Skip to main content
CNS Neuroscience & Therapeutics logoLink to CNS Neuroscience & Therapeutics
. 2024 Jul 23;30(7):e14865. doi: 10.1111/cns.14865

Mechanism of ferroptosis regulating ischemic stroke and pharmacologically inhibiting ferroptosis in treatment of ischemic stroke

Zhaohui Chai 1, Jiesheng Zheng 1, Jian Shen 1,
PMCID: PMC11265528  PMID: 39042604

Abstract

Ferroptosis is a newly discovered form of programmed cell death that is non‐caspase‐dependent and is characterized by the production of lethal levels of iron‐dependent lipid reactive oxygen species (ROS). In recent years, ferroptosis has attracted great interest in the field of cerebral infarction because it differs morphologically, physiologically, and genetically from other forms of cell death such as necrosis, apoptosis, autophagy, and pyroptosis. In addition, ROS is considered to be an important prognostic factor for ischemic stroke, making it a promising target for stroke treatment. This paper summarizes the induction and defense mechanisms associated with ferroptosis, and explores potential treatment strategies for ischemic stroke in order to lay the groundwork for the development of new neuroprotective drugs.

Keywords: ferroptosis, iron, ischemic stroke, lipid peroxide, ROS


Ferroptosis plays a pivotal role in the pathogenesis of cerebral infarction, where the delicate balance between pro‐ and anti‐ferroptotic factors critically influences the outcomes of an ischemic stroke. Our review aims to comprehensively elucidate the underlying mechanisms of ferroptosis and explore therapeutic strategies targeting ferroptosis in ischemic stroke.

graphic file with name CNS-30-e14865-g002.jpg

1. INTRODUCTION

The term ferroptosis was coined in 2012 by Dixon et al. 1 It is characterized by the generation of lethal levels of iron‐dependent lipid ROS and is associated with oxidative stress and inflammation. Lipid peroxides are considered to be the primary inducers of ferroptosis, while other compounds that stimulate the production of cytoplasmic or mitochondrial ROS do not trigger ferroptosis. 2 Ferroptosis is different from other forms of programmed cell death such as necrosis, apoptosis, autophagy, and pyroptosis in morphology, physiology, and genetics. For instance, from a morphological perspective, ferroptotic cells lack characteristic features such as chromatin aggregation and margination, organelle swelling, and plasma membrane rupture. The primary morphological hallmarks include shrunken mitochondria, increased membrane density, and decreased mitochondrial cristae numbers. 1 , 3 Ferroptosis occurs when the balance between pro‐ferroptotic and anti‐ferroptotic factors is disrupted. 4 Ferroptosis can be induced by the accumulation of glutamate, iron, or polyunsaturated fatty acid (PUFA) − phospholipids, or the consumption of endogenous ferroptosis inhibitors such as GSH, NADPH, glutathione peroxidase 4 (GPX4), or vitamin E. 5

Ferroptosis, as a unique form of cell death, has aroused great attention in the medical community, and has been associated with various diseases, including neurological diseases (such as ischemic stroke, 6 , 7 cerebral hemorrhage, 8 subarachnoid hemorrhage, 9 amyotrophic lateral sclerosis, 10 Parkinson's disease, 11 etc.), cancer, 12 , 13 cardiovascular diseases, 14 and metabolic diseases. 15 Globally, stroke is the third leading cause of death and the primary cause of disability worldwide, with ischemic stroke accounting for more than 70% of strokes. 16 Currently, treatment goals for ischemic stroke focus on salvaging the penumbra, such as thrombolysis, mechanical thrombolysis, etc.; however, these approaches have limited efficacy due to their time window constraints. There is an urgent need for more research into stroke. This review paper summarizes recent advancements in understanding potential regulatory mechanisms of ferroptosis. Additionally, we discuss the potential pathophysiological role of ferroptosis in ischemic stroke and possible treatment strategies.

2. THE DISCOVERY OF FERROPTOSIS

Stockwell and his colleagues conducted a synthetic lethal screening of key RAS‐related targets through high‐throughput sequencing. Approximately 24,000 compounds were screened, leading to the identification of erastin, which was found to induce non‐apoptotic cell death in Ras‐expressing cells. 17 They further discovered that erastin induces non‐apoptotic cell death in tumor cells with activating mutations in the RAS–RAF–MEK pathway by binding to voltage‐dependent anion channels 2 and 3 (VDAC2/3), although VDAC alone is not sufficient for erastin function. 18 Subsequently, Stockwell expanded the screening to approximately 48,000 compounds and identified two small molecules, named RSL3 and RSL5, which increased mortality in the presence of cancer‐causing RAS. While RDL5 acts through VDAC, RSL3 functions independently of VDAC due to its association with elevated iron levels observed in many cancer patients. 19 The authors studied erastin, RSL3, and RSL5 together. Antioxidants and iron chelators were found to inhibit cell death caused by these three lethal compounds, independent of caspase, and all three compounds induced iron‐dependent, RAS–RAF–MEK‐dependent, oxidative, non‐apoptotic cell death. 20

The selenoprotein family of glutathione peroxidases (GPx's) converts lipid peroxides and hydrogen into their corresponding alcohols, 21 Seiler et al. 22 found that specific knockout of GPX4, a member of the selenoprotein family of glutathione peroxidases, triggers a unique cell death pathway associated with lipid peroxides. Eagle 23 proposed in 1955 that cysteine is necessary for cellular growth. The Xc‐system is a cystine/glutamate exchange transporter, a heterodimer composed of Solute carrier family 7 member 11 (SLC7A11, also known as xCT) and its chaperone CD98/4F2hc (SLC3A2), 24 xCT expression on the cell membrane is essential for intracellular GSH synthesis through cystine uptake, and Banjac et al. 25 demonstrated that the overexpression of xCT provides cellular protection against lipid peroxidation‐induced cell death. Erastin disrupts the uptake of cystine by cells through binding to the Xc‐ system, leading to the accumulation of lipid ROS. This accumulation of ROS, which is heavily reliant on iron, results in a form of cell death distinct from apoptosis, necrosis and autophagy, termed ferroptosis by Stockwell et al. 1

3. THE MOLECULAR MECHANISM OF FERROPTOSIS

Ferroptosis is related to three primary hallmarks: iron metabolism, lipid peroxidation and mitochondrial metabolism. The dysregulation of the balance between ferroptosis defense systems and inducers of ferroptosis leads to the initiation of ferroptotic cell death (Figure 1).

FIGURE 1.

FIGURE 1

Effects of metabolic processes on cellular susceptibility to ferroptosis. Inadequate regulation of PUFA‐enriched phospholipids and intracellular iron storage is essential for the initiation of ferroptosis, resulting in cell death. The activation of the TCA cycle in mitochondria through glutaminolysis may significantly contribute to the induction of ferroptosis. Ferroptosis is governed by a minimum of four distinct defense mechanisms that shield cells from undergoing ferroptotic cell death.

3.1. Iron homeostasis

Iron is an essential nutrient primarily absorbed by duodenal cells and essential for various biological processes, including enzymatic reactions, mitochondrial function, and DNA synthesis. The interconversion between Fe2+ and Fe3+ contributes to its effectiveness in biological systems. 26 However, unbound iron can lead to oxidative damage at subcellular, cellular, and organ levels if it does not bind to specific iron storage proteins or serum carriers such as transferrin or ferritin. 27 Studies by Stockwell et al. 1 have shown that erastin or RSL3 disrupt the antioxidant system by promoting intracellular iron accumulation, leading to increased oxidative damage by producing harmful levels of hydroxyl radicals through Fenton reaction. 28 The oxidation capacity of Fe2+ is five times greater than that of Fe3+, and the consentration of hydroxyl radicals is proportional to the concentration of Fe2+. 29 Fe2+ is oxidized to Fe3+ by ceruloplasmin or hephaestin in plasma and binds to transferrin (Tf) in the blood. 30 Ceruloplasmin (Cp), a glycoprotein with six tightly bound copper ions, belongs to the multicopper oxidase family. 31 The ferroxidase activity of Cp was initially reported in the early 1960s by Curzon. 32 Ceruloplasmin inhibits the production of reactive oxygen species mediated by Fe2+ through Fenton reaction, serving as a protective agent against oxidative stress. 33 Additionally, Cp facilitates the excretion of Fe2+ from cells via ferroportin exporter and plays a crucial role in iron homeostasis. 34 Transferrin, a glycoprotein capable of reversibly binding iron in the blood for transport to various tissues and organs, maintains normal physiological activities within these structures. TF can bind not only iron ions (Fe3+) but also other metal ions such as copper, manganese, and zinc with different affinities. The iron‐binding properties of TF are pH‐dependent; at neutral pH, iron tightly binds to extracellular TF, while it is released from TF under acidic conditions. 35 Almost all cellular iron uptake is dependent on the TF/transferrin receptor 1 (TFR1) complex, which undergoes endocytosis after binding to TFR1. 36 Within endosomes characterized by an acidic environment, Fe3+ dissociates from TF before being immediately reduced to Fe2+ by ferric reductases six transmembrane epithelial antigen of prostate 3 (STEAP3) or duodenal cytochrome b (DCYTB). 37 Subsequently transported across endosomal membranes into cytosol via bivalent metal transporter 1 (DMT1), 38 free Fe2+ forms a labile iron pool (LIP) upon binding with GSH within the cytoplasm. Iron chaperones poly (rC)‐binding protein 1/2 (PCBP1/2) are indispensable multifunctional proteins that regulate the storage, utilization, and efflux of iron. 39 , 40 Iron levels in the brain are primarily controlled by the blood–brain barrier (BBB). 41 Iron enters brain microvascular endothelial cells in two forms: holo‐Tf and non‐transferrin binding iron. The binding of the holo‐T complex to TfR and its endocytosis mediated by the reticular protein are the main pathways of iron transport across the luminal membrane. 42 Iron can form non‐transferrin‐bound complex with citrate and other compounds, which cross the blood–brain barrier faster than transferrin‐bound iron, 43 however, their physiological relevance remains unclear. 28 Similarly, Fe3+ can also bind to ferritin, there are two subtypes of ferritin, heavy chain FtH and light chain FtL. FtH possesses iron oxide enzyme activity that oxidizes ferrous (Fe2+) to Fe3+ to facilitate the rapid absorption of iron ions, while FtL is involved in assembling the ferritin core and mainly functions in long‐term storage. Iron is stored as Fe3+ within the ferritin molecule, effectively preventing it from participating in the oxidation react. 44 Ferritinophagy degrades the ferritin, increases the labile iron pool in cells and drives ferroptosis. 45

Upon entry into the cell, a significant portion of iron is transported into the mitochondria, traversing both the outer membrane (OMM) and inner membrane (IMM) to reach the mitochondrial matrix for heme and iron–sulfur (Fe–S) cluster synthesis. The translocation of iron across the outer mitochondrial membrane is believed to be mediated by DMT1 (also known as SLC11A2), 46 , 47 while its passage through the inner mitochondrial membrane is thought to be associated with mitochondrial iron transporter‐1 (MFRN1, also known as SLC25A37) and mitochondrial iron transporter‐2 (MFRN2, also known as SLC25A28). 48 , 49 However, there may exist alternative mechanisms for iron transport into mitochondria. 50 Additionally, iron is absorbed into the bloodstream from intestinal cells, macrophages, and hepatocytes via the iron transporter FPN (also known as SLC40A1). 51

Iron homeostasis is maintained by a complex network of regulation systems. The absorption, transportation and storage of iron at the translational level are mediated by iron regulatory protein (IRP) 1 or cytoplasmic aconitase (encoded by ACO1) and the iron‐responsive element binding protein 2 or IRP2 (Regulated by the IREB2) pathway. 52 IRPs are RNA‐binding proteins that bind to cis‐regulated iron response elements (IREs), which are stem‐loop RNA structures located at the 3′ or 5′ UTR of target transcripts, to regulate ferritin, FPN translation, and TfR mRNA stability. IRP1/2 binds to the conserved IREs of FTH, FTL, and FPN mRNAs, and acts as both a translation enhancer and \inhibitor to regulate their translation. 53 Additionally, IRP interacts with multiple IREs on TFRC mRNAs to protect the mRNAs from degradation. 54

As previously mentioned, ferritin‐bound iron can be released through the process of ferritinophagy. In conditions of iron deficiency, the multifunctional autophagy receptor nuclear coactivator receptor 4 (NCOA4) recognizes ferritin and binds to the autophagic protein MAP1LC3/LC3 (microtubule associated protein 1 light chain 3). The phagosomes merge with lysosomes to degrade ferritin, release labile iron, and maintain the intracellular labile iron pool. 55 Therefore, ferritinophagy is largely influenced by the amount of cellular NCOA4, which fluctuate with changes in iron level. When iron is sufficient, HERC2 ubiquitin ligase recruits the ferritinophagy receptor NCOA4 for ubiquitination and subsequent proteasomal degradation, thereby regulating ferritin autophagy at a basal level. 56 There are also alternative mechanisms for modulating NCOA4 levels. For example, analysis of the NCOA4 promoter reveals typical hypoxia response elements through which hypoxia inducible factor 2a (HIF2a) can regulate NCOA4 levels during ischemic hypoxia and iron deficiency.

Oxygen free radicals generated by Fenton reaction pose a significant threat to cellular and organismal health. NRF2 serves as an important transcription factor for maintaining metabolic, protein and REDOX homeostasis. 57 Normally, NRF2 is bound to Kelch‐like epichlorohydrin‐associated protein 1 (Keap1), which interacts with actin and undergoes degradation via ubiquitination. However, under condition of oxidative stress or other toxic insults, oxidation or alkylation of cysteine thiol in KEAP1 58 prevents the degradation of NRF2 by ubiquitination, leading to its translocation into the nucleus where it binds to antioxidant responsive elements (AREs). These AREs are present in the promoters of numerous genes encoding antioxidant and detoxification enzymes that regulate xCT. Nrf2 influences the translation of ARE‐related genes about glutathione synthesis and the expression of HO‐1, and Gpx4 and NADPH quinone oxidoreductase 1 (NQO1) related to REDOX regulation. 59 , 60 Furthermore, NRF2 plays a role not only in antioxidation but also in iron storage, exportation, heme synthesis, and hemoglobin catabolism. As a transcription factor, Nrf2 governs the transcription of transferrin receptor (TFR1), FTH1 and Ftl related to iron storage while reducing labile iron levels, 61 simultaneously regulating the expression of iron transporter protein (FPN1) related to iron exportation. 62 ATP‐binding cassette subfamily B member 6 (ABCB6) is involved in heme anabolism by facilitating the transport of porphyrins to mitochondria. In hepatogenic cell lines, NRF2 binds to the promoter regions of ABCB6 and regulates its transcription. Ferrochelatase (FECH) catalyzes the insertion of ferrous iron into protoporphyrin IX to generate heme cofactor; similarly, NRF2 modulates the transcriptional activity of FECH. 63 Senescent erythrocytes undergo phagocytosis and degradation by macrophages, leading to the catalysis of heme into free iron (Fe2+), carbon monoxide (CO), and biliverdin by heme oxygenase (HO‐1). The released ferrous iron is recycled while biliverdin undergoes further metabolism. NRF2 functions as a transcription factor to regulate the expression of HO‐1. 64

As mentioned above, HIF can modulate the level of NCOA4, thereby influencing ferritinophagy. Under normal conditions, the α chains of HIF1 and HIF2 undergo hydroxylation by proline hydroxylase domain enzyme (PHD) and subsequent degradation through the ubiquitin‐proteasome pathway. 65 , 66 However, under hypoxic and iron‐deficient conditions where PHD function is limited, stable α‐β dimers of HIF are formed to regulate iron metabolism, erythropoiesis, angiogenesis, glycolysis. 66 , 67 , 68 HIF1 and HIF2 act as transcriptional regulators for TF (encoding transferrin), DMT1 (also known as SLC11A2), FPN (also known as SLC40A1), TFRC, HO‐1, and thus affect iron metabolism. 69 , 70 , 71 , 72 , 73

The hepcidin–ferroportin axis: Hepatic, a hepatic hormone, is a small peptide produced in the liver and secreted into the bloodstream. It plays a crucial role in controlling the iron homeostasis. 74 FPN (ferroportin, also known as SLC40A1) is the sole known mammalian iron exporter and hepcidin induces its degradation of ferroportin by ubiquitination. 75 FPN is primarily expressed on the reticuloendothelial system where macrophages recycle iron from senescent erythrocytes' hemoglobin degradation. 74 Hepcidin restricts iron entry from intestinal enterocytes, macrophages and other cells into the plasma while facilitating its removal from the plasma membrane by binding to the ferroportin, the sole iron‐export protein. 73 , 76 , 77 The hepcidin–FPN regulatory axis controls systemic iron homeostasis and ensures sufficient cellular iron supply. Hepcidin expression decreases in several diseases associated with iron overload such as inefficient erythropoiesis (e.g., thalassemia) or hereditary hemochromatosis (HH), whereas excessive hepcidin can lead to anemia of inflammation or iron‐refractory iron deficiency anemia (IRIDA). 78 RNF217 is an E3 ubiquitin ligase that induces polyubiquitination and degradation of FPN, meanwhile Tet1 (a member of the 10‐11 translocation methylcytosine dioxygenase family of enzymes) mediates the demethylation of the Rnf217 promoter leading to the up‐regulation of RNF217 expression. 79

3.2. Lipid peroxidation

Phospholipid peroxides (PLOOH) represent a distinct category of ROS, its' accumulation can result in the disruption of plasma membrane integrity, leading to ferroptosis. 80 Iron‐dependent lipid peroxidation involves free radical reactions, wherein hydrogen atoms are extracted from phospholipids containing polyunsaturated fatty acids (PUFA‐PL) to produce phospholipid peroxide (PLOOH). This process is conducted by enzymatic or non‐enzymatic lipid peroxidation reactions. 81 Enzymes involved in the incorporation of PUFAs into phospholipids (PLs) play an important role in ferroptosis. ACSL4 (a member of the Acyl‐CoA synthetase long‐chain family) serves as a critical drivers of ferroptosis. PUFAs, particularly arachidonic acid (C20:4) and adrenic acid (C22:4), are conjugated with CoA by ACSL4 to produce acyl‐CoA, which is then re‐esterified in phospholipids to generate PL through lysophosphatidylcholine acyltransferase (LPCATs). 82 , 83 The mammalian arachidonate lipoxygenases (ALOXs), comprising 6 members containing non‐heme iron (ALOXE3, ALOX5, ALOX12, ALOX12B, ALOX15, and ALOX15B), catalyze the oxidation of PUFAs. 84 For example, ALOX5 can convert arachidonic acid (AA) into 5‐hydroperoxyeicosatetraenoicacid (5‐HPETE), a precursor of leukotrienes. 80 In addition, Cytochrome P450 reductase (POR)‐mediated lipid peroxidation serves as a surrogate mediator of ferroptosis. POR is a NADPH‐dependent redox enzyme containing flavin mononucleotide, previously implicated in the maintenance of cellular redox homeostasis and the regulation of lipid desaturase 85 and elongase 86 activities. POR facilitates electron transfer from NADPH to microsomal cytochrome P450. 87 , 88 Additionally, POR, NADPH oxidase (NOX), NADH‐cytochrome b5 reductase (CYB5R1), mitochondrial electron transport chain pathways and iron‐dependent Fenton reaction collectively generate H2O2, which subsequently yields free hydroxyl radicals in the presence of ferrous iron and disrupts membrane integrity by peroxidation of PUFA‐PL. 89 LPO or its derivatives such as 4‐HNE, MDA perforate the plasma membrane or organelle membrane, ultimately instigating cell death. 90 , 91 , 92

3.3. Mitochondrial metabolism

Several metabolic processes in mitochondria play crucial roles in triggering ferroptosis. 93 Mitochondria contribute to ferroptosis through involvement in biosynthesis, bioenergetics and ROS regulation. First, mitochondrial ROS production may contribute to ferroptosis by promoting lipid peroxidation as they are the primary source of ROS in cells.. Specifically, electron leakage from ETC complexes I and III produces superoxide, which is subsequently converted to H2O2 by superoxide dismutase (SOD)‐mediated dismutase. H2O2 reacts with labile ferrous ion (Fe2+) via the Fenton reaction to form hydroxyl radicals (•OH), leading to extraction of diallyl hydrogen in PUFA and formation of PUFA radical (PUFA•). These unstable carbon‐centered radicals rapidly react with oxygen to form PUFA hydroperoxides (PUFA‐OOH). 93 In addition, electron transport and proton pumping in mitochondria play a significant role in inducing ferroptosis. 94 Futhermore, mitochondria are pivotal organelles for ATP production. In short, electrons transported through the ETC complex generate proton power that is then utilized by ATP synthase for energy generation. 95 In the context of energy depletion, activation of the energy sensor AMP‐activated protein kinase (AMPK) leads to phosphorylation and inactivation of acetyl‐CoA carboxylase (ACC), ultimately inhibits the synthesis of some PUFAs and ferroptosis. Conversely, under conditions of abundant energy, AMPK is deactivated, thereby facilitating ferroptosis. 96 Furthermore, mitochondrial involvement in biosynthesis can also lead to ferroptosis. The tricarboxylic acid (TCA) cycle is the primary source of cellular energy and is involved in a variety of metabolic pathways. Maintenance of TCA cycle function relies on mitochondrial metabolism due to their close interrelation. 97 Supplementation with glutamine or various anaplerotic reactions that fuel the tricarboxylic acid cycle, such as glutaminolysis, can enhance ferroptosis. 98 Ketoglutaric acid (αKG), a downstream metabolite of glutaminolysis within the TCA cycle, has been shown to induce lipid ROS accumulation and promote ferroptosis by substituting for glutamine. Additionally, the TCA metabolites downstream of the αKG, including fumarate, succinate and malate, are capable of substituting for glutamine in ferroptosis. 94 , 99 (Figure 2).

FIGURE 2.

FIGURE 2

The role of mitochondria in ferroptosis. Fe2+ is transported into the mitochondria by SLC11A2, SLC25A37, and SLC25A28. Glutaminolysis and the TCA cycle in the mitochondria are associated with ETC activity and further promote ferroptosis. AMPK inhibits ACC‐mediated conversion of acetyl‐CoA to malonyl‐CoA, thereby ameliorating ferroptosis. Mitochondrial ETCs synthesize ATP to deactivate AMPK, thus promoting ferroptosis. Additionally, mitochondrial ETC complexes I and III generate O2•‐, which can enhance PUFA production and induce ferroptosis.

3.4. Ferroptosis defense systems

There are at least four such ferroptosis defense systems with unique subcellular localization as described below.

3.4.1. The GPX4–GSH system

Gpx4 is a member of the GPX protein family responsible for converting cytotoxic lipid peroxides to non‐toxic lipid alcohols by transforming glutathione (GSH) to oxidized glutathione (GSSG), 100 thereby preserving cell membrane integrity. 101 Glutathione reductase (GSR) can convert GSSH back to GSH by FAD (flavin adenine dinucleotide) prosthetic group and NADPH, thus contributing to the maintenance of glutathione (GSH) homeostasis and cellular protection against oxidative damage. 102 , 103 In various in vitro and in vivo conditions, genetic ablation or pharmacological inhibition of Gpx4 can lead to uncontrolled lipid peroxidation and trigger severe ferroptosis. 4 , 104 The levels of GPX4 protein are regulated by the transcription factor Nrf2 or SP2. Supplementation with intracellular glutathione or selenium upregulates GPX4 activity, while post‐translational modifications (PTMs) such as ubiquitination, phosphorylation, succinization, and glycosylation affect the protein level and activity of GPX4. 105 It was previously believed that only cytoplasmic GPX4 played a role in defending against ferroptosis. 106 However, recent studies have shown that mitochondrial GPX4 also plays a crucial role in defending against ferroptosis. 107

The Xc system comprise xCT and SLC3A2, with xCT serving as the transporter subunit. 108 xCT exhibits antiporter activity by importing extracellular cysteine into the intracellular space while exporting intracellular glutamate. 109 Through its promotion of glutathione biosynthesis, Xc system can protect cells from ferroptosis. 110

3.4.2. The FSP1‐CoQH2 system

Previous studies have indicated that ferroptosis is regulated only by GPX4 4 and free radical trapping antioxidants. 111 , 112 However, recent studies have shown that the FSP1‐CoQ10‐NAD (P)H pathway exists as a separate parallel system that operates in parallel with GPX4 and glutathione to inhibit phospholipid peroxidation and ferroptosis. 113 Ferroptosis suppressor protein 1 (FSP1, also known as AIFM2), located at the plasma membrane, is a transcriptional target for NRF2. 113 , 114 FSP1 acts as an NAD(P) H‐dependent oxidoreductase to convert Coenzyme Q10 (also known as ubiquinone) to CoQ10‐H2 (ubiquinol), 115 The redox activity of CoQ enables it to participate in the mitochondrial electron transport chain by transferring electrons from oxidative phosphorylated (OxPhos) complexes I and II to complex III. 115 In addition to its role in electron transport, CoQ can trap lipid peroxyl radicals, thereby inhibiting lipid peroxidation and ferroptosis. 113 While most CoQ is synthesized in the inner mitochondrial membrane, some may be produced outside the mitochondria. 115

3.4.3. The DHODH–CoQH2 system

The DHODH‐CoQH2 system is a mitochondrial defense mechanism that operates in parallel with the GPX4 system to prevent lipid peroxidation and ferroptosis when GPX4 is inhibited. 116 DHODH, located in the mitochondria, serves as a rate‐limiting enzyme for pyrimidine nucleotide synthesis, linking this process to the electron transport chain (ETC) at complex III via the CoQ (ubiquinone) pool. 117 Ubiquinol (CoQ10‐H2), an anti‐ferroptotic lipophilic antioxidant, is generated during the catalytic conversion of dihydroorotate to orotate by DHODH, 117 akin to the function of FSP‐1 mediated conversion of ubiquinone (CoQ10) to CoQ10‐H2 in the cytoplasm. 113

3.4.4. The GCH1–BH4 system

The guanosine 5′‐triphosphate (GTP) cyclohydrolase 1 (GCH1) tetrahydrobioterin (BH4) pathway represents an additional pivotal regulator of ferroptosis, effectively preventing the accumulation of lipid peroxides. 118 BH4, a pteridine derivative, serves as an endogenous cofactor for numerous enzymes such as nitric oxide synthase (NOS), tryptophan hydroxylase and phenylalanine carboxylase. 119 , 120 Biosynthesis of BH4 occurs via a de novo pathway at both peripheral and central levels, commencing with guanosine triphosphate (GTP) and involving three successive enzyme systems: GCH1, 6‐pyruvoyltetrahydrobiopterin synthase (PTPS) and sepiapterin reductase (SR), GCH1 acts as the rate‐limiting enzyme in BH4 synthesis. 121 , 122 Furthermore, BH4 functions as a radical‐trapping antioxidant independently from its role as a cofactor, 123 thus effectively safeguarding lipids against peroxidation. 118 , 121 , 122 Moreover, BH4 can be oxidized to dihydrobiopterin (BH2), and then regenerated by dihydrofolate reductase (DHFR), which plays an essential role in maintaining the BH4/BH2 ratio 124 (Figure 3).

FIGURE 3.

FIGURE 3

Mechanisms of ferroptosis. Ferroptosis is primarily induced by iron‐dependent lipid peroxidation. All facets of iron metabolism, encompassing absorption, export, storage and utilization, play a pivotal role in the regulation of ferroptosis. The cystine/glutamate transporter, also known as system Xc, facilitates the transport of cystine into the cell with 1:1 glutamate reverse transport. Once inside the cell, cystine can be oxidized to cysteine. The GSH‐GPX4 antioxidant system plays a crucial role in safeguarding cells against ferroptosis. ACSL4 and LPCAT3 promote the binding of PUFAs to phospholipids, forming phospholipids containing PL‐PUFA, which are susceptible to free radical‐induced oxidation mediated by lipoxygenases (ALOXs). In addition, coenzyme Q10 or tetrahydrobiopterin (BH4) inhibits ferroptosis independently of glutathione.

4. FERROPTOSIS IN ISCHEMIC STROKE

The primary pathological mechanism of IS involves the obstruction of cerebral blood flow. Current clinical treatment strategies focus on expediting the restoration of blood flow as early as possible to save the ischemic penumbra. With prolonged ischemia, there is a gradual exacerbation of neuronal cell injury and clinical symptoms, while brain injury persists during reperfusion. 125 Inflammation and excitotoxicity can further exacerbate ischemic injury. 126 , 127 Research indicated that ferroptosis plays a role in the progression of cerebral ischemia. 128 , 129 Recent studies have illustrated that ferroptosis primarily occurs during the reperfusion phase rather than the ischemia phase, as evidenced by a gradual increase in ACSL4 levels, iron content, and malondialdehyde levels with extended reperfusion time, alongside a decrease in GPX4 levels. Notably, these indicators of ferroptosis show no significant changes during ischemia. Furthermore, it has been observed that an iron chelator (deferoxamine) exerts protective effects specifically in tissue subjected to ischemia/reperfusion 130 (Figure 4).

FIGURE 4.

FIGURE 4

Mechanisms of ferroptosis in ischemic stroke. Accumulation of extracellular glutamate activates NMDAR, facilitates Ca2+ uptake, triggers cPLA2α to generate PUFA, and ACSL4 and LPCAT3 convert PUFA to PL‐PUFA. Dysregulation of glutamatergic neurotransmission leads to elevated extracellular glutamate levels, which impede cystine uptake and restrict GSH biosynthesis. Tau‐mediated reduction in APP trafficking and Fpn1 activity hinders iron export. This results in the accumulation of iron in the form of LIP, leading to toxic intracellular iron buildup and ultimately causing neuronal ferroptotic damage following ischemic stroke.

4.1. Iron metabolism dysregulation in ischemic stroke

Serum iron, ferritin, and transferrin saturation levels are correlated with an elevated risk of IS and a poorer prognosis. 131 Following cerebral ischemia/reperfusion injury (CI/RI), there is dysregulation of iron metabolism leading to iron accumulation in the brain. 132 The expression of transferrin, ferritin and transferrin receptor in the brain is upregulated under condition of cerebral ischemia and hypoxia. 133 , 134 , 135 Bivalent metal transporter 1 (DMT1) in microglia plays a pivotal role in transferrin‐dependent and non‐transferrin‐dependent iron uptake. 136 The acidic environment during cerebral ischemia and hypoxia can induce the expression of DMT1 while inhibiting the binding of iron to transferrin, resulting in dissociation between iron and transferrin, thereby increasing brain iron content. 137 , 138 Neurons readily uptake this unbound iron, leading to elevated intracellular iron. 139 Davalos et al. 140 conducted a study involving 67 cases of acute ischemic stroke which revealed that increased serum ferritin within 24 h of admission was associated with a poor prognosis for stroke patients. Subsequently measuring plasma ferritin at admission in 100 patients with cerebral infarction less than 24 h after onset indicated a significant association between plasma ferritin levels and early neurological impairment as well as stroke progression. 141

Tau is a protein predominantly expressed in the adult brain, and it plays a crucial role in neurodegenerative diseases such as Parkinson's disease and Alzheimer's disease, as well as stroke. 142 Several studies have revealed the association between Tau protein and ferroptosis. In 2017, Tuo et al. 6 illustrated that tau can modulate the trafficking of Amyloid precursor protein (APP), which stabilizes Fpn, 143 thereby promoting iron exportation and preventing ferroptotic damage.

Ischemia upregulates hepcidin expression through the JAK/STAT3 pathway, leading to FPN degradation and subsequent iron accumulation. 144 The use of iron chelators like deferoxamine can sequester labile iron pools, reducing brain iron content after ischemic stroke and contributing to decreased neuronal death. 145 , 146

4.2. Lipid metabolism in ischemic stroke

In addition to adipose tissue, the human brain exhibits the highest lipid index, constituting approximately 60% of the total dry mass of the brain. 147 Consequently, ischemic stroke can readily induce lipid peroxidation in the brain. 148 It is well‐established that ischemic stroke generates substantial amounts of ROS, leading to oxidative stress. 149 Various medicines or biomaterials acting as ROS scavengers have been shown to effectively restore the neurobiological and behavioral functions. 150 , 151 Cytoplasmic phospholipase A2α (cPLA2α) is a Ca2+ dependent cytoplasmic enzyme that plays a pivotal role in initiation of arachidonic acid (AA) metabolism. Overactivation of NMDA receptors during ischemia can activate cPLA2α by causing excessive Ca2+ intake. Thrombin activates cPLA2α to facilitate free AA release. 152 , 153

Increasing evidence has demonstrated the involvement of ACSL4 and LOX in ischemic stroke. 154 , 155 ACSL4 is a crucial isoenzyme in the metabolism of polyunsaturated fatty acids (PUFAs) such as AA, and it plays a pivotal role in determining the sensitivity to ferroptosis. 156 Studies have illustrated that oxygen and glucose deprivation (OGD) induces the expression of HIF‐1α, which then binds to the ACSL4 promoter region to inhibit its transcription. Knockdown of ACSL4 can attenuate neuronal cell death, while overexpression of ACSL4 can reduce cell viability, increase lipid peroxidation, and aggravate ischemic brain injury. Ferroptosis inhibitor liproxstatin‐1 can alleviate the adverse reactions caused by overexpression of ASCL4. 128 Tuo et al. 157 found that knockout of ACSL4 did not affect the blood flow in the cortex of rats after MCAO/R but reduced infarct volume and alleviated nerve injury by inhibiting ferroptosis. Chen et al. 158 demonstrated that inhibition of ACSL4 can mitigate the lipid peroxidation associated with ferroptosis after stroke, improve nerve function, and reduce infarct volume. Liu et al. 159 revealed that calycosin, a phytoestrogen extracted from the root of Astragali radix, connects stably with ACSL4 to suppress lipid peroxidation associated with ferroptosis in ischemic stroke.

LOX catalyzes lipid peroxidation and triggers ferroptosis in numerous diseases. 160 The expression of 12/15‐LOX is upregulated in neurons around the infarction area of brain, the inhibition of 12/15‐LOX, or the knockout of 12/15‐LOX gene can reduce the infarct size. 155 , 161 , 162 For example, baicalein has been reported to selectively inhibit 12/15‐LOX, thereby offering protection against neuronal cell death following ischemic stroke. 161

Nrf2 can regulate a wide range of downstream factors, playing diverse roles such as anti‐ferroptosis, anti‐apoptosis, reduction of calcium overload, anti‐oxidative stress, and anti‐inflammatory. It assists in maintaining the REDOX reaction of brain tissue and cells. 163 Nrf2 plays a crucial role in the management of excessive oxidative stress following ischemic stroke. During ischemia–reperfusion injury, Nrf2 translocates to the nucleus and subsequently activates HO‐1, further inhibiting inflammation. 164 Apelin, a peptide hormone released from adipose tissue, modulates oxidative stress in many tissues. Duan et al. 165 illustrated that apelin treatment significantly decreased ROS, lipid peroxidation and inflammation through the AMPK/GSK‐3β/Nrf2 signaling pathway, therefore mediating neuroprotective effects after I/R in the brain. Furthermore, another study showed that Rehmannioside A reduced oxidative stress, diminished infarct volume, and improved cognitive impairment in I/R rats by attenuating ferroptosis through the PI3K/Nrf2/SLC7A11 pathway. 166

4.3. Amino acid metabolism in ischemic stroke

Glutamate serves as the primary excitatory neurotransmitter in the central nervous system (CNS), regulating the neural function and closely related to cell activities, synaptic activity, plasticity, pain perception, learning and memory. 167 , 168 The toxic impact of excessive or prolonged extracellular glutamate activation of the excitatory amino acid receptors (particularly NMDARs and AMPARs) is referred to as excitotoxicity. 169 Glutamate excitotoxicity is widely regarded as one of the mechanisms underlying ferroptosis. 170 In ischemic stroke, membrane depolarization and potential disruption occur following energy failure, leading to the diffusion of glutamate into the extra‐synaptic region. 171 Impeded reuptake results in a rapid increase in glutamate levels within ischemic brain areas. 172 I Inhibiting glutamate release, accelerating its clearance, and blocking its receptors may offer therapeutic benefits for ischemic stroke. For instance, methionine sulfoximine can inhibit the synthesis of glutamine. 173 mGluR2 and mGluR3 belong to the Group II mGluR family, and their agonists suppress the release of glutamate from presynaptic nerve terminals. 174 Study have demonstrated that both knockdown of High mobility group box 1 (HMGB1) and suppression of the HMGB1‐TLR4 signaling pathway can increase the expression of glutamate aspartate transporter (GLAST), a specific glutamate transporter, thus enhancing the clearance of glutamate. 175 N‐methyl‐d‐aspartate receptors (NMDARs) are transmembrane proteins, NMDAR channel blockers constitute a class of antagonists with broad clinical and pharmacological significance. Channel blockers usually enter their binding sites only when the channel is open, hence they are also referred to as open channel blockers. NMDAR channel blockers used for treatment include Mg2+, ketamine, MK‐801, dextrorphan, memantine, and phencyclidine (PCP). 176 , 177 , 178 , 179 , 180 For instance, Ketamine and its metabolites can enhance AMPAR signaling by inhibiting neuronal extrasynaptic NMDARs. AMPARs can stimulate Brain derived neurotrophic factor (BDNF) secretion, thus influencing cell growth and differentiation, cell survival, and synaptic protein transcription. 181 Study has shown that Glutamate induces acid sphingomyelinase activation and sphingosine accumulation leading to increased generation of ROS which results in the opening of mitochondrial permeability transition pore, the dissipation of mitochondrial transmembrane potential and the rupture of outer membrane, ultimately resulting in system Xc dependent ferroptosis. 182 However, studies have found that system Xc activity rapidly increased after transient middle cerebral artery occlusion (tMCAO) in the rat brain, 183 and xCT protein levels in microglia/macrophages, neurons, and astrocytes were elevated. 184 , 185 Hewett et al. 186 compared infarcts in mice naturally null for SLC7a11 (SLC7a11sut/sut mice) with wild type (SLC7a11+/+) after middle cerebral artery photothrombotic ischemic stroke (PTI) and permanent middle cerebral artery occlusion (pMCAO). They found that the infarct volume in SLC7a11sut/sut mice decreased significantly 48 h after PTI, but no difference between the two groups when MCA was permanently occluded, suggesting that system Xc aggravated the damage in mice with non‐severe ischemic stroke. In conclusion, IS increased the expression of system Xc and promotes the export of glutamate from neurons, the excitotoxic effect caused by extracellular glutamate outweigh the protection effect of system Xc. 187

In addition, glutathione acts as an endogenous ferroptosis inhibitor. 188 Glutathione levels are depleted in oxidative stress disorders, including stroke, and diminished brain glutathione levels are associated with increased stroke risk. Conversely, replenishing glutathione levels has been shown to reduce neuronal cell death and improve animal function recovery following a stroke. Therefore, maintaining stable intracellular glutathione levels is crucial for ischemic stroke. 189 GPX4 protein expression was significantly reduced in both vivo and in vitro ischemic stroke models. 190 Li and colleagues 154 demonstrated that Baicalin, the main component of Scuetellaria baicalensis Georgi root, prevented ferroptotic injury in tMACO mice or OGD/R cells by enhancing the expression of GPX4. Liu et al. 191 found that edaravone, a free radical scavenger, can reduce infarct volume and functional impairment through anti‐ferroptotic mechanisms such as increasing GSH level and the expression of GPX4.

4.4. New hope for IS treatment—inhibition of ferroptosis

Treatment against ischemic stroke by targeting ferroptosis has gained much attention. 192 Vitexin, 193 Angong Niuhuang Wan, 194 selenium, 195 Srs11‐92, 196 Se‐methyl L‐selenocysteine, 197 Rehmannioside A, 166 Icariside II, 198 Galangin, 199 Carvacrol, 200 Carthamin yellow, 201 β‐Caryophyllene, 202 Astragaloside IV, 203 Salvia miltiorrhiza, 204 Naotaifang extract, 133 Naotaifang, 205 Baicalein, 154 Caffeic acid, 206 Danlou tablet, 207 Rhein, 208 HSYA and AHSYB, 209 Calycosin, 159 Ferrostatin‐1, 210 Compound Tongluo Decoction, 211 Loureirin C, 212 Edaravone, 191 Quercetin, 213 Cottonseed oil, 214 DHT, 215 Dl‐3‐n‐butylphthalide, 216 Dimethyl fumarate, 217 2‐(1‐(4‐(4‐methylpiperazin‐1‐yl)phenyl)ethyl)‐10H‐phenothiazine, 218 Kaempferol, 219 Danhong injection, 220 Neutral polysaccharide from Gastrodia elata, 221 Resveratrol 190 alleviate injury after IS by inhibiting ferroptosis (Table 1).

TABLE 1.

Summary of compounds Interfering with ferroptosis in ischemic stroke.

Authors Compounds Study models Changes Effects Mode of action
Guo et al. 193 Vitexin OGD/R neuron cells Inhibit ↓Cell apoptosis; ↓mitochondria injury; ↓SLC7A11; ↑SOD, GSH, Nrf2, Keap1, Tfr1 Keap1/Nrf2/HO‐1
Guo et al. 193 Vitexin MCAO/R rats Inhibit ↑Cell viability; ↓cell apoptosis; ↓neurological Scores; ↓infracted volume; ↓ROS, MDA; ↑SOD, GSH, Nrf2, Keap1, Tfr1; ↓SLC7A11 Keap1/Nrf2/HO‐1
Bai et al. 194 Angong Niuhuang Wan MCAO/R rats Inhibit ↑Nerve function; ↓cerebral infarct volume; ↓mitochondrial dysfunction and morphology disruption, ↑nerve function; ↓cerebral infarct volume; ↑structural integrity of the BBB; ↓ROS, LPO, Fe2+; ↑expressions of PPARγ, p‐AKT/AKT and GPX4 ↑PPARγ/AKT/GPX4
Shi et al. 195 Selenium MCAO/R mice Inhibit ↓MDA; ↑SOD; ↑GPx4; ↓iron content; ↓p22phox; ↑survival rate of mice; ↓cerebral infarction area; ↓brain cell apoptosis ↓LPO; ↓Fe2+
Shi et al. 195 Selenium OGD/R N2a cells Inhibit ↑Cell viability; ↓iron content; ↑GSH; ↑GSH/GSSG; ↑Fth1; ↓Cox2, MDA; ↑SOD, GPx4; ↓p22phox; ↑MMP, Mfn1, mtDNA level ↑Mfn1
Chen et al. 196 Srs11‐92 MCAO/R mice Inhibit ↓Neurological deficit score; ↑GSH/GSSG; ↑SOD; ↓ROS. MDA; ↑GPx4, Nrf2, HO‐1 ↑Nrf2/HO‐1
Fei et al. 197 Se‐methyl L‐selenocysteine MCAO/R mice Inhibit ↓Infarct volume; ↓brain edema; ↓neurological dysfunction; ↑morphology of neurons; ↑tight junction protein; ↑BBB function; ↑GPX4; ↓ACSL4; ↓4‐HNE; ↑p‐Akt and p‐GSK3β ↑Akt/GSK3β
Fei et al. 197 Se‐methyl L‐selenocysteine OGD/R bEnd.3 cells Inhibit ↑Cell viability; ↑GPX4; ↑BBB function; ↓ACSL4; ↓4‐HNE; ↑Nrf2; ↑p‐Akt and p‐GSK3β ↑Akt/GSK3β
Fu et al. 166 Rehmannioside A MCAO/R rats Inhibit ↓Infarct volume, neurological defects, cognitive impairment; ↑SOD; ↓MDA, MPO, p‐PI3K, p‐Akt; ↑Nrf2, HO‐1, SLC7A11, GPX4 PI3K/Nrf2/SLC7A11
Gao et al. 198 Icariside II MCAO/R mice Inhibit ↓Neurological deficits; ↑sensory motor function; ↓infarct volume; ↑Nrf2 level within nuclei; ↓Nrf2 level within cytoplasm;↑SIRT5, HO‐1, NQO‐1, GPX4, IκBα; ↓the phosphorylation level and activity of NF‐κBp65; ↓mitochondrial dysfunction; ↓mitochondrial ROS, MDA and iron content; ↑NADPH/NADP+ ratio, RCI, ATP, GPX4 level, SOD2 activity, SIRT5 activity; ↓IL‐1β, IL‐6, TNF‐α; ↓the number of GFAP‐positive cells ↑Nrf2; ↓NF‐Κb; ↑GPX4
Gao et al. 198 Icariside II OGD primary astrocytes Inhibit ↑Cell viability; ↓LDH; ↓mitochondrial ROS and iron content; ↑RCI, SIRT5, IκBα; ↓IL‐1β, IL‐6, TNF‐α, NF‐κBp65 ↑Nrf2; ↓NF‐Κb; ↑GPX4
Guan et al. 199 Galangin BCAL/R gerbils Inhibit ↓Learning and memory deficits; ↑the neurons of CA1; ↓cell death of hippocampal neurons; ↓the hippocampus impairment; ↑GSH, GSHPX, SOD; ↓Fe2+, 4‐HNE; ↑SLC7A11, GPX4, H2AX ↑SLC7A11/GPX4
Guan et al. 199 Galangin OGD/R hippocampal neurons Inhibit ↑Survival rate of neurons; ↑SLC7A11; ↓Fe2+, ↑GPX4; ↓MDA, H2AX ↑SLC7A11/GPX4
Guan et al. 200 Carvacrol BCAL/R gerbils Inhibit ↑SOD, GSH, GPx4 and Fpn1;↓MDA, TFR1; ↑memory and learning ability; ↓hippocampus impairment ↑GPx4 axis
Guan et al. 200 Carvacrol OGD/R Hippocampal neurons Inhibit ↑GPx4; ↑cell viability; ↓MDA, Fe2+ ↑GPx4 axis
Guo et al. 201 Carthamin yellow MCAO/R rats Inhibit ↓Neurological deficits, infarction volume, brain water content; ↑MAP‐2; ↑GSH, SOD; ↓MDA, ACSL4 and TFR1; ↑FTH1, GPX4 ↑GPx4 axis
Hu et al. 202 β‐Caryophyllene MCAO/R rats Inhibit ↓Neurological deficits; ↓cerebral infarction volume; ↓cerebral pathological damage; ↑NRF2, HO‐1, GPX4; ↓ACSL4, COX‐2; ↑GSH; ↓4‐HNE, MDA ↑NRF2/HO1
Hu et al. 202 β‐Caryophyllene OGD/R astrocytes Inhibit ↑Cell viability; ↑NRF2/HO1; ↓COX2; ↑GPX4 ↑NRF2/HO1
Jin et al. 203 Astragaloside IV MCAO/R mice Inhibit ↓infarct size and neurological impairment; ↓MDA, Fe2+, Acsl4; ↑GSH ↓Acsl4
Jin et al. 203 Astragaloside IV OGD/R neuron cells Inhibit ↑Cell viability; ↓MDA, LDH, Acsl4 and Fe2+; ↑GSH, Slc7a11 and Gpx4; ↑Atf3, Fto ↑Atf3/Fto, ↓Acsl4
Ko et al. 204 Salvia miltiorrhiza Transient MCAO mice Inhibit ↓cerebral infarction volume; ↓brain edema; ↓neuronal loss; ↓neurological deficits; ↓cognitive impairment;↑survival rate of mice;↓iNOS ROS and 4‐HNE; ↑GSH, GPX4, FPN1; ↓ACSL4, Ferritin, iron deposition in the brain ↓LPO; ↑GPX4
Lan et al. 133 Naotaifang extract MCAO rats Inhibit ↓Neurobehavioral scores; ↓iron deposition; ↑GSH; ↓ROS, MDA, TFR1 and DMT1; ↑SLC7A11 and GPX4 ↑SLC7A11, GPX4
Liao et al. 205 Naotaifang OGD/R BV2 cells Attenuate ↑Cell viability; ↓Fe2+, LPO, ROS, MDA; ↑FPN and GPX4; ↓BMP6, p‐SMADs, p‐STAT3, hepcidin ↓BMP6/SMADs; ↓STAT3
Li et al. 154 Baicalein Transient MCAO mice Inhibit ↓Infarct volume; ↓neurological deficits; ↓Fe2+, MDA; ↑GSH, GPX4, ACSL3 and xCT; ↓ACSL4; ↑FTH and mitochondrial ferritin (FTMT) ↑GPX4, ACSL3; ↓ACSL4
Li et al. 154 Baicalein OGD/R HT22 cells Inhibit ↑Cell viability; ↓LDH, ROS and superoxide anion ↓LPO
Li et al. 154 Baicalein RSL3 exposed HT22 cells Inhibit ↑Cell viability; smaller volume, higher mitochondrial membrane electron density and disrupted mitochondrial cristae were ameliorated; ↓ROS, MDA; ↑GSH, MMP, FTH, FTMT, GPX4 and xCT; ↓ACSL4; ↑ACSL3 ↑GPX4, ACSL3; ↓ACSL4
Li et al. 206 Caffeic acid Permanent MCAO rats Alleviate ↑Cerebral infarction volume; ↑nerve function; ↑GPx GSH, and SOD; ↑Nrf2, HO‐1, AKT and GSK3β; ↑SLC3A2; ↓TFR1, ACSL4; ↓COX2 ↑GPx4; ↑Nrf2
Li et al. 206 Caffeic acid OGD/R SK‐N‐SH cells Alleviate ↓MDA and ROS; ↑GPx4; ↑Nrf2, HO‐1, SLC3A2; ↓ACSL4, TFR1 ↑GPx4; ↑Nrf2
Liu et al. 207 Danlou tablet Transient MCAO mice Attenuate ↑Neurological deficits; ↓infarct volume; ↓brain water content; ↓BBB disruption; ↓MDA, GSSG, ROS;↑GSH;↓COX2; ↑GPX4 and SLC7A11 ↓LPO; ↑GPX4
Liu et al. 207 Danlou tablet OGD/R hy926 cells Inhibit ↓Cell death; ↓barrier damage; ↑mitochondrial membrane potential; ↓MDA, GSSG, ROS;↑GSH;↓COX2; ↑GPX4 and SLC7A11 ↓LPO; ↑GPX4
Liu et al. 208 Rhein MCAO/R rats Inhibit ↓Infarct volume; ↓neurological deficits; ↓BBB disruption; ↑GSH; ↓MDA, Fe2+, ROS; ↑GPX4, NRF2, SLC7A11 ↑NRF2/SLC7A11/GPX4
Liu et al. 208 Rhein OGD/R HT22 cells Inhibit ↑Cell viability; ↓ROS, MDA, Fe2+; ↑NRF2, GPX4, SLC7A11 ↑NRF2/SLC7A11/GPX4
Chen et al. 209 HSYA and AHSYB OGD/R PC12 cells Inhibit ↑Cell viability; ↓ROS, Fe2+, 4‐HNE, MDA; ↑GSH/GSSG level, GPX4, SLC7A11; ↓mitochondrial morphological damage ↑SLC7A11, GPX4; ↓LPO
Liu et al. 159 Calycosin MCAO/R rats Ameliorate ↓Neurological defect; ↓brain edema; ↓BBB damage; ↓cerebral infarction; ↓Fe2+, MDA, ROS; ↑SOD; ↓ACSL4 and TfR1; ↑FTH1 and GPX4 ↓ACSL4
Liu et al. 159 Calycosin OGD/R PC12 cells Inhibit ↑Cell viability; ↓cell apoptosis; ↓Fe2+, MDA, ROS; ↑SOD; ↓ACSL4 and TfR1; ↑FTH1 and GPX4 ↓ACSL4
Liu et al. 210 Ferrostatin‐1 MCAO/R mice Inhibit ↓Total iron level; ↑GSH, SLC7A11, GPX4; ↓mitochondrial membrane density; ↑mitochondrial cristae; ↓infarct volume, neurological deficits; ↑p‐AKT and p‐GSK3β ↑AKT
Liu et al. 210 Ferrostatin‐1 OGD/R HT22 cells Inhibit ↑Cell viability; ↑SLC7A11, GPX4, GSH; ↓Fe2+; ↑p‐AKT and p‐GSK3β ↑AKT
Hui et al. 211 Compound Tongluo Decoction MCAO/R rats Inhibit ↓Cerebral ischemia area; ↓ROS, MDA; ↑SOD; ↓ACSL4, ALOX5; ↑GPX4; ↑angiogenesis; ↑Sonic Hedgehog pathway Sonic Hedgehog pathway
Hui et al. 211 Compound Tongluo Decoction OGD/R PC12 cells Inhibit ↑Cell viability; ↑Sonic Hedgehog pathway; ↓ACSL4, ALOX5; ↑GPX4;↓ATF4, PERK and cleaved caspase 3; ↑angiogenesis Sonic Hedgehog pathway
Liu et al. 212 Loureirin C MCAO/R mice Inhibit ↓Neurological scores, brain edema, infarct volume; ↓Fe2+; ↑GSH, GPX4, SOD; ↓MDA; ↓mitochondrial membrane density; ↑mitochondrial cristae; ↑Nrf2, HO‐1 ↑Nrf2
Liu et al. 212 Loureirin C OGD/R SH‐SY5Y cells Inhibit ↑Cell viability; ↓TFR1; ↑GSH, GPX4, SLC7A11, SOD; ↓DHE, MDA, Fe2+, ROS, LPO, Keap1; ↑nuclear Nrf2, GPX4, HO‐1 ↑Nrf2
Liu et al. 191 Edaravone MCAO/R rats Inhibit ↑Sensorimotor function; ↓cerebral infarct volume; ↓Fe2+, MDA, LPO; ↑GSH, GPX4, total Nrf2, nuclear Nrf2, FPN; ↓IL‐6, IL‐1β, TNF‐α and MPO ↑Nrf2/FPN
Peng et al. 213 Quercetin Permanent MCAO rats Inhibit ↓Infarct volume, neurological scores; ↓Fe2+, MDA; ↑GSH, SOD, GPX4, FTH1; ↓ACSL4; ↑Nrf2, HO‐1, nuclear translocation of Nrf2 ↑Nrf2/HO‐1
Sun et al. 214 Cottonseed oil MCAO/R rats Attenuate ↓Infarct volume; ↓neurological deficit; ↓BBB disruption, brain water content; ↓transferrin; ↑GPX4, xCT and FTH1; ↓ACSL4; ↑HO‐1, GSH, ROS; ↓MDA, LPO; ↑the proportion of normal mitochondria ↑GPX4; ↓LPO
Wu et al. 215 DHT Permanent MCAO rats Inhibit ↓Infarct volume, neurological score, cerebral edema; ↑Gpx4, GSH, mitochondrial viability; ↓Fe2+ ↑Gpx4; ↓Fe2+
Wu et al. 215 DHT t‐BHP PC12 cells Inhibit ↑Cell viability; ↓ROS, Fe2+; ↑Gpx4, GSH/GSSG; ↑MMP; ↓mitochondrial ROS; ↑Nrf2, HO‐1 ↑Nrf2/HO‐1
Xu et al. 216 Dl‐3‐n‐butylphthalide MCAO/R rats Attenuate ↓Neurological deficit score; ↓brain water content; ↓cerebral infarct area; ↑neuronal pathology; ↑number of Nissl bodies; ↓neuronal apoptosis, BBB damage; ↑p‐Akt and p‐PI3K; ↓p‐PDGFRβ; ↓TFRC, ROS, MDA; ↑GSH, SLC7A11, GPX4 ↑SLC7A11/GSH/GPX4
Yan et al. 217 Dimethyl fumarate Chronic cerebral hypoperfusion rats Inhibit ↓Cognitive impairment; ↓neuronal damage; ↓neuronal loss in hippocampal CA1 regions; ↓Fe2+, MDA; ↑GSH, SOD; ↓PTGS2; ↑FTH1, xCT; ↓IL‐1β, TNF‐α, IL‐6, NF‐κB‐p65; ↑IκBα; ↑HO‐1, NQO1, GPX4, NRF2 ↑NRF2
Yang et al. 218 2‐(1‐(4‐(4‐methylpiperazin‐1‐yl)phenyl)ethyl)‐10H‐phenothiazine MCAO/R rats Inhibit ↓Cerebral infarction volume; ↓neurological score; ↑GSH; ↓MDA ↑GSH
Yuan et al. 219 Kaempferol OGD/R neuron cell Inhibit ↑SLC7A11, GPX4, nuclear Nrf2; ↓cytoplasmic Nrf2; ↑GSH/GSSG, NADPH/NADP+, SOD; ↓Fe2+, LPO, 4‐HNE; ↓morphological changes of mitochondria ↑Nrf2; ↓LPO
Zhan et al. 220 Danhong injection Permanent MCAO mice Inhibit ↓Cerebral infarct volume; ↓neurological scores; ↑numbers of Nissl‐positive neurons; ↓TfR1, TF; ↑Ferritin; ↑SATB1, p‐SATB1, SLC7A11, GPX4, HO‐1 ↑SATB1/SLC7A11/HO‐1
Zhan et al. 220 Danhong injection OGD HT22 cells Inhibit ↑Cell viability; ↓MDA; ↑SOD, GSH, SATB1, p‐SATB1, SLC7A11, HO‐1 ↑SATB1/SLC7A11/HO‐1
Zhang et al. 221 Neutral polysaccharide from Gastrodia elata MCAO/R mice Inhibit ↓Infarct volume; ↓neurological deficit score; ↓cerebral edema, aquaporin 4; ↑GPX4, GSH; ↓Fe2+; ↑nuclear NRF2, HO‐1 ↑NRF2/HO‐1
Zhang et al. 221 Neutral polysaccharide from Gastrodia elata OGD/R HT22 cells Inhibit ↑Cell viability; ↓ROS, MDA; ↑SOD; ↓Fe2+; ↑GSH, SLC7A11, GPX4; ↑NRF2, HO‐1 ↑NRF2/HO‐1
Zhu et al. 190 Resveratrol MCAO/R rats Inhibit ↓Mitochondrial damage; ↓cerebral infarct volume; ↓neuronal degeneration; ↓ACSL4; ↑ferritin and GPX4 ↓ACSL4; ↑GPX4
Zhu et al. 190 Resveratrol OGD/R primary cortical neurons Inhibit ↑Neuronal viability; ↓Fe2+, MDA; ↑GSH; ↓ACSL4; ↑ferritin and GPX4; ↓mitochondrial damage ↓ACSL4; ↑GPX4; ↓Fe2+

Abbreviations: AHSYB, anhydrosafflor yellow B; BBB, blood−brain barrier; BCAL/R, bilateral carotid artery ligation/reperfusion; DHE, Dihydroethidium; DHT, 15, 16‐Dihydrotanshinone I; Fth1, Ferritin Heavy Chain 1; GSH, glutathione; GSSG, oxidized glutathione; HSYA, Hydroxysafflor yellow A; Mfn 1, mitofusin 1; MMP, mitochondrial membrane potential; mtDNA, mitochondrial DNA; p‐Akt, Phosphorylated AKT protein; p‐GSK3β, phosphorylated‐GSK‐3 Beta; p‐PI3K, phosphorylation phosphotylinosital 3 kinase; t‐BHP, tert‐Butyl hydroperoxide; TF, transferrin; TfR1, transferrin receptor 1; TFRC, Transferrin receptor.

5. CONCLUSION AND PERSPECTIVES

Ischemic stroke is a prevalent condition and represents a primary contributor to both mortality and disability. 222 The underlying pathological mechanisms remain to be clarified. 223 Ferroptosis has been implicated in the secondary brain injury associated with ischemic stroke. In the acute phase of ischemic stroke, disruption of the blood–brain barrier leads to aggregation of iron ions in the brain parenchyma, triggering the conversion of hydrogen peroxide to hydroxyl radicals through Fenton reaction, concomitant with reduced GSH levels and increased lipid peroxidation. 224 This review provides an overview of recent advancements in understanding the pathogenic pathways and regulatory mechanisms of ferroptosis in ischemic stroke, as well as discusses potential therapeutic applications targeting ferroptosis inhibition for treating ischemic stroke. Nevertheless, our understanding of the role of ferroptosis in ischemic stroke remains limited, leaving several questions unanswered. For instance, further clarification is needed regarding the involvement of ferroptosis in neurons, astrocytes, microglia, and oligodendrocytes during ischemic brain injury. Additionally, while other forms of programmed cell death have distinct markers (e.g., caspase activation for apoptosis and gasdermin D for pyroptosis), 225 specific markers for ferroptosis are still lacking despite current studies being primarily focused on iron levels, GPX4 expression, lipid hydroperoxides, ROS production, and cell viability. Therefore, it is imperative to identify specific markers for ferroptosis. Furthermore, clinical studies investigating ferroptosis in ischemic stroke are warranted given that most research efforts thus far have predominantly centered around cellular and animal models.

AUTHOR CONTRIBUTIONS

Zhaohui Chai was responsible for writing the manuscript, and Jian Shen and Jiesheng Zheng were responsible for research design. All the authors have contributed to the completion of this paper.

CONFLICT OF INTEREST STATEMENT

The authors declare that they have no competing interests.

ACKNOWLEDGMENTS

This study was financially supported by grants from the National Natural Science Foundation of China (82071285) and the Medical Science and Technology Project of Zhejiang Province (2022505533).

Chai Z, Zheng J, Shen J. Mechanism of ferroptosis regulating ischemic stroke and pharmacologically inhibiting ferroptosis in treatment of ischemic stroke. CNS Neurosci Ther. 2024;30:e14865. doi: 10.1111/cns.14865

DATA AVAILABILITY STATEMENT

Data sharing not applicable to this article as no datasets were generated or analysed during the current study.

REFERENCES

  • 1. Dixon SJ, Lemberg KM, Lamprecht MR, et al. Ferroptosis: an iron‐dependent form of nonapoptotic cell death. Cell. 2012;149:1060‐1072. doi: 10.1016/j.cell.2012.03.042 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Feng H, Stockwell BR. Unsolved mysteries: how does lipid peroxidation cause ferroptosis? PLoS Biol. 2018;16:e2006203. doi: 10.1371/journal.pbio.2006203 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Lei G, Zhuang L, Gan B. Targeting ferroptosis as a vulnerability in cancer. Nat Rev Cancer. 2022;22:381‐396. doi: 10.1038/s41568-022-00459-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Yang WS, SriRamaratnam R, Welsch ME, et al. Regulation of ferroptotic cancer cell death by GPX4. Cell. 2014;156:317‐331. doi: 10.1016/j.cell.2013.12.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Stockwell BR, Friedmann Angeli JP, Bayir H, et al. Ferroptosis: a regulated cell death nexus linking metabolism, redox biology, and disease. Cell. 2017;171:273‐285. doi: 10.1016/j.cell.2017.09.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Tuo QZ, Lei P, Jackman KA, et al. Tau‐mediated iron export prevents ferroptotic damage after ischemic stroke. Mol Psychiatry. 2017;22:1520‐1530. doi: 10.1038/mp.2017.171 [DOI] [PubMed] [Google Scholar]
  • 7. Weiland A, Wang Y, Wu W, et al. Ferroptosis and its role in diverse brain diseases. Mol Neurobiol. 2019;56:4880‐4893. doi: 10.1007/s12035-018-1403-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Xiao Z, Shen D, Lan T, et al. Reduction of lactoferrin aggravates neuronal ferroptosis after intracerebral hemorrhagic stroke in hyperglycemic mice. Redox Biol. 2022;50:102256. doi: 10.1016/j.redox.2022.102256 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Huang Y, Wu H, Hu Y, et al. Puerarin attenuates oxidative stress and ferroptosis via AMPK/PGC1α/Nrf2 pathway after subarachnoid hemorrhage in rats. Antioxidants (Basel, Switzerland). 2022;11:1‐17. doi: 10.3390/antiox11071259 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Wang T, Tomas D, Perera ND, et al. Ferroptosis mediates selective motor neuron death in amyotrophic lateral sclerosis. Cell Death Differ. 2022;29:1187‐1198. doi: 10.1038/s41418-021-00910-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Tian Y, Lu J, Hao X, et al. FTH1 inhibits ferroptosis through ferritinophagy in the 6‐OHDA model of Parkinson's disease. Neurotherapeutics. 2020;17:1796‐1812. doi: 10.1007/s13311-020-00929-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Jiang L, Kon N, Li T, et al. Ferroptosis as a p53‐mediated activity during tumour suppression. Nature. 2015;520:57‐62. doi: 10.1038/nature14344 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Tsoi J, Robert L, Paraiso K, et al. Multi‐stage differentiation defines melanoma subtypes with differential vulnerability to drug‐induced iron‐dependent oxidative stress. Cancer Cell. 2018;33:890‐904.e895. doi: 10.1016/j.ccell.2018.03.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Qin Y, Qiao Y, Wang D, Tang C, Yan G. Ferritinophagy and ferroptosis in cardiovascular disease: mechanisms and potential applications. Biomed Pharmacother. 2021;141:111872. doi: 10.1016/j.biopha.2021.111872 [DOI] [PubMed] [Google Scholar]
  • 15. Wang Y, Tang M. PM2.5 induces ferroptosis in human endothelial cells through iron overload and redox imbalance. Environ Pollut. 2019;254:112937. doi: 10.1016/j.envpol.2019.07.105 [DOI] [PubMed] [Google Scholar]
  • 16. Global, regional, and national burden of stroke and its risk factors, 1990‐2019: a systematic analysis for the global burden of disease study 2019. Lancet Neurol. 2021;20:795‐820. doi: 10.1016/s1474-4422(21)00252-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Dolma S, Lessnick SL, Hahn WC, Stockwell BR. Identification of genotype‐selective antitumor agents using synthetic lethal chemical screening in engineered human tumor cells. Cancer Cell. 2003;3:285‐296. doi: 10.1016/s1535-6108(03)00050-3 [DOI] [PubMed] [Google Scholar]
  • 18. Yagoda N, von Rechenberg M, Zaganjor E, et al. RAS‐RAF‐MEK‐dependent oxidative cell death involving voltage‐dependent anion channels. Nature. 2007;447:864‐868. doi: 10.1038/nature05859 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Shterman N, Kupfer B, Moroz C. Comparison of transferrin receptors, iron content and isoferritin profile in normal and malignant human breast cell lines. Pathobiology. 1991;59:19‐25. doi: 10.1159/000163611 [DOI] [PubMed] [Google Scholar]
  • 20. Yang WS, Stockwell BR. Synthetic lethal screening identifies compounds activating iron‐dependent, nonapoptotic cell death in oncogenic‐RAS‐harboring cancer cells. Chem Biol. 2008;15:234‐245. doi: 10.1016/j.chembiol.2008.02.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Loscalzo J. Membrane redox state and apoptosis: death by peroxide. Cell Metab. 2008;8:182‐183. doi: 10.1016/j.cmet.2008.08.004 [DOI] [PubMed] [Google Scholar]
  • 22. Seiler A, Schneider M, Förster H, et al. Glutathione peroxidase 4 senses and translates oxidative stress into 12/15‐lipoxygenase dependent‐ and AIF‐mediated cell death. Cell Metab. 2008;8:237‐248. doi: 10.1016/j.cmet.2008.07.005 [DOI] [PubMed] [Google Scholar]
  • 23. Eagle H. Nutrition needs of mammalian cells in tissue culture. Science. 1955;122:501‐514. doi: 10.1126/science.122.3168.501 [DOI] [PubMed] [Google Scholar]
  • 24. Guo W, Zhao Y, Zhang Z, et al. Disruption of xCT inhibits cell growth via the ROS/autophagy pathway in hepatocellular carcinoma. Cancer Lett. 2011;312:55‐61. doi: 10.1016/j.canlet.2011.07.024 [DOI] [PubMed] [Google Scholar]
  • 25. Banjac A, Perisic T, Sato H, et al. The cystine/cysteine cycle: a redox cycle regulating susceptibility versus resistance to cell death. Oncogene. 2008;27:1618‐1628. doi: 10.1038/sj.onc.1210796 [DOI] [PubMed] [Google Scholar]
  • 26. Dutt S, Hamza I, Bartnikas TB. Molecular mechanisms of iron and heme metabolism. Annu Rev Nutr. 2022;42:311‐335. doi: 10.1146/annurev-nutr-062320-112625 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Corradini E, Buzzetti E, Pietrangelo A. Genetic iron overload disorders. Mol Asp Med. 2020;75:100896. doi: 10.1016/j.mam.2020.100896 [DOI] [PubMed] [Google Scholar]
  • 28. Halcrow PW, Lynch ML, Geiger JD, Ohm JE. Role of endolysosome function in iron metabolism and brain carcinogenesis. Semin Cancer Biol. 2021;76:74‐85. doi: 10.1016/j.semcancer.2021.06.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Rauhala P, Chiueh CC. Effects of atypical antioxidative agents, S‐nitrosoglutathione and manganese, on brain lipid peroxidation induced by iron leaking from tissue disruption. Ann N Y Acad Sci. 2000;899:238‐254. doi: 10.1111/j.1749-6632.2000.tb06190.x [DOI] [PubMed] [Google Scholar]
  • 30. Sokolov AV, Voynova IV, Kostevich VA, Vlasenko AY, Zakharova ET, Vasilyev VB. Comparison of interaction between ceruloplasmin and lactoferrin/transferrin: to bind or not to bind. Biochemistry (Mosc). 2017;82:1073‐1078. doi: 10.1134/s0006297917090115 [DOI] [PubMed] [Google Scholar]
  • 31. Shang Y, Luo M, Yao F, Wang S, Yuan Z, Yang Y. Ceruloplasmin suppresses ferroptosis by regulating iron homeostasis in hepatocellular carcinoma cells. Cell Signal. 2020;72:109633. doi: 10.1016/j.cellsig.2020.109633 [DOI] [PubMed] [Google Scholar]
  • 32. Curzon G. Some properties of coupled iron‐caeruloplasmin oxidation systems. Biochem J. 1961;79:656‐663. doi: 10.1042/bj0790656 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Bakhautdin B, Goksoy Bakhautdin E, Fox PL. Ceruloplasmin has two nearly identical sites that bind myeloperoxidase. Biochem Biophys Res Commun. 2014;453:722‐727. doi: 10.1016/j.bbrc.2014.09.134 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Zanardi A, Conti A, Cremonesi M, et al. Ceruloplasmin replacement therapy ameliorates neurological symptoms in a preclinical model of aceruloplasminemia. EMBO Mol Med. 2018;10:91‐106. doi: 10.15252/emmm.201708361 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Chen X, Yu C, Kang R, Tang D. Iron metabolism in ferroptosis. Front Cell Dev Biol. 2020;8:590226. doi: 10.3389/fcell.2020.590226 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Wu Y, Ma Z, Mai X, et al. Identification of a novel inhibitor of TfR1 from designed and synthesized muriceidine a derivatives. Antioxidants (Basel, Switzerland). 2022;11:1‐2. doi: 10.3390/antiox11050834 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Kosman DJ. A holistic view of mammalian (vertebrate) cellular iron uptake. Metallom: Integr Biometal Sci. 2020;12:1323‐1334. doi: 10.1039/d0mt00065e [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Sendamarai AK, Ohgami RS, Fleming MD, Lawrence CM. Structure of the membrane proximal oxidoreductase domain of human Steap3, the dominant ferrireductase of the erythroid transferrin cycle. Proc Natl Acad Sci USA. 2008;105:7410‐7415. doi: 10.1073/pnas.0801318105 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Jadhav S, Protchenko O, Li F, et al. Mitochondrial dysfunction in mouse livers depleted of iron chaperone PCBP1. Free Radic Biol Med. 2021;175:18‐27. doi: 10.1016/j.freeradbiomed.2021.08.232 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Toyokuni S, Kong Y, Zheng H, Maeda Y, Motooka Y, Akatsuka S. Iron as spirit of life to share under monopoly. J Clin Biochem Nutr. 2022;71:78‐88. doi: 10.3164/jcbn.22-43 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Duck KA, Neely EB, Simpson IA, Connor JR. A role for sex and a common HFE gene variant in brain iron uptake. J Cereb Blood Flow Metab. 2018;38:540‐548. doi: 10.1177/0271678x17701949 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. You L, Yu PP, Dong T, et al. Astrocyte‐derived hepcidin controls iron traffic at the blood‐brain‐barrier via regulating ferroportin 1 of microvascular endothelial cells. Cell Death Dis. 2022;13:667. doi: 10.1038/s41419-022-05043-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Tripathi AK, Karmakar S, Asthana A, et al. Transport of non‐transferrin bound iron to the brain: implications for Alzheimer's disease. J Alzheimers Dis. 2017;58:1109‐1119. doi: 10.3233/jad-170097 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Reinert A, Reinert T, Arendt T, Morawski M. High iron and iron household protein contents in perineuronal net‐Ensheathed neurons ensure energy metabolism with safe iron handling. Int J Mol Sci. 2022;23:1‐3. doi: 10.3390/ijms23031634 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Hou W, Xie Y, Song X, et al. Autophagy promotes ferroptosis by degradation of ferritin. Autophagy. 2016;12:1425‐1428. doi: 10.1080/15548627.2016.1187366 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Bogdan AR, Miyazawa M, Hashimoto K, Tsuji Y. Regulators of iron homeostasis: new players in metabolism, cell death, and disease. Trends Biochem Sci. 2016;41:274‐286. doi: 10.1016/j.tibs.2015.11.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Wolff NA, Garrick MD, Zhao L, Garrick LM, Ghio AJ, Thévenod F. A role for divalent metal transporter (DMT1) in mitochondrial uptake of iron and manganese. Sci Rep. 2018;8:211. doi: 10.1038/s41598-017-18584-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Shaw GC, Cope JJ, Li L, et al. Mitoferrin is essential for erythroid iron assimilation. Nature. 2006;440:96‐100. doi: 10.1038/nature04512 [DOI] [PubMed] [Google Scholar]
  • 49. Ali MY, Griguer CE, Flor S, Oliva CR. Mitoferrin‐1 promotes proliferation and abrogates protein oxidation via the glutathione pathway in glioblastoma. Antioxidants (Basel, Switzerland). 2023;12:1‐2. doi: 10.3390/antiox12020349 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Seguin A, Jia X, Earl AM, et al. The mitochondrial metal transporters mitoferrin1 and mitoferrin2 are required for liver regeneration and cell proliferation in mice. J Biol Chem. 2020;295:11002‐11020. doi: 10.1074/jbc.RA120.013229 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Muckenthaler MU, Rivella S, Hentze MW, Galy B. A red carpet for iron metabolism. Cell. 2017;168:344‐361. doi: 10.1016/j.cell.2016.12.034 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Zhou ZD, Tan EK. Iron regulatory protein (IRP)‐iron responsive element (IRE) signaling pathway in human neurodegenerative diseases. Mol Neurodegener. 2017;12:75. doi: 10.1186/s13024-017-0218-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Piccinelli P, Samuelsson T. Evolution of the iron‐responsive element. RNA. 2007;13:952‐966. doi: 10.1261/rna.464807 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Yoshinaga M, Nakatsuka Y, Vandenbon A, et al. Regnase‐1 maintains iron homeostasis via the degradation of transferrin receptor 1 and prolyl‐hydroxylase‐domain‐containing protein 3 mRNAs. Cell Rep. 2017;19:1614‐1630. doi: 10.1016/j.celrep.2017.05.009 [DOI] [PubMed] [Google Scholar]
  • 55. Mi Y, Wei C, Sun L, et al. Melatonin inhibits ferroptosis and delays age‐related cataract by regulating SIRT6/p‐Nrf2/GPX4 and SIRT6/NCOA4/FTH1 pathways. Biomed Pharmacother. 2023;157:114048. doi: 10.1016/j.biopha.2022.114048 [DOI] [PubMed] [Google Scholar]
  • 56. Zhao H, Lu Y, Zhang J, et al. NCOA4 requires a [3Fe‐4S] to sense and maintain the iron homeostasis. J Biol Chem. 2023;300:105612. doi: 10.1016/j.jbc.2023.105612 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Anandhan A, Dodson M, Schmidlin CJ, Liu P, Zhang DD. Breakdown of an ironclad defense system: the critical role of NRF2 in mediating ferroptosis. Cell Chemical Biology. 2020;27:436‐447. doi: 10.1016/j.chembiol.2020.03.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Cuadrado A, Rojo AI, Wells G, et al. Therapeutic targeting of the NRF2 and KEAP1 partnership in chronic diseases. Nat Rev Drug Discov. 2019;18:295‐317. doi: 10.1038/s41573-018-0008-x [DOI] [PubMed] [Google Scholar]
  • 59. Liu Z, Lv X, Song E, Song Y. Fostered Nrf2 expression antagonizes iron overload and glutathione depletion to promote resistance of neuron‐like cells to ferroptosis. Toxicol Appl Pharmacol. 2020;407:115241. doi: 10.1016/j.taap.2020.115241 [DOI] [PubMed] [Google Scholar]
  • 60. Lin D, Zhang M, Luo C, Wei P, Cui K, Chen Z. Targeting ferroptosis attenuates inflammation, fibrosis, and mast cell activation in chronic prostatitis. J Immunol Res. 2022;2022:6833867. doi: 10.1155/2022/6833867 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Cairo G, Tacchini L, Pogliaghi G, Anzon E, Tomasi A, Bernelli‐Zazzera A. Induction of ferritin synthesis by oxidative stress. Transcriptional and post‐transcriptional regulation by expansion of the “free” iron pool. J Biol Chem. 1995;270:700‐703. doi: 10.1074/jbc.270.2.700 [DOI] [PubMed] [Google Scholar]
  • 62. Donovan A, Lima CA, Pinkus JL, et al. The iron exporter ferroportin/Slc40a1 is essential for iron homeostasis. Cell Metab. 2005;1:191‐200. doi: 10.1016/j.cmet.2005.01.003 [DOI] [PubMed] [Google Scholar]
  • 63. Kerins MJ, Ooi A. The roles of NRF2 in modulating cellular iron homeostasis. Antioxid Redox Signal. 2018;29:1756‐1773. doi: 10.1089/ars.2017.7176 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Loboda A, Damulewicz M, Pyza E, Jozkowicz A, Dulak J. Role of Nrf2/HO‐1 system in development, oxidative stress response and diseases: an evolutionarily conserved mechanism. Cell Mol Life Sci. 2016;73:3221‐3247. doi: 10.1007/s00018-016-2223-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Gassmann M, Muckenthaler MU. Adaptation of iron requirement to hypoxic conditions at high altitude. J Appl Physiol (1985). 2015;119:1432‐1440. doi: 10.1152/japplphysiol.00248.2015 [DOI] [PubMed] [Google Scholar]
  • 66. Haase VH. HIF‐prolyl hydroxylases as therapeutic targets in erythropoiesis and iron metabolism. Hemodial Int. 2017;21(Suppl 1):S110‐S124. doi: 10.1111/hdi.12567 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Zhang J, Qin Y, Martinez M, et al. HIF‐1α and HIF‐2α redundantly promote retinal neovascularization in patients with ischemic retinal disease. J Clin Invest. 2021;131:e139202. doi: 10.1172/jci139202 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Jaśkiewicz M, Moszyńska A, Króliczewski J, et al. The transition from HIF‐1 to HIF‐2 during prolonged hypoxia results from reactivation of PHDs and HIF1A mRNA instability. Cell Mol Biol Lett. 2022;27:109. doi: 10.1186/s11658-022-00408-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Dunn LL, Kong SMY, Tumanov S, et al. Hmox1 (Heme Oxygenase‐1) protects against ischemia‐mediated injury via stabilization of HIF‐1α (hypoxia‐inducible factor‐1α). Arterioscler Thromb Vasc Biol. 2021;41:317‐330. doi: 10.1161/atvbaha.120.315393 [DOI] [PubMed] [Google Scholar]
  • 70. Bianchi L, Tacchini L, Cairo G. HIF‐1‐mediated activation of transferrin receptor gene transcription by iron chelation. Nucleic Acids Res. 1999;27:4223‐4227. doi: 10.1093/nar/27.21.4223 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Tacchini L, Fusar Poli D, Bernelli‐Zazzera A, Cairo G. Transferrin receptor gene expression and transferrin‐bound iron uptake are increased during postischemic rat liver reperfusion. Hepatology. 2002;36:103‐111. doi: 10.1053/jhep.2002.33997 [DOI] [PubMed] [Google Scholar]
  • 72. Li M, Tang X, Liao Z, et al. Hypoxia and low temperature upregulate transferrin to induce hypercoagulability at high altitude. Blood. 2022;140:2063‐2075. doi: 10.1182/blood.2022016410 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Anderson ER, Taylor M, Xue X, et al. Intestinal HIF2α promotes tissue‐iron accumulation in disorders of iron overload with anemia. Proc Natl Acad Sci USA. 2013;110:E4922‐E4930. doi: 10.1073/pnas.1314197110 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Nicolas G, Bennoun M, Devaux I, et al. Lack of hepcidin gene expression and severe tissue iron overload in upstream stimulatory factor 2 (USF2) knockout mice. Proc Natl Acad Sci USA. 2001;98:8780‐8785. doi: 10.1073/pnas.151179498 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Sagar P, Angmo S, Sandhir R, Rishi V, Yadav H, Singhal NK. Effect of hepcidin antagonists on anemia during inflammatory disorders. Pharmacol Ther. 2021;226:107877. doi: 10.1016/j.pharmthera.2021.107877 [DOI] [PubMed] [Google Scholar]
  • 76. Darshan D, Frazer DM, Anderson GJ. Molecular basis of iron‐loading disorders. Expert Rev Mol Med. 2010;12:e36. doi: 10.1017/s1462399410001687 [DOI] [PubMed] [Google Scholar]
  • 77. Rochette L, Gudjoncik A, Guenancia C, Zeller M, Cottin Y, Vergely C. The iron‐regulatory hormone hepcidin: a possible therapeutic target? Pharmacol Ther. 2015;146:35‐52. doi: 10.1016/j.pharmthera.2014.09.004 [DOI] [PubMed] [Google Scholar]
  • 78. Galy B, Conrad M, Muckenthaler M. Mechanisms controlling cellular and systemic iron homeostasis. Nat Rev Mol Cell Biol. 2023;25:133‐155. doi: 10.1038/s41580-023-00648-1 [DOI] [PubMed] [Google Scholar]
  • 79. Jiang L, Wang J, Wang K, et al. RNF217 regulates iron homeostasis through its E3 ubiquitin ligase activity by modulating ferroportin degradation. Blood. 2021;138:689‐705. doi: 10.1182/blood.2020008986 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Liang D, Minikes AM, Jiang X. Ferroptosis at the intersection of lipid metabolism and cellular signaling. Mol Cell. 2022;82:2215‐2227. doi: 10.1016/j.molcel.2022.03.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Maiorino M, Conrad M, Ursini F. GPx4, lipid peroxidation, and cell death: discoveries, rediscoveries, and open issues. Antioxid Redox Signal. 2018;29:61‐74. doi: 10.1089/ars.2017.7115 [DOI] [PubMed] [Google Scholar]
  • 82. Hadian K, Stockwell BR. SnapShot: ferroptosis. Cell. 2020;181:1188.e1181. doi: 10.1016/j.cell.2020.04.039 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Zhang HL, Hu BX, Li ZL, et al. PKCβII phosphorylates ACSL4 to amplify lipid peroxidation to induce ferroptosis. Nat Cell Biol. 2022;24:88‐98. doi: 10.1038/s41556-021-00818-3 [DOI] [PubMed] [Google Scholar]
  • 84. Li C, Zhang Y, Liu J, Kang R, Klionsky DJ, Tang D. Mitochondrial DNA stress triggers autophagy‐dependent ferroptotic death. Autophagy. 2021;17:948‐960. doi: 10.1080/15548627.2020.1739447 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Iqbal T, Das D. Biochemical investigation of membrane‐bound cytochrome b5 and the catalytic domain of cytochrome b5 reductase from Arabidopsis thaliana . Biochemistry. 2022;61:909‐921. doi: 10.1021/acs.biochem.2c00002 [DOI] [PubMed] [Google Scholar]
  • 86. Vázquez M, Alegret M, López MC, et al. Different effects of fibrates on the microsomal fatty acid chain elongation and the acyl composition of phospholipids in Guinea‐pigs. Br J Pharmacol. 1995;116:3337‐3343. doi: 10.1111/j.1476-5381.1995.tb15144.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Dey A, Parmar D, Dhawan A, Dash D, Seth PK. Cytochrome P450 2E1 dependent catalytic activity and lipid peroxidation in rat blood lymphocytes. Life Sci. 2002;71:2509‐2519. doi: 10.1016/s0024-3205(02)02084-2 [DOI] [PubMed] [Google Scholar]
  • 88. Rezende F, Prior KK, Löwe O, et al. Cytochrome P450 enzymes but not NADPH oxidases are the source of the NADPH‐dependent lucigenin chemiluminescence in membrane assays. Free Radic Biol Med. 2017;102:57‐66. doi: 10.1016/j.freeradbiomed.2016.11.019 [DOI] [PubMed] [Google Scholar]
  • 89. Yan B, Ai Y, Sun Q, et al. Membrane damage during ferroptosis is caused by oxidation of phospholipids catalyzed by the oxidoreductases POR and CYB5R1. Mol Cell. 2021;81:355‐369.e310. doi: 10.1016/j.molcel.2020.11.024 [DOI] [PubMed] [Google Scholar]
  • 90. Wang Y, Wan R, Peng W, Zhao X, Bai W, Hu C. Quercetin alleviates ferroptosis accompanied by reducing M1 macrophage polarization during neutrophilic airway inflammation. Eur J Pharmacol. 2023;938:175407. doi: 10.1016/j.ejphar.2022.175407 [DOI] [PubMed] [Google Scholar]
  • 91. Zhang Y, Wu J, Jiang L, Lu C, Huang Z, Liu B. Prospects for the role of ferroptosis in fluorosis. Front Physiol. 2021;12:773055. doi: 10.3389/fphys.2021.773055 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Chen Y, Wang S, Ma Q, et al. Utilizing endosomal capture for tumor therapy via membrane‐lytic mechanism‐based Pickering emulsion. J Control Release. 2023;354:523‐537. doi: 10.1016/j.jconrel.2023.01.035 [DOI] [PubMed] [Google Scholar]
  • 93. Gan B. Mitochondrial regulation of ferroptosis. J Cell Biol. 2021;220:1‐10. doi: 10.1083/jcb.202105043 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Gao M, Yi J, Zhu J, et al. Role of mitochondria in ferroptosis. Mol Cell. 2019;73:354‐363.e353. doi: 10.1016/j.molcel.2018.10.042 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Vasan K, Werner M, Chandel NS. Mitochondrial metabolism as a target for cancer therapy. Cell Metab. 2020;32:341‐352. doi: 10.1016/j.cmet.2020.06.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Li C, Dong X, du W, et al. LKB1‐AMPK axis negatively regulates ferroptosis by inhibiting fatty acid synthesis. Signal Transduct Target Ther. 2020;5:187. doi: 10.1038/s41392-020-00297-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Kang W, Suzuki M, Saito T, Miyado K. Emerging role of TCA cycle‐related enzymes in human diseases. Int J Mol Sci. 2021;22:1‐2. doi: 10.3390/ijms222313057 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Gao M, Monian P, Quadri N, Ramasamy R, Jiang X. Glutaminolysis and transferrin regulate ferroptosis. Mol Cell. 2015;59:298‐308. doi: 10.1016/j.molcel.2015.06.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Shin D, Lee J, You JH, Kim D, Roh JL. Dihydrolipoamide dehydrogenase regulates cystine deprivation‐induced ferroptosis in head and neck cancer. Redox Biol. 2020;30:101418. doi: 10.1016/j.redox.2019.101418 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Li K, Lin C, Li M, et al. Multienzyme‐like reactivity cooperatively impairs glutathione peroxidase 4 and ferroptosis suppressor protein 1 pathways in triple‐negative breast cancer for sensitized ferroptosis therapy. ACS Nano. 2022;16:2381‐2398. doi: 10.1021/acsnano.1c08664 [DOI] [PubMed] [Google Scholar]
  • 101. Zhu L, Cao P, Yang S, Lin F, Wang J. Prolonged exposure to environmental levels of microcystin‐LR triggers ferroptosis in brain via the activation of Erk/MAPK signaling pathway. Ecotoxicol Environ Saf. 2023;267:115651. doi: 10.1016/j.ecoenv.2023.115651 [DOI] [PubMed] [Google Scholar]
  • 102. Han C, Kim MJ, Ding D, et al. GSR is not essential for the maintenance of antioxidant defenses in mouse cochlea: possible role of the thioredoxin system as a functional backup for GSR. PLoS One. 2017;12:e0180817. doi: 10.1371/journal.pone.0180817 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Xu FL, Wu XH, Chen C, et al. SLC27A5 promotes sorafenib‐induced ferroptosis in hepatocellular carcinoma by downregulating glutathione reductase. Cell Death Dis. 2023;14:22. doi: 10.1038/s41419-023-05558-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Friedmann Angeli JP, Schneider M, Proneth B, et al. Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nat Cell Biol. 2014;16:1180‐1191. doi: 10.1038/ncb3064 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Cui C, Yang F, Li Q. Post‐translational modification of GPX4 is a promising target for treating ferroptosis‐related diseases. Front Mol Biosci. 2022;9:901565. doi: 10.3389/fmolb.2022.901565 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Schneider M, Forster H, Boersma A, et al. Mitochondrial glutathione peroxidase 4 disruption causes male infertility. FASEB J. 2009;23:3233‐3242. doi: 10.1096/fj.09-132795 [DOI] [PubMed] [Google Scholar]
  • 107. Ding Y, Chen X, Liu C, et al. Identification of a small molecule as inducer of ferroptosis and apoptosis through ubiquitination of GPX4 in triple negative breast cancer cells. J Hematol Oncol. 2021;14:19. doi: 10.1186/s13045-020-01016-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Koppula P, Zhuang L, Gan B. Cystine transporter SLC7A11/xCT in cancer: ferroptosis, nutrient dependency, and cancer therapy. Protein Cell. 2021;12:599‐620. doi: 10.1007/s13238-020-00789-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Koppula P, Zhang Y, Zhuang L, Gan B. Amino acid transporter SLC7A11/xCT at the crossroads of regulating redox homeostasis and nutrient dependency of cancer. Cancer Commun (London, England). 2018;38:12. doi: 10.1186/s40880-018-0288-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Fang X, Cai Z, Wang H, et al. Loss of cardiac ferritin H facilitates cardiomyopathy via Slc7a11‐mediated ferroptosis. Circ Res. 2020;127:486‐501. doi: 10.1161/circresaha.120.316509 [DOI] [PubMed] [Google Scholar]
  • 111. Zilka O, Shah R, Li B, et al. On the mechanism of cytoprotection by Ferrostatin‐1 and Liproxstatin‐1 and the role of lipid peroxidation in ferroptotic cell death. ACS Cent Sci. 2017;3:232‐243. doi: 10.1021/acscentsci.7b00028 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Shah R, Shchepinov MS, Pratt DA. Resolving the role of lipoxygenases in the initiation and execution of ferroptosis. ACS Cent Sci. 2018;4:387‐396. doi: 10.1021/acscentsci.7b00589 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Doll S, Freitas FP, Shah R, et al. FSP1 is a glutathione‐independent ferroptosis suppressor. Nature. 2019;575:693‐698. doi: 10.1038/s41586-019-1707-0 [DOI] [PubMed] [Google Scholar]
  • 114. Koppula P, Lei G, Zhang Y, et al. A targetable CoQ‐FSP1 axis drives ferroptosis‐ and radiation‐resistance in KEAP1 inactive lung cancers. Nat Commun. 2022;13:2206. doi: 10.1038/s41467-022-29905-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Stefely JA, Pagliarini DJ. Biochemistry of mitochondrial coenzyme Q biosynthesis. Trends Biochem Sci. 2017;42:824‐843. doi: 10.1016/j.tibs.2017.06.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Mao C, Liu X, Zhang Y, et al. DHODH‐mediated ferroptosis defence is a targetable vulnerability in cancer. Nature. 2021;593:586‐590. doi: 10.1038/s41586-021-03539-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Amos A, Amos A, Wu L, Xia H. The Warburg effect modulates DHODH role in ferroptosis: a review. Cell Commun Signal. 2023;21:100. doi: 10.1186/s12964-022-01025-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Kraft VAN, Bezjian CT, Pfeiffer S, et al. GTP Cyclohydrolase 1/tetrahydrobiopterin counteract ferroptosis through lipid remodeling. ACS Cent Sci. 2020;6:41‐53. doi: 10.1021/acscentsci.9b01063 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Eichwald T, da Silva LB, Staats Pires AC, et al. Tetrahydrobiopterin: beyond its traditional role as a cofactor. Antioxidants (Basel, Switzerland). 2023;12:1037. doi: 10.3390/antiox12051037 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Fanet H, Capuron L, Castanon N, Calon F, Vancassel S. Tetrahydrobioterin (BH4) pathway: from metabolism to neuropsychiatry. Curr Neuropharmacol. 2021;19:591‐609. doi: 10.2174/1570159x18666200729103529 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Cronin SJF, Seehus C, Weidinger A, et al. The metabolite BH4 controls T cell proliferation in autoimmunity and cancer. Nature. 2018;563:564‐568. doi: 10.1038/s41586-018-0701-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Zhao Q, Zheng K, Ma C, et al. PTPS facilitates compartmentalized LTBP1 S‐Nitrosylation and promotes tumor growth under hypoxia. Mol Cell. 2020;77:95‐107.e105. doi: 10.1016/j.molcel.2019.09.018 [DOI] [PubMed] [Google Scholar]
  • 123. Soula M, Weber RA, Zilka O, et al. Metabolic determinants of cancer cell sensitivity to canonical ferroptosis inducers. Nat Chem Biol. 2020;16:1351‐1360. doi: 10.1038/s41589-020-0613-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Liang D, Shu R, Jiang S, et al. Expression of BmDHFR is up‐regulated to trigger an increase in the BH4/BH2 ratio when the de novo synthesis of BH4 is blocked in silkworm, Bombyx mori . Int J Biol Macromol. 2023;225:625‐633. doi: 10.1016/j.ijbiomac.2022.11.124 [DOI] [PubMed] [Google Scholar]
  • 125. Wang Q, Xu S, Wang B, et al. Chemokine receptor 7 mediates miRNA‐182 to regulate cerebral ischemia/reperfusion injury in rats. CNS Neurosci Ther. 2023;29:712‐726. doi: 10.1111/cns.14056 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Simats A, Liesz A. Systemic inflammation after stroke: implications for post‐stroke comorbidities. EMBO Mol Med. 2022;14:e16269. doi: 10.15252/emmm.202216269 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Zhu T, Wang L, Wang LP, Wan Q. Therapeutic targets of neuroprotection and neurorestoration in ischemic stroke: applications for natural compounds from medicinal herbs. Biomed Pharmacother. 2022;148:112719. doi: 10.1016/j.biopha.2022.112719 [DOI] [PubMed] [Google Scholar]
  • 128. Cui Y, Zhang Y, Zhao X, et al. ACSL4 exacerbates ischemic stroke by promoting ferroptosis‐induced brain injury and neuroinflammation. Brain Behav Immun. 2021;93:312‐321. doi: 10.1016/j.bbi.2021.01.003 [DOI] [PubMed] [Google Scholar]
  • 129. Zheng D, Liu J, Piao H, Zhu Z, Wei R, Liu K. ROS‐triggered endothelial cell death mechanisms: focus on pyroptosis, parthanatos, and ferroptosis. Front Immunol. 2022;13:1039241. doi: 10.3389/fimmu.2022.1039241 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Tang LJ, Luo XJ, Tu H, et al. Ferroptosis occurs in phase of reperfusion but not ischemia in rat heart following ischemia or ischemia/reperfusion. Naunyn Schmiedeberg's Arch Pharmacol. 2021;394:401‐410. doi: 10.1007/s00210-020-01932-z [DOI] [PubMed] [Google Scholar]
  • 131. Gill D, Monori G, Tzoulaki I, Dehghan A. Iron status and risk of stroke. Stroke. 2018;49:2815‐2821. doi: 10.1161/strokeaha.118.022701 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. She R, Liu D, Liao J, Wang G, Ge J, Mei Z. Mitochondrial dysfunctions induce PANoptosis and ferroptosis in cerebral ischemia/reperfusion injury: from pathology to therapeutic potential. Front Cell Neurosci. 2023;17:1191629. doi: 10.3389/fncel.2023.1191629 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Lan B, Ge JW, Cheng SW, et al. Extract of Naotaifang, a compound Chinese herbal medicine, protects neuron ferroptosis induced by acute cerebral ischemia in rats. J Integr Med. 2020;18:344‐350. doi: 10.1016/j.joim.2020.01.008 [DOI] [PubMed] [Google Scholar]
  • 134. Cojocaru IM, Cojocaru M, Sapira V, Ionescu A. Evaluation of oxidative stress in patients with acute ischemic stroke. Rom J Intern Med. 2013;51:97‐106. [PubMed] [Google Scholar]
  • 135. Tang X, Fang M, Cheng R, et al. Iron‐deficiency and estrogen are associated with ischemic stroke by up‐regulating transferrin to induce hypercoagulability. Circ Res. 2020;127:651‐663. doi: 10.1161/circresaha.119.316453 [DOI] [PubMed] [Google Scholar]
  • 136. Xu SY, Ni SM, Zeng CL, Peng YJ. Role of ferroptosis in glial cells after ischemic stroke. Front Biosci (Landmark Edition). 2023;28:208. doi: 10.31083/j.fbl2809208 [DOI] [PubMed] [Google Scholar]
  • 137. Lipscomb DC, Gorman LG, Traystman RJ, Hurn PD. Low molecular weight iron in cerebral ischemic acidosis in vivo. Stroke. 1998;29:487‐492. doi: 10.1161/01.str.29.2.487 [DOI] [PubMed] [Google Scholar]
  • 138. McCarthy RC, Sosa JC, Gardeck AM, Baez AS, Lee CH, Wessling‐Resnick M. Inflammation‐induced iron transport and metabolism by brain microglia. J Biol Chem. 2018;293:7853‐7863. doi: 10.1074/jbc.RA118.001949 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Palmer C, Menzies SL, Roberts RL, Pavlick G, Connor JR. Changes in iron histochemistry after hypoxic‐ischemic brain injury in the neonatal rat. J Neurosci Res. 1999;56:60‐71. [DOI] [PubMed] [Google Scholar]
  • 140. Dávalos A, Fernandez‐Real JM, Ricart W, et al. Iron‐related damage in acute ischemic stroke. Stroke. 1994;25:1543‐1546. doi: 10.1161/01.str.25.8.1543 [DOI] [PubMed] [Google Scholar]
  • 141. Dávalos A, Castillo J, Marrugat J, et al. Body iron stores and early neurologic deterioration in acute cerebral infarction. Neurology. 2000;54:1568‐1574. doi: 10.1212/wnl.54.8.1568 [DOI] [PubMed] [Google Scholar]
  • 142. Chen X, Jiang H. Tau as a potential therapeutic target for ischemic stroke. Aging (Albany NY). 2019;11:12827‐12843. doi: 10.18632/aging.102547 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. McCarthy RC, Park YH, Kosman DJ. sAPP modulates iron efflux from brain microvascular endothelial cells by stabilizing the ferrous iron exporter ferroportin. EMBO Rep. 2014;15:809‐815. doi: 10.15252/embr.201338064 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Petrova J, Manolov V, Vasilev V, Tzatchev K, Marinov B. Ischemic stroke, inflammation, iron overload – connection to a hepcidin. Int J Stroke. 2016;11:Np16‐17. doi: 10.1177/1747493015607509 [DOI] [PubMed] [Google Scholar]
  • 145. Millán M, DeGregorio‐Rocasolano N, Pérez de la Ossa N, et al. Targeting pro‐oxidant iron with deferoxamine as a treatment for ischemic stroke: safety and optimal dose selection in a randomized clinical trial. Antioxidants (Basel, Switzerland). 2021;10:1270. doi: 10.3390/antiox10081270 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Kosyakovsky J, Fine JM, Frey WH 2nd, Hanson LR. Mechanisms of intranasal deferoxamine in neurodegenerative and neurovascular disease. Pharmaceuticals (Basel). 2021;14:95. doi: 10.3390/ph14020095 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Barón‐Mendoza I, González‐Arenas A. Relationship between the effect of polyunsaturated fatty acids (PUFAs) on brain plasticity and the improvement on cognition and behavior in individuals with autism spectrum disorder. Nutr Neurosci. 2022;25:387‐410. doi: 10.1080/1028415x.2020.1755793 [DOI] [PubMed] [Google Scholar]
  • 148. Wang Y, Wu S, Li Q, Sun H, Wang H. Pharmacological inhibition of ferroptosis as a therapeutic target for neurodegenerative diseases and strokes. Adv Sci (Weinheim, Baden‐Wurttemberg, Germany). 2023;10:e2300325. doi: 10.1002/advs.202300325 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149. Ye H, Ma Z, Liu L, et al. Thrombus inhibition and neuroprotection for ischemic stroke treatment through platelet regulation and ROS scavenging. ChemMedChem. 2022;17:e202200317. doi: 10.1002/cmdc.202200317 [DOI] [PubMed] [Google Scholar]
  • 150. Kong J, Zou R, Chu R, et al. An ultrasmall Cu/Cu(2)O nanoparticle‐based diselenide‐bridged nanoplatform mediating reactive oxygen species scavenging and neuronal membrane enhancement for targeted therapy of ischemic stroke. ACS Nano. 2023;18:4140‐4158. doi: 10.1021/acsnano.3c08734 [DOI] [PubMed] [Google Scholar]
  • 151. Yan W, Sun W, Fan J, et al. Sirt1‐ROS‐TRAF6 signaling‐induced pyroptosis contributes to early injury in ischemic mice. Neurosci Bull. 2020;36:845‐859. doi: 10.1007/s12264-020-00489-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Xu S, Tuo QZ, Meng J, Wu XL, Li CL, Lei P. Thrombin induces ferroptosis in triple‐negative breast cancer through the cPLA2α/ACSL4 signaling pathway. Transl Oncol. 2024;39:101817. doi: 10.1016/j.tranon.2023.101817 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Murakami M, Kudo I. Phospholipase A2. J Biochem. 2002;131:285‐292. doi: 10.1093/oxfordjournals.jbchem.a003101 [DOI] [PubMed] [Google Scholar]
  • 154. Li M, Meng Z, Yu S, et al. Baicalein ameliorates cerebral ischemia‐reperfusion injury by inhibiting ferroptosis via regulating GPX4/ACSL4/ACSL3 axis. Chem Biol Interact. 2022;366:110137. doi: 10.1016/j.cbi.2022.110137 [DOI] [PubMed] [Google Scholar]
  • 155. Singh NK, Rao GN. Emerging role of 12/15‐lipoxygenase (ALOX15) in human pathologies. Prog Lipid Res. 2019;73:28‐45. doi: 10.1016/j.plipres.2018.11.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. Doll S, Proneth B, Tyurina YY, et al. ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat Chem Biol. 2017;13:91‐98. doi: 10.1038/nchembio.2239 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Tuo QZ, Liu Y, Xiang Z, et al. Thrombin induces ACSL4‐dependent ferroptosis during cerebral ischemia/reperfusion. Signal Transduct Target Ther. 2022;7:59. doi: 10.1038/s41392-022-00917-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Chen J, Yang L, Geng L, et al. Inhibition of acyl‐CoA synthetase long‐chain family member 4 facilitates neurological recovery after stroke by regulation ferroptosis. Front Cell Neurosci. 2021;15:632354. doi: 10.3389/fncel.2021.632354 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. Liu H, Zhao Z, Yan M, Zhang Q, Jiang T, Xue J. Calycosin decreases cerebral ischemia/reperfusion injury by suppressing ACSL4‐dependent ferroptosis. Arch Biochem Biophys. 2023;734:109488. doi: 10.1016/j.abb.2022.109488 [DOI] [PubMed] [Google Scholar]
  • 160. Shintoku R, Takigawa Y, Yamada K, et al. Lipoxygenase‐mediated generation of lipid peroxides enhances ferroptosis induced by erastin and RSL3. Cancer Sci. 2017;108:2187‐2194. doi: 10.1111/cas.13380 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161. van Leyen K, Kim HY, Lee SR, Jin G, Arai K, Lo EH. Baicalein and 12/15‐lipoxygenase in the ischemic brain. Stroke. 2006;37:3014‐3018. doi: 10.1161/01.STR.0000249004.25444.a5 [DOI] [PubMed] [Google Scholar]
  • 162. Zhang Y, Wang H, Li J, et al. Peroxynitrite‐induced neuronal apoptosis is mediated by intracellular zinc release and 12‐lipoxygenase activation. J Neurosci. 2004;24:10616‐10627. doi: 10.1523/jneurosci.2469-04.2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Wang L, Zhang X, Xiong X, et al. Nrf2 regulates oxidative stress and its role in cerebral ischemic stroke. Antioxidants (Basel, Switzerland). 2022;11:2377. doi: 10.3390/antiox11122377 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164. Sun YY, Zhu HJ, Zhao RY, et al. Remote ischemic conditioning attenuates oxidative stress and inflammation via the Nrf2/HO‐1 pathway in MCAO mice. Redox Biol. 2023;66:102852. doi: 10.1016/j.redox.2023.102852 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165. Duan J, Cui J, Yang Z, et al. Neuroprotective effect of Apelin 13 on ischemic stroke by activating AMPK/GSK‐3β/Nrf2 signaling. J Neuroinflamm. 2019;16:24. doi: 10.1186/s12974-019-1406-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166. Fu C, Wu Y, Liu S, et al. Rehmannioside A improves cognitive impairment and alleviates ferroptosis via activating PI3K/AKT/Nrf2 and SLC7A11/GPX4 signaling pathway after ischemia. J Ethnopharmacol. 2022;289:115021. doi: 10.1016/j.jep.2022.115021 [DOI] [PubMed] [Google Scholar]
  • 167. Arteaga Cabeza O, Zhang Z, Smith Khoury E, et al. Neuroprotective effects of a dendrimer‐based glutamate carboxypeptidase inhibitor on superoxide dismutase transgenic mice after neonatal hypoxic‐ischemic brain injury. Neurobiol Dis. 2021;148:105201. doi: 10.1016/j.nbd.2020.105201 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Byrnes KR, Loane DJ, Faden AI. Metabotropic glutamate receptors as targets for multipotential treatment of neurological disorders. Neurotherapeutics. 2009;6:94‐107. doi: 10.1016/j.nurt.2008.10.038 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. McCaughey‐Chapman A, Connor B. Rat cortico‐striatal sagittal organotypic slice cultures as ex vivo excitotoxic striatal lesion models. Heliyon. 2022;8:e10819. doi: 10.1016/j.heliyon.2022.e10819 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170. Zhang X, Yu K, Ma L, et al. Endogenous glutamate determines ferroptosis sensitivity via ADCY10‐dependent YAP suppression in lung adenocarcinoma. Theranostics. 2021;11:5650‐5674. doi: 10.7150/thno.55482 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171. Chao N, Li ST. Synaptic and extrasynaptic glutamate signaling in ischemic stroke. Curr Med Chem. 2014;21:2043‐2064. doi: 10.2174/0929867321666131228204533 [DOI] [PubMed] [Google Scholar]
  • 172. Castillo J, Dávalos A, Naveiro J, Noya M. Neuroexcitatory amino acids and their relation to infarct size and neurological deficit in ischemic stroke. Stroke. 1996;27:1060‐1065. doi: 10.1161/01.str.27.6.1060 [DOI] [PubMed] [Google Scholar]
  • 173. Ghoddoussi F, Galloway MP, Jambekar A, Bame M, Needleman R, Brusilow WSA. Methionine sulfoximine, an inhibitor of glutamine synthetase, lowers brain glutamine and glutamate in a mouse model of ALS. J Neurol Sci. 2010;290:41‐47. doi: 10.1016/j.jns.2009.11.013 [DOI] [PubMed] [Google Scholar]
  • 174. Li SH, Abd‐Elrahman KS, Ferguson SSG. Targeting mGluR2/3 for treatment of neurodegenerative and neuropsychiatric diseases. Pharmacol Ther. 2022;239:108275. doi: 10.1016/j.pharmthera.2022.108275 [DOI] [PubMed] [Google Scholar]
  • 175. Lin CH, Chen HY, Wei KC. Role of HMGB1/TLR4 Axis in ischemia/reperfusion‐impaired extracellular glutamate clearance in primary astrocytes. Cells. 2020;9:1‐13. doi: 10.3390/cells9122585 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176. Kulik K, Żyżyńska‐Granica B, Kowalczyk A, Kurowski P, Gajewska M, Bujalska‐Zadrożny M. Magnesium and morphine in the treatment of chronic neuropathic pain‐a biomedical mechanism of action. Int J Mol Sci. 2021;22:1‐2. doi: 10.3390/ijms222413599 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Ma S, Chen M, Jiang Y, et al. Sustained antidepressant effect of ketamine through NMDAR trapping in the LHb. Nature. 2023;622:802‐809. doi: 10.1038/s41586-023-06624-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178. Song X, Jensen MØ, Jogini V, et al. Mechanism of NMDA receptor channel block by MK‐801 and memantine. Nature. 2018;556:515‐519. doi: 10.1038/s41586-018-0039-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179. Šterk M, Križančić Bombek L, Skelin Klemen M, et al. NMDA receptor inhibition increases, synchronizes, and stabilizes the collective pancreatic beta cell activity: insights through multilayer network analysis. PLoS Comput Biol. 2021;17:e1009002. doi: 10.1371/journal.pcbi.1009002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180. Adell A. Brain NMDA receptors in schizophrenia and depression. Biomol Ther. 2020;10:1. doi: 10.3390/biom10060947 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181. Hess EM, Riggs LM, Michaelides M, Gould TD. Mechanisms of ketamine and its metabolites as antidepressants. Biochem Pharmacol. 2022;197:114892. doi: 10.1016/j.bcp.2021.114892 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182. Novgorodov SA, Voltin JR, Gooz MA, Li L, Lemasters JJ, Gudz TI. Acid sphingomyelinase promotes mitochondrial dysfunction due to glutamate‐induced regulated necrosis. J Lipid Res. 2018;59:312‐329. doi: 10.1194/jlr.M080374 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183. Soria FN, Pérez‐Samartín A, Martin A, et al. Extrasynaptic glutamate release through cystine/glutamate antiporter contributes to ischemic damage. J Clin Invest. 2014;124:3645‐3655. doi: 10.1172/jci71886 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184. Domercq M, Szczupak B, Gejo J, et al. PET imaging with [(18)F]FSPG evidences the role of system xc(−) on brain inflammation following cerebral ischemia in rats. Theranostics. 2016;6:1753‐1767. doi: 10.7150/thno.15616 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185. Hsieh CH, Lin YJ, Chen WL, et al. HIF‐1α triggers long‐lasting glutamate excitotoxicity via system x(c)(−) in cerebral ischaemia‐reperfusion. J Pathol. 2017;241:337‐349. [DOI] [PubMed] [Google Scholar]
  • 186. He Y, Hewett SJ. The cystine/glutamate antiporter, system x(c) (−), contributes to cortical infarction after moderate but not severe focal cerebral ischemia in mice. Front Cell Neurosci. 2022;16:821036. doi: 10.3389/fncel.2022.821036 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187. Polewski MD, Reveron‐Thornton RF, Cherryholmes GA, Marinov GK, Cassady K, Aboody KS. Increased expression of system xc‐ in glioblastoma confers an altered metabolic state and temozolomide resistance. Mol Cancer Res. 2016;14:1229‐1242. doi: 10.1158/1541-7786.mcr-16-0028 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188. Zhang Y, Lu X, Tai B, Li W, Li T. Ferroptosis and its multifaceted roles in cerebral stroke. Front Cell Neurosci. 2021;15:615372. doi: 10.3389/fncel.2021.615372 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189. Liu Y, Min JW, Feng S, et al. Therapeutic role of a cysteine precursor, OTC, in ischemic stroke is mediated by improved Proteostasis in mice. Transl Stroke Res. 2020;11:147‐160. doi: 10.1007/s12975-019-00707-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190. Zhu H, Huang J, Chen Y, et al. Resveratrol pretreatment protects neurons from oxygen‐glucose deprivation/reoxygenation and ischemic injury through inhibiting ferroptosis. Biosci Biotechnol Biochem. 2022;86:704‐716. doi: 10.1093/bbb/zbac048 [DOI] [PubMed] [Google Scholar]
  • 191. Liu W, Wang L, Liu C, Dai Z, Li T, Tang B. Edaravone ameliorates cerebral ischemia‐reperfusion injury by downregulating ferroptosis via the Nrf2/FPN pathway in rats. Biol Pharm Bull. 2022;45:1269‐1275. doi: 10.1248/bpb.b22-00186 [DOI] [PubMed] [Google Scholar]
  • 192. Chen G, Li L, Tao H. Bioinformatics identification of ferroptosis‐related biomarkers and therapeutic compounds in ischemic stroke. Front Neurol. 2021;12:745240. doi: 10.3389/fneur.2021.745240 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193. Guo L, Shi L. Vitexin improves cerebral ischemia‐reperfusion injury by attenuating oxidative injury and ferroptosis via Keap1/Nrf2/HO‐1 signaling. Neurochem Res. 2023;48:980‐995. doi: 10.1007/s11064-022-03829-0 [DOI] [PubMed] [Google Scholar]
  • 194. Bai X, Zheng E, Tong L, et al. Angong Niuhuang Wan inhibit ferroptosis on ischemic and hemorrhagic stroke by activating PPARγ/AKT/GPX4 pathway. J Ethnopharmacol. 2024;321:117438. doi: 10.1016/j.jep.2023.117438 [DOI] [PubMed] [Google Scholar]
  • 195. Shi Y, Han L, Zhang X, Xie L, Pan P, Chen F. Selenium alleviates cerebral ischemia/reperfusion injury by regulating oxidative stress, mitochondrial fusion and ferroptosis. Neurochem Res. 2022;47:2992‐3002. doi: 10.1007/s11064-022-03643-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196. Chen Y, He W, Wei H, et al. Srs11‐92, a ferrostatin‐1 analog, improves oxidative stress and neuroinflammation via Nrf2 signal following cerebral ischemia/reperfusion injury. CNS Neurosci Ther. 2023;29:1667‐1677. doi: 10.1111/cns.14130 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197. Fei Y, Li T, Wu R, et al. Se‐(methyl)‐selenocysteine ameliorates blood‐brain barrier disruption of focal cerebral ischemia mice via ferroptosis inhibition and tight junction upregulation in an Akt/GSK3β‐dependent manner. Psychopharmacology. 2023;241:379‐399. doi: 10.1007/s00213-023-06495-4 [DOI] [PubMed] [Google Scholar]
  • 198. Gao J, Ma C, Xia D, et al. Icariside II preconditioning evokes robust neuroprotection against ischaemic stroke, by targeting Nrf2 and the OXPHOS/NF‐κB/ferroptosis pathway. Br J Pharmacol. 2023;180:308‐329. doi: 10.1111/bph.15961 [DOI] [PubMed] [Google Scholar]
  • 199. Guan X, Li Z, Zhu S, et al. Galangin attenuated cerebral ischemia‐reperfusion injury by inhibition of ferroptosis through activating the SLC7A11/GPX4 axis in gerbils. Life Sci. 2021;264:118660. doi: 10.1016/j.lfs.2020.118660 [DOI] [PubMed] [Google Scholar]
  • 200. Guan X, Li X, Yang X, et al. The neuroprotective effects of carvacrol on ischemia/reperfusion‐induced hippocampal neuronal impairment by ferroptosis mitigation. Life Sci. 2019;235:116795. doi: 10.1016/j.lfs.2019.116795 [DOI] [PubMed] [Google Scholar]
  • 201. Guo H, Zhu L, Tang P, et al. Carthamin yellow improves cerebral ischemia‐reperfusion injury by attenuating inflammation and ferroptosis in rats. Int J Mol Med. 2021;47:1‐10. doi: 10.3892/ijmm.2021.4885 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202. Hu Q, Zuo T, Deng L, et al. β‐Caryophyllene suppresses ferroptosis induced by cerebral ischemia reperfusion via activation of the NRF2/HO‐1 signaling pathway in MCAO/R rats. Phytomedicine. 2022;102:154112. doi: 10.1016/j.phymed.2022.154112 [DOI] [PubMed] [Google Scholar]
  • 203. Jin Z, Gao W, Guo F, et al. Astragaloside IV alleviates neuronal ferroptosis in ischemic stroke by regulating fat mass and obesity‐associated‐N6‐methyladenosine‐acyl‐CoA synthetase long‐chain family member 4 axis. J Neurochem. 2023;166:328‐345. doi: 10.1111/jnc.15871 [DOI] [PubMed] [Google Scholar]
  • 204. Ko G, Kim J, Jeon YJ, Lee D, Baek HM, Chang KA. Salvia miltiorrhiza alleviates memory deficit induced by ischemic brain injury in a transient MCAO mouse model by inhibiting ferroptosis. Antioxidants (Basel, Switzerland). 2023;12:1‐17. doi: 10.3390/antiox12040785 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205. Liao J, Wei M, Wang J, et al. Naotaifang formula attenuates OGD/R‐induced inflammation and ferroptosis by regulating microglial M1/M2 polarization through BMP6/SMADs signaling pathway. Biomed Pharmacother. 2023;167:115465. doi: 10.1016/j.biopha.2023.115465 [DOI] [PubMed] [Google Scholar]
  • 206. Li XN, Shang NY, Kang YY, et al. Caffeic acid alleviates cerebral ischemic injury in rats by resisting ferroptosis via Nrf2 signaling pathway. Acta Pharmacol Sin. 2023;45:248‐267. doi: 10.1038/s41401-023-01177-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207. Liu C, Liu E, Li Z, et al. Danlou tablet attenuates ischemic stroke injury and blood–brain barrier damage by inhibiting ferroptosis. J Ethnopharmacol. 2023;322:117657. doi: 10.1016/j.jep.2023.117657 [DOI] [PubMed] [Google Scholar]
  • 208. Liu H, Zhang TA, Zhang WY, Huang SR, Hu Y, Sun J. Rhein attenuates cerebral ischemia‐reperfusion injury via inhibition of ferroptosis through NRF2/SLC7A11/GPX4 pathway. Exp Neurol. 2023;369:114541. doi: 10.1016/j.expneurol.2023.114541 [DOI] [PubMed] [Google Scholar]
  • 209. Chen G, Li C, Zhang L, et al. Hydroxysafflor yellow a and anhydrosafflor yellow B alleviate ferroptosis and parthanatos in PC12 cells injured by OGD/R. Free Radic Biol Med. 2022;179:1‐10. doi: 10.1016/j.freeradbiomed.2021.12.262 [DOI] [PubMed] [Google Scholar]
  • 210. Liu X, du Y, Liu J, Cheng L, He W, Zhang W. Ferrostatin‐1 alleviates cerebral ischemia/reperfusion injury through activation of the AKT/GSK3β signaling pathway. Brain Res Bull. 2023;193:146‐157. doi: 10.1016/j.brainresbull.2022.12.009 [DOI] [PubMed] [Google Scholar]
  • 211. Hui Z, Wang S, Li J, Wang J, Zhang Z. Compound Tongluo decoction inhibits endoplasmic reticulum stress‐induced ferroptosis and promoted angiogenesis by activating the sonic hedgehog pathway in cerebral infarction. J Ethnopharmacol. 2022;283:114634. doi: 10.1016/j.jep.2021.114634 [DOI] [PubMed] [Google Scholar]
  • 212. Liu Y, Mi Y, Wang Y, et al. Loureirin C inhibits ferroptosis after cerebral ischemia reperfusion through regulation of the Nrf2 pathway in mice. Phytomedicine. 2023;113:154729. doi: 10.1016/j.phymed.2023.154729 [DOI] [PubMed] [Google Scholar]
  • 213. Peng C, Ai Q, Zhao F, et al. Quercetin attenuates cerebral ischemic injury by inhibiting ferroptosis via Nrf2/HO‐1 signaling pathway. Eur J Pharmacol. 2023;963:176264. doi: 10.1016/j.ejphar.2023.176264 [DOI] [PubMed] [Google Scholar]
  • 214. Sun M, Liu M, Li Q, et al. Cottonseed oil alleviates ischemic stroke injury by inhibiting ferroptosis. Brain Behav. 2023;13:e3179. doi: 10.1002/brb3.3179 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215. Wu C, Duan F, Yang R, Dai Y, Chen X, Li S. 15, 16‐Dihydrotanshinone I protects against ischemic stroke by inhibiting ferroptosis via the activation of nuclear factor erythroid 2‐related factor 2. Phytomedicine. 2023;114:154790. doi: 10.1016/j.phymed.2023.154790 [DOI] [PubMed] [Google Scholar]
  • 216. Xu S, Li X, Li Y, et al. Neuroprotective effect of Dl‐3‐n‐butylphthalide against ischemia‐reperfusion injury is mediated by ferroptosis regulation via the SLC7A11/GSH/GPX4 pathway and the attenuation of blood‐brain barrier disruption. Front Aging Neurosci. 2023;15:1028178. doi: 10.3389/fnagi.2023.1028178 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217. Yan N, Xu Z, Qu C, Zhang J. Dimethyl fumarate improves cognitive deficits in chronic cerebral hypoperfusion rats by alleviating inflammation, oxidative stress, and ferroptosis via NRF2/ARE/NF‐κB signal pathway. Int Immunopharmacol. 2021;98:107844. doi: 10.1016/j.intimp.2021.107844 [DOI] [PubMed] [Google Scholar]
  • 218. Yang W, Liu X, Song C, et al. 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. 2021;209:112842. doi: 10.1016/j.ejmech.2020.112842 [DOI] [PubMed] [Google Scholar]
  • 219. Yuan Y, Zhai Y, Chen J, Xu X, Wang H. Kaempferol ameliorates oxygen‐glucose deprivation/reoxygenation‐induced neuronal ferroptosis by activating Nrf2/SLC7A11/GPX4 Axis. Biomol Ther. 2021;11:1‐14. doi: 10.3390/biom11070923 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220. Zhan S, Liang J, Lin H, et al. SATB1/SLC7A11/HO‐1 Axis ameliorates ferroptosis in neuron cells after ischemic stroke by Danhong injection. Mol Neurobiol. 2023;60:413‐427. doi: 10.1007/s12035-022-03075-z [DOI] [PubMed] [Google Scholar]
  • 221. Zhang Y, Ye P, Zhu H, et al. Neutral polysaccharide from Gastrodia elata alleviates cerebral ischemia‐reperfusion injury by inhibiting ferroptosis‐mediated neuroinflammation via the NRF2/HO‐1 signaling pathway. CNS Neurosci Ther. 2023;30:e14456. doi: 10.1111/cns.14456 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222. Liang F, Wu Y, Wang X, et al. General anesthesia vs conscious sedation for endovascular treatment in patients with posterior circulation acute ischemic stroke: An exploratory randomized clinical trial. JAMA Neurol. 2023;80:64‐72. doi: 10.1001/jamaneurol.2022.3018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223. Nakamura A, Sakai S, Taketomi Y, et al. PLA2G2E‐mediated lipid metabolism triggers brain‐autonomous neural repair after ischemic stroke. Neuron. 2023;111:2995‐3010.e2999. doi: 10.1016/j.neuron.2023.06.024 [DOI] [PubMed] [Google Scholar]
  • 224. Liu J, Guo ZN, Yan XL, et al. Crosstalk between autophagy and ferroptosis and its putative role in ischemic stroke. Front Cell Neurosci. 2020;14:577403. doi: 10.3389/fncel.2020.577403 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225. Ai Y, Meng Y, Yan B, Zhou Q, Wang X. The biochemical pathways of apoptotic, necroptotic, pyroptotic, and ferroptotic cell death. Mol Cell. 2024;84:170‐179. doi: 10.1016/j.molcel.2023.11.040 [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Data sharing not applicable to this article as no datasets were generated or analysed during the current study.


Articles from CNS Neuroscience & Therapeutics are provided here courtesy of Wiley

RESOURCES