IMR Press / JIN / Volume 24 / Issue 4 / DOI: 10.31083/JIN25845
Open Access Review
Ferroptosis in Alzheimer’s Disease: The Regulatory Role of Glial Cells
Show Less
Affiliation
1 Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, 646000 Luzhou, Sichuan, China
2 Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
3 Laboratory of Neurological Diseases and Brain Function, Institute of Epigenetics and Brain Science, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
4 Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University, 646000 Luzhou, Sichuan, China
*Correspondence: yuyang80@swmu.edu.cn (Yang Yu); dongwei@swmu.edu.cn (Wei Dong)
J. Integr. Neurosci. 2025, 24(4), 25845; https://doi.org/10.31083/JIN25845
Submitted: 25 July 2024 | Revised: 11 October 2024 | Accepted: 30 October 2024 | Published: 24 April 2025
Copyright: © 2025 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Alzheimer’s disease (AD) is a neurodegenerative disease characterized by the formation of amyloid plaques, neurofibrillary tangles and progressive cognitive decline. Amyloid-beta peptide (Aβ) monoclonal antibody therapeutic clinical trials have nearly failed, raising significant concerns about other etiological hypotheses about AD. Recent evidence suggests that AD patients also exhibit persistent neuronal loss and neuronal death accompanied by brain iron deposition or overload-related oxidative stress. Ferroptosis is a type of cell death that depends on iron, unlike autophagy and apoptosis. Inhibiting neuronal ferroptosis function is effective in improving cognitive impairment in AD. Notably, new research shows that ferroptosis in AD is crucially dependent on glial cell activation. This review examines the relationship between the imbalance of iron metabolism, the regulation of iron homeostasis in glial cells and neuronal death in AD pathology. Finally, the review summarizes some current drug research in AD targeting iron homeostasis, many novel iron-chelating compounds and natural compounds showing potential AD-modifying properties that may provide therapeutic targets for treating AD.

Keywords
ferroptosis
Alzheimer’s disease
glial cells
neuron
lipid peroxidation
1. Introduction

Iron is an essential metal cofactor that plays a vital role in many physiological functions in the body. Various physiological processes are regulated by iron in the brain, including functions that contribute to myelin formation, neurotransmitter synthesis and antioxidant enzymes [1]. Maintaining homeostasis of iron is crucial for maintaining human health, as both iron deficiency and iron overload can be harmful. It has been demonstrated that iron is essential for the normal function of neurons, oligodendrocytes, astrocytes and microglia in the central nervous system (CNS) [2]. Abnormal iron metabolism can increase blood-brain barrier (BBB) permeability and neuroinflammation, promoting neurodegenerative processes [3]. Ferroptosis is a novel type of iron-dependent cell death caused by iron overload, which results in iron-dependent accumulation of lipid peroxides and the production of reactive oxygen species (ROS) [4]. This may be a novel mechanism for inducing neuronal death in neurodegenerative diseases such as Alzheimer’s disease (AD).

There is increasing evidence that iron overload and homeostasis dysregulation of iron contribute to neurodegeneration in AD and that dysregulation of metal homeostasis is associated with cell activation and death processes [5, 6]. In an earlier study, histochemistry showed a striking accumulation of iron (III), localized with iron (II) cyanide, which was closely related to senile plaques, neurofibrillary tangles, and neuropil threads; the iron associated with this lesion promotes in situ oxidation and catalyze H2O2-dependent oxidation, indicating that iron accumulation may be an important factor in oxidative damage in AD [7]. Patients with moderate cognitive impairment (MCI) and AD have higher levels of brain iron deposition, according to quantification using susceptibility-weighted imaging [8]. Further evidence suggests that disruption of iron homeostasis and elevated ferritin levels in the cerebrospinal fluid (CSF) are linked to the progression from MCI to AD [9]. Additionally, a meta-analysis involving 2174 patients with AD and 2931 healthy controls showed decreased levels of iron in the blood and increased ferritin in the CSF of patients with AD when compared to levels in controls, suggesting that iron and iron-associated proteins are associated with the risk of AD [10]. Notably, whole-exome sequencing identified protein-coding variants associated with brain iron in 29,828 individuals, revealing several insights into the relationship between brain iron accumulation and AD and identifying multiple genes associated with brain iron accumulation levels [11]. These studies demonstrated a strong correlation between the pathophysiology of AD and iron homeostasis disturbance.

Recently, most research on iron-mediated neurodegenerative diseases has focused on neurons, ignoring the role of non-neuronal cells, in regulating pathological processes. Indeed, in an environment of iron metabolism disorder AD, the crosstalk between glial cells and neurons may form positive feedback, astrocyte reactive hyperplasia and amyloid-beta peptide (Aβ) activation of microglia, resulting in a self-amplifying inflammatory response in the AD process [2, 12, 13]. This suggests that iron deposition-induced ferroptosis, glial cell activation and neuroinflammation may be potential underpinnings of AD pathogenesis.

2. AD Pathogenesis

AD is a neurodegenerative disease that stands out as a prominent health problem for elderly individuals worldwide. It has become evident that AD is not only linked to its hallmark lesions—the accumulation of Aβ and development of neurofibrillary tangles (NFT) consequent to the hyperphosphorylation of tau protein but also to other concurrent pathologies [14]. Currently, the exact pathogenesis of AD is complex and still not fully understood, as it is influenced by factors such as genetics, aging, nutrition and a person’s environment [15]. The complexity of the clinical measurement of underlying pathogenesis also poses a challenge to characterizing the pathology of AD and the fact that most medications only alleviate the symptoms and usually fail to achieve the desired outcome [15]. Indeed, the underlying biological mechanisms implicated in the pathogenesis of AD are also involved in abnormal protein dynamics, oxidative stress, neuroinflammation, disturbances of metal-ion homeostasis and bioenergetics [16].

3. Iron Dysregulation in AD

Aberrant iron uptake, excretion and storage in neurons increases intracellular labile iron pools (LIP) and Fenton responses during the regulation of iron homeostasis [17]. The primary regulator of intracellular iron metabolism is the iron regulatory protein (IRP) response element (IRE) regulatory system [18]. In an intracellular environment deficient in iron, IRP binds to ferritin and homologous IRE in the untranslated region of transferrin receptor 1 (TfR1) mRNA to promote ferritin mRNA translation inhibition and stabilize TfR1 mRNA to prevent intranuclear catabolic events [19]. However, in iron-rich cells, IRP does not bind to the IRE, enhances ferritin synthesis, permitting degradation of TfR1 mRNA and the extracellular transport of Fe3+ by transferrin (Tf) to cells, subsequently reducing Fe2+ [20, 21]. Fe2+ are then transported from endosomes to the cytoplasm via divalent metal transporter 1 (DMT1), which is regulated by homeostasis of LIP [22]. DMT1 is used for normal neuronal metabolism and is partially stored in ferritin to protect neurons from excess iron [23]. When excess H2O2 in the cell interacts with Fe2+, it starts the Fenton reaction, which causes intracellular lipid peroxidation to produce ROS and ferroptosis [24]. Notably, iron export in neurons is mediated by iron-exporting transmembrane protein ferroportin1 (Fpn1) through the Fpn1/Hephaestin and Fpn1/Ceruloplasmin (CP) pathways [25, 26]. Alterations to this pathway induce iron retention, leading to memory impairment [27, 28]. Simultaneous Fe2+ and Fe3+ imaging shows their enrichment in the brains of mice with AD [29]. Moreover, increased ferritin heavy chain (FTH) and ferritin light chain expression were found in the brains of AD patients, suggesting that LIP is increased in AD, confirming that iron can be used as a biomarker for AD [5].

Aβ deposition and hyperphosphorylated Tau protein tangles are AD’s two main pathological features. Autopsy and magnetic resonance imaging have shown significant iron deposition around senile plaques [30] and at sites of cortical Tau accumulation [31], suggesting potential crosstalk between iron and senile plaques and neurogenic fiber tangles. Iron-Aβ complexes are formed in the Amyloid Precursor Protein (APP)/Presenilin genes 1 (PS1) animal, as shown by submicron resolution X-ray microscopy, indicating a direct correlation between Aβ and iron [32]. Aβ is produced by the amyloid precursor protein (APP) and an IRE in the 5 untranslated region of the APP transcript regulates APP levels [33]. It has been shown that higher cellular iron levels in AD pathology cause increased APP translation and Aβ generation [34]. Interestingly, APP can interact with and stabilize Fpn1 to facilitate iron export and maintain the homeostasis of iron concentration [35]. Silencing/mutation of APP leads to abnormal Fpn1 function and inefficient neuronal output of iron, which further leads to iron accumulation, suggesting that APP/Fpn1 plays an important role in the regulation of iron homeostasis in the brain [36]. Furthermore, in the context of aging, Fpn1 exhibits age-dependent down-regulation [37], as demonstrated in the APPswe/PS1dE9 mouse model and in brain tissue from AD patients [28]. The degradation of APP involves two pathways, mainly involving three secretases: α, β, and γ [38]. Under normal physiological conditions, APP is processed by α-secretase to produce non-toxic forms of P3, Aβ16 and Aβ17-40/42; on the downside, when iron is overloaded in cells, β and γ secretases promote APP cleavage to form Aβ1-40 and Aβ1-42 fragments, leading to neurotoxic amyloid proteins, inducing cell damage and death [39]. It is also noteworthy that Aβ also promotes the reduction of Fe3+ to Fe2+ to be a potential source of low-oxidation-state iron, leading to oxidative damage to Aβ and surrounding molecules and further deposition of Aβ into senile plaques [40]. Additionally, the affinity of Aβ for iron increases after aggregation, leading to additional neuronal cytotoxicity [41]. The above studies suggest that AD pathophysiology is significantly influenced by the interplay between APP, Aβ and iron metabolism.

The tau protein is a significant component of neurogenic fibrillary tangles, mediating iron efflux from cells and excess iron colocalizes with tau in neurogenic fibrillary tangles [31]. Autopsy study finds colocalization of iron and tau in neurons carrying NFT associated with progressive neurodegeneration [42]. Neuronal iron overload causes tau hyperphosphorylation by activating protein kinases, such as glycogen synthase kinase-3β, cyclin-dependent protein kinase-5 and mitogen-activated protein kinases amongst others, inhibiting protein phosphatase 2A [43, 44], while treatment with the iron chelator desferrioxamine alleviates iron-induced tau hyperphosphorylation [45]. Under normal physiological conditions, tau protein can transport APP to cell membranes, thereby stabilizing Fpn1 and promoting iron efflux [46]. However, due to over-modification and cleavage of APP by secreted enzymes, APP is unable to reach the cell surface properly, which results in a decrease in Fpn1 levels and iron accumulation within neurons and NFT [47, 48]. Notably, increased levels of oxidative stress mediated by iron regulatory imbalance may also promote tau oligomerization by binding to cysteine containing mercapto-cysteine [49]. Moreover, in a cell culture model, iron may lead to hyperphosphorylation and aggregation of tau proteins through iron binding motifs in tau proteins as well as dysregulation of insulin signaling [50]. More importantly, it has been demonstrated that age-induced neurodegeneration is caused by iron accumulation in the cortex, substantia nigra and hippocampus of tau mutant mice [47]. APP/PS1 mice with iron feeding showed impaired cognitive function, accompanied by increased in Aβ accumulation and phosphorylation of tau expression [51]. Inhibition of iron-induced hippocampal tau phosphorylation [52] and APP production processing in APP/PS1 mice is induced by treatment with the iron chelator desferrioxamine (DFO), thereby inhibiting Aβ deposit formation [53] and reversing iron-induced memory impairment [52]. Different models of AD show a strong association between iron accumulation with Aβ deposition and tau phosphorylation. However, the up- and downstream molecular mechanisms among these three in this potentially relentless cycle remain unclear. Despite insufficient data from human studies, targeting iron homeostasis with therapies improves AD pathology, as one important mechanism controlling the early pathological development of AD is iron metabolism.

Dysregulation of iron metabolism in AD has been demonstrated mainly in preclinical studies. Studies have found that Fpn1 is downregulated in AD patients and APP/PS1 animal models, while genetic absence of Fpn1 resulted in hippocampus atrophy and memory impairment. However, administration of specific iron chelating inducer significantly reduces neuronal death and memory deficits caused by Aβ aggregation, while restoration of Fpn1 ferroptosis and memory deficits in APP/PS1 mice [28]. Additionally, ferritinophagy-mediated ferroptosis is a key mechanism leading to neurodegenerative disease [54]. An essential component of the body’s regulation of iron metabolism is the iron store protein ferritin; nuclear receptor coactivator 4 (NCOA4) and ferritin-containing iron cooperate in iron-deficient cells to mediate the release of iron from the ferritin storage pool through the ferritinophagy pathway [55]. The ferritinophagy process promotes ferroptosis and ferritin degradation within the lysosome and dysfunction of ferritinophagy is an important factor contributing to iron accumulation in senescent cells, this can lead to elevated total iron levels within affected brain regions, especially in AD pathology [56, 57].

Calcium-mediated oxidative stress is largely inseparable from iron, as both iron and Ca2+ play key roles in neuronal function, such as ROS production, mitochondrial homeostasis, and synaptic plasticity [58]. Iron-induced neuronal ROS production alters Ca2+ signaling, which in turn affects synaptic plasticity and hippocampus-dependent memory formation [59, 60]. In the mouse hippocampal neuronal cell line HT-22, excess iron leads to mitochondrial fragmentation and increased Ca2+ levels, which in turn stimulates the Ca2+-dependent phosphatase calcineurin and neuronal cell death [61]. In primary hippocampal neurons, iron-induced ROS increase promotes the release of calcium in the endoplasmic reticulum (ER) and subsequent mitochondrial fission may lead to neuronal dysfunction [62]. Thus, mitochondria as a potential target for the treatment of neurodegenerative processes induced by abnormal iron metabolism. Notably, the crosstalk between iron, ROS, and Ca2+ signaling is bidirectional, as many proteins involved in cellular antioxidant defense and ROS production are dependent on Ca2+. The aberrant increase in intracellular Ca2+ promotes the uptake of mitochondrial Ca2+ by mitochondrial Ca2+ unidirectional transporters, resulting in more ROS production, which may further increase an unstable iron pool [63]. A primary hippocampal neuronal study has reported an influx of iron through Ca2+ channels, resulting in altered intracellular iron content [64]. A sustained increase in Ca2+ flux regulates the immunological profile of ferroptosis [65]. Overall, current evidence suggests that the coordinated interrelationship between excess iron and abnormal Ca2+ signaling is most likely to occur in neurodegeneration, particularly AD, as abnormal calcium signaling is a central issue in AD pathology [66, 67, 68].

4. Ferroptosis in AD

Several pathological features found in AD patients are highly correlated with ferroptosis, including elevated levels of free iron [8], lipid peroxidation [69] and defects in antioxidant systems such as glutathione peroxidase 4 (GPX4), glutathione (GSH), and cystine/glutamate antiporter system (system Xc) [70]. System Xc, composed of the light chain Solute Carrier Family 7 Member 11 (SLC7A11) and heavy chain Solute Carrier Family 3 Member 2 (SLC3A2), is an important glutamate transporter in the CNS for the cellular uptake of cystine in exchange for intracellular glutamate [71]. In the context of AD, compensatory uptake of cystine leads to poor glutamate clearance in the synaptic cleft, causes excitatory neurotoxicity through system Xc, enhances lipid peroxidation and leads to ferroptosis and the development of AD [72, 73]. Additionally, GSH is an important intracellular antioxidant that prevents iron-dependent oxidation, which is a crucial biological process in ferroptosis, primarily by binding to Fe2+ in unstable LIP [74]. Reduced GSH levels have been found in the occurrence and progression of AD [75]. Further, γ-glutamylcysteine ethyl ester, a precursor to GSH, was found to increase intracellular GSH content, thereby inhibiting neuronal toxicity triggered by an Aβ-induced decline in GSH levels [76] and these findings suggest a strong link between dysregulation of GSH levels and AD pathology. Another predisposing factor associated with ferroptosis in AD is GPX4. GPX4 has been shown to be a key regulatory enzyme in AD-associated ferroptosis and has been shown to repair lipid hydroperoxides in membranes and inhibition of GPX4 is one of the key factors in the pathogenesis of ferroptosis-associated AD [77]. Indeed, knockout of the GPX4 gene in the brain leads to a number of features associated with AD pathology, for example, Gpx4BIKO mice exhibit cognitive deficits and hippocampal neurodegeneration, including reduced levels of NeuN, synaptophysin and Synaptosome Associated Protein 25 (SNAP25) and a significant decline in learning ability and memory in mice over time [78]. Additionally, studies have shown that GPX4 deletion upregulates β-secretase activity and leads to increased Aβ production [79], overexpression of the GPX4 gene in 5xFAD mice reduced cognitive impairment and markedly enhanced learning and memory [77]. Further, mutations in presenilin genes 1 and 2 (PS1 and PS2) mediate autosomal dominant familial AD lesions; mutations in PS1 and PS2 cause AD lesions by repressing GPX4 expression and deregulating ferroptosis [80]. These findings suggest that ferroptosis is involved in AD lesions and the search for drugs t targeting Xc, GSH and GPX4 systems represents an important direction in AD research. Notably, the aging brain expresses high quantities of polyunsaturated fatty acids, which are favored substrates for lipid peroxidation during ferroptosis and increase vulnerability to the consequences of ferroptosis [81]. Moreover, one important regulator of ferroptosis is nuclear factor erythroid 2-related factor 2 (Nrf2), Nrf2 activates to protect cells and tissues from ferroptosis by regulating significant genes, such as those involved in iron and glutathione metabolism, against oxidative stress caused by excess iron [82]. Research has demonstrated that Nrf2 mRNA expression is significantly lower in AD patients and animal models, and that Nrf2 deficiency speeds up the production of Aβ and β-secretase 1 (BACE1) expression [83]. More importantly, the expression of Nrf2 decreases with age, suggesting that it may contribute to the development of AD by promoting ferroptosis stress sensitivity [84].

Apolipoprotein E (APOE) allele variants are the most significant genetic risk for disseminated AD and APOE variants, leading to Aβ deposition, hippocampal atrophy and abnormal brain metabolic function [85]. Ferritin levels in CSF have been found to be positively correlated with APOE-ε4 expression and negatively correlated with both MCI and AD [86]. In this context, Duro et al. [87] found an association between APOE-ε4, intracortical iron and brain function. Indeed, elevated extracellular iron levels upregulate APOE-ε4 expression in neurons and accelerate intracellular polyunsaturated fatty acids (PUFA) accumulation [88], providing a suitable environment for lipid peroxidation and ferroptosis, which leads to increased extracellular Aβ deposition and exacerbation of AD [89]. Notably, glial cell dysfunction is thought to contribute to amyloid accumulation and synaptic loss in AD and Haney et al. [90] found that both a large number of lipid droplets accumulate in glial cells and their synaptic pruning function may be regulated by the APOE allele. Moreover, a recent research has revealed that APOE is a potent inhibitor of ferroptosis that activates the Phosphatidylinositol-3-kinase/ Protein kinase B (PI3K/AKT) pathway and inhibits the autophagic degradation of ferritin (ferritinophagy) thereby avoiding lipid peroxidation mediated by iron [57]. Although the study of APOE in brain iron regulation needs to be further developed, iron seems to provide a bridge between APOE and AD, which supports a further theoretical basis for the study of the pathomechanisms of AD.

Ferroptosis in AD is accompanied by multiple pathologic changes. A variety of other manifestations associated with ferroptosis have been identified in various neurological diseases mediated by neuroinflammation, and damage-associated molecular patterns. These include the ROS generated during ferroptosis events that activate glial cells by activating neuroimmune pathways, which subsequently lead to the production of various inflammatory cytokines that promote the development of neurological diseases [91]. Novel drugs that target inflammation have shown effective intervention against ferroptosis. One study found that dimethyl fumarate attenuates neuroinflammation and ferroptosis by mediating the inflammatory central node nuclear factor-κB (NF-κB) signaling pathway and ameliorates bivascular occlusion-induced cognitive impairment in rat chronic cerebral hypoperfusion models [92]. Similarly, silybin exerts neuroprotective effects on STZ-induced sporadic AD models by downregulating ferroptosis injury and thereby downregulating downstream stimulator of interferon genes (STING)-mediated neuroinflammation [93]. Indeed, a recent study indicates that ferroptosis is associated with the activation of inflammation, including the activation of multiple inflammation-related signaling pathways, including the Janus kinase-signal transducer and activator of transcription (JAK-STAT), NF-κB, inflammasome, cyclic Guanosine Monophosphate–Adenosine Monophosphate (GMP-AMP) synthase-stimulator of IFN genes (cGAS-STING) and Mitogen-Activated Protein Kinase (MAPK) pathways [94]. Targeting these inflammation-related pathways may provide intervention for ferroptosis, especially in AD, as one of the important features of AD pathogenesis is neuroinflammation [95].

Various pathways, including iron metabolism, redox status, lipid and sugar metabolism, the metabolism of cytotoxic amino acids and mitochondrial activity may regulate the process of ferroptosis [4]. Major markers of ferroptosis, dysregulation of ferritosis, lipid peroxidation and ROS production have long been identified in the brain and animal models of AD patients [96]. A recent transcriptomic study has identified differential expression of genes associated with ferroptosis in AD in different glial cells and neurons [97], suggesting that the pathogenic process of ferroptosis in AD may include glial cells. Though it’s yet unknown exactly how ferroptosis occurs in neurons, non-neuronal cells such as glia are integral to mediating ferroptosis to neurons [17]. When activated, glial cells can induce ferroptosis by disrupting iron homeostasis, releasing proinflammatory factors, altering amino acid metabolism and raising levels of oxidative stress, which are associated with many organ injuries and degenerative diseases [98, 99]. For instance, neurofilament light chain, a pathologic marker for various neurological disorders, including AD, is part of the axonal cytoskeletal protein complex and FTH may be released into the extracellular space while reflecting the senescent state of microglia. After acting on microglia with various neurofilament light chain concentrations, the secretion of FTH-containing exosomes by microglia increased and stimulated peroxidation of membrane lipid and neuronal loss involved in neuronal ferroptosis [100]. Although iron homeostasis and ferroptosis are crucial in causing AD neuronal death, how they interact and lead to neurodegeneration remains poorly understood. Therefore, the pathological processes of iron metabolism disruption and associated cell death urgently require further investigation in the brain.

5. The Role of Glial Cells in Regulating Ferroptosis in AD

Microvascular endothelial cells (BMECs), astrocytes, microglia and pericytes together constitute the BBB [101]. The BBB is an effective protective filter for the brain as it regulates iron entry, separating cerebral iron homeostasis from whole-body homeostasis. Expressed on the luminal side of cerebral capillaries, TfR1 mediates iron uptake in the brain and binds circulating transferrin to promote the transfer of iron to BMECs through the TfR1-mediated endocytosis mechanism [3]. Notably, hepcidin is a peptide hormone implicated in the regulation of Fpn1 and brain iron metabolism. It has been shown that hepcidin is expressed in cortical neurons, glial cells and BMECs and plays an important role in the regulation of iron homeostasis [102]. Hepcidin binds to Fpn1 in the cell membrane of BMECs, promotes the internalization and degradation of Fpn1 and blocks the passage of iron into the brain through the BBB [103]. It was shown that hepcidin expression levels were reduced in AD patients and APP transgenic mice, whereas hepcidin treatment of cultured BMECs and neurons resulted in a reduction of iron input (TfR1 and DMT1) and output (Fpn1) proteins, which reduced iron uptake in neurons [104]. Furthermore, in a recent study of the APP/PS1 mouse model, it has been shown that hepcidin overexpression in astrocytes induces a reduction in iron levels in cortical and hippocampal neurons, with significant improvements in cognition and Aβ plaque aggregation, leading to a reduction in oxidative stress and neuroinflammation, but more importantly, a reduction in neuronal death [105]. Additionally, it was found that iron, transferrin and ferritin levels in astrocytes, microglia and oligodendrocytes change during aging [106]. This means that glial cells play an important role in the regulation of iron in the brain. In fact, aging mediates inflammation and BBB damage, which is responsible for the uneven and unbalanced distribution of brain iron and the main cause of neurodegeneration, while aging-dependent brain iron accumulation may be due to changes in ferritin levels that impair its homeostasis, which in turn activates glial cells, induces cytokine release and propagates neuroinflammation.

Glial cells are not only the primary support cells for neurons but are active participants in neuronal network formation and information processing in the CNS [107]. Extensive research has demonstrated that the crucial role of ferroptosis in the inflammatory response. Ferritin synthesis is regulated by proinflammatory cytokines, including interleukin-1β (IL-1β), interleukin-6 (IL-6), tumor necrosis factor α (TNF-α), and interferon-gamma (IFN-γ), which affect iron storage in cells and tissues [108]. Microglia first induce ferroptosis and may do so by producing inflammatory factors, including interleukin-8 or IL-1β [109]. Astrocyte-neuron interactions protect neurons from ferroptosis, these include enhanced expression of GSH, Nrf2 and catalase to counteract redox stress associated with excess free iron [110, 111]. For example, in the resting state, the ferroptosis inhibitor Nrf2 is efficiently activated by brain-derived neurotrophic factor in astrocytes and regulates metabolic cooperation between astrocytes and neurons [112]. Thus, dysregulation of neuronal interactions in astrocytes may induce neuronal ferroptosis. Additionally, in the setting of iron deficiency, astrocytes and microglia are able to release iron bound to ferritin and support oligodendrocytes during myelination [113, 114]. Conversely, in the setting of iron overload, oligodendrocytes overexpress ferritin, providing important antioxidant defenses to neurons [115]. Uncontrolled glial activation and neuroinflammation are involved in the pathogenesis of AD [116]. In the brains of AD patients, reactive astrocytes accumulate around Aβ by phagocytosis of locally degenerated dendrites and synapses [117] and surround Aβ deposition in the form of glial scars [118]. Activated microglia and astrocytes induce neuronal death in vitro by releasing proinflammatory factors, nitric oxide (NO), ROS, and glutamate [119]. Moreover, in sporadic and preclinical AD patients, focal loss of oligodendrocytes in the neocortical gray matter is associated with Aβ plaque cores [120]. Indeed, glial cells regulate iron metabolism by expressing iron regulatory proteins to maintain normal myelin sheath function and neuronal recombination processes; when activated, they can influence AD pathology by inducing neuronal death through pathological processes such as disruption of iron homeostasis [2], release of various cytokines, alteration of protein metabolism, and increased levels of oxidative stress (Fig. 1). Thus, there is a need to gain insight into the interactions between glial cells and iron metabolism and neurodegenerative diseases, which may facilitate the exploration of therapeutic strategies for AD by restoring iron homeostasis and inhibiting oxidative stress.

Fig. 1.

Molecular mechanisms of glial cell ferroptosis regulation in AD. In an AD pathological setting, glial cells are exposed to elevated levels of iron, lipopolysaccharide and extracellular Aβ released by damaged neurons. (i) DMT1 and ferritin expression are upregulated in microglia, leading to increased intracellular iron stores. With continued iron uptake, LIP is formed. Iron induces ROS production and activates inflammatory factor release, promoting neuroinflammation, infiltrating Aβ plaques and promoting neuronal iron accumulation. Increased iron promotes microglia glycolytic pathways, reduces phagocytosis and leads to Aβ deposition. Extracellular Aβ promotes ROS production via NADPH-oxidase, further increasing DMT1 expression and promoting iron sequestration, forming a relentless cycle of iron, ROS and Aβ. (ii) High iron levels induce reactive astrocytes to produce ROS and release inflammatory factors to promote Aβ deposition and NFT formation. Inflammatory factors upregulate DMT1 expression and downregulate Fpn1 expression, further promoting iron sequestration. Hepcidin in astrocytes resist ROS-mediated inflammatory factor release and upregulate Fpn1 expression. CP protein regulates the dynamic balance of iron entering and leaving the cell. (iii) Activated oligodendrocytes release ROS to promote neuronal oxidative stress, upregulate ferritin expression, increase intracellular iron stores and provide antioxidant defense against iron-induced cytotoxicity by secreting ferritin heavy chains. Three types of glial cells change neuronal iron homeostasis in neuroinflammation, promote iron accumulation in neurons by releasing inflammatory factors, infiltrating Aβ plaques and NFT formation, triggering neuronal ferroptosis, resulting in neuronal loss. Abbreviations: ROS, reactive oxygen species; HO-1, heme oxygenase 1; DMT1, divalent metal transporter 1; NOX, NADPH-oxidase; LPS, lipopolysaccharide; IFN-γ, interferon-γ; TNF-α, tumor necrosis factor α; IL-1β, interleukin-1β; IL-6, interleukin 6; iNOS, inducible nitric oxidesynthase; Lf, lactoferrin; APP, amyloid precursor protein; Aβ, amyloid β-protein; NFT, neurofibrillary tangles; LIP, labile iron pool; Lipro-1, liproxstatin-1; FTH, ferritin heavy chains; BChE, butyrylcholinesterase; AChE, acetylcholinesterase; TGF-β1, transforming growth factor-β1; NOX4, NADPH Oxidase 4; GPI-CP, glycosylphosphatidylinositol-anchored proteins; MTFT, Mitochondrial ferritin; Fpn1, ferroportin1; TfR, transferrin receptors. This figure was drawn using Biorender (https://www.biorender.com).

6. Glia Activation Regulates Ferroptosis by Disrupting Iron Homeostasis
6.1 Microglia

As highly reactive immune cells in the brain, microglia play a critical role in regulating the iron metabolism in brain networks [121]. Proteins that regulate iron metabolism in microglia, including transferrin receptors (TfR), ferritin, divalent metal DMT1 and Fpn1, which mediate iron influx from BMECs and the brain interstitium control the amount of iron in neurons [122, 123]. After being released from Tf, iron is translocated to the cytoplasm via DMT1 or other transporters [124]. Thus, microglia can internalize non-Tf-bound iron (NTBI) and Tf iron through different uptake pathways [125]. Microglia regulate their iron levels through the IRP and IRE signaling pathways, which control the expression of proteins related to iron metabolism; when intracellular iron levels are low, IRP binds to transferrin receptors and iron IRE in ferritin mRNA, leading to an increase in the expression of these proteins and promoting iron uptake and storage [125]. Under conditions of neuroimmune activation, the abundance of DMT1 on the surface of microglia and neurons is increased, and these cells are susceptible to ferroptosis due to iron overload induced by high levels of NTBI [126].

First, the microglia of activated cells can accumulate iron, leading to increased iron accumulation in the CNS within the brain [127]. The study reported that iron supplementation in newborn mice promotes neurodegeneration, leading to microglia activation, increased volume and reduced process length [128]. Second, proinflammatory cytokines mediated by NF-κB activate microglia in response to iron excess; Subsequently, microglia exacerbate iron accumulation in neurons by releasing proinflammatory cytokines [129]. When cultures of organotypic hippocampal cells were treated with ferrous ammonium sulfate, ferrous ammonium citrate, or ferrocene, there was a significant activation of microglia, accompanied by increased levels of ferritin expression in microglia and oligodendrocytes, as well as proinflammatory factors such as TNF-α, IL-1β and IL-6 [130, 131]. Furthermore, ultra-high-resolution magnetic resonance imaging (MRI) showed the presence of microglia with iron-positive staining in AD patients’ hippocampus [132]. Iron accumulation is also consistently found in activated microglia in the frontal cortex of AD patients and is normally located near the Aβ plaques [133]. Further research has found that increased iron levels may trigger and lead to iron-rich Aβ plaque formations, which promote microglia proliferation and toxicity accumulation [127]. Iron feeding elevates Aβ levels in SH-SY5Y (neuroblastoma cells) co-culture senescent microglia cells [134] and co-deposition of Aβ with iron has also been found in microglia of APP/PS1 mice [135]. Moreover, an earlier autopsy study has found strong immunopositivity for microglia ferritin in senile plaques in the hippocampal region of AD patients and subsequent in-depth studies found that increased microglia ferritin, as well as intracellular iron deposition, increased the level of oxidative stress in the cells themselves, leading to a tendency toward apoptosis [136]. Notably, when neurodegeneration has occurred, activated microglia remove extracellular iron by expressing more ferritin [137], which is accompanied by an increase in expression of the intracellular ferritin light chain iron storage protein [127], an increase in the release of proinflammatory factors [135, 138] and finally infiltration with Aβ [127, 139], meaning that that iron overload may trigger microglia metabolic and/or inflammatory responses [140].

In the APP/PS1 mouse, an inflammatory microglia phenotype was induced with IFN-γ and Aβ [135], inflammatory cytokines IL-6 and TNF-α increased DMT1 expression in microglia, thereby promoting iron sequestration in cells [141, 142]. Subsequently, the expression of ferritin mRNA on the cell surface was upregulated, and transferrin mRNA was down-regulated, which further promoted iron accumulation in activated microglia; iron accumulation induced a shift toward a glycolytic metabolism, impaired their phagocytosis and weakened the clearance rate of Aβ and eventually led to Aβ deposition [135], resulting in a relentless cycle of AD. Interestingly, activated microglia also secrete the acute-phase protein lactoferrin, which interacts with APP to promote Aβ formation [143], promoting an increase in IL-1β expression in microglia in the setting of iron overload, thereby enhancing pro-inflammatory effects [138]. Additionally, through regulating the NF-κB signaling pathway, Aβ causes microglia to synthesize and secrete inflammatory IL-1β; iron treatment further enhanced the inflammatory response in microglia, while DMT1 inhibition protects microglia from Aβ-induced inflammation responses and IL-1β increases induced by Aβ were prevented by ROS inhibitors [138]. Intriguingly, in another study, iron deprivation culture experiments inhibited LPS-induced secretion of IL-1β and TNF-α from microglia and increased the expression of anti-inflammatory cytokine IL-10 [144]. In particular, IL-10 improves iron metabolism by inhibiting inflammatory factors, regulating the STAT3 signaling pathway, downregulating hepcidin expression and suppressing TfR expression [145].

6.2 Astrocytes

Astrocytes, a component of the BBB, are thought to be critical regulators of redox-active metal-iron metabolism within the brain and they influence iron homeostasis by regulating the transfer of iron from the outer body into the CNS, as well as the inflow and outflow of iron between neurons and glial cells [146]. Since the terminal foot protrusions of astrocytes are closely linked to the luminal creation of BMECs, this may be the primary pathway for iron to enter brain tissue via the Tf/TfR1 pathway [147]. Studies show astrocytes exhibit high levels of DMT1 and TfR1, as well as the iron oxidase CP, iron inflow and iron efflux proteins [148, 149]. Among them, DMT1 is predominantly expressed in astrocytes associated with vascular endothelial cells, which play a major role in iron uptake [149]. Moreover, CP acts as an iron oxidase, oxidizes Fe2+ to Fe3+, promotes iron efflux from cells to regulate iron homeostasis, inhibits lipid peroxidation mediated by the instability of the pool of intracellular Fe2+ and prevents neurotoxicity [150]; in the absence of physiological activity of CP proteins, Fe2+ cannot be oxidized, leading to a significant iron accumulation in astrocytes and loss of glial-derived growth factors essential for neurons [151]. CP knockout mice lead to progressive astrocyte and neuronal death and contribute to neurodegenerative processes [152]. According to recent research, the CSF of AD patients contains substantially less CP than normal [153]. In Aβ25-35-induced or transgenic APP expression-induced AD mouse models, knockout of the gene encoding CP increased memory impairment and iron accumulation and Aβ-induced hippocampal neuronal damage and iron levels were attenuated by injection of exogenous CP expression plasmids into the ventricles [154]. Notably, astrocytes appear to be primarily involved in the transport of iron rather than its accumulation, as they have the lowest metabolic prerequisites for iron in neuroglia [155]. However, in the case of brain aging, an increase in the number of iron-positive astrocytes can be observed, perhaps because iron is stored not only in ferritin but also in the mitochondria of astrocytes, iron overload mediates mitochondrial dysfunction and triggers oxidative stress [156].

Unlike CP, heme oxygenase-1 (HO-1) breaks down heme into carbon monoxide, free iron and biliverdin [157]. It acts as a sensor of mitochondrial iron chelation in astrocytes, and overexpression of HO-1 in astrocytes contributes to the pathological deposition of non-transferrin iron and adjacent neuronal damage. In contrast, using the iron chelator DFO effectively reduces the death of cultured astrocytes [158]. Furthermore, it was discovered that, in comparison to controls, HO-1 was substantially more immunoreactive in astrocytes and neurons in the cerebral cortex and hippocampus and it colocalized with neurogenic fibrous tangles and amyloid senile plaques in AD patients [159]. This suggests neuronal death in AD pathogenesis is related to elevated astrocyte expression of HO-1 and consequent iron accumulation. Within the brain’s glia and white matter, butyrylcholinesterase (BChE) participates in cholinergic neurotransmission alongside acetylcholinesterase [160]. In AD mice, pathogenic Aβ and NFT were discovered to colocalize with BChE, indicating that this protein may be implicated in the etiology of AD [161]. In particular, the recent identification of a possible IRE in the 3-UTR of BChE based on mRNA sequence analysis suggests that BChE may be regulated by iron metabolism, while further studies have found that BChE levels are iron-dependent and are expressed predominantly in brain astrocytes and BChE generated by reactive astrocytes can hasten the breakdown of acetylcholine in the aging brain, which causes cognitive function to deteriorate [160, 162]. Additionally, ferroptosis is induced in astrocytes and may contribute to pathogenesis in the inner olfactory brain according to scRNA-seq study and astrocyte ferroptosis further accelerates mood and cognitive deficits in AD [163]. Moreover, TNF-α treatment of astrocytes increased DMT1 expression and decreased Fpn1 expression in vitro, whereas TGF-β1 treatment had no effect on DMT1 expression but increased Fpn1 expression in astrocytes [141], suggesting that iron metabolism in astrocytes is also regulated by cellular inflammatory factors in the CNS. Based on the previously listed AD models, astrocyte-bound or released proteins affect iron homeostasis, which in turn may affect ferroptosis. Thus, strategies targeting different protein expression in astrocytes may help prevent neuronal death, particularly via ferroptosis.

Further, given that astrocytes are Cu depots, dysregulation of Cu metabolism in the brain is characteristic of neurodegenerative diseases, including AD [164]. The cytotoxicity of copper disrupts mitochondrial integrity by aggregating lipoylated proteins and causing the loss of Fe-S cluster proteins, thereby triggering cuproptosis, leads to oxidative stress and mitochondrial dysfunction, which has something in common with the pathological mechanism of ferroptosis [165]. In fact, in addition to the mitochondrial tricarboxylic acid cycle, GSH is a common hub of ferroptosis and cuproptosis, albeit with different roles, in ferroptosis GSH acts as an antioxidant, preventing lipid peroxidation, while in cuproptosis it acts as a copper companion, binding copper to mitigate the aggregation of lipoylated proteins [166]. The depletion of GSH levels is a feature of brain aging and has been associated with the progression of neurodegenerative diseases, including impaired cognitive function and ferroptosis in AD patients [167]. Therefore, it may be speculated that cuproptosis and ferroptosis may have a cross talk in AD, but the upstream and downstream molecules that mediate the common pathological mechanism need to be further studied.

6.3 Oligodendrocytes

The brain’s oligodendrocytes are the cells with the highest iron content. Iron is bound by oligodendrocytes as metabolic substrates for myelin synthesis and maintenance; the formation of myelin sheaths is correlated with both direct and indirect iron uptake [168]. Indeed, ferritin is found in AD patients, mainly in oligodendrocytes and myelinated axons of glial dystrophy [169]. By comparing cortical myelin formation profiles with Aβ deposition profiles, it was found that age-related myelin breakdown liberates iron release from oligodendrocytes, which promotes Aβ oligomerization in brain parenchyma [170]. White matter abnormalities due to myelin and oligodendrocyte damage have been shown to promote cognitive impairment and AD pathogenesis [171]. Notably, one of the most susceptible cell types to oxidative stress is the oligodendrocyte due to its intrinsic iron-rich composition and low antioxidant concentration and as age increases, iron levels in the brain gradually increase and oligodendrocytes are highly susceptible to increased damage due to oxidative stress and DNA damage [172]. A recent MRI study of AD patients found a negative correlation between iron levels and myelin levels in key brain regions with age, suggesting that myelin breakdown may release large amounts of accumulated iron, further promoting myelin breakdown and neurodegeneration [173]. Additionally, FTH secreted by oligodendrocytes prevents oxidative damage and death caused by neuronal iron deposition [115]. In a multiple sclerosis model induced by cuprizone, ferritin’s quick mobilization of iron causes ferroptosis, leading to oligodendrocyte loss and iron-mediated lipid peroxidation and demyelination [174]. This suggests that ferroptosis in AD may also be caused by lipid peroxidation and rapid ferritin mobilization-mediated oligodendrocyte loss, as oligodendrocyte demyelination is a natural source of iron for loading iron into neurons under conditions such as aging and pathological inflammation.

7. Glia Activation Induces Ferroptosis by Increasing Oxidative Stress
7.1 Microglia

Molecular mechanisms of ferroptosis in AD have been implicated in increased oxidative stress. Neuronal networks are attenuated by glial cells via a variety of pathways that inhibit the entry of excess ROS into neurons, with the targets they provide being valuable for restoring the degenerating metabolic status of neurons. Activation of microglia induces neuronal ferroptosis in AD through increased oxidative stress. One study found that Aβ25-35 induces excessive amounts of ROS in BV2 microglia through NADPH oxidase; when intracellular iron is depleted, Aβ25-35-induced ROS is inhibited [175]. Further research found that when primary neurons and microglia were co-cultured with iron and LPS, there was a significant decrease in the number of neuronal cells and a dose-dependent increase in ROS generation in the treated microglia, suggesting that iron significantly increased microglia ROS production and induced neurotoxicity and targeting NADPH oxidase may be a therapeutic direction for iron-induced microglia-related inflammation and neurotoxicity [176]. Moreover, in acute inflammatory injury and LPS-induced inflammation, increased microglia HO-1 expression, iron deposition and iron toxicity accumulation further increases ROS, decreased GPX4 expression, and enhanced oxidative stress in the brain, leading to cognitive impairment and behavioral disorder; in this context, these features are significantly improved and partially restored when the HO-1 gene is knocked out, or iron chelators are used in mice, suggesting that inhibition of microglia HO-1 activity or application of iron chelators might be a potential therapeutic in slowing down AD progression [177]. According to a recent study, ferroptosis susceptibility is modulated by inducible nitric oxide synthase (iNOS)/NO•-enrichment in activated M1 macrophages/microglia. iNOS knockdown increased the sensitivity of macrophages and microglia to RAS-selective lethal 3 (RSL3) (ferroptosis agonist)-induced ferroptosis, however, this phenomenon was effectively inhibited by the use of ferrostatin-1 (a ferroptosis inhibitor), indicating that iNOS is a potent target for the regulation of inflammation and ferroptosis [178].

7.2 Astrocytes

When reactive, astrocytes release a variety of antioxidant molecules such as GSH, hepcidin and Nrf2, which are critical regulators in iron metabolism and oxidative stress neuroprotection [112, 179, 180]. Hepcidin is one of the key regulators of systemic iron homeostasis; astrocyte hepcidin regulates iron uptake at the BBB and hepcidin injection into the brain attenuates iron deposition [105]. In an Aβ-induced AD model, hepcidin treatment reduces Aβ-induced secretion of TNF-α and IL-6 in astrocytes, reduces neuroinflammation and pro-oxidative processes, the Aβ burden and AD symptoms can be alleviated by astrocyte hepcidin thereby protecting neighboring neurons from neurotoxicity [181, 182]. Another study found that hepcidin overexpression in APP/PS1 transgenic mice astrocytes reduced iron deposition caused by Aβ25-35, significantly reduced oxidative damage and neuroinflammation in the hippocampus, further improving cognitive performance [105]. Additionally, ferroptosis of astrocytes caused by oxidative stress in AD such as lipid peroxidation, DNA oxidation and mitochondrial breakage in mouse astrocytes is encouraged by Nrf2 deficiency [183]. Another study found that NADPH oxidase 4 (NOX4) serves as a significant generator of ROS, that NOX4 is significantly elevated in astrocytes in the cerebral cortex and mitochondrial metabolism was impaired in APP/PS1 double transgenic mice through oxidative stress-induced lipid peroxidation, which promoted iron toxicity in astrocytes [12], NOX4 silencing alleviated mitochondrial abnormalities, decreased Aβ and p-Tau levels, enhanced cognition and lessened ferroptosis [184].

7.3 Oligodendrocytes

Oligodendrocyte death, dysfunction and/or demyelination occur in AD [185]. It has been estimated that oligodendrocytes operate at the highest metabolic rate among brain cells that consume a lot of oxygen to generate adenosine triphosphate to provide energy for the production of myelin sheaths, resulting in the production of hydrogen peroxide and other ROS [186, 187]. Iron serves as an important cofactor in myelin synthesis; it provides raw material for myelin production and tends to trigger the Fenton reaction, leading to enhanced lipid peroxidation and promotion of ferroptosis [188]. Paradoxically, low antioxidant content in oligodendrocytes such as GSH further amplifies ferroptosis. A study has shown that liproxstatin-1 prevents ferroptosis; the use of liproxstatin-1 not only restored the expression of GPX4, GSH and iron death inhibitory proteins, but also inhibited oligodendrocyte mitochondrial lipid peroxidation and GPX4-induced iron death in oligodendrocytes [189]. This suggests that antioxidants targeting ferroptosis with age may be the direction of research to target non-neuronal cells. Since iron accumulation is associated with many neurological diseases, exploring oligodendrocytes and AD pathogenesis in future studies should be encouraged.

8. Prospects of Targeted Ferroptosis in AD Therapy
8.1 Clinical Trials of Iron Chelators for the Treatment of AD

One key mechanism causing AD neurodegeneration is ferroptosis. Therefore, in patients with AD, targeting ferroptosis may be a promising treatment option. Inhibitors of ferroptosis with current applications for treating and preventing AD are emerging (Table 1, Ref. [73, 105, 190, 191, 192, 193, 194, 195, 196, 197, 198, 199, 200, 201, 202, 203, 204, 205, 206, 207, 208, 209, 210, 211, 212, 213, 214]). Therapeutic effects in AD models have been demonstrated by iron chelators that are used in clinical settings, such as DFO, deferiprone (DFP) and deferasirox [215, 216]. In a 24-month, a single-blind trial of 48 patients with AD, intramuscular injection of DFO was found to reduce the rate of cognitive decline in these patients by 50% when compared to the normal controls [190]. DFO is challenging to transport across the BBB, has low bioavailability, the nasally administered DFO nanoparticles under investigation facilitate brain transport [217] and have a potential role in AD treatment. DFP readily crosses the BBB and chelates intracellular iron. It is currently being studied in a phase II randomized, double-blind, placebo-controlled clinical trial (ClinicalTrials.gov ID NCT03234686) designed to investigate the safety and efficacy of DFP in subjects with MCI, prodromal AD and mild AD [218]. DFP has a high safety profile, low systemic toxicity and is a viable strategy for anti-ferroptosis therapy in AD. Additionally, other iron chelators, such as chloroioquine and its derivatives, may also help with the treatment of AD by reducing tau and Aβ deposition and enhancing memory and cognition (Table 1). Chloroiodoquinoline and its derivatives are moderately mild iron chelators capable of chelating iron, zinc and copper and polybutylene terephthalate 2 (PBT2) is one of the chloroiodoquinoline derivatives that promotes Aβ degradation [219]. The tolerability and safety of PBT2 in AD treatment has been demonstrated in a randomized, double-blind, placebo-controlled phase IIa trial with reduced Aβ42 concentrations in CSF and improved cognition [220, 221]. Due to the potential of chloroiodoquinoline in AD treatment, researchers have synthesized several derivatives or hybrids to treat AD that are effective [222]; For example, the flurbiprofen-chloroiodonol hybrid, which has recently been used for AD treatment, has been shown to prevent the accumulation of Aβ and it resists oxidation and anti-neuroinflammation [223]. Additionally, antioxidants targeting ferroptosis compounds currently under development with therapeutic potential for AD, including Vitamin E, Alpha-lipoic acid, Selenium, Ferrostatin-1, Hepcidin, Coenzyme Q10 (CoQ10), N-acetylcysteine and Dexmedetomidine. They have all been shown to inhibit lipid peroxidation and exhibit beneficial aspects in cell and animal models of AD therapy (Table 1).

Table 1. Overview of available ferroptosis inhibitors in AD.
Sort Drug candidates Key results References
Iron Chelators Deferoxamine Cognitive decline in AD patients slowed by 50% [190]
Deferiprone Phase AD II clinical trials are ongoing; Reverses pathological brain iron accumulation; Reduces iron deposition, Aβ levels and cognitive impairment in rats [191, 192]
Deferasirox Blocks iron accumulation, reduces expression of ferritin and transferrin receptors; reverses changes in Aβ metabolism; alleviates Aβ-induced learning defects in AD rat model [193, 194]
Clioquinol Prevention of cognitive decline in 32 AD patients and reduction of plasma Aβ42 levels; reduces iron-associated Aβ42 aggregation [195, 196]
M30 Reduces APP expression and Aβ production; alleviates cognitive impairment in APP/PS1 mice; Reduces brain iron accumulation, Aβ and tau levels [197, 198]
HPO Neuroprotective effect on Aβ-induced mouse cortical neurons [199]
Hydroxylated chalcones Fights human neuroblastoma SH-SY5Y cells Aβ peptide aggregation and ferroptosis [200]
HLA20A Reduces APP expression and Aβ production and reduces iron-induced Aβ aggregation [201]
Tacrine Neuroprotective in neuroblastoma cells treatment with Aβ1-42 and ascorbate/iron stressors. AChE inhibition and antioxidant activity [202, 203]
Schiff bases Inhibit redox active metals and metal-induced Aβ aggregation [204]
Antioxidants Selenium Inhibit ferroptosis. Key regulator of the activity of GPX4. Improvement of cognitive impairment in AD animal models [205, 206]
Vitamin E Lipophilic free radicals capture antioxidants, fight ferroptosis, improve lipid ROS to prevent iron denaturation in GPX4-deficient cells, slows cognitive deterioration in patients with AD [207]
Alpha-Lipoic Acid Blocks tau induced iron overload and lipid peroxidation and improves cognitive function in AD patients. Slowed cognitive decline in 129 probable AD patients [208, 209]
Ferrostatin-1 Prevents neuronal death and memory loss. Reduces iron deposition and neuronal degeneration. Improves long-term motor and cognitive function [73, 210]
Liproxstatin-1 Aβ-induced memory improved in AD mice [73]
Hepcidin Reduces iron and Aβ levels in the hippocampus and cortex of APP/PS mice, improves cognitive function in mice [105]
Polyphenols Reduces iron accumulation, clears ROS, inhibits Aβ aggregation and P-tau, improves memory and cognitive function in APP/PS1 mice [211, 212]
FSP1 A FSP1-CoQ10-NAD(P)H pathway, along with GPX4 and GSH, inhibits ferroptosis and phospholipid peroxidation [213]
NQO1 Inhibits lipid peroxidation and maintains antioxidant forms of α-tocopherol, ascorbic acid and CoQ10 [214]

AD, Alzheimer’s disease; CoQ10, Coenzyme Q10; GSH, glutathione; GPX4, glutathione peroxidase 4; NQO1, NAD(P)H Quinone Dehydrogenase 1; FSP1, Ferroptosis suppressor protein-1; HPO, hydroxypyridin-4-ones.

8.2 The Role and Mechanism of Other Drugs in Regulating Ferroptosis in AD

Traditional metal chelators have a single function in terms of treatment and as previously mentioned that there may be crosstalk in the pathogenesis of AD with cuproptosis and ferroptosis, dual chelating agents like Cu chelators along with Fe chelatore, have also been studied in the treatment of AD. α-Lipoic acid is a metal ion (iron, copper) chelator with targeted ferroptosis, anti-inflammatory and antioxidant effects, which improves scopolamine-induced cognitive and memory deficits in rats by attenuating reactive astrocyte proliferation [224]. Additionally, it has been shown to act as a copper chelator to transfer copper from the extracellular to the intracellular milleu, to alleviate intracellular copper deficiency in AD neurons [225]. Similarly, salicylic acid has been widely used as an anti-inflammatory drug, and given its anti-neuroinflammatory effects in AD, researchers have developed a series of salicylamide derivatives with metal chelating Cu2+, Fe2+ capabilities, exhibiting good activity to inhibit self and Cu2+-induced Aβ aggregation [226]. Further, some natural compounds also have a potential role in targeting ferroptosis in AD. For example, curcumin has chelating activity against Fe3+, Cu2+ and Zn2+ and has been shown to attenuate pathological alterations in AD, while curcumin inhibits neuroinflammation in APOE4 transgenic mice via the endoplasmic reticulum stress pathway [227] and regulates microglial (M1/M2) polarization by inhibiting the TLR4/NF-κB pathway and TREM2 expression in BV2 cells, thereby attenuating LPS-induced inflammation [228]. Ginkgolide B prevents cognitive impairment by reducing oxidative stress, inflammation and ferroptosis in senescence-accelerated P8 mice [229]. Multifunctional metal chelators are able to modulate multiple targets simultaneously and have shown promise in the treatment of metal-induced neurotoxicity, however, their exact mechanism has not been determined clinically and parameters such as chelation specificity, mode of administration, bioavailability, BBB penetration ability and drug toxicity, must be considered when selecting appropriate multiple metal chelation therapies, especially for the case of drug toxicity, which must exhibit the lowest possible toxicity levels when providing durable intervention for metal-related neurodegenerative diseases.

Notably, some genes targeting Fe chelators along with the gene responsible for promoting ferroptosis or anti-ferroptosis genes should be considered. For example, NCOA4, as a selective autophagy receptor, has been identified to colocalize with endogenous LC3B and intracellular iron storage ferritin complexes. However, NCOA4-mediated ferritosis-induced ferroptosis dysfunction can accelerate the pathological process of AD [230], which suggests that targeting NCOA4-mediated ferritinophagy holds promise for preventing and treating AD. Recently, the role of Nrf2 as a key regulator against oxidative stress has attracted much attention. Nrf2 exerts an antiferroptosis role by regulating the expression of a large number of ferroptosis-related genes and key aspects of lipid metabolism, which play an important role in the development of AD, as brain cells are rich in lipid [231]. Future studies should examine the mechanisms of iron homeostasis dysregulation and ferroptosis, as well as the role of the Nrf2-regulated ferroptosis-related signaling pathway in the development of AD, both to determine new drugs that effectively treat AD and identify their key targets.

Despite the compelling evidence supporting iron’s involvement in the pathogenesis of AD, it is unclear what roles iron play both upstream and downstream in the pathophysiology of AD. More recently, a growing number of iron-chelating compounds and antioxidants have been found to show potential therapeutic benefits. Although it is still unclear as to whether these ferroptosis inhibitors are effective in treating AD patients, their performance has demonstrated encouraging results in AD models and future research into the use of drugs with these potential therapeutic properties (e.g., iron chelators and multifunctional metal chelators) and their to entry into clinical trials, improved bioavailability and ability to effectively cross the BBB into the brain for practical physiological functions will be a significant issue in ferroptosis-mediated AD therapeutic pharmacology research.

9. Conclusions

Given an aging population, early detection of AD is essential. Notwithstanding substantial progress in understanding the etiology of AD, drug development therapeutic strategies based on these discoveries have very limited efficacy in altering the course of the disease. Consequently, there are compelling reasons to investigate new therapeutic approaches. Ferroptosis research has recently made rapid progress in understanding the pathophysiology of AD. Dysregulation of brain iron homeostasis exacerbates neurotoxicity and cognitive impairment. Thus, targeted ferroptosis inhibitors gain new momentum in the treatment of AD. Nevertheless, further study is required to pinpoint the exact cause of neurodegenerative diseases linked to ferroptosis. The current review elucidates the function of iron homeostasis between neuronal and glial cells in AD pathogenesis. Disturbed iron metabolism in glial cells mediates inflammation, oxidative stress and other pathological processes damaging neurons; iron deposition in neurons also leads to glial cell hyperactivation, accompanied by the release of proinflammatory factors and iron metabolism molecules by overactive microglia and astrocytes, mediating neuronal cell death in AD pathology. There are few studies on the mechanism of ferroptosis in oligodendrocytes in AD. Oligodendrocytes are high in iron content and tightly wrapped around neuronal axons; further study is necessary to investigate the ferroptosis induced by oligodendrocytes in AD neurons during aging.

In summary, ferroptosis is involved in the interaction between glial cells and neurons, thereby regulating the pathogenesis of AD. However, experimental animal models are the primary focus of current ferroptosis research and changes in ferroptosis in AD patients cannot be fully simulated. Comprehensive preclinical investigations are necessary to completely understand the precise molecular and cellular pathways of iron metabolism in AD neuronal degeneration, even while clinical trials assess the potential treatment benefits focused on ferroptosis.

Author Contributions

YY and WD conceived the perspective of the work. JX, RS, MQ, and BX drafted the manuscript. JX, ZZ, BX, and YJ designed the figure. RS and MQ collect and sort references. All authors contributed to editorial changes in the manuscript. All authors read and approved the final manuscript. All authors have participated sufficiently in the work and agreed to be accountable for all aspects of the work.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

Not applicable.

Funding

This work was supported by the National Natural Science Foundation of China (31871031, WD), Ministry of Science and Technology of the People’s Republic of China (2019YFE0120600), and Sichuan Science and Technology Program (2023NSFSC0593, YY; 2022YFS0615, BX), the Science and Technology Planning Project of Luzhou (2022-JYJ-140 to YY).

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Ward RJ, Zucca FA, Duyn JH, Crichton RR, Zecca L. The role of iron in brain ageing and neurodegenerative disorders. The Lancet. Neurology. 2014; 13: 1045–1060. https://doi.org/10.1016/S1474-4422(14)70117-6.
[2]
Kenkhuis B, Bush AI, Ayton S. How iron can drive neurodegeneration. Trends in Neurosciences. 2023; 46: 333–335. https://doi.org/10.1016/j.tins.2023.02.003.
[3]
Chiou B, Neal EH, Bowman AB, Lippmann ES, Simpson IA, Connor JR. Endothelial cells are critical regulators of iron transport in a model of the human blood-brain barrier. Journal of Cerebral Blood Flow and Metabolism: Official Journal of the International Society of Cerebral Blood Flow and Metabolism. 2019; 39: 2117–2131. https://doi.org/10.1177/0271678X18783372.
[4]
Li J, Cao F, Yin HL, Huang ZJ, Lin ZT, Mao N, et al. Ferroptosis: past, present and future. Cell Death & Disease. 2020; 11: 88. https://doi.org/10.1038/s41419-020-2298-2.
[5]
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 Biology. 2020; 32: 101494. https://doi.org/10.1016/j.redox.2020.101494.
[6]
Ayton S, Portbury S, Kalinowski P, Agarwal P, Diouf I, Schneider JA, et al. Regional brain iron associated with deterioration in Alzheimer’s disease: A large cohort study and theoretical significance. Alzheimer’s & Dementia: the Journal of the Alzheimer’s Association. 2021; 17: 1244–1256. https://doi.org/10.1002/alz.12282.
[7]
Smith MA, Harris PL, Sayre LM, Perry G. Iron accumulation in Alzheimer disease is a source of redox-generated free radicals. Proceedings of the National Academy of Sciences of the United States of America. 1997; 94: 9866–9868. https://doi.org/10.1073/pnas.94.18.9866.
[8]
Gao L, Jiang Z, Cai Z, Cai M, Zhang Q, Ma Y, et al. Brain iron deposition analysis using susceptibility weighted imaging and its association with body iron level in patients with mild cognitive impairment. Molecular Medicine Reports. 2017; 16: 8209–8215. https://doi.org/10.3892/mmr.2017.7668.
[9]
Ayton S, Faux NG, Bush AI, Alzheimer’s Disease Neuroimaging Initiative. Ferritin levels in the cerebrospinal fluid predict Alzheimer’s disease outcomes and are regulated by APOE. Nature Communications. 2015; 6: 6760. https://doi.org/10.1038/ncomms7760.
[10]
Gong L, Sun J, Cong S. Levels of iron and iron-related proteins in Alzheimer’s disease: A systematic review and meta-analysis. Journal of Trace Elements in Medicine and Biology: Organ of the Society for Minerals and Trace Elements (GMS). 2023; 80: 127304. https://doi.org/10.1016/j.jtemb.2023.127304.
[11]
Gong W, Fu Y, Wu BS, Du J, Yang L, Zhang YR, et al. Whole-exome sequencing identifies protein-coding variants associated with brain iron in 29,828 individuals. Nature Communications. 2024; 15: 5540. https://doi.org/10.1038/s41467-024-49702-2.
[12]
Park MW, Cha HW, Kim J, Kim JH, Yang H, Yoon S, et al. NOX4 promotes ferroptosis of astrocytes by oxidative stress-induced lipid peroxidation via the impairment of mitochondrial metabolism in Alzheimer’s diseases. Redox Biology. 2021; 41: 101947. https://doi.org/10.1016/j.redox.2021.101947.
[13]
Liddell JR, Hilton JBW, Kysenius K, Billings JL, Nikseresht S, McInnes LE, et al. Microglial ferroptotic stress causes non-cell autonomous neuronal death. Molecular Neurodegeneration. 2024; 19: 14. https://doi.org/10.1186/s13024-023-00691-8.
[14]
Coulthard EJ, Love S. A broader view of dementia: multiple co-pathologies are the norm. Brain: a Journal of Neurology. 2018; 141: 1894–1897. https://doi.org/10.1093/brain/awy153.
[15]
Zhang J, Zhang Y, Wang J, Xia Y, Zhang J, Chen L. Recent advances in Alzheimer’s disease: Mechanisms, clinical trials and new drug development strategies. Signal Transduction and Targeted Therapy. 2024; 9: 211. https://doi.org/10.1038/s41392-024-01911-3.
[16]
Plascencia-Villa G, Perry G. Preventive and Therapeutic Strategies in Alzheimer’s Disease: Focus on Oxidative Stress, Redox Metals, and Ferroptosis. Antioxidants & Redox Signaling. 2021; 34: 591–610. https://doi.org/10.1089/ars.2020.8134.
[17]
Maniscalchi A, Benzi Juncos ON, Conde MA, Funk MI, Fermento ME, Facchinetti MM, et al. New insights on neurodegeneration triggered by iron accumulation: Intersections with neutral lipid metabolism, ferroptosis, and motor impairment. Redox Biology. 2024; 71: 103074. https://doi.org/10.1016/j.redox.2024.103074.
[18]
Ma J, Haldar S, Khan MA, Sharma SD, Merrick WC, Theil EC, et al. Fe2+ binds iron responsive element-RNA, selectively changing protein-binding affinities and regulating mRNA repression and activation. Proceedings of the National Academy of Sciences of the United States of America. 2012; 109: 8417–8422. https://doi.org/10.1073/pnas.1120045109.
[19]
Rouault TA. The role of iron regulatory proteins in mammalian iron homeostasis and disease. Nature Chemical Biology. 2006; 2: 406–414. https://doi.org/10.1038/nchembio807.
[20]
Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell. 2012; 149: 1060–1072. https://doi.org/10.1016/j.cell.2012.03.042.
[21]
Wilkinson N, Pantopoulos K. The IRP/IRE system in vivo: insights from mouse models. Frontiers in Pharmacology. 2014; 5: 176. https://doi.org/10.3389/fphar.2014.00176.
[22]
Barra J, Crosbourne I, Roberge CL, Bossardi-Ramos R, Warren JSA, Matteson K, et al. DMT1-dependent endosome-mitochondria interactions regulate mitochondrial iron translocation and metastatic outgrowth. Oncogene. 2024; 43: 650–667. https://doi.org/10.1038/s41388-023-02933-x.
[23]
Li LB, Chai R, Zhang S, Xu SF, Zhang YH, Li HL, et al. Iron Exposure and the Cellular Mechanisms Linked to Neuron Degeneration in Adult Mice. Cells. 2019; 8: 198. https://doi.org/10.3390/cells8020198.
[24]
Rochette L, Dogon G, Rigal E, Zeller M, Cottin Y, Vergely C. Lipid Peroxidation and Iron Metabolism: Two Corner Stones in the Homeostasis Control of Ferroptosis. International Journal of Molecular Sciences. 2022; 24: 449. https://doi.org/10.3390/ijms24010449.
[25]
Qian ZM, Ke Y. Brain iron transport. Biological Reviews of the Cambridge Philosophical Society. 2019; 94: 1672–1684. https://doi.org/10.1111/brv.12521.
[26]
Dlouhy AC, Bailey DK, Steimle BL, Parker HV, Kosman DJ. Fluorescence resonance energy transfer links membrane ferroportin, hephaestin but not ferroportin, amyloid precursor protein complex with iron efflux. The Journal of Biological Chemistry. 2019; 294: 4202–4214. https://doi.org/10.1074/jbc.RA118.005142.
[27]
Zheng J, Jiang R, Chen M, Maimaitiming Z, Wang J, Anderson GJ, et al. Multi-Copper Ferroxidase-Deficient Mice Have Increased Brain Iron Concentrations and Learning and Memory Deficits. The Journal of Nutrition. 2018; 148: 643–649. https://doi.org/10.1093/jn/nxy012.
[28]
Bao WD, Pang P, Zhou XT, Hu F, Xiong W, Chen K, et al. Correction: Loss of ferroportin induces memory impairment by promoting ferroptosis in Alzheimer’s disease. Cell Death and Differentiation. 2024; 31: 1099. https://doi.org/10.1038/s41418-024-01290-w.
[29]
Wu Y, Torabi SF, Lake RJ, Hong S, Yu Z, Wu P, et al. Simultaneous Fe2+/Fe3+ imaging shows Fe3+ over Fe2+ enrichment in Alzheimer’s disease mouse brain. Science Advances. 2023; 9: eade7622. https://doi.org/10.1126/sciadv.ade7622.
[30]
James SA, Churches QI, de Jonge MD, Birchall IE, Streltsov V, McColl G, et al. Iron, Copper, and Zinc Concentration in Aβ Plaques in the APP/PS1 Mouse Model of Alzheimer’s Disease Correlates with Metal Levels in the Surrounding Neuropil. ACS Chemical Neuroscience. 2017; 8: 629–637. https://doi.org/10.1021/acschemneuro.6b00362.
[31]
Spotorno N, Acosta-Cabronero J, Stomrud E, Lampinen B, Strandberg OT, van Westen D, et al. Relationship between cortical iron and tau aggregation in Alzheimer’s disease. Brain: a Journal of Neurology. 2020; 143: 1341–1349. https://doi.org/10.1093/brain/awaa089.
[32]
Telling ND, Everett J, Collingwood JF, Dobson J, van der Laan G, Gallagher JJ, et al. Iron Biochemistry is Correlated with Amyloid Plaque Morphology in an Established Mouse Model of Alzheimer’s Disease. Cell Chemical Biology. 2017; 24: 1205–1215.e3. https://doi.org/10.1016/j.chembiol.2017.07.014.
[33]
Cho HH, Cahill CM, Vanderburg CR, Scherzer CR, Wang B, Huang X, et al. Selective translational control of the Alzheimer amyloid precursor protein transcript by iron regulatory protein-1. The Journal of Biological Chemistry. 2010; 285: 31217–31232. https://doi.org/10.1074/jbc.M110.149161.
[34]
Long JM, Maloney B, Rogers JT, Lahiri DK. Novel upregulation of amyloid-β precursor protein (APP) by microRNA-346 via targeting of APP mRNA 5’-untranslated region: Implications in Alzheimer’s disease. Molecular Psychiatry. 2019; 24: 345–363. https://doi.org/10.1038/s41380-018-0266-3.
[35]
McCarthy RC, Park YH, Kosman DJ. sAPP modulates iron efflux from brain microvascular endothelial cells by stabilizing the ferrous iron exporter ferroportin. EMBO Reports. 2014; 15: 809–815. https://doi.org/10.15252/embr.201338064.
[36]
Belaidi AA, Gunn AP, Wong BX, Ayton S, Appukuttan AT, Roberts BR, et al. Marked Age-Related Changes in Brain Iron Homeostasis in Amyloid Protein Precursor Knockout Mice. Neurotherapeutics: the Journal of the American Society for Experimental NeuroTherapeutics. 2018; 15: 1055–1062. https://doi.org/10.1007/s13311-018-0656-x.
[37]
Mezzanotte M, Ammirata G, Boido M, Stanga S, Roetto A. Activation of the Hepcidin-Ferroportin1 pathway in the brain and astrocytic-neuronal crosstalk to counteract iron dyshomeostasis during aging. Scientific Reports. 2022; 12: 11724. https://doi.org/10.1038/s41598-022-15812-4.
[38]
Afram E, Lauritzen I, Bourgeois A, El Manaa W, Duplan E, Chami M, et al. The η-secretase-derived APP fragment ηCTF is localized in Golgi, endosomes and extracellular vesicles and contributes to Aβ production. Cellular and Molecular Life Sciences: CMLS. 2023; 80: 97. https://doi.org/10.1007/s00018-023-04737-4.
[39]
Everett J, Céspedes E, Shelford LR, Exley C, Collingwood JF, Dobson J, et al. Ferrous iron formation following the co-aggregation of ferric iron and the Alzheimer’s disease peptide β-amyloid (1-42). Journal of the Royal Society, Interface. 2014; 11: 20140165. https://doi.org/10.1098/rsif.2014.0165.
[40]
Everett J, Brooks J, Lermyte F, O’Connor PB, Sadler PJ, Dobson J, et al. Iron stored in ferritin is chemically reduced in the presence of aggregating Aβ(1-42). Scientific Reports. 2020; 10: 10332. https://doi.org/10.1038/s41598-020-67117-z.
[41]
Boopathi S, Kolandaivel P. Fe(2+) binding on amyloid β-peptide promotes aggregation. Proteins. 2016; 84: 1257–1274. https://doi.org/10.1002/prot.25075.
[42]
Hansra GK, Popov G, Banaczek PO, Vogiatzis M, Jegathees T, Goldsbury CS, et al. The neuritic plaque in Alzheimer’s disease: perivascular degeneration of neuronal processes. Neurobiology of Aging. 2019; 82: 88–101. https://doi.org/10.1016/j.neurobiolaging.2019.06.009.
[43]
Vossel KA, Xu JC, Fomenko V, Miyamoto T, Suberbielle E, Knox JA, et al. Tau reduction prevents Aβ-induced axonal transport deficits by blocking activation of GSK3β. The Journal of Cell Biology. 2015; 209: 419–433. https://doi.org/10.1083/jcb.201407065.
[44]
Wang S, Jiang Y, Liu Y, Liu Q, Sun H, Mei M, et al. Ferroptosis promotes microtubule-associated protein tau aggregation via GSK-3β activation and proteasome inhibition. Molecular Neurobiology. 2022; 59: 1486–1501. https://doi.org/10.1007/s12035-022-02731-8.
[45]
Guo C, Wang P, Zhong ML, Wang T, Huang XS, Li JY, et al. Deferoxamine inhibits iron induced hippocampal tau phosphorylation in the Alzheimer transgenic mouse brain. Neurochemistry International. 2013; 62: 165–172. https://doi.org/10.1016/j.neuint.2012.12.005.
[46]
Tuo QZ, Lei P, Jackman KA, Li XL, Xiong H, Li XL, et al. Tau-mediated iron export prevents ferroptotic damage after ischemic stroke. Molecular Psychiatry. 2017; 22: 1520–1530. https://doi.org/10.1038/mp.2017.171.
[47]
Lei P, Ayton S, Finkelstein DI, Spoerri L, Ciccotosto GD, Wright DK, et al. Tau deficiency induces parkinsonism with dementia by impairing APP-mediated iron export. Nature Medicine. 2012; 18: 291–295. https://doi.org/10.1038/nm.2613.
[48]
Mezzanotte M, Stanga S. Brain Iron Dyshomeostasis and Ferroptosis in Alzheimer’s Disease Pathophysiology: Two Faces of the Same Coin. Aging and Disease. 2024. (online ahead of print)
[49]
Chai B, Wu Y, Yang H, Fan B, Cao S, Zhang X, et al. Tau Aggregation-Dependent Lipid Peroxide Accumulation Driven by the hsa_circ_0001546/14-3-3/CAMK2D/Tau Complex Inhibits Epithelial Ovarian Cancer Peritoneal Metastasis. Advanced Science (Weinheim, Baden-Wurttemberg, Germany). 2024; 11: e2310134. https://doi.org/10.1002/advs.202310134.
[50]
Wan W, Cao L, Kalionis B, Murthi P, Xia S, Guan Y. Iron Deposition Leads to Hyperphosphorylation of Tau and Disruption of Insulin Signaling. Frontiers in Neurology. 2019; 10: 607. https://doi.org/10.3389/fneur.2019.00607.
[51]
Chen M, Zheng J, Liu G, Zeng C, Xu E, Zhu W, et al. High Dietary Iron Disrupts Iron Homeostasis and Induces Amyloid-β and Phospho-τ Expression in the Hippocampus of Adult Wild-Type and APP/PS1 Transgenic Mice. The Journal of Nutrition. 2019; 149: 2247–2254. https://doi.org/10.1093/jn/nxz168.
[52]
Guo C, Wang T, Zheng W, Shan ZY, Teng WP, Wang ZY. Intranasal deferoxamine reverses iron-induced memory deficits and inhibits amyloidogenic APP processing in a transgenic mouse model of Alzheimer’s disease. Neurobiology of Aging. 2013; 34: 562–575. https://doi.org/10.1016/j.neurobiolaging.2012.05.009.
[53]
Zhang Y, He ML. Deferoxamine enhances alternative activation of microglia and inhibits amyloid beta deposits in APP/PS1 mice. Brain Research. 2017; 1677: 86–92. https://doi.org/10.1016/j.brainres.2017.09.019.
[54]
Tang M, Chen Z, Wu D, Chen L. Ferritinophagy/ferroptosis: Iron-related newcomers in human diseases. Journal of Cellular Physiology. 2018; 233: 9179–9190. https://doi.org/10.1002/jcp.26954.
[55]
Fuhrmann DC, Mondorf A, Beifuß J, Jung M, Brüne B. Hypoxia inhibits ferritinophagy, increases mitochondrial ferritin, and protects from ferroptosis. Redox Biology. 2020; 36: 101670. https://doi.org/10.1016/j.redox.2020.101670.
[56]
LeVine SM. Examining the Role of a Functional Deficiency of Iron in Lysosomal Storage Disorders with Translational Relevance to Alzheimer’s Disease. Cells. 2023; 12: 2641. https://doi.org/10.3390/cells12222641.
[57]
Belaidi AA, Masaldan S, Southon A, Kalinowski P, Acevedo K, Appukuttan AT, et al. Apolipoprotein E potently inhibits ferroptosis by blocking ferritinophagy. Molecular Psychiatry. 2024; 29: 211–220. https://doi.org/10.1038/s41380-022-01568-w.
[58]
Núñez MT, Hidalgo C. Noxious Iron-Calcium Connections in Neurodegeneration. Frontiers in Neuroscience. 2019; 13: 48. https://doi.org/10.3389/fnins.2019.00048.
[59]
Hidalgo C, Carrasco MA, Muñoz P, Núñez MT. A role for reactive oxygen/nitrogen species and iron on neuronal synaptic plasticity. Antioxidants & Redox Signaling. 2007; 9: 245–255. https://doi.org/10.1089/ars.2007.9.245.
[60]
Kishida KT, Hoeffer CA, Hu D, Pao M, Holland SM, Klann E. Synaptic plasticity deficits and mild memory impairments in mouse models of chronic granulomatous disease. Molecular and Cellular Biology. 2006; 26: 5908–5920. https://doi.org/10.1128/MCB.00269-06.
[61]
Lee DG, Park J, Lee HS, Lee SR, Lee DS. Iron overload-induced calcium signals modulate mitochondrial fragmentation in HT-22 hippocampal neuron cells. Toxicology. 2016; 365: 17–24. https://doi.org/10.1016/j.tox.2016.07.022.
[62]
Sanmartín CD, Paula-Lima AC, García A, Barattini P, Hartel S, Núñez MT, et al. Ryanodine receptor-mediated Ca(2+) release underlies iron-induced mitochondrial fission and stimulates mitochondrial Ca(2+) uptake in primary hippocampal neurons. Frontiers in Molecular Neuroscience. 2014; 7: 13. https://doi.org/10.3389/fnmol.2014.00013.
[63]
Marmolejo-Garza A, Krabbendam IE, Luu MDA, Brouwer F, Trombetta-Lima M, Unal O, et al. Negative modulation of mitochondrial calcium uniporter complex protects neurons against ferroptosis. Cell Death & Disease. 2023; 14: 772. https://doi.org/10.1038/s41419-023-06290-1.
[64]
Pelizzoni I, Macco R, Morini MF, Zacchetti D, Grohovaz F, Codazzi F. Iron handling in hippocampal neurons: activity-dependent iron entry and mitochondria-mediated neurotoxicity. Aging Cell. 2011; 10: 172–183. https://doi.org/10.1111/j.1474-9726.2010.00652.x.
[65]
Pedrera L, Espiritu RA, Ros U, Weber J, Schmitt A, Stroh J, et al. Ferroptotic pores induce Ca2+ fluxes and ESCRT-III activation to modulate cell death kinetics. Cell Death and Differentiation. 2021; 28: 1644–1657. https://doi.org/10.1038/s41418-020-00691-x.
[66]
Berridge MJ. Calcium hypothesis of Alzheimer’s disease. Pflugers Archiv: European Journal of Physiology. 2010; 459: 441–449. https://doi.org/10.1007/s00424-009-0736-1.
[67]
Alzheimer’s Association Calcium Hypothesis Workgroup. Calcium Hypothesis of Alzheimer’s disease and brain aging: A framework for integrating new evidence into a comprehensive theory of pathogenesis. Alzheimer’s & Dementia: the Journal of the Alzheimer’s Association. 2017; 13: 178–182.e17. https://doi.org/10.1016/j.jalz.2016.12.006.
[68]
Bezprozvanny I, Mattson MP. Neuronal calcium mishandling and the pathogenesis of Alzheimer’s disease. Trends in Neurosciences. 2008; 31: 454–463. https://doi.org/10.1016/j.tins.2008.06.005.
[69]
Ayton S, Wang Y, Diouf I, Schneider JA, Brockman J, Morris MC, et al. Brain iron is associated with accelerated cognitive decline in people with Alzheimer pathology. Molecular Psychiatry. 2020; 25: 2932–2941. https://doi.org/10.1038/s41380-019-0375-7.
[70]
Huang L, McClatchy DB, Maher P, Liang Z, Diedrich JK, Soriano-Castell D, et al. Intracellular amyloid toxicity induces oxytosis/ferroptosis regulated cell death. Cell Death & Disease. 2020; 11: 828. https://doi.org/10.1038/s41419-020-03020-9.
[71]
Verbruggen L, Ates G, Lara O, De Munck J, Villers A, De Pauw L, et al. Lifespan extension with preservation of hippocampal function in aged system xc-deficient male mice. Molecular Psychiatry. 2022; 27: 2355–2368. https://doi.org/10.1038/s41380-022-01470-5.
[72]
D’Ezio V, Colasanti M, Persichini T. Amyloid-β 25-35 Induces Neurotoxicity through the Up-Regulation of Astrocytic System Xc. Antioxidants (Basel, Switzerland). 2021; 10: 1685. https://doi.org/10.3390/antiox10111685.
[73]
Bao WD, Pang P, Zhou XT, Hu F, Xiong W, Chen K, et al. Loss of ferroportin induces memory impairment by promoting ferroptosis in Alzheimer’s disease. Cell Death and Differentiation. 2021; 28: 1548–1562. https://doi.org/10.1038/s41418-020-00685-9.
[74]
O’Keeffe R, Latunde-Dada GO, Chen YL, Kong XL, Cilibrizzi A, Hider RC. Glutathione and the intracellular labile heme pool. Biometals: an International Journal on the Role of Metal Ions in Biology, Biochemistry, and Medicine. 2021; 34: 221–228. https://doi.org/10.1007/s10534-020-00274-w.
[75]
Chen JJ, Thiyagarajah M, Song J, Chen C, Herrmann N, Gallagher D, et al. Altered central and blood glutathione in Alzheimer’s disease and mild cognitive impairment: a meta-analysis. Alzheimer’s Research & Therapy. 2022; 14: 23. https://doi.org/10.1186/s13195-022-00961-5.
[76]
Boyd-Kimball D, Sultana R, Abdul HM, Butterfield DA. Gamma-glutamylcysteine ethyl ester-induced up-regulation of glutathione protects neurons against Abeta(1-42)-mediated oxidative stress and neurotoxicity: implications for Alzheimer’s disease. Journal of Neuroscience Research. 2005; 79: 700–706. https://doi.org/10.1002/jnr.20394.
[77]
Chen L, Dar NJ, Na R, McLane KD, Yoo K, Han X, et al. Enhanced defense against ferroptosis ameliorates cognitive impairment and reduces neurodegeneration in 5xFAD mice. Free Radical Biology & Medicine. 2022; 180: 1–12. https://doi.org/10.1016/j.freeradbiomed.2022.01.002.
[78]
Hambright WS, Fonseca RS, Chen L, Na R, Ran Q. Ablation of ferroptosis regulator glutathione peroxidase 4 in forebrain neurons promotes cognitive impairment and neurodegeneration. Redox Biology. 2017; 12: 8–17. https://doi.org/10.1016/j.redox.2017.01.021.
[79]
Padhi D, Baruah P, Ramesh M, Moorthy H, Govindaraju T. Hybrid molecules synergistically mitigate ferroptosis and amyloid-associated toxicities in Alzheimer’s disease. Redox Biology. 2024; 71: 103119. https://doi.org/10.1016/j.redox.2024.103119.
[80]
Greenough MA, Lane DJR, Balez R, Anastacio HTD, Zeng Z, Ganio K, et al. Selective ferroptosis vulnerability due to familial Alzheimer’s disease presenilin mutations. Cell Death and Differentiation. 2022; 29: 2123–2136. https://doi.org/10.1038/s41418-022-01003-1.
[81]
Domínguez-González M, Puigpinós M, Jové M, Naudi A, Portero-Otín M, Pamplona R, et al. Regional vulnerability to lipoxidative damage and inflammation in normal human brain aging. Experimental Gerontology. 2018; 111: 218–228. https://doi.org/10.1016/j.exger.2018.07.023.
[82]
Lim PJ, Duarte TL, Arezes J, Garcia-Santos D, Hamdi A, Pasricha SR, et al. Nrf2 controls iron homeostasis in haemochromatosis and thalassaemia via Bmp6 and hepcidin. Nature Metabolism. 2019; 1: 519–531. https://doi.org/10.1038/s42255-019-0063-6.
[83]
Bahn G, Park JS, Yun UJ, Lee YJ, Choi Y, Park JS, et al. NRF2/ARE pathway negatively regulates BACE1 expression and ameliorates cognitive deficits in mouse Alzheimer’s models. Proceedings of the National Academy of Sciences of the United States of America. 2019; 116: 12516–12523. https://doi.org/10.1073/pnas.1819541116.
[84]
Zhang Y, Wang M, Chang W. Iron dyshomeostasis and ferroptosis in Alzheimer’s disease: Molecular mechanisms of cell death and novel therapeutic drugs and targets for AD. Frontiers in Pharmacology. 2022; 13: 983623. https://doi.org/10.3389/fphar.2022.983623.
[85]
Gonzales MM, O’Donnell A, Ghosh S, Thibault E, Tanner J, Satizabal CL, et al. Associations of cerebral amyloid beta and tau with cognition from midlife. Alzheimer’s & Dementia: the Journal of the Alzheimer’s Association. 2024; 20: 5901–5911. https://doi.org/10.1002/alz.14060.
[86]
Ayton S, Faux NG, Bush AI. Association of Cerebrospinal Fluid Ferritin Level With Preclinical Cognitive Decline in APOE-ε4 Carriers. JAMA Neurology. 2017; 74: 122–125. https://doi.org/10.1001/jamaneurol.2016.4406.
[87]
Duro MV, Ebright B, Yassine HN. Lipids and brain inflammation in APOE4-associated dementia. Current Opinion in Lipidology. 2022; 33: 16–24. https://doi.org/10.1097/MOL.0000000000000801.
[88]
Xu H, Perreau VM, Dent KA, Bush AI, Finkelstein DI, Adlard PA. Iron Regulates Apolipoprotein E Expression and Secretion in Neurons and Astrocytes. Journal of Alzheimer’s Disease: JAD. 2016; 51: 471–487. https://doi.org/10.3233/JAD-150797.
[89]
Faraji P, Kühn H, Ahmadian S. Multiple Roles of Apolipoprotein E4 in Oxidative Lipid Metabolism and Ferroptosis During the Pathogenesis of Alzheimer’s Disease. Journal of Molecular Neuroscience: MN. 2024; 74: 62. https://doi.org/10.1007/s12031-024-02224-4.
[90]
Haney MS, Pálovics R, Munson CN, Long C, Johansson PK, Yip O, et al. APOE4/4 is linked to damaging lipid droplets in Alzheimer’s disease microglia. Nature. 2024; 628: 154–161. https://doi.org/10.1038/s41586-024-07185-7.
[91]
Li Y, Ruan X, Sun M, Yuan M, Song J, Zhou Z, et al. Iron deposition participates in LPS-induced cognitive impairment by promoting neuroinflammation and ferroptosis in mice. Experimental Neurology. 2024; 379: 114862. https://doi.org/10.1016/j.expneurol.2024.114862.
[92]
Yan N, Xu Z, Qu C, Zhang J. Dimethyl fumarate improves cognitive deficits in chronic cerebral hypoperfusion rats by alleviating inflammation, oxidative stress, and ferroptosis via NRF2/ARE/NF-κB signal pathway. International Immunopharmacology. 2021; 98: 107844. https://doi.org/10.1016/j.intimp.2021.107844.
[93]
Liu P, Chen W, Kang Y, Wang C, Wang X, Liu W, et al. Silibinin ameliorates STING-mediated neuroinflammation via downregulation of ferroptotic damage in a sporadic Alzheimer’s disease model. Archives of Biochemistry and Biophysics. 2023; 744: 109691. https://doi.org/10.1016/j.abb.2023.109691.
[94]
Chen Y, Fang ZM, Yi X, Wei X, Jiang DS. The interaction between ferroptosis and inflammatory signaling pathways. Cell Death & Disease. 2023; 14: 205. https://doi.org/10.1038/s41419-023-05716-0.
[95]
Mohan S, Alhazmi HA, Hassani R, Khuwaja G, Maheshkumar VP, Aldahish A, et al. Role of ferroptosis pathways in neuroinflammation and neurological disorders: From pathogenesis to treatment. Heliyon. 2024; 10: e24786. https://doi.org/10.1016/j.heliyon.2024.e24786.
[96]
Choi DH, Kwon KC, Hwang DJ, Koo JH, Um HS, Song HS, et al. Treadmill Exercise Alleviates Brain Iron Dyshomeostasis Accelerating Neuronal Amyloid-β Production, Neuronal Cell Death, and Cognitive Impairment in Transgenic Mice Model of Alzheimer’s Disease. Molecular Neurobiology. 2021; 58: 3208–3223. https://doi.org/10.1007/s12035-021-02335-8.
[97]
Majerníková N, den Dunnen WFA, Dolga AM. The Potential of Ferroptosis-Targeting Therapies for Alzheimer’s Disease: From Mechanism to Transcriptomic Analysis. Frontiers in Aging Neuroscience. 2021; 13: 745046. https://doi.org/10.3389/fnagi.2021.745046.
[98]
Qin D, Wang J, Le A, Wang TJ, Chen X, Wang J. Traumatic Brain Injury: Ultrastructural Features in Neuronal Ferroptosis, Glial Cell Activation and Polarization, and Blood-Brain Barrier Breakdown. Cells. 2021; 10: 1009. https://doi.org/10.3390/cells10051009.
[99]
Santambrogio P, Ripamonti M, Cozzi A, Raimondi M, Cavestro C, Di Meo I, et al. Massive iron accumulation in PKAN-derived neurons and astrocytes: light on the human pathological phenotype. Cell Death & Disease. 2022; 13: 185. https://doi.org/10.1038/s41419-022-04626-x.
[100]
Gong L, Yu Q, Wang H, Xu C, Dou Y, Mao B, et al. Neurofilament Light Chain (NF-L) Stimulates Lipid Peroxidation to Neuronal Membrane through Microglia-Derived Ferritin Heavy Chain (FTH) Secretion. Oxidative Medicine and Cellular Longevity. 2022; 2022: 3938940. https://doi.org/10.1155/2022/3938940.
[101]
Stanca S, Rossetti M, Bokulic Panichi L, Bongioanni P. The Cellular Dysfunction of the Brain-Blood Barrier from Endothelial Cells to Astrocytes: The Pathway towards Neurotransmitter Impairment in Schizophrenia. International Journal of Molecular Sciences. 2024; 25: 1250. https://doi.org/10.3390/ijms25021250.
[102]
Zechel S, Huber-Wittmer K, von Bohlen und Halbach O. Distribution of the iron-regulating protein hepcidin in the murine central nervous system. Journal of Neuroscience Research. 2006; 84: 790–800. https://doi.org/10.1002/jnr.20991.
[103]
Billesbølle CB, Azumaya CM, Kretsch RC, Powers AS, Gonen S, Schneider S, et al. Structure of hepcidin-bound ferroportin reveals iron homeostatic mechanisms. Nature. 2020; 586: 807–811. https://doi.org/10.1038/s41586-020-2668-z.
[104]
Du F, Qian ZM, Luo Q, Yung WH, Ke Y. Hepcidin Suppresses Brain Iron Accumulation by Downregulating Iron Transport Proteins in Iron-Overloaded Rats. Molecular Neurobiology. 2015; 52: 101–114. https://doi.org/10.1007/s12035-014-8847-x.
[105]
Xu Y, Zhang Y, Zhang JH, Han K, Zhang X, Bai X, et al. Astrocyte hepcidin ameliorates neuronal loss through attenuating brain iron deposition and oxidative stress in APP/PS1 mice. Free Radical Biology & Medicine. 2020; 158: 84–95. https://doi.org/10.1016/j.freeradbiomed.2020.07.012.
[106]
Benkovic SA, Connor JR. Ferritin, transferrin, and iron in selected regions of the adult and aged rat brain. The Journal of Comparative Neurology. 1993; 338: 97–113. https://doi.org/10.1002/cne.903380108.
[107]
Verkhratsky A. Physiology of neuronal-glial networking. Neurochemistry International. 2010; 57: 332–343. https://doi.org/10.1016/j.neuint.2010.02.002.
[108]
Lee J, Hyun DH. The Interplay between Intracellular Iron Homeostasis and Neuroinflammation in Neurodegenerative Diseases. Antioxidants (Basel, Switzerland). 2023; 12: 918. https://doi.org/10.3390/antiox12040918.
[109]
Pandur E, Varga E, Tamási K, Pap R, Nagy J, Sipos K. Effect of Inflammatory Mediators Lipopolysaccharide and Lipoteichoic Acid on Iron Metabolism of Differentiated SH-SY5Y Cells Alters in the Presence of BV-2 Microglia. International Journal of Molecular Sciences. 2018; 20: 17. https://doi.org/10.3390/ijms20010017.
[110]
Cui Z, Zhong Z, Yang Y, Wang B, Sun Y, Sun Q, et al. Ferrous Iron Induces Nrf2 Expression in Mouse Brain Astrocytes to Prevent Neurotoxicity. Journal of Biochemical and Molecular Toxicology. 2016; 30: 396–403. https://doi.org/10.1002/jbt.21803.
[111]
Liddell JR, Robinson SR, Dringen R. Endogenous glutathione and catalase protect cultured rat astrocytes from the iron-mediated toxicity of hydrogen peroxide. Neuroscience Letters. 2004; 364: 164–167. https://doi.org/10.1016/j.neulet.2004.04.042.
[112]
Ishii T, Warabi E, Mann GE. Circadian control of BDNF-mediated Nrf2 activation in astrocytes protects dopaminergic neurons from ferroptosis. Free Radical Biology & Medicine. 2019; 133: 169–178. https://doi.org/10.1016/j.freeradbiomed.2018.09.002.
[113]
Cheli VT, Santiago González DA, Wan Q, Denaroso G, Wan R, Rosenblum SL, et al. H-ferritin expression in astrocytes is necessary for proper oligodendrocyte development and myelination. Glia. 2021; 69: 2981–2998. https://doi.org/10.1002/glia.24083.
[114]
Carden TR, Correale J, Pasquini JM, Pérez MJ. Transferrin Enhances Microglial Phagocytic Capacity. Molecular Neurobiology. 2019; 56: 6324–6340. https://doi.org/10.1007/s12035-019-1519-0.
[115]
Mukherjee C, Kling T, Russo B, Miebach K, Kess E, Schifferer M, et al. Oligodendrocytes Provide Antioxidant Defense Function for Neurons by Secreting Ferritin Heavy Chain. Cell Metabolism. 2020; 32: 259–272.e10. https://doi.org/10.1016/j.cmet.2020.05.019.
[116]
Jha MK, Lee WH, Suk K. Functional polarization of neuroglia: Implications in neuroinflammation and neurological disorders. Biochemical Pharmacology. 2016; 103: 1–16. https://doi.org/10.1016/j.bcp.2015.11.003.
[117]
Nagele RG, D’Andrea MR, Lee H, Venkataraman V, Wang HY. Astrocytes accumulate A beta 42 and give rise to astrocytic amyloid plaques in Alzheimer disease brains. Brain Research. 2003; 971: 197–209. https://doi.org/10.1016/s0006-8993(03)02361-8.
[118]
Akiyama H, Mori H, Saido T, Kondo H, Ikeda K, McGeer PL. Occurrence of the diffuse amyloid beta-protein (Abeta) deposits with numerous Abeta-containing glial cells in the cerebral cortex of patients with Alzheimer’s disease. Glia. 1999; 25: 324–331. https://doi.org/10.1002/(sici)1098-1136(19990215)25:4<324::aid-glia2>3.0.co;2-5.
[119]
Liddelow SA, Guttenplan KA, Clarke LE, Bennett FC, Bohlen CJ, Schirmer L, et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature. 2017; 541: 481–487. https://doi.org/10.1038/nature21029.
[120]
Mitew S, Kirkcaldie MTK, Halliday GM, Shepherd CE, Vickers JC, Dickson TC. Focal demyelination in Alzheimer’s disease and transgenic mouse models. Acta Neuropathologica. 2010; 119: 567–577. https://doi.org/10.1007/s00401-010-0657-2.
[121]
Ryan SK, Zelic M, Han Y, Teeple E, Chen L, Sadeghi M, et al. Microglia ferroptosis is regulated by SEC24B and contributes to neurodegeneration. Nature Neuroscience. 2023; 26: 12–26. https://doi.org/10.1038/s41593-022-01221-3.
[122]
Urrutia P, Aguirre P, Esparza A, Tapia V, Mena NP, Arredondo M, et al. Inflammation alters the expression of DMT1, FPN1 and hepcidin, and it causes iron accumulation in central nervous system cells. Journal of Neurochemistry. 2013; 126: 541–549. https://doi.org/10.1111/jnc.12244.
[123]
Thomsen MS, Andersen MV, Christoffersen PR, Jensen MD, Lichota J, Moos T. Neurodegeneration with inflammation is accompanied by accumulation of iron and ferritin in microglia and neurons. Neurobiology of Disease. 2015; 81: 108–118. https://doi.org/10.1016/j.nbd.2015.03.013.
[124]
Bishop GM, Dang TN, Dringen R, Robinson SR. Accumulation of non-transferrin-bound iron by neurons, astrocytes, and microglia. Neurotoxicity Research. 2011; 19: 443–451. https://doi.org/10.1007/s12640-010-9195-x.
[125]
McCarthy RC, Sosa JC, Gardeck AM, Baez AS, Lee CH, Wessling-Resnick M. Inflammation-induced iron transport and metabolism by brain microglia. The Journal of Biological Chemistry. 2018; 293: 7853–7863. https://doi.org/10.1074/jbc.RA118.001949.
[126]
Sharawy N, Imam AAA, Aboulhoda BE, Khalifa MM, Morcos GNB, Abd Algaleel WA, et al. Iron dyshomeostasis and time-course changes in iron-uptake systems and ferritin level in relation to pro-inflammatory microglia polarization in sepsis-induced encephalopathy. Frontiers in Physiology. 2022; 13: 953206. https://doi.org/10.3389/fphys.2022.953206.
[127]
Kenkhuis B, Somarakis A, de Haan L, Dzyubachyk O, IJsselsteijn ME, de Miranda NFCC, et al. Iron loading is a prominent feature of activated microglia in Alzheimer’s disease patients. Acta Neuropathologica Communications. 2021; 9: 27. https://doi.org/10.1186/s40478-021-01126-5.
[128]
Donley DW, Realing M, Gigley JP, Fox JH. Iron activates microglia and directly stimulates indoleamine-2,3-dioxygenase activity in the N171-82Q mouse model of Huntington’s disease. PloS One. 2021; 16: e0250606. https://doi.org/10.1371/journal.pone.0250606.
[129]
Meng FX, Hou JM, Sun TS. In vivo evaluation of microglia activation by intracranial iron overload in central pain after spinal cord injury. Journal of Orthopaedic Surgery and Research. 2017; 12: 75. https://doi.org/10.1186/s13018-017-0578-z.
[130]
Wang J, Song N, Jiang H, Wang J, Xie J. Pro-inflammatory cytokines modulate iron regulatory protein 1 expression and iron transportation through reactive oxygen/nitrogen species production in ventral mesencephalic neurons. Biochimica et Biophysica Acta. 2013; 1832: 618–625. https://doi.org/10.1016/j.bbadis.2013.01.021.
[131]
Healy S, McMahon J, Owens P, FitzGerald U. Significant glial alterations in response to iron loading in a novel organotypic hippocampal slice culture model. Scientific Reports. 2016; 6: 36410. https://doi.org/10.1038/srep36410.
[132]
Zeineh MM, Chen Y, Kitzler HH, Hammond R, Vogel H, Rutt BK. Activated iron-containing microglia in the human hippocampus identified by magnetic resonance imaging in Alzheimer disease. Neurobiology of Aging. 2015; 36: 2483–2500. https://doi.org/10.1016/j.neurobiolaging.2015.05.022.
[133]
van Duijn S, Bulk M, van Duinen SG, Nabuurs RJA, van Buchem MA, van der Weerd L, et al. Cortical Iron Reflects Severity of Alzheimer’s Disease. Journal of Alzheimer’s Disease: JAD. 2017; 60: 1533–1545. https://doi.org/10.3233/JAD-161143.
[134]
Angelova DM, Brown DR. Altered Processing of β-Amyloid in SH-SY5Y Cells Induced by Model Senescent Microglia. ACS Chemical Neuroscience. 2018; 9: 3137–3152. https://doi.org/10.1021/acschemneuro.8b00334.
[135]
McIntosh A, Mela V, Harty C, Minogue AM, Costello DA, Kerskens 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 Pathology (Zurich, Switzerland). 2019; 29: 606–621. https://doi.org/10.1111/bpa.12704.
[136]
Lopes KO, Sparks DL, Streit WJ. Microglial dystrophy in the aged and Alzheimer’s disease brain is associated with ferritin immunoreactivity. Glia. 2008; 56: 1048–1060. https://doi.org/10.1002/glia.20678.
[137]
Streit WJ, Rotter J, Winter K, Müller W, Khoshbouei H, Bechmann I. Droplet Degeneration of Hippocampal and Cortical Neurons Signifies the Beginning of Neuritic Plaque Formation. Journal of Alzheimer’s Disease: JAD. 2022; 85: 1701–1720. https://doi.org/10.3233/JAD-215334.
[138]
Nnah IC, Lee CH, Wessling-Resnick M. Iron potentiates microglial interleukin-1β secretion induced by amyloid-β. Journal of Neurochemistry. 2020; 154: 177–189. https://doi.org/10.1111/jnc.14906.
[139]
Peters DG, Pollack AN, Cheng KC, Sun D, Saido T, Haaf MP, et al. Dietary lipophilic iron alters amyloidogenesis and microglial morphology in Alzheimer’s disease knock-in APP mice. Metallomics: Integrated Biometal Science. 2018; 10: 426–443. https://doi.org/10.1039/c8mt00004b.
[140]
Wu T, Ning S, Zhang H, Cao Y, Li X, Hao J, et al. Role of ferroptosis in neuroimmunity and neurodegeneration in multiple sclerosis revealed by multi-omics data. Journal of Cellular and Molecular Medicine. 2024; 28: e18396. https://doi.org/10.1111/jcmm.18396.
[141]
Rathore KI, Redensek A, David S. Iron homeostasis in astrocytes and microglia is differentially regulated by TNF-α and TGF-β1. Glia. 2012; 60: 738–750. https://doi.org/10.1002/glia.22303.
[142]
Zhou S, Du X, Xie J, Wang J. Interleukin-6 regulates iron-related proteins through c-Jun N-terminal kinase activation in BV2 microglial cell lines. PloS One. 2017; 12: e0180464. https://doi.org/10.1371/journal.pone.0180464.
[143]
Tsatsanis A, McCorkindale AN, Wong BX, Patrick E, Ryan TM, Evans RW, et al. The acute phase protein lactoferrin is a key feature of Alzheimer’s disease and predictor of Aβ burden through induction of APP amyloidogenic processing. Molecular Psychiatry. 2021; 26: 5516–5531. https://doi.org/10.1038/s41380-021-01248-1.
[144]
Pérez MJ, Carden TR, Dos Santos Claro PA, Silberstein S, Páez PM, Cheli VT, et al. Transferrin Enhances Neuronal Differentiation. ASN Neuro. 2023; 15: 17590914231170703. https://doi.org/10.1177/17590914231170703.
[145]
Huang P, Wang J, Lin X, Yang FF, Tan JH. Effects of IL-10 on iron metabolism in LPS-induced inflammatory mice via modulating hepcidin expression. European Review for Medical and Pharmacological Sciences. 2017; 21: 3469–3475.
[146]
Li B, Xia M, Zorec R, Parpura V, Verkhratsky A. Astrocytes in heavy metal neurotoxicity and neurodegeneration. Brain Research. 2021; 1752: 147234. https://doi.org/10.1016/j.brainres.2020.147234.
[147]
McCarthy RC, Kosman DJ. Glial cell ceruloplasmin and hepcidin differentially regulate iron efflux from brain microvascular endothelial cells. PloS One. 2014; 9: e89003. https://doi.org/10.1371/journal.pone.0089003.
[148]
Zarruk JG, Berard JL, Passos dos Santos R, Kroner A, Lee J, Arosio P, et al. Expression of iron homeostasis proteins in the spinal cord in experimental autoimmune encephalomyelitis and their implications for iron accumulation. Neurobiology of Disease. 2015; 81: 93–107. https://doi.org/10.1016/j.nbd.2015.02.001.
[149]
Pelizzoni I, Zacchetti D, Campanella A, Grohovaz F, Codazzi F. Iron uptake in quiescent and inflammation-activated astrocytes: a potentially neuroprotective control of iron burden. Biochimica et Biophysica Acta. 2013; 1832: 1326–1333. https://doi.org/10.1016/j.bbadis.2013.04.007.
[150]
Liu H, Hua Y, Keep RF, Xi G. Brain Ceruloplasmin Expression After Experimental Intracerebral Hemorrhage and Protection Against Iron-Induced Brain Injury. Translational Stroke Research. 2019; 10: 112–119. https://doi.org/10.1007/s12975-018-0669-0.
[151]
Li ZD, Li H, Kang S, Cui YG, Zheng H, Wang P, et al. The divergent effects of astrocyte ceruloplasmin on learning and memory function in young and old mice. Cell Death & Disease. 2022; 13: 1006. https://doi.org/10.1038/s41419-022-05459-4.
[152]
Santiago González DA, Cheli VT, Rosenblum SL, Denaroso G, Paez PM. Ceruloplasmin deletion in myelinating glial cells induces myelin disruption and oxidative stress in the central and peripheral nervous systems. Redox Biology. 2021; 46: 102118. https://doi.org/10.1016/j.redox.2021.102118.
[153]
Jouini N, Saied Z, Ben Sassi S, Nebli F, Messaoud T, Hentati F, et al. Impacts of Iron Metabolism Dysregulation on Alzheimer’s Disease. Journal of Alzheimer’s Disease: JAD. 2021; 80: 1439–1450. https://doi.org/10.3233/JAD-201250.
[154]
Zhao YS, Zhang LH, Yu PP, Gou YJ, Zhao J, You LH, et al. Ceruloplasmin, a Potential Therapeutic Agent for Alzheimer’s Disease. Antioxidants & Redox Signaling. 2018; 28: 1323–1337. https://doi.org/10.1089/ars.2016.6883.
[155]
Kress GJ, Dineley KE, Reynolds IJ. The relationship between intracellular free iron and cell injury in cultured neurons, astrocytes, and oligodendrocytes. The Journal of Neuroscience: the Official Journal of the Society for Neuroscience. 2002; 22: 5848–5855. https://doi.org/10.1523/JNEUROSCI.22-14-05848.2002.
[156]
Santambrogio P, Cozzi A, Balestrucci C, Ripamonti M, Berno V, Cammarota E, et al. Mitochondrial iron deficiency triggers cytosolic iron overload in PKAN hiPS-derived astrocytes. Cell Death & Disease. 2024; 15: 361. https://doi.org/10.1038/s41419-024-06757-9.
[157]
Wu YH, Hsieh HL. Roles of Heme Oxygenase-1 in Neuroinflammation and Brain Disorders. Antioxidants (Basel, Switzerland). 2022; 11: 923. https://doi.org/10.3390/antiox11050923.
[158]
Görg B, Karababa A, Schütz E, Paluschinski M, Schrimpf A, Shafigullina A, et al. O-GlcNAcylation-dependent upregulation of HO1 triggers ammonia-induced oxidative stress and senescence in hepatic encephalopathy. Journal of Hepatology. 2019; 71: 930–941. https://doi.org/10.1016/j.jhep.2019.06.020.
[159]
Song W, Zukor H, Liberman A, Kaduri S, Arvanitakis Z, Bennett DA, et al. Astroglial heme oxygenase-1 and the origin of corpora amylacea in aging and degenerating neural tissues. Experimental Neurology. 2014; 254: 78–89. https://doi.org/10.1016/j.expneurol.2014.01.006.
[160]
Darreh-Shori T, Vijayaraghavan S, Aeinehband S, Piehl F, Lindblom RPF, Nilsson B, et al. Functional variability in butyrylcholinesterase activity regulates intrathecal cytokine and astroglial biomarker profiles in patients with Alzheimer’s disease. Neurobiology of Aging. 2013; 34: 2465–2481. https://doi.org/10.1016/j.neurobiolaging.2013.04.027.
[161]
Rejc L, Gómez-Vallejo V, Joya A, Moreno O, Egimendia A, Castellnou P, et al. Longitudinal evaluation of a novel BChE PET tracer as an early in vivo biomarker in the brain of a mouse model for Alzheimer disease. Theranostics. 2021; 11: 6542–6559. https://doi.org/10.7150/thno.54589.
[162]
Campillos M, Cases I, Hentze MW, Sanchez M. SIREs: searching for iron-responsive elements. Nucleic Acids Research. 2010; 38: W360–W367. https://doi.org/10.1093/nar/gkq371.
[163]
Dang Y, He Q, Yang S, Sun H, Liu Y, Li W, et al. FTH1- and SAT1-Induced Astrocytic Ferroptosis Is Involved in Alzheimer’s Disease: Evidence from Single-Cell Transcriptomic Analysis. Pharmaceuticals (Basel, Switzerland). 2022; 15: 1177. https://doi.org/10.3390/ph15101177.
[164]
Zhang Y, Zhou Q, Lu L, Su Y, Shi W, Zhang H, et al. Copper Induces Cognitive Impairment in Mice via Modulation of Cuproptosis and CREB Signaling. Nutrients. 2023; 15: 972. https://doi.org/10.3390/nu15040972.
[165]
Tang D, Chen X, Kroemer G. Cuproptosis: a copper-triggered modality of mitochondrial cell death. Cell Research. 2022; 32: 417–418. https://doi.org/10.1038/s41422-022-00653-7.
[166]
Tsvetkov P, Coy S, Petrova B, Dreishpoon M, Verma A, Abdusamad M, et al. Copper induces cell death by targeting lipoylated TCA cycle proteins. Science (New York, N.Y.). 2022; 375: 1254–1261. https://doi.org/10.1126/science.abf0529.
[167]
Maher P. Potentiation of glutathione loss and nerve cell death by the transition metals iron and copper: Implications for age-related neurodegenerative diseases. Free Radical Biology & Medicine. 2018; 115: 92–104. https://doi.org/10.1016/j.freeradbiomed.2017.11.015.
[168]
Emamnejad R, Pagnin M, Petratos S. The iron maiden: Oligodendroglial metabolic dysfunction in multiple sclerosis and mitochondrial signaling. Neuroscience and Biobehavioral Reviews. 2024; 164: 105788. https://doi.org/10.1016/j.neubiorev.2024.105788.
[169]
Quintana C. Contribution of analytical microscopies to human neurodegenerative diseases research (PSP and AD). Mini Reviews in Medicinal Chemistry. 2007; 7: 961–975. https://doi.org/10.2174/138955707781662654.
[170]
Bartzokis G, Lu PH, Mintz J. Human brain myelination and amyloid beta deposition in Alzheimer’s disease. Alzheimer’s & Dementia: the Journal of the Alzheimer’s Association. 2007; 3: 122–125. https://doi.org/10.1016/j.jalz.2007.01.019.
[171]
Nasrabady SE, Rizvi B, Goldman JE, Brickman AM. White matter changes in Alzheimer’s disease: a focus on myelin and oligodendrocytes. Acta Neuropathologica Communications. 2018; 6: 22. https://doi.org/10.1186/s40478-018-0515-3.
[172]
Tse KH, Herrup K. DNA damage in the oligodendrocyte lineage and its role in brain aging. Mechanisms of Ageing and Development. 2017; 161: 37–50. https://doi.org/10.1016/j.mad.2016.05.006.
[173]
Khattar N, Triebswetter C, Kiely M, Ferrucci L, Resnick SM, Spencer RG, et al. Investigation of the association between cerebral iron content and myelin content in normative aging using quantitative magnetic resonance neuroimaging. NeuroImage. 2021; 239: 118267. https://doi.org/10.1016/j.neuroimage.2021.118267.
[174]
Jhelum P, Santos-Nogueira E, Teo W, Haumont A, Lenoël I, Stys PK, et al. Ferroptosis Mediates Cuprizone-Induced Loss of Oligodendrocytes and Demyelination. The Journal of Neuroscience: the Official Journal of the Society for Neuroscience. 2020; 40: 9327–9341. https://doi.org/10.1523/JNEUROSCI.1749-20.2020.
[175]
Part K, Künnis-Beres K, Poska H, Land T, Shimmo R, Zetterström Fernaeus S. Amyloid β25-35 induced ROS-burst through NADPH oxidase is sensitive to iron chelation in microglial Bv2 cells. Brain Research. 2015; 1629: 282–290. https://doi.org/10.1016/j.brainres.2015.09.034.
[176]
Yauger YJ, Bermudez S, Moritz KE, Glaser E, Stoica B, Byrnes KR. Iron accentuated reactive oxygen species release by NADPH oxidase in activated microglia contributes to oxidative stress in vitro. Journal of Neuroinflammation. 2019; 16: 41. https://doi.org/10.1186/s12974-019-1430-7.
[177]
Fernández-Mendívil C, Luengo E, Trigo-Alonso P, García-Magro N, Negredo P, López MG. Protective role of microglial HO-1 blockade in aging: Implication of iron metabolism. Redox Biology. 2021; 38: 101789. https://doi.org/10.1016/j.redox.2020.101789.
[178]
Kapralov AA, Yang Q, Dar HH, Tyurina YY, Anthonymuthu TS, Kim R, et al. Redox lipid reprogramming commands susceptibility of macrophages and microglia to ferroptotic death. Nature Chemical Biology. 2020; 16: 278–290. https://doi.org/10.1038/s41589-019-0462-8.
[179]
Li S, Zhou C, Zhu Y, Chao Z, Sheng Z, Zhang Y, et al. Ferrostatin-1 alleviates angiotensin II (Ang II)- induced inflammation and ferroptosis in astrocytes. International Immunopharmacology. 2021; 90: 107179. https://doi.org/10.1016/j.intimp.2020.107179.
[180]
Qian ZM, Ke Y. Hepcidin and its therapeutic potential in neurodegenerative disorders. Medicinal Research Reviews. 2020; 40: 633–653. https://doi.org/10.1002/med.21631.
[181]
Urrutia PJ, Hirsch EC, González-Billault C, Núñez MT. Hepcidin attenuates amyloid beta-induced inflammatory and pro-oxidant responses in astrocytes and microglia. Journal of Neurochemistry. 2017; 142: 140–152. https://doi.org/10.1111/jnc.14005.
[182]
Zhang X, Gou YJ, Zhang Y, Li J, Han K, Xu Y, et al. Hepcidin overexpression in astrocytes alters brain iron metabolism and protects against amyloid-β induced brain damage in mice. Cell Death Discovery. 2020; 6: 113. https://doi.org/10.1038/s41420-020-00346-3.
[183]
Tang Z, Chen Z, Guo M, Peng Y, Xiao Y, Guan Z, et al. NRF2 Deficiency Promotes Ferroptosis of Astrocytes Mediated by Oxidative Stress in Alzheimer’s Disease. Molecular Neurobiology. 2024; 61: 7517–7533. https://doi.org/10.1007/s12035-024-04023-9.
[184]
Maimaiti Y, Su T, Zhang Z, Ma L, Zhang Y, Xu H. NOX4-mediated astrocyte ferroptosis in Alzheimer’s disease. Cell & Bioscience. 2024; 14: 88. https://doi.org/10.1186/s13578-024-01266-w.
[185]
Chen P, Guo Z, Zhou B. Disease-Associated Oligodendrocyte: New Player in Alzheimer’s Disease and CNS Pathologies. Journal of Integrative Neuroscience. 2023; 22: 90. https://doi.org/10.31083/j.jin2204090.
[186]
Mouzannar R, Miric SJ, Wiggins RC, Konat GW. Hydrogen peroxide induces rapid digestion of oligodendrocyte chromatin into high molecular weight fragments. Neurochemistry International. 2001; 38: 9–15. https://doi.org/10.1016/s0197-0186(00)00066-8.
[187]
McTigue DM, Tripathi RB. The life, death, and replacement of oligodendrocytes in the adult CNS. Journal of Neurochemistry. 2008; 107: 1–19. https://doi.org/10.1111/j.1471-4159.2008.05570.x.
[188]
Cheli VT, Correale J, Paez PM, Pasquini JM. Iron Metabolism in Oligodendrocytes and Astrocytes, Implications for Myelination and Remyelination. ASN Neuro. 2020; 12: 1759091420962681. https://doi.org/10.1177/1759091420962681.
[189]
Fan BY, Pang YL, Li WX, Zhao CX, Zhang Y, Wang X, et al. Liproxstatin-1 is an effective inhibitor of oligodendrocyte ferroptosis induced by inhibition of glutathione peroxidase 4. Neural Regeneration Research. 2021; 16: 561–566. https://doi.org/10.4103/1673-5374.293157.
[190]
Crapper McLachlan DR, Dalton AJ, Kruck TP, Bell MY, Smith WL, Kalow W, et al. Intramuscular desferrioxamine in patients with Alzheimer’s disease. Lancet (London, England). 1991; 337: 1304–1308. https://doi.org/10.1016/0140-6736(91)92978-b.
[191]
Pandolfo M, Arpa J, Delatycki MB, Le Quan Sang KH, Mariotti C, Munnich A, et al. Deferiprone in Friedreich ataxia: a 6-month randomized controlled trial. Annals of Neurology. 2014; 76: 509–521. https://doi.org/10.1002/ana.24248.
[192]
Fawzi SF, Menze ET, Tadros MG. Deferiprone ameliorates memory impairment in Scopolamine-treated rats: The impact of its iron-chelating effect on β-amyloid disposition. Behavioural Brain Research. 2020; 378: 112314. https://doi.org/10.1016/j.bbr.2019.112314.
[193]
Kamalinia G, Khodagholi F, Atyabi F, Amini M, Shaerzadeh F, Sharifzadeh M, et al. Enhanced brain delivery of deferasirox-lactoferrin conjugates for iron chelation therapy in neurodegenerative disorders: in vitro and in vivo studies. Molecular Pharmaceutics. 2013; 10: 4418–4431. https://doi.org/10.1021/mp4002014.
[194]
Banerjee P, Sahoo A, Anand S, Bir A, Chakrabarti S. The Oral Iron Chelator, Deferasirox, Reverses the Age-Dependent Alterations in Iron and Amyloid-β Homeostasis in Rat Brain: Implications in the Therapy of Alzheimer’s Disease. Journal of Alzheimer’s Disease: JAD. 2016; 49: 681–693. https://doi.org/10.3233/JAD-150514.
[195]
Ritchie CW, Bush AI, Mackinnon A, Macfarlane S, Mastwyk M, MacGregor L, et al. Metal-protein attenuation with iodochlorhydroxyquin (clioquinol) targeting Abeta amyloid deposition and toxicity in Alzheimer disease: a pilot phase 2 clinical trial. Archives of Neurology. 2003; 60: 1685–1691. https://doi.org/10.1001/archneur.60.12.1685.
[196]
Tahmasebinia F, Emadi S. Effect of metal chelators on the aggregation of beta-amyloid peptides in the presence of copper and iron. Biometals: an International Journal on the Role of Metal Ions in Biology, Biochemistry, and Medicine. 2017; 30: 285–293. https://doi.org/10.1007/s10534-017-0005-2.
[197]
Kupershmidt L, Amit T, Bar-Am O, Youdim MBH, Weinreb O. The novel multi-target iron chelating-radical scavenging compound M30 possesses beneficial effects on major hallmarks of Alzheimer’s disease. Antioxidants & Redox Signaling. 2012; 17: 860–877. https://doi.org/10.1089/ars.2011.4279.
[198]
Amit T, Bar-Am O, Mechlovich D, Kupershmidt L, Youdim MBH, Weinreb O. The novel multitarget iron chelating and propargylamine drug M30 affects APP regulation and processing activities in Alzheimer’s disease models. Neuropharmacology. 2017; 123: 359–367. https://doi.org/10.1016/j.neuropharm.2017.05.026.
[199]
Gaeta A, Molina-Holgado F, Kong XL, Salvage S, Fakih S, Francis PT, et al. Synthesis, physical-chemical characterisation and biological evaluation of novel 2-amido-3-hydroxypyridin-4(1H)-ones: Iron chelators with the potential for treating Alzheimer’s disease. Bioorganic & Medicinal Chemistry. 2011; 19: 1285–1297. https://doi.org/10.1016/j.bmc.2010.12.007.
[200]
Cong L, Dong X, Wang Y, Deng Y, Li B, Dai R. On the role of synthesized hydroxylated chalcones as dual functional amyloid-β aggregation and ferroptosis inhibitors for potential treatment of Alzheimer’s disease. European Journal of Medicinal Chemistry. 2019; 166: 11–21. https://doi.org/10.1016/j.ejmech.2019.01.039.
[201]
Zheng H, Youdim MBH, Fridkin M. Selective acetylcholinesterase inhibitor activated by acetylcholinesterase releases an active chelator with neurorescuing and anti-amyloid activities. ACS Chemical Neuroscience. 2010; 1: 737–746. https://doi.org/10.1021/cn100069c.
[202]
Chand K, Alsoghier HM, Chaves S, Santos MA. Tacrine-(hydroxybenzoyl-pyridone) hybrids as potential multifunctional anti-Alzheimer’s agents: AChE inhibition, antioxidant activity and metal chelating capacity. Journal of Inorganic Biochemistry. 2016; 163: 266–277. https://doi.org/10.1016/j.jinorgbio.2016.05.005.
[203]
Chand K, Rajeshwari, Candeias E, Cardoso SM, Chaves S, Santos MA. Tacrine-deferiprone hybrids as multi-target-directed metal chelators against Alzheimer’s disease: a two-in-one drug. Metallomics: Integrated Biometal Science. 2018; 10: 1460–1475. https://doi.org/10.1039/c8mt00143j.
[204]
D’Acunto CW, Kaplánek R, Gbelcová H, Kejík Z, Bříza T, Vasina L, et al. Metallomics for Alzheimer’s disease treatment: Use of new generation of chelators combining metal-cation binding and transport properties. European Journal of Medicinal Chemistry. 2018; 150: 140–155. https://doi.org/10.1016/j.ejmech.2018.02.084.
[205]
Ingold I, Berndt C, Schmitt S, Doll S, Poschmann G, Buday K, et al. Selenium Utilization by GPX4 Is Required to Prevent Hydroperoxide-Induced Ferroptosis. Cell. 2018; 172: 409–422.e21. https://doi.org/10.1016/j.cell.2017.11.048.
[206]
Zhang ZH, Wu QY, Chen C, Zheng R, Chen Y, Ni JZ, et al. Comparison of the effects of selenomethionine and selenium-enriched yeast in the triple-transgenic mouse model of Alzheimer’s disease. Food & Function. 2018; 9: 3965–3973. https://doi.org/10.1039/c7fo02063e.
[207]
Hu Q, Zhang Y, Lou H, Ou Z, Liu J, Duan W, et al. GPX4 and vitamin E cooperatively protect hematopoietic stem and progenitor cells from lipid peroxidation and ferroptosis. Cell Death & Disease. 2021; 12: 706. https://doi.org/10.1038/s41419-021-04008-9.
[208]
Fava A, Pirritano D, Plastino M, Cristiano D, Puccio G, Colica C, et al. The Effect of Lipoic Acid Therapy on Cognitive Functioning in Patients with Alzheimer’s Disease. Journal of Neurodegenerative Diseases. 2013; 2013: 454253. https://doi.org/10.1155/2013/454253.
[209]
Song X, Long D. Nrf2 and Ferroptosis: A New Research Direction for Neurodegenerative Diseases. Frontiers in Neuroscience. 2020; 14: 267. https://doi.org/10.3389/fnins.2020.00267.
[210]
Xie BS, Wang YQ, Lin Y, Mao Q, Feng JF, Gao GY, et al. Inhibition of ferroptosis attenuates tissue damage and improves long-term outcomes after traumatic brain injury in mice. CNS Neuroscience & Therapeutics. 2019; 25: 465–475. https://doi.org/10.1111/cns.13069.
[211]
Ates G, Goldberg J, Currais A, Maher P. CMS121, a fatty acid synthase inhibitor, protects against excess lipid peroxidation and inflammation and alleviates cognitive loss in a transgenic mouse model of Alzheimer’s disease. Redox Biology. 2020; 36: 101648. https://doi.org/10.1016/j.redox.2020.101648.
[212]
Ege D. Action Mechanisms of Curcumin in Alzheimer’s Disease and Its Brain Targeted Delivery. Materials (Basel, Switzerland). 2021; 14: 3332. https://doi.org/10.3390/ma14123332.
[213]
Doll S, Freitas FP, Shah R, Aldrovandi M, da Silva MC, Ingold I, et al. FSP1 is a glutathione-independent ferroptosis suppressor. Nature. 2019; 575: 693–698. https://doi.org/10.1038/s41586-019-1707-0.
[214]
Ross D, Siegel D. The diverse functionality of NQO1 and its roles in redox control. Redox Biology. 2021; 41: 101950. https://doi.org/10.1016/j.redox.2021.101950.
[215]
Gleason A, Bush AI. Iron and Ferroptosis as Therapeutic Targets in Alzheimer’s Disease. Neurotherapeutics: the Journal of the American Society for Experimental NeuroTherapeutics. 2021; 18: 252–264. https://doi.org/10.1007/s13311-020-00954-y.
[216]
Wang F, Wang J, Shen Y, Li H, Rausch WD, Huang X. Iron Dyshomeostasis and Ferroptosis: A New Alzheimer’s Disease Hypothesis? Frontiers in Aging Neuroscience. 2022; 14: 830569. https://doi.org/10.3389/fnagi.2022.830569.
[217]
Rassu G, Soddu E, Cossu M, Brundu A, Cerri G, Marchetti N, et al. Solid microparticles based on chitosan or methyl-β-cyclodextrin: a first formulative approach to increase the nose-to-brain transport of deferoxamine mesylate. Journal of Controlled Release: Official Journal of the Controlled Release Society. 2015; 201: 68–77. https://doi.org/10.1016/j.jconrel.2015.01.025.
[218]
Deferiprone to Delay Dementia (The 3D Study). ClinicalTrials.gov identifier: NCT03234686. Accessed 8 October, 2024. https://clinicaltrials.gov/study/NCT03234686?cond=NCT03234686&rank=1
[219]
Crouch PJ, Savva MS, Hung LW, Donnelly PS, Mot AI, Parker SJ, et al. The Alzheimer’s therapeutic PBT2 promotes amyloid-β degradation and GSK3 phosphorylation via a metal chaperone activity. Journal of Neurochemistry. 2011; 119: 220–230. https://doi.org/10.1111/j.1471-4159.2011.07402.x.
[220]
Lannfelt L, Blennow K, Zetterberg H, Batsman S, Ames D, Harrison J, et al. Safety, efficacy, and biomarker findings of PBT2 in targeting Abeta as a modifying therapy for Alzheimer’s disease: a phase IIa, double-blind, randomised, placebo-controlled trial. The Lancet. Neurology. 2008; 7: 779–786. https://doi.org/10.1016/S1474-4422(08)70167-4.
[221]
Faux NG, Ritchie CW, Gunn A, Rembach A, Tsatsanis A, Bedo J, et al. PBT2 rapidly improves cognition in Alzheimer’s Disease: additional phase II analyses. Journal of Alzheimer’s Disease: JAD. 2010; 20: 509–516. https://doi.org/10.3233/JAD-2010-1390.
[222]
Perez DR, Sklar LA, Chigaev A. Clioquinol: To harm or heal. Pharmacology & Therapeutics. 2019; 199: 155–163. https://doi.org/10.1016/j.pharmthera.2019.03.009.
[223]
Yang Z, Song Q, Cao Z, Yu G, Liu Z, Tan Z, et al. Design, synthesis and evaluation of flurbiprofen-clioquinol hybrids as multitarget-directed ligands against Alzheimer’s disease. Bioorganic & Medicinal Chemistry. 2020; 28: 115374. https://doi.org/10.1016/j.bmc.2020.115374.
[224]
Memudu AE, Adewumi AE. Alpha lipoic acid ameliorates scopolamine induced memory deficit and neurodegeneration in the cerebello-hippocampal cortex. Metabolic Brain Disease. 2021; 36: 1729–1745. https://doi.org/10.1007/s11011-021-00720-9.
[225]
Metsla K, Kirss S, Laks K, Sildnik G, Palgi M, Palumaa T, et al. α-Lipoic Acid Has the Potential to Normalize Copper Metabolism, Which Is Dysregulated in Alzheimer’s Disease. Journal of Alzheimer’s Disease: JAD. 2022; 85: 715–728. https://doi.org/10.3233/JAD-215026.
[226]
Song Q, Li Y, Cao Z, Qiang X, Tan Z, Deng Y. Novel salicylamide derivatives as potent multifunctional agents for the treatment of Alzheimer’s disease: Design, synthesis and biological evaluation. Bioorganic Chemistry. 2019; 84: 137–149. https://doi.org/10.1016/j.bioorg.2018.11.022.
[227]
Kou J, Wang M, Shi J, Zhang H, Pu X, Song S, et al. Curcumin Reduces Cognitive Deficits by Inhibiting Neuroinflammation through the Endoplasmic Reticulum Stress Pathway in Apolipoprotein E4 Transgenic Mice. ACS Omega. 2021; 6: 6654–6662. https://doi.org/10.1021/acsomega.0c04810.
[228]
Zhang J, Zheng Y, Luo Y, Du Y, Zhang X, Fu J. Curcumin inhibits LPS-induced neuroinflammation by promoting microglial M2 polarization via TREM2/ TLR4/ NF-κB pathways in BV2 cells. Molecular Immunology. 2019; 116: 29–37. https://doi.org/10.1016/j.molimm.2019.09.020.
[229]
Shao L, Dong C, Geng D, He Q, Shi Y. Ginkgolide B protects against cognitive impairment in senescence-accelerated P8 mice by mitigating oxidative stress, inflammation and ferroptosis. Biochemical and Biophysical Research Communications. 2021; 572: 7–14. https://doi.org/10.1016/j.bbrc.2021.07.081.
[230]
Wang D, Wu Y, Zhou X, Liang C, Ma Y, Yuan Q, et al. Cadmium exposure induced neuronal ferroptosis and cognitive deficits via the mtROS-ferritinophagy pathway. Environmental Pollution (Barking, Essex: 1987). 2024; 349: 123958. https://doi.org/10.1016/j.envpol.2024.123958.
[231]
Shakya A, McKee NW, Dodson M, Chapman E, Zhang DD. Anti-Ferroptotic Effects of Nrf2: Beyond the Antioxidant Response. Molecules and Cells. 2023; 46: 165–175. https://doi.org/10.14348/molcells.2023.0005.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top