Open Access

The role of ferroptosis in Alzheimer's disease: Mechanisms and therapeutic potential (Review)

  • Authors:
    • Heng Zeng
    • Zhaohui Jin
  • View Affiliations

  • Published online on: May 2, 2025     https://doi.org/10.3892/mmr.2025.13557
  • Article Number: 192
  • Copyright: © Zeng et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Alzheimer's disease (AD) is a prevalent neurodegenerative disorder characterized by insidious onset and progressive symptom deterioration. It extends beyond a simple aging process, involving irreversible and progressive neurological degeneration that impairs brain function through multiple etiologies. Iron dysregulation is implicated in the pathophysiology of AD; however, the precise mechanisms remain unclear. Additionally, vitamin E and selenium are key in regulating ferroptosis through their antioxidant properties. The present review examined the mechanistic pathways by which ferroptosis contributes to AD, the regulatory roles of vitamin E, selenium, ferrostatin‑1, N‑acetylcysteine and curcumin, and their potential as therapeutic agents to mitigate neurodegeneration.

Introduction

Alzheimer's disease (AD), the most prevalent form of dementia in the elderly, leads to a gradual decline in cognitive function. It is characterized by memory loss, language difficulty, impaired judgment, mood swings and, in advanced stages, loss of self-care ability (1). Epidemiological data reveals that >50 million individuals worldwide are affected by AD, with age-standardized prevalence of dementia in patients aged >60 is ~5–7% worldwide, making it one of the most expensive and fatal diseases globally (the absolute numbers of deaths have increased by 39%) (2). China has the highest number of patients with AD, with ~9.83 million individuals >60 years old diagnosed with AD (3). While the exact etiology of AD remains unclear, its development is linked to a range of factors, including genetic predisposition, abnormal protein aggregation, neurotransmitter imbalance and neuronal damage (46). Chinese Food and Drug Administration-approved treatments for AD include memantine, rivastigmine, galantamine and donepezil (7). In China, treatment recommendations for cognitive symptoms involve cholinesterase inhibitors, glutamate receptor antagonists such as memantine and combination therapy with both classes of drugs. Psychobehavioral symptoms are commonly managed with medications such as atypical antipsychotics and selective 5-hydroxytryptamine receptor agonists (8). The rise of artificial intelligence has led to the increasing use of data mining techniques, with complex network analysis based on graph theory offering a promising approach for clinical application (9,10). Topological indices expedite AD drug discovery by enabling rapid computational screening of compounds. They highlight promising candidates for further testing, bridging computational predictions and therapeutic development. Their integration with multi-omics data and machine learning holds promise for future breakthroughs in understanding and treating AD. Ashraf et al (11) employed quantitative structure-property association analysis to explore topological indices and drug properties for AD treatment. The analysis identified key structural features(such as topological indices) associated with drug efficacy, providing valuable insight for the design of more effective AD therapeutics (11). Despite the variety of mechanisms through which current drugs operate, most approved treatments fail to prevent the pathological progression of AD, and often exhibit limited efficacy or notable side effects (12,13). Thus, further research is key to improve understanding of the underlying mechanisms of AD and develop more effective therapies.

Ferroptosis, a distinct form of cell death driven by iron-dependent lipid peroxidation, serves a key role in several biological processes, including development, aging, immune regulation and cancer (1416). Previous studies suggest that oxidative stress and iron overload contribute to neuronal death in AD (17,18). Iron, an essential trace element for the human body, is involved in numerous physiological functions such as erythropoiesis, energy metabolism, muscle function and cell cycle regulation (19). Elevated iron levels in the gray matter of patients with AD have been documented (1), along with dysregulated iron homeostasis and lipid peroxidation, hallmarks of ferroptosis that are implicated in AD pathology (20). Therapeutic strategies targeting ferroptosis to prevent or mitigate organ damage have gained attention (21,22).

Ferroptosis

Ferroptosis typically results from disruptions in iron metabolism, lipid peroxidation and decreased glutathione (GSH) levels or inactivation of GSH peroxidase 4 (GPX4) (14). In ferroptotic cells, mitochondria are smaller, with ruffled and reduced cristae and membrane rupture, while non-ferroptotic cells exhibit swollen mitochondria. Vitamin E, ferrostatin-1 (Fer-1) and liproxstatin-1 (Lip-1) inhibit ferroptotic cell death without affecting other cell death pathways (23). The molecular mechanisms underlying ferroptosis primarily involve lipid, iron and amino acid metabolism (Fig. 1). Lipid metabolism is key for ferroptosis, which is driven by the accumulation of lipid peroxides resulting from the oxidation of polyunsaturated fatty acids (PUFAs) (24). Enzymes regulating lipid metabolism serve a key role in ferroptosis during lipid peroxidation. Acyl-CoA synthetase long-chain family member 4, a key enzyme in phospholipid metabolism, facilitates the conversion of PUFAs, such as arachidonoyl and adrenic acid, into PUFA-CoA (25). GSH, synthesized from glutamate, cysteine and glycine via glutamine cysteine ligase and GSH synthetase, serves as the primary antioxidant in mammalian cells. During cellular transport, glutamate and cystine are exchanged between cells through system Xc, which is key for GSH synthesis (26). Cysteine, due to its limited intracellular availability, is the rate-limiting precursor in GSH synthesis. System Xc, consisting of subunits solute carrier family 7 member 11 (SLC7A11) and SLC3A2, exports glutamate when GSH is consumed in excess (27). Disruptions in iron metabolism lead to pathological conditions, with transferrin-Fe3+ complex formation occurring when transferrin binds external Fe3+ on the cell membrane, which is subsequently internalized by transferrin receptor 1 (24). Divalent metal transporter 1 mediates release of Fe3+ ions from the six-transmembrane epithelial antigen of prostate 3 endosome into the cytosol (28). Ferritin releases Fe2+ ions via ferroportin 1. In ferroptosis, free Fe2+ interacts with hydrogen peroxide to generate highly reactive lipid peroxides, resulting from the disruption of the balance between ferrous iron absorption, depletion and recycling (29).

Ferroptosis and AD pathology

Amyloid (A)β plaques and neurofibrillary tangles (NFTs)

Aβ plaque accumulation is a hallmark pathological feature of AD, where abnormal Aβ aggregation disrupts synaptic function and impairs memory (30,31). Aβ, composed of 39–43 amino acids, is cleaved from amyloid precursor protein (APP) (32). APP is a highly conserved protein involved in synapse formation, dendritic growth and neuronal migration (33). Iron facilitates the dissociation of iron regulatory protein (IRP) 1 from the iron-responsive element (IRE) (34). Elevated intracellular iron levels disrupt the IRP/IRE signaling pathway, leading to increased expression of APP (35,36) and Aβ production. Additionally, Fe2+ binds to the N-terminal domain of Aβ, destabilizing its helical structure and promoting peptide aggregation by enhancing peptide-peptide interactions (37). Concurrently, τ hyperphosphorylation and its abnormal accumulation, coupled with impaired clearance, lead to the formation of NFTs, further compromising neuronal function. NFT formation is a key pathological hallmark of AD (3841). Dysregulated iron homeostasis has been linked to τ hyperphosphorylation and NFT development (42). In the cortex and hippocampus of patients with AD, NFTs accumulate in response to increased iron levels (43). Excessive neuronal iron promotes NFT formation via the activation of CDK5 (Cyclin-dependent kinase 5)/P25 complexes and GSK-3β (Glycogen synthase kinase-3 beta) kinase pathways. Fe3+ also induces hyperphosphorylated τ aggregation by binding to the histidine residues of τ (4446).

Microglia activation

Microglia are essential components of the central nervous system (CNS), serving key roles in energy metabolism, synaptic plasticity and ion homeostasis. In addition, microglia serve as resident immune cells, engaging in immune responses with memory-like behavior and maintaining brain homeostasis. During neurodegenerative processes, microglial activation is frequently observed, with increasing evidence suggesting that iron overload and disrupted iron homeostasis contribute to neurodegeneration in AD (47,48). As such, microglia serve a key role in neurological disorder. In response to infection or tissue injury, microglia rapidly adapt to the local environment, undergoing activation that can result in either beneficial or harmful outcomes. Kroner et al (49) demonstrated that elevated iron levels in microglia promote phagocytosis and drive a harmful M1 phenotype, which triggers the release of pro-inflammatory factors such as TNF-α, IL-1β, IL-6 and nitric oxide (NO), causing neuronal damage. Similarly, Rao et al (50) found that increased intracellular iron disrupts the neuromelanin-iron complex in neurons, releasing free iron ions that damage neurons and lead to neuromelanin leakage. This leakage further activates microglia towards the M1 phenotype, promoting release of neurotoxic agents, including TNF-α and IL-6 (50). Moreover, iron accumulation in activated microglia contributes to iron deposition within the CNS (51,52). In the M1 state, microglia express inducible NO synthase (iNOS), which converts arginine into NO. The resulting NO accumulation exacerbates glutamate-induced neurotoxicity, contributing to neuronal ferroptosis (53).

Oxidative stress and neuronal loss

Ferroptosis, an iron-dependent form of cell death distinct from apoptosis and necrosis, is primarily triggered by oxidative stress, a key pathological process in AD. The accumulation of reactive oxygen species (ROS), a hallmark of oxidative stress, serves a key role in initiating ferroptosis. Oxidative stress affects numerous molecular pathways, including inhibition of the cystine/glutamate antiporter system, decreased expression of GPX4, disruption of iron homeostasis and lipid peroxidation, which is a major driver of ferroptosis activation (54). Excess lipid peroxide accumulation in cells, a key feature of ferroptosis, results from a free radical chain reaction. Oxygen radicals insert into the C-H bonds of PUFAs, generating lipid hydroperoxides and elevating levels of ROS, which induce ferroptosis (55). Malondialdehyde, a byproduct of lipid peroxidation, is a marker for both ferroptosis and oxidative stress (56). Furthermore, oxidative stress impairs the antioxidant defense system by decreasing expression of key enzymes such as GSH, catalase, superoxide dismutase and GPX, thus accelerating ferroptosis (56,57). Neuronal loss, a defining characteristic of neurodegenerative disease, is associated with cognitive decline in AD (58). Elevated iron levels promote ROS production, depleting intracellular GSH levels and accelerating lipid peroxidation. This cascade leads to ferroptosis, contributing to neuronal death (59). Bao et al (60) found downregulation of the ferroptosis regulator GPX4 in both Fpnfl/fl/NEXcre (NEX-Cre mice were mated with Fpn-floxed (Fpnfl/fl) mice to generate conditional Fpnfl/fl/NEXcre mice.) and APPswe/PS1dE9 (Carrying genetically modified mice with AD-related mutations: a chimeric mouse/human APP with the Swedish mutation and human PSEN1 lacking exon 9) mouse models compared with controls. Additionally, mRNA expression of iron response element binding protein 2, encodes a master regulator of iron metabolism), and CS (citrate synthase, regulating the mitochondrial fatty acid metabolism) was upregulated in both models, while ACSF2 ((acyl-CoA synthetase family member 2, regulating the mitochondrial fatty acid metabolism) was upregulated only in APPswe/PS1dE9 mice. These findings suggest that ferroptosis is activated in the hippocampus of both mouse models.

Pathways regulating ferroptosis in AD

Kelch-like ECH-associated protein 1 (Keap1)/Nrf2)/antioxidant response element (ARE) pathway

In cellular defense against oxidative stress, the Keap1/Nrf2/ARE signaling pathway regulates the expression of various proteins involved in detoxification and antioxidant defense, positioning it as a potential target for AD treatment (61). Nrf2, a transcription factor that is highly responsive to oxidative stress, serves a key role in mitigating lipid peroxidation and ferroptosis (62). Under physiological conditions, Keap1 suppresses Nrf2 by facilitating its ubiquitination and degradation via the ubiquitin-proteasome system. By contrast, during oxidative stress, Nrf2 dissociates from Keap1, translocates to the nucleus, forms a heterodimer with small musculoaponeurotic fibrosarcoma oncogene homolog proteins and binds to ARE, thereby enhancing the transcription of antioxidant genes (6365). Nrf2 regulates key components of anti-ferroptotic pathways, positioning it as a central modulator of lipid peroxidation and ferroptosis (62). In the nucleus, Nrf2 induces the expression of cytoprotective genes that mitigate ferroptosis by regulating iron metabolism and enhancing antioxidant defenses. This includes the upregulation of ferritin heavy and light chain, ferroportin, transferrin receptor and heme oxygenase-1 (HO-1), alongside increased production of NADPH, GSH and CoQ10 (coenzyme Q10) which counter lipid peroxidation and suppress ferroptosis (66,67). Moreover, the detachment of the DLG motif of Nrf2 from Keap1 prevents its ubiquitination and degradation, thus strengthening antioxidant defenses and inhibiting ferroptosis (68).

p53/SLC7A11 pathway

This process is initiated when unsaturated FAs in cell membranes undergo catalytic lipid peroxidation, driven by divalent iron or esteroxygenases, leading to cell death (6971). The p53 protein, a key human tumor suppressor, regulates the expression of oncogenes and downstream signaling pathways, contributing to biological effects (7275): Beyond its role in cancer, p53 is highly expressed in the brain, where it influences dendritic growth, oxidative stress response, apoptosis and autophagy, making p53 dysfunction and associated pathways noteworthy in the pathogenesis of AD (7678). SLC7A11, a key ferroptosis regulator, is a transmembrane protein that is part of the system Xc, responsible for cystine import into cells for cysteine synthesis and GSH production (79,80). Downregulation of SLC7A11 disrupts cysteine metabolism, leading to decreased intracellular cystine and GSH levels, impairing GPX4 activity and triggering lipid peroxide accumulation and ferroptosis (79,81,82). Studies indicate that p53 binds the SLC7A11 promoter, suppressing its expression and limiting GSH production, thereby promoting ferroptosis (83,84). Aristolochic acid, mediated by p53, may limit ferroptosis in liver cancer to enhance tumor growth. The p53(3KR) mutant, lacking acetylation due to lysine-to-arginine substitutions at three residues, decreases expression of SLC7A11 without affecting other p53 targets such as CDKN1A/p21 (involved in cell cycle progression) or BAX (involved in apoptosis). By contrast, the p53(4KR98) mutant, with an additional lysine 98 substitution, does not downregulate SLC7A11 (8587).

GSH/GPX4 pathway

In AD, excessive lipid peroxide accumulation is a key initiator of ferroptosis, with elevated markers of lipid peroxidation observed in neurons. Iron accumulation drives the Fenton and Haber-Weiss reactions, generating ROS that induce lipid peroxidation, leading to oxidative damage to subcellular structures (88). GPX4 and GSH are key regulators of ferroptosis. GSH, containing a thiol group derived from cysteine, serves as a vital antioxidant, neutralizing ROS and reactive nitrogen species, maintaining cellular redox balance and detoxifying xenobiotics (89). GPX4, a selenium (Se)-dependent enzyme, relies on a Se-containing amino acid residue to execute its reductive function (90). It converts lipid hydroperoxides into less harmful lipid alcohols, preventing oxidative damage (91,92). The active site of GPX4, selenocysteine, alternates between reduced and oxidized states to sustain its catalytic activity. In the presence of peroxides, the selenolate form of GPX4 is oxidized to selenic acid, which is regenerated to its active form by two molecules of reduced GSH, converting lipid hydroperoxides into non-toxic lipid alcohols and generating oxidized GSH (16,9294). This highlights the key role of GPX4 synthesis and its associated pathways in the regulation of ferroptosis.

Ferroptosis inhibitors and AD

Given the mechanisms underlying ferroptosis in AD, researchers have focused on its inhibition as a potential therapeutic strategy. Growing evidence suggests that targeting ferroptosis may offer benefits for CNS disorders, driving the development of effective inhibitors (24,95). This approach presents promising therapeutic opportunities for AD management (Table I; Fig. 2). As AD is characterized by iron accumulation in brain cells, exacerbating oxidative damage and cognitive decline (96100), these inhibitors show therapeutic promise. Vitamin E, Se, Fer-1, N-acetylcysteine (NAC) and curcumin exhibit antioxidant and neuroprotective properties (101131).

Table I.

Ferroptosis inhibitors and their mechanisms.

Table I.

Ferroptosis inhibitors and their mechanisms.

First author/s, yearSubjectMechanism(Refs.)
Hinman et al, 2018Vitamin EInhibits ferroptosis by inhibiting lipid peroxidation(106)
Ashram et al, 2020 (107)
Gugliandolo et al, 2017 (101)
Kryscio et al, 2017 (108)
Conrad et al, 2020SeleniumAntioxidant activity; inhibits ferroptosis by upregulating GPX4(109)
Alim et al, 2019 (102)
Ingold et al, 2018 (110)
Cardoso et al, 2017 (111)
Dixon et al, 2012Ferrostatin-1Prevents the accumulation of lipid reactive oxygen species, inhibits oxidative stress and decreases lipid peroxidation(27)
Skouta et al, 2014 (112)
Li et al, 2017 (103)
Fang et al, 2019 (113)
Miotto et al, 2020 (114)
Asano et al, 2017 (115)
Kalyanaraman et al, 2022 N-acetylcysteineActivates Nrf2; controls the expression of metallothionein, ferritin and iron transporter(116)
Fan et al, 2017 (117)
Kerins et al, 2018 (118)
Rojo de la Vega et al, 2018 (119)
Hara et al, 2017 (120)
Pocernich et al, 2000 (121)
Koppal et al, 1999 (122)
Pocernich et al, 2001 (123)
Dodson et al, 2019CurcuminDecreases expression levels of GPX4 and increase levels of HO-1 and Nrf2(62)
Prasad et al, 2014 (124)
Wei et al, 2022 (125)
Hirata et al, 2019 (126)
Ikawa et al, 2021 (104)
Hirata et al, 2021 (127)
Hirata et al, 2022 (128)
Friedmann Angeli et al, 2014Liproxstatin-1Decreases oxidative stress and blocks lipid peroxidation(129)
Li et al, 2022 (130)
Fan et al, 2021 (131)

[i] GPX4, glutathione GSH peroxidase 4; HO-1, heme oxygenase-1.

Vitamin E

Vitamin E, an antioxidant in AD treatment, inhibits ferroptosis primarily by preventing lipid peroxidation (101). It consists of two subclasses: Tocotrienols and tocopherols, with four saturated analogs (α, β, γ and δ) (132,133). These lipophilic compounds, along with their derivatives, serve as radical-trapping antioxidants, preventing the formation of phospholipid hydroperoxides. As the dominant form in tissue, α-tocopherol exerts its antioxidant effects by interrupting the chain reaction of lipid oxidation. Specifically, the oxidative conversion of α-tocopherol produces α-tocopherol quinone, which is reduced to α-tocopherol hydroquinone. This reduction is key for inhibiting the enzymatic activity of 15-lipoxygenase, primarily by converting its non-heme Fe3+ to an inactive Fe2+ state. This inhibition disrupts the ferroptotic signaling cascade, effectively preventing lipid peroxidation and mitigating ferroptosis (106). α-tocopherol transfer protein (TTP), which is abundant in the brain, regulates α-tocopherol levels and distribution (107). Vitamin E or TTP deficiency induce oxidative stress in the brain. Studies show that patients with AD exhibit reduced vitamin E levels in plasma, serum and cerebrospinal fluid, and those receiving vitamin E supplementation experience slower cognitive decline and decreased oxidative stress compared with placebo-treated individuals (101,108). These findings suggest that vitamin E deficiency contributes to neurodegeneration, while supplementation may offer protection against ferroptotic stress.

Se

Se, an essential trace element involved in GPX4 synthesis, is incorporated into several proteins in the body (109). Known for its antioxidant properties, Se also serves a role in inhibiting ferroptosis (102,110). Clinical studies suggest Se may have potential in mitigating cognitive decline (111,134). In a mouse model of stroke, intracerebroventricular sodium selenate treatment elevates GPX4 levels by activating transcription factors activating enhancer binding protein 2γ and specificity protein 1, while also providing protection against excitotoxicity and endoplasmic reticulum stress-induced cell death independent of GPX4 (102). Decreased Se levels in the brains of patients with AD are associated with disease progression. In primary neuronal cultures, Se was shown to decrease Aβ production by downregulating 4-hydroxy-2-nonenal-induced β-secretase transcription, thus preventing Aβ-associated toxicity (102). Se-containing compounds inhibit ferroptosis by upregulating GPX4. A clinical trial demonstrated that oral sodium selenate supplementation increases brain Se levels without notable side effects, and participants with improved Se levels do not exhibit worsening Mini-Mental State Examination scores over time (108). However, Kryscio et al (108) revealed that both Se and vitamin E have adverse effects on progression of AD in male patients. Thus, current evidence does not definitively support a potential therapeutic role of Se in AD, and further clinical trials are needed to clarify its effects.

Fer-1

Fer-1, the first synthetic ferroptosis inhibitor, has served as a pivotal reference compound (27). It effectively prevents oxidative lipid damage and cell death in disease models, including Huntington's disease, periventricular leukomalacia, renal dysfunction, cerebral hemorrhage and cardiomyopathy (103,112,113). As a highly specific ferroptosis inhibitor, Fer-1 surpasses phenolic antioxidants in its ability to suppress ferroptosis, particularly by inhibiting lipid ROS accumulation induced by erastin or RSL3 in HT-1080 human fibrosarcoma cells) (27). The anti-ferroptotic action of Fer-1 primarily arises from its capacity to scavenge alkoxyl radicals and reactive species generated by ferrous iron in lipid hydroperoxides. Additionally, it reduces labile iron, as confirmed by calcein fluorescence assays, further supporting its iron-complexing properties (114). In HT-22 (mouse hippocampal neurons cells) cells, Fer-1 notably decreases both cytoplasmic and lipid ROS levels, counteracts glutamate-induced suppression of GSH and GPX activity and protects against oxidative toxicity. Furthermore, Fer-1 mitigates oxidative stress by downregulating prostaglandin endoperoxide synthase 2 while upregulating GPX4 and Nrf2, thereby decreasing lipid peroxidation (115). Despite its documented efficacy in vitro and in vivo in alleviating oxidative stress and preventing iron deficiency, Fer-1 remains untested in clinical trials (114). Further investigation is required to assess its therapeutic potential, safety and clinical effectiveness for treating AD.

NAC

NAC, a cysteine precursor, is used to treat acetaminophen overdose and is listed as an essential medicine by the World Health Organization (116). In addition to its role in treating acute poisoning, NAC is recognized for its pro-neurogenic and neuroprotective effects in neurodegenerative and psychiatric disorder (120). NAC can cross the blood-brain barrier and mitigate age-associated memory decline (117,120). NAC may prevent ferroptosis by activating Nrf2, which regulates the expression of metallothioneins, ferritins and ferroportins, thereby preventing iron accumulation. Other Nrf2-dependent genes, including GPX4, GSH and NADPH synthesis genes, as well as epigenetic regulators of lipid hydroperoxides, contribute to its ferroptosis-inhibitory effects (117119). In AD, NAC has potential as a GSH precursor, enhancing antioxidant defenses (120). Rat and gerbil studies have demonstrated that intraperitoneal NAC administration protects against oxidative damage induced by acrolein, peroxynitrite, hydroxyl radicals and 3-nitropropionic acid while increasing GSH levels in the brain and synaptosomes (119123).

Curcumin

Curcumin, a polyphenolic compound initially identified as diferuloylmethane in turmeric rhizomes, has demonstrated protective effects against oxidative damage resulting from iron accumulation in cells (124). Curcumin treatment has been shown to decrease expression of GPX4 while increasing levels of heme-oxygenase (HO-1) and Nrf2 (124). As a key regulator of oxidative stress, Nrf2 governs the expression of ARE-dependent genes (62,125). Hirata et al (126) developed hybrid molecules that combine the oxidized indole structure of neuroprotective agents with the polyphenolic structure of curcumin, applying them to mouse hippocampal HT22 cells. These hybrids exhibited superior neuroprotective effects and lower cytotoxicity compared with curcumin alone, activated ARE, chelated ferrous ions and scavenged ROS, thereby protecting cells from oxidative stress and ferroptosis, ultimately promoting neuronal survival (104,127,128).

Lip-1

Lip-1, a derivative of quinoxaline spirocyclic compounds, is a potent ferroptosis inhibitor first identified in 2014 through small molecule compound library screening (129). Lip-1 primarily exerts its effects by inhibiting lipid peroxidation. Li et al (130) demonstrated that Lip-1 alleviates memory deficits in a mouse model of lipopolysaccharide (LPS)-induced cognitive impairment. Moreover, Lip-1 reduced microglial activation and the secretion of pro-inflammatory cytokines such as IL-6 and TNF-α, while mitigating oxidative stress, lipid peroxidation, mitochondrial damage and neuronal injury following LPS exposure. Further studies have revealed that Lip-1 not only prevents mitochondrial lipid peroxidation but also restores the expression of key molecules involved in ferroptosis regulation, including GSH, GPX4 and ferroptosis suppressor protein 1 (135,136). These findings suggest that GPX4 inhibition may induce ferroptosis in oligodendrocytes, with Lip-1 serving as a potent ferroptosis antagonist. Therefore, Lip-1 holds promise as a therapeutic candidate for CNS disease (131).

Conclusion

Research on iron-lowering strategies in AD has been limited, likely due to the predominant focus on amyloid-lowering treatments, which have generally yielded unfavorable results (137,138). Nevertheless, studies involving iron chelators in cell and animal models of AD have demonstrated promising outcomes (95,139). While iron metabolism and lipid peroxidation trigger ferroptosis under pathological conditions, the precise mechanisms remain incompletely understood and warrant further investigation. Additionally, iron chelating agents, including chloroiodohydroxyquine and its derivatives, as well as antioxidants, have shown efficacy in animal models of AD, though clinical trials are yet to be conducted (27,106,116). Ferroptosis may serve as a key target pathway for advancing AD treatment strategies.

In conclusion, the present review summarized the role of ferroptosis in the pathology of AD and how mechanisms such as iron metabolism disorders, lipid peroxidation and GSH depletion lead to neuronal damage, as well as the role of ferroptosis inhibitors such as vitamin E, Se and Fer-1 as potential therapeutic strategies. Keap1/Nrf2/ARE, p53/SLC7A11 and GSH/GPX4 signaling pathways underlie the pathological mechanism of AD, positioning them as a potential direction for future research.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

HZ and ZJ wrote and reviewed the manuscript. All authors have read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Zhao D, Yang K, Guo H, Zeng J, Wang S, Xu H, Ge A, Zeng L, Chen S and Ge J: Mechanisms of ferroptosis in Alzheimer's disease and therapeutic effects of natural plant products: A review. Biomed Pharmacother. 164:1143122023. View Article : Google Scholar : PubMed/NCBI

2 

Graff-Radford J, Yong KXX, Apostolova LG, Bouwman FH, Carrillo M, Dickerson BC, Rabinovici GD, Schott JM, Jones DT and Murray ME: New insights into atypical Alzheimer's disease in the era of biomarkers. Lancet Neurol. 20:222–234. 2021. View Article : Google Scholar : PubMed/NCBI

3 

Wang Q, Sun J, Chen T, Song S, Hou Y, Feng L, Fan C and Li M: Ferroptosis, pyroptosis, and cuproptosis in Alzheimer's disease. ACS Chem Neurosci. 14:3564–3587. 2023. View Article : Google Scholar : PubMed/NCBI

4 

Lane DJR, Metselaar B, Greenough M, Bush AI and Ayton SJ: Ferroptosis and NRF2: An emerging battlefield in the neurodegeneration of Alzheimer's disease. Essays Biochem. 65:925–940. 2021. View Article : Google Scholar : PubMed/NCBI

5 

Huang Q, Wu W, Wen Y, Lu S and Zhao C: Potential therapeutic natural compounds for the treatment of Alzheimer's disease. Phytomedicine. 132:1558222024. View Article : Google Scholar : PubMed/NCBI

6 

Nayak V, Patra S, Rout S, Jena AB, Sharma R, Pattanaik KP, Singh J, Pandey SS, Singh RP, Majhi S, et al: Regulation of neuroinflammation in Alzheimer's disease via nanoparticle-loaded phytocompounds with anti-inflammatory and autophagy-inducing properties. Phytomedicine. 122:1551502024. View Article : Google Scholar : PubMed/NCBI

7 

Weintraub D, Aarsland D, Chaudhuri KR, Dobkin RD, Leentjens AF, Rodriguez-Violante M and Schrag A: The neuropsychiatry of Parkinson's disease: Advances and challenges. Lancet Neurol. 21:89–102. 2022. View Article : Google Scholar : PubMed/NCBI

8 

Markus HS, van Der Flier WM, Smith EE, Bath P, Biessels GJ, Briceno E, Brodtman A, Chabriat H, Chen C, de Leeuw FE, et al: Framework for clinical trials in cerebral small vessel disease (FINESSE): A Review. JAMA Neurol. 79:1187–1198. 2022. View Article : Google Scholar : PubMed/NCBI

9 

Liu JB, Wang X and Cao J: The coherence and properties analysis of balanced 2 p-Ary tree networks. IEEE Trans Netw Sci Eng. 11:4719–4728. 2024. View Article : Google Scholar

10 

Liu JB, Zhang X, Cao J and Chen L: Mean first-passage time and robustness of complex cellular mobile communication network. IEEE Trans Netw Sci Eng. 11:3066–3076. 2024. View Article : Google Scholar

11 

Ashraf T, Idrees N and Belay MB: Regression analysis of topological indices for predicting efficacy of Alzheimer's drugs. PLoS One. 19:e03094772024. View Article : Google Scholar : PubMed/NCBI

12 

Wang Y, Song X, Wang R, Xu X, Du Y, Chen G and Mei J: Genome-wide mendelian randomization identifies ferroptosis-related drug targets for Alzheimer's disease. J Alzheimers Dis Rep. 8:1185–1197. 2024. View Article : Google Scholar : PubMed/NCBI

13 

Soni P, Ammal Kaidery N, Sharma SM, Gazaryan I, Nikulin SV, Hushpulian DM and Thomas B: A critical appraisal of ferroptosis in Alzheimer's and Parkinson's disease: New insights into emerging mechanisms and therapeutic targets. Front Pharmacol. 15:13907982024. View Article : Google Scholar : PubMed/NCBI

14 

Jiang X, Stockwell BR and Conrad M: Ferroptosis: Mechanisms, biology and role in disease. Nat Rev Mol Cell Biol. 22:266–282. 2021. View Article : Google Scholar : PubMed/NCBI

15 

Li J, Cao F, Yin HL, Huang ZJ, Lin ZT, Mao N, Sun B and Wang G: Ferroptosis: Past, present and future. Cell Death Dis. 11:882020. View Article : Google Scholar : PubMed/NCBI

16 

Tang D, Chen X, Kang R and Kroemer G: Ferroptosis: Molecular mechanisms and health implications. Cell Res. 31:107–125. 2021. View Article : Google Scholar : PubMed/NCBI

17 

Lei P, Ayton S and Bush AI: The essential elements of Alzheimer's disease. J Biol Chem. 296:1001052021. View Article : Google Scholar : PubMed/NCBI

18 

Yong YY, Yan L, Wang BD, Fan DS, Guo MS, Yu L, Wu JM, Qin DL, Law BY, Wong VK, et al: Penthorum chinense Pursh inhibits ferroptosis in cellular and Caenorhabditis elegans models of Alzheimer's disease. Phytomedicine. 127:1554632024. View Article : Google Scholar : PubMed/NCBI

19 

Andrews NC: Disorders of iron metabolism. N Engl J Med. 341:1986–1995. 1999. View Article : Google Scholar : PubMed/NCBI

20 

Buijs M, Doan NT, van Rooden S, Versluis MJ, van Lew B, Milles J, van der Grond J and van Buchem MA: In vivo assessment of iron content of the cerebral cortex in healthy aging using 7-Tesla T2*-weighted phase imaging. Neurobiol Aging. 53:20–26. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Chu J, Li J, Sun L and Wei J: The role of cellular defense systems of ferroptosis in Parkinson's disease and Alzheimer's disease. Int J Mol Sci. 24:141082023. View Article : Google Scholar : PubMed/NCBI

22 

Yu J and Wang JQ: Research mechanisms of and pharmaceutical treatments for ferroptosis in liver diseases. Biochimie. 180:149–157. 2021. View Article : Google Scholar : PubMed/NCBI

23 

Ajoolabady A, Aslkhodapasandhokmabad H, Libby P, Tuomilehto J, Lip GYH, Penninger JM, Richardson DR, Tang D, Zhou H, Wang S, et al: Ferritinophagy and ferroptosis in the management of metabolic diseases. Trends Endocrinol Metab. 32:444–462. 2021. View Article : Google Scholar : PubMed/NCBI

24 

Jakaria M, Belaidi AA, Bush AI and Ayton S: Ferroptosis as a mechanism of neurodegeneration in Alzheimer's disease. J Neurochem. 159:804–825. 2021. View Article : Google Scholar : PubMed/NCBI

25 

Sun Y, Chen P, Zhai B, Zhang M, Xiang Y, Fang J, Xu S, Gao Y, Chen X, Sui X and Li G: The emerging role of ferroptosis in inflammation. Biomed Pharmacother. 127:1101082020. View Article : Google Scholar : PubMed/NCBI

26 

Couto N, Wood J and Barber J: The role of glutathione reductase and related enzymes on cellular redox homoeostasis network. Free Radic Biol Med. 95:27–42. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS, et al: Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell. 149:1060–1072. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Hu H, Chen Y, Jing L, Zhai C and Shen L: The link between ferroptosis and cardiovascular diseases: A novel target for treatment. Front Cardiovasc Med. 8:7109632021. View Article : Google Scholar : PubMed/NCBI

29 

Bu ZQ, Yu HY, Wang J, He X, Cui YR, Feng JC and Feng J: Emerging role of ferroptosis in the pathogenesis of ischemic stroke: A new therapeutic target? ASN Neuro. 13:175909142110375052021. View Article : Google Scholar : PubMed/NCBI

30 

Foley KE and Wilcock DM: Vascular considerations for amyloid immunotherapy. Curr Neurol Neurosci Rep. 22:709–719. 2022. View Article : Google Scholar : PubMed/NCBI

31 

Liu Y, Chen Z, Li B, Yao H, Zarka M, Welch J, Sachdev P, Bridge W and Braidy N: Supplementation with γ-glutamylcysteine (γ-GC) lessens oxidative stress, brain inflammation and amyloid pathology and improves spatial memory in a murine model of AD. Neurochem Int. 144:1049312021. View Article : Google Scholar : PubMed/NCBI

32 

Chen LL, Fan YG, Zhao LX, Zhang Q and Wang ZY: The metal ion hypothesis of Alzheimer's disease and the anti-neuroinflammatory effect of metal chelators. Bioorg Chem. 131:1063012023. View Article : Google Scholar : PubMed/NCBI

33 

Müller UC, Deller T and Korte M: Not just amyloid: physiological functions of the amyloid precursor protein family. Nat Rev Neurosci. 18:281–298. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Zhou ZD and Tan EK: Iron regulatory protein (IRP)-iron responsive element (IRE) signaling pathway in human neurodegenerative diseases. Mol Neurodegener. 12:752017. View Article : Google Scholar : PubMed/NCBI

35 

Goel P, Chakrabarti S, Goel K, Bhutani K, Chopra T and Bali S: Neuronal cell death mechanisms in Alzheimer's disease: An insight. Front Mol Neurosci. 15:9371332022. View Article : Google Scholar : PubMed/NCBI

36 

Wang J, Fu J, Zhao Y, Liu Q, Yan X and Su J: Iron and targeted iron therapy in Alzheimer's disease. Int J Mol Sci. 24:163532023. View Article : Google Scholar : PubMed/NCBI

37 

Boopathi S and Kolandaivel P: Fe2+ binding on amyloid β-peptide promotes aggregation. Proteins. 84:1257–1274. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Faraji P, Kühn H and Ahmadian S: Multiple roles of apolipoprotein E4 in oxidative lipid metabolism and ferroptosis during the pathogenesis of Alzheimer's disease. J Mol Neurosci. 74:622024. View Article : Google Scholar : PubMed/NCBI

39 

Gao Y and Tan L, Yu JT and Tan L: Tau in Alzheimer's disease: Mechanisms and therapeutic strategies. Curr Alzheimer Res. 15:283–300. 2018. View Article : Google Scholar : PubMed/NCBI

40 

Sinsky J, Pichlerova K and Hanes J: Tau protein interaction partners and their roles in Alzheimer's disease and other tauopathies. Int J Mol Sci. 22:92072021. View Article : Google Scholar : PubMed/NCBI

41 

Cody KA, Langhough RE, Zammit MD, Clark L, Chin N, Christian BT, Betthauser TJ and Johnson SC: Characterizing brain tau and cognitive decline along the amyloid timeline in Alzheimer's disease. Brain. 147:2144–2157. 2024. View Article : Google Scholar : PubMed/NCBI

42 

Wang F, Wang J, Shen Y, Li H, Rausch WD and Huang X: Iron dyshomeostasis and ferroptosis: A new Alzheimer's disease hypothesis? Front Aging Neurosci. 14:8305692022. View Article : Google Scholar : PubMed/NCBI

43 

Spotorno N, Acosta-Cabronero J, Stomrud E, Lampinen B, Strandberg OT, van Westen D and Hansson O: Relationship between cortical iron and tau aggregation in Alzheimer's disease. Brain. 143:1341–1349. 2020. View Article : Google Scholar : PubMed/NCBI

44 

Guo C, Wang P, Zhong ML, Wang T, Huang XS, Li JY and Wang ZY: Deferoxamine inhibits iron induced hippocampal tau phosphorylation in the Alzheimer transgenic mouse brain. Neurochem Int. 62:165–172. 2013. View Article : Google Scholar : PubMed/NCBI

45 

Vossel KA, Xu JC, Fomenko V, Miyamoto T, Suberbielle E, Knox JA, Ho K, Kim DH, Yu GQ and Mucke L: Tau reduction prevents Aβ-induced axonal transport deficits by blocking activation of GSK3β. J Cell Biol. 209:419–433. 2015. View Article : Google Scholar : PubMed/NCBI

46 

Kim AC, Lim S and Kim YK: Metal ion effects on Aβ and tau aggregation. Int J Mol Sci. 19:1282018. View Article : Google Scholar : PubMed/NCBI

47 

Mills E, Dong XP, Wang F and Xu H: Mechanisms of brain iron transport: Insight into neurodegeneration and CNS disorders. Future Med Chem. 2:51–64. 2010. View Article : Google Scholar : PubMed/NCBI

48 

Masaldan S, Bush AI, Devos D, Rolland AS and Moreau C: Striking while the iron is hot: Iron metabolism and ferroptosis in neurodegeneration. Free Radic Biol Med. 133:221–233. 2019. View Article : Google Scholar : PubMed/NCBI

49 

Kroner A, Greenhalgh AD, Zarruk JG, Passos Dos Santos R, Gaestel M and David S: TNF and increased intracellular iron alter macrophage polarization to a detrimental M1 phenotype in the injured spinal cord. Neuron. 83:1098–1116. 2014. View Article : Google Scholar : PubMed/NCBI

50 

Rao KS, Hegde ML, Anitha S, Musicco M, Zucca FA, Turro NJ and Zecca L: Amyloid β and neuromelanin-toxic or protective molecules?: The cellular context makes the difference. Prog Neurobiol. 78:364–373. 2006. View Article : Google Scholar : PubMed/NCBI

51 

Guo JJ, Yue F, Song DY, Bousset L, Liang X, Tang J, Yuan L, Li W, Melki R, Tang Y, et al: Intranasal administration of α-synuclein preformed fibrils triggers microglial iron deposition in the substantia nigra of Macaca fascicularis. Cell Death Dis. 12:812021. View Article : Google Scholar : PubMed/NCBI

52 

Kenkhuis B, Somarakis A, de Haan L, Dzyubachyk O, IJsselsteijn ME, de Miranda NFCC, Lelieveldt BPF, Dijkstra J, van Roon-Mom WMC, Höllt T and van der Weerd L: Iron loading is a prominent feature of activated microglia in Alzheimer's disease patients. Acta Neuropathol Commun. 9:272021. View Article : Google Scholar : PubMed/NCBI

53 

Wang M, Tang G, Zhou C, Guo H, Hu Z, Hu Q and Li G: Revisiting the intersection of microglial activation and neuroinflammation in Alzheimer's disease from the perspective of ferroptosis. Chem Biol Interact. 375:1103872023. View Article : Google Scholar : PubMed/NCBI

54 

Li S, Wen P, Zhang D, Li D, Gao Q, Liu H and Di Y: PGAM5 expression levels in heart failure and protection ROS-induced oxidative stress and ferroptosis by Keap1/Nrf2. Clin Exp Hypertens. 45:21625372023. View Article : Google Scholar : PubMed/NCBI

55 

Conrad M, Kagan VE, Bayir H, Pagnussat GC, Head B, Traber MG and Stockwell BR: Regulation of lipid peroxidation and ferroptosis in diverse species. Genes Dev. 32:602–619. 2018. View Article : Google Scholar : PubMed/NCBI

56 

Ayala A, Muñoz MF and Argüelles S: Lipid peroxidation: Production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. Oxid Med Cell Longev. 2014:3604382014. View Article : Google Scholar : PubMed/NCBI

57 

Endale HT, Tesfaye W and Mengstie TA: ROS induced lipid peroxidation and their role in ferroptosis. Front Cell Dev Biol. 11:12260442023. View Article : Google Scholar : PubMed/NCBI

58 

Hanseeuw BJ, Betensky RA, Jacobs HI, Schultz AP, Sepulcre J, Becker JA, Cosio DMO, Farrell M, Quiroz YT, Mormino EC, et al: Association of amyloid and tau with cognition in preclinical Alzheimer disease: A longitudinal study. JAMA Neurol. 76:915–924. 2019. View Article : Google Scholar : PubMed/NCBI

59 

Maher P: Potentiation of glutathione loss and nerve cell death by the transition metals iron and copper: Implications for age-related neurodegenerative diseases. Free Radic Biol Med. 115:92–104. 2018. View Article : Google Scholar : PubMed/NCBI

60 

Bao WD, Pang P, Zhou XT, Hu F, Xiong W, Chen K, Wang J, Wang F, Xie D, Hu YZ, et al: Loss of ferroportin induces memory impairment by promoting ferroptosis in Alzheimer's disease. Cell Death Differ. 28:1548–1562. 2021. View Article : Google Scholar : PubMed/NCBI

61 

Tu W, Wang H, Li S, Liu Q and Sha H: The anti-inflammatory and anti-oxidant mechanisms of the Keap1/Nrf2/ARE signaling pathway in chronic diseases. Aging Dis. 10:637–651. 2019. View Article : Google Scholar : PubMed/NCBI

62 

Dodson M, Castro-Portuguez R and Zhang DD: NRF2 plays a critical role in mitigating lipid peroxidation and ferroptosis. Redox Biol. 23:1011072019. View Article : Google Scholar : PubMed/NCBI

63 

Ma Q: Role of nrf2 in oxidative stress and toxicity. Annu Rev Pharmacol Toxicol. 53:401–426. 2013. View Article : Google Scholar : PubMed/NCBI

64 

Baird L, Swift S, Llères D and Dinkova-Kostova AT: Monitoring Keap1-Nrf2 interactions in single live cells. Biotechnol Adv. 32:1133–1144. 2014. View Article : Google Scholar : PubMed/NCBI

65 

Kumar A and Mittal R: Nrf2: A potential therapeutic target for diabetic neuropathy. Inflammopharmacology. 25:393–402. 2017. View Article : Google Scholar : PubMed/NCBI

66 

Sun Y, Xia X, Basnet D, Zheng JC, Huang J and Liu J: Mechanisms of ferroptosis and emerging links to the pathology of neurodegenerative diseases. Front Aging Neurosci. 14:9041522022. View Article : Google Scholar : PubMed/NCBI

67 

Shin CS, Mishra P, Watrous JD, Carelli V, D'Aurelio M, Jain M and Chan DC: The glutamate/cystine xCT antiporter antagonizes glutamine metabolism and reduces nutrient flexibility. Nat Commun. 8:150742017. View Article : Google Scholar : PubMed/NCBI

68 

Bellezza I, Giambanco I, Minelli A and Donato R: Nrf2-Keap1 signaling in oxidative and reductive stress. Biochim Biophys Acta Mol Cell Res. 1865:721–733. 2018. View Article : Google Scholar : PubMed/NCBI

69 

Liang D, Minikes AM and Jiang X: Ferroptosis at the intersection of lipid metabolism and cellular signaling. Mol Cell. 82:2215–2227. 2022. View Article : Google Scholar : PubMed/NCBI

70 

Ursini F and Maiorino M: Lipid peroxidation and ferroptosis: The role of GSH and GPx4. Free Radic Biol Med. 152:175–185. 2020. View Article : Google Scholar : PubMed/NCBI

71 

Pope LE and Dixon SJ: Regulation of ferroptosis by lipid metabolism. Trends Cell Biol. 33:1077–1087. 2023. View Article : Google Scholar : PubMed/NCBI

72 

Finlay CA, Hinds PW and Levine AJ: The 53 proto-oncogene can act as a suppressor of transformation. Cell. 57:1083–1093. 1989. View Article : Google Scholar : PubMed/NCBI

73 

Kenzelmann Broz D and Attardi LD: In vivo analysis of p53 tumor suppressor function using genetically engineered mouse models. Carcinogenesis. 31:1311–1318. 2010. View Article : Google Scholar : PubMed/NCBI

74 

Kamada R, Toguchi Y, Nomura T, Imagawa T and Sakaguchi K: Tetramer formation of tumor suppressor protein p53: Structure, function, and applications. Biopolymers. 106:598–612. 2016. View Article : Google Scholar : PubMed/NCBI

75 

Joerger AC and Fersht AR: Structural biology of the tumor suppressor p53. Annu Rev Biochem. 77:557–582. 2008. View Article : Google Scholar : PubMed/NCBI

76 

Li H, Zhang Z, Li H, Pan X and Wang Y: New insights into the roles of p53 in central nervous system diseases. Int J Neuropsychopharmacol. 26:465–473. 2023. View Article : Google Scholar : PubMed/NCBI

77 

Ohyagi Y, Asahara H, Chui DH, Tsuruta Y, Sakae N, Miyoshi K, Yamada T, Kikuchi H, Taniwaki T, Murai H, et al: Intracellular Abeta42 activates p53 promoter: A pathway to neurodegeneration in Alzheimer's disease. FASEB J. 19:255–257. 2005. View Article : Google Scholar : PubMed/NCBI

78 

Masaldan S, Belaidi AA, Ayton S and Bush AI: Cellular senescence and iron dyshomeostasis in Alzheimer's disease. Pharmaceuticals (Basel). 12:932019. View Article : Google Scholar : PubMed/NCBI

79 

Wang C, Liu H, Xu S, Deng Y, Xu B, Yang T and Liu W: Ferroptosis and neurodegenerative diseases: Insights into the regulatory roles of SLC7A11. Cell Mol Neurobiol. 43:2627–2642. 2023. View Article : Google Scholar : PubMed/NCBI

80 

Lee J and Roh JL: SLC7A11 as a gateway of metabolic perturbation and ferroptosis vulnerability in cancer. Antioxidants (Basel). 11:24442022. View Article : Google Scholar : PubMed/NCBI

81 

Koppula P, Zhuang L and Gan B: Cystine transporter SLC7A11/xCT in cancer: Ferroptosis, nutrient dependency, and cancer therapy. Protein Cell. 12:599–620. 2021. View Article : Google Scholar : PubMed/NCBI

82 

Iida Y, Okamoto-Katsuyama M, Maruoka S, Mizumura K, Shimizu T, Shikano S, Hikichi M, Takahashi M, Tsuya K, Okamoto S, et al: Effective ferroptotic small-cell lung cancer cell death from SLC7A11 inhibition by sulforaphane. Oncol Lett. 21:712021. View Article : Google Scholar : PubMed/NCBI

83 

Shin D, Lee J and Roh JL: Pioneering the future of cancer therapy: Deciphering the p53-ferroptosis nexus for precision medicine. Cancer Lett. 585:2166452024. View Article : Google Scholar : PubMed/NCBI

84 

Hou CY, Suo YH, Lv P, Yuan HF, Zhao LN, Wang YF, Zhang HH, Sun J, Sun LL, Lu W, et al: Aristolochic acids-hijacked p53 promotes liver cancer cell growth by inhibiting ferroptosis. Acta Pharmacol Sin. 46:208–221. 2025. View Article : Google Scholar : PubMed/NCBI

85 

Kang R, Kroemer G and Tang D: The tumor suppressor protein p53 and the ferroptosis network. Free Radic Biol Med. 133:162–168. 2019. View Article : Google Scholar : PubMed/NCBI

86 

Jiang L, Kon N, Li T, Wang SJ, Su T, Hibshoosh H, Baer R and Gu W: Ferroptosis as a p53-mediated activity during tumour suppression. Nature. 520:57–62. 2015. View Article : Google Scholar : PubMed/NCBI

87 

Wang SJ, Li D, Ou Y, Jiang L, Chen Y, Zhao Y and Gu W: Acetylation is crucial for p53-mediated ferroptosis and tumor suppression. Cell Rep. 17:366–373. 2016. View Article : Google Scholar : PubMed/NCBI

88 

Zhang Y, Wang M and Chang W: Iron dyshomeostasis and ferroptosis in Alzheimer's disease: Molecular mechanisms of cell death and novel therapeutic drugs and targets for AD. Front Pharmacol. 13:9836232022. View Article : Google Scholar : PubMed/NCBI

89 

Dwivedi D, Megha K, Mishra R and Mandal PK: Glutathione in brain: Overview of its conformations, functions, biochemical characteristics, quantitation and potential therapeutic role in brain disorders. Neurochem Res. 45:1461–1480. 2020. View Article : Google Scholar : PubMed/NCBI

90 

Rohr-Udilova N, Sieghart W, Eferl R, Stoiber D, Björkhem-Bergman L, Eriksson LC, Stolze K, Hayden H, Keppler B, Sagmeister S, et al: Antagonistic effects of selenium and lipid peroxides on growth control in early hepatocellular carcinoma. Hepatology. 55:1112–1121. 2012. View Article : Google Scholar : PubMed/NCBI

91 

Imai H, Matsuoka M, Kumagai T, Sakamoto T and Koumura T: Lipid peroxidation-dependent cell death regulated by GPx4 and ferroptosis. Curr Top Microbiol Immunol. 403:143–170. 2017.PubMed/NCBI

92 

Xu Y, Li K, Zhao Y, Zhou L, Liu Y and Zhao J: Role of ferroptosis in stroke. Cell Mol Neurobiol. 43:205–222. 2023. View Article : Google Scholar : PubMed/NCBI

93 

Magtanong L, Ko PJ and Dixon SJ: Emerging roles for lipids in non-apoptotic cell death. Cell Death Differ. 23:1099–1109. 2016. View Article : Google Scholar : PubMed/NCBI

94 

Lin KJ, Chen SD, Lin KL, Liou CW, Lan MY, Chuang YC, Wang PW, Lee JJ, Wang FS, Lin HY, et al: Iron brain menace: The involvement of ferroptosis in Parkinson disease. Cells. 11:38292022. View Article : Google Scholar : PubMed/NCBI

95 

Shen W, Li C, Liu Q, Cai J, Wang Z, Pang Y, Ning G, Yao X, Kong X and Feng S: Celastrol inhibits oligodendrocyte and neuron ferroptosis to promote spinal cord injury recovery. Phytomedicine. 128:1553802024. View Article : Google Scholar : PubMed/NCBI

96 

Ward RJ, Zucca FA, Duyn JH, Crichton RR and Zecca L: The role of iron in brain ageing and neurodegenerative disorders. Lancet Neurol. 13:1045–1060. 2014. View Article : Google Scholar : PubMed/NCBI

97 

Yan HF, Zou T, Tuo QZ, Xu S, Li H, Belaidi AA and Lei P: Ferroptosis: Mechanisms and links with diseases. Signal Transduct Target Ther. 6:492021. View Article : Google Scholar : PubMed/NCBI

98 

Chaudhary S, Ashok A, McDonald D, Wise AS, Kritikos AE, Rana NA, Harding CV and Singh N: Upregulation of local hepcidin contributes to iron accumulation in Alzheimer's disease brains. J Alzheimers Dis. 82:1487–1497. 2021. View Article : Google Scholar : PubMed/NCBI

99 

Li B, Xia M, Zorec R, Parpura V and Verkhratsky A: Astrocytes in heavy metal neurotoxicity and neurodegeneration. Brain research. 1752:1472342021. View Article : Google Scholar : PubMed/NCBI

100 

Villalón-García I, Povea-Cabello S, Álvarez-Córdoba M, Talaverón-Rey M, Suárez-Rivero JM, Suárez-Carrillo A, Munuera-Cabeza M, Reche-López D, Cilleros-Holgado P, Piñero-Pérez R and Sánchez-Alcázar JA: Vicious cycle of lipid peroxidation and iron accumulation in neurodegeneration. Neural Regen Res. 18:1196–1202. 2023. View Article : Google Scholar : PubMed/NCBI

101 

Gugliandolo A, Bramanti P and Mazzon E: Role of vitamin E in the treatment of Alzheimer's disease: Evidence from animal models. Int J Mol Sci. 18:25042017. View Article : Google Scholar : PubMed/NCBI

102 

Alim I, Caulfield JT, Chen Y, Swarup V, Geschwind DH, Ivanova E, Seravalli J, Ai Y, Sansing LH, Ste Marie EJ, et al: Selenium drives a transcriptional adaptive program to block ferroptosis and treat stroke. Cell. 177:1262–1279. e252019. View Article : Google Scholar : PubMed/NCBI

103 

Li Q, Han X, Lan X, Gao Y, Wan J, Durham F, Cheng T, Yang J, Wang Z, Jiang C, et al: Inhibition of neuronal ferroptosis protects hemorrhagic brain. JCI insight. 2:e907772017. View Article : Google Scholar : PubMed/NCBI

104 

Ikawa T, Sato M, Oh-Hashi K, Furuta K and Hirata Y: Oxindole-curcumin hybrid compound enhances the transcription of γ-glutamylcysteine ligase. Eur J Pharmacol. 896:1738982021. View Article : Google Scholar : PubMed/NCBI

105 

Deepmala, Slattery J, Kumar N, Delhey L, Berk M, Dean O, Spielholz C and Frye R: Clinical trials of N-acetylcysteine in psychiatry and neurology: A systematic review. Neurosci Biobehav Rev. 55:294–321. 2015. View Article : Google Scholar : PubMed/NCBI

106 

Hinman A, Holst CR, Latham JC, Bruegger JJ, Ulas G, McCusker KP, Amagata A, Davis D, Hoff KG, Kahn-Kirby AH, et al: Vitamin E hydroquinone is an endogenous regulator of ferroptosis via redox control of 15-lipoxygenase. PLoS One. 13:e02013692018. View Article : Google Scholar : PubMed/NCBI

107 

Ashraf A and So PW: Spotlight on ferroptosis: Iron-dependent cell death in Alzheimer's disease. Front Aging Neurosci. 12:1962020. View Article : Google Scholar : PubMed/NCBI

108 

Kryscio RJ, Abner EL, Caban-Holt A, Lovell M, Goodman P, Darke AK, Yee M, Crowley J and Schmitt FA: Association of antioxidant supplement use and dementia in the prevention of Alzheimer's disease by vitamin E and selenium trial (PREADViSE). JAMA Neurol. 74:567–573. 2017. View Article : Google Scholar : PubMed/NCBI

109 

Conrad M and Proneth B: Selenium: Tracing another essential element of ferroptotic cell death. Cell Chem Biol. 27:409–419. 2020. View Article : Google Scholar : PubMed/NCBI

110 

Ingold I, Berndt C, Schmitt S, Doll S, Poschmann G, Buday K, Roveri A, Peng X, Porto Freitas F, Seibt T, et al: Selenium utilization by GPX4 is required to prevent hydroperoxide-induced ferroptosis. Cell. 172:409–422. e212018. View Article : Google Scholar : PubMed/NCBI

111 

R Cardoso B, Hare DJ, Lind M, McLean CA, Volitakis I, Laws SM, Masters CL, Bush AI and Roberts BR: The APOE ε4 allele is associated with lower selenium levels in the brain: Implications for Alzheimer's disease. ACS Chem Neurosci. 8:1459–1464. 2017. View Article : Google Scholar : PubMed/NCBI

112 

Skouta R, Dixon SJ, Wang J, Dunn DE, Orman M, Shimada K, Rosenberg PA, Lo DC, Weinberg JM, Linkermann A and Stockwell BR: Ferrostatins inhibit oxidative lipid damage and cell death in diverse disease models. J Am Chem Soc. 136:4551–4556. 2014. View Article : Google Scholar : PubMed/NCBI

113 

Fang X, Wang H, Han D, Xie E, Yang X, Wei J, Gu S, Gao F, Zhu N, Yin X, et al: Ferroptosis as a target for protection against cardiomyopathy. Proc Natl Acad Sci USA. 116:2672–2680. 2019. View Article : Google Scholar : PubMed/NCBI

114 

Miotto G, Rossetto M, Di Paolo ML, Orian L, Venerando R, Roveri A, Vučković AM, Bosello Travain V, Zaccarin M, Zennaro L, et al: Insight into the mechanism of ferroptosis inhibition by ferrostatin-1. Redox Biol. 28:1013282020. View Article : Google Scholar : PubMed/NCBI

115 

Asano M, Yamasaki K, Yamauchi T, Terui T and Aiba S: Epidermal iron metabolism for iron salvage. J Dermatol Sci. 87:101–109. 2017. View Article : Google Scholar : PubMed/NCBI

116 

Kalyanaraman B: NAC, NAC, Knockin' on Heaven's door: Interpreting the mechanism of action of N-acetylcysteine in tumor and immune cells. Redox Biol. 57:1024972022. View Article : Google Scholar : PubMed/NCBI

117 

Fan Z, Wirth AK, Chen D, Wruck CJ, Rauh M, Buchfelder M and Savaskan N: Nrf2-Keap1 pathway promotes cell proliferation and diminishes ferroptosis. Oncogenesis. 6:e3712017. View Article : Google Scholar : PubMed/NCBI

118 

Kerins MJ and Ooi A: The roles of NRF2 in modulating cellular iron homeostasis. Antioxid Redox Signal. 29:1756–1773. 2018. View Article : Google Scholar : PubMed/NCBI

119 

Rojo de la Vega M, Chapman E and Zhang DD: NRF2 and the Hallmarks of cancer. Cancer Cell. 34:21–43. 2018. View Article : Google Scholar : PubMed/NCBI

120 

Hara Y, McKeehan N, Dacks PA and Fillit HM: Evaluation of the neuroprotective potential of N-acetylcysteine for prevention and treatment of cognitive aging and dementia. J Prev Alzheimers Dis. 4:201–206. 2017.PubMed/NCBI

121 

Pocernich CB, La Fontaine M and Butterfield DA: In-vivo glutathione elevation protects against hydroxyl free radical-induced protein oxidation in rat brain. Neurochem Int. 36:185–191. 2000. View Article : Google Scholar : PubMed/NCBI

122 

Koppal T, Drake J and Butterfield DA: In vivo modulation of rodent glutathione and its role in peroxynitrite-induced neocortical synaptosomal membrane protein damage. Biochim Biophys Acta. 1453:407–411. 1999. View Article : Google Scholar : PubMed/NCBI

123 

Pocernich CB, Cardin AL, Racine CL, Lauderback CM and Butterfield DA: Glutathione elevation and its protective role in acrolein-induced protein damage in synaptosomal membranes: Relevance to brain lipid peroxidation in neurodegenerative disease. Neurochem Int. 39:141–149. 2001. View Article : Google Scholar : PubMed/NCBI

124 

Prasad S, Tyagi AK and Aggarwal BB: Recent developments in delivery, bioavailability, absorption and metabolism of curcumin: the golden pigment from golden spice. Cancer Res Treat. 46:2–18. 2014. View Article : Google Scholar : PubMed/NCBI

125 

Wei Z, Shaohuan Q, Pinfang K and Chao S: Curcumin attenuates ferroptosis-induced myocardial injury in diabetic cardiomyopathy through the Nrf2 pathway. Cardiovasc Ther. 2022:31597172022. View Article : Google Scholar : PubMed/NCBI

126 

Hirata Y, Ito Y, Takashima M, Yagyu K, Oh-Hashi K, Suzuki H, Ono K, Furuta K and Sawada M: novel oxindole-curcumin hybrid compound for antioxidative stress and neuroprotection. ACS Chem Neurosci. 11:76–85. 2020. View Article : Google Scholar : PubMed/NCBI

127 

Hirata Y, Tsunekawa Y, Takahashi M, Oh-Hashi K, Kawaguchi K, Hayazaki M, Watanabe M, Koga KI, Hattori Y, Takemori H and Furuta K: Identification of novel neuroprotective N, N-dimethylaniline derivatives that prevent oxytosis/ferroptosis and localize to late endosomes and lysosomes. Free Radic Biol Med. 174:225–235. 2021. View Article : Google Scholar : PubMed/NCBI

128 

Hirata Y, Okazaki R, Sato M, Oh-Hashi K, Takemori H and Furuta K: Effect of ferroptosis inhibitors oxindole-curcumin hybrid compound and N, N-dimethylaniline derivatives on rotenone-induced oxidative stress. Eur J Pharmacol. 928:1751192022. View Article : Google Scholar : PubMed/NCBI

129 

Friedmann Angeli JP, Schneider M, Proneth B, Tyurina YY, Tyurin VA, Hammond VJ, Herbach N, Aichler M, Walch A, Eggenhofer E, et al: Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nat Cell Biol. 16:1180–1191. 2014. View Article : Google Scholar : PubMed/NCBI

130 

Li Y, Sun M, Cao F, Chen Y, Zhang L, Li H, Cao J, Song J, Ma Y, Mi W and Zhang X: The ferroptosis inhibitor liproxstatin-1 ameliorates LPS-induced cognitive impairment in mice. Nutrients. 14:45992022. View Article : Google Scholar : PubMed/NCBI

131 

Fan BY, Pang YL, Li WX, Zhao CX, Zhang Y, Wang X, Ning GZ, Kong XH, Liu C, Yao X and Feng SQ: Liproxstatin-1 is an effective inhibitor of oligodendrocyte ferroptosis induced by inhibition of glutathione peroxidase 4. Neural Regen Res. 16:561–566. 2021. View Article : Google Scholar : PubMed/NCBI

132 

Singh VK, Beattie LA and Seed TM: Vitamin E: Tocopherols and tocotrienols as potential radiation countermeasures. J Radiat Res. 54:973–988. 2013. View Article : Google Scholar : PubMed/NCBI

133 

Angeli JPF, Shah R, Pratt DA and Conrad M: Ferroptosis inhibition: mechanisms and opportunities. Trends Pharmacol Sci. 38:489–498. 2017. View Article : Google Scholar : PubMed/NCBI

134 

Zhang ZH, Chen C, Jia SZ, Cao XC, Liu M, Tian J, Hoffmann PR, Xu HX, Ni JZ and Song GL: Selenium restores synaptic deficits by modulating NMDA receptors and selenoprotein K in an Alzheimer's disease model. Antioxid Redox Signal. 35:863–884. 2021. View Article : Google Scholar : PubMed/NCBI

135 

Bao C, Liu C, Liu Q, Hua L, Hu J, Li Z and Xu S: Liproxstatin-1 alleviates LPS/IL-13-induced bronchial epithelial cell injury and neutrophilic asthma in mice by inhibiting ferroptosis. Int Immunopharmacol. 109:1087702022. View Article : Google Scholar : PubMed/NCBI

136 

Pei Z, Qin Y, Fu X, Yang F, Huo F, Liang X, Wang S, Cui H, Lin P, Zhou G, et al: Inhibition of ferroptosis and iron accumulation alleviates pulmonary fibrosis in a bleomycin model. Redox Biol. 57:1025092022. View Article : Google Scholar : PubMed/NCBI

137 

Li Z, Lu Y, Zhen Y, Jin W, Ma X, Yuan Z, Liu B, Zhou XL and Zhang L: Avicularin inhibits ferroptosis and improves cognitive impairments in Alzheimer's disease by modulating the NOX4/Nrf2 axis. Phytomedicine. 135:1562092024. View Article : Google Scholar : PubMed/NCBI

138 

Wu Y, Wei M, Wang M, Guo M, Yu H, Chen Y, Xu T and Zhou Y: Schisandra total lignans ameliorate neuronal ferroptosis in 3×Tg-AD mice via regulating NADK/NADPH/GSH pathway. Phytomedicine. 140:1566122025. View Article : Google Scholar : PubMed/NCBI

139 

Li X, Chen J, Feng W, Wang C, Chen M, Li Y, Chen J, Liu X, Liu Q and Tian J: Berberine ameliorates iron levels and ferroptosis in the brain of 3 × Tg-AD mice. Phytomedicine. 118:1549622023. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2025
Volume 32 Issue 1

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zeng H and Jin Z: The role of ferroptosis in Alzheimer's disease: Mechanisms and therapeutic potential (Review). Mol Med Rep 32: 192, 2025.
APA
Zeng, H., & Jin, Z. (2025). The role of ferroptosis in Alzheimer's disease: Mechanisms and therapeutic potential (Review). Molecular Medicine Reports, 32, 192. https://doi.org/10.3892/mmr.2025.13557
MLA
Zeng, H., Jin, Z."The role of ferroptosis in Alzheimer's disease: Mechanisms and therapeutic potential (Review)". Molecular Medicine Reports 32.1 (2025): 192.
Chicago
Zeng, H., Jin, Z."The role of ferroptosis in Alzheimer's disease: Mechanisms and therapeutic potential (Review)". Molecular Medicine Reports 32, no. 1 (2025): 192. https://doi.org/10.3892/mmr.2025.13557