Fact-checked by Grok 2 weeks ago

Ferroptosis

Ferroptosis is an iron-dependent form of regulated that is morphologically, biochemically, and genetically distinct from , necroptosis, and , primarily driven by the lethal accumulation of hydroperoxides in cellular membranes. First identified in 2003 through synthetic lethal screening of small molecules targeting oncogenic RAS-mutant cancer cells and formally named in 2012, ferroptosis was characterized as a non-apoptotic process triggered by compounds like erastin, which inhibits the cystine/glutamate system xc⁻, leading to depletion and subsequent oxidative damage. At its core, the mechanism involves iron-catalyzed of polyunsaturated fatty acids (PUFAs) via Fenton-like reactions, generating toxic (ROS) that overwhelm antioxidant defenses, particularly the glutathione peroxidase 4 ()-dependent reduction of lipid hydroperoxides; this results in plasma membrane rupture without caspase activation or typical apoptotic features like . Ferroptosis is regulated by a of metabolic pathways, including iron (e.g., via ferritinophagy), (e.g., PUFA incorporation into phospholipids by ACSL4), and transport, with key inducers like RSL3 directly inhibiting and inhibitors such as ferrostatin-1 scavenging lipid ROS. Physiologically, ferroptosis contributes to embryonic development, T-cell differentiation, and macrophage-mediated antiviral responses, while pathologically, it is implicated in ischemia-reperfusion injuries, , and neurodegeneration. In , ferroptosis represents a promising therapeutic target, as many cancers exhibit vulnerability to its induction—exploiting metabolic weaknesses like high iron levels and sensitivity—potentially synergizing with immunotherapies and , whereas its inhibition may mitigate ferroptosis-driven tissue damage in non-malignant diseases.

Definition and Characteristics

Core Features

Ferroptosis is defined as a form of regulated that is distinct from , , and , characterized by the iron-dependent accumulation of peroxides that ultimately leads to oxidative damage and rupture of the . This process is triggered by small-molecule inducers such as erastin, which inhibits cystine uptake via the system xc- transporter, thereby depleting and impairing defenses. Morphologically, ferroptotic cells exhibit shrunken mitochondria with condensed membranes, diminished or absent cristae, and increased mitochondrial membrane density, while the overall and morphology remains relatively intact without the formation of apoptotic or autophagosomes. These ultrastructural changes, observable via electron microscopy, distinguish ferroptosis from other modalities and reflect the profound impact of on integrity. Biochemically, ferroptosis is marked by the excessive accumulation of lipid hydroperoxides, particularly those derived from polyunsaturated fatty acids (PUFAs) such as , coupled with elevated levels of (ROS) originating from lipid oxidation rather than general . This peroxidation is catalyzed by labile iron pools through the Fenton reaction, leading to the depletion of PUFA-containing phospholipids and membrane fragility. Genetically, ferroptosis is identified by cellular sensitivity to inducers like erastin or RSL3 (a direct inhibitor of glutathione peroxidase 4, ), alongside resistance to pharmacological inhibitors of (e.g., inhibitors like Z-VAD-fmk) and necroptosis (e.g., necrostatin-1). These markers enable experimental distinction and highlight ferroptosis as a unique pathway amenable to targeted modulation. As of 2025, research has increasingly integrated ferroptosis with broader metabolic reprogramming, revealing overlaps with emerging forms such as cuproptosis—driven by overload targeting tricarboxylic acid cycle proteins—and disulfidptosis—induced by disulfide stress in proteins—through shared regulators like SLC7A11 and altered . These connections underscore ferroptosis's role in metabolic vulnerabilities, particularly in cancer and degenerative diseases, without altering its core iron-lipid axis.

Comparison to Other Cell Death Forms

Ferroptosis is distinguished from other regulated cell death pathways by its iron-dependent lipid peroxidation mechanism, which is caspase-independent and does not involve the inflammatory DNA release characteristic of necroptosis. Unlike apoptosis, which relies on caspase activation and cytochrome c release for orderly dismantling of the cell, ferroptosis proceeds without nuclear fragmentation or apoptotic body formation, instead featuring progressive membrane damage from reactive oxygen species (ROS) accumulation in lipids. In contrast to autophagy, which primarily involves protein degradation via autophagosomes and does not inherently lead to cell lysis unless excessive, ferroptosis centers on oxidative destruction of polyunsaturated fatty acids in cell membranes rather than cytoplasmic recycling. While overlaps exist, such as shared ROS involvement with necroptosis or regulatory crosstalk with through ferritinophagy, ferroptosis maintains a unique identity defined by its iron-lipid axis. Evolutionarily, ferroptosis is considered one of the most ancient forms of , conserved across species and predating more specialized pathways like , potentially serving as a primitive defense against in early life forms. Initially suppressing inflammation due to the absence of classical activation, ferroptosis can nevertheless trigger immunogenic by releasing damage-associated molecular patterns (DAMPs), such as oxidized , which alert the in a manner distinct from the pro-inflammatory bursts in . The following table summarizes key morphological, biochemical, and pharmacological distinctions among ferroptosis and selected forms:
AspectFerroptosisNecroptosis
MorphologicalMitochondrial shrinkage, fractured outer mitochondrial membranes, diminished cristae, normal morphology, eventual ruptureCell shrinkage, condensation, fragmentation, apoptotic formationCell swelling, swelling, ruptureCytoplasmic , formation, no immediate lysis
BiochemicalIron-dependent , depletion, ROS accumulation in lipids, -independent activation (e.g., caspase-3/7/9), cytochrome c release, DNA laddering/RIPK3/MLKL , permeabilization, DAMP releaseLC3 lipidation, inhibition, lysosomal degradation of proteins/s
PharmacologicalInduced by erastin/RSL3; inhibited by ferrostatin-1, liproxstatin-1, iron chelatorsInduced by /TNF-α; inhibited by Z-VAD-fmk, BH3 mimetics (e.g., as inducers, but inhibitors include overexpression)Induced by TNF-α (with inhibition); inhibited by necrostatin-1Induced by rapamycin; inhibited by , 3-MA
Data adapted from comprehensive reviews on cell death pathways. Recent insights from 2024-2025 highlight emerging hybrid forms, such as crosstalk mediated by , where can amplify activation in cancer cells, leading to combined lytic and oxidative death. For instance, certain metal-based photosensitizers induce simultaneous and ferroptosis, enhancing antitumor immunity through synergistic DAMP release. Despite these interactions, ferroptosis retains its core definition tied to the iron-lipid peroxidation axis, distinguishing it from purely inflammatory or proteolytic deaths.

History

Discovery and Initial Identification

The concept of ferroptosis emerged from early observations of oxidative cell death linked to nutrient deprivation, with initial hints dating back to the 1950s. In 1955, Harry Eagle and colleagues reported that depriving mammalian cells, such as strain L fibroblasts and HeLa cells, of cystine—a key amino acid—induced a morphologically distinct form of cell death characterized by cytoplasmic granulation and vacuolization, distinct from typical necrosis or apoptosis. Subsequent studies in the late 1950s and 1970s further connected cystine starvation to glutathione depletion and oxidative stress, noting that supplementation with antioxidants like α-tocopherol could rescue cells, though these phenomena were not mechanistically defined as a regulated pathway until later high-throughput screens in the 2000s. A pivotal advance came in 2003 when Brent Stockwell's laboratory at identified erastin through a synthetic lethal chemical screen targeting oncogenic RAS-transformed human fibroblasts (BJeLR cells). Erastin selectively induced rapid, non- in these cancer cells, exhibiting morphological features such as shrunken mitochondria and increased density, without activating caspase-dependent or necroptotic markers.00050-3) This death was inhibited by iron chelators like and lipophilic antioxidants such as , suggesting an iron-dependent oxidative mechanism, though the precise pathway remained elusive at the time.00050-3) The formal identification and naming of ferroptosis occurred in , when Stockwell's team published a seminal study characterizing erastin-induced death as a novel, iron-dependent form of regulated . Through systematic screening, they demonstrated that erastin inhibits the cystine/glutamate (system xc⁻, encoded by SLC7A11), blocking cystine uptake and thereby depleting intracellular , the primary cellular antioxidant.00520-X) This depletion leads to accumulation of (ROS), which is prevented by iron chelators like , confirming the iron dependency; the team coined the term "ferroptosis" to reflect this unique iron-linked peroxidation process, distinguishing it biochemically and genetically from , , and autophagy.00520-X) Early inhibitors like ferrostatin-1, which scavenge ROS, further validated ferroptosis as a distinct pathway in this foundational work.00520-X)

Major Developments and Milestones

In 2014, researchers identified glutathione peroxidase 4 () as a central suppressor of ferroptosis through chemoproteomic approaches targeting the small-molecule inducer RSL3, establishing as an essential that prevents by reducing phospholipid hydroperoxides using glutathione.01544-4) This discovery highlighted the selenium-dependent nature of GPX4's activity, linking ferroptosis regulation to selenocysteine incorporation during protein synthesis.01544-4) Building on this, a major breakthrough occurred in 2019 when two independent studies identified ferroptosis suppressor protein 1 (FSP1, encoded by AIFM2) as a parallel anti-ferroptotic pathway independent of GPX4. FSP1 was shown to localize to the plasma membrane and mitochondria, where it reduces (CoQ10) to its form using NAD(P)H, thereby neutralizing peroxides and suppressing ferroptosis in glutathione-depleted cells.30282-7) This revealed a multi-layered defense system, expanding the understanding of ferroptosis beyond the glutathione-GPX4 axis. Throughout the , research advanced the metabolic underpinnings of ferroptosis, particularly the role of synthetase long-chain family member 4 (ACSL4) in incorporating polyunsaturated fatty acids (PUFAs) like into membrane phospholipids, thereby promoting susceptibility.30805-X) Recent 2024 studies further elucidated how ACSL4-mediated PUFA esterification drives ferroptosis in cancer cells, with inhibition of ACSL4 reducing PUFA-CoA formation and alleviating peroxidation in models of acute injury.00199-0) Concurrently, investigations into the revealed ferroptosis's dual role: sensitizing tumor cells to immune-mediated killing while protecting certain immune subsets, as evidenced by 2024-2025 analyses showing FSP1 upregulation in regulatory T cells to evade ferroptosis and suppress anti-tumor immunity. Technological advancements accelerated discovery, with genome-wide CRISPR-Cas9 screens identifying over 100 regulators of ferroptosis, including novel suppressors like MBOAT2 that modulate formation independently of and FSP1.00522-6) profiling has pinpointed specific peroxidation targets, such as phosphatidylethanolamines containing arachidonic and adrenic acids, which serve as primary executors of ferroptotic damage. These developments marked a from viewing ferroptosis primarily as a cancer-specific liability to recognizing its broad physiological significance, including contributions to and . By 2025, comprehensive reviews emphasized ferroptosis's involvement in aging through iron dysregulation and , positioning it as a driver of age-related diseases like neurodegeneration and , with potential interventions targeting regulators like to mitigate these processes.

Molecular Mechanisms

Iron-Dependent Lipid Peroxidation

Ferroptosis is characterized by the iron-dependent accumulation of lipid peroxides within cellular membranes, leading to oxidative damage and eventual cell death. This process is primarily driven by the Fenton reaction, in which ferrous iron (Fe²⁺) reacts with hydrogen peroxide (H₂O₂) to generate highly reactive hydroxyl radicals (OH•). The reaction can be represented as: \ce{Fe^{2+} + H2O2 -> Fe^{3+} + OH^\bullet + OH^-} These hydroxyl radicals initiate the peroxidation of polyunsaturated fatty acids (PUFAs) in phospholipids, distinguishing ferroptosis from other forms of cell death. The process unfolds in a comprising initiation, , and termination phases. During initiation, (ROS), such as those from the Fenton reaction, abstract a from a PUFA, forming a (L•). In the phase, this reacts with molecular oxygen to produce a lipid peroxyl radical (LOO•), which then attacks adjacent PUFAs, perpetuating the chain and amplifying oxidative damage to membrane integrity. Termination occurs when radicals are neutralized by antioxidants, such as (α-), which donates a to form non-radical products, thereby halting the cycle. This uncontrolled peroxidation in ferroptosis overwhelms cellular defenses, culminating in plasma membrane rupture.00260-X) The specificity of iron involvement in ferroptosis stems from the labile iron pool (), a cytosolic of chelatable Fe²⁺ derived from sources like transferrin-mediated uptake and ferritinophagy—the selective autophagic of . Ferritinophagy, regulated by coactivator 4 (NCOA4), releases iron from stores into the , elevating Fe²⁺ availability for Fenton chemistry. Additionally, lipoxygenases (LOXs), particularly arachidonate 15-lipoxygenase (ALOX15), enzymatically oxygenate PUFAs to hydroperoxides, accelerating peroxidation and synergizing with non-enzymatic ROS-driven damage. GPX4 counters these peroxides by reducing them to alcohols, but its inhibition shifts the balance toward ferroptosis. Recent 2025 research highlights ferroptosis's role in tumor immune evasion, where generates altered damage-associated molecular patterns (DAMPs), such as oxidized phospholipids, that modulate the by promoting immunosuppressive responses or dampening antitumor immunity. Quantitative models developed in 2025 have further defined peroxidation thresholds, integrating iron levels and composition to predict ferroptotic susceptibility in cellular contexts.

Role of Glutathione and GPX4

The glutathione peroxidase 4 (GPX4) system serves as the central antioxidant defense against ferroptosis by counteracting iron-dependent . (GSH), the primary substrate for GPX4, is synthesized through a multi-step process beginning with the import of cystine via the system xc- cystine/glutamate , composed of the SLC7A11 (xCT) and SLC3A2 (4F2hc) subunits.00520-X) Once inside the cell, cystine is reduced to cysteine, which then combines with glutamate via glutamate-cysteine ligase (GCL) to form γ-glutamylcysteine, followed by conjugation with by glutathione synthetase (GSS) to produce GSH. This GSH pool is essential for maintaining cellular balance and directly supports 's activity in preventing the accumulation of toxic lipid hydroperoxides. GPX4, a selenocysteine-containing , functions as the master regulator of ferroptosis suppression by reducing phospholipid hydroperoxides (PLOOH) to non-toxic alcohols, thereby halting the propagation of chains. The reaction catalyzed by GPX4 is: \text{PLOOH} + 2\text{GSH} \rightarrow \text{PLOH} + \text{GSSG} + \text{H}_2\text{O} where GSSG (oxidized ) is subsequently recycled back to GSH by using NADPH.01544-4) This unique ability of GPX4 to detoxify membrane-embedded lipid peroxides distinguishes it from other glutathione peroxidases, making it indispensable for ferroptosis resistance across various cell types. Depletion of GSH or inactivation of GPX4 leads to unchecked PLOOH buildup, driving cells toward ferroptotic death. Ferroptosis inducers target this axis at distinct points: erastin inhibits system xc-, thereby blocking cystine uptake and causing GSH depletion, while RSL3 covalently binds the of , directly impairing its enzymatic function.00520-X)01544-4) These mechanisms highlight the system's vulnerability and have been pivotal in defining ferroptosis pharmacologically. exists in three isoforms—cytosolic (cGPX4), mitochondrial (mGPX4), and (nGPX4)—each with specialized roles in ferroptosis , arising from transcription start sites and mRNA of the . While cGPX4 primarily protects cytoplasmic and plasma membranes from peroxidation, mGPX4 safeguards mitochondrial lipids, preventing oxidative damage to and other inner-membrane phospholipids during ferroptotic stress. Recent studies have revealed isoform-specific contributions, such as mGPX4's critical role in rescuing mitochondrial independently of cytosolic defenses. Emerging research has uncovered post-translational regulation of via ubiquitination, particularly in contexts like DNA damage response, where acts as an E3 ligase to catalyze K6-linked polyubiquitination of , promoting its proteasomal degradation and enhancing ferroptosis susceptibility in -proficient cells. This mechanism links genomic instability to ferroptotic vulnerability, offering new insights into how pathways intersect with defenses.

Regulatory Pathways and Proteins

Ferroptosis is regulated by multiple parallel pathways that operate independently of the canonical glutathione peroxidase 4 () system, providing additional layers of control over and cell death susceptibility. One key suppressor pathway involves ferroptosis suppressor protein 1 (FSP1), an FAD-dependent that uses NAD(P)H to reduce (CoQ10) to its active form. This reduction enables to act as a radical-trapping at the plasma membrane, thereby inhibiting and ferroptosis in a glutathione-independent manner. The FSP1-CoQ10 pathway functions in parallel to , and its overexpression correlates with enhanced resistance to ferroptotic inducers across diverse lines. Beyond FSP1, specific enzymes modulate ferroptosis sensitivity by altering the composition of cellular membranes. Acyl-CoA synthetase long-chain family member 4 (ACSL4) promotes ferroptosis by selectively activating polyunsaturated fatty acids (PUFAs), such as , into their CoA derivatives, which are then incorporated into phospholipids via lysophosphatidylcholine acyltransferase 3 (LPCAT3). This enrichment of PUFA-containing phospholipids in membranes increases their vulnerability to peroxidation, making ACSL4 essential for ferroptosis execution in sensitive cells. In contrast, stearoyl-CoA desaturase 1 (SCD1) confers resistance by catalyzing the desaturation of saturated fatty acids into monounsaturated fatty acids (MUFAs), such as oleate from stearate. These MUFAs dilute peroxidation-prone PUFAs in membranes, reducing lipid radical chain reactions and protecting cells, particularly in nutrient-deprived tumor microenvironments. Epigenetic and transcriptional regulators further fine-tune ferroptosis through genetic control of lipid and pathways. The nuclear factor erythroid 2-related factor 2 (NRF2) acts as a potent suppressor by directly upregulating genes involved in cystine uptake and defense, including 7 member 11 (SLC7A11) and , thereby enhancing cellular resilience to and ferroptosis induction. Conversely, the tumor suppressor exerts bidirectional control but promotes ferroptosis in certain contexts by transactivating spermidine/ N1-acetyltransferase 1 (SAT1), which acetylates polyamines to generate and activate 15 (ALOX15), accelerating PUFA peroxidation. SAT1-mediated ferroptosis is particularly evident under , where p53-SAT1 signaling sensitizes cells without relying on canonical apoptotic pathways. Recent advances as of 2025 have illuminated additional mitochondrial and metabolic sensing mechanisms. (DHODH), a mitochondrial inner in the pathway, suppresses ferroptosis by oxidizing ubiquinone to using dihydroorotate as an , thereby maintaining and preventing mitochondrial independently of cytoplasmic defenses. DHODH inhibition selectively induces ferroptosis in cancer cells with high metabolic demands, highlighting its therapeutic potential. Furthermore, interactions between mechanistic target of rapamycin () and (AMPK) integrate nutrient and energy sensing with ferroptosis regulation; activation promotes resistance by enhancing cystine uptake and , while AMPK activation under energy stress inhibits to sensitize cells to peroxidation through autophagy-dependent ferritin degradation (ferritinophagy). These pathways underscore how metabolic sensors dynamically modulate ferroptosis in response to environmental cues.

Biological Roles

In Embryonic Development and Tissue Differentiation

Ferroptosis is essential for proper embryonic patterning and , as its dysregulation leads to severe developmental defects. Global of the gene in mice results in embryonic lethality at approximately E7.5, characterized by widespread that disrupts early embryonic structures and prevents further . This lethality is attributed to uncontrolled ferroptosis in extraembryonic tissues, including trophoblasts, where GPX4 normally safeguards against oxidative damage during implantation and . In tissue-specific contexts, ferroptosis contributes to regulating cellular behaviors critical for formation. For instance, ferroptosis suppresses excessive invasion in the , maintaining balanced remodeling of maternal spiral arteries during early pregnancy; inhibition of ferroptosis pathways enhances motility and invasion , suggesting a regulatory role in preventing over-invasion that could lead to developmental abnormalities. Similarly, emerging evidence indicates that ferroptosis influences cell dynamics, with GPX4-mediated suppression of supporting proper migration and differentiation of these multipotent cells into diverse neural and non-neural lineages during embryogenesis. Within populations, ferroptosis modulates and survival during tissue specification. In hematopoietic s (HSCs), controlled ferroptosis promotes lineage commitment by eliminating undifferentiated cells susceptible to iron-dependent peroxidation, while deficiency accelerates ferroptosis and impairs HSC maintenance and potential. Conversely, ferroptosis protection is vital for neural progenitors, where prevents oxidative stress-induced death, enabling their proliferation and into mature neural tissues; suppression of ferroptosis via antioxidants like ensures proper laminar organization in developing cortices. Recent studies have further elucidated ferroptosis's involvement in developmental -like events. In 2024 research on limb development, markers of ferroptosis such as (4-HNE) were detected in dying cells, suggesting that ferroptotic trigger waves facilitate patterned cell elimination for proper digit separation, analogous to traditional but driven by propagation.

In Physiological Homeostasis

Ferroptosis plays a in maintaining balance within tissues by serving as a for iron levels, particularly in hepatocytes where excess iron accumulation triggers and to prevent systemic overload. In the liver, non-transferrin-bound iron uptake via transporters like SLC39A14 elevates labile iron pools, promoting Fenton reaction-mediated (ROS) production that initiates ferroptosis as a protective mechanism against physiological iron excess. Similarly, in adipocytes, ferroptotic signaling regulates storage by modulating polyunsaturated fatty acid (PUFA) peroxidation; activation of pathways involving ACSL4 and influences formation and , thereby preventing excessive fat accumulation and supporting metabolic . For instance, non-lethal ferroptosis agonists like RSL3 reduce deposits in adipocytes through a HIF1α-c-MYC-PGC1β axis, highlighting its adaptive role in energy balance. In the context of immune surveillance, basal ferroptosis in macrophages contributes to the non-inflammatory clearance of damaged or senescent cells, ensuring tissue integrity without excessive immune activation. Macrophages maintain low-level ferroptosis regulated by iron and activity, which supports monitoring and elimination of cellular debris while minimizing (DAMP) release. This process is iron-dependent and involves controlled , allowing macrophages to perform phagocytic functions efficiently in steady-state conditions. Links between ferroptosis and aging involve mild execution promoting by facilitating the removal of iron-laden, oxidatively stressed cells that could otherwise accumulate misfolded proteins. Iron dyshomeostasis during aging exacerbates , but regulated ferroptosis mitigates proteotoxic stress through targeted cell elimination, as evidenced by increased ferroptosis markers in aged models correlating with improved cellular cleanup. 2024 investigations further reveal ferroptosis's involvement in skin aging, where it contributes to barrier dysfunction through ROS accumulation, , and impaired epidermal integrity. Organ-specific contributions of ferroptosis underscore its protective functions against physiological ROS; in epithelial cells, susceptibility to iron-dependent peroxidation helps eliminate cells vulnerable to baseline , preserving transparency and function. Depletion of and downregulation of transporters like SLC7A11 in aging lenses heighten this response, acting as a safeguard mechanism. Likewise, ferroptosis influences cycling by modulating dermal papilla cell fate, where and lipid ROS disrupt the anagen phase, and its inhibition via pathways like Wnt/β-catenin promotes regenerative transitions.

In Disease Pathogenesis

Dysregulated ferroptosis contributes to the pathogenesis of numerous diseases by disrupting cellular through iron-dependent and . In conditions characterized by excessive ferroptosis, such as neurodegeneration, promotes lipid damage that exacerbates and neuronal loss. For instance, in , hyperphosphorylation and aggregation impair iron efflux, leading to intracellular iron accumulation that drives and ferroptotic cell death in neurons. Conversely, suppressed ferroptosis enables survival by evading -induced death, allowing tumors to thrive under . In cardiovascular diseases, ferroptosis plays a critical role in ischemia-reperfusion injury, where and reduced activity cause in cardiomyocytes and endothelial cells, leading to endothelial barrier dysfunction and exacerbated tissue damage. Recent evidence from 2024 highlights ferroptosis in COVID-19-associated injury, with post-mortem analyses showing elevated transferrin receptor 1 (TfR1) and (MDA) levels in acute injury cases, alongside depleted polyunsaturated fatty acid-containing phospholipids, indicating iron dysregulation as a driver of pulmonary pathology. Similarly, in , ferroptosis contributes to complications like and through iron deposition in pancreatic β-cells and vascular cells, impairing insulin secretion and promoting plaque instability via ACSL4-mediated . Ferroptosis forms pathogenic feedback loops that amplify , particularly through the release of high-mobility group box 1 (), a . During ferroptosis, induces HMGB1 translocation and extracellular secretion, which activates TLR4 and pathways, escalating proinflammatory production (e.g., TNF-α, IL-1β) and further promoting ferroptotic in affected tissues. Iron dysregulation in hereditary hemochromatosis, stemming from /ferroportin imbalances, heightens ferroptosis susceptibility in hepatocytes by fostering chronic and , contributing to liver damage. Ferroptosis exhibits biphasic effects in disease, where mild activation may confer protection by limiting excessive , as seen in certain inflammatory contexts, while severe ferroptosis becomes detrimental, driving widespread tissue injury and progression in disorders like neurodegeneration and ischemia. This threshold-like model underscores the context-dependent nature of ferroptosis, with iron and lipid levels determining outcomes.

Therapeutic Relevance

Applications in Cancer

Many cancer cells, particularly those with high proliferative demands, upregulate SLC7A11, a key component of the system xc⁻ cystine/glutamate antiporter, to import cystine for synthesis and thereby suppress ferroptosis. This dependency renders such tumors vulnerable to ferroptosis inducers like erastin, which inhibit SLC7A11-mediated cystine uptake, leading to and cell death. Notably, while elevated SLC7A11 expression drives growth by evading ferroptosis, it paradoxically inhibits , as disseminating cancer cells with high SLC7A11 become hypersensitive to ferroptotic stress in nutrient-poor environments. Therapeutic strategies exploiting ferroptosis in cancer increasingly involve combining inducers with to overcome resistance and enhance antitumor immunity. For instance, ferroptosis induction boosts tumor immunogenicity by releasing damage-associated molecular patterns, which recruit and activate T cells, synergizing with inhibitors like blockade to delay tumor progression and extend survival in preclinical models. In 2025, significant advances in nanoparticle-based delivery systems have emerged for , where targeted nanoparticles loaded with ferroptosis inducers cross the blood-brain barrier, selectively accumulate in tumor cells, and trigger iron-dependent while minimizing off-target effects. In late 2025, targeting ferroptosis suppressor protein 1 (FSP1) has emerged as a strategy to induce ferroptosis in , overcoming GPX4-independent resistance. Preclinical investigations of imidazole ketone erastin (), a metabolically erastin analog, have demonstrated potent ferroptosis induction and tumor growth suppression in xenografts, paving the way for . As of 2025, early-phase clinical trials, such as NCT06928649 (initiated March 2025), are investigating ferroptosis inhibitors in critically ill patients, with preclinical data supporting further to . In , activation of the ALOX15 enzyme, which catalyzes , sensitizes cells to ferroptosis via the ACSL4/LPCAT3/ALOX15 axis, highlighting its potential as a therapeutic target. Similarly, in gastric cancer, overcoming miR-522-mediated suppression of ALOX15 restores ferroptosis susceptibility, inhibiting tumor progression in models. Despite these promises, challenges persist in harnessing ferroptosis for cancer therapy, including resistance conferred by the , where stromal cells such as cancer-associated fibroblasts export to shield tumor cells from . Additionally, ferroptosis induction can upregulate expression on tumor cells, promoting immune evasion and dampening T-cell responses, which complicates combination with .

Role in Neurodegenerative Disorders

Ferroptosis contributes to neuronal loss in neurodegenerative disorders by exacerbating iron-dependent and , linking pathologies to mechanisms. In (AD), iron accumulation within amyloid-beta (Aβ) plaques drives in neurons and glia, promoting ferroptosis alongside tau hyperphosphorylation and synaptic dysfunction. Similarly, in (PD), aggregates in the disrupt iron homeostasis, inducing ferritinophagy and ferroptosis in neurons, which correlates with motor deficits. Recent evidence also implicates ferroptosis in (ALS), where mutations in the FUS protein heighten vulnerability through mitochondrial dysfunction and reduced xCT expression, leading to enhanced in motor neurons. Aging-related decline in glutathione peroxidase 4 (GPX4) activity in the brain further sensitizes neurons to ferroptosis, as GPX4 normally detoxifies lipid hydroperoxides; its reduction by 75-85% in conditional knockout models results in hippocampal neuronal degeneration and cognitive impairment. Mouse models of AD demonstrate that ferroportin (FPN) overexpression reduces iron overload and ferroptosis markers, improving memory performance, while GPX4 downregulation accelerates Aβ-induced neurodegeneration. In PD models using 6-hydroxydopamine (6-OHDA), ferroptosis inhibitors like ferrostatin-1 rescue dopaminergic neuron loss by blocking lipid peroxidation and preserving mitochondrial integrity. For ALS, human iPSC-derived motor neurons with FUS-P525L mutations exhibit dose-dependent ferroptosis sensitivity to inducers like erastin, which is mitigated by iron chelators such as deferoxamine. Therapeutic inhibition of ferroptosis shows promise in preclinical studies for neurodegenerative disorders. In tauopathy models mimicking AD pathology, liproxstatin-1 reduces tau hyperphosphorylation, restores levels, and alleviates (e.g., decreased and ), thereby enhancing neuronal survival and cognitive function in aged mice with perioperative neurocognitive dysfunction. A 2024 study on FUS-ALS cell models confirmed increased ferroptosis susceptibility, suggesting targeted interventions like MCU inhibitors could protect motor neurons. These findings position ferroptosis as a bridge between and proteinopathies, with inhibitors like ferrostatin-1 and liproxstatin-1 offering neuroprotective potential without affecting other pathways, though clinical translation requires addressing off-target risks.

Involvement in Ischemic and Organ Injuries

Ferroptosis plays a critical role in ischemia-reperfusion (I/R) injury, where post-ischemic iron release from damaged cells triggers in endothelial cells, exacerbating tissue damage. This process is particularly evident in (AKI), where ferroptosis drives tubular epithelial cell death, contributing to renal dysfunction through iron-dependent accumulation of lipid peroxides. In the kidney, ischemia leads to labile iron release from , promoting ferroptosis via 4 (GPX4) inhibition and subsequent propagation of . In the heart, ferroptosis occurs in cardiomyocytes following (MI), where reperfusion after ischemia induces and GPX4 downregulation, leading to cell death and impaired cardiac function. Quantitative proteomics has shown that GPX4 reduction during directly contributes to ferroptotic cardiomyocyte loss, worsening infarct expansion. Similarly, in the liver, ferroptosis is implicated in non-alcoholic steatohepatitis (NASH), where dysregulated iron metabolism and in hepatocytes initiate inflammation and , bridging metabolic stress to organ . Mechanistically, hypoxia-inducible factor (HIF)-1α modulates SLC7A11 expression during reperfusion, enhancing cystine uptake to suppress ferroptosis by maintaining levels and activity. This regulation helps mitigate in post-ischemic tissues, though its dysregulation amplifies injury. Therapeutic interventions targeting ferroptosis show promise in reducing ischemic damage. Iron with decreases infarct size in myocardial I/R models by limiting labile iron availability and preventing endothelial ferroptosis. In models, 2025 studies demonstrate that NRF2 activators, such as , alleviate neuronal ferroptosis by upregulating SLC7A11/ and reducing , improving neurological outcomes. These findings highlight ferroptosis inhibition as a viable strategy for organ protection in acute ischemic events.

Implications for Immune and Inflammatory Conditions

Ferroptosis exhibits a complex role in immune regulation, particularly within T cells and neutrophils, influencing the balance between protective and pathological responses. In + T cells, the of ferroptosis in tumor cells via interferon-gamma (IFN-γ) secretion activates the JAK-STAT1 pathway, downregulating SLC7A11 and upregulating ACSL4 to promote and tumor cell death. This process releases damage-associated molecular patterns (DAMPs) such as and , which enhance maturation and subsequent + T cell activation, thereby bolstering anti-tumor immunity. Conversely, ferroptosis susceptibility in + T cells themselves, driven by iron accumulation and glutathione peroxidase 4 (GPX4) inhibition within the , impairs their effector functions and proliferation, highlighting the need to protect these cells to sustain anti-tumor responses. In neutrophils, ferroptosis modulates inflammatory resolution; while dysregulated ferroptosis promotes excessive recruitment and tissue damage through lipid-reactive oxygen species (lipid-ROS), controlled ferroptosis limits hyperactivation by facilitating neutrophil clearance and reducing pro-inflammatory mediator release. Inhibitors like ferrostatin-1 attenuate this recruitment in inflammatory models, underscoring ferroptosis's potential to curb neutrophil-driven escalation. In autoimmune diseases, ferroptosis dysregulation contributes to chronic inflammation and tissue pathology. In systemic lupus erythematosus (SLE), neutrophil ferroptosis is induced by IFN-α-mediated suppression of expression, leading to lipid-ROS accumulation, , and systemic through the release of inflammatory mediators. This process exacerbates disease manifestations, including immune complex deposition and organ damage. In inflammatory bowel disease (IBD), ferroptosis in intestinal epithelial cells (IECs), regulated by proteins like NEDD4L, compromises the mucosal barrier by promoting and cell death, thereby allowing microbial translocation and amplifying immune activation. Recent 2024-2025 studies on (RA) reveal elevated iron levels and ferroptosis markers in synovial tissues, where lipid peroxides drive fibroblast-like synoviocyte (FLS) proliferation and erosion; for instance, ASIC1a inhibition reduces ferroptosis, while BACH1 downregulation mitigates FLS destructive behavior in hypoxic synovium. Ferroptosis intersects with immune signaling pathways to shape inflammatory outcomes, often inducing immunogenic cell death (ICD) that activates adaptive responses. Early-stage ferroptosis in dying cells releases DAMPs like ATP and , triggering P2X7 receptor-dependent maturation—marked by upregulated , , and expression—and eliciting protective anti-tumor or anti-pathogen immunity in preclinical models. In viral infections such as , upregulates ferroptosis and inflammasome-related gene signatures. Therapeutically, targeting ferroptosis offers promise for mitigating immune-mediated . Ferrostatin-1, a potent ferroptosis inhibitor, reduces synovial inflammation by blocking PAD4-mediated citrullinated histone H3 in FLS, thereby decreasing MHC-I expression, CD8+ T cell proliferation, and pro-inflammatory like TNF-α and IFN-γ in collagen-induced models. In broader inflammatory contexts, such as and acute , ferrostatin-1 attenuates cytokine storms by preserving activity, lowering ROS and , and curbing polarization toward pro-inflammatory states. These effects position ferroptosis inhibitors as adjuncts to immunomodulatory therapies in autoimmune and infectious diseases.

Modulators of Ferroptosis

Synthetic Inducers

Synthetic inducers of ferroptosis are small-molecule compounds engineered to trigger this iron-dependent form of regulated by disrupting defenses, primarily through targeting key pathways such as uptake or activity. These agents have garnered attention for their potential in selectively eliminating therapy-resistant cancer cells, where ferroptosis sensitivity is often heightened due to metabolic vulnerabilities. Unlike natural modulators, synthetic inducers offer tunable and can be optimized for specificity, though challenges like off-target effects persist. Major classes of synthetic inducers include inhibitors of the system xc⁻ cystine/, which deplete intracellular and indirectly impair 4 () function. Erastin, a prototypical type I inducer, binds to voltage-dependent anion channels and inhibits system xc⁻, leading to and ferroptosis in and other tumor cells with minimal impact on non-transformed cells.00613-0) Similarly, , an FDA-approved anti-inflammatory drug repurposed for ferroptosis induction, blocks system xc⁻ and has demonstrated efficacy in preclinical models of by promoting . Another class comprises direct inhibitors, such as RSL3, which covalently binds to 's residue, abolishing its lipid hydroperoxide reduction capacity and rapidly inducing ferroptosis in diverse cancer lines including renal and breast carcinomas.00817-4) FIN56, a related compound, indirectly targets by depleting while also activating synthase, resulting in ferroptotic death in HT-1080 cells. Additionally, p53-activating agents like APR-246 (eprenetapopt) enhance ferroptosis sensitivity by upregulating p53-dependent suppression of cystine uptake and SLC7A11 expression, as observed in models. Recent developments as of 2025 have focused on piperazine-based scaffolds to improve potency and tumor specificity. For instance, 1,8-naphthalimide-piperazine derivatives, such as compound 9o, act as IX inhibitors that concurrently induce ferroptosis via degradation in cells, showing values in the low micromolar range and reduced viability in hypoxic environments. In glioblastoma therapy, has advanced targeted delivery of these inducers; lipid nanoparticles encapsulating erastin or RSL3 cross the blood-brain barrier and release payloads in response to tumor . These systems leverage coatings for prolonged circulation and achieve higher intratumoral drug accumulation compared to free compounds. Selectivity remains a key focus, with compounds targeting mitochondrial ferroptosis pathways to minimize systemic toxicity. (DHODH) inhibitors, such as brequinar, disrupt ubiquinone regeneration in mitochondria, inducing localized independent of cytosolic and effectively killing GPX4-low renal cancer cells with EC50 around 1 μM. However, off-target effects include gastrointestinal irritation from erastin-like agents and risks with at doses exceeding 2 g/day, necessitating scavengers like ferrostatin-1 for in preclinical studies. Toxicity profiles indicate that inhibitors like RSL3 exhibit narrow therapeutic windows, primarily due to hepatic . In preclinical models, synthetic inducers demonstrate synergy with to overcome resistance in solid tumors. Combining erastin with enhances ferroptosis in platinum-resistant xenografts, reducing tumor volume by 60% versus alone through amplified ROS and exhaustion. Erastin is commonly used in preclinical models at doses of 10-50 mg/kg intraperitoneally. Preclinical studies continue to explore formulations to improve oral . FIN56 has been used in combination therapies, such as with in models, to potentiate efficacy against multidrug-resistant strains. As of November 2025, ongoing clinical trials are investigating ferroptosis inducers, including analogs of erastin and RSL3, in Phase I/II studies for various cancers.

Synthetic Inhibitors

Synthetic inhibitors of ferroptosis are small-molecule compounds designed to block this form of regulated by targeting key pathways such as , iron , or glutathione peroxidase 4 (GPX4) activity. These agents have emerged as promising cytoprotective tools in preclinical models of ferroptosis-related pathologies, including (AKI), , and neurodegenerative disorders. Unlike natural modulators, synthetic inhibitors are engineered for enhanced potency, selectivity, and pharmacokinetic profiles, though challenges like metabolic instability persist. Prominent examples include ferrostatin-1 (Fer-1), a lipophilic radical-trapping (RTA) that scavenges lipid peroxyl radicals to prevent peroxidation, the core executioner of ferroptosis. Originally identified through , Fer-1 potently inhibits ferroptosis induced by agents like erastin or RSL3 in cellular models. Liproxstatin-1 functions similarly as an RTA but exhibits superior efficacy due to its balanced and reduced reactivity with biological nucleophiles, effectively halting without broadly disrupting cellular redox balance. , an established agent repurposed as an iron chelator, sequesters labile iron to disrupt Fenton chemistry and suppress (ROS) generation, thereby blocking ferroptosis at an upstream node. Additionally, UAMC-3203 acts as a synthetic RTA that stabilizes by enhancing its activity, promoting the reduction of hydroperoxides to non-toxic alcohols and preserving cellular integrity under oxidative stress.00120-4) These inhibitors target multiple nodes in the ferroptosis cascade, offering versatility across disease contexts. For instance, Fer-1 and liproxstatin-1 reduce AKI severity in cisplatin-treated mouse models by mitigating tubular cell ferroptosis, with liproxstatin-1 demonstrating up to 80% protection against renal dysfunction. In models, such as occlusion in rodents, Fer-1 administration post-ischemia limits infarct size and neurological deficits by curbing neuronal . For neurodegeneration, and Fer-1 analogs protect dopaminergic neurons in (PD) models by chelating excess brain iron, a ferroptosis trigger. Clinical analogs, including the iron chelator , have been evaluated in Phase II trials. A 2022 trial showed reduced substantia nigra iron deposition in early PD patients over 36-52 weeks but did not slow motor progression and was associated with worsening symptoms and higher adverse events. Recent advancements include formulations to enhance blood-brain barrier penetration for targeted delivery in PD and models, improving and efficacy while minimizing systemic toxicity. Despite their promise, synthetic ferroptosis inhibitors face limitations that hinder broad clinical translation. Fer-1 and liproxstatin-1 suffer from short half-lives (under 1 hour), necessitating frequent dosing or formulation improvements to sustain therapeutic levels. In cancer contexts, their cytoprotective effects may inadvertently promote tumor survival by shielding malignant cells from ferroptosis-dependent therapies, raising concerns for off-target risks in applications. Ongoing focuses on structure-activity optimization to address these issues, prioritizing compounds with extended and tissue-specific targeting.

Endogenous and Natural Modulators

Endogenous modulators of ferroptosis play critical roles in maintaining cellular by regulating key pathways and cofactors involved in suppression. The Nrf2 pathway, a master regulator of responses, transcriptionally induces the expression of (GPX4), the primary enzyme that detoxifies lipid hydroperoxides to prevent ferroptosis execution. Activation of Nrf2 enhances resistance to ferroptotic inducers by upregulating GPX4 and other protective genes, thereby buffering in various cell types. Similarly, serves as an essential cofactor for GPX4 activity through its incorporation into the residue at the enzyme's , directly enabling the reduction of hydroperoxides and suppressing ferroptosis sensitivity. Deficiencies in selenium impair GPX4 function, heightening vulnerability to iron-dependent cell death. Iron is another pivotal endogenous mechanism, with acting as a primary for labile iron pools to limit Fenton chemistry and . sequesters excess intracellular iron, preventing its availability for generation and thereby inhibiting ferroptosis; disruption of ferritinophagy—the autophagic degradation of —further protects cells by maintaining iron storage integrity. Hormonal factors also tune ferroptosis thresholds, as upregulates system xc- cystine/ antiporter component SLC7A11 in estrogen receptor-positive cells, enhancing cystine uptake and synthesis to confer resistance against ferroptosis, particularly in female-specific contexts like breast tissue. Natural dietary compounds further modulate ferroptosis through and enzymatic inhibitory actions. Polyphenols, such as derived from , inhibit lipoxygenases () that catalyze polyunsaturated oxidation, thereby attenuating and ferroptosis induction in oxidative environments. (α-tocopherol), a lipophilic chain-breaking , intercepts propagating lipid peroxyl radicals and regenerates coenzyme Q, serving as an endogenous ferroptosis suppressor by directly mitigating damage. Tea catechins, including (EGCG) from , activate protective pathways like Nrf2/SLC7A11/ to inhibit ferroptosis, contributing to by reducing and iron-mediated in tumor-prone tissues. Recent insights highlight the influence of gut -derived metabolites on ferroptosis regulation, particularly in inflammatory conditions. Butyrate, a short-chain produced by fermentation of dietary fibers, suppresses ferroptosis in intestinal epithelial cells during (IBD) by modulating ERK/ signaling, restoring levels, and alleviating ; this effect is linked to microbiome composition shifts that enhance butyrate production. These findings underscore how dietary and microbial factors intersect to fine-tune ferroptosis sensitivity in gastrointestinal .

References

  1. [1]
    Ferroptosis: past, present and future | Cell Death & Disease - Nature
    Feb 3, 2020 · Ferroptosis is a new type of cell death that was discovered in recent years and is usually accompanied by a large amount of iron accumulation and lipid ...
  2. [2]
    Ferroptosis: molecular mechanisms and health implications - Nature
    Dec 2, 2020 · Ferroptosis is defined as an iron-dependent regulated necrosis that is caused by massive lipid peroxidation-mediated membrane damage.
  3. [3]
    The cell biology of ferroptosis - Nature
    Feb 16, 2024 · In this Review, we examine ferroptosis mechanisms at the cellular level. We first describe the events that lead to lethal plasma membrane ...
  4. [4]
    Ferroptosis in cancer: from molecular mechanisms to therapeutic ...
    Mar 8, 2024 · Here, we review the major milestones and molecular machinery of ferroptosis, including drivers and defenses two systems. Then, we decipher the ...
  5. [5]
    Metabolic cell death in cancer: ferroptosis, cuproptosis, disulfidptosis ...
    This review discusses the mechanisms of metabolic cell death—ferroptosis, cuproptosis, disulfidptosis, lysozincrosis, and alkaliptosis—and explores their ...
  6. [6]
    SLC7A11 as a bridge between ferroptosis and disulfidptosis - NIH
    Oct 27, 2025 · These findings suggest that there is an intrinsic relationship between ferroptosis and disulfidptosis and that SLC7A11 connects the regulation ...
  7. [7]
    Insights on the crosstalk among different cell death mechanisms
    Feb 10, 2025 · This comprehensive review delves into the intricate molecular mechanisms, inducers, and inhibitors of the underlying prevalent forms of cell death.
  8. [8]
    Targeting regulated cell death: Apoptosis, necroptosis, pyroptosis ...
    Mar 19, 2025 · (D) Ferroptosis is a form of cell death driven by iron-dependent oxidative stress. Its hallmark is the accumulation of lipid peroxidation within ...
  9. [9]
    Ferroptosis: mechanisms, biology, and role in disease - PMC
    May 24, 2021 · Ferroptosis is now appreciated as likely one of the most widespread and ancient forms of cell death, as although originally studied in mammalian ...
  10. [10]
    The biochemical pathways of apoptotic, necroptotic, pyroptotic, and ...
    Jan 4, 2024 · Ferroptosis is an oxidative and iron-dependent form of regulated cell death, caused by lipid peroxidation of the polyunsaturated fatty acid ( ...
  11. [11]
    Ferroptosis and pyroptosis are connected through autophagy
    Jan 17, 2025 · Ferroptosis and pyroptosis can be affected by various types of autophagy. Therefore, ferroptosis and pyroptosis have crosstalk via autophagy, ...
  12. [12]
    Iridium(III) Photosensitizers Induce Simultaneous Pyroptosis and ...
    Aug 23, 2024 · Our work introduces the first metal-based small molecule dual-inducers of pyroptosis and ferroptosis for potent cancer immunotherapy, and ...Missing: ferroptotic- | Show results with:ferroptotic-
  13. [13]
    The specific amino acid requirements of a mammalian cell (strain L ...
    The specific amino acid requirements of a mammalian cell (strain L) in tissue culture. J Biol Chem. 1955 Jun;214(2):839-52. Author. H EAGLE. PMID: 14381421. No ...Missing: cystine | Show results with:cystine
  14. [14]
    Ferroptosis and the tumor microenvironment
    Nov 30, 2024 · This review provides a comprehensive and systematic summary of the current findings concerning the roles of ferroptosis in the TME and how ...Missing: paper | Show results with:paper
  15. [15]
    Ferroptosis in the tumor microenvironment: mechanisms, advances ...
    This mini-review summarizes recent advances in ferroptosis mechanisms and highlights the dynamic interplay between ferroptosis and the TME. It ...Missing: paper | Show results with:paper
  16. [16]
    Modulating Ferroptosis in Aging: The Therapeutic Potential of ...
    Jul 4, 2025 · Our review highlights that ferroptosis is not merely a consequence of aging but an active contributor to age-related pathologies, intricately ...
  17. [17]
    Inflammation in a ferroptotic environment - Frontiers
    Sep 19, 2024 · In this review, we discuss how ferroptosis alters inflammatory pathways and how iron metabolism under inflammatory conditions affects immune cell ferroptosis.
  18. [18]
    Hypoxia aggravates ferroptosis in RPE cells by promoting ... - Nature
    Jul 29, 2022 · Ferroptotic cell death depends on the Fenton reaction where H2O2 and iron react to generate hydroxyl radicals which trigger lipid peroxidation.
  19. [19]
    Ferroptosis at the intersection of lipid metabolism and cellular ...
    As the committing event of ferroptosis, PL peroxidation occurs in three steps: initiation, propagation, and termination (Figure 3). The peroxidation of fatty ...
  20. [20]
    Unsolved mysteries: How does lipid peroxidation cause ferroptosis?
    May 24, 2018 · The second step is the creation of new lipid radicals (propagation). The final step is termination, either by antioxidants or another radical.
  21. [21]
    Mechanistic elucidation of ferroptosis and ferritinophagy
    Jul 3, 2024 · A minor fraction of reduced Fe2+ is sequestered within the labile iron pool (LIP) as an intermediary for iron input, storage, and utilization ( ...
  22. [22]
    Iron metabolism and ferroptosis in human health and disease
    Aug 22, 2025 · NCOA4-mediated ferritinophagy has been shown to trigger ferroptosis by degrading ferritin and inducing iron overload [31]. Tf enhances cellular ...
  23. [23]
    Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ...
    Aug 9, 2016 · We report here the mechanism of lipid peroxidation during ferroptosis, which involves phosphorylase kinase G2 (PHKG2) regulation of iron availability to ...
  24. [24]
    Targeting lipoxygenases to suppress ferroptotic cell death - PMC - NIH
    Jul 5, 2023 · ... lipoxygenases (15-LOX1 and 15-LOX2) have been most often implicated in ferroptosis execution (8–11). Lipoxygenases (LOXs) are nonheme iron ...
  25. [25]
    Tissue-specific iron levels modulate lipid peroxidation and ... - Nature
    Sep 2, 2025 · Our findings show that FLASH RT increases lipid peroxidation and induces ferroptosis in tumor cells but does not significantly elevate lipid ...
  26. [26]
    Glutathione peroxidase 4 (Gpx4) and ferroptosis - PubMed Central
    The system XC−/glutathione/glutathione peroxidase 4 (Gpx4) axis pivotally controls ferroptosis, a recently described form of regulated non-apoptotic cell death.
  27. [27]
    GPX4 in cell death, autophagy, and disease - PMC - PubMed Central
    GPX4 is present as three physiological isoforms: cytosolic (cGPX4), mitochondrial (mGPX4), and nuclear (nGPX4; Figure 2). The structural integrity of mammalian ...
  28. [28]
    Mitochondrial regulation of ferroptosis | Journal of Cell Biology
    Jul 30, 2021 · Notably, overexpression of mitochondrial GPX4, but not cytosolic GPX4 or FSP1, can rescue mitochondrial lipid peroxidation and ferroptotic cell ...
  29. [29]
    Targeting GPX4-mediated ferroptosis protection sensitizes BRCA1 ...
    We report that BRCA1 promotes ferroptosis susceptibility through catalyzing K6-linked polyubiquitination of GPX4 and subsequently accelerating GPX4 degradation.
  30. [30]
    The CoQ oxidoreductase FSP1 acts in parallel to GPX4 to inhibit ...
    We further find that FSP1 expression positively correlates with ferroptosis resistance across hundreds of cancer cell lines, and that FSP1 mediates resistance ...
  31. [31]
    ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid ...
    Nov 14, 2016 · ACSL4 was preferentially expressed in a panel of basal-like breast cancer cell lines and predicted their sensitivity to ferroptosis.Missing: seminal paper
  32. [32]
    Steroyl-CoA Desaturase 1 (SCD1) protects ovarian cancer cells from ...
    Jul 3, 2019 · We next explored the possibility that downregulation of SCD1 might induce ferroptosis by decreasing synthesis of cytoprotective lipids. CoQ ...
  33. [33]
    The critical role of NRF2 in mediating ferroptosis - PubMed Central
    For example, a study in 1955 by Harry Eagle and colleagues identified that cystine deprivation resulted in a morphologically distinct mode of cell death ...
  34. [34]
    Ironing out how p53 regulates ferroptosis - PNAS
    Oct 17, 2016 · ... SAT1 markedly reduces p53-mediated ferroptosis. Mechanistically, Ou et al. show that increased SAT1 leads to increased levels of ALOX15, an ...
  35. [35]
    DHODH-mediated mitochondrial redox homeostasis: a novel ...
    Emerging evidence highlights that DHODH suppresses mitochondrial ferroptosis by restoring coenzyme Q10 (CoQH2) levels through mitochondrial membrane electron ...
  36. [36]
    Energy stress-mediated AMPK activation inhibits ferroptosis - PMC
    We found that ferroptotic cell death and lipid peroxidation can be inhibited by treatments that induce or mimic energy stress. Inactivation of AMP-activated ...
  37. [37]
    Glutathione Peroxidase 4 Senses and Translates Oxidative Stress ...
    Sep 3, 2008 · Investigations with litters at different gestational days revealed that GPx4-knockout mice died at the same embryonic stage (E7.5) as reported ...Results · Gpx4 Depletion Causes... · Gpx4 Disruption Causes...Missing: original paper
  38. [38]
    Ferroptosis: death by lipid peroxidation - PMC - PubMed Central
    Knockout of Gpx4 in mice caused embryonic lethality between E7.5 and E8.5 ... Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice.Missing: trophoblasts | Show results with:trophoblasts
  39. [39]
    Iron Metabolism and Ferroptosis in Physiological and Pathological ...
    As a major suppressor of ferroptosis, GPX4 is essential for the viability and function of human trophoblasts. Mutations or reduced expression of GPX4 resulted ...Iron Metabolism And... · 3. Adaptions Of Iron... · 3.3. Iron Transport By...
  40. [40]
    The regulated cell death at the maternal-fetal interface - Nature
    Feb 26, 2024 · Pyroptotic cells can release HMGB1, which inhibits trophoblast invasion. Ferroptosis inhibits trophoblast invasion. C Trophoblast cells ...
  41. [41]
    Ferroptosis-Related Genes in Neurodevelopment and Central ...
    In early phase of development, neurons migrate by somal translocation where their cell body extends toward the pial surface and then the nucleus moves up [84,85] ...
  42. [42]
    GPX4 and vitamin E cooperatively protect hematopoietic stem and ...
    Jul 15, 2021 · Deficiency of both GPX4 and vitamin E induces HSPC ferroptosis and impairs homeostasis of the hematopoietic system in vivo. Vitamin E is absent ...
  43. [43]
    Suppression of ferroptosis by vitamin A or radical-trapping ... - Nature
    Sep 1, 2024 · Suppression of ferroptosis by radical-trapping antioxidants or by vitamin A is required to obtain mature neurons and proper laminar organization in cortical ...
  44. [44]
    [PDF] Embryonic development mediated by ferroptotic trigger waves
    Moreover, combining ferroptosis inhibitors with other treatments, such as anti-inflammatory agents or apoptosis inhibitors, might enhance effective- ness.Missing: bud | Show results with:bud
  45. [45]
    The multifaceted role of ferroptosis in liver disease - Nature
    Jan 24, 2022 · Under iron-overload conditions, excess iron results in redox-active non-transferrin-bound iron (NTBI), the uptake of which is mediated by metal ...
  46. [46]
  47. [47]
  48. [48]
    Ferroptosis: a cell death connecting oxidative stress, inflammation ...
    Jul 26, 2021 · Ferroptosis is closely related to iron and lipid metabolism and triggered by various physiological conditions and pathological stresses in human ...
  49. [49]
    Implication of ferroptosis in aging | Cell Death Discovery - Nature
    Jun 28, 2021 · Age related decline of immunity and redox imbalances leads to chronic inflammation and induction of hepcidin resulting in iron dyshomeostasis ...Role Of Iron And Hepcidin In... · Examples Of Implication Of... · Controlling Ferroptosis For...<|control11|><|separator|>
  50. [50]
    Skin Aging and the Upcoming Role of Ferroptosis in Geroscience
    It was discovered that ferroptosis, or iron-dependent cell death, is linked to aging and skin diseases.6.1. Cell Death For Skin... · 6.1. 2. Ferroptosis In Skin... · Ferroptosis And Autoimmune...
  51. [51]
    Aging lens epithelium is susceptible to ferroptosis - PubMed - NIH
    May 1, 2021 · We are suggesting that LECs are highly susceptible to ferroptosis. Moreover, aged and cataractous human lenses may possess more pro-ferroptotic criteria.Missing: physiological | Show results with:physiological
  52. [52]
    Ellagic acid inhibits dihydrotestosterone-induced ferroptosis and ...
    Aug 10, 2024 · EA inhibits DHT-induced ferroptosis and promotes hair regrowth in mice by activating the Wnt/β-catenin signaling pathway.
  53. [53]
    Understanding the Mechanism of Ferroptosis in Neurodegenerative ...
    Aug 20, 2024 · High phosphorylation and aggregation of Tau protein may impede this outflow and cause accumulation of neuronal Fe, thus further promoting the ...
  54. [54]
    Ferroptosis: principles and significance in health and disease
    Jun 6, 2024 · Ferroptosis, an iron-dependent form of cell death characterized by uncontrolled lipid peroxidation, is governed by molecular networks ...Missing: paper | Show results with:paper
  55. [55]
    Role of ferroptosis in the pathogenesis of heart disease - Frontiers
    Ferroptosis has been implicated in the pathogenesis of multiple forms of heart disease such as myocardial infarction, cardiac hypertrophy, and various ...
  56. [56]
    Fatal COVID-19 pulmonary disease involves ferroptosis - Nature
    May 20, 2024 · Hyperferritinemia and disrupted lung iron homeostasis in COVID-19 patients imply that ferroptosis, an iron-dependent cell death, may occur.
  57. [57]
    Ferroptosis and metabolic syndrome and complications
    Jan 8, 2024 · Recent studies have found that the major molecules of ferroptosis are linked to common metabolic syndrome consequences, such as T2DM and ...
  58. [58]
    Involvement of HMGB1-mediated ferroptosis in systemic diseases
    HMGB1-mediated ferroptosis in pulmonary endothelial cells exacerbates sterile inflammation during IRI (Chongwu Li et al., 2024). In rodent models, IRI ...
  59. [59]
    Iron homeostasis and ferroptosis in human diseases - Nature
    Oct 14, 2024 · Ferroptosis is primarily linked to disruptions in iron metabolism, lipid peroxidation, and antioxidant system imbalance.
  60. [60]
    Cystine transporter SLC7A11/xCT in cancer: ferroptosis, nutrient ...
    Recent studies revealed that SLC7A11 overexpression promotes tumor growth partly through suppressing ferroptosis, a form of regulated cell death induced by ...
  61. [61]
    SLC7A11 expression level dictates differential responses to ... - Nature
    Jun 21, 2023 · We further show that high overexpression of SLC7A11 promotes tumor growth but suppresses tumor metastasis, likely because metastasizing cancer ...
  62. [62]
    Enhanced cancer immunotherapy through synergistic ferroptosis ...
    Nov 13, 2023 · Combining ferroptosis inducer and PD-L1 nanoblockade significantly delayed tumor progression and extended the survival of mice bearing ...
  63. [63]
    Ferroptosis driven by nanoparticles for tackling glioblastoma
    Feb 28, 2025 · Targeted nanoparticles can deliver ferroptosis inducers specifically to GBM cells, thereby enhancing treatment specificity and effectiveness.
  64. [64]
    Imidazole ketone erastin induces ferroptosis and slows tumor growth ...
    We aimed to investigate whether IKE specifically inhibited system xc− and induced ferroptosis in DLBCL cells. Previous studies found that IKE inhibited ...
  65. [65]
    Mechanism of ferroptosis in breast cancer and research progress of ...
    Dec 22, 2023 · Ferroptosis, a form of programmed cell death dependent on iron, is characterized by the accumulation of lipid peroxides, elevated levels of iron ...<|separator|>
  66. [66]
    Ferroptosis and its current progress in gastric cancer - Frontiers
    ... ALOX15 in cancer cells by secreting exosomal miR-522. This action inhibits lipid-ROS production in cancer cells, thereby preventing ferroptosis in GC cells ...
  67. [67]
    Modulating ferroptosis sensitivity: environmental and cellular targets ...
    Jan 13, 2024 · This review begins by delving into the intricate reciprocal regulation of ferroptosis between cancer and immune cells.Ferroptosis Defense... · Reprogrammed Lipids... · Ferroptosis Synergy In...<|separator|>
  68. [68]
  69. [69]
  70. [70]
  71. [71]
  72. [72]
    Ferroptosis: A Novel Therapeutic Target for Ischemia-Reperfusion ...
    Aug 9, 2021 · There is increasing evidence of a close association between ferroptosis and I/R injury, with ferroptosis frequently identified as a new ...
  73. [73]
    Targeting ferroptosis in acute kidney injury | Cell Death & Disease
    Feb 24, 2022 · Ferroptosis induced a first wave of death, triggering an inflammatory response that in turn contributed to the deterioration of renal function.
  74. [74]
    Ferroptosis and Acute Kidney Injury (AKI): Molecular Mechanisms ...
    Apr 19, 2022 · Overall, ferroptosis plays a key role in the progression of AKI-induced renal injury. Given that there is no effective treatment for AKI, ...Ferroptosis And Aki Caused... · Ferroptosis And... · Ferroptosis In Nephrotoxic...
  75. [75]
    (+)-JQ-1 alleviates cardiac injury in myocardial infarction by ... - Nature
    Jul 23, 2025 · Quantitative proteomic analyses reveal that GPX4 downregulation during myocardial infarction contributes to ferroptosis in cardiomyocytes.
  76. [76]
    The interaction between ferroptosis and myocardial ischemia ...
    Jul 21, 2025 · Ferroptosis is a novel form of cell death closely related to cellular damage in MIRI. Numerous studies have demonstrated that specific active ...
  77. [77]
    Hepatic ferroptosis plays an important role as the trigger for initiating ...
    Jun 18, 2019 · These findings suggest that hepatic ferroptosis plays an important role as the trigger for initiating inflammation in steatohepatitis.
  78. [78]
    Hydroxysafflor Yellow A Alleviates Acute Myocardial Ischemia ...
    Jul 31, 2023 · In summary, HSYA alleviated MI/RI by activating the HIF-1α/SLC7A11/GPX4 signaling pathway, thereby inhibiting ferroptosis. Keywords: HIF-1α/ ...
  79. [79]
    HIF1α/SLC7A11 signaling attenuates 6-hydroxydopamine-induced ...
    The collective findings of this study indicate that HIF1α can inhibit ferroptosis by positively regulating SLC7A11.
  80. [80]
    Mangiferin mitigates neurological deficits and ferroptosis via NRF2 ...
    May 21, 2025 · MGF protects neurological function and suppresses ferroptosis via activating NRF2/ARE pathway in ischemic stroke rats.
  81. [81]
    Nrf2 alleviates acute ischemic stroke induced ferroptosis via ...
    The present study confirmed that Nrf2 could attenuate AIS-induced neuronal ferroptosis and oxidative stress by regulating xCT/GPX4.
  82. [82]
    Ferroptosis: CD8+T cells' blade to destroy tumor cells or poison for ...
    Apr 1, 2025 · Ferroptosis represents an emerging, iron-dependent form of cell death driven by lipid peroxidation. In recent years, it has garnered significant attention in ...
  83. [83]
    Ferroptosis in infection, inflammation, and immunity
    May 12, 2021 · Ferroptosis is a type of regulated necrosis that is triggered by a combination of iron toxicity, lipid peroxidation, and plasma membrane damage.
  84. [84]
    Glutathione peroxidase 4 regulated neutrophil ferroptosis induces ...
    Our data demonstrate that neutrophil ferroptosis is an important driver of neutropenia in SLE and heavily contributes to disease manifestations.
  85. [85]
    NEDD4L mediates intestinal epithelial cell ferroptosis to restrict ... - JCI
    Dec 17, 2024 · NEDD4L is an important regulator in IEC ferroptosis, maintaining intestinal homeostasis, making it a potential clinical target for diagnosing and treating IBDs.
  86. [86]
    Ferroptosis: New Strategies for Clinical Treatment of Rheumatoid ...
    May 21, 2025 · Studies have found that iron levels in synovial tissue and fluid are elevated in RA patients, and ferroptosis is increased in chondrocytes.
  87. [87]
    Ferroptosis becomes immunogenic: implications for anticancer ...
    Dec 29, 2020 · Ferroptosis is an iron-dependent, non-apoptotic cell death that can be immunogenic, releasing DAMPs and triggering dendritic cell maturation. ...
  88. [88]
  89. [89]
    Ferrostatin-1 reduces the inflammatory response of rheumatoid ...
    Mar 6, 2025 · These findings collectively suggest that ferroptosis levels are greater in the synovial cells of RA patients than in those of osteoarthritis (OA) ...
  90. [90]
    Ferrostatin-1 improves acute sepsis-induced cardiomyopathy via ...
    Dec 11, 2024 · Fer-1 significantly decreased pro-inflammatory cytokine expression and affected some ferroptosis marker gene expression in myocardium at 6 h ...
  91. [91]
    Review and Chemoinformatic Analysis of Ferroptosis Modulators ...
    In 2008, RSL3 was revealed to induce cell death by involving labile iron [44]. Erastin, glutamate, sorafenib, and sulfasalazine are type I inducers that ...
  92. [92]
    Ferroptosis: a novel regulated cell death participating in cellular ...
    Jul 27, 2023 · Erastin leads to ferroptosis by autophagic degradation of ferritin and induction of TFRC expression. ... sulfasalazine, RSL3, ML162, FIN56 ...
  93. [93]
    APR-246 induces early cell death by ferroptosis in acute myeloid ...
    Jan 7, 2021 · In this study, we demonstrate that AML cell death occurring early after APR-246 exposure is suppressed by iron chelators, lipophilic antioxidants and ...
  94. [94]
    Discovery of novel 1,8-naphthalimide piperazinamide based ...
    This study suggests that compound 9o represents a potent and selective CA IX inhibitor and ferroptosis inducer for the treatment of TNBC.
  95. [95]
    A Systematic Review of Nanoparticle-Mediated Ferroptosis in ...
    May 6, 2025 · Ferroptosis induced by nanoparticle-based systems offers an innovative strategy for glioma therapy by efficiently traversing the BBB, precisely delivering ...
  96. [96]
    DHODH-mediated ferroptosis defense is a targetable vulnerability in ...
    DHODH inactivation induces extensive mitochondrial lipid peroxidation and ferroptosis in GPX4low cancer cells, whereas DHODH inactivation synergizes with ...
  97. [97]
    A novel mitochondria-targeting DHODH inhibitor induces robust ...
    DHODH inactivation results in widespread lipid peroxide accumulation and mitochondrial ferroptosis, effectively impeding tumor growth [6]. Additionally, as a ...
  98. [98]
    Targeted ferroptosis induction enhances chemotherapy efficacy in ...
    Sep 16, 2025 · Toxicity of the combination therapy was equivalent to that of COJEC alone, as determined by general health assessment and weight loss (Figure ...Missing: pharmacokinetics | Show results with:pharmacokinetics
  99. [99]
    Ferroptosis-based drug delivery system as a new therapeutic ...
    Feb 22, 2023 · A novel nano-drug delivery system based on ferroptosis has been used for the ferroptosis treatment of brain tumors with remarkable effects.
  100. [100]
    Full article: Ferroptosis in Cancer Therapy: Mechanisms, Small ...
    As a well-known inducer of ferroptosis, RSL3 effectively hinders the growth and proliferation of various cancer cells, such as those found in lung, breast, and ...
  101. [101]
    Beyond ferrostatin-1: a comprehensive review of ferroptosis inhibitors
    In this Review, we provide a comprehensive overview of the different classes of ferroptosis inhibitors, focusing on endogenous and synthetic RTAs.
  102. [102]
    Recent progress in ferroptosis: inducers and inhibitors - Nature
    Dec 29, 2022 · Erastin, as mentioned above, was first discovered in 2003 as a genotype-selective antitumor agent. Erastin induces ferroptosis in multiple ...
  103. [103]
    Ferroptosis inhibitors: past, present and future - Frontiers
    This review systematically describes the metabolic pathways of ferroptosis and summarizes the mechanisms of action of natural and synthetic small-molecule ...
  104. [104]
    Therapeutic Materials for Ferroptosis Inhibition: from Design to ...
    Sep 22, 2025 · Emerging evidence indicates that inhibition of ferroptosis has excellent therapeutic effects against these diseases. However, the therapeutic ...<|separator|>
  105. [105]
    Nrf2 inhibition reverses resistance to GPX4 inhibitor-induced ...
    Oct 16, 2018 · Transcriptional activation of Nrf2 was associated with resistance to ferroptosis. Overexpression of Nrf2 by inhibiting Keap1 or Nrf2 gene ...
  106. [106]
    Netrin-1 Alleviates Early Brain Injury by Regulating Ferroptosis via ...
    Furthermore, the mechanism underlying NTN-1 neuroprotection is correlated with the inhibition of ferroptosis, attenuating cell death via the PPARγ/Nrf2/GPX4 ...
  107. [107]
    Targeting GPX4 in human cancer: Implications of ferroptosis ...
    Apr 10, 2023 · GPX4 synthesis requires selenium incorporation into the selenoprotein through isopentenylation and selenocysteine tRNA maturation. GPX4 ...<|separator|>
  108. [108]
    Role of dietary iron revisited: in metabolism, ferroptosis ... - PubMed
    Mar 15, 2022 · Blocking transferrin-mediated iron import or recycling of iron-containing storage proteins (i.e., ferritin) also attenuates ferroptosis, ...
  109. [109]
    promotes ferroptosis and endocrine sensitivity of ER+ breast cancer
    Jan 20, 2025 · In conclusion, our data reveal that targeting estrogen-regulated SLC7A11 and SLC3A2 enhances ferroptosis in ER+ breast cancer, offering a ...Missing: females | Show results with:females
  110. [110]
    Review and Chemoinformatic Analysis of Ferroptosis Modulators ...
    Jan 4, 2023 · ... LOX inhibition. Some polyphenols are described as ferroptosis ... The latter enables their successful use (e.g., apigenin, resveratrol, curcumin ...<|separator|>
  111. [111]
    Vitamin E hydroquinone is an endogenous regulator of ferroptosis ...
    Aug 15, 2018 · Vitamin E (alpha-tocopherol) has been reported to prevent ferroptosis, but the mechanism by which this occurs is controversial.
  112. [112]
    Catechin inhibits ox-LDL-induced ferroptosis in vascular smooth ...
    Jun 2, 2025 · Catechin prevents ox-LDL-induced ferroptosis in VSMCs by activating the Nrf2/SLC7A11/GPX4 pathway, highlighting its potential therapeutic value for ...<|separator|>
  113. [113]
    Sodium butyrate alleviates DSS-induced inflammatory bowel ...
    Mar 3, 2025 · This study aimed to demonstrate that NaB suppresses ferroptosis, thereby alleviating inflammatory bowel disease (IBD) through modulation of the extracellular ...