Abstract
Ferroptosis is a regulated form of cell death characterized by iron-dependent lipid peroxidation. It plays a crucial role in various pathological conditions, including neurodegenerative diseases, cancer, ischemia–reperfusion injury, and organ failure. This review systematically explores the key mechanisms underlying ferroptosis, including polyunsaturated fatty acid-containing phospholipid (PUFA-PL) peroxidation, iron metabolism, and mitochondrial dysfunction. Additionally, we summarize major endogenous ferroptosis defense systems, including the SLC7A11-glutathione (GSH)-glutathione peroxidase 4 (GPX4) axis, the ferroptosis suppressor protein 1 (FSP1)-ubiquinol (CoQH₂) system, the mitochondrial dihydroorotate dehydrogenase (DHODH)-CoQH₂ pathway, and the guanosine triphosphate cyclohydrolase 1 (GCH1)-tetrahydrobiopterin (BH4) pathway, which act as critical brakes on ferroptosis. Furthermore, we discuss various small-molecule inhibitors targeting ferroptosis, categorized by their mechanisms of action, including iron chelators, lipid peroxidation inhibitors, antioxidants, and regulatory pathway modulators. Recent advances in pharmacological strategies and their potential therapeutic applications are also highlighted.
Keywords: Ferroptosis, Lipid peroxidation, Iron metabolism, Mitochondrial dysfunction, Small-molecule inhibitors
Introduction
Ferroptosis is a regulated form of cell death characterized by the iron-dependent accumulation of lipid peroxides within cellular membrane [1]. Mechanistically, cells undergo ferroptosis when antioxidant defense systems—most prominently glutathione peroxidase 4 (GPX4) —fail to detoxify lipid peroxyl radicals, leading to uncontrolled lipid peroxidation, membrane damage, and collapse of redox balance. Morphologically, ferroptosis is characterized by unique ultrastructural features, including shrunken mitochondria with increased membrane density, reduced or absent cristae, and rupture of the outer mitochondrial membrane, while nuclear morphology remains largely preserved. Although it shares similarities with necrotic cell death, such as plasma membrane rupture, ferroptosis is mechanistically defined as a non-apoptotic pathway distinct from classical necrosis and autophagy. Its execution can be specifically prevented by inhibitors of lipid peroxidation, including ferrostatin-1 and liproxstatin-1 [1].
Since its identification in 2012, ferroptosis has emerged as a key player in a wide spectrum of pathological conditions, including neurodegenerative diseases, ischemia–reperfusion injury, metabolic disorders, and malignancies [2, 3]. This metabolic vulnerability makes ferroptosis not only a major driver of tissue damage, but also a promising therapeutic target in diseases characterized by oxidative stress or therapy resistance [4, 5].
Importantly, ferroptosis is tightly regulated by multiple intrinsic defense systems that counteract lipid peroxidation and maintain cellular integrity. Beyond the canonical GPX4-glutathione (GSH) axis, recent studies have identified additional protective mechanisms such as ferroptosis suppressor protein 1 (FSP1), guanosine triphosphate cyclohydrolase 1 (GCH1), and mitochondrial dihydroorotate dehydrogenase (DHODH), which provide complementary safeguards against ferroptotic death. The complexity and redundancy of these pathways underscore the need to better define their relative contributions across different cell types and disease contexts [6, 7].
Given its involvement in various pathologies, pharmacological modulation of ferroptosis has become an attractive therapeutic strategy. Small molecule inhibitors have been extensively explored as potential agents to suppress ferroptosis in conditions where oxidative damage exacerbates disease progression, such as neurodegeneration, acute kidney injury (AKI), and cardiovascular disease [8, 9]. These inhibitors can be broadly categorized based on their mechanisms of action, including iron chelators that limit the availability of redox-active iron, lipid peroxidation inhibitors that suppress oxidative stress, and activators of endogenous antioxidant pathways such as nuclear factor erythroid 2‑related factor 2 (NRF2) and GPX4 stabilization [10].
This review provides a comprehensive analysis of ferroptosis, focusing on its molecular underpinnings, regulatory networks, and opportunities for therapeutic modulation. The following sections outline the principal processes driving ferroptosis, including iron metabolism, oxidative stress, and mitochondrial regulation. Endogenous defense pathways that counteract ferroptosis are subsequently examined, with a focus on their redundancy and context-specific activity. Recent advances in small-molecule inhibitors are then highlighted, addressing their mechanisms of action, pharmacological potential, and challenges in clinical application. Collectively, this review integrates current knowledge of ferroptosis drivers and suppressors to inform future therapeutic strategies in human diseases.
The engines of ferroptosis
Ferroptosis is a form of regulated cell death driven by the iron-catalyzed peroxidation of polyunsaturated fatty acid (PUFA)-containing phospholipids (PUFA-PLs). When lipid peroxidation surpasses the capacity of ferroptosis defense mechanisms, oxidative damage leads to membrane rupture and subsequent cell death. The principal drivers of ferroptosis include PUFA-PL synthesis and peroxidation, iron metabolism, and mitochondrial metabolism. Clarifying these processes is essential for understanding the regulatory network of ferroptosis and identifying potential therapeutic targets (Fig. 1).
Fig. 1.
The molecular mechanism of ferroptosis. Reactive oxygen species (ROS) accumulation triggers ferroptosis, a form of regulated cell death characterized by membrane damage through lipid peroxidation. The biosynthesis of polyunsaturated fatty acid-containing phospholipids (PUFA-PLs) requires two key enzymes: ACSL4 activates free PUFAs to form PUFA-CoAs, while LPCAT3 catalyzes the incorporation of PUFA-CoAs into membrane phospholipids. These PUFA-PLs are subsequently oxidized through lipoxygenase (LOX)-catalyzed enzymatic reactions, initiating lipid peroxidation cascades. Iron metabolism critically regulates ferroptosis via three coordinated processes: (1) TFR1-mediated Fe3⁺ uptake, (2) STEAP3-dependent reduction of Fe3⁺ to Fe2⁺, and (3) DMT1-facilitated Fe2⁺ transport. This iron flux elevates the labile iron pool (LIP), thereby promoting Fenton reactions that amplify ROS production. Mitochondrial contributions to ferroptosis involve glutaminolysis, wherein glutamine (Gln) is converted to glutamate (Glu) and subsequently to α-ketoglutarate (α-KG) to fuel the TCA cycle. This metabolic reprogramming sustains oxidative phosphorylation, resulting in electron leakage from the ETC and consequent ROS accumulation. There are at least three antioxidant systems that counteract ferroptosis: (1) SLC7A11-GSH-GPX4 axis: System Xc⁻ imports cystine for glutathione (GSH) biosynthesis, enabling GPX4 to reduce lipid hydroperoxides. (2) FSP1-CoQH₂ system: Independent of GPX4, FSP1 regenerates reduced ubiquinol (CoQH₂) to neutralize lipid radicals. (3) Mitochondrial DHODH pathway: Dihydroorotate dehydrogenase (DHODH) maintains CoQH₂ levels in the inner mitochondrial membrane, protecting against lipid peroxidation
PUFA-PL synthesis and peroxidation
PUFA-PLs serve as the critical substrates in ferroptosis, as their oxidative modification destabilizes membranes and precipitates cell death. Unlike saturated and monounsaturated fatty acids, PUFAs, such as arachidonic acid (AA) and adrenic acid (AdA), contain multiple double bonds, making them highly susceptible to peroxidation [11]. Prior to oxidation, PUFAs must undergo enzymatic activation and incorporation into membrane phospholipids. Acyl-CoA synthetase long-chain family member 4 (ACSL4) converts free PUFAs into acyl-CoA derivatives (PUFA-CoAs), which are subsequently esterified into lysophosphatidylcholine acyltransferase 3 (LPCAT3) [12, 13]. This process generates PUFA-PLs that are embedded in membranes and become the direct targets of oxidative attack. Lipid peroxidation of PUFA-PLs is initiated primarily by reactive oxygen species (ROS).
A major contributor is iron-dependent Fenton chemistry, in which ferrous iron (Fe2⁺) reacts with hydrogen peroxide to generate hydroxyl radicals capable of oxidizing PUFA-PLs into lipid peroxides (PUFA-OOH) [14]. The accumulation of PUFA-OOH disrupts membrane architecture, enhances permeability, and yields reactive aldehydes such as 4-hydroxynonenal (4-HNE) and 4-hydroxyhexenal (4-HHE), all of which intensify cellular injury. In addition to non-enzymatic reactions, enzymatic peroxidation catalyzed by lipoxygenases (LOXs) amplifies lipid oxidative generation [15]. These processes highlight the dual contribution of enzymatic and non-enzymatic pathways to ferroptosis in a context-dependent manner (Fig. 1).
Iron metabolism
Iron is indispensable for numerous physiological processes, yet its redox activity makes it a potent catalyst of ferroptosis. Ferric iron (Fe3⁺), transported in complex with transferrin, is internalized via transferrin receptor (TFR1) and reduced to Fe2⁺ by the metalloreductase STEAP3 within endosomes [16]. Fe2⁺ subsequently enters the cytosolic labile iron pool (LIP) through divalent metal transporter 1 (DMT1) and other non-transferrin-bound iron (NTBI) carriers [17]. When excessive, Fe2⁺ fuels Fenton chemistry and escalates ROS production, thereby amplifying lipid peroxidation and ferroptosis.
Intracellular iron balance is safeguarded by ferritin, which sequesters excess iron, and ferroportin (FPN), the sole known iron exporter. Ferritin degradation via NCOA4-mediated ferritinophagy releases stored iron into the LIP, whereas FPN mediates iron efflux but is negatively regulated by hepcidin. Disruption of these pathways, through enhanced uptake, excessive ferritin degradation, or reduced FPN activity, elevates intracellular Fe2⁺ levels and drives ferroptotic susceptibility.
Mitochondrial metabolism
ROS generation from the electron transport chain
Mitochondria are the primary source of intracellular reactive oxygen species (ROS), mainly derived from electron leakage within the electron transport chain (ETC). Under physiological conditions, electrons transferred through Complexes I and III occasionally escape and react with molecular oxygen, producing superoxide anions (O₂•⁻). These are subsequently converted by superoxide dismutase (SOD) into hydrogen peroxide (H₂O₂), which can further react with ferrous iron (Fe2⁺) via the Fenton reaction to generate highly reactive hydroxyl radicals (•OH). Excessive ROS accumulation damages mitochondrial membranes, proteins, and lipids, ultimately driving ferroptotic cell death. Thus, ETC leakage represents a central mechanism linking mitochondrial activity to oxidative stress and ferroptosis.
metabolic regulation: glutamine catabolism and AMPK-ACC axis
In addition to ROS generation, mitochondrial metabolism influences ferroptosis through nutrient decomposition and metabolic signaling pathways. Glutamine decomposition supplies α-ketoglutarate into the tricarboxylic acid (TCA) cycle, fueling mitochondrial respiration and supporting lipid biosynthesis. Enhanced glutamine catabolism not only sustains energy production but also increases the availability of substrates for PUFA-PL synthesis, thereby sensitizing cells to lipid peroxidation. The AMPK–ACC axis further regulates lipid metabolism in ferroptosis. Activation of AMP-activated protein kinase (AMPK) under energy stress conditions inhibits acetyl-CoA carboxylase (ACC), reducing fatty acid biosynthesis and limiting PUFA-PL incorporation into membranes. This metabolic checkpoint thus acts as a protective mechanism against ferroptosis by lowering the supply of oxidizable lipid substrates. Importantly, mitochondrial iron accumulation has emerged as another key determinant of ferroptotic sensitivity. Iron is indispensable for TCA cycle enzymes and ETC complexes; however, disruption of mitochondrial iron homeostasis enlarges the labile iron pool (LIP), intensifying ROS production through iron-catalyzed reactions. Insufficient sequestration by ferritin or impaired iron export via FPN promotes mitochondrial iron overload, exacerbating lipid peroxidation and reinforcing ferroptosis progression.
Non-GPX4 defense systems: FSP1–CoQ10 vs DHODH–CoQH₂
Recent advances have identified FSP1–CoQ10 and DHODH–CoQH₂ as critical non-GPX4 defense systems that broaden the landscape of ferroptosis regulation. Ferroptosis suppressor protein 1 (FSP1) (also known as AIFM2) localizes to cellular membranes in a myristoylation-dependent manner, where it reduces ubiquinone (CoQ10) to ubiquinol (CoQH₂) using NAD(P)H, thereby generating a potent radical-trapping antioxidant pool that suppresses lipid peroxidation outside mitochondria. In contrast, DHODH is anchored within the inner mitochondrial membrane and couples the oxidation of dihydroorotate to orotate with CoQ reduction, producing a localized reservoir of CoQH₂ that specifically protects mitochondrial membranes from ferroptotic damage. While these two pathways act in distinct cellular compartments, they function in a complementary rather than redundant manner, collectively safeguarding membrane integrity when GPX4 activity is compromised. Their therapeutic potential is context-dependent: augmentation of FSP1 activity or CoQ10 supplementation may benefit diseases characterized by widespread membrane lipid peroxidation, whereas enhancing DHODH function may be particularly relevant in mitochondria-rich, OXPHOS-active tissues vulnerable to ischemia–reperfusion injury or neurodegeneration. Conversely, pharmacological inhibition of FSP1 or DHODH can sensitize tumors to ferroptosis, especially in cancers with high FSP1 expression or reliance on mitochondrial metabolism, raising the possibility of synthetic lethality when combined with GPX4 inhibition. Nevertheless, unresolved issues remain, including the extent to which these systems compensate for GPX4 loss, the influence of CoQ biosynthetic capacity on their effectiveness, and the role of mitochondrial iron and ROS in amplifying lipid peroxidation. Furthermore, translational challenges such as tissue targeting (e.g., blood–brain barrier penetration), metabolic stability (e.g., optimization of CoQ analogs), and the need for predictive biomarkers underscore the importance of continued mechanistic studies to refine therapeutic strategies exploiting these emerging ferroptosis defense nodes.
The brakes of ferroptosis
Ferroptosis is regulated by multiple endogenous defense mechanisms that counteract lipid peroxidation and maintain redox homeostasis. These protective mechanisms mitigate oxidative stress, safeguard membrane integrity, and ultimately determine cellular susceptibility to ferroptotic death. The major ferroptosis defense systems include the SLC7A11-GSH-GPX4 axis, the FSP1-CoQH₂ system, the DHODH-CoQH₂ pathway, and the GCH1-BH4 pathway. These networks serve as crucial “brakes” on ferroptosis and illustrate the complexity of its regulation.
The SLC7A11-GSH-GPX4 axis
The SLC7A11-GSH-GPX4 axis constitutes the canonical defense against ferroptosis. SLC7A11 (also known as xCT) is a component of the cystine/glutamate antiporter system (system xₙ⁻) that imports cystine in exchange for glutamate [18]. Intracellular cystine is rapidly reduced to cysteine via an NADPH-dependent manner, providing the precursor for GSH biosynthesis [19].
GSH is a major cofactor for GPX4, a selenium-dependent enzyme that reduces lipid hydroperoxides (PUFA-PL-OOH) to their corresponding non-toxic lipid alcohols (PUFA-PL-OH), thereby preventing membrane destabilization [20]. During this process, GSH is oxidized to glutathione disulfide (GSSG), which is subsequently recycled back to GSH by glutathione reductase (GR) in an NADPH-dependent manner [21]. The SLC7A11-GSH-GPX4 System is considered the central gatekeeper of ferroptosis, and its pharmacological regulation has been a key focus in ferroptosis-related research.
The FSP1–CoQH2 system
CoQ10 is a lipophilic antioxidant that plays a key role in preventing membrane lipid peroxidation. FSP1 functions independently of GPX4 to maintain ferroptosis resistance [22–24]. FSP1 is localized predominantly to the plasma membrane but can also translocate to mitochondria. Through N-myristoylation, FSP1 anchors to membrane surfaces, where it catalyzes the NAD(P)H-dependent reduction of CoQ10 to CoQH₂. CoQH₂ acts as a potent radical-trapping antioxidant that directly scavenges lipid peroxyl radicals, thereby attenuating lipid peroxidation and ferroptosis [9]. Additionally, CoQH₂ supports the regeneration of α-tocopherol (vitamin E), further enhancing cellular antioxidant capacity. Therefore, the FSP-CoQH2 system represents an essential parallel defense mechanism that complements GPX4 activity.
The DHODH–CoQH2 system
The DHODH-CoQH₂ system, an inner mitochondrial membrane enzyme required for pyrimidine biosynthesis, has recently been identified as a mitochondria-specific ferroptosis defense factor [25]. DHODH catalyzes the oxidation of dihydroorotate (DHO) to orotate, utilizing CoQ10 as an electron acceptor. This reaction generates mitochondrial CoQH₂, which locally suppresses lipid peroxidation and protects against ferroptosis [25].
When GPX4 is inactivated, DHODH activity becomes increasingly critical for maintaining mitochondrial redox stability. Elevated CoQH₂ in this context enhances lipid peroxyl radical scavenging and restricts ferroptotic progression. However, simultaneous inhibition of GPX4 and DHODH abolishes this protective capacity, resulting in profound mitochondrial lipid peroxidation and extensive ferroptotic cell death.
The GCH1–BH4 system
The GCH1–BH4 pathway has been identified as an alternative ferroptosis defense mechanism independent of GPX4 and CoQH₂ [26, 27]. GCH1 catalyzes the rate-limiting step in the biosynthesis of BH4. BH4 acts as a radical-trapping antioxidant that directly neutralizes lipid peroxyl radicals and stabilizes membranes against oxidative damage. Cells with high GCH1 expression or enhanced BH4 biosynthesis show increased resistance to ferroptosis.
Beyond its direct antioxidant effects, BH4 contributes to redox regulation by supporting CoQ10 biosynthesis through the production of 4-hydroxybenzoate, a precursor in CoQ10 synthesis [26, 28]. Additionally, dihydrofolate reductase (DHFR) is required for the regeneration of BH4 from its oxidized form, dihydrobiopterin (BH2). Inhibition of DHFR depletes BH4 levels and enhances ferroptotic sensitivity, further underscoring its importance in ferroptosis resistance [29].
Small molecule inhibitors targeting ferroptosis
Pharmacological modulation of ferroptosis has attracted substantial interest as a therapeutic strategy across cancer, neurodegeneration, and ischemic injury. Small-molecule inhibitors at multiple nodes of the ferroptotic network, primarily by altering iron metabolism, suppressing lipid peroxidation, or reinforcing antioxidant defense systems (Table 1).
Table 1.
Summary of ferroptosis inhibitors
| Category | Inhibitor | Mechanism | Disease | References |
|---|---|---|---|---|
| Iron metabolism | Deferoxamine (DFO) | Iron chelator | SCI | [30] |
| Deferiprone (DFP) | Iron chelator | Multiple sclerosis | [31] | |
| Deferasirox (DFX) | Iron chelator | Ulcerative colitis | [32] | |
| CN128 | Iron chelator | β-thalassemia | [35] | |
| DFA1 | Iron chelator | - | [36] | |
| Compound 9c | Iron chelator; Scavenges free radicals | AKI | [31] | |
| Dexrazoxane (DXZ) | Iron chelator; upregulates GPX4, FTH1 | Cardiomyopathies | [37] | |
| 2,2′-bipyridine | Iron chelator | - | [9] | |
| Phenanthroline | Iron chelator | - | [9] | |
| M-30 | Iron chelator | AD | [38] | |
| Baicalein | Iron chelator; upregulates GPX4 and GSH | - | [39] | |
| Hinokitiol | Iron chelator; activates Nrf2; upregulates SLC7A11, GPX4, HO-1 | Parkinson’s disease | [40] | |
| Tannins (TA) | Iron chelator | Iron overload | [41] | |
| BMS536924 | Iron chelator | - | [42] | |
| Thymosin β4 | Iron chelator; upregulates BAX, HO-1 and HSP70 | - | [43] | |
| Ciclopirox (CPX) | Iron chelator | Neurological disorders | [45] | |
| AKI-02 | Iron chelator | AKI | [44] | |
| YL-939 | Binds 2PHB2; upregulates ferritin; reduces iron | Liver injury | [8] | |
| Compound 9a | Stabilizes Fe2⁺ levels | IS | [46] | |
| NSC306711 | Blocks Tf-TfR pathway; inhibits iron uptake | - | [47] | |
| Pyrrolidine dithiocarbamate (PDTC) | Inhibits DMT1 | - | [48] | |
| Benzylisothiourea | Inhibits DMT1 | - | [49] | |
| Carthamin yellow (CY) | Inhibits Fe2+ and ROS; reverses ACSL4, TFR1, GPX4, FTH1 | MIRI | [50] | |
| Farrerol (FA) | Reduces lipid peroxidation | Hypoxic-ischemic encephalopathy | [51] | |
| Lipid metabolism |
Vitamin E- α-tocopherol |
Scavenges free radicals | Epilepsy | [52] |
| Melatonin (MLT) | Scavenges free radicals; activates Nrf2/HO-1 | Subarachnoid hemorrhage | [53] | |
| Vitamin K1 (VK1) | Scavenges free radicals | AKI | [54] | |
| Menaquinone-4 (MK-4) | Scavenges free radicals; inhibits p53/SLC7A11 | Sepsis-associated acute lung injury | [55] | |
| Trolox | Ameliorates oxygenated PE | NASH | [56] | |
| Ferrostatin-1 (Fer-1) | Scavenges free radicals | IS | [57] | |
| SRS11-92 | Scavenges free radicals via activating Nrf2 | IS | [59] | |
|
UAMC-3203 UAMC-2418 |
Scavenges free radicals | - | [60] | |
| Liproxstatin-1 (Lip-1) | Inhibits lipid peroxidation | Ferroptosis-induced cardiovascular disease | [61] | |
| Lip-2 | Upregulates GPX4 | Lupus nephritis | [62] | |
| Compound 51 | Scavenges free radicals | IS | [57] | |
| Compound 7 J | Scavenges free radicals | DOX-induced cardiomyopathy | [63] | |
| Edaravone (EDA) | Activates Nrf2-FPN; upregulates GPX4 | IS, CIRI | [64] | |
| Olanzapine (OLZ) | Scavenges free radicals | - | [65] | |
| XJB-5–131 | Scavenges free radicals | Renal I/R injury, osteoarthritis | [66] | |
| JP4-039 | Scavenges free radicals | - | [66] | |
| (S)−6c | Scavenges free radicals | - | [67] | |
| CuATSM | Scavenges free radicals | IS | [68] | |
| Compound 25 | Scavenges free radicals | - | [69] | |
| Zileuton | Inhibits LOX-5 and lipid peroxidation | Acute retinal damage | [74] | |
| MK-886 | Inhibits ALOX-5 | MIRI | [71] | |
| BWA4C | Inhibits ALOX-5 | Inflammatory bowel diseases | [72] | |
| PD146176 | Inhibits ALOX-5 and lipid peroxidation | Defective sperm function | [73] | |
| ML351 | Inhibits ALOX-5 | MIRI | [75] | |
| Docebenone | Inhibits ALOX-5/12 and lipid peroxidation | IS | [58] | |
| Troglitazone (TRO) | Inhibits ACSL4 | - | [12] | |
| BRD4770 | Downregulates LPO and MDA; upregulates SLC7A11, 4-HNE | Aortic dissection | [79] | |
| 7- dehydrochole-sterol (7-DHC) | Inhibits lipid peroxide-tion | - | [80] | |
| Gossypol acetic acid (GAA) | Upregulates GPX4, downregulates ACSL4 and Nrf2 | MIRI | [82] | |
| Antioxidant | 2-amino-5-chloro-N, 3-dimethylbenza-mide (CDDO) | Inhibits GPX4 degradation, lipid peroxidation and ROS | - | [83] |
| Disulfiram (DSF), Fursultiamine | Disrupts GPX4 with HSC70; inhibits GPX4 degradation | - | [84] | |
| ADA-409–052 | Inhibits lipid peroxidation | Embolic stroke | [85] | |
| Mitoglitazone (MGZ) | Upregulates GPX4; Inhibits lipid peroxidation | Renal I/R injury | [86] | |
| Sodium selenite | Upregulates GPX4 | SCI | [87] | |
| PKUMDL-LC-101/LC101-D04 | Upregulates GPX4 | - | [58] | |
| Seratrodast | Activates system xₙ⁻/GSH/GPX4 | Seizures | [88] | |
| Paeoniflorin (PF) | Downregulates Fe2+, ROS; upregulates SOD | AD | [89] | |
| Carvacrol (CAR) | Upregulates GPX4 | IS | [90] | |
| Ginkgolide B (GB) | Regulates TFR1 and NOCA4; upregulates Nrf2/GPX4 | AD | [91] | |
| Pachymic acid (PA) | Upregulates GSH, SLC7A11 and GPX4; Downregulates Fe2+, MDA, ROS | MIRI | [92] | |
| Kaempferol (KF) | Inhibits lipid peroxidation accumulation via Nrf2/SLC7A11/GPX4 | IS | [93] | |
| Flavonoids (TFA) | Decreases ROS via SLC7A11/GPX4 | Parkinson’s disease | [94] | |
| Galangin (Gal) | Decreases lipid peroxidation via SLC7A11/GPX4 | Cerebral ischemia | [95] | |
| Alpha-lipoic acid (α-LA) | Increases xCT/GPX4 | AKI | [96] | |
| Capsiate | Activates TRPV1/GPX4 | Inestinal I/R injury | [97] | |
| San-Huang-Yi-Shen (SHYS) | Reduced iron overload via GSH/GPX4 | Diabetic nephropathy | [98] | |
| Modified Shoutai Pill | Upregulates GPX4 and SLC7A11; decreases MDA and ACSL4 | Recurrent pregnancy loss | [99] | |
| Angong Niuhuang Wan (AGNHW) | Activates PPARγ/AKT/GPX4 | IS | [100] | |
| Uridine | Activates Nrf2/SLC7 A11/GPX4/HO-1 | Sepsis-induced ALI | [101] | |
| Dithiolethiones (ACDT, D3T) | Activates Nrf2/xCT/GSH; reduced lipid peroxidation | Iron overload-induced cytotoxicity | [103] | |
| Propofol | Regulates SLC16A13/AMPK/GPX4 | MIRI | [105] | |
| Metformin (Met) | Upregulates Nrf2, GPX4; decreases MDA | Vascular calcification | [107] | |
| Ajudecunoid C | Scavenges free radicals via activating Nrf2/AREs | Neurological diseases | [109] | |
| Dehydroabietic acid | Reduced ROS and lipid peroxidation; upregulates HO-1, GSH, GPX4 and FSP1 via Nrf2/ARE | Nonalcoholic fatty liver disease | [110] | |
| Proanthocyanidins (PACs) | Upregulates GSH, GPX4, SLC7A11 via Nrf2-HO-1 | SCI | [110] | |
| Geraniin | Iron chelator; inhibits lipid peroxidation | - | [112] | |
| Quercetin (QCT) | Activates Nrf2/HO-1; uprgulates GPX4 | IS | [113] | |
| Eriodictyol | Activates Nrf2/HO-1 | AD | [38] | |
| Naringenin | Activates Nrf2/HO-1 | Lung injury | [114] | |
| Tectorigenin | Suppresses Smad3; restores GPX4 | Myocardial injury | [115] | |
| Biochanin A | Scavenges free radicals via Nrf2/System xc/GPX4 | Knee osteoarthritis | [116] | |
| Isoliquiritin apioside | Downregulates Hif-1α and HO-1 | Acute lung injury | [117] | |
| Puerarin | Upregulates Nrf2, SLC7A11, GPX4; inhibits COX-2 | Retinal degradation | [118] | |
| Resveratrol | Activates Nrf2/GPX4 | SCI | [119] | |
| Aloe-emodin | Activates Nrf2/SLC7A11/GPX4 | Cardiac toxicity | [120] | |
| Withaferin A | Activates Nrf2/HO-1 | Intracerebral hemorrhage | [121] | |
| Arbutin | Upregulates SLC7A11 | Non-alcoholic fatty liver disease | [122] | |
| Dl-3-n-butylphthalide | Upregulates Nrf2 and GPX4 | Parkinson’s disease | [123] | |
| Tetrahydroxy stilbene glycoside | Activates Keap1/Nrf2/ARE axis | AD | [124] | |
| β-caryophyllene | Decreased ROS and iron accumulation via Nrf2/HO-1 | IS | [127] | |
| 15,16-dihydrotanshinone I | Upregulates GSH/GSSG ratio, Nrf2/GPX4; reduced ROS | IS | [128] | |
| Dihydromyricetin | Upregulates GPX4; reduced Fe2⁺ and ACSL4 | Cerebral I/R injury | [126] | |
| Forsythoside A | Activates Nrf2/GPX4; upregulates GSH | AD | [125] | |
| Rosiglitazone | Inhibits ACSL4; restores GPX4 and GSH | Renal I/R injury | [76] | |
| Pioglitazone | Inhibits ACSL4 | - | [77] | |
| Compound 3f | Upregulates FSP1 | IS | [129] | |
| Ginsenoside Rg1 | Reverses xCT, GPX4 and FSP1; activates Nrf2 | Neurodegenerative diseases | [130] | |
| Cardamonin | Regulates p53/SLC7A11/GPX4 | Osteochondral injury | [131] |
Inhibitors targeting iron metabolism
Iron chelators
Iron chelators attenuate Fenton chemistry by limiting redox-active iron and thereby restraining lipid peroxidation. Deferoxamine (DFO), an FDA-approved iron chelator, effectively inhibits ferroptosis by binding free Fe3⁺, decreasing ROS production, and upregulating ferroptosis-protective proteins such as GPX4, ferritin heavy chain 1 (FTH1), and system xₙ⁻ [30]. Clinical translation is constrained by DFO’s short half-life and poor oral bioavailability..
Oral chelators, including deferiprone (DFP) and deferasirox (DFX), were developed to overcome these limitations and showed improved pharmacokinetics [31, 32]. Their use is tempered by adverse effects such as granulocyte deficiency (DFP) and renal toxicity (DFX) [33, 34]. Next-generation agents aim to enhance efficacy and safety: CN128, which incorporates a glucuronidation “sacrificial” site, demonstrates superior oral activity and is under clinical evaluation for β-thalassemia [35]. DFA1, a deferric amine compound, exhibits potent chelation and ferroptosis suppression in vivo and in vitro [36].
Structure-guided optimization has yielded hybrid chelators with intrinsic radical-trapping capacity. Cinnamamide-hydroxypyridone derivatives bearing phenolic acid motifs increase both Fe-binding and ROS scavenging; among them, compound 9c surpasses DFP in preventing ferroptosis [31]. Dexrazoxane (DXZ), the only FDA-approved agent for preventing doxorubicin (DOX)-induced cardiotoxicity, exerts its protective effects by chelating mitochondrial iron and mitigating oxidative stress [37]. Membrane-permeable Fe2⁺ chelators such as 2,2′-bipyridine and phenanthroline reduce mitochondrial iron-dependent ROS and inhibit nanoparticle-induced ferroptosis in vitro [9].
Natural or repurposed chelators also show promise. M-30 shows the ability to permeate the blood–brain barrier and reduce amyloid precursor protein and Aβ levels, highlighting its potential application in Alzheimer’s disease (AD) [38]. Baicalein reverses the elastin-induced decrease in iron accumulation, GPX4, and GSH levels [39]. Hinokitiol chelates iron and activates Nrf2, suggesting a dual mechanism of suppression [40].
Tannins (TA) selectively binds free iron without interfering with endogenous iron-containing molecules [41]. BMS536924, a dual inhibitor of insulin-like growth factor (IGF) and insulin receptors, has been found to exert iron-chelating and ferroptosis-inhibitory effects [42]. Moreover, Thymosin β4, an endogenous iron chelator, plays a role in preventing ferroptosis by regulating intracellular free iron and ROS levels [43]. More recently, AKI-02, a hydroxypyridinone-based chelator, has shown promise in reducing iron overload-induced AKI and ferroptosis-related renal damage [44]. The FDA-approved antifungal drug ciclopirox functions as an iron chelator to evaluate the effect of ferroptosis stress in mild traumatic brain injury models [45] (Table 2).
Table 2.
Iron Chelators – Comparison of Efficiency, Oral Bioavailability, and Toxicity
| Compound | Class | Reported Anti-ferroptosis Efficiency | Oral Bioavailability | Key Toxicities/Limitations | Advantages/Notes |
|---|---|---|---|---|---|
| Deferoxamine (DFO) | Classical chelator (hexadentate) | Moderate (benchmark) | Poor (IV/IM only) | Short half-life; injection-site reactions | Gold-standard Fe3⁺ chelator; robust anti-ferroptosis evidence |
| Deferiprone (DFP) | Bidentate chelator | Moderate–High | High (oral) | Agranulocytosis/neutropenia | Oral alternative to DFO; CNS penetration reported |
| Deferasirox (DFX) | Tridentate chelator | Moderate–High | High (oral) | Renal/hepatic toxicity; GI upset | Once-daily dosing; long half-life |
| CN128 | Next-gen hydroxypyridinone | High (vs DFP) | High (oral) | Favorable in early reports | Sacrificial glucuronidation site; superior oral efficacy; clinical trials for β-thalassemia |
| DFA1 | Deferric amine compound | High | Oral (reported) | Limited data | Enhanced chelation efficiency in vitro/in vivo |
| Compound 9c | Cinnamamide-hydroxypyridone derivative | Very high (≈10 × DFP) | Unknown/likely oral | Limited data | Dual chelation + radical scavenging |
| Dexrazoxane (DXZ) | EDTA analogue/pro-chelating agent | High (mitochondrial iron) | IV | Myelosuppression | Only FDA-approved for DOX cardiotoxicity prevention |
| Ciclopirox (CPX) | Hydroxypyridone antifungal | Moderate | High (oral/topical) | GI upset; off-label safety concerns | Repurposable; studied in brain injury models |
| AKI-02 | Hydroxypyridinone chelator | High (AKI models) | Oral (preclinical) | Limited data | Reduces iron overload–induced AKI |
| M-30 | Hydroxyquinoline chelator | Moderate | High (oral; BBB-permeable) | Limited data | Reduces APP/Aβ; potential neuro applications |
| Hinokitiol | α-Hydroxy ketone; ionophore-like | Moderate | High (oral) | Limited data | Chelation + Nrf2 activation; ↑SLC7A11/GPX4/HO-1 |
| Tannins (TA) | Polyphenolic chelators | Moderate | Oral (dietary) | GI intolerance (high dose) | Selectively bind free iron; spare holo-proteins |
| 2,2′-Bipyridine | Fe2⁺ chelator (research tool) | High (in vitro) | N/A | Not for clinical use | Reduces mitochondrial Fe-ROS; used as a lab reagent |
| Phenanthroline | Fe2⁺ chelator (research tool) | High (in vitro) | N/A | Not for clinical use | Often paired with bipyridine; blocks Fe-dependent ROS |
| Baicalein | Natural flavonoid (chelator + antioxidant) | Moderate | High (oral, natural) | Generally safe; GI upset at high doses | Chelates Fe; ↑GPX4/GSH; dual anti-ferroptosis activity |
| BMS536924 | Dual IGF/insulin receptor inhibitor with chelation activity | Moderate | Oral (reported) | Limited safety data | Iron-chelating + ferroptosis-inhibitory effects |
Non-iron chelators
Several small molecules attenuate ferroptosis by altering iron trafficking rather than directly chelating metal ions. YL-939 has been identified as a ferroptosis inhibitor through its interaction with 2PHB2, a protein involved in iron homeostasis. This interaction enhances ferritin expression and reduces intracellular iron levels, thereby providing protective effects against ferroptosis-induced liver injury [8]. Similarly, compound 9a, derived from benzimidazole scaffolds, stabilizes intracellular Fe2⁺ and improves neurological outcomes in the middle cerebral artery occlusion (MCAO) model of ischemic stroke (IS) [46].
NSC306711 blocks transferrin-transferrin receptor (Tf-TfR)-mediated uptake to limit iron influx [47]. DMT1 inhibitors, including pyrrolidine dithiocarbamate (PDTC) and benzylisothiourea, reduce NTBI levels, thereby slowing ferroptosis-related disease progression [48, 49].
Natural flavonoids have emerged as promising ferroptosis regulators. Carthamin yellow (CY) attenuates myocardial ischemia–reperfusion injury (MIRI) and reduces ROS accumulation in both in vivo and in vitro models [50]. Farrerol (FA) suppresses iron accumulation and lipid peroxidation, limiting ferroptosis-induced damage [51] (Fig. 2).
Fig. 2.
Ferroptosis inhibitors that target iron metabolic pathways. Ferric iron (Fe3⁺) is taken up by transferrin receptor 1 (TFR1) and internalized into endosomes, where it is reduced to Fe2⁺ by STEAP3 and transported into the cytosol via DMT1. Cytosolic Fe2⁺ forms the labile iron pool (LIP), which can be sequestered by chelators (e.g., DFP or DFX), stored in ferritin, or exported by ferroportin (FPN). Ferritinophagy, mediated by NCOA4, degrades ferritin to release Fe2⁺. Free Fe2⁺ catalyzes Fenton reactions, generating reactive oxygen species (ROS) that induce lipid peroxidation, leading to membrane damage and ferroptotic cell death. Ferroptosis inhibitors that activate the signaling are indicated by arrows. Ferroptosis inhibitors that inhibit the signaling are indicated by blunt arrows
Inhibition of ferroptosis via the lipid metabolism pathway
Endogenous free radical trapping antioxidants
Endogenous antioxidants neutralize lipid peroxyl radicals and reinforce cellular defenses. Vitamin E and its isoforms, particularly α-tocopherol (α-TOH), inhibit ferroptosis by reducing Fe3⁺ level and preventing lipid peroxidation [52]. Melatonin (MLT) exerts its ferroptosis-inhibitory effects by regulating ferritin levels, reducing lipid peroxidation, and activating the Nrf2/heme oxygenase-1 (HO-1) pathway, thereby providing neuroprotective effects in subarachnoid hemorrhage-related neuronal injury [53].
Vitamin K (VK) is a redox-active naphthoquinone that suppresses ferroptosis in GPX4-deficient mouse models [54]; menaquinone-4 (MK-4) acts as an electron donor to regenerate reduced antioxidants [55]. Additionally, recent findings suggest that vitamin K1 (VK1) may function as an effective endogenous antioxidant in mitigating AKI [54]. Trolox, a water-soluble vitamin E analog, reduces inflammatory cytokines and oxidative stress in early-stage non-alcoholic steatohepatitis (NASH) [56].
Exogenous free radical trapping antioxidants
Synthetic-free RTAs represent a major class of ferroptosis inhibitors. Ferrostatin-1 (Fer-1) stabilizes radicals and reduces ROS accumulation across various disease models [57]. Structure–activity relationship (SAR) studies have indicated that the N-cyclohexyl group serves as a critical lipophilic anchor for Fer-1’s activity [58]. SRS11-92, a Fer-1 analog, enhances metabolic stability but reduces plasma potency [59]. Further optimization yielded UAMC-2418 (improved stability but limited solubility) and UAMC-3203 (enhanced solubility, stability, and PK), which protects against organ damage without overt toxicity in mice [60].
Liproxstatin-1 (Lip-1), bearing a spiroquinoxaline amine scaffold, is efficacious in neurological disorders and cancers by inhibiting lipid peroxidation [61]. Lip-2 improves PK properties and prevents ferroptosis in lupus nephritis-derived human proximal tubular epithelial cells [62].
Phenothiazine derivatives also display RTA activity: compound 51 reduces elastin-induced ferroptosis and infarct volume in an MCAO model [57]; compound 7 J lowers DOX-induced cardiotoxicity with enhanced bioavailability and safety [63].
Clinically used agents with RTA properties include edaravone (EDA), which modulates GPX4/ACSL4/5-LOX and reduces neuroinflammation to promote recovery after spinal cord injury (SCI) [64], and Olanzapine (OLZ), which traps free radicals and suppresses RSL3-induced ferroptosis [65]; thiophene benzodiazepine derivatives, D:\WorkDIR\OPERATORS\QY31\2025\October\10-27\CAMLS_18_2025_5958_AMP-shared\Done exhibits 16-fold potency with minimal cytotoxicity [65].
Mitochondria-targeted RTAs-XJB-5–131 and JP4-039-suppress cytosolic ferroptosis in HT-1080 cells [66]; the refined compound (S)−6c, improves inhibitory activity approximately 30-fold vs JP4-039 [67]. Diacetylbis(N(4)-methylthiosemicarbazonato) copper(II) (CuATSM) reduces cerebral infarct size and oxidative stress in the transient MCAO model [68]. HW-3-based-4-hydroxy-pyrazoles yielded multiple inhibitors; compound 25 emerged as the most promising candidate [69].
Lipoxygenase (LOX) inhibitors
ALOX5 and ALOX15 catalyze PUFA peroxidation and promote ferroptosis [70]. ALOX5 inhibitors-zileuton, MK-886, BWA4C, and PD146176-exert anti-ferroptotic effects [71–73]. Zileuton reduces ROS and oxidative stress in retinal pigment epithelium (RPE) cells [74]. ML351, a specific ALOX15 inhibitor, limits erastin-induced cardiac ischemia–reperfusion injury [75]. Dual LOX inhibitors are also effective: docebenone (LOX-5/12 inhibitor) and baicalein (LOX-12/15 inhibitor) suppress lipid peroxidation [58].
ACSL4 inhibitors
ACSL4 promotes incorporation of PUFAs into phospholipids, sensitizing membranes to peroxidation and ferroptosis. Thiazolidinedione derivatives, including troglitazone (TRO), rosiglitazone, and pioglitazone (PIO), have been identified as effective ACSL4 inhibitors [12, 76, 77]. Notably, rosiglitazone enhances suppression of ferroptosis in GPX4-deficient mice [12]. Thrombin inhibitors have been reported to downregulate ACSL4 and ferroptosis [78].
Other inhibitors
Histone modulators can influence ferroptosis. The histone methyltransferase inhibitor BRD4770 lowers malondialdehyde (MDA) while increasing 4-HNE expression, reduces aortic lipid peroxidation, and improves cognitive function [79]. 7-dehydrocholesterol (7-DHC), prone to autooxidation, mitigates metabolic stress and acts as a ferroptosis inhibitor [80].
Abolane-type sesquiterpenoids (PBSs) from the deep-sea fungus Aspergillus floridus YPH1 include compound 7, which selectively blocks Erastin/RSL3-induced ferroptosis [81]. Gossypol acetic acid (GAA) decreases chelatable iron and lipid peroxidation in vitro and upregulates GPX4 expression in vivo, reinforcing its potential as a ferroptosis inhibitor [82].
Inhibition of ferroptosis via antioxidant action
The system Xc⁻–GSH–GPX4 axis
Reinforcement of the system Xc⁻–GSH–GPX4 axis remains a central anti-ferroptotic strategy. The triterpenoid compound 2-amino-5-chloro-N, 3-dimethylbenzamide (CDDO) inhibits heat shock protein 90 (HSP90), stabilizes GPX4, reduces lipid peroxidation, and limits ROS accumulation, thereby offering cellular protection [83]. Additionally, disulfide compounds such as disulfiram (DSF) and fursultiamine disrupt the GPX4-HSC70 interaction, inhibiting GPX4 degradation and enhancing ferroptosis resistance [84]. ADA-409–052 effectively suppresses tert-butyl hydroperoxide (TBHP)-induced lipid peroxidation and prevents ferroptosis triggered by GSH or GPX4 depletion. In a thrombotic mouse model, ADA-409–052 administration significantly reduced cerebral infarction, cerebral edema, and pro-inflammatory factor levels [85]. The antidiabetic drug mitoglitazone (MGZ) has also been found to enhance GPX4 expression while reducing lipid peroxidation, leading to significant renal protection in mice following I/R injury [86].
Se has been recognized as an essential regulator of ferroptosis through its ability to upregulate GPX4 and mitigate oxidative damage. In a rat model of SCI, sodium selenite administration significantly reduces iron, MDA, and 4-HNE levels while enhancing the FSP1/GPX4 pathway, ultimately improving motor function [87]. Recently, a metastable activation site on GPX4 has been identified, leading to the discovery of several GPX4 activators. Among them, PKUMDL-LC-101 and its analogue PKUMDL-LC101-D04 are the most effective in suppressing ferroptosis by increasing GPX4 enzymatic activity [58].
Additional small molecules and natural compounds have demonstrated ferroptosis-inhibitory potential through this pathway. Seratrodast, a thromboxane A2 receptor antagonist, significantly upregulates GPX4 levels and reduces lipid ROS, effectively preventing erastin-induced neuronal ferroptosis. In a mouse epilepsy model, seratrodast administration prolongs seizure latency and decreases seizure duration by enhancing GPX4 expression [88]. Similarly, paeoniflorin (PF), a monoterpene glycoside, improves cognitive function and reduces oxidative stress markers in brain tissue, thereby decreasing oxidative damage and alleviating neurological impairments in AD mice [89].
Carvacrol (CAR), a monoterpene phenol, effectively reduces lipid peroxidation markers and increases GPX4 expression, providing neuroprotection in IS models [90]. Ginkgolide B (GB), a terpene lactone derivative from Ginkgo biloba, modulates key ferroptosis markers, such as TFR1 and NOCA4, while enhancing Nrf2 and GPX4 expression, offering neuroprotective effects in AD mice [91]. Pachymic acid (PA), a lanolin-type triterpenoid, demonstrates ferroptosis-inhibitory properties by reducing MDA, ROS, and Fe2⁺ levels while upregulating GSH, SLC7A11, and GPX4, thereby alleviating MIRI [92].
Further studies have identified kaempferol (KF), a bioflavonoid, as a potent ferroptosis inhibitor. It enhances antioxidant defense mechanisms via the Nrf2/SLC7A11/GPX4 pathway, effectively preventing neuronal damage in oxygen–glucose deprivation/reperfusion (OGDR)-induced injury [93]. Additionally, total flavonoids from Aspergillus membranaceus (TFA) have shown neuroprotective effects in Parkinson’s disease (PD) models by increasing GSH levels and decreasing ROS accumulation [94]. Similarly, galangin (Gal) significantly reduces lipid peroxidation and enhances SLC7A11 and GPX4 expression in ischemia–reperfusion injury, ultimately protecting hippocampal neurons [95].
A-lipoic acid (LA), a mitochondrial cofactor, has been reported to enhance GSH and GPX4 expression while reducing ROS and lipid peroxidation. In folic acid-induced AKI models, LA effectively inhibited ferroptosis by reducing iron overload, mitigating ROS accumulation and lipid peroxidation, and increasing GSH and GPX4 levels [96]. Capsiate, a gut microbiota-derived metabolite, inhibits ferroptosis by activating the transient receptor potential vanilloid 1 (TRPV1) channel and upregulating GPX4 expression, thereby alleviating intestinal ischemia–reperfusion injury [97].
Traditional herbal medicines have also shown promise in ferroptosis inhibition. The San-Huang-Yi-Shen capsule (SHYS) significantly reduces Fe2⁺ levels while enhancing SLC7A11, GPX4, and GSH/GSSG levels, providing renal protection in diabetic nephropathy models [98]. Similarly, the Modified Shoutai Pill (Jianwei Shoutai Pill, JSP) upregulates GPX4 and GSH levels while decreasing MDA and ACSL4 protein expression, thereby mitigating lipid metabolism disorders in recurrent pregnancy loss models [99]. Another well-studied traditional formulation, Angong Niuhuang Wan (AGNHW), has demonstrated neuroprotective effects by modulating GPX4-dependent pathways and inhibiting ferroptosis [100] (Table 3).
Table 3.
Natural products and their ferroptosis-related targets
| Compound/Natural Product | Clarified Target/Mechanism (ferroptosis-related) | Disease/Model (example) |
|---|---|---|
| Myricetin | Inhibits OGDR injury via Nrf2/SLC7A11/GPX4 axis | Neuroprotection (OGDR) |
| Shikonin | Modulates Nrf2/HO-1 and SLC7A11/GPX4; context-dependent ROS regulation | Multiple (model-dependent) |
| Kaempferol (KF) | Activates Nrf2 → ↑SLC7A11/GPX4; ↓lipid peroxidation | OGDR/neuronal models |
| Quercetin (QCT) | Activates Nrf2/HO-1; ↑GPX4; ↓ACSL4 | Ischemic stroke |
| Resveratrol (RES) | Enhances Nrf2/GPX4 signaling; limits lipid ROS | Spinal cord injury (SCI) |
| Aloe-emodin (AE) | Activates Nrf2; ↑SLC7A11/GPX4; ↓oxidative stress | Cardiac toxicity |
| Arbutin (ARB) | Targets FTO/SLC7A11 axis; ↑SLC7A11 | NAFLD |
| Paeoniflorin (PF) | Reduces oxidative stress; ↑GPX4, ↓ROS | Alzheimer’s disease models |
| Carvacrol (CAR) | ↑GPX4; ↓lipid peroxidation | Ischemic models |
| Ginkgolide B (GB) | Regulates TFR1/NCOA4; activates Nrf2/GPX4 | Alzheimer’s disease models |
| Pachymic acid (PA) | ↑GSH/SLC7A11/GPX4; ↓Fe2⁺/ROS/MDA | Myocardial I/R injury (MIRI) |
| Capsiate | Activates TRPV1 → ↑GPX4 | Intestinal I/R injury |
| Ginsenoside Rg1 | Inhibits AIM2 inflammasome; activates Nrf2 | Neuroinflammation |
| Cardamonin (CAD) | Modulates p53/SLC7A11/GPX4 | Osteochondral injury |
| Galangin (Gal) | ↓Lipid peroxidation via Nrf2/SLC7A11/GPX4 axis | Cerebral ischemia |
| TFA (Total flavonoids from A. membranaceus) | ↑GSH, ↑GPX4; ↓ROS via Nrf2/SLC7A11 axis | Parkinson’s disease models |
| α-Lipoic acid (LA) | Enhances GSH/GPX4; ↓ROS and lipid peroxidation | Acute kidney injury (AKI) |
| SHYS (San-Huang-Yi-Shen capsule) | ↓Fe2⁺; ↑SLC7A11, GPX4, GSH/GSSG ratio | Diabetic nephropathy |
| JSP (Modified Shoutai Pill) | ↑GPX4, ↑GSH; ↓MDA, ↓ACSL4 | Recurrent pregnancy loss |
| AGNHW (Angong Niuhuang Wan) | Activates PPARγ/AKT/GPX4; inhibits ferroptosis | Ischemic stroke |
These findings highlight the intricate regulatory mechanisms of ferroptosis and underscore the therapeutic potential of targeting the System Xc⁻–GSH–GPX4 axis. Further research is needed to optimize these inhibitors and explore their clinical applications in ferroptosis-associated diseases.
Nrf2 pathway and ferroptosis inhibition
Uridine, a nucleoside composed of uracil and ribose, has demonstrated antioxidant, anti-inflammatory, and anti-aging properties [101]. Recent studies indicate that uridine suppresses macrophage ferroptosis by activating the Nrf2 signaling pathway, with the ferroptosis inhibitor Fer-1 further enhancing its protective effects [102]. Dithiolethiones, a class of lipophilic organosulfur compounds, including 5-amino-3-thioxo-3H-(1,2)dithiole-4-carboxylic acid ethyl ester (ACDT) and 3H-1,2-dithiole-3-thione (D3T), have been found to activate Nrf2 signaling pathway and increase cystine/glutamate antiporter system (System Xc⁻) and GSH levels in ferroptosis models [103, 104]. These compounds also elevate Nrf2-mediated ferritin and FPN expression, offering protection to glioblastoma cells against iron overload-induced cytotoxicity.
The anesthetic propofol has been shown to exhibit antioxidant properties by modulating the Nrf2/HO-1 pathway [105]. In a rat model of MIRI, propofol effectively mitigated ferroptosis by inhibiting SLC16A13 expression, activating the AMPK/GPX4 pathway, and protecting cardiac function [106]. Metformin (Met), a widely used first-line treatment for type 2 diabetes, attenuates oxidative stress by activating Nrf2/ARE pathway and promotes neural regeneration in SCI models [107]. Additionally, metformin upregulates GPX4 expression, activates Nrf2 signaling, reduces lipid peroxidation, and inhibits ferroptosis in vascular smooth muscle cells, thereby attenuating hyperlipidemia-induced vascular calcification [108]. Ajudecunoid C, a novel chlorane diterpenoid analogue, exhibits potent ferroptosis inhibition by activating the Nrf2-ARE pathway and scavenging free radicals [109]. Similarly, dehydroabietic acid, another diterpenoid, has been found to modulate the Keap1-Nrf2 axis and prevent non-alcoholic fatty liver disease [110].
Polyphenolic compounds have also been widely studied for their ability to regulate ferroptosis through the Nrf2 pathway. Proanthocyanidins (PACs) effectively scavenge free radicals and regulate lipoxygenase expression, increasing levels of GPX4, GSH, Nrf2, and HO-1, while downregulating ACSL4, thereby promoting functional recovery in SCI models [111]. The phenolic compound geraniin has been reported to inhibit ferroptosis in bmMSCs by suppressing lipid peroxidation, chelating Fe2⁺, and exhibiting strong antioxidant activity, without forming radical adducts [112]. Quercetin (QCT) activates the Nrf2-HO-1 signaling pathway, upregulates GPX4, downregulates ACSL4, and inhibits ferroptosis, thereby reducing neuronal damage and oxidative stress in cerebral IS [113].
Eriodictyol, a flavonoid, protects against ferroptosis-related neuronal damage in AD by activating Nrf2/HO-1 signaling pathway, reducing oxidative stress, and alleviating AD-like pathological changes [38]. Naringenin, a citrus-derived flavonoid, protects against silver nanoparticle-induced lung injury by reducing ferroptosis via the Nrf2/HO-1 signaling pathway [114]. Tectorigenin alleviates myocardial injury in sepsis by inhibiting ferroptosis through Smad3 suppression, restoring GPX4 expression, and mitigating oxidative stress and inflammation [115]. Biochanin A has also been identified as a potent ferroptosis inhibitor, suppressing iron accumulation and lipid peroxidation via Nrf2 signaling and mitigating osteoarthritis progression [116].
Isoliquiritin apioside, extracted from licorice, upregulates HIF-α and HO-1, thereby alleviating acute lung injury [117]. Puerarin mitigates ferroptosis by regulating iron-handling proteins, reducing lipid peroxidation via Nrf2 activation, and inhibiting COX-2, thereby protecting against iron overload-induced retinal degeneration [118]. The polyphenolic compound Resveratrol (RES) has been shown to promote ferroptosis-mediated recovery of motor function in SCI models by modulating Nrf2/GPX4 pathway [119]. Notably, both quercetin and resveratrol prevent iron-catalyzed hydroxyl radical formation, providing ferroptosis inhibition independent of Nrf2/ARE pathway. Aloe-emodin (AE), a natural anthraquinone derivative, activates Nrf2 and enhances SLC7A11 and GPX4 expression while reducing oxidative stress, thereby mitigating adriamycin-induced cardiotoxicity [120].
Several other compounds have been identified as Nrf2 pathway modulators with potential therapeutic applications. Withaferin A, a bioactive steroid ester, decreases oxidative stress and protects neuronal cells following intracerebral hemorrhage [121]. Arbutin (ARB), a natural phenolic glycoside, inhibits ferroptosis and alleviates non-alcoholic fatty liver disease by targeting the FTO/SLC7A11 axis, regulating gene methylation to enhance SLC7A11 expression [122]. Dl-3-n-butylphthalide (NBP), derived from celery extract, inhibits erastin-induced ROS accumulation and ferroptosis, offering protection against dopaminergic neuron loss [123]. Tetrahydroxy stilbene glycoside (TSG), an active component of Polygonum tigrinum, protects hippocampal neurons from ferroptosis-induced damage in AD by activating the Keap1/Nrf2/ARE axis [124]. Other small molecules, including β-caryophyllene (BCP), 15,16-dihydrotanshinone I (DHT), dihydromyricetin (DHM), and forsythoside A (FA), have demonstrated neuroprotective effects in neurological disorders by modulating Nrf2 signaling [125–128].
Other antioxidant pathways
Researchers have synthesized 14 analogs based on the structure of diphenylthiazole to enhance its inhibitory effects. Among them, compound 3f exhibits the highest biological activity, significantly alleviating neurological impairment in a rat model of MCAO following cerebral ischemia [129]. Similarly, ginsenoside Rg1, a bioactive ingredient derived from ginseng, has demonstrated protective effects against chronic neuroinflammation-induced neuronal ferroptosis by reducing oxidative stress, inhibiting the AIM2 inflammasome, and activating Nrf2 signaling pathway [130]. Cardamonin (CAD), a natural chalcone, has been reported to inhibit ferroptosis and prevent cartilage damage in rats by modulating the p53/SLC7A11/GPX4 signaling pathway [131] (Fig. 3).
Fig. 3.
Ferroptosis inhibitors that target antioxidant defense pathways. Ferroptosis is tightly regulated by multiple antioxidant systems that counteract phospholipid peroxidation. The primary cytosolic defense system is the SLC7A11–GSH–GPX4 axis: System Xc⁻ (composed of SLC7A11 and SLC3A2) imports cystine for glutathione (GSH) synthesis. GSH acts as a cofactor for GPX4, which detoxifies lipid hydroperoxides (PL-PUFA-OOH) into harmless lipid alcohols, thereby suppressing ferroptosis. Beyond this central pathway, complementary mechanisms provide backup protection: (1) The FSP1–CoQ₁₀ axis regenerates ubiquinol (CoQH₂) to neutralize lipid radicals at the plasma membrane; (2) In mitochondria, DHODH–CoQH₂ converts CoQ10 to CoQH₂, preventing mitochondrial lipid peroxidation; and (3) The GCH1–BH4 pathway generates tetrahydrobiopterin (BH4), a radical-trapping antioxidant that scavenges lipid peroxyl radicals and modulates CoQ synthesis. Additionally, oxidative stress triggers NRF2 release from KEAP1, activating antioxidant genes (e.g., SLC7A11, FSP1, HO-1) to prevent cellular oxidative damage. Ferroptosis inhibitors that activate the signaling are indicated by arrows. Ferroptosis inhibitors that inhibit the signaling are indicated by blunt arrows
Conclusions and perspectives
The interplay between lipid metabolism, iron homeostasis, and antioxidant defense mechanisms determines ferroptotic susceptibility, making it a highly regulated and therapeutically relevant process. Targeting ferroptosis offers opportunities to mitigate tissue damage in acute and chronic diseases while enhancing ferroptotic vulnerability in cancers resistant to conventional therapies [132, 133]. Importantly, ferroptosis plays a dual role in disease contexts: excessive activation contributes to ischemic stroke, neurodegeneration, and organ injury, whereas its induction provides tumor-suppressive benefits by eliminating metabolically stressed cancer cells.
Despite the remarkable progress, several challenges remain in ferroptosis research. The relative contributions of enzymatic versus non-enzymatic lipid peroxidation, as well as the precise role of mitochondria, continue to be debated. Beyond GPX4, emerging defense nodes such as FSP1–CoQ10 and DHODH–CoQH₂ operate in distinct cellular compartments and provide complementary protection, yet the extent of their compensatory activity when GPX4 is inhibited requires further clarification.
Clinical translation of ferroptosis modulators also faces significant bottlenecks, including bioavailability, limited tissue specificity, and potential toxicity or off-target [134, 135]. Tissue targeting is particularly challenging for neurological applications due to the blood–brain barrier, while optimization of CoQH₂ analogs exemplifies the need to improve metabolic stability. Innovative strategies such as nanocarrier-based delivery systems and prodrug design may help overcome these barriers. Moreover, integration of ferroptosis modulation with existing therapeutic regimens, coupled with a deeper understanding of its interplay with apoptosis, necroptosis, and autophagy, will likely broaden its clinical utility. Future directions should focus on optimizing pharmacokinetics and safety profiles, with strategies such as nanotechnology-based delivery systems and prodrugs offering promising solutions [142, 143]. Beyond mechanistic insights, the pharmacokinetic and pharmacodynamic properties of ferroptosis modulators present critical hurdles for clinical translation. Classical agents such as deferoxamine are hampered by poor oral bioavailability, limited tissue penetration, and off-target toxicities. Improving metabolic stability, tissue specificity, and dosing profiles will therefore be essential. Innovative approaches, including nanocarrier-based drug delivery platforms and prodrug strategies, are emerging as promising solutions to enhance the pharmacological performance of ferroptosis inhibitors and inducers, potentially bridging the gap between experimental efficacy and clinical application. Moreover, another critical challenge for clinical translation is the lack of standardized biomarkers to reliably monitor ferroptosis inhibition in patients. Current evaluations rely largely on indirect measures, including lipid peroxidation products such as malondialdehyde (MDA) and 4-hydroxynonenal (4-HNE), glutathione depletion, and alterations in iron metabolism. Emerging approaches, such as non-invasive iron imaging and the detection of oxidized phospholipid species, have shown promise but remain experimental. Establishing sensitive and disease-specific biomarkers will be essential to assess therapeutic efficacy, optimize patient selection, and guide the clinical application of ferroptosis modulators.
In conclusion, ferroptosis represents a powerful but double-edged sword in human disease. Addressing mechanistic uncertainties and translational barriers will be critical for harnessing its therapeutic potential and realizing effective interventions against some of the most challenging medical conditions (Table 4).
Table 4.
Abbreviation table
| Abbreviation | Full Term |
|---|---|
| PUFA | Polyunsaturated fatty acid |
| PUFA-PL | Polyunsaturated fatty acid-containing phospholipid |
| PUFA-CoA | Polyunsaturated fatty acid acyl-CoA derivative |
| PUFA-OOH | Lipid peroxides derived from PUFA-PLs |
| GSH | Glutathione |
| GPX4 | Glutathione peroxidase 4 |
| GSSG | Glutathione disulfide |
| GR | Glutathione reductase |
| FSP1 | Ferroptosis suppressor protein 1 |
| CoQH₂ | Ubiquinol (reduced form of Coenzyme Q10) |
| CoQ10 | Coenzyme Q10 (ubiquinone) |
| GCH1 | Guanosine triphosphate cyclohydrolase 1 |
| BH4 | Tetrahydrobiopterin |
| DHODH | Dihydroorotate dehydrogenase |
| NRF2 | Nuclear factor erythroid 2-related factor 2 |
| AKI | Acute kidney injury |
| AA | Arachidonic acid |
| AdA | Adrenic acid |
| ACSL4 | Acyl-CoA synthetase long-chain family member 4 |
| LPCAT3 | Lysophosphatidylcholine acyltransferase 3 |
| 4-HNE | 4-hydroxynonenal |
| 4-HHE | 4-hydroxyhexenal |
| LOX | Lipoxygenase |
| ROS | Reactive oxygen species |
| TFR1 | Transferrin receptor 1 |
| STEAP3 | Six-transmembrane epithelial antigen of prostate 3 (metalloreductase) |
| DMT1 | Divalent metal transporter 1 |
| NTBI | Non-transferrin-bound iron |
| LIP | Labile iron pool |
| FPN | Ferroportin |
| ETC | Electron transport chain |
| SOD | Superoxide dismutase |
| TCA | Tricarboxylic acid cycle |
| AMPK | AMP-activated protein kinase |
| ACC | Acetyl-CoA carboxylase |
| AIFM2 | Apoptosis-inducing factor mitochondria-associated 2 |
| IMM | Inner mitochondrial membrane |
| DHO | Dihydroorotate |
| BH2 | Dihydrobiopterin |
| DHFR | Dihydrofolate reductase |
| FDA | U.S. Food and Drug Administration |
| DFP | Deferiprone |
| DFX | Deferasirox |
| CN128 | A novel hydroxypyridinone-based oral iron chelator |
| DFA1 | Deferric amine compound |
| DXZ | Dexrazoxane |
| DOX | Doxorubicin |
| AD | Alzheimer’s disease |
| TA | Tannins |
| IGF | Insulin-like growth factor |
| Aβ | Amyloid-beta peptide |
| 2PHB2 | Prohibitin 2 |
| AGNHW | Angong Niuhuang Wan |
| ALOX15 | Arachidonate 15-lipoxygenase |
| ALOX5 | Arachidonate 5-lipoxygenase |
| ALS | Amyotrophic lateral sclerosis |
| BCP | Beta-caryophyllene |
| CAD | Cardamonin |
| CuATSM | Diacetylbis(N(4)-methylthiosemicarbazonato) copper(II) |
| DHM | Dihydromyricetin |
| DHT | 15,16-dihydrotanshinone I |
| FA | Farrerol |
| I/R | Ischemia/Reperfusion |
| IS | Ischemic stroke |
| JSP | Modified Shoutai Pill (Jianwei Shoutai Pill) |
| MCAO | Middle cerebral artery occlusion |
| MDA | Malondialdehyde |
| MGZ | Mitoglitazone |
| MIRI | Myocardial ischemia–reperfusion injury |
| MK-4 | Menaquinone-4 |
| NASH | Non-alcoholic steatohepatitis |
| NBP | Dl-3-n-butylphthalide |
| OLZ | Olanzapine |
| PDTC | Pyrrolidine dithiocarbamate |
| PIO | Pioglitazone |
| RPE | Retinal pigment epithelium |
| SAR | Structure–activity relationship |
| Se | Selenium |
| TBHP | Tert-butyl hydroperoxide |
| TRO | Troglitazone |
| TSG | Tetrahydroxy stilbene glycoside |
| Tf-TfR | Transferrin-Transferrin receptor |
| VK | Vitamin K |
| VK1 | Vitamin K1 |
| α-TOH | Alpha-tocopherol |
| SCI | Spinal cord injury |
| System Xc⁻ | Cystine/Glutamate Antiporter System |
| AGNHW | Angong Niuhuang Wan |
| ACDT | 5-amino-3-thioxo-3H-(1,2)dithiole-4-carboxylic acid ethyl ester |
| D3T | 3H-1,2-dithiole-3-thione |
| Met | Metformin |
| PACs | Proanthocyanidins |
| QCT | Quercetin |
| RES | Resveratrol |
| AE | Aloe-emodin |
| ARB | Arbutin |
| NBP | Dl-3-n-butylphthalide |
| TSG | Tetrahydroxy stilbene glycoside |
| BCP | β-caryophyllene |
| DHT | 15,16-dihydrotanshinone I |
| DHM | Dihydromyricetin |
| FA | Forsythoside A |
| CAD | Cardamonin |
| Lip-1 | Liproxstatin-1 |
| EDA | Edaravone |
| OLZ | Olanzapine |
| TRO | Troglitazone |
| PIO | Pioglitazone |
| 7-DHC | 7-dehydrocholesterol |
| GAA | Gossypol acetic acid |
| CDDO | 2-amino-5-chloro-N,3-dimethylbenzamide |
| DSF | Disulfiram |
| MGZ | Mitoglitazone |
| PF | Paeoniflorin |
| CAR | Carvacrol |
| GB | Ginkgolide B |
| PA | Pachymic acid |
| KF | Kaempferol |
| TFA | Total flavonoids from Aspergillus membranaceus |
| Gal | Galangin |
| LA | α-Lipoic acid |
| TRPV1 | Transient receptor potential vanilloid 1 |
| SHYS | San-Huang-Yi-Shen capsule |
| JSP | Modified Shoutai Pill (Jianwei Shoutai Pill) |
Acknowledgements
This work was supported by grants from the National Key Research and Development
Program of China (No. 2021YFA1302200); the Key Research and Development Program of Ningxia (No.2022BFH02012), National Natural Science Foundation of China (No.32170748 and No. 82204481); Shanghai Committee of Science and Technology (No. 21490714300); and Fundamental Research Funds for the Central Universities.
Author contributions
ZC had the idea for the review article; YH, KD, XF, XT, FW, YZ, JY, and YL performed the literature search; YH, KD and XF drafted the manuscript; ZC critically revised the manuscript.
Data availability
Not applicable.
Declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Conflict of interest
The authors declare no conflict of interest.
Footnotes
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Kun Duo, Xiaona Feng and Xiaofang Tian contributed equally to this work.
Contributor Information
Yuan He, Email: hy050344@163.com.
Zhenyu Cai, Email: drcaizhenyu@tongji.edu.cn.
References
- 1.Chen X, Li J, Kang R, Klionsky DJ, Tang D (2021) Ferroptosis: machinery and regulation. Autophagy 17:2054–2081 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Jiang X, Stockwell BR, Conrad M (2021) Ferroptosis: mechanisms, biology and role in disease. Nat Rev Mol Cell Biol 22:266–282 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Chen X, Kang R, Kroemer G, Tang D (2021) Broadening horizons: the role of ferroptosis in cancer. Nat Rev Clin Oncol 18:280–296 [DOI] [PubMed] [Google Scholar]
- 4.Liang D, Minikes AM, Jiang X (2022) Ferroptosis at the intersection of lipid metabolism and cellular signaling. Mol Cell 82:2215–2227 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Hassannia B, Vandenabeele P, Vanden Berghe T (2019) Targeting ferroptosis to iron out cancer. Cancer Cell 35:830–849 [DOI] [PubMed] [Google Scholar]
- 6.Kim JW, Min DW, Kim D et al (2023) GPX4 overexpressed non-small cell lung cancer cells are sensitive to RSL3-induced ferroptosis. Sci Rep 13:8872 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Ursini F, Maiorino M (2020) Lipid peroxidation and ferroptosis: the role of GSH and GPx4. Free Radic Biol Med 152:175–185 [DOI] [PubMed] [Google Scholar]
- 8.Yang W, Mu B, You J et al (2022) Non-classical ferroptosis inhibition by a small molecule targeting PHB2. Nat Commun 13:7473 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Chen X, Yu C, Kang R, Tang D (2020) Iron metabolism in ferroptosis. Front Cell Dev Biol 8:590226 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Ye T, Yang W, Gao T et al (2023) Trastuzumab-induced cardiomyopathy via ferroptosis-mediated mitochondrial dysfunction. Free Radic Biol Med 206:143–161 [DOI] [PubMed] [Google Scholar]
- 11.Xin S, Schick JA (2023) Pufas dictate the balance of power in ferroptosis. Cell Calcium 110:102703 [DOI] [PubMed] [Google Scholar]
- 12.Doll S, Proneth B, Tyurina YY et al (2017) ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat Chem Biol 13:91–98 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Markovic M, Ben-Shabat S, Keinan S, Aponick A, Zimmermann EM, Dahan A (2019) Lipidic prodrug approach for improved oral drug delivery and therapy. Med Res Rev 39:579–607 [DOI] [PubMed] [Google Scholar]
- 14.Delesderrier E, Monteiro JDC, Freitas S, Pinheiro IC, Batista MS, Citelli M (2023) Can iron and polyunsaturated fatty acid supplementation induce ferroptosis? Cell Physiol Biochem 57:24–41 [DOI] [PubMed] [Google Scholar]
- 15.Yang WS, Kim KJ, Gaschler MM, Patel M, Shchepinov MS, Stockwell BR (2016) Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis. Proc Natl Acad Sci U S A 113:E4966-4975 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Long H, Zhu W, Wei L, Zhao J (2023) Iron homeostasis imbalance and ferroptosis in brain diseases. MedComm 4:e298 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Chen L, Ma Y, Ma X et al (2023) TFEB regulates cellular labile iron and prevents ferroptosis in a TfR1-dependent manner. Free Radic Biol Med 208:445–457 [DOI] [PubMed] [Google Scholar]
- 18.Koppula P, Zhang Y, Zhuang L, Gan B (2018) Amino acid transporter SLC7A11/xCT at the crossroads of regulating redox homeostasis and nutrient dependency of cancer. Cancer Commun 38:12 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Koppula P, Zhuang L, Gan B (2021) Cystine transporter SLC7A11/xCT in cancer: ferroptosis, nutrient dependency, and cancer therapy. Protein Cell 12:599–620 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Seibt TM, Proneth B, Conrad M (2019) Role of GPX4 in ferroptosis and its pharmacological implication. Free Radic Biol Med 133:144–152 [DOI] [PubMed] [Google Scholar]
- 21.Imai H, Matsuoka M, Kumagai T, Sakamoto T, Koumura T (2017) Lipid peroxidation-dependent cell death regulated by GPx4 and ferroptosis. Curr Top Microbiol Immunol 403:143–170 [DOI] [PubMed] [Google Scholar]
- 22.Bersuker K, Hendricks JM, Li Z et al (2019) The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature 575:688–692 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Doll S, Freitas FP, Shah R et al (2019) FSP1 is a glutathione-independent ferroptosis suppressor. Nature 575:693–698 [DOI] [PubMed] [Google Scholar]
- 24.Susin SA, Lorenzo HK, Zamzami N et al (1999) Molecular characterization of mitochondrial apoptosis-inducing factor. Nature 397:441–446 [DOI] [PubMed] [Google Scholar]
- 25.Mao C, Liu X, Zhang Y et al (2021) DHODH-mediated ferroptosis defence is a targetable vulnerability in cancer. Nature 593:586–590 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Kraft VAN, Bezjian CT, Pfeiffer S et al (2020) GTP cyclohydrolase 1/tetrahydrobiopterin counteract ferroptosis through lipid remodeling. ACS Cent Sci 6:41–53 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Soula M, Weber RA, Zilka O et al (2020) Metabolic determinants of cancer cell sensitivity to canonical ferroptosis inducers. Nat Chem Biol 16:1351–1360 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Pierrel F, Burgardt A, Lee JH, Pelosi L, Wendisch VF (2022) Recent advances in the metabolic pathways and microbial production of coenzyme Q. World J Microbiol Biotechnol 38:58 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Brantley KD, Douglas TD, Singh RH (2018) One-year follow-up of B vitamin and iron status in patients with phenylketonuria provided tetrahydrobiopterin (BH4). Orphanet J Rare Dis 13:192 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Zhang Y, Fan BY, Pang YL et al (2020) Neuroprotective effect of deferoxamine on erastininduced ferroptosis in primary cortical neurons. Neural Regen Res 15:1539–1545 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Rayatpour A, Foolad F, Heibatollahi M, Khajeh K, Javan M (2022) Ferroptosis inhibition by deferiprone, attenuates myelin damage and promotes neuroprotection in demyelinated optic nerve. Sci Rep 12:19630 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Wu Y, Ran L, Yang Y et al (2023) Deferasirox alleviates DSS-induced ulcerative colitis in mice by inhibiting ferroptosis and improving intestinal microbiota. Life Sci 314:121312 [DOI] [PubMed] [Google Scholar]
- 33.Lecornec N, Castex MP, Reguerre Y et al (2022) Agranulocytosis in patients with Diamond-Blackfan anaemia (DBA) treated with deferiprone for post-transfusion iron overload: a retrospective study of the French DBA cohort. Br J Haematol 199:285–288 [DOI] [PubMed] [Google Scholar]
- 34.Kattamis A (2019) Renal function abnormalities and deferasirox. Lancet Child Adolesc Health 3:2–3 [DOI] [PubMed] [Google Scholar]
- 35.Chen W, Yuan X, Li Z et al (2020) CN128: a new orally active hydroxypyridinone iron chelator. J Med Chem 63:4215–4226 [DOI] [PubMed] [Google Scholar]
- 36.Feng W, Xiao Y, Zhao C et al (2022) New deferric amine compounds efficiently chelate excess iron to treat iron overload disorders and to prevent ferroptosis. Adv Sci 9:e2202679 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Huang C, Guo Y, Li T et al (2024) Pharmacological activation of GPX4 ameliorates doxorubicin-induced cardiomyopathy. Redox Biol 70:103024 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Zhang Y, Wang M, Chang W (2022) Iron dyshomeostasis and ferroptosis in Alzheimer’s disease: molecular mechanisms of cell death and novel therapeutic drugs and targets for AD. Front Pharmacol 13:983623 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Xie Y, Song X, Sun X et al (2016) Identification of baicalein as a ferroptosis inhibitor by natural product library screening. Biochem Biophys Res Commun 473:775–780 [DOI] [PubMed] [Google Scholar]
- 40.Xi J, Zhang Z, Wang Z et al (2022) Hinokitiol functions as a ferroptosis inhibitor to confer neuroprotection. Free Radic Biol Med 190:202–215 [DOI] [PubMed] [Google Scholar]
- 41.Phiwchai I, Yuensook W, Sawaengsiriphon N, Krungchanuchat S, Pilapong C (2018) Tannic acid (TA): a molecular tool for chelating and imaging labile iron. Eur J Pharm Sci 114:64–73 [DOI] [PubMed] [Google Scholar]
- 42.Kuganesan N, Dlamini S, McDaniel J, Tillekeratne VLM, Taylor WR (2021) Identification and initial characterization of a potent inhibitor of ferroptosis. J Cell Biochem 122:413–424 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Lachowicz JI, Pichiri G, Piludu M, et al (2022) Thymosin beta4 is an endogenous iron chelator and molecular switcher of ferroptosis. Int J Mol Sci 23(1):551 [DOI] [PMC free article] [PubMed]
- 44.Zhu H, Cen J, Hong C et al (2023) Targeting labile iron-mediated ferroptosis provides a potential therapeutic strategy for rhabdomyolysis-induced acute kidney injury. ACS Chem Biol 18:1294–1304 [DOI] [PubMed] [Google Scholar]
- 45.Zheng L, Pang Q, Huang R et al (2025) Stress-mediated activation of ferroptosis, pyroptosis, and apoptosis following mild traumatic brain injury exacerbates neurological dysfunctions. Mol Neurobiol 62:4055–4075 [DOI] [PubMed] [Google Scholar]
- 46.Fang Y, Chen X, Tan Q, Zhou H, Xu J, Gu Q (2021) Inhibiting ferroptosis through disrupting the NCOA4-FTH1 interaction: a new mechanism of action. ACS Cent Sci 7:980–989 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Horonchik L, Wessling-Resnick M (2008) The small-molecule iron transport inhibitor ferristatin/NSC306711 promotes degradation of the transferrin receptor. Chem Biol 15:647–653 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Wetli HA, Buckett PD, Wessling-Resnick M (2006) Small-molecule screening identifies the selanazal drug ebselen as a potent inhibitor of DMT1-mediated iron uptake. Chem Biol 13:965–972 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Zhang Z, Kodumuru V, Sviridov S et al (2012) Discovery of benzylisothioureas as potent divalent metal transporter 1 (DMT1) inhibitors. Bioorg Med Chem Lett 22:5108–5113 [DOI] [PubMed] [Google Scholar]
- 50.Guo H, Zhu L, Tang P et al (2021) Carthamin yellow improves cerebral ischemia-reperfusion injury by attenuating inflammation and ferroptosis in rats. Int J Mol Med. 10.3892/ijmm.2021.4885 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Wu Y, Qian J, Li K, Li W, Yin W, Jiang H (2022) Farrerol alleviates collagenase-induced tendinopathy by inhibiting ferroptosis in rats. J Cell Mol Med 26:3483–3494 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Zhang X, Wu S, Guo C et al (2022) Vitamin E exerts neuroprotective effects in pentylenetetrazole kindling epilepsy via suppression of ferroptosis. Neurochem Res 47(3):739–747 [DOI] [PubMed] [Google Scholar]
- 53.Ma SJ, Li C, Yan C et al (2023) Melatonin alleviates early brain injury by inhibiting the NRF2-mediated ferroptosis pathway after subarachnoid hemorrhage. Free Radic Biol Med 208:555–570 [DOI] [PubMed] [Google Scholar]
- 54.Kolbrink B, von Samson-Himmelstjerna FA, Messtorff ML et al (2022) Vitamin K1 inhibits ferroptosis and counteracts a detrimental effect of phenprocoumon in experimental acute kidney injury. Cell Mol Life Sci 79:387 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Gao N, Liu XY, Chen J, Hu TP, Wang Y, Zhang GQ (2024) Menaquinone-4 alleviates sepsis-associated acute lung injury via activating SIRT3-p53/SLC7A11 pathway. J Inflamm Res 17:7675–7685 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Tsurusaki S, Tsuchiya Y, Koumura T et al (2019) Hepatic ferroptosis plays an important role as the trigger for initiating inflammation in nonalcoholic steatohepatitis. Cell Death Dis 10:449 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Yang W, Liu X, Song C et al (2021) Structure-activity relationship studies of phenothiazine derivatives as a new class of ferroptosis inhibitors together with the therapeutic effect in an ischemic stroke model. Eur J Med Chem 209:112842 [DOI] [PubMed] [Google Scholar]
- 58.Scarpellini C, Klejborowska G, Lanthier C, Hassannia B, Vanden Berghe T, Augustyns K (2023) Beyond ferrostatin-1: a comprehensive review of ferroptosis inhibitors. Trends Pharmacol Sci 44:902–916 [DOI] [PubMed] [Google Scholar]
- 59.Chen Y, He W, Wei H et al (2023) Srs11-92, a ferrostatin-1 analog, improves oxidative stress and neuroinflammation via Nrf2 signal following cerebral ischemia/reperfusion injury. CNS Neurosci Ther 29:1667–1677 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Devisscher L, Van Coillie S, Hofmans S et al (2018) Discovery of novel, drug-like ferroptosis inhibitors with in vivo efficacy. J Med Chem 61:10126–10140 [DOI] [PubMed] [Google Scholar]
- 61.Liu W, Ostberg N, Yalcinkaya M et al (2022) Erythroid lineage Jak2V617F expression promotes atherosclerosis through erythrophagocytosis and macrophage ferroptosis. J Clin Invest. 10.1172/JCI155724 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Alli AA, Desai D, Elshika A et al (2023) Kidney tubular epithelial cell ferroptosis links glomerular injury to tubulointerstitial pathology in lupus nephritis. Clin Immunol 248:109213 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.You J, Yang W, Ma R et al (2022) Discovery of 2-vinyl-10H-phenothiazine derivatives as a class of ferroptosis inhibitors with minimal human ether-a-go-go related gene (hERG) activity for the treatment of DOX-induced cardiomyopathy. Bioorg Med Chem Lett 74:128911 [DOI] [PubMed] [Google Scholar]
- 64.Pang Y, Liu X, Wang X et al (2022) Edaravone modulates neuronal GPX4/ACSL4/5-LOX to promote recovery after spinal cord injury. Front Cell Dev Biol 10:849854 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Jiang X, Teng X, Shi H, Cao L, He L, Gu Q (2023) Discovery and optimization of olanzapine derivatives as new ferroptosis inhibitors. Bioorg Chem 133:106393 [DOI] [PubMed] [Google Scholar]
- 66.Krainz T, Gaschler MM, Lim C, Sacher JR, Stockwell BR, Wipf P (2016) A mitochondrial-targeted nitroxide is a potent inhibitor of ferroptosis. ACS Cent Sci 2:653–659 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Charaschanya M, Maskrey TS, LaPorte MG, Janjic JM, Wipf P (2022) Synthesis and optimization of nitroxide-based inhibitors of ferroptotic cell death in cancer cells and macrophages. ACS Med Chem Lett 13:403–408 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Shi X, Ohta Y, Nakano Y et al (2021) Neuroprotective effect of CuATSM in mice stroke model by ameliorating oxidative stress. Neurosci Res 166:55–61 [DOI] [PubMed] [Google Scholar]
- 69.Ying D, Shen X, Wang S et al (2024) Discovery of 4-hydroxyl pyrazole derivatives as potent ferroptosis inhibitors. Eur J Med Chem 263:115913 [DOI] [PubMed] [Google Scholar]
- 70.Shah R, Shchepinov MS, Pratt DA (2018) Resolving the role of lipoxygenases in the initiation and execution of ferroptosis. ACS Cent Sci 4:387–396 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Shi KN, Li PB, Su HX, Gao J, Li HH (2023) MK-886 protects against cardiac ischaemia/reperfusion injury by activating proteasome-Keap1-NRF2 signalling. Redox Biol 62:102706 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Franchi-Miller C, Saffar JL (1995) The 5-lipoxygenase inhibitor BWA4C impairs osteoclastic resorption in a synchronized model of bone remodeling. Bone 17:185–191 [DOI] [PubMed] [Google Scholar]
- 73.Walters JLH, De Iuliis GN, Dun MD et al (2018) Pharmacological inhibition of arachidonate 15-lipoxygenase protects human spermatozoa against oxidative stress. Biol Reprod 98:784–794 [DOI] [PubMed] [Google Scholar]
- 74.Lee JJ, Chang-Chien GP, Lin S et al (2022) 5-lipoxygenase inhibition protects retinal pigment epithelium from sodium iodate-induced ferroptosis and prevents retinal degeneration. Oxid Med Cell Longev 2022:1792894 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Cai W, Liu L, Shi X et al (2023) Alox15/15-HpETE aggravates myocardial ischemia-reperfusion injury by promoting cardiomyocyte ferroptosis. Circulation 147:1444–1460 [DOI] [PubMed] [Google Scholar]
- 76.Tao WH, Shan XS, Zhang JX et al (2022) Dexmedetomidine attenuates ferroptosis-mediated renal ischemia/reperfusion injury and inflammation by inhibiting ACSL4 via alpha2-AR. Front Pharmacol 13:782466 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Kung YA, Chiang HJ, Li ML et al (2022) Acyl-coenzyme a synthetase long-chain family member 4 is involved in viral replication organelle formation and facilitates virus replication via ferroptosis. MBio 13:e0271721 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Zhang L, Bai XY, Sun KY et al (2024) A new perspective in the treatment of ischemic stroke: ferroptosis. Neurochem Res 49:815–833 [DOI] [PubMed] [Google Scholar]
- 79.Chen Y, Yi X, Huo B et al (2022) Brd4770 functions as a novel ferroptosis inhibitor to protect against aortic dissection. Pharmacol Res 177:106122 [DOI] [PubMed] [Google Scholar]
- 80.Freitas FP, Alborzinia H, Dos Santos AF et al (2024) 7-dehydrocholesterol is an endogenous suppressor of ferroptosis. Nature 626:401–410 [DOI] [PubMed] [Google Scholar]
- 81.Li Y, Shi J, Liu R et al (2023) Structure revisions of phenolic bisabolane sesquiterpenes and a ferroptosis inhibitor from the marine-derived fungus Aspergillus versicolor YPH93. J Nat Prod 86:830–841 [DOI] [PubMed] [Google Scholar]
- 82.Lin JH, Yang KT, Ting PC et al (2021) Gossypol acetic acid attenuates cardiac ischemia/reperfusion injury in rats via an antiferroptotic mechanism. Biomolecules. 10.3390/biom11111667 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Wu Z, Geng Y, Lu X et al (2019) Chaperone-mediated autophagy is involved in the execution of ferroptosis. Proc Natl Acad Sci USA 116:2996–3005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Liu JP, Cen SY, Xue Z et al (2022) A class of disulfide compounds suppresses ferroptosis by stabilizing GPX4. ACS Chem Biol 17:3389–3406 [DOI] [PubMed] [Google Scholar]
- 85.Keuters MH, Keksa-Goldsteine V, Dhungana H et al (2021) An arylthiazyne derivative is a potent inhibitor of lipid peroxidation and ferroptosis providing neuroprotection in vitro and in vivo. Sci Rep 11:3518 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Qi Y, Hu M, Qiu Y et al (2023) Mitoglitazone ameliorates renal ischemia/reperfusion injury by inhibiting ferroptosis via targeting mitoNEET. Toxicol Appl Pharmacol 465:116440 [DOI] [PubMed] [Google Scholar]
- 87.Chen YX, Zuliyaer T, Liu B et al (2022) Sodium selenite promotes neurological function recovery after spinal cord injury by inhibiting ferroptosis. Neural Regen Res 17:2702–2709 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Hao Y, Ou Y, Zhang C et al (2022) Seratrodast, a thromboxane A2 receptor antagonist, inhibits neuronal ferroptosis by promoting GPX4 expression and suppressing JNK phosphorylation. Brain Res 1795:148073 [DOI] [PubMed] [Google Scholar]
- 89.Zhai L, Pei H, Shen H, Yang Y, Han C, Guan Q (2023) Paeoniflorin suppresses neuronal ferroptosis to improve the cognitive behaviors in Alzheimer’s disease mice. Phytother Res 37:4791–4800 [DOI] [PubMed] [Google Scholar]
- 90.Abdul Ghani MA, Ugusman A, Latip J, Zainalabidin S (2023) Role of terpenophenolics in modulating inflammation and apoptosis in cardiovascular diseases: a review. Int J Mol Sci. 10.3390/ijms24065339 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Hebert M, Bellavance G, Barriault L (2022) Total synthesis of Ginkgolide C and formal syntheses of Ginkgolides A and B. J Am Chem Soc 144:17792–17796 [DOI] [PubMed] [Google Scholar]
- 92.Liu D, Ding J, Li Z, Lu Y (2024) Pachymic acid (PA) inhibits ferroptosis of cardiomyocytes via activation of the AMPK in mice with ischemia/reperfusion-induced myocardial injury. Cell Biol Int 48:46–59 [DOI] [PubMed] [Google Scholar]
- 93.Yuan Y, Zhai Y, Chen J, Xu X, Wang H (2021) Kaempferol ameliorates oxygen-glucose deprivation/reoxygenation-induced neuronal ferroptosis by activating Nrf2/SLC7A11/GPX4 axis. Biomolecules. 10.3390/biom11070923 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Gao Z, Wang G, Chen Y et al (2024) Total flavonoids of Astragalus membranaceus protect against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced neurotoxicity in mice by inhibiting ferroptosis through SLC7A11/GPX-4 signaling pathway. Food Sci Hum Wellness 13:414–420 [Google Scholar]
- 95.Guan X, Li Z, Zhu S et al (2021) Galangin attenuated cerebral ischemia-reperfusion injury by inhibition of ferroptosis through activating the SLC7A11/GPX4 axis in gerbils. Life Sci 264:118660 [DOI] [PubMed] [Google Scholar]
- 96.Li X, Zou Y, Fu YY et al (2021) A-lipoic acid alleviates folic acid-induced renal damage through inhibition of ferroptosis. Front Physiol 12:680544 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Deng F, Zhao BC, Yang X et al (2021) The gut microbiota metabolite capsiate promotes Gpx4 expression by activating TRPV1 to inhibit intestinal ischemia reperfusion-induced ferroptosis. Gut Microbes 13:1–21 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Lv S, Li H, Zhang T et al (2023) San-huang-yi-shen capsule ameliorates diabetic nephropathy in mice through inhibiting ferroptosis. Biomed Pharmacother 165:115086 [DOI] [PubMed] [Google Scholar]
- 99.Lai Y, Zhang Y, Zhang H et al (2024) Modified shoutai pill inhibited ferroptosis to alleviate recurrent pregnancy loss. J Ethnopharmacol 319:117028 [DOI] [PubMed] [Google Scholar]
- 100.Bai X, Zheng E, Tong L et al (2024) Angong Niuhuang Wan inhibit ferroptosis on ischemic and hemorrhagic stroke by activating PPARgamma/AKT/GPX4 pathway. J Ethnopharmacol 321:117438 [DOI] [PubMed] [Google Scholar]
- 101.Jeengar MK, Thummuri D, Magnusson M, Naidu VGM, Uppugunduri S (2017) Uridine ameliorates dextran sulfate sodium (DSS)-induced colitis in mice. Sci Rep 7:3924 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Lai K, Song C, Gao M et al (2023) Uridine alleviates sepsis-induced acute lung injury by inhibiting ferroptosis of macrophage. Int J Mol Sci. 10.3390/ijms24065093 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Ansari MI, Khan MM, Saquib M, Khatoon S, Hussain MK (2018) Dithiolethiones: a privileged pharmacophore for anticancer therapy and chemoprevention. Future Med Chem 10:1241–1260 [DOI] [PubMed] [Google Scholar]
- 104.Kulkarni N, Gadde R, Betharia S (2023) Dithiolethiones D3T and ACDT protect against iron overload-induced cytotoxicity and serve as ferroptosis inhibitors in U-87 MG cells. Neurochem Res 48:2542–2551 [DOI] [PubMed] [Google Scholar]
- 105.Zhou Y, Yang Y, Yi L, Pan M, Tang W, Duan H (2024) Propofol mitigates sepsis-induced brain injury by inhibiting ferroptosis via activation of the Nrf2/HO-1axis. Neurochem Res 49:2131–2147 [DOI] [PubMed] [Google Scholar]
- 106.Sun F, He Y, Yang Z et al (2024) Propofol pretreatment inhibits ferroptosis and alleviates myocardial ischemia-reperfusion injury through the SLC16A13-AMPK-GPX4 pathway. Biomed Pharmacother 179:117345 [DOI] [PubMed] [Google Scholar]
- 107.Wang H, Zheng Z, Han W et al (2020) Metformin promotes axon regeneration after spinal cord injury through inhibiting oxidative stress and stabilizing microtubule. Oxid Med Cell Longev 2020:9741369 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Ma WQ, Sun XJ, Zhu Y, Liu NF (2021) Metformin attenuates hyperlipidaemia-associated vascular calcification through anti-ferroptotic effects. Free Radic Biol Med 165:229–242 [DOI] [PubMed] [Google Scholar]
- 109.Tan Q, Fang Y, Peng X, Zhou H, Xu J, Gu Q (2021) A new ferroptosis inhibitor, isolated from Ajuga nipponensis, protects neuronal cells via activating NRF2-antioxidant response elements (AREs) pathway. Bioorg Chem 115:105177 [DOI] [PubMed] [Google Scholar]
- 110.Gao G, Xie Z, Li EW et al (2021) Dehydroabietic acid improves nonalcoholic fatty liver disease through activating the Keap1/Nrf2-ARE signaling pathway to reduce ferroptosis. J Nat Med 75:540–552 [DOI] [PubMed] [Google Scholar]
- 111.Zhou H, Yin C, Zhang Z et al (2020) Proanthocyanidin promotes functional recovery of spinal cord injury via inhibiting ferroptosis. J Chem Neuroanat 107:101807 [DOI] [PubMed] [Google Scholar]
- 112.Chen B, Li X, Liu J et al (2021) Ferroptosis-inhibitory effect and possible mechanisms of ellagitannin geraniin. ChemistryOpen 10:737–739 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Peng C, Ai Q, Zhao F et al (2024) Quercetin attenuates cerebral ischemic injury by inhibiting ferroptosis via Nrf2/HO-1 signaling pathway. Eur J Pharmacol 963:176264 [DOI] [PubMed] [Google Scholar]
- 114.Zhang X, Li M, Wu H et al (2022) Naringenin attenuates inflammation, apoptosis, and ferroptosis in silver nanoparticle-induced lung injury through a mechanism associated with Nrf2/HO-1 axis: in vitro and in vivo studies. Life Sci 311:121127 [DOI] [PubMed] [Google Scholar]
- 115.Fang X, Fu W, Zou B, Zhang F (2023) Tectorigenin relieved sepsis-induced myocardial ferroptosis by inhibiting the expression of Smad3. Toxicol Res 12:520–526 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.He Q, Yang J, Pan Z et al (2023) Biochanin A protects against iron overload associated knee osteoarthritis via regulating iron levels and NRF2/System xc-/GPX4 axis. Biomed Pharmacother 157:113915 [DOI] [PubMed] [Google Scholar]
- 117.Zhongyin Z, Wei W, Juan X, Guohua F (2022) Isoliquiritin apioside relieves intestinal ischemia/reperfusion-induced acute lung injury by blocking Hif-1alpha-mediated ferroptosis. Int Immunopharmacol 108:108852 [DOI] [PubMed] [Google Scholar]
- 118.Song Q, Jian W, Zhang Y et al (2024) Puerarin attenuates iron overload-induced ferroptosis in retina through a Nrf2-mediated mechanism. Mol Nutr Food Res 68:e2300123 [DOI] [PubMed] [Google Scholar]
- 119.Ni C, Ye Q, Mi X et al (2023) Resveratrol inhibits ferroptosis via activating NRF2/GPX4 pathway in mice with spinal cord injury. Microsc Res Tech 86:1378–1390 [DOI] [PubMed] [Google Scholar]
- 120.He Y, Xi J, Fang J, Zhang B, Cai W (2023) Aloe-emodin alleviates doxorubicin-induced cardiotoxicity via inhibition of ferroptosis. Free Radic Biol Med 206:13–21 [DOI] [PubMed] [Google Scholar]
- 121.Zhou ZX, Cui Q, Zhang YM et al (2023) Withaferin a inhibits ferroptosis and protects against intracerebral hemorrhage. Neural Regen Res 18:1308–1315 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Jiang T, Xiao Y, Zhou J et al (2023) Arbutin alleviates fatty liver by inhibiting ferroptosis via FTO/SLC7A11 pathway. Redox Biol 68:102963 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Ye Z, Li C, Liu S et al (2023) Dl-3-n-butylphthalide activates Nrf2, inhibits ferritinophagy, and protects MES23.5 dopaminergic neurons from ferroptosis. Chem Biol Interact 382:110604 [DOI] [PubMed] [Google Scholar]
- 124.Gao Y, Li J, Wu Q et al (2021) Tetrahydroxy stilbene glycoside ameliorates Alzheimer’s disease in APP/PS1 mice via glutathione peroxidase related ferroptosis. Int Immunopharmacol 99:108002 [DOI] [PubMed] [Google Scholar]
- 125.Zhang L, Lang F, Feng J, Wang J (2024) Review of the therapeutic potential of Forsythiae Fructus on the central nervous system: active ingredients and mechanisms of action. J Ethnopharmacol 319:117275 [DOI] [PubMed] [Google Scholar]
- 126.Xie J, Zhang T, Li P, Wang D, Liu T, Xu S (2022) Dihydromyricetin attenuates cerebral ischemia reperfusion injury by inhibiting SPHK1/mTOR signaling and targeting ferroptosis. Drug Des Devel Ther 16:3071–3085 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Hu Q, Zuo T, Deng L et al (2022) beta-Caryophyllene suppresses ferroptosis induced by cerebral ischemia reperfusion via activation of the NRF2/HO-1 signaling pathway in MCAO/R rats. Phytomedicine 102:154112 [DOI] [PubMed] [Google Scholar]
- 128.Wu C, Duan F, Yang R, Dai Y, Chen X, Li S (2023) 15, 16-dihydrotanshinone I protects against ischemic stroke by inhibiting ferroptosis via the activation of nuclear factor erythroid 2-related factor 2. Phytomedicine 114:154790 [DOI] [PubMed] [Google Scholar]
- 129.Fang Y, Tan Q, Zhou H, Gu Q, Xu J (2022) Discovery of novel diphenylbutene derivative ferroptosis inhibitors as neuroprotective agents. Eur J Med Chem 231:114151 [DOI] [PubMed] [Google Scholar]
- 130.Kong L, Liu Y, Li J et al (2024) Ginsenoside Rg1 alleviates chronic inflammation-induced neuronal ferroptosis and cognitive impairments via regulation of AIM2 - Nrf2 signaling pathway. J Ethnopharmacol 330:118205 [DOI] [PubMed] [Google Scholar]
- 131.Gong Z, Wang Y, Li L, Li X, Qiu B, Hu Y (2023) Cardamonin alleviates chondrocytes inflammation and cartilage degradation of osteoarthritis by inhibiting ferroptosis via p53 pathway. Food Chem Toxicol 174:113644 [DOI] [PubMed] [Google Scholar]
- 132.Zhang C, Liu X, Jin S, Chen Y, Guo R (2022) Ferroptosis in cancer therapy: a novel approach to reversing drug resistance. Mol Cancer 21:47 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Li J, Cao F, Yin HL et al (2020) Ferroptosis: past, present and future. Cell Death Dis 11:88 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Stockwell BR (2022) Ferroptosis turns 10: emerging mechanisms, physiological functions, and therapeutic applications. Cell 185:2401–2421 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Wu X, Li Y, Zhang S, Zhou X (2021) Ferroptosis as a novel therapeutic target for cardiovascular disease. Theranostics 11:3052–3059 [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Not applicable.



