Ferroptosis is a unique form of programmed cell death pathway that is mechanistically different from necrosis, apoptosis, or autophagy.1 Ferroptosis is induced by reactive oxygen species (ROS) originating from multiple sources, especially accumulation of free intracellular divalent iron (Fe2+), leading to peroxidation of polyunsaturated fatty acids (PUFAs) and resulting in membrane damage (Figure). One major trigger of ferroptosis is decreased availability of intracellular antioxidant enzymes, particularly glutathione peroxidases (GPXs).1-7 Because of its high lipid concentration and oxygen consumption rate, the nervous tissue is extremely vulnerable to oxidative damage and ferroptosis. With aging, iron accumulates in the brain, which predisposes to and exacerbates these processes.8 Iron promotes ferroptosis both directly and as a cofactor required for the activity of enzymes mediating redox reactions and lipid peroxidation.9,10 Epigenetic regulation can determine cell sensitivity to ferroptosis by affecting intracellular iron levels, oxidative stress, and lipid metabolism.11 Ferroptosis affects all cell types in the nervous system, including neurons, glial cells, and pericytes.12 Microglia are the primary iron-accumulating cells in disease.13,14 Recent evidence indicates that iron-laden microglia have a critical role in ferroptosis associated with neurodegeneration.15 Studies in vitro, in experimental disease models, and in human brain tissue indicate that iron accumulation, oxidative stress, and lipid peroxidation are major disease mechanisms in a wide range of neurologic disorders. These studies also suggest that the different components of the ferroptosis pathway are potential targets for therapeutic intervention. The mechanisms of ferroptosis and its role in neurologic disorders have been the subject of several recent comprehensive reviews.7,8,16-21 Some of these concepts pertinent to neurodegeneration will be emphasized in this study.
Figure. Iron Metabolisms and Mechanisms of Ferroptosis.
A highly complex network including various transporters, storage, and exporter proteins control iron levels in cells. Ferric iron (Fe3+) present in biological fluids binds to transferrin, which is then recognized by the transferrin receptor 1 (TfR1) in cell membranes. This complex transports Fe3+ into the endosome where it is converted to ferrous iron (Fe2+) by the metalloreductases STEAP2. Ferrous iron is then released through the divalent metal transporter 1 (DMT1) into the labile Fe2+ iron pool in the cytoplasm, exported to brain interstitial fluid through ferroportin after oxidation to Fe3+ by ceruloplasmin, and then loaded onto transferrin. Ferroportin level is controlled by its interaction with hepcidin. When available iron exceeds the biological needs, it is sequestered by the iron storage protein ferritin. Dynamic changes in the Fe3+/Fe2+ redox potential determine the sensitivity of cells to ferroptosis. The reaction of Fe2+ and hydrogen peroxide (H2O2) through the Fenton reaction leads to the formation of hydroxyl radicals (OH−), which elicit lipid peroxidation required for the activity of metabolic enzymes used in the redox reaction. The reactive oxygen species triggering ferroptosis may also originate from the mitochondrial respiratory chain through the production of superoxide (O2*), which is then converted to H2O2 by superoxide dismutases (SODs). The membrane-associated nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) also provides reactive oxygen species. The primary substrates for lipid peroxidation in the process of ferroptosis are polyunsaturated fatty acids (PUFAs) that are incorporated into phospholipids such as phosphatidylethanolamine (PE-PUFA) by sequential action of acyl-CoA synthetase long-chain family member 4 (ACSL4) and lysophosphatidylcholine acyltransferase 3 (LPCAT3). Lipid peroxidation occurs not only through the iron-dependent Fenton reaction but also by enzymatic pathways, primarily mediated by lipoxygenases (LOXs), which use iron as a cofactor. Peroxidation of membrane lipids (PE-PUFA-OOH) affects transport along cell membranes, mitochondrial bioenergetics, vesicular traffic, and the machinery for autophagy and mitophagy. This promotes the accumulation of lipofuscin and iron primarily in lysosomes. Ferroptosis is also associated with membrane rupture and release of damage-associated molecular patterns that trigger inflammation. Toxic metabolites resulting from lipid peroxidation, such as 4-hydroxynonenal or malondialdehyde, are also cytotoxic molecules that form adducts with DNA bases, proteins, and other nucleophilic molecules. The primary protective mechanism against ferroptosis is the glutathione (GSH)–glutathione peroxidase 4 (GPx4) system. The availability of GSH in cells depends on the activity of the cystine (Cys)/glutamate (Glu) antiporter (system Xc−), which transports Cys into the cell, where it is reduced to cysteine (CysH) and then combined with glutamate and glycine (Gly) to produce GSH by the action of glutathione synthetase (GS). GPx4 converts GSH to oxidized glutathione (GSSG) and while reducing lipid peroxides to lipid alcohols. The reconstitution of GSH from GSSG is mediated by glutathione reductase (GR). In the plasma membrane, coenzyme Q10 (CoQ10) blocks lipid peroxidation and ferroptosis through an NADPH-mediated action mediated by the oxidoreductase ferroptosis inhibitory protein 1 (FSP1). Nuclear factor erythroid 2–related factor 2 (NRF2) improves cellular tolerance to oxidative stress, lipid peroxidation, and ferroptosis by promoting transcription of proteins related to iron transport and metabolism, NADPH, and GSH metabolism. The calcium-independent phospholipase A2 type G6 (iPLA2G6) degrades hydroxy peroxy-phosphatidylethanolamine also preventing ferroptosis.
Mechanisms of Ferroptosis
Overview of Iron Metabolism
A major trigger of ferroptosis is abnormal intracellular iron accumulation. Iron participates in many fundamental functions in the CNS,16,22 and its metabolism is a dynamic process involving multiple steps (Figure).16,23,24 A highly complex network including various transporters, storage, and exporter proteins control iron levels in cells. Ferric iron (Fe3+) present in biological fluids binds to transferrin, which is then recognized by the transferrin receptor 1 (TfR1) in cell membranes including those forming the blood-brain barrier (BBB); this complex transports Fe3+ into the endosome.25 In the acid milieu of endosomes, Fe3+ undergoes reduction to Fe2+ by metalloreductases, and Fe2+ is then released through the divalent metal transporter 1 (DMT1) into the labile Fe2+ iron pool in the cytoplasm.26 Trafficking from the endoplasmic reticulum to the Golgi apparatus may also contribute to the labile iron pool.27 Ferrous iron is oxidized to Fe3+ by ceruloplasmin, exported to the brain interstitial fluid through ferroportin, and then loaded onto transferrin.24,28 Ferric iron bound to transferrin is taken up by neurons and glial cells through TfR1-mediated endocytosis. Ferroportin is a major regulator of intracellular iron levels; in the presence of iron, ferroportin binds to hepcidin, forming a complex that is internalized and degraded in lysosomes.29 When available iron exceeds the biological needs, it is sequestered by the iron-storage protein ferritin. Ferritin is the main iron buffering and storage protein and is found in glial cells but generally not in neurons.30 Catecholaminergic neurons can buffer free iron accumulation through the formation of neuromelanin from oxidized dopamine or norepinephrine metabolites.31 In response to change in cellular levels, iron homeostasis is regulated mainly posttranscriptionally by iron regulatory proteins; these proteins bind to specific messenger RNA (mRNA) sequences called iron-responsive elements in untranslated regions of mRNAs encoding TfR1, DMT1, ferritin, and other iron export, storage, and import proteins.32
Role of Iron and Oxidative Stress in Ferroptosis
Dynamic changes in the Fe3+/Fe2+ redox potential mediate several fundamental cell processes such as mitochondrial respiration and have a major role in determining the sensitivity of cells to ferroptosis.16 The reaction of Fe2+ and hydrogen peroxide through the Fenton reaction leads to the formation of hydroxides and hydroxyl radicals, which elicits lipid peroxidation.23,33 Iron is also a cofactor required for the activity of metabolic enzymes used in redox reactions and lipid peroxidation, such as lipoxygenase (LOX).9,10 In addition to Fenton reaction, the ROS triggering ferroptosis may originate from the mitochondria, which are regulatory hubs for cell death.34 Mitochondrial ROS production by the electron transport chain, fatty acid synthesis, and DNA stress all promote lipid peroxidation.35,36 Mitochondrial DNA stress also triggers autophagic-dependent ferroptotic cell death.35,37 Selective forms of autophagy, such as ferritinophagy and lipophagy, drive cells toward ferroptosis.38 For example, ROS-induced autophagy promotes the degradation of ferritin and induction of TfR1, resulting in intracellular iron accumulation.39 Mitochondrial energy sensors such as adenosine monophosphate–activated protein kinase may bidirectionally regulate ferroptosis by affecting lipid synthesis40 and autophagy.41 Another source of ROS for ferroptosis is the activity of membrane-associated nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX),5 which is primarily expressed in microglia.42,43 NOX family members, like other enzymes involved in ferroptosis, require iron for their normal function.44
Mechanisms and Consequences of Lipid Peroxidation
The primary substrates for lipid peroxidation are PUFAs, such as arachidonic acid and adrenic acid.10,45 These PUFAs are incorporated into cellular phospholipids, especially phosphatidylethanolamine, by sequential action of acyl-CoA synthetase long-chain family member 4 and lysophosphatidylcholine acyltransferase 3.10,45-47 Other sources of lipid substrates for peroxidation include polyunsaturated ether phospholipids derived from peroxisomes48 and fatty acids produced by lipophagy.49 Lipid peroxidation occurs not only through hydroxyl radicals generated during the iron-dependent Fenton reaction33 but also through enzymatic pathways primarily mediated by LOXs, which use iron as a cofactor.50 Cytochrome P450 oxidoreductase transfers electrons from NADPH to the mitochondria, which also promotes lipid peroxidation.51
Peroxidation of membrane lipids results in increased membrane rigidity and affects membrane transport, mitochondrial bioenergetics, vesicular traffic, and the machinery for autophagy and mitophagy.52-54 These processes interact with each other to promote ferroptosis. Impaired mitophagy promotes the accumulation of ROS and damaged proteins and lipids in mitochondria; release of iron by mitochondria into autolysosomes triggers further Fenton reactions, lipid peroxidation, and impaired autophagy.53,54 This leads to the accumulation of lipofuscin and iron which, in turn, increase oxidative stress and lipid peroxidation.55 Ferroptosis is a type of lytic cell death associated with membrane rupture and release of damage-associated molecular patterns that trigger inflammation.38,56 Cathepsin B, a lysosomal cysteine protease, is one of the mediators of ferroptosis, causing DNA damage after translocation from lysosomes to the nucleus.57,58 Toxic metabolites resulting from lipid peroxidation, such as 4-hydroxynonenal or malondialdehyde, are also cytotoxic molecules that form adducts with DNA bases, proteins, and other nucleophilic molecules, including proteins involved in autophagosome and autolysosome formation, thereby further decreasing autophagic flux.45,59
Defense Mechanisms Against Ferroptosis
The primary mechanism that protects cells from ferroptosis is the glutathione (GSH)-GPX4 system (Figure).1,2,7,17 In the brain, GPX4 is expressed in neurons and glial cells and has a free radical–scavenging and antioxidant function.60 The availability of GSH in cells depends on the activity of the cystine/glutamate antiporter (system Xc-), which transports cystine into the cell; cystine is reduced to cysteine and then combined with glutamate and glycine to produce GSH by the action of glutathione synthetase.61 Because neurons lack this cystine transport system, they primarily depend on GSH synthesis and release from surrounding astrocytes.62 Glutathione peroxidase 4 converts GSH to oxidized glutathione (GSSG) while reducing lipid peroxides to lipid alcohols.63 The reconstitution of GSH from GSSG is mediated by glutathione reductase using NADPH. In addition to the GSH-GPX4 system, there are other mechanisms that protect against oxidative stress and ferroptosis. In the plasma membrane, coenzyme Q10 (CoQ10) functions as a lipophilic free radical–trapping antioxidant that blocks lipid peroxidation through an NADPH-mediated action mediated by the oxidoreductase ferroptosis inhibitory protein 1.5,64 The mevalonate pathway provides substrates for the synthesis of both GPX4 and CoQ10.65 Other ferroptosis protection mechanisms include the mitochondrial dihydroorotate dehydrogenase–dihydroubiquinone system66 and the guanosine triphosphate cyclohydrolase–1–tetrahydrobiopterin pathway.67,68 The relative contribution of each antioxidant mechanism may vary in different cells and tissues.69
Nuclear factor erythroid 2–related factor 2 (NRF2) is a transcription factor that has a major role in promoting cellular tolerance to oxidative stress, lipid peroxidation, and ferroptosis.70-73 NRF2 is expressed mainly in astrocytes and promotes the transcription of proteins related to iron transport and metabolism, such as ferroportin 1 and ferritin light and heavy chains; enzymes related to NADPH regeneration such as glucose-6-phosphate dehydrogenase; and proteins and enzymes related to GSH metabolism such as subunits of the cystine/glutamate transporter protein and glutathione synthetase.62,70,71 Other mechanisms can prevent ferroptosis by affecting lipid composition and repair of cell membranes. For example, the calcium-independent phospholipase A2 type G6 (PLA2G6) degrades membrane hydroxyperoxy-phosphatidylethanolamine.74 The membrane scission machinery that depends on endosomal sorting complex required for transport-III can repair the cell membrane.75
Role of Ferroptosis in Neurologic Disease
Biochemical and transcriptomic analyses of mediators of iron metabolism, antioxidant defenses, oxidative stress, and lipid peroxidation in experimental disease models, autopsy material, and induced pluripotent stem cell (iPSCs) derived from patients indicate that ferroptosis is a major mechanism of cell death in a wide range of neurologic disorders. These include Alzheimer disease (AD),64,72,76-82 Parkinson disease (PD),55,83-87 amyotrophic lateral sclerosis (ALS),88-92 Huntington disease,93-95 Friedreich ataxia,96-98 multiple sclerosis,99-101 ischemic stroke,47,102-105 intracerebral hemorrhage,106,107 traumatic brain injury,108 spinal cord injury,109,110 epilepsy,111-113 and glioma,114 among others. This topic has been extensively reviewed,16-21,115,116 and only few concepts will be emphasized in this study.
Cerebral iron deposition is a typical finding associated with aging and neurodegeneration.8,117 Studies in experimental models and human autopsy material indicate that several interacting mechanisms may result in iron accumulation and oxidative stress in both neurons and glial cells in vulnerable areas. Mitochondrial dysfunction with excessive ROS production, autophagy disruption, lipofuscin accumulation, and lipid peroxidation are common pathomechanisms in these disorders.34,55,115,116,118,119 Indirect evidence of ferroptosis include abnormal levels of iron import, storage, and export proteins indicating iron dyshomeostasis, reduced expression of GSH and other antioxidants, upregulation of the NRF2, and accumulation of products from lipid peroxidation, such as malonyldialdehyde and 4-hydroxynonenal.16-21
A set of iron-responsive elements in 5′untranslated regions of mRNA can bind excessive iron and fold mRNA into loops that affect transcription of proteins such as amyloid precursor protein (APP), α-synuclein, and prion protein; many of these proteins interact with iron, which may initiate a potential feedback to accelerate ferroptosis.120 Excessive iron accumulation correlates with accelerated cognitive decline in AD.121 In this disorder, iron accumulates in β-amyloid (Aβ) aggregates and neurofibrillary tangles77,122,123 and promotes the aggregation of both toxic Aβ oligomers and hyperphosphorylated tau.124 Consistent with the role of ferroptosis in AD, studies in human autopsy material and experimental models show decreased GSH levels, GPX4 downregulation, and accumulation of malonyldialdehyde in brain tissue.122 Ferroptosis may be a prominent cell death mechanism in PD. Both oxidative stress due to impaired mitochondrial complex I and decreased CoQ, GSH, and GPX4 levels and iron dyshomeostasis, reflected by increased TfR1 and DMT1 and decreased ceruloplasmin and ferritin expression, promote abnormal iron accumulation and ferroptosis in both dopaminergic neurons and glial cells.28,83,125 α-Synuclein can form complexes with iron, act as a ferrireductase, and increase DMT1-mediated iron uptake in the cytosol.126 In human dopaminergic neuron models, endogenous levels of α-synuclein can affect the availability of ether-linked phospholipids required for ferroptosis.127,128 Aggregated α-synuclein also induces the production of ROS and formation of lipid peroxidation products, which in turn, promote α-synuclein aggregation, thus perpetuating the pathogenic cycle.129,130 Misfolded α-synuclein also activates microglia, which release interleukin-6 that induces changes in the neuronal transcriptome promoting iron uptake and decreases iron export.131
Glial cells also have a major role in the mechanisms of ferroptosis. Astrocytes express TfR1, store iron through DMT1, and release iron through ferroportin, thereby regulating iron distribution in neural tissues in both normal and pathologic conditions.132,133 Production of GSH by astrocytes may also have neuroprotective effects both by attenuating neuroinflammation134 and maintaining BBB stability.135 Recent evidence indicates that microglia are especially vulnerable to ferroptosis and iron accumulation in microglia has deleterious effects of neuronal viability.14,15 Single-cell RNA sequencing studies in both transgenic models and microglia derived from human iPSCs revealed that iron-laded microglia express markers of ferroptosis activation, upregulation of oxidative stress pathways, dampening of transcriptional activation of proinflammatory and anti-inflammatory mediators, and impaired phagocytosis.14,15 This iron-induced transcriptomic signature is similar to that found in microglia from postmortem tissue from patients with PD, AD, ALS, and MS.15,136,137 In transgenic mice that overexpress APP and presenilin 1 (PS1; APP/PS1 mice), stimulation of cultured microglia with interferon gamma and Aβ induced these cells to retain iron and have reduced phagocytic and chemotactic functions.138 A study using human iPSC–derived triculture containing microglia, astrocytes, and neurons showed that exposure to iron and a GPX4 inhibitor resulted in marked microglial cell death followed by neuronal cell death.15 Omission of microglia from the culture markedly reduced neuronal death.15 These studies thus show that iron accumulation in microglia induces oxidative stress, dampens inflammatory activation, and triggers neurodegeneration.14,15 Iron also accumulates in the choroid plexus and may contribute to the damage of the BBB and blood-CSF barrier.139
Some forms of neurodegeneration with brain iron accumulation (NBIA), such as hereditary ferritinopathy and aceruloplasminemia, are caused by pathogenic variants in genes coding for proteins directly involved in the iron regulatory pathway.117 However, the relationship between iron accumulation and neurodegeneration in these disorders is incompletely understood. For example, in neuroferritinopathy, abnormal ferritin light chain aggregates lead to impaired proteostasis while derangement of iron metabolism may occur as a secondary event. Other disorders in the NBIA group are caused by variants in genes not directly related to iron metabolism. Many of these disorders share features such as mitochondrial dysfunction, oxidative stress, altered phospholipid metabolism, and neuroinflammation.140-142 Of potential relevance is NBIA caused by pathogenic variants of the PLA2G6 gene, causing PLA2G6-associated neurodegeneration.55,74,140,141,143,144 As mentioned earlier in this review, PLA2G6 degrades membrane hydroxyperoxy-phosphatidylethanolamine, thus protecting against ferroptosis.74 In cellular models, reduced PLA2G6 function alters vesicular transport (including retromer formation), impairs mitophagy and autophagy, and leads to lysosomal dysfunction and accumulation of lipofuscin.145,146 Lipofuscin recruits iron, promoting the formation of free radicals and thus lipid peroxidation in lysosomes.147,148
Perspective
The evidence that intracellular free iron accumulation, oxidative stress, and lipid peroxidation resulting in ferroptosis have a major role in neurodegeneration has led to multiple approaches to interrupt these processes. Studies in experimental models suggest that iron chelators such as deferoxamine and deferiprone; antioxidants and inhibitors of lipid peroxidation such as α-tocopherol, CoQ10, tetrahydrobiopterin, or edaravone; GPX4 inducers such as ferrostatin-1 and lipostatin-1; GSH precursors such as N-acetylcysteine; or activators of NRF2 signaling such as dimethyl fumarate could potentially have a neuroprotective effect.19,149 However, the role of iron chelation as a neuroprotective approach in human neurologic disorders is yet to be established. For example, a recent multicenter phase 2 randomized double-blind trial in patients with newly diagnosed PD showed that patients treated with oral deferiprone over the course of 36 weeks had worse scores in measures of parkinsonism despite a higher reduction of iron in the substantia nigra compared with those receiving placebo.150 Whereas iron chelation has also been suggested as a potential neuroprotective approach for AD,151 it has proven unsuccessful in patients with ALS.152 In a placebo-controlled double-blind multicenter trial in patients with pantothenate kinase–associated neurodegeneration, deferiprone reduced iron load in the basal ganglia and showed a trend to slowing of disease progression but did not produce any significant change in symptoms.153 Similarly, despite evidence from preclinical models, strong clinical evidence is still lacking for the benefit of antioxidant therapy as a neuroprotective approach, although edaravone may provide potential benefits in patients with ALS.154 Given the complex interactions among iron metabolism, oxidative stress, organelle dysfunction, and neuroinflammation at different disease stages, it is unlikely that targeting an individual component of the ferroptosis pathway will provide substantial clinical benefit. Novel therapies, such as epigenetic approaches11 or multitargeting drugs with iron-chelating, antioxidant, antiferroptotic, and anti-inflammatory properties may provide potential alternatives.
Glossary
- Aβ
β-amyloid
- AD
Alzheimer disease
- ALS
amyotrophic lateral sclerosis
- APP
amyloid precursor protein
- BBB
blood-brain barrier
- CoQ10
coenzyme Q10
- DMT1
divalent metal transporter 1
- Fe2+
iron
- Fe3+
ferric iron
- GPX
glutathione peroxidase
- GSH
glutathione
- GSSG
oxidized glutathione
- iPSC
induced pluripotent stem cell
- LOX
lipoxygenase
- mRNA
messenger RNA
- NADPH
nicotinamide adenine dinucleotide phosphate
- NBIA
neurodegeneration with brain iron accumulation
- NOX
nicotinamide oxidase
- NRF2
nuclear factor erythroid 2–related factor 2
- PD
Parkinson disease
- PLA2G6
phospholipase A2 type G6
- PS1
presenilin 1
- PUFA
polyunsaturated fatty acid
- ROS
reactive oxygen species
- TfR1
transferrin receptor 1
Study Funding
No targeted funding reported.
Disclosure
The authors report no relevant disclosures. Go to Neurology.org/N for full disclosures.
References
- 1.Dixon SJ, Lemberg KM, Lamprecht MR, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell. 2012;149(5):1060-1072. doi: 10.1016/j.cell.2012.03.042 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Seibt TM, Proneth B, Conrad M. Role of GPX4 in ferroptosis and its pharmacological implication. Free Radic Biol Med. 2019;133:144-152. doi: 10.1016/j.freeradbiomed.2018.09.014 [DOI] [PubMed] [Google Scholar]
- 3.Conrad M, Pratt DA. The chemical basis of ferroptosis. Nat Chem Biol. 2019;15(12):1137-1147. doi: 10.1038/s41589-019-0408-1 [DOI] [PubMed] [Google Scholar]
- 4.Jiang X, Stockwell BR, Conrad M. Ferroptosis: mechanisms, biology and role in disease. Nat Rev Mol Cell Biol. 2021;22(4):266-282. doi: 10.1038/s41580-020-00324-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Liu J, Kang R, Tang D. Signaling pathways and defense mechanisms of ferroptosis. FEBS J. 2022;289(22):7038-7050. doi: 10.1111/febs.16059 [DOI] [PubMed] [Google Scholar]
- 6.Kuang F, Liu J, Tang D, Kang R. Oxidative damage and antioxidant defense in ferroptosis. Front Cell Dev Biol. 2020;8:586578. doi: 10.3389/fcell.2020.586578 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Tang D, Chen X, Kang R, Kroemer G. Ferroptosis: molecular mechanisms and health implications. Cell Res. 2021;31(2):107-125. doi: 10.1038/s41422-020-00441-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Ward RJ, Zucca FA, Duyn JH, Crichton RR, Zecca L. The role of iron in brain ageing and neurodegenerative disorders. Lancet Neurol. 2014;13(10):1045-1060. doi: 10.1016/s1474-4422(14)70117-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Wise CE, Grant JL, Amaya JA, et al. Divergent mechanisms of iron-containing enzymes for hydrocarbon biosynthesis. J Biol Inorg Chem. 2017;22(2-3):221-235. doi: 10.1007/s00775-016-1425-0 [DOI] [PubMed] [Google Scholar]
- 10.Zheng J, Conrad M. The metabolic underpinnings of ferroptosis. Cell Metab. 2020;32(6):920-937. doi: 10.1016/j.cmet.2020.10.011 [DOI] [PubMed] [Google Scholar]
- 11.Lan T, Sun TT, Wei C, et al. Epigenetic regulation of ferroptosis in central nervous system diseases. Mol Neurobiol. 2023;60(7):3584-3599. doi: 10.1007/s12035-023-03267-1 [DOI] [PubMed] [Google Scholar]
- 12.Li Y, Xiao D, Wang X. The emerging roles of ferroptosis in cells of the central nervous system. Front Neurosci. 2022;16:1032140. doi: 10.3389/fnins.2022.1032140 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Kenkhuis B, Somarakis A, de Haan L, et al. Iron loading is a prominent feature of activated microglia in Alzheimer's disease patients. Acta Neuropathol Commun. 2021;9(1):27. doi: 10.1186/s40478-021-01126-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Kenkhuis B, van Eekeren M, Parfitt DA, et al. Iron accumulation induces oxidative stress, while depressing inflammatory polarization in human iPSC-derived microglia. Stem Cell Rep. 2022;17(6):1351-1365. doi: 10.1016/j.stemcr.2022.04.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Ryan SK, Zelic M, Han Y, et al. Microglia ferroptosis is regulated by SEC24B and contributes to neurodegeneration. Nat Neurosci. 2023;26(1):12-26. doi: 10.1038/s41593-022-01221-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Nakamura T, Naguro I, Ichijo H. Iron homeostasis and iron-regulated ROS in cell death, senescence and human diseases. Biochim Biophys Acta Gen Subj. 2019;1863(9):1398-1409. doi: 10.1016/j.bbagen.2019.06.010 [DOI] [PubMed] [Google Scholar]
- 17.Yan HF, Zou T, Tuo QZ, et al. Ferroptosis: mechanisms and links with diseases. Signal Transduct Target Ther. 2021;6(1):49. doi: 10.1038/s41392-020-00428-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Du L, Wu Y, Fan Z, et al. The role of ferroptosis in nervous system disorders. J Integr Neurosci. 2023;22(1):19. doi: 10.31083/j.jin2201019 [DOI] [PubMed] [Google Scholar]
- 19.Costa I, Barbosa DJ, Benfeito S, et al. Molecular mechanisms of ferroptosis and their involvement in brain diseases. Pharmacol Ther. 2023;244:108373. doi: 10.1016/j.pharmthera.2023.108373 [DOI] [PubMed] [Google Scholar]
- 20.Xu Y, Zhao J, Zhao Y, et al. The role of ferroptosis in neurodegenerative diseases. Mol Biol Rep. 2023;50(2):1655-1661. doi: 10.1007/s11033-022-08048-y [DOI] [PubMed] [Google Scholar]
- 21.Reichert CO, de Freitas FA, Sampaio-Silva J, et al. Ferroptosis mechanisms involved in neurodegenerative diseases. Int J Mol Sci. 2020;21(22):8765. doi: 10.3390/ijms21228765 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Cheli VT, Correale J, Paez PM, Pasquini JM. Iron metabolism in oligodendrocytes and astrocytes, implications for myelination and remyelination. ASN Neuro. 2020;12:1759091420962681. doi: 10.1177/1759091420962681 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.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]
- 24.Donovan A, Roy CN, Andrews NC. The ins and outs of iron homeostasis. Physiology (Bethesda). 2006;21(2):115-123. doi: 10.1152/physiol.00052.2005 [DOI] [PubMed] [Google Scholar]
- 25.Gao M, Monian P, Quadri N, Ramasamy R, Jiang X. Glutaminolysis and transferrin regulate ferroptosis. Mol Cell. 2015;59(2):298-308. doi: 10.1016/j.molcel.2015.06.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Yanatori I, Kishi F. DMT1 and iron transport. Free Radic Biol Med. 2019;133:55-63. doi: 10.1016/j.freeradbiomed.2018.07.020 [DOI] [PubMed] [Google Scholar]
- 27.Wendeler MW, Paccaud JP, Hauri HP. Role of Sec24 isoforms in selective export of membrane proteins from the endoplasmic reticulum. EMBO Rep. 2007;8(3):258-264. doi: 10.1038/sj.embor.7400893 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Ayton S, Lei P, Duce JA, et al. Ceruloplasmin dysfunction and therapeutic potential for Parkinson disease. Ann Neurol. 2013;73(4):554-559. doi: 10.1002/ana.23817 [DOI] [PubMed] [Google Scholar]
- 29.Billesbolle CB, Azumaya CM, Kretsch RC, et al. Structure of hepcidin-bound ferroportin reveals iron homeostatic mechanisms. Nature. 2020;586(7831):807-811. doi: 10.1038/s41586-020-2668-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Zecca L, Youdim MB, Riederer P, Connor JR, Crichton RR. Iron, brain ageing and neurodegenerative disorders. Nat Rev Neurosci. 2004;5(11):863-873. doi: 10.1038/nrn1537 [DOI] [PubMed] [Google Scholar]
- 31.Zucca FA, Capucciati A, Bellei C, et al. Neuromelanins in brain aging and Parkinson's disease: synthesis, structure, neuroinflammatory, and neurodegenerative role. IUBMB Life. 2023;75(1):55-65. doi: 10.1002/iub.2654 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Muckenthaler MU, Galy B, Hentze MW. Systemic iron homeostasis and the iron-responsive element/iron-regulatory protein (IRE/IRP) regulatory network. Annu Rev Nutr. 2008;28(1):197-213. doi: 10.1146/annurev.nutr.28.061807.155521 [DOI] [PubMed] [Google Scholar]
- 33.Winterbourn CC. Toxicity of iron and hydrogen peroxide: the Fenton reaction. Toxicol Lett. 1995;82-83:969-974. doi: 10.1016/0378-4274(95)03532-x [DOI] [PubMed] [Google Scholar]
- 34.Bock FJ, Tait SWG. Mitochondria as multifaceted regulators of cell death. Nat Rev Mol Cell Biol. 2020;21(2):85-100. doi: 10.1038/s41580-019-0173-8 [DOI] [PubMed] [Google Scholar]
- 35.Li C, Zhang Y, Liu J, Kang R, Klionsky DJ, Tang D. Mitochondrial DNA stress triggers autophagy-dependent ferroptotic death. Autophagy. 2021;17(4):948-960. doi: 10.1080/15548627.2020.1739447 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Gao M, Yi J, Zhu J, et al. Role of mitochondria in ferroptosis. Mol Cell. 2019;73(2):354-363.e3. doi: 10.1016/j.molcel.2018.10.042 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Chen PH, Tseng WH, Chi JT. The intersection of DNA damage response and ferroptosis-A rationale for combination therapeutics. Biology (Basel). 2020;9(8):187. doi: 10.3390/biology9080187 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Liu J, Kuang F, Kroemer G, Klionsky DJ, Kang R, Tang D. Autophagy-dependent ferroptosis: machinery and regulation. Cell Chem Biol. 2020;27(4):420-435. doi: 10.1016/j.chembiol.2020.02.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Park E, Chung SW. ROS-mediated autophagy increases intracellular iron levels and ferroptosis by ferritin and transferrin receptor regulation. Cell Death Dis. 2019;10(11):822. doi: 10.1038/s41419-019-2064-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Lee H, Zhuang L, Gan B. Energy stress inhibits ferroptosis via AMPK. Mol Cell Oncol. 2020;7(4):1761242. doi: 10.1080/23723556.2020.1761242 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Song X, Zhu S, Chen P, et al. AMPK-mediated BECN1 phosphorylation promotes ferroptosis by directly blocking system X(c)(-) activity. Curr Biol. 2018;28(15):2388-2399.e5. doi: 10.1016/j.cub.2018.05.094 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Ma MW, Wang J, Zhang Q, et al. NADPH oxidase in brain injury and neurodegenerative disorders. Mol Neurodegener. 2017;12(1):7. doi: 10.1186/s13024-017-0150-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Tu D, Velagapudi R, Gao Y, Hong JS, Zhou H, Gao HM. Activation of neuronal NADPH oxidase NOX2 promotes inflammatory neurodegeneration. Free Radic Biol Med. 2023;200:47-58. doi: 10.1016/j.freeradbiomed.2023.03.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Bedard K, Krause KH. The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology. Physiol Rev. 2007;87(1):245-313. doi: 10.1152/physrev.00044.2005 [DOI] [PubMed] [Google Scholar]
- 45.Kagan VE, Tyurina YY, Vlasova II, et al. Redox epiphospholipidome in programmed cell death signaling: catalytic mechanisms and regulation. Front Endocrinol (Lausanne). 2020;11:628079. doi: 10.3389/fendo.2020.628079 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Yuan H, Li X, Zhang X, Kang R, Tang D. Identification of ACSL4 as a biomarker and contributor of ferroptosis. Biochem Biophys Res Commun. 2016;478(3):1338-1343. doi: 10.1016/j.bbrc.2016.08.124 [DOI] [PubMed] [Google Scholar]
- 47.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]
- 48.Zou Y, Henry WS, Ricq EL, et al. Plasticity of ether lipids promotes ferroptosis susceptibility and evasion. Nature. 2020;585(7826):603-608. doi: 10.1038/s41586-020-2732-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Bai Y, Meng L, Han L, et al. Lipid storage and lipophagy regulates ferroptosis. Biochem Biophys Res Commun. 2019;508(4):997-1003. doi: 10.1016/j.bbrc.2018.12.039 [DOI] [PubMed] [Google Scholar]
- 50.Yang WS, Kim KJ, Gaschler MM, Patel M, Shchepinov MS, Stockwell BR. Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis. Proc Natl Acad Sci USA. 2016;113(34):E4966-E4975. doi: 10.1073/pnas.1603244113 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Zou Y, Li H, Graham ET, et al. Cytochrome P450 oxidoreductase contributes to phospholipid peroxidation in ferroptosis. Nat Chem Biol. 2020;16(3):302-309. doi: 10.1038/s41589-020-0472-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Ademowo OS, Dias HKI, Burton DGA, Griffiths HR. Lipid (per) oxidation in mitochondria: an emerging target in the ageing process? Biogerontology. 2017;18(6):859-879. doi: 10.1007/s10522-017-9710-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Su LJ, Zhang JH, Gomez H, et al. Reactive oxygen species-induced lipid peroxidation in apoptosis, autophagy, and ferroptosis. Oxid Med Cell Longev. 2019;2019:5080843. doi: 10.1155/2019/5080843 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Moreno-Garcia A, Kun A, Calero O, Medina M, Calero M. An overview of the role of lipofuscin in age-related neurodegeneration. Front Neurosci. 2018;12:464. doi: 10.3389/fnins.2018.00464 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Villalon-Garcia I, Povea-Cabello S, Alvarez-Cordoba M, et al. Vicious cycle of lipid peroxidation and iron accumulation in neurodegeneration. Neural Regen Res. 2023;18(6):1196-1202. doi: 10.4103/1673-5374.358614 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Wen Q, Liu J, Kang R, Zhou B, Tang D. The release and activity of HMGB1 in ferroptosis. Biochem Biophys Res Commun. 2019;510(2):278-283. doi: 10.1016/j.bbrc.2019.01.090 [DOI] [PubMed] [Google Scholar]
- 57.Kuang F, Liu J, Li C, Kang R, Tang D. Cathepsin B is a mediator of organelle-specific initiation of ferroptosis. Biochem Biophys Res Commun. 2020;533(4):1464-1469. doi: 10.1016/j.bbrc.2020.10.035 [DOI] [PubMed] [Google Scholar]
- 58.Nagakannan P, Islam MI, Conrad M, Eftekharpour E. Cathepsin B is an executioner of ferroptosis. Biochim Biophys Acta Mol Cell Res. 2021;1868(3):118928. doi: 10.1016/j.bbamcr.2020.118928 [DOI] [PubMed] [Google Scholar]
- 59.Dodson M, Wani WY, Redmann M, et al. Regulation of autophagy, mitochondrial dynamics, and cellular bioenergetics by 4-hydroxynonenal in primary neurons. Autophagy. 2017;13(11):1828-1840. doi: 10.1080/15548627.2017.1356948 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Cardoso BR, Hare DJ, Bush AI, Roberts BR. Glutathione peroxidase 4: a new player in neurodegeneration? Mol Psychiatry. 2017;22(3):328-335. doi: 10.1038/mp.2016.196 [DOI] [PubMed] [Google Scholar]
- 61.Parker JL, Deme JC, Kolokouris D, et al. Molecular basis for redox control by the human cystine/glutamate antiporter system xc. Nat Commun. 2021;12(1):7147. doi: 10.1038/s41467-021-27414-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Asanuma M, Miyazaki I. Glutathione and related molecules in parkinsonism. Int J Mol Sci. 2021;22(16):8689. doi: 10.3390/ijms22168689 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Forcina GC, Dixon SJ. GPX4 at the crossroads of lipid homeostasis and ferroptosis. Proteomics. 2019;19(18):e1800311. doi: 10.1002/pmic.201800311 [DOI] [PubMed] [Google Scholar]
- 64.Santoro MM. The antioxidant role of non-mitochondrial CoQ10: mystery solved! Cell Metab. 2020;31(1):13-15. doi: 10.1016/j.cmet.2019.12.007 [DOI] [PubMed] [Google Scholar]
- 65.Warner GJ, Berry MJ, Moustafa ME, Carlson BA, Hatfield DL, Faust JR. Inhibition of selenoprotein synthesis by selenocysteine tRNA[Ser]Sec lacking isopentenyladenosine. J Biol Chem. 2000;275(36):28110-28119. doi: 10.1074/jbc.m001280200 [DOI] [PubMed] [Google Scholar]
- 66.Mao C, Liu X, Zhang Y, et al. DHODH-mediated ferroptosis defence is a targetable vulnerability in cancer. Nature. 2021;593(7860):586-590. doi: 10.1038/s41586-021-03539-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Kraft VAN, Bezjian CT, Pfeiffer S, et al. GTP cyclohydrolase 1/tetrahydrobiopterin counteract ferroptosis through lipid remodeling. ACS Cent Sci. 2020;6(1):41-53. doi: 10.1021/acscentsci.9b01063 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Vasquez-Vivar J, Shi Z, Tan S. Tetrahydrobiopterin in cell function and death mechanisms. Antioxid Redox Signal. 2022;37(1-3):171-183. doi: 10.1089/ars.2021.0136 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Nehring H, Meierjohann S, Friedmann Angeli JP. Emerging aspects in the regulation of ferroptosis. Biochem Soc Trans. 2020;48(5):2253-2259. doi: 10.1042/bst20200523 [DOI] [PubMed] [Google Scholar]
- 70.He F, Ru X, Wen T. NRF2, a transcription factor for stress response and beyond. Int J Mol Sci. 2020;21(13):4777. doi: 10.3390/ijms21134777 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Dodson M, Castro-Portuguez R, Zhang DD. NRF2 plays a critical role in mitigating lipid peroxidation and ferroptosis. Redox Biol. 2019;23:101107. doi: 10.1016/j.redox.2019.101107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Lane DJR, Metselaar B, Greenough M, Bush AI, Ayton SJ. Ferroptosis and NRF2: an emerging battlefield in the neurodegeneration of Alzheimer's disease. Essays Biochem. 2021;65(7):925-940. doi: 10.1042/ebc20210017 [DOI] [PubMed] [Google Scholar]
- 73.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 Chem Biol. 2020;27(4):436-447. doi: 10.1016/j.chembiol.2020.03.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Chen D, Chu B, Yang X, et al. iPLA2β-mediated lipid detoxification controls p53-driven ferroptosis independent of GPX4. Nat Commun. 2021;12(1):3644. doi: 10.1038/s41467-021-23902-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Dai E, Meng L, Kang R, Wang X, Tang D. ESCRT-III-dependent membrane repair blocks ferroptosis. Biochem Biophys Res Commun. 2020;522(2):415-421. doi: 10.1016/j.bbrc.2019.11.110 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Liu JL, Fan YG, Yang ZS, Wang ZY, Guo C. Iron and Alzheimer's disease: from pathogenesis to therapeutic implications. Front Neurosci. 2018;12:632. doi: 10.3389/fnins.2018.00632 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Ashraf A, Jeandriens J, Parkes HG, So PW. Iron dyshomeostasis, lipid peroxidation and perturbed expression of cystine/glutamate antiporter in Alzheimer's disease: evidence of ferroptosis. Redox Biol. 2020;32:101494. doi: 10.1016/j.redox.2020.101494 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Wu L, Xian X, Tan Z, et al. The role of iron metabolism, lipid metabolism, and redox homeostasis in Alzheimer's disease: from the perspective of ferroptosis. Mol Neurobiol. 2023;60(5):2832-2850. doi: 10.1007/s12035-023-03245-7 [DOI] [PubMed] [Google Scholar]
- 79.Wang M, Tang G, Zhou C, et al. Revisiting the intersection of microglial activation and neuroinflammation in Alzheimer's disease from the perspective of ferroptosis. Chem Biol Interact. 2023;375:110387. doi: 10.1016/j.cbi.2023.110387 [DOI] [PubMed] [Google Scholar]
- 80.Lane DJR, Ayton S, Bush AI. Iron and Alzheimer's disease: an update on emerging mechanisms. J Alzheimers Dis. 2018;64(suppl 1):S379-S395. doi: 10.3233/jad-179944 [DOI] [PubMed] [Google Scholar]
- 81.Yan N, Zhang J. Iron metabolism, ferroptosis, and the links with Alzheimer's disease. Front Neurosci. 2019;13:1443. doi: 10.3389/fnins.2019.01443 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Chen K, Jiang X, Wu M, Cao X, Bao W, Zhu LQ. Ferroptosis, a potential therapeutic target in Alzheimer's disease. Front Cell Dev Biol. 2021;9:704298. doi: 10.3389/fcell.2021.704298 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Jiang X, Wu K, Ye XY, et al. Novel druggable mechanism of Parkinson's disease: potential therapeutics and underlying pathogenesis based on ferroptosis. Med Res Rev. 2023;43(4):872-896. doi: 10.1002/med.21939 [DOI] [PubMed] [Google Scholar]
- 84.Lin KJ, Chen SD, Lin KL, et al. Iron brain menace: the involvement of ferroptosis in Parkinson disease. Cells. 2022;11(23):3829. doi: 10.3390/cells11233829 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Guiney SJ, Adlard PA, Bush AI, Finkelstein DI, Ayton S. Ferroptosis and cell death mechanisms in Parkinson's disease. Neurochem Int. 2017;104:34-48. doi: 10.1016/j.neuint.2017.01.004 [DOI] [PubMed] [Google Scholar]
- 86.Thomas GEC, Zarkali A, Ryten M, et al. Regional brain iron and gene expression provide insights into neurodegeneration in Parkinson's disease. Brain. 2021;144(6):1787-1798. doi: 10.1093/brain/awab084 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Mahoney-Sanchez L, Bouchaoui H, Ayton S, Devos D, Duce JA, Devedjian JC. Ferroptosis and its potential role in the physiopathology of Parkinson's disease. Prog Neurobiol. 2021;196:101890. doi: 10.1016/j.pneurobio.2020.101890 [DOI] [PubMed] [Google Scholar]
- 88.Devos D, Moreau C, Kyheng M, et al. A ferroptosis-based panel of prognostic biomarkers for amyotrophic lateral sclerosis. Sci Rep. 2019;9(1):2918. doi: 10.1038/s41598-019-39739-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Chen L, Hambright WS, Na R, Ran Q. Ablation of the ferroptosis inhibitor glutathione peroxidase 4 in neurons results in rapid motor neuron degeneration and paralysis. J Biol Chem. 2015;290(47):28097-28106. doi: 10.1074/jbc.m115.680090 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Wang T, Tomas D, Perera ND, et al. Ferroptosis mediates selective motor neuron death in amyotrophic lateral sclerosis. Cell Death Differ. 2022;29(6):1187-1198. doi: 10.1038/s41418-021-00910-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Wang D, Liang W, Huo D, et al. SPY1 inhibits neuronal ferroptosis in amyotrophic lateral sclerosis by reducing lipid peroxidation through regulation of GCH1 and TFR1. Cell Death Differ. 2023;30(2):369-382. doi: 10.1038/s41418-022-01089-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Evans RC, Chen L, Na R, Yoo K, Ran Q. The Gpx4NIKO mouse is a versatile model for testing interventions targeting ferroptotic cell death of spinal motor neurons. Neurotox Res. 2022;40(2):373-383. doi: 10.1007/s12640-021-00469-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Agrawal S, Fox J, Thyagarajan B, Fox JH. Brain mitochondrial iron accumulates in Huntington's disease, mediates mitochondrial dysfunction, and can be removed pharmacologically. Free Radic Biol Med. 2018;120:317-329. doi: 10.1016/j.freeradbiomed.2018.04.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Lee J, Kosaras B, Del Signore SJ, et al. Modulation of lipid peroxidation and mitochondrial function improves neuropathology in Huntington's disease mice. Acta Neuropathol. 2011;121(4):487-498. doi: 10.1007/s00401-010-0788-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Chen J, Marks E, Lai B, et al. Iron accumulates in Huntington's disease neurons: protection by deferoxamine. PLoS One. 2013;8(10):e77023. doi: 10.1371/journal.pone.0077023 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Chen K, Lin G, Haelterman NA, et al. Loss of Frataxin induces iron toxicity, sphingolipid synthesis, and Pdk1/Mef2 activation, leading to neurodegeneration. Elife. 2016;5:e16043. doi: 10.7554/elife.16043 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Turchi R, Faraonio R, Lettieri-Barbato D, Aquilano K. An overview of the ferroptosis hallmarks in Friedreich's ataxia. Biomolecules. 2020;10(11):1489. doi: 10.3390/biom10111489 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.La Rosa P, Petrillo S, Turchi R, et al. The Nrf2 induction prevents ferroptosis in Friedreich's ataxia. Redox Biol. 2021;38:101791. doi: 10.1016/j.redox.2020.101791 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Hu CL, Nydes M, Shanley KL, Morales Pantoja IE, Howard TA, Bizzozero OA. Reduced expression of the ferroptosis inhibitor glutathione peroxidase-4 in multiple sclerosis and experimental autoimmune encephalomyelitis. J Neurochem. 2019;148(3):426-439. doi: 10.1111/jnc.14604 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Jhelum P, Santos-Nogueira E, Teo W, et al. Ferroptosis mediates cuprizone-induced loss of oligodendrocytes and demyelination. J Neurosci. 2020;40(48):9327-9341. doi: 10.1523/jneurosci.1749-20.2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Luoqian J, Yang W, Ding X, et al. Ferroptosis promotes T-cell activation-induced neurodegeneration in multiple sclerosis. Cell Mol Immunol. 2022;19(8):913-924. doi: 10.1038/s41423-022-00883-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Ahmad S, Elsherbiny NM, Haque R, et al. Sesamin attenuates neurotoxicity in mouse model of ischemic brain stroke. Neurotoxicology. 2014;45:100-110. doi: 10.1016/j.neuro.2014.10.002 [DOI] [PubMed] [Google Scholar]
- 103.Bu ZQ, Yu HY, Wang J, et al. Emerging role of ferroptosis in the pathogenesis of ischemic stroke: a new therapeutic target? ASN Neuro. 2021;13:17590914211037505. doi: 10.1177/17590914211037505 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Guo J, Tuo QZ, Lei P. Iron, ferroptosis, and ischemic stroke. J Neurochem. 2023;165(4):487-520. doi: 10.1111/jnc.15807 [DOI] [PubMed] [Google Scholar]
- 105.Cui Y, Zhou X, Meng P, et al. Identifying potential therapeutic targets for ischemic stroke through immune infiltration analysis and construction of a programmed cell death-related ceRNA network. Exp Ther Med. 2022;24(5):680. doi: 10.3892/etm.2022.11616 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Bai Q, Liu J, Wang G. Ferroptosis, a regulated neuronal cell death type after intracerebral hemorrhage. Front Cell Neurosci. 2020;14:591874. doi: 10.3389/fncel.2020.591874 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Pan F, Xu W, Ding J, Wang C. Elucidating the progress and impact of ferroptosis in hemorrhagic stroke. Front Cell Neurosci. 2022;16:1067570. doi: 10.3389/fncel.2022.1067570 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Magtanong L, Dixon SJ. Ferroptosis and brain injury. Dev Neurosci. 2018;40(5-6):382-395. doi: 10.1159/000496922 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Zhong FM, Yao FY, Liu J, et al. Ferroptosis-related molecular patterns reveal immune escape, inflammatory development and lipid metabolism characteristics of the tumor microenvironment in acute myeloid leukemia. Front Oncol. 2022;12:888570. doi: 10.3389/fonc.2022.888570 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Shi Z, Yuan S, Shi L, et al. Programmed cell death in spinal cord injury pathogenesis and therapy. Cell Prolif. 2021;54(3):e12992. doi: 10.1111/cpr.12992 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Du K, He M, Zhao D, et al. Mechanism of cell death pathways in status epilepticus and related therapeutic agents. Biomed Pharmacother. 2022;149:112875. doi: 10.1016/j.biopha.2022.112875 [DOI] [PubMed] [Google Scholar]
- 112.Cai Y, Yang Z. Ferroptosis and its role in epilepsy. Front Cell Neurosci. 2021;15:696889. doi: 10.3389/fncel.2021.696889 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Chen S, Chen Y, Zhang Y, et al. Iron metabolism and ferroptosis in epilepsy. Front Neurosci. 2020;14:601193. doi: 10.3389/fnins.2020.601193 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Liu Y, Xu Z, Jin T, Xu K, Liu M, Xu H. Ferroptosis in low-grade glioma: a new marker for diagnosis and prognosis. Med Sci Monit. 2020;26:e921947. doi: 10.12659/msm.921947 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Pena-Bautista C, Vento M, Baquero M, Chafer-Pericas C. Lipid peroxidation in neurodegeneration. Clin Chim Acta. 2019;497:178-188. doi: 10.1016/j.cca.2019.07.037 [DOI] [PubMed] [Google Scholar]
- 116.Chafer-Pericas C. Lipid peroxidation in neurodegeneration. Antioxidants (Basel). 2021;10(3):484. doi: 10.3390/antiox10030484 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Dusek P, Hofer T, Alexander J, Roos PM, Aaseth JO. Cerebral iron deposition in neurodegeneration. Biomolecules. 2022;12(5):714. doi: 10.3390/biom12050714 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Angelova PR, Esteras N, Abramov AY. Mitochondria and lipid peroxidation in the mechanism of neurodegeneration: finding ways for prevention. Med Res Rev. 2021;41(2):770-784. doi: 10.1002/med.21712 [DOI] [PubMed] [Google Scholar]
- 119.Lyamzaev KG, Panteleeva AA, Simonyan RA, Avetisyan AV, Chernyak BV. Mitochondrial lipid peroxidation is responsible for ferroptosis. Cells. 2023;12(4):611. doi: 10.3390/cells12040611 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Rogers JT, Cahill CM. Iron-responsive-like elements and neurodegenerative ferroptosis. Learn Mem. 2020;27(9):395-413. doi: 10.1101/lm.052282.120 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Ayton S, Portbury S, Kalinowski P, et al. Regional brain iron associated with deterioration in Alzheimer's disease: a large cohort study and theoretical significance. Alzheimers Dement. 2021;17(7):1244-1256. doi: 10.1002/alz.12282 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Bao WD, Pang P, Zhou XT, et al. Loss of ferroportin induces memory impairment by promoting ferroptosis in Alzheimer's disease. Cell Death Differ. 2021;28(5):1548-1562. doi: 10.1038/s41418-020-00685-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Tao Y, Wang Y, Rogers JT, Wang F. Perturbed iron distribution in Alzheimer's disease serum, cerebrospinal fluid, and selected brain regions: a systematic review and meta-analysis. J Alzheimers Dis. 2014;42(2):679-690. doi: 10.3233/jad-140396 [DOI] [PubMed] [Google Scholar]
- 124.Kim AC, Lim S, Kim YK. Metal ion effects on Aβ and tau aggregation. Int J Mol Sci. 2018;19(1):128. doi: 10.3390/ijms19010128 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Vallerga CL, Zhang F, Fowdar J, et al. Analysis of DNA methylation associates the cystine-glutamate antiporter SLC7A11 with risk of Parkinson's disease. Nat Commun. 2020;11(1):1238. doi: 10.1038/s41467-020-15065-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Lingor P, Carboni E, Koch JC. Alpha-synuclein and iron: two keys unlocking Parkinson's disease. J Neural Transm (Vienna). 2017;124(8):973-981. doi: 10.1007/s00702-017-1695-x [DOI] [PubMed] [Google Scholar]
- 127.Mahoney-Sanchez L, Bouchaoui H, Boussaad I, et al. Alpha synuclein determines ferroptosis sensitivity in dopaminergic neurons via modulation of ether-phospholipid membrane composition. Cell Rep. 2022;40(8):111231. doi: 10.1016/j.celrep.2022.111231 [DOI] [PubMed] [Google Scholar]
- 128.Bouchaoui H, Mahoney-Sanchez L, Garcon G, et al. ACSL4 and the lipoxygenases 15/15B are pivotal for ferroptosis induced by iron and PUFA dyshomeostasis in dopaminergic neurons. Free Radic Biol Med. 2023;195:145-157. doi: 10.1016/j.freeradbiomed.2022.12.086 [DOI] [PubMed] [Google Scholar]
- 129.Angelova PR, Choi ML, Berezhnov AV, et al. Alpha synuclein aggregation drives ferroptosis: an interplay of iron, calcium and lipid peroxidation. Cell Death Differ. 2020;27(10):2781-2796. doi: 10.1038/s41418-020-0542-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Shamoto-Nagai M, Hisaka S, Naoi M, Maruyama W. Modification of alpha-synuclein by lipid peroxidation products derived from polyunsaturated fatty acids promotes toxic oligomerization: its relevance to Parkinson disease. J Clin Biochem Nutr. 2018;62(3):207-212. doi: 10.3164/jcbn.18-25 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Sterling JK, Kam TI, Guttha S, et al. Interleukin-6 triggers toxic neuronal iron sequestration in response to pathological alpha-synuclein. Cell Rep. 2022;38(7):110358. doi: 10.1016/j.celrep.2022.110358 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Guan W, Xia M, Ji M, et al. Iron induces two distinct Ca(2+) signalling cascades in astrocytes. Commun Biol. 2021;4(1):525. doi: 10.1038/s42003-021-02060-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Mochizuki H, Choong CJ, Baba K. Parkinson's disease and iron. J Neural Transm (Vienna). 2020;127(2):181-187. doi: 10.1007/s00702-020-02149-3 [DOI] [PubMed] [Google Scholar]
- 134.Braidy N, Zarka M, Jugder BE, et al. The precursor to glutathione (GSH), γ-glutamylcysteine (GGC), can ameliorate oxidative damage and neuroinflammation induced by Aβ40 oligomers in human astrocytes. Front Aging Neurosci. 2019;11:177. doi: 10.3389/fnagi.2019.00177 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Huang SF, Othman A, Koshkin A, et al. Astrocyte glutathione maintains endothelial barrier stability. Redox Biol. 2020;34:101576. doi: 10.1016/j.redox.2020.101576 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Absinta M, Maric D, Gharagozloo M, et al. A lymphocyte-microglia-astrocyte axis in chronic active multiple sclerosis. Nature. 2021;597(7878):709-714. doi: 10.1038/s41586-021-03892-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Mathys H, Adaikkan C, Gao F, et al. Temporal tracking of microglia activation in neurodegeneration at single-cell resolution. Cell Rep. 2017;21(2):366-380. doi: 10.1016/j.celrep.2017.09.039 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.McIntosh A, Mela V, Harty C, et al. Iron accumulation in microglia triggers a cascade of events that leads to altered metabolism and compromised function in APP/PS1 mice. Brain Pathol. 2019;29(5):606-621. doi: 10.1111/bpa.12704 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Zheng W, Aschner M, Ghersi-Egea JF. Brain barrier systems: a new frontier in metal neurotoxicological research. Toxicol Appl Pharmacol. 2003;192:1-11. doi: 10.1016/s0041-008x(03)00251-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Hinarejos I, Machuca-Arellano C, Sancho P, Espinos C. Mitochondrial dysfunction, oxidative stress and neuroinflammation in neurodegeneration with brain iron accumulation (NBIA). Antioxidants (Basel). 2020;9(10):1020. doi: 10.3390/antiox9101020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Hayflick SJ, Kurian MA, Hogarth P. Neurodegeneration with brain iron accumulation. Handb Clin Neurol. 2018;147:293-305. doi: 10.1016/B978-0-444-63233-3.00019-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Tello C, Darling A, Lupo V, Perez-Duenas B, Espinos C. On the complexity of clinical and molecular bases of neurodegeneration with brain iron accumulation. Clin Genet. 2018;93(4):731-740. doi: 10.1111/cge.13057 [DOI] [PubMed] [Google Scholar]
- 143.Sun WY, Tyurin VA, Mikulska-Ruminska K, et al. Phospholipase iPLA2β averts ferroptosis by eliminating a redox lipid death signal. Nat Chem Biol. 2021;17(4):465-476. doi: 10.1038/s41589-020-00734-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Kinghorn KJ, Castillo-Quan JI, Bartolome F, et al. Loss of PLA2G6 leads to elevated mitochondrial lipid peroxidation and mitochondrial dysfunction. Brain. 2015;138(7):1801-1816. doi: 10.1093/brain/awv132 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Arber CE, Li A, Houlden H, Wray S. Review: insights into molecular mechanisms of disease in neurodegeneration with brain iron accumulation: unifying theories. Neuropathol Appl Neurobiol. 2016;42(3):220-241. doi: 10.1111/nan.12242 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Lin G, Lee PT, Chen K, et al. Phospholipase PLA2G6, a parkinsonism-associated gene, affects Vps26 and Vps35, retromer function, and ceramide levels, similar to alpha-synuclein gain. Cell Metab. 2018;28(4):605-618.e6. doi: 10.1016/j.cmet.2018.05.019 [DOI] [PubMed] [Google Scholar]
- 147.Hohn A, Jung T, Grimm S, Grune T. Lipofuscin-bound iron is a major intracellular source of oxidants: role in senescent cells. Free Radic Biol Med. 2010;48(8):1100-1108. doi: 10.1016/j.freeradbiomed.2010.01.030 [DOI] [PubMed] [Google Scholar]
- 148.Villalon-Garcia I, Alvarez-Cordoba M, Povea-Cabello S, et al. Vitamin E prevents lipid peroxidation and iron accumulation in PLA2G6-associated neurodegeneration. Neurobiol Dis. 2022;165:105649. doi: 10.1016/j.nbd.2022.105649 [DOI] [PubMed] [Google Scholar]
- 149.Cheng H, Wang P, Wang N, et al. Neuroprotection of NRF2 against ferroptosis after traumatic brain injury in mice. Antioxidants (Basel). 2023;12(3):731. doi: 10.3390/antiox12030731 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Devos D, Labreuche J, Rascol O, et al. Trial of deferiprone in Parkinson's disease. N Engl J Med. 2022;387(22):2045-2055. doi: 10.1056/nejmoa2209254 [DOI] [PubMed] [Google Scholar]
- 151.Gleason A, Bush AI. Iron and ferroptosis as therapeutic targets in Alzheimer's disease. Neurotherapeutics. 2021;18(1):252-264. doi: 10.1007/s13311-020-00954-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Bu XL, Xiang Y, Guo Y. The role of iron in amyotrophic lateral sclerosis. Adv Exp Med Biol. 2019;1173:145-152. doi: 10.1007/978-981-13-9589-5_8 [DOI] [PubMed] [Google Scholar]
- 153.Klopstock T, Tricta F, Neumayr L, et al. Safety and efficacy of deferiprone for pantothenate kinase-associated neurodegeneration: a randomised, double-blind, controlled trial and an open-label extension study. Lancet Neurol. 2019;18(7):631-642. doi: 10.1016/s1474-4422(19)30142-5 [DOI] [PubMed] [Google Scholar]
- 154.Gao M, Zhu L, Chang J, et al. Safety and efficacy of edaravone in patients with amyotrophic lateral sclerosis: a systematic review and meta-analysis. Clin Drug Investig. 2023;43:1-11. doi: 10.1007/s40261-022-01229-4 [DOI] [PubMed] [Google Scholar]

