophils may have an immunosuppressive role, tumor microenvironmental ferroptosis is immunosuppressive and pro-tumorigenic. Rather, it is likely that context and cellular composition of the tumor microenvironment is critical to whether ferroptosis is immunosuppressive or immunogenic. This provides additional complications to inducing ferroptosis to inhibit tumor growth, as other cell types are susceptible to ferroptosis, and may inhibit anti-tumor immunity. Currently, it is impossible to induce ferroptosis in a cell type specific manner without genetic modulation, but cell type targeting is likely required before ferroptosis induction is adapted widely as a cancer fighting strategy. Similar concerns can be implied in inducing cancer apoptosis and pyroptosis for therapy.
In addition to neutrophils, ferroptosis of T cell subsets has been observed in different settings, including in tumors. Interestingly, human lymphocytes have surprisingly low cystine transport activity even after activation125. Loss of GPX4 in regulatory T cells (Tregs) leads to lipid peroxidation and ferroptosis in Treg cells after T-cell-receptor signaling126. As expected, ablation of Treg cell GPX4 represses tumor growth and potentiates antitumor immunity126. In contrast, genetic deletion of GPX4 in CD8+ T cells led to the failure of antigen-stimulated T cell expansion and reduced pathogen immunity103. This is attributed to the rapid accumulation of membrane lipid peroxides and ferroptosis in CD8+ T cells103. Genetic deletion of GPX4 in T cells also prevents the generation of Tfh cells and germinal center responses54. This potentially curtails antibody responses. Together, these data suggest that GPX4 is critical to protect T cells from lipid peroxidation and ferroptosis. Notably, although T cell proliferation in vitro is dependent on system xc-, the cystine-glutamate antiporter, it appears that system xc- is not essential for T cell proliferation in vivo and the generation of primary and memory immune responses to tumors127. It is possible that the importance of extracellular cystine in T cells is compensated by alternative mechanism(s)127. However, the differences in ferroptosis induced by cystine depletion or xCT inhibition compared to ferroptosis induced by GPX4 depletion are not well characterized.
Tumors often have aberrantly regulated lipid production. How this dysregulation mediates ferroptosis in tumor cells or the anti-tumor immune response is poorly understood. It has been reported that tumor-infiltrating T cells accumulate lipids128,129. This accumulation is dependent on CD36, a scavenger receptor for oxidized lipids129. Knocking out CD36 allowed T cells to maintain their effector function in the tumor compared to wildtype T cells129. CD36 promotes the uptake of oxidized low-density lipoproteins into T cells, which induces lipid peroxidation129. In support of this work, ablation of CD36 in effector CD8+ T cells led to an increase in cytotoxic cytokines and increased tumor clearance128. Thus, blocking CD36 or inhibiting ferroptosis in CD8+ T cells could restore antitumor immunity and potentially increase the efficacy of ICB128,129.
The functional importance of ferroptosis-regulatory genes in different cell types in the TME is slowly being dissected. However, there are many unanswered questions: In what contexts is ferroptosis immunogenic; what damage-associated molecular patterns are present in cells undergoing ferroptosis; what TME cells initiate the response to cells dying of ferroptosis; what propagates immune response to the tumor over time; what and how potential crosstalk among different death pathways is in the TME; and how can we optimize pro- vs anti-ferroptotic agents to control the balance between help and harm in tumor suppression. Answering these questions will reveal the nature of ferroptosis in the TME and identify novel cancer treatment modalities.
Therapeutically Targeting Ferroptosis
No current ferroptosis-modulating drugs are used to treat infectious diseases. Proof-of-concept studies have demonstrated that inducing bacterial ferroptosis may lead to the development of a new class of antibiotics. Treatment with ferrous-sulfate-loaded hydrogel induces aa variety of reactive oxygen species, lipid peroxidation, and ferroptosis in Staphylococcus (S.) aureus130. This iron-sulfate-based drug kills S. aureus persister cells and biofilms, both of which are canonically resistant to antibiotics, prevents the dissemination of S. aureus to the lung, and reduces systemic inflammation130. Moreover, arachidonic acid induces lipid peroxidation and death in S. aureus131. Mechanistically, arachidonic acid is oxidized and modifies macromolecules in the bacteria, inducing toxicity131. Treatment with antioxidants prevents S. aureus death after exposure to arachidonic acid131. Thus, inducing ferroptosis in bacteria may treat infections resistant to traditional antibiotics and is an approach to develop a new class of antibiotics.
Anti-ferroptotic modulation, partly via dietary supplementation, has been investigated to modulate autoimmune disease progression. Several studies report that antioxidants do not change clinical indexes of inflammation in RA132,133. However, N-acetylcysteine, a cysteine donor, improves the clinical presentation of RA and SLE132,134. Selenium supplementation improves RA patients’ rating of their health compared to a placebo and leads to normalized oxidative stress markers of granulocytes and improved clinical conditions in psoriatic patients113,135. The current literature does not provide a consensus about how modulating ferroptosis through diet could prevent the pathology of autoimmune disease. Understanding the temporal ferroptosis involvement, cell types undergoing ferroptosis, and ferroptotic molecular pathways will be required to achieve translational impact.
Preclinical studies have shown that targeting tumor ferroptosis pathways is a promising anti-cancer therapeutic strategy. In fact, erastin was identified as a ferroptosis inducer, in a screen for anti-cancer drugs. Treatment with imidazole ketone erastin, a potent ferroptosis inducer, induces GSH depletion, increases lipid peroxidation, and slows down tumor growth in murine models of diffuse large B cell lymphomas136. As cysteine is critical for the survival of some cancer cells in oxidative conditions, treatment with cysteinase, an agent that depletes cysteine and cystine, inhibited tumor growth in vivo39,136. Sorafenib is the first-line drug for advanced hepatocellular carcinoma (HCC)137. Some studies suggest that Sorafenib induces an iron-dependent ferroptotic tumor cell death138,139. Solute carrier family 27 member 5 (SLC27A5) is downregulated in sorafenib-resistant HCC138. SLC27A5 is involved in fatty acid elongation and complex lipid synthesis, and its deficiency suppresses ferroptosis and drives sorafenib resistance. However, whether sorafenib truly induces ferroptosis has recently been up for debate. A recent study utilizing genetically engineered cell lines and canonical ferroptosis inhibitors, showed that sorafenib does not appear to induce ferroptosis in a panel of tumor cell lines139. Whether sorafenib can induce ferroptosis in certain in vitro contexts or in vivo, and the underlying reasons for the discrepancy between these reports is unknown.
As noted above, immune cell subsets are subjected to ferroptosis regulation in the TME. Ferroptosis-inducing drugs may indiscriminately induce ferroptosis in both cancer cells and other cells in the TME, such as immune cells. Recent drug screening has identified, N6F11, a previously unstudied small molecule, can trigger selective degradation of GPX4 in cancer cells, but not in DCs, T cells, and NK cells140. Treatment with N6F11 induces ferroptotic cancer cell death and sensitizes pancreatic tumor response to ICB140. It remains to assess how N6F11 mediates tumor-selective targeting. Given that increased microenvironmental iron can mediate ferroptosis, magnetic fields can be used to deliver iron oxide to the TME to trigger tumor cell ferroptosis and inhibit tumor progression141.
Combining ferroptosis induction with well-established cancer therapies, such as immunotherapy, radiotherapy, and chemotherapy has been experimentally evaluated36,142,143. Several approaches to induce ferroptosis in tumors are actively being explored. Biomimetic magnetic nanoparticles generate a mild immune response and improve the efficacy of immunotherapy in a murine metastatic breast cancer model144. Another potential approach is to combine several approaches, including thermal energy, ferroptosis, and immunotherapy. A gel delivery therapy platform combined with embedded gold nanorods, and iron oxide nanoparticles allows for targeted photothermal therapy and ferroptosis to trigger both innate and adaptive immunity145. Recently, a novel approach of a six enzyme co-expressed nanoplatform, including lipoxygenase and phospholipase A2 co-loaded FeCo/Fe-Co dual metal atom nanozyme, induces initial ferroptosis and upregulates arachidonic acid expression and synergizes with IFNγ to induce immunogenic ferroptosis146. Inducing ferroptosis by depleting cysteine, intermittent methionine intermittent deprivation, inhibiting system xc-, or targeting GPX4, can increase the therapeutic efficacy of ICB in tumor-bearing animal models39,136,147,148. Interestingly, human obesity is associated with an increased response to ICB, the phenomenon termed the “obesity-immunotherapy paradox149.” This paradox may be partially explained because ICB-activated immune cells promote and induce cancer ferroptosis via IFNγ, in collaboration with microenvironmental fatty acids16. No direct evidence has yet demonstrated that obese patients with cancer have a distinctly favorable lipidome. However, fatty acids serum and feces profiles do correlate with ICB efficacy in patients with cancer150,151. Thus, it is possible that obese patients with cancer may manifest lipid metabolism favorable for immune-induced tumor ferroptosis although this has not been directly demonstrated. Overall, current basic and translational studies have generated rationales for the development of ferroptosis therapeutics in infectious diseases, autoimmune diseases, and cancers.
Concluding Remarks and Future Directions
Compelling evidence demonstrates that ferroptosis participates in the regulation of immune responses. Several molecular pathways, including system xc-, GPX4, the lipid peroxidation and detoxification system, ACSL-mediated fatty acid activation, the FSP1-Coenzme Q10 system, and DHODH-associated mitochondrial pathway, can regulate ferroptotic cell death152. Unlike apoptosis and pyroptosis, there is no identified “execution gene” for ferroptosis. This not only highlights mechanistic distinctions among the different types of regulated cell death, but also demands the necessity of further understanding the relationship between ferroptosis-promoting and ferroptosis-inhibiting signals and environmental contexts in physiological and pathological settings.
Current studies point toward the involvement of several cellular organelles in ferroptosis. The roles of mitochondria, ER, Golgi, lysosome, and plasma membrane have been examined in ferroptotic cell death in different experimental settings, but the specific cell types, timing, instigators, microenvironmental factors, and intervention points, need to be clarified58. We posit that different ferroptosis-modulating determinants alter different organelles, as some act through the mitochondria, some through the ER, and some through the Golgi. Nonetheless, no matter the organellar path to get there, the final and irreversible mechanism of ferroptosis may be cellular membrane damage due to lipid peroxidation (Figure 1). Potentially and arguably, the mitochondria act as a central hub for the induction of cell death and communication between different cell death pathways.
It will be important to further elucidate the precise molecular and cellular regulation used by the immune system to regulate ferroptotic cell death. In the presence of specific fatty acids, effector T cells inhibit system xc- and stimulate ACSL4 via IFNγ to promote and induce tumor cell ferroptosis16,39. This contributes to T cell tumor killing and ICB. Based on these discoveries, it is reasoned that activated immune cells, such as T cells, NK cells, NKT cells, may similarly induce ferroptotic cell death in the disease microenvironments of infection and autoimmune disorders. Ferroptosis generates thousands of diverse lipid peroxidation byproducts from hydroperoxyl lipids to secondary electrophilic oxidatively truncated species. It is known that some lipid species may form covalent adducts with proteins and attach to biomolecules. On the one hand, this may generate potential neo-antigens, offering an opportunity to harness a response to these neo-antigens against cancer. On the other hand, these lipid-peroxidation-induced neoantigens may stimulate autoimmune responses via self-antigen cross reactivity. Thus, exploration of the interaction between oxidized lipids and proteins in ferroptosis may yield novel insight into ferroptosis biology.
Finally, oxidative stress is common to different forms of regulated cell death, including ferroptosis, apoptosis, and pyroptosis. However, phospholipid peroxidation is specific to ferroptosis. The unique feature of ferroptosis provides additional access points to modulate cell death. Mitochondrial and ER stress, induced by cellular metabolism, immune stress, and therapeutic intervention, may be a crossroad for interaction among different forms of regulated cell death. All forms of cell death end with the disintegration of the plasma membrane and dissolution of the cell, but the triggering pathways to this end differ wildly. Iron dependency and lipid peroxidation is specific to ferroptosis. Functionally, the role of apoptosis is initially and predominantly defined in the development of tissue and organ, and cell differentiation, while pyroptosis is heavily involved in infection control, and ferroptosis lies somewhere in between—with developing roles in homeostasis and established roles in autoimmunity, pathogenicity, and tumor. A future research direction would be to explore whether, how, and when, crosstalk among different forms of regulated cell death, determines the fate of infectious diseases, autoimmune diseases, and tumors.
Highlights:
Ferroptosis, a type of regulated cell death dependent on lipid peroxidation and iron, plays critical roles in immunity.
Ferroptosis is a double-edged sword in immunity against infection, as pathogens can be either limited or promoted by ferroptotic death.
Patients with autoimmune disease frequently exhibit increased oxidation and dysregulated ferroptosis pathway. How to use ferroptosis modulating agents as treatment is being actively investigated.
IFNγ+ immune cells, such as T cells, can induce tumor ferroptosis in the presence of specific fatty acids in the tumor microenvironment. The immunogenicity of ferroptosis of different cell types, the role of crosstalk between cellular death pathways, and the therapeutic potential of ferroptosis modulation need further investigation.
Acknowledgements:
This work was supported by the NIH/NCI R35 (CA209896) (B.R.S.), R01 grants (CA217648, CA123088, CA19136, and CA152470) (W.Z.), and the NIH/NCI through the University of Michigan Rogel Cancer Center (CA46592).
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Declarations of Interest:
B.R.S. is an inventor on patents and patent applications involving ferroptosis; co-founded and serves as a consultant to ProJenX, Inc. and Exarta Therapeutics; holds equity in Sonata Therapeutics; serves as a consultant to Weatherwax Biotechnologies Corporation and Akin Gump Strauss Hauer & Feld LLP. W.Z. has served as a scientific advisor or consultant for Cstone, NextCure, and HanchorBio Inc. H.N.B declares no competing interests.
References:
- 1.Elmore S (2007). Apoptosis: A Review of Programmed Cell Death. Toxicol. Pathol 35, 495–516. 10.1080/01926230701320337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Green DR, Ferguson T, Zitvogel L, and Kroemer G (2009). Immunogenic and tolerogenic cell death. Nat. Rev. Immunol 9, 353–363. 10.1038/nri2545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Kroemer G, Galluzzi L, Kepp O, and Zitvogel L (2013). Immunogenic Cell Death in Cancer Therapy. Annu. Rev. Immunol 31, 51–72. 10.1146/annurev-immunol-032712-100008. [DOI] [PubMed] [Google Scholar]
- 4.Galluzzi L, Buqué A, Kepp O, Zitvogel L, and Kroemer G (2017). Immunogenic cell death in cancer and infectious disease. Nat. Rev. Immunol 17, 97–111. 10.1038/nri.2016.107. [DOI] [PubMed] [Google Scholar]
- 5.Workenhe ST, Inkol JM, Westerveld MJ, Verburg SG, Worfolk SM, Walsh SR, and Kallio KLF (2023). Determinants for Antitumor and Protumor Effects of Programmed Cell Death. Cancer Immunol. Res, OF1–OF10. 10.1158/2326-6066.cir-23-0321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Kolb JP, Oguin TH, Oberst A, and Martinez J (2017). Programmed Cell Death and Inflammation: Winter Is Coming. Trends Immunol. 38, 705–718. 10.1016/j.it.2017.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Koonin EV, and Krupovic M (2019). Origin of programmed cell death from antiviral defense? Proc. Natl. Acad. Sci 116, 16167–16169. 10.1073/pnas.1910303116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Nagata S, and Tanaka M (2017). Programmed cell death and the immune system. Nat. Rev. Immunol 17, 333–340. 10.1038/nri.2016.153. [DOI] [PubMed] [Google Scholar]
- 9.Wang W, and Zou W (2020). Amino Acids and Their Transporters in T Cell Immunity and Cancer Therapy. Mol. Cell 80, 384–395. 10.1016/j.molcel.2020.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Zou W, and Green DR (2023). Beggars banquet: Metabolism in the tumor immune microenvironment and cancer therapy. Cell Metab. 35, 1101–1113. 10.1016/j.cmet.2023.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.ZHANG N, HARTIG H, DZHAGALOV I, DRAPER D, and HE YW (2005). The role of apoptosis in the development and function of T lymphocytes. Cell Res. 15, 749–769. 10.1038/sj.cr.7290345. [DOI] [PubMed] [Google Scholar]
- 12.Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS, et al. (2012). Ferroptosis: An Iron-Dependent Form of Nonapoptotic Cell Death. Cell 149, 1060–1072. 10.1016/j.cell.2012.03.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Gan B (2021). Mitochondrial regulation of ferroptosis. J. Cell Biol 220, e202105043. 10.1083/jcb.202105043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Yagoda N, Rechenberg M von, Zaganjor E, Bauer AJ, Yang WS, Fridman DJ, Wolpaw AJ, Smukste I, Peltier JM, Boniface JJ, et al. (2007). RAS–RAF–MEK-dependent oxidative cell death involving voltage-dependent anion channels. Nature 447, 865–869. 10.1038/nature05859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Riegman M, Sagie L, Galed C, Levin T, Steinberg N, Dixon SJ, Wiesner U, Bradbury MS, Niethammer P, Zaritsky A, et al. (2020). Ferroptosis occurs through an osmotic mechanism and propagates independently of cell rupture. Nat Cell Biol 22, 1042–1048. 10.1038/s41556-020-0565-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Liao P, Wang W, Wang W, Kryczek I, Li X, Bian Y, Sell A, Wei S, Grove S, Johnson JK, et al. (2022). CD8+ T cells and fatty acids orchestrate tumor ferroptosis and immunity via ACSL4. Cancer Cell. 10.1016/j.ccell.2022.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Zhang X, Yu K, Ma L, Qian Z, Tian X, Miao Y, Niu Y, Xu X, Guo S, Yang Y, et al. (2021). Endogenous glutamate determines ferroptosis sensitivity via ADCY10-dependent YAP suppression in lung adenocarcinoma. Theranostics 11, 5650–5674. 10.7150/thno.55482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Yang WS, Kim KJ, Gaschler MM, Patel M, Shchepinov MS, and Stockwell BR (2016). Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis. Proc National Acad Sci 113, E4966–E4975. 10.1073/pnas.1603244113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Ingold I, Berndt C, Schmitt S, Doll S, Poschmann G, Buday K, Roveri A, Peng X, Freitas FP, Seibt T, et al. (2018). Selenium Utilization by GPX4 Is Required to Prevent Hydroperoxide-Induced Ferroptosis. Cell 172, 409–422.e21. 10.1016/j.cell.2017.11.048. [DOI] [PubMed] [Google Scholar]
- 20.Ursini F, Maiorino M, and Gregolin C (1985). The selenoenzyme phospholipid hydroperoxide glutathione peroxidase. Biochim. Biophys. Acta (BBA) - Gen. Subj 839, 62–70. 10.1016/0304-4165(85)90182-5. [DOI] [PubMed] [Google Scholar]
- 21.Seiler A, Schneider M, Förster H, Roth S, Wirth EK, Culmsee C, Plesnila N, Kremmer E, Rådmark O, Wurst W, et al. (2008). Glutathione Peroxidase 4 Senses and Translates Oxidative Stress into 12/15-Lipoxygenase Dependent- and AIF-Mediated Cell Death. Cell Metab 8, 237–248. 10.1016/j.cmet.2008.07.005. [DOI] [PubMed] [Google Scholar]
- 22.Ursini F, and Maiorino M (2020). Lipid peroxidation and ferroptosis: The role of GSH and GPx4. Free Radic. Biol. Med 152, 175–185. 10.1016/j.freeradbiomed.2020.02.027. [DOI] [PubMed] [Google Scholar]
- 23.Angeli JPF, Schneider M, Proneth B, Tyurina YY, Tyurin VA, Hammond VJ, Herbach N, Aichler M, Walch A, Eggenhofer E, et al. (2014). Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nat Cell Biol 16, 1180–1191. 10.1038/ncb3064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Stockwell BR (2022). Ferroptosis turns 10: Emerging mechanisms, physiological functions, and therapeutic applications. Cell 185, 2401–2421. 10.1016/j.cell.2022.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Miao R, Jiang C, Chang WY, Zhang H, An J, Ho F, Chen P, Zhang H, Junqueira C, Amgalan D, et al. (2023). Gasdermin D permeabilization of mitochondrial inner and outer membranes accelerates and enhances pyroptosis. Immunity 56, 2523–2541.e8. 10.1016/j.immuni.2023.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Bock FJ, and Tait SWG (2020). Mitochondria as multifaceted regulators of cell death. Nat. Rev. Mol. Cell Biol 21, 85–100. 10.1038/s41580-019-0173-8. [DOI] [PubMed] [Google Scholar]
- 27.Gao M, Yi J, Zhu J, Minikes AM, Monian P, Thompson CB, and Jiang X (2019). Role of Mitochondria in Ferroptosis. Mol. Cell 73, 354–363.e3. 10.1016/j.molcel.2018.10.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Yang WS, and Stockwell BR (2008). Synthetic Lethal Screening Identifies Compounds Activating Iron-Dependent, Nonapoptotic Cell Death in Oncogenic-RAS-Harboring Cancer Cells. Chem. Biol 15, 234–245. 10.1016/j.chembiol.2008.02.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Dixon SJ, Patel DN, Welsch M, Skouta R, Lee ED, Hayano M, Thomas AG, Gleason CE, Tatonetti NP, Slusher BS, et al. (2014). Pharmacological inhibition of cystine–glutamate exchange induces endoplasmic reticulum stress and ferroptosis. eLife 3, e02523. 10.7554/elife.02523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Sato M, Kusumi R, Hamashima S, Kobayashi S, Sasaki S, Komiyama Y, Izumikawa T, Conrad M, Bannai S, and Sato H (2018). The ferroptosis inducer erastin irreversibly inhibits system xc− and synergizes with cisplatin to increase cisplatin’s cytotoxicity in cancer cells. Sci. Rep 8, 968. 10.1038/s41598-018-19213-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Cheff DM, Huang C, Scholzen KC, Gencheva R, Ronzetti MH, Cheng Q, Hall MD, and Arnér ESJ (2023). The ferroptosis inducing compounds RSL3 and ML162 are not direct inhibitors of GPX4 but of TXNRD1. Redox Biol. 62, 102703. 10.1016/j.redox.2023.102703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Vučković A, Travain VB, Bordin L, Cozza G, Miotto G, Rossetto M, Toppo S, Venerando R, Zaccarin M, Maiorino M, et al. (2020). Inactivation of the glutathione peroxidase GPx4 by the ferroptosis-inducing molecule RSL3 requires the adaptor protein 14-3-3ε. FEBS Lett. 594, 611–624. 10.1002/1873-3468.13631. [DOI] [PubMed] [Google Scholar]
- 33.Chen Y, Liu Y, Lan T, Qin W, Zhu Y, Qin K, Gao J, Wang H, Hou X, Chen N, et al. (2018). Quantitative Profiling of Protein Carbonylations in Ferroptosis by an Aniline-Derived Probe. J. Am. Chem. Soc 140, 4712–4720. 10.1021/jacs.8b01462. [DOI] [PubMed] [Google Scholar]
- 34.Chen J, Li X, Ge C, Min J, and Wang F (2022). The multifaceted role of ferroptosis in liver disease. Cell Death Differ. 29, 467–480. 10.1038/s41418-022-00941-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Jeridi A, Günsel GG, Novikava M, Doll S, Lang NJ, Verleden SE, Rehberg M, Stoeger T, Schiller HB, Conrad M, et al. (2022). Ferroptosis, induced by macrophages drives COPD pathogenesis. Airw. cell Biol. Immunopathol, 177. 10.1183/23120541.lsc-2022.177. [DOI] [Google Scholar]
- 36.Lang X, Green MD, Wang W, Yu J, Choi JE, Jiang L, Liao P, Zhou J, Zhang Q, Dow A, et al. (2019). Radiotherapy and Immunotherapy Promote Tumoral Lipid Oxidation and Ferroptosis via Synergistic Repression of SLC7A11. Cancer Discov 9, 1673–1685. 10.1158/2159-8290.cd-19-0338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Paraskevaidis IA, Iliodromitis EK, Vlahakos D, Tsiapras DP, Nikolaidis A, Marathias A, Michalis A, and Kremastinos D.Th. (2005). Deferoxamine infusion during coronary artery bypass grafting ameliorates lipid peroxidation and protects the myocardium against reperfusion injury: immediate and long-term significance. Eur. Hear. J 26, 263–270. 10.1093/eurheartj/ehi028. [DOI] [PubMed] [Google Scholar]
- 38.Ryan SK, Ugalde CL, Rolland A-S, Skidmore J, Devos D, and Hammond TR (2023). Therapeutic inhibition of ferroptosis in neurodegenerative disease. Trends Pharmacol. Sci 44, 674–688. 10.1016/j.tips.2023.07.007. [DOI] [PubMed] [Google Scholar]
- 39.Wang W, Green M, Choi JE, Gijón M, Kennedy PD, Johnson JK, Liao P, Lang X, Kryczek I, Sell A, et al. (2019). CD8+ T cells regulate tumour ferroptosis during cancer immunotherapy. Nature 569, 270–274. 10.1038/s41586-019-1170-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Wang Y, Tang B, Zhu J, Yu J, Hui J, Xia S, and Ji J (2022). Emerging Mechanisms and Targeted Therapy of Ferroptosis in Neurological Diseases and Neuro-oncology. Int. J. Biol. Sci 18, 4260–4274. 10.7150/ijbs.72251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Yang L, Cao L, Zhang X, and Chu B (2022). Targeting ferroptosis as a vulnerability in pulmonary diseases. Cell Death Dis. 13, 649. 10.1038/s41419-022-05070-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Jiang X, Stockwell BR, and Conrad M (2021). Ferroptosis: mechanisms, biology and role in disease. Nat. Rev. Mol. Cell Biol 22, 266–282. 10.1038/s41580-020-00324-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Dolma S, Lessnick SL, Hahn WC, and Stockwell BR (2003). Identification of genotype-selective antitumor agents using synthetic lethal chemical screening in engineered human tumor cells. Cancer Cell 3, 285–296. 10.1016/s1535-6108(03)00050-3. [DOI] [PubMed] [Google Scholar]
- 44.Yang WS, SriRamaratnam R, Welsch ME, Shimada K, Skouta R, Viswanathan VS, Cheah JH, Clemons PA, Shamji AF, Clish CB, et al. (2014). Regulation of Ferroptotic Cancer Cell Death by GPX4. Cell 156, 317–331. 10.1016/j.cell.2013.12.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Koppula P, Zhuang L, and Gan B (2021). Cystine transporter SLC7A11/xCT in cancer: ferroptosis, nutrient dependency, and cancer therapy. Protein Cell 12, 599–620. 10.1007/s13238-020-00789-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Sato H, Tamba M, Ishii T, and Bannai S (1999). Cloning and Expression of a Plasma Membrane Cystine/Glutamate Exchange Transporter Composed of Two Distinct Proteins*. J. Biol. Chem 274, 11455–11458. 10.1074/jbc.274.17.11455. [DOI] [PubMed] [Google Scholar]
- 47.Jyotsana N, Ta KT, and DelGiorno KE (2022). The Role of Cystine/Glutamate Antiporter SLC7A11/xCT in the Pathophysiology of Cancer. Front. Oncol 12, 858462. 10.3389/fonc.2022.858462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Gao M, Monian P, Quadri N, Ramasamy R, and Jiang X (2015). Glutaminolysis and Transferrin Regulate Ferroptosis. Mol. Cell 59, 298–308. 10.1016/j.molcel.2015.06.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Doll S, Proneth B, Tyurina YY, Panzilius E, Kobayashi S, Ingold I, Irmler M, Beckers J, Aichler M, Walch A, et al. (2017). ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat Chem Biol 13, 91–98. 10.1038/nchembio.2239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Krusenstiern AN von, Robson RN, Qian N, Qiu B, Hu F, Reznik E, Smith N, Zandkarimi F, Estes VM, Dupont M, et al. (2023). Identification of essential sites of lipid peroxidation in ferroptosis. Nat. Chem. Biol 19, 719–730. 10.1038/s41589-022-01249-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Shchepinov MS (2020). Polyunsaturated Fatty Acid Deuteration against Neurodegeneration. Trends Pharmacol. Sci 41, 236–248. 10.1016/j.tips.2020.01.010. [DOI] [PubMed] [Google Scholar]
- 52.Coillie SV, San EV, Goetschalckx I, Wiernicki B, Mukhopadhyay B, Tonnus W, Choi SM, Roelandt R, Dumitrascu C, Lamberts L, et al. (2022). Targeting ferroptosis protects against experimental (multi)organ dysfunction and death. Nat. Commun 13, 1046. 10.1038/s41467-022-28718-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Alim I, Caulfield JT, Chen Y, Swarup V, Geschwind DH, Ivanova E, Seravalli J, Ai Y, Sansing LH, Ste.Marie EJ, et al. (2019). Selenium Drives a Transcriptional Adaptive Program to Block Ferroptosis and Treat Stroke. Cell 177, 1262–1279.e25. 10.1016/j.cell.2019.03.032. [DOI] [PubMed] [Google Scholar]
- 54.Yao Y, Chen Z, Zhang H, Chen C, Zeng M, Yunis J, Wei Y, Wan Y, Wang N, Zhou M, et al. (2021). Selenium–GPX4 axis protects follicular helper T cells from ferroptosis. Nat. Immunol 22, 1127–1139. 10.1038/s41590-021-00996-0. [DOI] [PubMed] [Google Scholar]
- 55.Mancias JD, Wang X, Gygi SP, Harper JW, and Kimmelman AC (2014). Quantitative proteomics identifies NCOA4 as the cargo receptor mediating ferritinophagy. Nature 509, 105–109. 10.1038/nature13148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Gryzik M, Asperti M, Denardo A, Arosio P, and Poli M (2021). NCOA4-mediated ferritinophagy promotes ferroptosis induced by erastin, but not by RSL3 in HeLa cells. Biochim. Biophys. Acta (BBA) - Mol. Cell Res 1868, 118913. 10.1016/j.bbamcr.2020.118913. [DOI] [PubMed] [Google Scholar]
- 57.Kremer DM, Nelson BS, Lin L, Yarosz EL, Halbrook CJ, Kerk SA, Sajjakulnukit P, Myers A, Thurston G, Hou SW, et al. (2021). GOT1 inhibition promotes pancreatic cancer cell death by ferroptosis. Nat. Commun 12, 4860. 10.1038/s41467-021-24859-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Mao C, Liu X, Zhang Y, Lei G, Yan Y, Lee H, Koppula P, Wu S, Zhuang L, Fang B, et al. (2021). DHODH-mediated ferroptosis defence is a targetable vulnerability in cancer. Nature 593, 586–590. 10.1038/s41586-021-03539-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Mishima E, Nakamura T, Zheng J, Zhang W, Mourão ASD, Sennhenn P, and Conrad M (2023). DHODH inhibitors sensitize to ferroptosis by FSP1 inhibition. Nature 619, E9–E18. 10.1038/s41586-023-06269-0. [DOI] [PubMed] [Google Scholar]
- 60.Bersuker K, Hendricks JM, Li Z, Magtanong L, Ford B, Tang PH, Roberts MA, Tong B, Maimone TJ, Zoncu R, et al. (2019). The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature 575, 688–692. 10.1038/s41586-019-1705-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Doll S, Freitas FP, Shah R, Aldrovandi M, Silva MC da, Ingold I, Grocin AG, X. da Silva TN, Panzilius E, Scheel CH, et al. (2019). FSP1 is a glutathione-independent ferroptosis suppressor. Nature 575, 693–698. 10.1038/s41586-019-1707-0. [DOI] [PubMed] [Google Scholar]
- 62.Cui S, Ghai A, Deng Y, Li S, Zhang R, Egbulefu C, Liang G, Achilefu S, and Ye J (2023). Identification of hyperoxidized PRDX3 as a ferroptosis marker reveals ferroptotic damage in chronic liver diseases. Mol. Cell 83, 3931–3939.e5. 10.1016/j.molcel.2023.09.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Scarpellini C, Klejborowska G, Lanthier C, Hassannia B, Berghe TV, and Augustyns K (2023). Beyond ferrostatin-1: a comprehensive review of ferroptosis inhibitors. Trends Pharmacol. Sci 44, 902–916. 10.1016/j.tips.2023.08.012. [DOI] [PubMed] [Google Scholar]
- 64.Sato H, Fujiwara K, Sagara J, and Bannai S (1995). Induction of cystine transport activity in mouse peritoneal macrophages by bacterial lipopolysaccharide. Biochem. J 310, 547–551. 10.1042/bj3100547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Wu J, Minikes AM, Gao M, Bian H, Li Y, Stockwell BR, Chen Z-N, and Jiang X (2019). Intercellular interaction dictates cancer cell ferroptosis via NF2–YAP signalling. Nature 572, 402–406. 10.1038/s41586-019-1426-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Yang W-H, Ding C-KC, Sun T, Rupprecht G, Lin C-C, Hsu D, and Chi J-T (2019). The Hippo Pathway Effector TAZ Regulates Ferroptosis in Renal Cell Carcinoma. Cell Reports 28, 2501–2508.e4. 10.1016/j.celrep.2019.07.107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Nishizawa H, Matsumoto M, Chen G, Ishii Y, Tada K, Onodera M, Kato H, Muto A, Tanaka K, and Igarashi K (2021). Lipid peroxidation and the subsequent cell death transmitting from ferroptotic cells to neighboring cells. Cell Death Dis. 12, 332. 10.1038/s41419-021-03613-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Freitas FP, Alborzinia H, dos Santos AF, Nepachalovich P, Pedrera L, Zilka O, Inague A, Klein C, Aroua N, Kaushal K, et al. (2024). 7-Dehydrocholesterol is an endogenous suppressor of ferroptosis. Nature 626, 401–410. 10.1038/s41586-023-06878-9. [DOI] [PubMed] [Google Scholar]
- 69.Oh S-J, Ikeda M, Ide T, Hur KY, and Lee M-S (2022). Mitochondrial event as an ultimate step in ferroptosis. Cell Death Discov. 8, 414. 10.1038/s41420-022-01199-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Gaschler MM, Andia AA, Liu H, Csuka JM, Hurlocker B, Vaiana CA, Heindel DW, Zuckerman DS, Bos PH, Reznik E, et al. (2018). FINO2 initiates ferroptosis through GPX4 inactivation and iron oxidation. Nat. Chem. Biol 14, 507–515. 10.1038/s41589-018-0031-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Zou Y, Henry WS, Ricq EL, Graham ET, Phadnis VV, Maretich P, Paradkar S, Boehnke N, Deik AA, Reinhardt F, et al. (2020). Plasticity of ether lipids promotes ferroptosis susceptibility and evasion. Nature 585, 603–608. 10.1038/s41586-020-2732-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Alborzinia H, Ignashkova TI, Dejure FR, Gendarme M, Theobald J, Wölfl S, Lindemann RK, and Reiling JH (2018). Golgi stress mediates redox imbalance and ferroptosis in human cells. Commun. Biol 1, 210. 10.1038/s42003-018-0212-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Feng H, Schorpp K, Jin J, Yozwiak CE, Hoffstrom BG, Decker AM, Rajbhandari P, Stokes ME, Bender HG, Csuka JM, et al. (2020). Transferrin Receptor Is a Specific Ferroptosis Marker. Cell Rep. 30, 3411–3423.e7. 10.1016/j.celrep.2020.02.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Jin J, Schorpp K, Samaga D, Unger K, Hadian K, and Stockwell BR (2022). Machine Learning Classifies Ferroptosis and Apoptosis Cell Death Modalities with TfR1 Immunostaining. ACS Chem. Biol 17, 654–660. 10.1021/acschembio.1c00953. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Antoszczak M, Müller S, Cañeque T, Colombeau L, Dusetti N, Santofimia-Castaño P, Gaillet C, Puisieux A, Iovanna JL, and Rodriguez R (2022). Iron-Sensitive Prodrugs That Trigger Active Ferroptosis in Drug-Tolerant Pancreatic Cancer Cells. J. Am. Chem. Soc 144, 11536–11545. 10.1021/jacs.2c03973. [DOI] [PubMed] [Google Scholar]
- 76.Mai TT, Hamaï A, Hienzsch A, Cañeque T, Müller S, Wicinski J, Cabaud O, Leroy C, David A, Acevedo V, et al. (2017). Salinomycin kills cancer stem cells by sequestering iron in lysosomes. Nat. Chem 9, 1025–1033. 10.1038/nchem.2778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Gaschler MM, Hu F, Feng H, Linkermann A, Min W, and Stockwell BR (2018). Determination of the Subcellular Localization and Mechanism of Action of Ferrostatins in Suppressing Ferroptosis. Acs Chem Biol 13, 1013–1020. 10.1021/acschembio.8b00199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Gao H, Bai Y, Jia Y, Zhao Y, Kang R, Tang D, and Dai E (2018). Ferroptosis is a lysosomal cell death process. Biochem. Biophys. Res. Commun 503, 1550–1556. 10.1016/j.bbrc.2018.07.078. [DOI] [PubMed] [Google Scholar]
- 79.Wang J, and Han X (2019). Analytical challenges of shotgun lipidomics at different resolution of measurements. TrAC Trends Anal. Chem 121, 115697. 10.1016/j.trac.2019.115697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.da Silva KM, Wölk M, Nepachalovich P, Iturrospe E, Covaci A, van Nuijs ALN, and Fedorova M (2023). Investigating the Potential of Drift Tube Ion Mobility for the Analysis of Oxidized Lipids. Anal. Chem 95, 13566–13574. 10.1021/acs.analchem.3c02213. [DOI] [PubMed] [Google Scholar]
- 81.Tyurina YY, Kapralov AA, Tyurin VA, Shurin G, Amoscato AA, Rajasundaram D, Tian H, Bunimovich YL, Nefedova Y, Herrick WG, et al. (2023). Redox phospholipidomics discovers pro-ferroptotic death signals in A375 melanoma cells in vitro and in vivo. Redox Biol. 61, 102650. 10.1016/j.redox.2023.102650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Tyurina YY, Tyurin VA, Anthonymuthu T, Amoscato AA, Sparvero LJ, Nesterova AM, Baynard ML, Sun W, He R, Khaitovich P, et al. (2019). “Redox lipidomics technology: Looking for a needle in a haystack.” Chem. Phys. Lipids 221, 93–107. 10.1016/j.chemphyslip.2019.03.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Rajbhandari P, Neelakantan TV, Hosny N, and Stockwell BR (2023). Spatial pharmacology using mass spectrometry imaging. Trends Pharmacol. Sci 10.1016/j.tips.2023.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Dixon SJ, Winter GE, Musavi LS, Lee ED, Snijder B, Rebsamen M, Superti-Furga G, and Stockwell BR (2015). Human Haploid Cell Genetics Reveals Roles for Lipid Metabolism Genes in Nonapoptotic Cell Death. ACS Chem. Biol 10, 1604–1609. 10.1021/acschembio.5b00245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Kagan VE, Mao G, Qu F, Angeli JPF, Doll S, Croix CS, Dar HH, Liu B, Tyurin VA, Ritov VB, et al. (2017). Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis. Nat Chem Biol 13, 81–90. 10.1038/nchembio.2238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Magtanong L, Ko P-J, To M, Cao JY, Forcina GC, Tarangelo A, Ward CC, Cho K, Patti GJ, Nomura DK, et al. (2019). Exogenous Monounsaturated Fatty Acids Promote a Ferroptosis-Resistant Cell State. Cell Chem Biol 26, 420–432.e9. 10.1016/j.chembiol.2018.11.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Mishima E, Ito J, Wu Z, Nakamura T, Wahida A, Doll S, Tonnus W, Nepachalovich P, Eggenhofer E, Aldrovandi M, et al. (2022). A non-canonical vitamin K cycle is a potent ferroptosis suppressor. Nature 608, 778–783. 10.1038/s41586-022-05022-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Magtanong L, Mueller GD, Williams KJ, Billmann M, Chan K, Armenta DA, Pope LE, Moffat J, Boone C, Myers CL, et al. (2022). Context-dependent regulation of ferroptosis sensitivity. Cell Chem. Biol 29, 1568. 10.1016/j.chembiol.2022.08.008. [DOI] [PubMed] [Google Scholar]
- 89.Zhao J, Jia Y, Mahmut D, Deik AA, Jeanfavre S, Clish CB, and Sankaran VG (2023). Human hematopoietic stem cell vulnerability to ferroptosis. Cell 186, 732–747.e16. 10.1016/j.cell.2023.01.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Jin X, He X, Cao X, Xu P, Xing Y, Sui S, Wang L, Meng J, Lu W, Cui R, et al. (2018). Iron overload impairs normal hematopoietic stem and progenitor cells through reactive oxygen species and shortens survival in myelodysplastic syndrome mice. Haematologica 103, 1627–1634. 10.3324/haematol.2018.193128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Zheng H, Jiang L, Tsuduki T, Conrad M, and Toyokuni S (2021). Embryonal erythropoiesis and aging exploit ferroptosis. Redox Biol. 48, 102175. 10.1016/j.redox.2021.102175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Hu Q, Zhang Y, Lou H, Ou Z, Liu J, Duan W, Wang H, Ge Y, Min J, Wang F, et al. (2021). GPX4 and vitamin E cooperatively protect hematopoietic stem and progenitor cells from lipid peroxidation and ferroptosis. Cell Death Dis. 12, 706. 10.1038/s41419-021-04008-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Jenkins NL, James SA, Salim A, Sumardy F, Speed TP, Conrad M, Richardson DR, Bush AI, and McColl G (2020). Changes in ferrous iron and glutathione promote ferroptosis and frailty in aging Caenorhabditis elegans. eLife 9, e56580. 10.7554/elife.56580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Youssef LA, Rebbaa A, Pampou S, Weisberg SP, Stockwell BR, Hod EA, and Spitalnik SL (2018). Increased erythrophagocytosis induces ferroptosis in red pulp macrophages in a mouse model of transfusion. Blood 131, 2581–2593. 10.1182/blood-2017-12-822619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Kapralov AA, Yang Q, Dar HH, Tyurina YY, Anthonymuthu TS, Kim R, St.Croix CM, Mikulska-Ruminska K, Liu B, Shrivastava IH, et al. (2020). Redox lipid reprogramming commands susceptibility of macrophages and microglia to ferroptotic death. Nat. Chem. Biol 16, 278–290. 10.1038/s41589-019-0462-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Liu G-Z, Xu X-W, Tao S-H, Gao M-J, and Hou Z-H (2021). HBx facilitates ferroptosis in acute liver failure via EZH2 mediated SLC7A11 suppression. J. Biomed. Sci 28, 67. 10.1186/s12929-021-00762-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Yamane D, McGivern DR, Wauthier E, Yi M, Madden VJ, Welsch C, Antes I, Wen Y, Chugh PE, McGee CE, et al. (2014). Regulation of the hepatitis C virus RNA replicase by endogenous lipid peroxidation. Nat. Med 20, 927–935. 10.1038/nm.3610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Peleman C, Coillie SV, Ligthart S, Choi SM, Waele JD, Depuydt P, Benoit D, Schaubroeck H, Francque SM, Dams K, et al. (2023). Ferroptosis and pyroptosis signatures in critical COVID-19 patients. Cell Death Differ. 30, 2066–2077. 10.1038/s41418-023-01204-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Amaral EP, Costa DL, Namasivayam S, Riteau N, Kamenyeva O, Mittereder L, Mayer-Barber KD, Andrade BB, and Sher A (2019). A major role for ferroptosis in Mycobacterium tuberculosis–induced cell death and tissue necrosis. J. Exp. Med 216, 556–570. 10.1084/jem.20181776. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Dar HH, Tyurina YY, Mikulska-Ruminska K, Shrivastava I, Ting H-C, Tyurin VA, Krieger J, St.Croix CM, Watkins S, Bayir E, et al. (2018). Pseudomonas aeruginosa utilizes host polyunsaturated phosphatidylethanolamines to trigger theft-ferroptosis in bronchial epithelium. J. Clin. Investig 128, 4639–4653. 10.1172/jci99490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Shen Q, Liang M, Yang F, Deng YZ, and Naqvi NI (2020). Ferroptosis contributes to developmental cell death in rice blast. N. Phytol 227, 1831–1846. 10.1111/nph.16636. [DOI] [PubMed] [Google Scholar]
- 102.Gao J, Wang Q, Tang Y-D, Zhai J, Hu W, and Zheng C (2023). When ferroptosis meets pathogenic infections. Trends Microbiol. 31, 468–479. 10.1016/j.tim.2022.11.006. [DOI] [PubMed] [Google Scholar]
- 103.Matsushita M, Freigang S, Schneider C, Conrad M, Bornkamm GW, and Kopf M (2015). T cell lipid peroxidation induces ferroptosis and prevents immunity to infection. J. Exp. Med 212, 555–568. 10.1084/jem.20140857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Nishida T, Miwa H, Shigematsu A, Yamamoto M, Iida M, and Fujishima M (1987). Increased arachidonic acid composition of phospholipids in colonic mucosa from patients with active ulcerative colitis. Gut 28, 1002. 10.1136/gut.28.8.1002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Xu M, Tao J, Yang Y, Tan S, Liu H, Jiang J, Zheng F, and Wu B (2020). Ferroptosis involves in intestinal epithelial cell death in ulcerative colitis. Cell Death Dis. 11, 86. 10.1038/s41419-020-2299-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Mayr L, Grabherr F, Schwärzler J, Reitmeier I, Sommer F, Gehmacher T, Niederreiter L, He G-W, Ruder B, Kunz KTR, et al. (2020). Dietary lipids fuel GPX4-restricted enteritis resembling Crohn’s disease. Nat. Commun 11, 1775. 10.1038/s41467-020-15646-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Datta S, Kundu S, Ghosh P, De S, Ghosh A, and Chatterjee M (2014). Correlation of oxidant status with oxidative tissue damage in patients with rheumatoid arthritis. Clin. Rheumatol 33, 1557–1564. 10.1007/s10067-014-2597-z. [DOI] [PubMed] [Google Scholar]
- 108.Wu J, Feng Z, Chen L, Li Y, Bian H, Geng J, Zheng Z-H, Fu X, Pei Z, Qin Y, et al. (2022). TNF antagonist sensitizes synovial fibroblasts to ferroptotic cell death in collagen-induced arthritis mouse models. Nat. Commun 13, 676. 10.1038/s41467-021-27948-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Hu C, Nydes M, Shanley KL, Pantoja IEM, Howard TA, and Bizzozero OA (2019). Reduced expression of the ferroptosis inhibitor glutathione peroxidase-4 in multiple sclerosis and experimental autoimmune encephalomyelitis. J. Neurochem 148, 426–439. 10.1111/jnc.14604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Luoqian J, Yang W, Ding X, Tuo Q, Xiang Z, Zheng Z, Guo Y, Li L, Guan P, Ayton S, et al. (2022). Ferroptosis promotes T-cell activation-induced neurodegeneration in multiple sclerosis. Cell. Mol. Immunol 19, 913–924. 10.1038/s41423-022-00883-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.San EV, Debruyne AC, Veeckmans G, Tyurina YY, Tyurin VA, Zheng H, Choi SM, Augustyns K, van Loo G, Michalke B, et al. (2023). Ferroptosis contributes to multiple sclerosis and its pharmacological targeting suppresses experimental disease progression. Cell Death Differ. 30, 2092–2103. 10.1038/s41418-023-01195-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Alli AA, Desai D, Elshika A, Conrad M, Proneth B, Clapp W, Atkinson C, Segal M, Searcy LA, Denslow ND, et al. (2023). Kidney tubular epithelial cell ferroptosis links glomerular injury to tubulointerstitial pathology in lupus nephritis. Clin. Immunol 248, 109213. 10.1016/j.clim.2022.109213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Li P, Jiang M, Li K, Li H, Zhou Y, Xiao X, Xu Y, Krishfield S, Lipsky PE, Tsokos GC, et al. (2021). Glutathione peroxidase 4–regulated neutrophil ferroptosis induces systemic autoimmunity. Nat. Immunol 22, 1107–1117. 10.1038/s41590-021-00993-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Liu L, and Kang X (2022). ACSL4 is overexpressed in psoriasis and enhances inflammatory responses by activating ferroptosis. Biochem. Biophys. Res. Commun 623, 1–8. 10.1016/j.bbrc.2022.07.041. [DOI] [PubMed] [Google Scholar]
- 115.Zhang Z, Zhang Y, Xia S, Kong Q, Li S, Liu X, Junqueira C, Meza-Sosa KF, Mok TMY, Ansara J, et al. (2020). Gasdermin E suppresses tumour growth by activating anti-tumour immunity. Nature 579, 415–420. 10.1038/s41586-020-2071-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Zhou Z, He H, Wang K, Shi X, Wang Y, Su Y, Wang Y, Li D, Liu W, Zhang Y, et al. (2020). Granzyme A from cytotoxic lymphocytes cleaves GSDMB to trigger pyroptosis in target cells. Science 368. 10.1126/science.aaz7548. [DOI] [PubMed] [Google Scholar]
- 117.Wang W, Kryczek I, Dostál L, Lin H, Tan L, Zhao L, Lu F, Wei S, Maj T, Peng D, et al. (2016). Effector T Cells Abrogate Stroma-Mediated Chemoresistance in Ovarian Cancer. Cell 165, 1092–1105. 10.1016/j.cell.2016.04.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Cubillos-Ruiz JR, Silberman PC, Rutkowski MR, Chopra S, Perales-Puchalt A, Song M, Zhang S, Bettigole SE, Gupta D, Holcomb K, et al. (2015). ER Stress Sensor XBP1 Controls Anti-tumor Immunity by Disrupting Dendritic Cell Homeostasis. Cell 161, 1527–1538. 10.1016/j.cell.2015.05.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Yee PP, Wei Y, Kim S-Y, Lu T, Chih SY, Lawson C, Tang M, Liu Z, Anderson B, Thamburaj K, et al. (2020). Neutrophil-induced ferroptosis promotes tumor necrosis in glioblastoma progression. Nat. Commun 11, 5424. 10.1038/s41467-020-19193-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Luo X, Gong H-B, Gao H-Y, Wu Y-P, Sun W-Y, Li Z-Q, Wang G, Liu B, Liang L, Kurihara H, et al. (2021). Oxygenated phosphatidylethanolamine navigates phagocytosis of ferroptotic cells by interacting with TLR2. Cell Death Differ. 28, 1971–1989. 10.1038/s41418-020-00719-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Efimova I, Catanzaro E, der Meeren LV, Turubanova VD, Hammad H, Mishchenko TA, Vedunova MV, Fimognari C, Bachert C, Coppieters F, et al. (2020). Vaccination with early ferroptotic cancer cells induces efficient antitumor immunity. J. Immunother. Cancer 8, e001369. 10.1136/jitc-2020-001369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Wiernicki B, Maschalidi S, Pinney J, Adjemian S, Berghe TV, Ravichandran KS, and Vandenabeele P (2022). Cancer cells dying from ferroptosis impede dendritic cell-mediated anti-tumor immunity. Nat. Commun 13, 3676. 10.1038/s41467-022-31218-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Kim R, Hashimoto A, Markosyan N, Tyurin VA, Tyurina YY, Kar G, Fu S, Sehgal M, Garcia-Gerique L, Kossenkov A, et al. (2022). Ferroptosis of tumour neutrophils causes immune suppression in cancer. Nature 612, 338–346. 10.1038/s41586-022-05443-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Zhao Y, Liu Z, Liu G, Zhang Y, Liu S, Gan D, Chang W, Peng X, Sung ES, Gilbert K, et al. (2023). Neutrophils resist ferroptosis and promote breast cancer metastasis through aconitate decarboxylase 1. Cell Metab. 35, 1688–1703.e10. 10.1016/j.cmet.2023.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.GMÜNDER H, ECK H, and DRÖGE W (1991). Low membrane transport activity for cystine in resting and mitogenically stimulated human lymphocyte preparations and human T cell clones. Eur. J. Biochem 201, 113–117. 10.1111/j.1432-1033.1991.tb16263.x. [DOI] [PubMed] [Google Scholar]
- 126.Xu C, Sun S, Johnson T, Qi R, Zhang S, Zhang J, and Yang K (2021). The glutathione peroxidase Gpx4 prevents lipid peroxidation and ferroptosis to sustain Treg cell activation and suppression of antitumor immunity. Cell Rep. 35, 109235. 10.1016/j.celrep.2021.109235. [DOI] [PubMed] [Google Scholar]
- 127.Arensman MD, Yang XS, Leahy DM, Toral-Barza L, Mileski M, Rosfjord EC, Wang F, Deng S, Myers JS, Abraham RT, et al. (2019). Cystine–glutamate antiporter xCT deficiency suppresses tumor growth while preserving antitumor immunity. Proc. Natl. Acad. Sci 116, 9533–9542. 10.1073/pnas.1814932116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Ma X, Xiao L, Liu L, Ye L, Su P, Bi E, Wang Q, Yang M, Qian J, and Yi Q (2021). CD36-mediated ferroptosis dampens intratumoral CD8+ T cell effector function and impairs their antitumor ability. Cell Metab 33, 1001–1012.e5. 10.1016/j.cmet.2021.02.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Xu S, Chaudhary O, Rodríguez-Morales P, Sun X, Chen D, Zappasodi R, Xu Z, Pinto AFM, Williams A, Schulze I, et al. (2021). Uptake of oxidized lipids by the scavenger receptor CD36 promotes lipid peroxidation and dysfunction in CD8+ T cells in tumors. Immunity 54, 1561–1577.e7. 10.1016/j.immuni.2021.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Wang Z, Li H, Zhou W, Lee J, Liu Z, An Z, Xu D, Mo H, Hu L, and Zhou X (2022). Ferrous sulfate-loaded hydrogel cures Staphylococcus aureus infection via facilitating a ferroptosis-like bacterial cell death in a mouse keratitis model. Biomaterials 290, 121842. 10.1016/j.biomaterials.2022.121842. [DOI] [PubMed] [Google Scholar]
- 131.Beavers WN, Monteith AJ, Amarnath V, Mernaugh RL, Roberts LJ, Chazin WJ, Davies SS, and Skaar EP (2019). Arachidonic Acid Kills Staphylococcus aureus through a Lipid Peroxidation Mechanism. mBio 10, e01333–19. 10.1128/mbio.01333-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Batooei M, Tahamoli-Roudsari A, Basiri Z, Yasrebifar F, Shahdoust M, Eshraghi A, Mehrpooya M, and Ataei S (2018). Evaluating the Effect of Oral N-acetylcysteine as an Adjuvant Treatment on Clinical Outcomes of Patients with Rheumatoid Arthritis: A Randomized, Double Blind Clinical Trial. Rev. Recent Clin. Trials 13, 132–138. 10.2174/1574887113666180307151937. [DOI] [PubMed] [Google Scholar]
- 133.Edmonds SE, Winyard PG, Guo R, Kidd B, Merry P, Langrish-Smith A, Hansen C, Ramm S, and Blake DR (1997). Putative analgesic activity of repeated oral doses of vitamin E in the treatment of rheumatoid arthritis. Results of a prospective placebo controlled double blind trial. Ann. Rheum. Dis 56, 649. 10.1136/ard.56.11.649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Lai Z, Hanczko R, Bonilla E, Caza TN, Clair B, Bartos A, Miklossy G, Jimah J, Doherty E, Tily H, et al. (2012). N-acetylcysteine reduces disease activity by blocking mammalian target of rapamycin in T cells from systemic lupus erythematosus patients: A randomized, double-blind, placebo-controlled trial. Arthritis Rheum. 64, 2937–2946. 10.1002/art.34502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Kharaeva Z, Gostova E, Luca CD, Raskovic D, and Korkina L (2009). Clinical and biochemical effects of coenzyme Q10, vitamin E, and selenium supplementation to psoriasis patients. Nutrition 25, 295–302. 10.1016/j.nut.2008.08.015. [DOI] [PubMed] [Google Scholar]
- 136.Zhang Y, Tan H, Daniels JD, Zandkarimi F, Liu H, Brown LM, Uchida K, O’Connor OA, and Stockwell BR (2019). Imidazole Ketone Erastin Induces Ferroptosis and Slows Tumor Growth in a Mouse Lymphoma Model. Cell Chem. Biol 26, 623–633.e9. 10.1016/j.chembiol.2019.01.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc J-F, Oliveira A.C. de, Santoro A, Raoul J-L, Forner A, et al. (2008). Sorafenib in Advanced Hepatocellular Carcinoma. N. Engl. J. Med 359, 378–390. 10.1056/nejmoa0708857. [DOI] [PubMed] [Google Scholar]
- 138.Xu F, Wu X, Chen C, Wang K, Huang L, Xia J, Liu Y, Shan X, and Tang N (2023). SLC27A5 promotes sorafenib-induced ferroptosis in hepatocellular carcinoma by downregulating glutathione reductase. Cell Death Dis. 14, 22. 10.1038/s41419-023-05558-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Zheng J, Sato M, Mishima E, Sato H, Proneth B, and Conrad M (2021). Sorafenib fails to trigger ferroptosis across a wide range of cancer cell lines. Cell Death Dis. 12, 698. 10.1038/s41419-021-03998-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Li J, Liu J, Zhou Z, Wu R, Chen X, Yu C, Stockwell B, Kroemer G, Kang R, and Tang D (2023). Tumor-specific GPX4 degradation enhances ferroptosis-initiated antitumor immune response in mouse models of pancreatic cancer. Sci. Transl. Med 15, eadg3049. 10.1126/scitranslmed.adg3049. [DOI] [PubMed] [Google Scholar]
- 141.Yu B, Choi B, Li W, and Kim D-H (2020). Magnetic field boosted ferroptosis-like cell death and responsive MRI using hybrid vesicles for cancer immunotherapy. Nat. Commun 11, 3637. 10.1038/s41467-020-17380-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Fan F, Liu P, Bao R, Chen J, Zhou M, Mo Z, Ma Y, Liu H, Zhou Y, Cai X, et al. (2021). A Dual PI3K/HDAC Inhibitor Induces Immunogenic Ferroptosis to Potentiate Cancer Immune Checkpoint Therapy. Cancer Res. 81, 6233–6245. 10.1158/0008-5472.can-21-1547. [DOI] [PubMed] [Google Scholar]
- 143.Yang F, Xiao Y, Ding J-H, Jin X, Ma D, Li D-Q, Shi J-X, Huang W, Wang Y-P, Jiang Y-Z, et al. (2023). Ferroptosis heterogeneity in triple-negative breast cancer reveals an innovative immunotherapy combination strategy. Cell Metab. 35, 84–100.e8. 10.1016/j.cmet.2022.09.021. [DOI] [PubMed] [Google Scholar]
- 144.Jiang Q, Wang K, Zhang X, Ouyang B, Liu H, Pang Z, and Yang W (2020). Platelet Membrane-Camouflaged Magnetic Nanoparticles for Ferroptosis-Enhanced Cancer Immunotherapy. Small 16, e2001704. 10.1002/smll.202001704. [DOI] [PubMed] [Google Scholar]
- 145.Guo P, Wang L, Shang W, Chen J, Chen Z, Xiong F, Wang Z, Tong Z, Wang K, Yang L, et al. (2020). Intravesical In Situ Immunostimulatory Gel for Triple Therapy of Bladder Cancer. ACS Appl. Mater. Interfaces 12, 54367–54377. 10.1021/acsami.0c15176. [DOI] [PubMed] [Google Scholar]
- 146.Liu Y, Niu R, Deng R, Song S, Wang Y, and Zhang H (2023). Multi-enzyme Co-expressed Dual-Atom Nanozymes Induce Cascade Immunogenic Ferroptosis via Activating Interferon-γ and Targeting Arachidonic Acid Metabolism. J. Am. Chem. Soc 145, 8965–8978. 10.1021/jacs.2c13689. [DOI] [PubMed] [Google Scholar]
- 147.Xue Y, Lu F, Chang Z, Li J, Gao Y, Zhou J, Luo Y, Lai Y, Cao S, Li X, et al. (2023). Intermittent dietary methionine deprivation facilitates tumoral ferroptosis and synergizes with checkpoint blockade. Nat. Commun 14, 4758. 10.1038/s41467-023-40518-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Jiang Z, Lim S-O, Yan M, Hsu JL, Yao J, Wei Y, Chang S-S, Yamaguchi H, Lee H-H, Ke B, et al. (2021). TYRO3 induces anti–PD-1/PD-L1 therapy resistance by limiting innate immunity and tumoral ferroptosis. J. Clin. Investig 131, e139434. 10.1172/jci139434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Wang Z, Aguilar EG, Luna JI, Dunai C, Khuat LT, Le CT, Mirsoian A, Minnar CM, Stoffel KM, Sturgill IR, et al. (2019). Paradoxical effects of obesity on T cell function during tumor progression and PD-1 checkpoint blockade. Nat Med 25, 141–151. 10.1038/s41591-018-0221-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Coutzac C, Jouniaux J-M, Paci A, Schmidt J, Mallardo D, Seck A, Asvatourian V, Cassard L, Saulnier P, Lacroix L, et al. (2020). Systemic short chain fatty acids limit antitumor effect of CTLA-4 blockade in hosts with cancer. Nat Commun 11, 2168. 10.1038/s41467-020-16079-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Nomura M, Nagatomo R, Doi K, Shimizu J, Baba K, Saito T, Matsumoto S, Inoue K, and Muto M (2020). Association of Short-Chain Fatty Acids in the Gut Microbiome With Clinical Response to Treatment With Nivolumab or Pembrolizumab in Patients With Solid Cancer Tumors. JAMA Netw. Open 3, e202895. 10.1001/jamanetworkopen.2020.2895. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Stockwell BR, Angeli JPF, Bayir H, Bush AI, Conrad M, Dixon SJ, Fulda S, Gascón S, Hatzios SK, Kagan VE, et al. (2017). Ferroptosis: A Regulated Cell Death Nexus Linking Metabolism, Redox Biology, and Disease. Cell 171, 273–285. 10.1016/j.cell.2017.09.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
