International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Cell death is a fundamental biological process that governs cell fate, tissue regeneration and host immune responses (1). Among the various forms of cell death, ferroptosis has recently emerged as a unique, iron-dependent modality of programmed cell death that has gained significant scientific interest (2). Morphologically, biochemically and genetically, ferroptosis is distinct from other well-established cell death modalities, including apoptosis, necrosis and autophagy (3). Rather than being a stochastic or uncontrolled process, ferroptosis is precisely orchestrated by complex signaling pathways and metabolic networks; its core mechanism involves the dysregulation and interplay of three pivotal modules: Iron, lipid and amino acid metabolism (4,5).
Cognitive impairment represents a major global public health challenge, encompassing a clinical spectrum from mild cognitive impairment to severe dementia. This condition profoundly affects the quality of life of patients and imposes a significant socioeconomic burden (6). This dysfunction serves as a common pathological endpoint for a multitude of neurological disorders. The present review focuses on the following key categories of disorders closely associated with cognitive impairment: Neurodegenerative diseases including Alzheimer's disease (AD) and Parkinson's disease (PD), which are pathologically characterized by the progressive loss of neurons in specific brain regions and the abnormal aggregation of functional proteins (7), and diabetes-associated cognitive impairment (DACI), which is caused mainly by factors such as insulin resistance and neuroinflammation (8).
Accumulating evidence indicates that ferroptosis plays a pivotal role in the pathophysiology of the aforementioned cognitive disorders. However, despite the rapid expansion of research into ferroptosis within the field of neuroscience, the existing body of knowledge remains relatively fragmented and unsystematized (9). Therefore, the present review aims to systematically dissect the core molecular mechanisms of ferroptosis, elucidate its specific pathogenic roles in cognitive disorders including AD, PD and DACI as well as provide a comprehensive overview of emerging diagnostic biomarkers and therapeutic strategies targeting this cell death pathway. By synthesizing current research advancements, the present review aims to provide new perspectives on the common pathological pathways underlying these diseases and to establish a theoretical foundation to guide the development of effective clinical interventions.
Although several reviews have discussed the role of ferroptosis in neurodegenerative diseases, most have focused on a single disorder, isolated pathways or specific mechanistic aspects (7,10,11). By contrast, the novelty of the present review is its cross-disease perspective. The present review summarizes ferroptosis-related mechanisms across AD, PD and DACI, emphasizing both the common molecular features and the unique triggers in each disease. Moreover, the present review integrates recent progress in ferroptosis-related biomarkers and evaluates the translational potential of ferroptosis-targeted therapeutic strategies. By discussing the practical challenges, clinical feasibility and workflow integration of these biomarkers, the present review aims to bridge the gap between basic mechanistic research and clinical application.
Ferroptosis is a complex biological process regulated by the interplay of multiple metabolic pathways (Fig. 1); its hallmark feature is the uncontrolled, iron-dependent accumulation of lipid peroxides on cellular membranes (12), ultimately leading to the destruction of membrane integrity and cell death. Understanding the interplay among lipid metabolism, iron metabolism and the antioxidant defense system is fundamental to elucidating the role of ferroptosis in disease.
Lipid peroxidation constitutes the final step of ferroptosis execution, the occurrence of which depends on specific lipid substrates and catalytic enzymes. Owing to its high lipid content, particularly its enrichment in polyunsaturated fatty acids (PUFAs), the brain is especially vulnerable to ferroptosis (13).
PUFAs, especially arachidonic acid and adrenic acid, are the primary substrates for lipid peroxidation during ferroptosis. The bis-allylic hydrogen atoms within the molecular chains of these fatty acids are chemically labile and highly susceptible to abstraction, thereby initiating the chain reaction of lipid peroxidation (14). The content and composition of PUFAs within the cell membrane directly determine a cell's sensitivity to ferroptosis.
PUFAs cannot be directly peroxidized; they must first be integrated into membrane phospholipids. This process is accomplished through the synergistic actions of two key enzymes: ACSL4 and LPCAT3. ACSL4 is responsible for esterifying PUFAs into their corresponding acyl-CoA derivatives, after which LPCAT3 catalyzes the incorporation of these activated PUFAs into membrane phospholipids to form phospholipid-containing polyunsaturated fatty acids (PL-PUFAs) (14). Consequently, ACSL4 is considered a critical 'gatekeeper' enzyme for ferroptosis, as its expression level and activity cellular sensitivity to ferroptosis; its high expression enriches the cell membrane with oxidizable PUFAs, effectively 'priming' the cell for ferroptosis (15).
Once incorporated into the membrane, PL-PUFAs are oxidized to phospholipid hydroperoxides (PL-PUFA-OOH) through enzymatic or non-enzymatic pathways. The enzymatic pathway is primarily catalyzed by members of the lipoxygenase family (16). The non-enzymatic pathway is driven by the Fenton reaction, which is catalyzed by intracellular labile Fe2+ (17). The accumulation of these PL-PUFA-OOH molecules disrupts the integrity and fluidity of the cell membrane, ultimately leading to membrane rupture, the release of the cellular contents and cell death (18).
Although iron is an essential trace element for life, its dyshomeostasis is a key catalyst for ferroptosis. Cells possess an intricate system to regulate iron uptake, storage, utilization and efflux; disruptions at any point can lead to ferroptosis.
Extracellular Fe3+ typically binds to transferrin and enters cells via endocytosis through transferrin receptor 1 on the cell membrane. Within the acidic endosome, Fe3+ is reduced to Fe2+ by the six-transmembrane epithelial antigen of prostate 3 and is then released into the cytoplasm by divalent metal transporter 1, forming the highly redox-active LIP (19).
Fe2+ within the LIP is central to the catalysis of ferroptosis; it participates in the Fenton reaction, which decomposes H2O2, a byproduct of cellular metabolism, into highly reactive hydroxyl radicals. Hydroxyl radicals are among the most potent reactive oxygen species (ROS) and can non-specifically attack membrane PUFAs, thereby triggering intense lipid peroxidation (20).
Cells store excess iron within ferritin, a spherical protein shell composed of 24 subunits, to mitigate the toxicity of free iron in the LIP (21,22). However, upon exposure to specific signaling cues, cells can degrade ferritin through a selective autophagic process known as ferritinophagy (23). This process is mediated by a specific cargo receptor, nuclear receptor coactivator 4, which recognizes and binds to ferritin, targeting it for lysosomal degradation (24). This process releases large amounts of stored iron into the LIP (23). Ferritinophagy is a key mechanism for rapidly increasing the iron content of the LIP when needed and is important for increasing cellular sensitivity to ferroptosis (25).
Cells have evolved a complex, multilayered antioxidant defense network to counteract the onset of ferroptosis. This network maintains cellular redox homeostasis by eliminating lipid peroxides or inhibiting their formation. These defense mechanisms can be broadly categorized into the canonical glutathione peroxidase 4 (GPX4)-dependent pathway and non-canonical, independent pathways.
The canonical axis defending against ferroptosis is centered on the GPX4/GSH system (26). This pathway begins with the cell membrane-bound cystine/glutamate antiporter, System Xc− (27). Composed of the light chain subunit solute carrier family 7 member 11 (SLC7A11) and the heavy chain subunit SLC3A2, System Xc− imports extracellular cystine, which is then rapidly reduced to cysteine intracellularly (28). Cysteine serves as the rate-limiting substrate for the synthesis of GSH (29). As the core effector molecule of this pathway, GPX4 is a unique selenoprotein and the only member of the GPX family capable of directly catalyzing the reduction of membrane phospholipid hydroperoxides (30). GPX4 utilizes GSH as a cofactor to efficiently reduce toxic lipid hydroperoxides into non-toxic phospholipid alcohols, thereby halting the lipid peroxidation chain reaction and effectively suppressing ferroptosis (31). Consequently, the inactivation of GPX4, either through the inhibition of System Xc− or the depletion of intracellular GSH, represents a classic experimental method for inducing ferroptosis.
In addition to the canonical GPX4/GSH axis, cells possess other parallel, GPX4-independent anti-ferroptotic pathways that provide additional layers of protection. One key non-canonical system is the ferroptosis suppressor protein 1 (FSP1)/coenzyme Q10 (CoQ10) pathway (32). Localized to the plasma membrane, FSP1 utilizes NADPH as an electron donor to reduce CoQ10 to its antioxidant form, ubiquinol. Ubiquinol acts as a lipophilic radical-trapping antioxidant that can effectively scavenge lipid peroxyl radicals, thus providing a critical alternative line of defense when GPX4 function is compromised. Another important independent anti-ferroptotic axis is the GTP cyclohydrolase-1/tetrahydrobiopterin (BH4) pathway (33). BH4 is not only a crucial enzyme cofactor but also a potent radical scavenger. Studies have shown that it exerts powerful anti-ferroptotic effects through two mechanisms: Protecting CoQ10 from depletion and directly inhibiting lipid peroxidation (34,35). In parallel to the plasma membrane-localized FSP1 system, a distinct, mitochondria-specific antioxidant axis that counteracts ferroptosis centered on the enzyme dihydroorotate dehydrogenase (DHODH) has been identified (36). DHODH is a flavoprotein of the electron transport chain that is anchored to the inner mitochondrial membrane, where it performs its canonical function in de novo pyrimidine synthesis (37). Crucially, this enzymatic process reduces CoQ10 to its active antioxidant form, ubiquinol (36). This dedicated mitochondrial pool of ubiquinol acts as a potent, radical-trapping antioxidant, directly neutralizing lipid peroxyl radicals within the inner mitochondrial membrane and thereby shielding the organelle from lipid peroxidation and subsequent ferroptotic collapse (38). Given that mitochondria are both a primary source of endogenous ROS and a central hub for iron metabolism, their membranes are uniquely vulnerable to ferroptotic insults. This defense system thus provides a critical layer of localized protection specifically within this susceptible organelle, offering new insights for understanding cellular resilience and identifying potential therapeutic targets in neurodegenerative disorders where mitochondrial dysfunction is a core pathological feature.
Nuclear factor erythroid 2-related factor 2 (NRF2) acts as the 'master regulator' of the cellular response to oxidative stress. Under oxidative or electrophilic stress, NRF2 is activated and translocates to the nucleus, where it binds to antioxidant response elements to initiate the transcription of a number of downstream genes. The proteins encoded by these genes have diverse functions, including promoting GSH synthesis, regenerating NADPH and upregulating iron storage proteins (ferritin), thereby constructing a robust, multilayered defense against ferroptosis (32).
The classic tumor suppressor p53 plays a complex, dual role in the regulation of ferroptosis. On the one hand, p53 can promote ferroptosis by inhibiting the expression of SLC7A11 (39). On the other hand, it can also suppress ferroptosis by regulating other metabolic pathways (such as activating CDKN1A/p21) (40). The ultimate effect of p53 is highly dependent on the cell type and the specific stress context (41).
The regulatory mechanisms of ferroptosis are not isolated but are intimately linked to the core metabolic activities of cells. The initiation of ferroptosis is deeply integrated with amino acid metabolism (glutamate/cystine balance), lipid metabolism (PUFA availability) and metal homeostasis (iron regulation). For instance, the activity of System Xc− is directly linked to the levels of the neurotransmitter glutamate and the synthesis of the antioxidant GSH (42). The execution phase depends on the biological properties of the cell membrane and the activity of lipid-modifying enzymes (ACSL4 and LPCAT3) (20). Moreover, iron is a critical cofactor for core bioenergetic processes such as mitochondrial respiration and oxygen transport and the Fe2+ required for ferroptosis (32). This intrinsic connectivity implies that ferroptosis is not merely a simple 'death switch' but rather a composite reflection of the overall metabolic state of the cell. Any pathological process that can disrupt these interconnected metabolic hubs, such as mitochondrial dysfunction in PD or oxidative stress in AD, may inadvertently lower the cellular threshold for resistance to ferroptosis. This explains why ferroptosis has emerged as a common pathological pathway in multiple, seemingly unrelated diseases; it represents a shared downstream execution point where diverse upstream damage signals converge.
The fundamental molecular mechanisms of ferroptosis are specifically activated and regulated in different disease contexts and are emerging as key factors driving the pathological progression of cognitive impairment (Table I). This section describes the specific mechanisms of ferroptosis in AD, PD and DACI.
The regulation of ferroptosis can impact AD through multiple mechanisms, including the regulation of GSH levels, the Fenton reaction, amyloid-β (Aβ), lipid metabolism, iron metabolism and the NRF2 signaling pathway (Fig. 2).
An abnormal increase in iron levels in the AD brain is a well-documented phenomenon, especially in the regions most severely affected by neurodegeneration (6). Iron dyshomeostasis is not only an epiphenomenon but is also strongly implicated in the core pathological processes of AD. On the one hand, excess iron ions can directly catalyze ROS production, exacerbating oxidative stress, while also promoting the generation and aggregation of Aβ and catalyzing the hyperphosphorylation of the tau protein. On the other hand, Aβ and tau aggregates can disrupt iron homeostasis, for instance, by interfering with the function of iron-efflux proteins, thereby further intensifying iron accumulation. This positive feedback loop, or 'vicious cycle', between iron and Aβ/tau significantly lowers the threshold for neuronal resistance to ferroptosis, rendering them exceptionally vulnerable (43).
The AD brain is in a state of chronic, sustained oxidative stress. Aβ oligomers and fibrils are themselves sources of ROS and can directly induce lipid peroxidation. Critically, a study has shown that the levels of GSH, a key antioxidant, are significantly reduced in the AD brain and that the extent of this depletion correlates positively with the severity of cognitive impairment (43). The depletion of GSH leads directly to the failure of the GPX4 antioxidant system, resulting in cells being unable to effectively neutralize lipid peroxides and thereby allowing the onset of ferroptosis.
In recent years, a new paradigm in AD pathology has emerged: The ferroptosis of microglia, the resident immune cells of the brain. In the pathological environment of AD, microglia are activated to clear Aβ plaques, damaged myelin, and cellular debris. However, this debris (particularly myelin) is rich in iron. During the phagocytosis and degradation of this iron-rich material, the microglia themselves can undergo ferroptosis due to iron overload (44). This process not only results in the loss of protective immune cells but also the release of proinflammatory cytokines and damage-associated molecular patterns during their death, further exacerbating the neuroinflammatory response (45). This process creates a self-amplifying cycle of damage that promotes neurodegeneration (46).
Genetic studies have provided direct evidence for the involvement of ferroptosis in AD (47,48). Transcriptomic analyses of brain tissue samples from patients with AD have revealed significant alterations in the expression of a range of ferroptosis-related genes (FRGs). For example, the genes encoding transferrin receptor (TFRC), glucose transporters and the master antioxidant regulator NRF2 are closely associated with the pathological progression of AD (49).
Ferroptosis is regulated by multiple interconnected pathways, including the GPX4/GSH system, iron and lipid metabolism, the NRF2 signaling pathway and mitochondrial function, in PD (Fig. 3).
The integrity of the GPX4/GSH antioxidant axis is critical for dopaminergic neuron survival. The oxidation product of dopamine can induce ferroptosis by covalently modifying GPX4 and mediating its ubiquitination and degradation (50). Conversely, experimental GPX4 overexpression exerts neuroprotective effects as it reduces neuronal death (50). This pathway is also targeted by other PD-related factors; the leucine-rich repeat kinase 2 (LRRK2) protein induces ferroptosis by acting on the System Xc−/GSH/GPX4 pathway (51) and the m6A demethylase fat mass and obesity-associated protein can promote ferroptosis via the p53/SLC7A11 axis (39).
Given the central role of iron, therapeutic strategies have focused on its chelation. The drug deferiprone has been shown to inhibit ferroptosis by chelating excess iron within the substantia nigra in vivo (52). This result is compounded by the fact that α-synuclein aggregation, a hallmark of PD, can also induce ferroptosis through mechanisms such as membrane disruption and altered calcium influx (53).
The regulation of lipid metabolism, particularly through the enzyme ACSL4, is a key control point. The PD-relevant neurotoxin salsolinol induces ferroptosis by upregulating ACSL4 expression (54). By contrast, the natural compound acteoside exerts a protective effect by downregulating ACSL4 (54). Downstream lipid peroxidation can be directly inhibited by radical-trapping antioxidants such as ferrostatin-1 (55).
The NRF2 antioxidant response pathway is a major defensive system whose impairment contributes to PD. Pathological α-synuclein upregulation induces ferroptosis by reducing NRF2 protein levels and impairing antioxidant defenses (56). The therapeutic activation of this pathway with compounds such as dimethyl fumarate, tert-butylhydroquinone and sulforaphane can inhibit ferroptosis by upregulating the expression of downstream antioxidant genes (57). Additionally, the activation of melatonin MT1 receptors can suppress ferroptosis through the sirtuin 1/NRF2/heme oxygenase 1 (HO-1)/GPX4 signaling cascade (58).
Mitochondrial dysfunction acts as a core driver of ferroptosis through a combination of iron homeostasis imbalance and oxidative stress (59). This oxidative environment is further exacerbated by neuroinflammatory processes. Microglial complement receptor 3 can induce ferroptosis via NADPH oxidase 2 (NOX2) (60), and other NADPH oxidases, including NOX1 (61) and NOX4 (62), are also key contributors to the ferroptotic process.
The regulation of ferroptosis can affect DACI through multiple mechanisms, including the modulation of the GPX4/GSH system, iron and ROS metabolism, mitochondrial homeostasis and the NRF2 and PKR-like endoplasmic reticulum kinase (PERK) signaling pathways (Fig. 4).
The metabolic state in DACI significantly affects the activity of the GPX4 antioxidant axis and the sensitivity of cells to ferroptosis. The AMP-activated protein kinase (AMPK) pathway, a key sensor of cellular energy, plays a protective role. AMPK agonists inhibit ferroptosis by activating AMPK, which subsequently upregulates GPX4 expression and downregulates lipocalin-2 expression in vivo (63). Additionally, berberine has been shown to protect against high glucose-induced damage to glomerular podocytes by activating the NRF2/HO-1/GPX4 pathway, thereby inhibiting ferroptosis (64).
The diabetic state creates a pro-ferroptotic environment characterized by iron dyshomeostasis (65). Extensive iron deposition is observed in the hippocampus of diabetic models and elevated plasma Fe2+ levels correlate with cognitive decline in patients (66,67). The increased iron levels can catalyze the Fenton reaction, leading to lipid peroxidation. Therapeutic interventions aim to mitigate this iron-induced stress. Erythropoietin has been shown to inhibit ferroptosis by reducing iron overload and lipid peroxidation both in vitro and in vivo (68). Similarly, the antioxidant liproxstatin-1 can effectively inhibit ferroptosis by attenuating iron accumulation and oxidative stress in vivo (63).
Triggering receptor expressed on myeloid cells 1 (TREM1) is a key driver of microglial-mediated ferroptosis in individuals with DACI (69). Under high-glucose conditions, the expression of TREM1 is upregulated, activating the PERK pathway (69). This process leads to the inactivation of antioxidant systems and increased lipid peroxidation and ROS levels, ultimately inducing ferroptosis in microglia and exacerbating neuroinflammation.
Mitochondrial dysfunction is a core feature of DACI (70), contributing to both the ROS production and iron dysregulation that drive ferroptosis. The scaffolding protein Caveolin-1 is crucial for maintaining mitochondrial integrity (71). In diabetic models, Caveolin-1 expression is significantly reduced. Restoring its function via Caveolin-1 overexpression has been shown to inhibit ferroptosis by improving mitochondrial homeostasis, thereby mitigating cognitive decline both in vitro and in vivo (67).
The transcription factor NRF2 is a master regulator of the antioxidant response and a key target for inhibiting ferroptosis in individuals with DACI. Several compounds have been shown to confer protection by modulating this pathway. Quercetin inhibits ferroptosis by activating the NRF2/HO-1 pathway in models of diabetic nephropathy, a mechanism that is potentially relevant to DACI (72). Berberine also leverages this pathway, activating the NRF2/HO-1/GPX4 axis to suppress ferroptosis (64). Furthermore, vitamin D can inhibit ferroptosis by activating the vitamin D receptor, which in turn upregulates the NRF2/HO-1 signaling cascade, as shown in in vitro and in vivo models (73).
Although the initial etiologies of AD, PD and DACI are distinct, they all ultimately converge on the common cell death pathway of ferroptosis. These diseases share several core pathological features: Oxidative stress, neuroinflammation and mitochondrial dysfunction (74). Oxidative stress directly generates ROS to initiate lipid peroxidation (32); neuroinflammation, particularly the activation of microglia, alters iron metabolism and promotes ROS release (44); and mitochondrial dysfunction is a major source of both ROS and the LIP (75). The specific upstream triggers of each disease (such as Aβ, ischemia and hemorrhage) activate these common downstream pathological processes, which in turn are the direct upstream activators of the core ferroptosis machinery (iron overload, lipid peroxidation and antioxidant system depletion). Therefore, ferroptosis is not merely another form of cell death, it acts as a 'central executioner', a common endpoint where diverse neural injury signals converge. This recognition highlights the potential of ferroptosis as a therapeutic target. Theoretically, an effective antiferroptotic agent could possess broad value as a treatment for multiple mechanistically distinct cognitive disorders.
Given the central role of ferroptosis in the pathological processes of various cognitive disorders, its associated molecules and metabolites hold immense potential as novel diagnostic and prognostic biomarkers. At present, diagnostic tools for neurodegenerative diseases primarily rely on the assessment of late-stage clinical symptoms or costly neuroimaging examinations and lack pathology-based biomarkers that would enable intervention in the early stages of the disease (76). The development of biomarkers that can reflect the state of ferroptosis in the brain is crucial for enabling early diagnosis, assessing disease progression and monitoring therapeutic responses (Table II).
Cerebrospinal fluid (CSF) and blood represent two vital biofluids for accessing pathological information regarding the central nervous system. Detecting changes in ferroptosis-related molecules within these biofluids holds promise for the development of minimally invasive diagnostic methods.
Measuring the levels of iron homeostasis-related proteins in CSF or serum is a direct strategy. For example, multiple studies have shown that the levels of ferritin in the CSF of patients with AD are elevated and this increase is associated with faster rates of cognitive decline and brain atrophy, suggesting that CSF ferritin levels could be a valuable prognostic biomarker (77,78). Urbano et al (79) demonstrated that CSF ferritin levels are associated with the conversion from mild cognitive impairment to dementia. Furthermore, CSF iron levels correlate positively with tau protein levels (80). In the BioFINDER cohort study, CSF ferritin levels reaching a threshold value of 12.47 ng/ml predicted cognitive deterioration in patients with mild cognitive impairment (81). Moreover, Apolipoprotein E4 carrier status strengthens the diagnostic association between brain iron content and AD pathology (81). A study also found that patients with PD with substantia nigra hyperechogenicity exhibit significantly lower cognitive function than those without hyperechogenicity; among patients with PD and poorer cognitive function, CSF iron levels are significantly elevated while transferrin levels are significantly reduced (82). Additionally, the levels of transferrin and non-transferrin-bound iron may also reflect the status of iron load in the brain (77).
Lipid peroxidation is the key step in the execution of ferroptosis and its metabolites are direct indicators of ferroptotic activity. Malondialdehyde and 4-hydroxynonenal are two major end-products of lipid peroxidation and their elevated levels in CSF or blood have been shown to be correlated with the pathological severity of AD and PD (33,83).
The state of the antioxidant system reflects the capacity of the cell to resist ferroptosis. GSH is central to the GPX4 system and its reduced levels in CSF are closely associated with severe cognitive impairment in patients with AD, suggesting that GSH could serve as a marker for the failure of the brain's antioxidant capacity and the risk of ferroptosis (43).
Using high-throughput sequencing technologies, researchers can identify ferroptosis-related molecular markers at the genetic and transcriptomic levels.
Through bioinformatics methods, the expression profile of a set of FRGs can be analyzed in patient tissues or peripheral blood cells to construct prognostic models. For example, in AD research, an FRG signature composed of genes such as DNA damage inducible transcript 4, mucin 1 and CD44 was found to not only distinguish patients with AD from healthy controls but also to predict disease severity and immune cell infiltration in the brain (43). Such multigene signatures offer greater robustness and predictive power than single-gene markers.
Certain genes that play key roles in the ferroptosis pathway may also serve as independent prognostic indicators based on their expression levels. Measuring the expression levels of genes such as TFRC or SLC7A11 in brain tissue or blood from patients with neurodegenerative diseases could provide clues for assessing ferroptotic activity and disease progression (49).
Neuroimaging techniques, particularly magnetic resonance imaging (MRI), represent a non-invasive, in vivo method for assessing iron deposition in the brain and can serve as an indirect means of evaluating ferroptosis risk. Advanced MRI sequences, such as T2* relaxometry, are highly sensitive to iron deposition. Compared with elderly individuals negative for AD biomarkers, cognitively normal elderly individuals positive for AD biomarkers show significantly higher R2* levels in the caudate nucleus, putamen and globus pallidus, suggesting increased iron content (84). In PD research, this technique can be used to precisely quantify the iron content in the substantia nigra region and has been found to be closely correlated with disease severity and motor deficits (85). This method is useful not only for diagnosis but also for tracking disease progression and evaluating the efficacy of iron-targeted therapies.
Although numerous potential biomarkers have been identified, the vast majority are still in the preclinical or early clinical research stages (11). Transforming these research findings into stable, reliable and easily accessible clinical diagnostic tools remains a significant challenge.
Given the insidious and progressive nature of neurodegenerative diseases, the utility of a single biomarker measurement is questionable. Early in the disease course, ferroptotic processes may be subtle and localized, potentially yielding false-negative results. Longitudinal monitoring, therefore, represents a more clinically meaningful approach. Tracking the trajectory of changes in a panel of ferroptosis-related markers within accessible biofluids offers greater prognostic value than a single snapshot measurement. This dynamic monitoring is essential for identifying high-risk individuals, tracking disease progression and evaluating therapeutic responses.
Biomarker sources profoundly impact clinical feasibility. While analysis of postmortem brain tissue is the gold standard for research, it is not relevant for clinical diagnosis. A study on human postmortem brain tissue has revealed that the pathological progression of AD is accompanied by decreased expression of nuclear receptor coactivator 4 and GPX4 in gray matter (86). Meanwhile, research using human brain organoid models has demonstrated that ferroptosis inhibitors (Ferrostatin-1) can block Aβ pathology, reduce lipid peroxidation and restore iron storage function in AD organoids (86).
By contrast, CSF provides a more direct window into central nervous system pathology, yet the invasiveness of lumbar puncture limits its utility for routine screening or frequent monitoring. Consequently, developing biomarkers in easily accessible peripheral biofluids, such as blood, is a key priority. However, this approach faces several challenges, including the potential for the blood-brain barrier (BBB) to mask or dilute CNS-specific signals and the difficulty in distinguishing brain-derived markers from those originating systemically (87,88). In particular, the development of peripheral blood biomarkers that can accurately reflect the state of local ferroptosis within the brain is key and represents a major hurdle for future research. Future research must therefore focus on identifying markers capable of specifically crossing the BBB and on developing ultrasensitive technologies for reliable peripheral detection.
A major challenge in translating ferroptosis research from the laboratory to clinical practice is how to integrate ferroptosis biomarkers into current diagnostic pathways for cognitive impairment. A stepwise clinical workflow may provide a practical framework for this translation (Fig. 5).
At the first stage, ferroptosis-related tests could be added to primary care as part of low-cost, population-wide screening. For individuals at increased risk, such as those >65 years of age, with a family history of dementia or with comorbid type 2 diabetes mellitus (T2DM) (89-91), routine blood tests may be expanded to include serum ferritin and transferrin levels. These tests could be combined with simple cognitive assessments, such as the Montreal Cognitive Assessment or the Mini-Mental State Examination (92,93). Individuals who show both mild cognitive changes and abnormal iron metabolism could be flagged as high-risk and referred for specialist evaluation. This approach is affordable and easy to implement, but its specificity is low as ferritin and related markers are affected by inflammation and liver disease (94).
At the specialist evaluation stage, diagnosis could be refined in memory clinics or neurology departments using more specific ferroptosis biomarkers. Key measurements include lipid peroxidation markers such as malondialdehyde and 4-hydroxynonenal, antioxidant defense markers such as GPX4 activity and the GSH/oxidized GSH ratio and advanced MRI-based assessments such as quantitative susceptibility mapping to quantify brain iron deposition (95,96). These ferroptosis markers should be interpreted together with established diagnostic tools, including CSF Aβ42, phosphorylated tau, total tau and Aβ PET imaging (97). Signs of increased ferroptosis activity, when combined with these biomarkers, may improve diagnostic accuracy and help with differential diagnosis. The main barriers at this stage are the lack of standardized laboratory assays and the high cost and limited availability of advanced imaging.
The advanced research stage focuses on precision subtyping and clinical trial applications. Multi-omics approaches, such as metabolomics to identify lipid peroxidation profiles and genomics to characterize polymorphisms in ferroptosis-regulating genes, may allow more accurate pathological classification. Although these approaches are currently limited by high cost and complex data processing, they show strong potential for identifying patient groups that may benefit from ferroptosis-targeted treatments.
Advanced age is the most significant non-modifiable risk factor for major cognitive disorders, including AD, PD and DACI (98-100). A compelling body of evidence suggests that the aging process itself fosters a 'pro-ferroptotic' state in the brain, thereby lowering the threshold for neurodegeneration (101,102). This connection is rooted in several age-related physiological changes that directly impinge on the core regulatory axes of ferroptosis.
Physiological aging systematically increases neuronal susceptibility to ferroptosis through the synergistic action of four interconnected mechanisms. First, normal aging is associated with the progressive dysregulation of iron homeostasis, often leading to iron accumulation in vulnerable brain regions such as the hippocampus and substantia nigra (52). This expands the labile, catalytically active iron pool, providing ample 'fuel' to initiate destructive lipid peroxidation via the Fenton reaction. Second, the efficiency of endogenous antioxidant systems decreases with age, a core feature of which is the reduction in the levels of the master intracellular antioxidant, GSH (103,104). As GPX4 is entirely dependent on GSH to neutralize toxic lipid peroxides, the depletion of GSH directly weakens the cell's most critical line of defense against ferroptosis (105). Third, age-related mitochondrial dysfunction is another key contributor. Senescent mitochondria not only produce more ROS due to inefficient electron transport but also exhibit impaired iron-sulfur cluster biogenesis, leading to improper iron handling and accumulation within the mitochondria, thus creating a potent internal source of oxidative stress (106,107). Finally, although the total brain lipid content may decrease with age, the enzymes responsible for incorporating PUFAs into membrane phospholipids remain active, ensuring the continued presence of the necessary substrates for the final execution step of ferroptosis (75).
In summary, age-dependent changes such as increased catalytic iron levels, weakened antioxidant defenses, elevated mitochondrial-derived oxidative stress and sustained membrane vulnerability to peroxidation, collectively place neurons in the aging brain in a fragile state. This underlying vulnerability explains why the pathological triggers of various neurodegenerative diseases can converge on ferroptosis in the aging brain. Therefore, ferroptosis emerges as a core mechanism that intimately links age, the most universal risk factor, to cognitive decline, the shared pathological outcome.
Given the core pathogenic role of ferroptosis in various cognitive disorders, targeting this pathway has emerged as a highly attractive and novel therapeutic strategy. To this end, researchers have developed or screened a range of drugs and compounds capable of inhibiting ferroptosis through various approaches and have validated their neuroprotective effects using various disease models (Table III).
Cognitive impairment in AD is closely associated with disrupted iron homeostasis, excessive oxidative stress and the accumulation of Aβ and tau proteins, which collectively promote ferroptosis. Several natural compounds have been found to be effective at modulating ferroptosis-related pathways to counteract these processes.
A common strategy involves activating the NRF2 signaling pathway, a master regulator of antioxidant responses. For example, ganoderic acid A activates the NRF2/SLC7A11/GPX4 axis (108), whereas saponins derived from Astragalus membranaceus act on the NOX4/NRF2 pathway to restore iron balance (109). Lignans from Schisandra chinensis activate the NRF2/FPN1 pathway, reducing phosphorylated tau levels and improving cognitive function (110). Similarly, avicularin enhances cognition by modulating NOX4/NRF2 signaling (111) and artemisinin derivatives increase NRF2 activity by targeting its inhibitor kelch-like ECH-associated protein 1 (Keap1) (112).
Some compounds work by directly supporting the GPX4/GSH antioxidant defense system. Thonningianin A, a newly identified ferroptosis inhibitor, activates GPX4 through the AMPK/NRF2 pathway (113). Total lignans from Schisandra increase the activity of the NADK/NADPH/GSH system to reduce Aβ deposition (114) and schisandrin B downregulates glycogen synthase kinase 3β (GSK3β) expression to promote NRF2/GPX4 signaling (115). Clinical studies suggest that selenium may help slow cognitive decline (116,117). Reduced selenium levels in the brains of patients with AD are associated with disease progression. In addition, selenium-loaded nanospheres can deliver selenium, an essential cofactor for GPX4, and markedly improve cognition in AD models (118).
Other strategies target molecules other than NRF2 and GPX4. Berberine reduces oxidative damage by inhibiting the JNK/p38 MAPK pathway (119). Neuritin protects against cognitive impairment by activating PI3K/Akt (120), whereas ghrelin mitigates neuroinflammation by acting on the bone morphogenetic protein 6/SMAD1 pathway (121). In addition, tetrahedral framework nucleic acids have been developed to directly neutralize Aβ toxicity and restore synaptic integrity (122).
The degeneration of dopaminergic neurons in PD is fueled by iron overload, mitochondrial dysfunction and oxidative stress, all of which are tightly linked to ferroptosis. Therapeutic strategies have therefore focused on disrupting these pathological cycles.
Iron chelation remains a straightforward intervention. Deferoxamine (DFO) has been shown to preserve dopaminergic neurons in animal models (123). However, the clinical outcomes associated with deferiprone, an oral iron chelator, have been inconsistent. For instance, one trial reported worsening motor symptoms in patients with early-stage PD, highlighting the complexity of iron-targeted therapy (52,60,124).
Several natural compounds activate endogenous antioxidant pathways to increase the resilience of the brain to oxidative stress. Acteoside inhibits ACSL4 (54) while simultaneously activating NRF2 (125). Granulathiazole A reduces α-synuclein accumulation through NRF2/HO-1 activation (126) and betulinic acid improves behavioral outcomes by reducing ROS levels (127). Dingzhen pills, a traditional herbal formulation, inhibit the cyclic GMP-AMP synthase/stimulator of interferon genes pathway to protect dopaminergic neurons (128).
Efforts have also been directed at modulating key proteins in the ferroptosis cascade. γ-aminobutyric acid from Lactobacillus reuteri alleviates pathology by activating the Akt/GSK3β/GPX4 axis (129), emodin upregulates the mitochondrial protein ubiquitin-cytochrome C reductase core protein 1 to suppress ferroptosis (130) and corilagin protects neurons by regulating the Toll-like receptor 4/Src/NOX2 pathway (131). Additionally, the inhibition of LRRK2 has been linked to reduced ferroptotic damage via the p62/Keap1/NRF2 axis (132) and teneligliptin, a dipeptidyl peptidase-4 inhibitor, decreases ROS levels and prevents ferroptosis (133).
Therapeutic interventions for DACI often target the metabolic dysregulation that creates a pro-ferroptotic environment. Several repurposed diabetes drugs and natural compounds have been shown to exert neuroprotective effects by inhibiting ferroptosis. The GLP-1 receptor agonist liraglutide improves cognition by repairing mitochondria and modulating the GPX4/SLC7A11 pathway (134).
Activation of the NRF2 antioxidant pathway is a common strategy. The natural product sinomenine improves cognition by regulating the EGF/NRF2/HO-1 pathway (135). Artemisinin has also been shown to ameliorate cognitive impairment in T2DM mice by activating NRF2 (136). Other compounds work by inhibiting inflammatory signaling; dihydromyricetin alleviates hippocampal ferroptosis by inhibiting the JNK pathway (137).
Other approaches aim to correct the iron and ROS imbalance directly. Erythropoietin has been shown to improve cognition by reducing iron overload and lipid peroxidation in diabetic models (68). Resveratrol inhibits the ability of microRNA-9-3p to upregulate SLC7A11 expression, thereby improving cognition in the context of DACI (138).
Ferroptosis, a distinct form of iron-dependent cell death driven by lipid peroxidation, has emerged as a key pathological mechanism underlying multiple cognitive disorders, including AD, PD and DACI. Preclinical studies have consistently shown that targeting ferroptosis with drugs such as iron chelators, lipid peroxidation inhibitors or NRF2 activators can produce significant neuroprotective effects. However, translating these promising findings into clinical success remains a formidable challenge.
One of the major pharmacological barriers is the BBB, which restricts the entry of most therapeutic agents into the central nervous system. A number of compounds with robust in vitro efficacy, such as DFO and certain natural products, fail to exert therapeutic effects in vivo due to their poor BBB permeability (139). Thus, improving drug delivery across the BBB (through molecular optimization or nanotechnology-based delivery systems) is a critical priority.
Target specificity and off-target effects represent other challenges. Iron homeostasis plays dual roles in maintaining normal brain function and contributes to pathology. Broad-spectrum iron chelation, as highlighted by the 'Deferiprone Paradox', risks impairing physiological processes while attempting to block ferroptosis. Similarly, electrophilic NRF2 activators may non-specifically react with other thiol-containing proteins, causing cytotoxicity (140). Future therapeutics must achieve high specificity and ideally be selectively activated under pathological conditions.
A number of currently available ferroptosis modulators also face limitations in drug-like properties, such as poor water solubility, instability and suboptimal pharmacokinetics, which hinder their development into clinically viable agents (141). Improving these characteristics through medicinal chemistry is essential.
Compounding these pharmacological issues is the lack of reliable biomarkers for monitoring ferroptosis in the human brain. In the absence of validated protein, metabolite or genetic markers in CSF or blood, identifying patients most likely to benefit from ferroptosis-targeting therapies, assessing the treatment response or dose adjustment are difficult (9). Most proposed biomarkers are still in the exploratory stage and require rigorous clinical validation.
Several innovative strategies should be prioritized to overcome these obstacles. Nanodelivery systems can facilitate BBB penetration, improve drug solubility and stability, and enable targeted delivery to pathological brain regions (142,143). Combination therapies that integrate ferroptosis inhibitors with other modalities, such as dopamine replacement therapy for PD, anti-Aβ or anti-tau agents for AD and anti-inflammatory drugs for chronic neuroinflammation, may yield synergistic benefits. Personalized medicine approaches that incorporate individual disease stages, genetic profiles and biomarker status are critical for optimizing therapeutic outcomes. Furthermore, mechanistic research is essential to elucidate the unresolved aspects of ferroptosis, including the final molecular executors and the crosstalk between ferroptosis and other cell death pathways, such as apoptosis and autophagy. These insights will inform the discovery of more precise and effective drug targets. At present, the field of ferroptosis is at a critical inflection point, shifting from mechanistic discovery to translational application. While preclinical data highlight its therapeutic potential (74), clinical progress has been slow due to unresolved delivery and diagnostic issues (144). Animal models are indispensable for mechanistic exploration but cannot fully replicate the long-term, multifactorial progression of human neurodegenerative diseases.
Therefore, the future of ferroptosis-targeted therapy depends not only on discovering new inhibitors or inducers but also on solving the core issues of how to deliver the right drug to the right brain region in the right patient and how to reliably monitor ferroptosis in clinical settings. Interdisciplinary collaboration among researchers from the neuroscience, pharmacology, medicinal chemistry and biomarker fields is essential. Without such concerted efforts, the enormous therapeutic promise of ferroptosis modulation may remain confined to the preclinical realm.
Not applicable.
This work was funded by the National Natural Science Foundation of China (grant no. 82271967), the Nanjing Medical Science and Technology Development Fund (grant no. ZKX22038) and the China Postdoctoral Science Foundation (grant no. 2024M751466).
Not applicable.
JM and WX provided supervision and assisted with writing and editing. SG conducted the literature search and drafted and compiled the manuscript and created the figures. DK, SF, YL, XY and ZJ provided constructive suggestions for the article and contributed to its design and structuring. JM and WX modified and adjusted the figures, ensuring their accuracy and clarity. All authors have read and approved the final version of the manuscript. Data authentication is not applicable.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
ACSL4 |
acyl-CoA synthetase long-chain family member 4 |
|
AD |
Alzheimer's disease |
|
CSF |
cerebrospinal fluid |
|
DACI |
diabetes-associated cognitive impairment |
|
GPX4 |
glutathione peroxidase 4 |
|
GSH |
glutathione |
|
GSK3β |
glycogen synthase kinase 3β |
|
LIP |
labile iron pool |
|
NRF2 |
nuclear factor erythroid 2-related factor 2 |
|
PD |
Parkinson's disease |
|
PUFAs |
polyunsaturated fatty acids |
|
ROS |
reactive oxygen species |
|
SLC7A11 |
solute carrier family 7 member 11 |
|
T2DM |
type 2 diabetes mellitus |
|
TREM1 |
triggering receptor expressed on myeloid cells 1 |
|
Chen X, Comish PB, Tang D and Kang R: Characteristics and biomarkers of ferroptosis. Front Cell Dev Biol. 9:6371622021. View Article : Google Scholar : PubMed/NCBI | |
|
Wang H and Xie Y: Advances in ferroptosis research: A comprehensive review of mechanism exploration, drug development, and disease treatment. Pharmaceuticals (Basel). 18:3342025. View Article : Google Scholar : PubMed/NCBI | |
|
Ma W, Jiang X, Jia R and Li Y: Mechanisms of ferroptosis and targeted therapeutic approaches in urological malignancies. Cell Death Discov. 10:4322024. View Article : Google Scholar : PubMed/NCBI | |
|
Yan HF, Zou T, Tuo QZ, Xu S, Li H, Belaidi AA and Lei P: Ferroptosis: Mechanisms and links with diseases. Signal Transduct Target Ther. 6:492021. View Article : Google Scholar : PubMed/NCBI | |
|
Zheng J and Conrad M: Ferroptosis: When metabolism meets cell death. Physiol Rev. 105:651–706. 2025. View Article : Google Scholar | |
|
Huang X: A concise review on oxidative stress-mediated ferroptosis and cuproptosis in Alzheimer's disease. Cells. 12:13692023. View Article : Google Scholar : PubMed/NCBI | |
|
Naderi S, Khodagholi F, Janahmadi M, Motamedi F, Torabi A, Batool Z, Heydarabadi MF and Pourbadie HG: Ferroptosis and cognitive impairment: Unraveling the link and potential therapeutic targets. Neuropharmacology. 263:1102102025. View Article : Google Scholar | |
|
Zhang Y, Wang Y, Li Y, Pang J, Höhn A, Dong W, Gao R, Liu Y, Wang D, She Y, et al: Methionine restriction alleviates Diabetes-associated cognitive impairment via activation of FGF21. Redox Biol. 77:1033902024. View Article : Google Scholar : PubMed/NCBI | |
|
Ryan SK, Ugalde CL, Rolland AS, Skidmore J, Devos D and Hammond TR: Therapeutic inhibition of ferroptosis in neurodegenerative disease. Trends Pharmacol Sci. 44:674–688. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Khan G, Hussain MS, Khan Y, Fatima R, Ahmad S, Sultana A and Alam P: Ferroptosis and its contribution to cognitive impairment in Alzheimer's disease: Mechanisms and therapeutic potential. Brain Res. 1864:1497762025. View Article : Google Scholar : PubMed/NCBI | |
|
Ma M, Jing G, Tian Y, Yin R and Zhang M: Ferroptosis in cognitive impairment associated with diabetes and Alzheimer's disease: Mechanistic insights and new therapeutic opportunities. Mol Neurobiol. 62:2435–2449. 2025. View Article : Google Scholar | |
|
Jacquemyn J, Ralhan I and Ioannou MS: Driving factors of neuronal ferroptosis. Trends Cell Biol. 34:535–546. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Yan N and Zhang JJ: The emerging roles of ferroptosis in vascular cognitive impairment. Front Neurosci. 13:8112019. View Article : Google Scholar : PubMed/NCBI | |
|
Li Y, Li Z, Ran Q and Wang P: Sterols in ferroptosis: From molecular mechanisms to therapeutic strategies. Trends Mol Med. 31:36–49. 2025. View Article : Google Scholar | |
|
Li QS and Jia YJ: Ferroptosis: A critical player and potential therapeutic target in traumatic brain injury and spinal cord injury. Neural Regen Res. 18:506–512. 2023. View Article : Google Scholar | |
|
Shui S, Zhao Z, Wang H, Conrad M and Liu G: Non-enzymatic lipid peroxidation initiated by photodynamic therapy drives a distinct ferroptosis-like cell death pathway. Redox Biol. 45:1020562021. View Article : Google Scholar : PubMed/NCBI | |
|
Chang S, Zhang M, Liu C, Li M, Lou Y and Tan H: Redox mechanism of glycerophospholipids and relevant targeted therapy in ferroptosis. Cell Death Discov. 11:3582025. View Article : Google Scholar : PubMed/NCBI | |
|
Fei W, Chen D, Tang H, Li C, Zheng W, Chen F, Song Q, Zhao Y, Zou Y and Zheng C: Targeted GSH-exhausting and hydroxyl radical self-producing manganese-silica nanomissiles for MRI guided ferroptotic cancer therapy. Nanoscale. 12:16738–16754. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Lee J and Hyun DH: The interplay between intracellular iron homeostasis and neuroinflammation in neurodegenerative diseases. Antioxidants (Basel). 12:9182023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang XD, Liu ZY, Wang MS, Guo YX, Wang XK, Luo K, Huang S and Li RF: Mechanisms and regulations of ferroptosis. Front Immunol. 14:12694512023. View Article : Google Scholar : | |
|
Kakhlon O and Cabantchik ZI: The labile iron pool: Characterization, measurement, and participation in cellular processes(1). Free Radic Biol Med. 33:1037–1046. 2002. View Article : Google Scholar : PubMed/NCBI | |
|
Arosio P, Elia L and Poli M: Ferritin, cellular iron storage and regulation. IUBMB Life. 69:414–422. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Mancias JD, Wang X, Gygi SP, Harper JW and Kimmelman AC: Quantitative proteomics identifies NCOA4 as the cargo receptor mediating ferritinophagy. Nature. 509:105–109. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Mancias JD, Pontano Vaites L, Nissim S, Biancur DE, Kim AJ, Wang X, Liu Y, Goessling W, Kimmelman AC and Harper JW: Ferritinophagy via NCOA4 is required for erythropoiesis and is regulated by iron dependent HERC2-mediated proteolysis. Elife. 4:e103082015. View Article : Google Scholar : PubMed/NCBI | |
|
Ajoolabady A, Aslkhodapasandhokmabad H, Libby P, Tuomilehto J, Lip GYH, Penninger JM, Richardson DR, Tang D, Zhou H, Wang S, et al: Ferritinophagy and ferroptosis in the management of metabolic diseases. Trends Endocrinol Metab. 32:444–462. 2021. View Article : Google Scholar | |
|
Forcina GC and Dixon SJ: GPX4 at the crossroads of lipid homeostasis and ferroptosis. Proteomics. 19:e18003112019. View Article : Google Scholar : PubMed/NCBI | |
|
Bridges RJ, Natale NR and Patel SA: System xc− Cystine/glutamate antiporter: An update on molecular pharmacology and roles within the CNS. Br J Pharmacol. 165:20–34. 2012. View Article : Google Scholar : | |
|
Conrad M and Sato H: The oxidative stress-inducible cystine/glutamate antiporter, system x (c) (-): Cystine supplier and beyond. Amino Acids. 42:231–246. 2012. View Article : Google Scholar | |
|
Xue Z, Nuerrula Y, Sitiwaerdi Y and Eli M: Nuclear factor erythroid 2-related factor 2 promotes radioresistance by regulating glutamate-cysteine ligase modifier subunit and its unique immunoinvasive pattern. Biomol Biomed. 24:545–559. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Ursini F and Maiorino M: Lipid peroxidation and ferroptosis: The role of GSH and GPx4. Free Radic Biol Med. 152:175–185. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Yang WS, SriRamaratnam R, Welsch ME, Shimada K, Skouta R, Viswanathan VS, Cheah JH, Clemons PA, Shamji AF, Clish CB and Brown LM: Regulation of ferroptotic cancer cell death by GPX4. Cell. 156:317–331. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Benarroch E: What is the role of ferroptosis in neurodegeneration? Neurology. 101:312–319. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou M, Xu K, Ge J, Luo X, Wu M, Wang N and Zeng J: Targeting ferroptosis in Parkinson's disease: Mechanisms and emerging therapeutic strategies. Int J Mol Sci. 25:130422024. View Article : Google Scholar : PubMed/NCBI | |
|
Kraft VAN, Bezjian CT, Pfeiffer S, Ringelstetter L, Müller C, Zandkarimi F, Merl-Pham J, Bao X, Anastasov N, Kössl J, et al: GTP cyclohydrolase 1/tetrahydrobiopterin counteract ferroptosis through lipid remodeling. ACS Cent Sci. 6:41–53. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Soula M, Weber RA, Zilka O, Alwaseem H, La K, Yen F, Molina H, Garcia-Bermudez J, Pratt DA, Birsoy K, et al: Metabolic determinants of cancer cell sensitivity to canonical ferroptosis inducers. Nat Chem Biol. 16:1351–1360. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Mao C, Liu X, Zhang Y, Lei G, Yan Y, Lee H, Koppula P, Wu S, Zhuang L, Fang B, et al: DHODH-mediated ferroptosis defence is a targetable vulnerability in cancer. Nature. 593:586–590. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Teng D, Swanson KD, Wang R, Zhuang A, Wu H, Niu Z, Cai L, Avritt FR, Gu L, Asara JM, et al: DHODH modulates immune evasion of cancer cells via CDP-choline dependent regulation of phospholipid metabolism and ferroptosis. Nat Commun. 16:38672025. View Article : Google Scholar : PubMed/NCBI | |
|
Cao J, Chen X, Chen L, Lu Y, Wu Y, Deng A, Pan F, Huang H, Liu Y, Li Y, et al: DHODH-mediated mitochondrial redox homeostasis: A novel ferroptosis regulator and promising therapeutic target. Redox Biol. 85:1037882025. View Article : Google Scholar : PubMed/NCBI | |
|
Li Z, Chen X, Xiang W, Tang T and Gan L: m6A demethylase FTO-mediated upregulation of BAP1 induces neuronal ferroptosis via the p53/SLC7A11 axis in the MPP+/MPTP-induced Parkinson's disease model. ACS Chem Neurosci. 16:405–416. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Tarangelo A, Magtanong L, Bieging-Rolett KT, Li Y, Ye J, Attardi LD and Dixon SJ: p53 suppresses metabolic stress-induced ferroptosis in cancer cells. Cell Rep. 22:569–575. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang M, Qiao M, Zhao C, Deng J, Li X and Zhou C: Targeting ferroptosis for cancer therapy: Exploring novel strategies from its mechanisms and role in cancers. Transl Lung Cancer Res. 9:1569–1584. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Lewerenz J, Hewett SJ, Huang Y, Lambros M, Gout PW, Kalivas PW, Massie A, Smolders I, Methner A, Pergande M, et al: The cystine/glutamate antiporter system x(c)(-) in health and disease: From molecular mechanisms to novel therapeutic opportunities. Antioxid Redox Signal. 18:522–555. 2013. View Article : Google Scholar : | |
|
Sun Y, Xiao Y, Tang Q, Chen W and Lin L: Genetic markers associated with ferroptosis in Alzheimer's disease. Front Aging Neurosci. 16:13646052024. View Article : Google Scholar : PubMed/NCBI | |
|
Streit WJ, Phan L and Bechmann I: Ferroptosis and pathogenesis of neuritic plaques in Alzheimer disease. Pharmacol Rev. 77:1000052025. View Article : Google Scholar : PubMed/NCBI | |
|
Proneth B and Conrad M: Ferroptosis and necroinflammation, a yet poorly explored link. Cell Death Differ. 26:14–24. 2019. View Article : Google Scholar | |
|
McIntosh A, Mela V, Harty C, Minogue AM, Costello DA, Kerskens C and Lynch MA: Iron accumulation in microglia triggers a cascade of events that leads to altered metabolism and compromised function in APP/PS1 mice. Brain Pathol. 29:606–621. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao H, Wang J, Li Z, Wang S, Yu G and Wang L: Identification Ferroptosis-related hub genes and diagnostic model in Alzheimer's disease. Front Mol Neurosci. 16:12806392023. View Article : Google Scholar : PubMed/NCBI | |
|
Tian M, Shen J, Qi Z, Feng Y and Fang P: Bioinformatics analysis and prediction of Alzheimer's disease and alcohol dependence based on Ferroptosis-related genes. Front Aging Neurosci. 15:12011422023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Y, Wang M and Chang W: Iron dyshomeostasis and ferroptosis in Alzheimer's disease: Molecular mechanisms of cell death and novel therapeutic drugs and targets for AD. Front Pharmacol. 13:9836232022. View Article : Google Scholar : PubMed/NCBI | |
|
Sun J, Lin XM, Lu DH, Wang M, Li K, Li SR, Li ZQ, Zhu CJ, Zhang ZM, Yan CY, et al: Midbrain dopamine oxidation links ubiquitination of glutathione peroxidase 4 to ferroptosis of dopaminergic neurons. J Clin Invest. 133:e1731102023. View Article : Google Scholar : PubMed/NCBI | |
|
Zheng Z, Zhang S, Liu X, Wang X, Xue C, Wu X, Zhang X, Xu X, Liu Z, Yao L and Lu G: LRRK2 regulates ferroptosis through the system xc-GSH-GPX4 pathway in the neuroinflammatory mechanism of Parkinson's disease. J Cell Physiol. 239:e312502024. View Article : Google Scholar : PubMed/NCBI | |
|
Negida A, Hassan NM, Aboeldahab H, Zain YE, Negida Y, Cadri S, Cadri N, Cloud LJ, Barrett MJ and Berman B: Efficacy of the Iron-chelating agent, deferiprone, in patients with Parkinson's disease: A systematic review and Meta-analysis. CNS Neurosci Ther. 30:e146072024. View Article : Google Scholar : PubMed/NCBI | |
|
Angelova PR, Choi ML, Berezhnov AV, Horrocks MH, Hughes CD, De S, Rodrigues M, Yapom R, Little D, Dolt KS, et al: Alpha synuclein aggregation drives ferroptosis: An Interplay of iron, calcium and lipid peroxidation. Cell Death Differ. 27:2781–2796. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Wang H, Wu S, Li Q, Sun H and Wang Y: Targeting ferroptosis: Acteoside as a neuroprotective agent in salsolinol-induced Parkinson's disease models. Front Biosci (Landmark Ed). 30:266792025. View Article : Google Scholar : PubMed/NCBI | |
|
Yan J, Bao L, Liang H, Zhao L, Liu M, Kong L, Fan X, Liang C, Liu T, Han X, et al: A druglike ferrostatin-1 analogue as a ferroptosis inhibitor and photoluminescent indicator. Angew Chem Int Ed Engl. 64:e2025021952025. View Article : Google Scholar : PubMed/NCBI | |
|
A A, W C, N N, L M, M D and Zhang DD: α-Syn overexpression, NRF2 suppression, and enhanced ferroptosis create a vicious cycle of neuronal loss in Parkinson's disease. Free Radic Biol Med. 192:130–140. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Gong K, Zhou S, Xiao L, Xu M, Zhou Y, Lu K, Yu X, Zhu J, Liu C and Zhu Q: Danggui shaoyao san ameliorates Alzheimer's disease by regulating lipid metabolism and inhibiting neuronal ferroptosis through the AMPK/Sp1/ACSL4 signaling pathway. Front Pharmacol. 16:15883752025. View Article : Google Scholar | |
|
Lv QK, Tao KX, Yao XY, Pang MZ, Cao BE, Liu CF and Wang F: Melatonin MT1 receptors regulate the Sirt1/Nrf2/ho-1/Gpx4 pathway to prevent α-synuclein-induced ferroptosis in Parkinson's disease. J Pineal Res. 76:e129482024. View Article : Google Scholar | |
|
Wang W, Thomas ER, Xiao R, Chen T, Guo Q, Liu K, Yang Y and Li X: Targeting mitochondria-regulated ferroptosis: A new frontier in Parkinson's disease therapy. Neuropharmacology. 274:1104392025. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Q, Liu J, Zhang Y, Li Z, Zhao Z, Jiang W, Zhao J, Hou L and Wang Q: Microglial CR3 promotes neuron ferroptosis via NOX2-mediated iron deposition in rotenone-induced experimental models of Parkinson's disease. Redox Biol. 77:1033692024. View Article : Google Scholar : PubMed/NCBI | |
|
Wang H, Mao W, Zhang Y, Feng W, Bai B, Ji B, Chen J, Cheng B and Yan F: NOX1 triggers ferroptosis and ferritinophagy, contributes to Parkinson's disease. Free Radic Biol Med. 222:331–343. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Lin Z, Ying C, Si X, Xue N, Liu Y, Zheng R, Chen Y, Pu J and Zhang B: NOX4 exacerbates Parkinson's disease pathology by promoting neuronal ferroptosis and neuroinflammation. Neural Regen Res. 20:2038–2052. 2025. View Article : Google Scholar | |
|
Xie Z, Wang X, Luo X, Yan J, Zhang J, Sun R, Luo A and Li S: Activated AMPK mitigates Diabetes-related cognitive dysfunction by inhibiting hippocampal ferroptosis. Biochem Pharmacol. 207:1153742023. View Article : Google Scholar | |
|
Wei M, Liu X, Tan Z, Tian X, Li M and Wei J: Ferroptosis: A new strategy for Chinese herbal medicine treatment of diabetic nephropathy. Front Endocrinol (Lausanne). 14:11880032023. View Article : Google Scholar : PubMed/NCBI | |
|
Sha W, Hu F, Xi Y, Chu Y and Bu S: Mechanism of ferroptosis and its role in type 2 diabetes mellitus. J Diabetes Res. 2021:99996122021. View Article : Google Scholar : PubMed/NCBI | |
|
Liu J, Hu X, Xue Y, Liu C, Liu D, Shang Y, Shi Y, Cheng L, Zhang J, Chen A and Wang J: Targeting hepcidin improves cognitive impairment and reduces iron deposition in a diabetic rat model. Am J Transl Res. 12:4830–4839. 2020.PubMed/NCBI | |
|
Tang W, Li Y, He S, Jiang T, Wang N, Du M, Cheng B, Gao W, Li Y and Wang Q: Caveolin-1 alleviates diabetes-associated cognitive dysfunction through modulating neuronal ferroptosis-mediated mitochondrial homeostasis. Antioxid Redox Signal. 37:867–886. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Guo T, Yu Y, Yan W, Zhang M, Yi X, Liu N, Cui X, Wei X, Sun Y, Wang Z, et al: Erythropoietin ameliorates cognitive dysfunction in mice with type 2 diabetes mellitus via inhibiting iron overload and ferroptosis. Exp Neurol. 365:1144142023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao Y, Guo H and Li Q, Wang N, Yan C, Zhang S, Dong Y, Liu C, Gao W, Zhu Y and Li Q: TREM1 induces microglial ferroptosis through the PERK pathway in diabetic-associated cognitive impairment. Exp Neurol. 383:1150312025. View Article : Google Scholar | |
|
Ma H, Jiang T, Tang W, Ma Z, Pu K, Xu F, Chang H, Zhao G, Gao W, Li Y and Wang Q: Transplantation of platelet-derived mitochondria alleviates cognitive impairment and mitochondrial dysfunction in db/db mice. Clin Sci (Lond). 134:2161–2175. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Asterholm IW, Mundy DI, Weng J, Anderson RGW and Scherer PE: Altered mitochondrial function and metabolic inflexibility associated with loss of caveolin-1. Cell Metab. 15:171–185. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Feng Q, Yang Y, Qiao Y, Zheng Y, Yu X, Liu F, Wang H, Zheng B, Pan S, Ren K, et al: Quercetin ameliorates diabetic kidney injury by inhibiting ferroptosis via activating Nrf2/HO-1 signaling pathway. Am J Chin Med. 51:997–1018. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Li J, Cao Y, Xu J, Li J, Lv C, Gao Q, Zhang C, Jin C, Wang R, Jiao R and Zhu H: Vitamin D improves cognitive impairment and alleviates ferroptosis via the Nrf2 signaling pathway in aging mice. Int J Mol Sci. 24:153152023. View Article : Google Scholar : PubMed/NCBI | |
|
Wei Z, Yu H, Zhao H, Wei M, Xing H, Pei J, Yang Y and Ren K: Broadening horizons: Ferroptosis as a new target for traumatic brain injury. Burns Trauma. 12:tkad0512024. View Article : Google Scholar : PubMed/NCBI | |
|
Abdukarimov N, Kokabi K and Kunz J: Ferroptosis and iron homeostasis: Molecular mechanisms and neurodegenerative disease implications. Antioxidants (Basel). 14:5272025. View Article : Google Scholar : PubMed/NCBI | |
|
Wang B, Fu C, Wei Y, Xu B, Yang R, Li C, Qiu M, Yin Y and Qin D: Ferroptosis-related biomarkers for alzheimer's disease: Identification by bioinformatic analysis in hippocampus. Front Cell Neurosci. 16:10239472022. View Article : Google Scholar : PubMed/NCBI | |
|
Ficiarà E, Munir Z, Boschi S, Caligiuri ME and Guiot C: Alteration of iron concentration in Alzheimer's disease as a possible diagnostic biomarker unveiling ferroptosis. Int J Mol Sci. 22:44792021. View Article : Google Scholar : PubMed/NCBI | |
|
Ayton S, Faux NG and Bush AI; Alzheimer's disease neuroimaging initiative: Ferritin levels in the cerebrospinal fluid predict Alzheimer's disease outcomes and are regulated by APOE. Nat Commun. 6:67602015. View Article : Google Scholar : PubMed/NCBI | |
|
Urbano T, Michalke B, Chiari A, Malagoli C, Bedin R, Tondelli M, Vinceti M and Filippini T: Iron species in cerebrospinal fluid and dementia risk in subjects with mild cognitive impairment: A cohort study. Neurotoxicology. 110:1–9. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Urbano T, Vinceti M, Carbone C, Wise LA, Malavolti M, Tondelli M, Bedin R, Vinceti G, Marti A, Chiari A, et al: Exposure to cadmium and other trace elements among individuals with mild cognitive impairment. Toxics. 12:9332024. View Article : Google Scholar : | |
|
Ayton S, Janelidze S, Kalinowski P, Palmqvist S, Belaidi AA, Stomrud E, Roberts A, Roberts B, Hansson O and Bush AI: CSF ferritin in the clinicopathological progression of Alzheimer's disease and associations with APOE and inflammation biomarkers. J Neurol Neurosurg Psychiatry. 94:211–219. 2023. View Article : Google Scholar | |
|
Yu SY, Cao CJ, Zuo LJ, Chen ZJ, Lian TH, Wang F, Hu Y, Piao YS, Li LX, Guo P, et al: Clinical features and dysfunctions of iron metabolism in Parkinson disease patients with hyper echogenicity in Substantia nigra: A Cross-sectional study. BMC Neurol. 18:92018. View Article : Google Scholar : PubMed/NCBI | |
|
Padurariu M, Ciobica A, Hritcu L, Stoica B, Bild W and Stefanescu C: Changes of some oxidative stress markers in the serum of patients with mild cognitive impairment and Alzheimer's disease. Neurosci Lett. 469:6–10. 2010. View Article : Google Scholar | |
|
Lin Q, Shahid S, Hone-Blanchet A, Huang S, Wu J, Bisht A, Loring D, Goldstein F, Levey A, Crosson B, et al: Magnetic resonance evidence of increased iron content in subcortical brain regions in asymptomatic Alzheimer's disease. Hum Brain Mapp. 44:3072–3083. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Dusek P, Hofer T, Alexander J, Roos PM and Aaseth JO: Cerebral iron deposition in neurodegeneration. Biomolecules. 12:7142022. View Article : Google Scholar : PubMed/NCBI | |
|
Majerníková N, Marmolejo-Garza A, Salinas CS, Luu MDA, Zhang Y, Trombetta-Lima M, Tomin T, Birner-Gruenberger R, Lehtonen Š, Koistinaho J, et al: The link between amyloid β and ferroptosis pathway in Alzheimer's disease progression. Cell Death Dis. 15:7822024. View Article : Google Scholar | |
|
Kang JH, Korecka M, Lee EB, Cousins KAQ, Tropea TF, Chen-Plotkin AA, Irwin DJ, Wolk D, Brylska M, Wan Y and Shaw LM: Alzheimer disease biomarkers: Moving from CSF to plasma for reliable detection of amyloid and tau pathology. Clin Chem. 69:1247–1259. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Hampel H, O'Bryant SE, Molinuevo JL, Zetterberg H, Masters CL, Lista S, Kiddle SJ, Batrla R and Blennow K: Blood-based biomarkers for Alzheimer disease: Mapping the road to the clinic. Nat Rev Neurol. 14:639–652. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Livingston G, Huntley J, Sommerlad A, Ames D, Ballard C, Banerjee S, Brayne C, Burns A, Cohen-Mansfield J, Cooper C, et al: Dementia prevention, intervention, and care: 2020 report of the lancet commission. Lancet. 396:413–446. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Biessels GJ and Despa F: Cognitive decline and dementia in diabetes mellitus: Mechanisms and clinical implications. Nat Rev Endocrinol. 14:591–604. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Andrews SJ, Tolosa-Tort P, Jonson C, Fulton-Howard B, Renton AE, Yokoyama JS and Yaffe K; Alzheimer's Disease Neuroimaging Initiative: The role of genomic-informed risk assessments in predicting dementia outcomes. Alzheimers Dement. 21:e708262025. View Article : Google Scholar : PubMed/NCBI | |
|
Nasreddine ZS, Phillips NA, Bédirian V, Charbonneau S, Whitehead V, Collin I, Cummings JL and Chertkow H: The Montreal cognitive assessment, MoCA: A brief screening tool for mild cognitive impairment. J Am Geriatr Soc. 53:695–699. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Folstein MF, Folstein SE and McHugh PR: 'Mini-mental state'. A practical method for grading the cognitive state of patients for the clinician J Psychiatr Res. 12:189–198. 1975. | |
|
Cullis JO, Fitzsimons EJ, Griffiths WJ, Tsochatzis E and Thomas DW; British Society for Haematology: Investigation and management of a raised serum ferritin. Br J Haematol. 181:331–340. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Stockwell BR, Friedmann Angeli JP, Bayir H, Bush AI, Conrad M, Dixon SJ, Fulda S, Gascón S, Hatzios SK, Kagan VE, et al: Ferroptosis: A regulated cell death nexus linking metabolism, redox biology, and disease. Cell. 171:273–285. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Y and Liu T: Quantitative susceptibility mapping (QSM): Decoding MRI data for a tissue magnetic biomarker. Magn Reson Med. 73:82–101. 2015. View Article : Google Scholar : | |
|
Jack CR, Bennett DA, Blennow K, Carrillo MC, Dunn B, Haeberlein SB, Holtzman DM, Jagust W, Jessen F, Karlawish J, et al: NIA-AA research framework: Toward a biological definition of Alzheimer's disease. Alzheimers Dement. 14:535–562. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Chen HJ, Dong X, Wang Y, Wang K, Feng G, Bai T, Zhang M, Gan K, Peng JJ, Huang W, et al: Polygenic risk for Alzheimer's disease in healthy aging: Age-related and APOE-driven effects on brain structures and cognition. Genome Med. 17:1262025. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Y, Chen M, Chen P, Lin X, Chen S, Liu C, Wang D, Deng H, Li Q and Wu Y: Machine learning-based stratification of mild cognitive impairment in Parkinson's disease: A multicenter cross-sectional analysis. BMC Med Inform Decis Mak. 25:3842025. View Article : Google Scholar : PubMed/NCBI | |
|
Ren Q, Niu S, Zhi L and Liu H: Risk factor analysis of cognitive frailty in older adults with type 2 diabetes mellitus: A cross-sectional study. Front Aging Neurosci. 17:16674052025. View Article : Google Scholar : PubMed/NCBI | |
|
Hambright WS, Fonseca RS, Chen L, Na R and Ran Q: Ablation of ferroptosis regulator glutathione peroxidase 4 in forebrain neurons promotes cognitive impairment and neurodegeneration. Redox Biol. 12:8–17. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Masaldan S, Bush AI, Devos D, Rolland AS and Moreau C: Striking while the iron is hot: Iron metabolism and ferroptosis in neurodegeneration. Free Radic Biol Med. 133:221–233. 2019. View Article : Google Scholar | |
|
Thorwald MA, Godoy-Lugo JA, Kerstiens E, Garcia G, Kim M, Shemtov SJ, Silva J, Durra S, O'Day PA, Mack WJ, et al: Down syndrome with Alzheimer's disease brains have increased iron and associated lipid peroxidation consistent with ferroptosis. Alzheimers Dement. 21:e703222025. View Article : Google Scholar : PubMed/NCBI | |
|
Ra F and K H: Oxidative stress in brain aging. Implications for therapeutics of neurodegenerative diseases. Neurobiol Aging. 23:795–807. 2002. View Article : Google Scholar | |
|
Ji Y, Zheng K, Li S, Ren C, Shen Y, Tian L, Zhu H, Zhou Z and Jiang Y: Insight into the potential role of ferroptosis in neurodegenerative diseases. Front Cell Neurosci. 16:10051822022. View Article : Google Scholar : PubMed/NCBI | |
|
Tian Y, Tian Y, Yuan Z, Zeng Y, Wang S, Fan X, Yang D and Yang M: Iron metabolism in aging and age-related diseases. Int J Mol Sci. 23:36122022. View Article : Google Scholar : PubMed/NCBI | |
|
Baier MP, Sharum SI, Wilson JL, Narasimhan S, Salim C and Logan S: Enhancing mitochondrial matrix antioxidant SOD2 in astrocytes mitigates cellular senescence and cognitive impairment in aging. Aging Dis. Oct 9–2025.Epub ahead of print. PubMed/NCBI | |
|
Lu Q, Shao N, Fang Z, Ouyang Z, Shen Y, Yang R, Liu H, Cai B and Wei T: The anti-Alzheimer's disease effects of ganoderic acid a by inhibiting Ferroptosis-lipid peroxidation via activation of the NRF2/SLC7A11/GPX4 signaling pathway. Chem Biol Interact. 412:1114592025. View Article : Google Scholar : PubMed/NCBI | |
|
Wang M, Li M, Jiang Y, Wang S, Yang X, Naseem A, Algradi AM, Hao Z, Guan W, Chen Q, et al: Saponins from Astragalus membranaceus(Fisch.) Bge alleviated neuronal ferroptosis in Alzheimer's disease by regulating the NOX4/Nrf2 signaling pathway. J Agric Food Chem. 73:7725–7740. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Meng X, Zhao W, Yang R, Xu SQ, Wang SY, Li MM, Jiang YK, Hao ZC, Guan W, Kuang HX, et al: Lignans from Schisandra chinensis(Turcz.) Baill ameliorates cognitive impairment in Alzheimer's disease and alleviates ferroptosis by activating the Nrf2/FPN1 signaling pathway and regulating iron levels. J Ethnopharmacol. 341:1193352025. View Article : Google Scholar | |
|
Li Z, Lu Y, Zhen Y, Jin W, Ma X, Yuan Z, Liu B, Zhou XL and Zhang L: Avicularin inhibits ferroptosis and improves cognitive impairments in Alzheimer's disease by modulating the NOX4/Nrf2 axis. Phytomedicine. 135:1562092024. View Article : Google Scholar : PubMed/NCBI | |
|
Deng PX, Silva M, Yang N, Wang Q, Meng X, Ye KQ, Gao HC and Zheng WH: Artemisinin inhibits neuronal ferroptosis in Alzheimer's disease models by targeting KEAP1. Acta Pharmacol Sin. 46:326–337. 2025. View Article : Google Scholar | |
|
Yong Y, Yan L, Wei J, Feng C, Yu L, Wu J, Guo M, Fan D, Yu C, Qin D, et al: A novel ferroptosis inhibitor, thonningianin a, improves Alzheimer's disease by activating GPX4. Theranostics. 14:6161–6184. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Wu Y, Wei M, Wang M, Guo M, Yu H, Chen Y, Xu T and Zhou Y: Schisandra total lignans ameliorate neuronal ferroptosis in 3xTg-AD mice via regulating NADK/NADPH/GSH pathway. Phytomedicine. 140:1566122025. View Article : Google Scholar : PubMed/NCBI | |
|
Ding T, Song M, Wu Y, Li Z, Zhang S and Fan X: Schisandrin B ameliorates Alzheimer's disease by suppressing neuronal ferroptosis and ensuing microglia M1 polarization. Phytomedicine. 142:1567802025. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang ZH, Chen C, Jia SZ, Cao XC, Liu M, Tian J, Hoffmann PR, Xu HX, Ni JZ and Song GL: Selenium restores synaptic deficits by modulating NMDA receptors and selenoprotein K in an Alzheimer's disease model. Antioxid Redox Signal. 35:863–884. 2021. View Article : Google Scholar | |
|
Zeng H and Jin Z: The role of ferroptosis in Alzheimer's disease: Mechanisms and therapeutic potential (review). Mol Med Rep. 32:1922025. View Article : Google Scholar : PubMed/NCBI | |
|
Wang J, Wang Z, Li Y, Hou Y, Yin C, Yang E, Liao Z, Fan C, Martin LL and Sun D: Blood brain Barrier-targeted delivery of double selenium nanospheres ameliorates neural ferroptosis in Alzheimer's disease. Biomaterials. 302:1223592023. View Article : Google Scholar | |
|
Sun C, Gao X, Sha S, Wang S, Shan Y, Li L, Xing C, Guan H and Du H: Berberine alleviates Alzheimer's disease by activating autophagy and inhibiting ferroptosis through the JNK-p38MAPK signaling pathway. Int Immunopharmacol. 155:1145502025. View Article : Google Scholar : PubMed/NCBI | |
|
Song D, Gui F, Li G, Zhuang S, Sun J, Tan X, Hong C and Huang J: Neuritin improves cognitive impairments in APP/PS1 Alzheimer's disease mice model by mitigating neuronal ferroptosis via PI3K/Akt activation. Int J Biol Macromol. 303:1406622025. View Article : Google Scholar : PubMed/NCBI | |
|
Guo Y, Zhao J, Liu X, Lu P, Liang F, Wang X, Wu J and Hai Y: Ghrelin induces ferroptosis resistance and M2 polarization of microglia to alleviate neuroinflammation and cognitive impairment in Alzheimer's disease. J Neuroimmune Pharmacol. 20:62025. View Article : Google Scholar : PubMed/NCBI | |
|
Tan L, Xie J, Liao C, Li X, Zhang W, Cai C, Cheng L and Wang X: Tetrahedral framework nucleic acids inhibit aβ-mediated ferroptosis and ameliorate cognitive and synaptic impairments in Alzheimer's disease. J Nanobiotechnology. 22:6822024. View Article : Google Scholar | |
|
Lei L, Yuan J, Dai Z, Xiang S, Tu Q, Cui X, Zhai S, Chen X, He Z, Fang B, et al: Targeting the labile iron pool with engineered DFO nanosheets to inhibit ferroptosis for Parkinson's disease therapy. Adv Mater. 36:e24093292024. View Article : Google Scholar : PubMed/NCBI | |
|
Ko CJ, Gao SL, Lin TK, Chu PY and Lin HY: Ferroptosis as a major factor and therapeutic target for neuroinflammation in Parkinson's disease. Biomedicines. 9:16792021. View Article : Google Scholar : PubMed/NCBI | |
|
Han Z, Wang B, Wen YQ, Li YN, Feng CX, Ding XS, Shen Y, Yang Q and Gao L: Acteoside alleviates lipid peroxidation by enhancing Nrf2-mediated mitophagy to inhibit ferroptosis for neuroprotection in Parkinson's disease. Free Radic Biol Med. 223:493–505. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Kong L, Wang Y, Tong Z, Dai R, Yusuf A, Du L, Liu B, Huang Z and Hu L: Granulathiazole a protects 6-OHDA-induced Parkinson's disease from ferroptosis via activating Nrf2/HO-1 pathway. Bioorg Chem. 147:1073992024. View Article : Google Scholar : PubMed/NCBI | |
|
Pandey SK, Nanda A, Gautam AS and Singh RK: Betulinic acid protects against lipopolysaccharide and ferrous sulfate-induced oxidative stress, ferroptosis, apoptosis, and neuroinflammation signaling relevant to Parkinson's disease. Free Radic Biol Med. 233:340–354. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Fu G, Li T, Zhao Y, Zhang S, Xue X, Yang Y, Li T, Luo S, Yue G and Lei T: Dingzhen pills inhibit neuronal ferroptosis and neuroinflammation by inhibiting the cGAS-STING pathway for Parkinson's disease mice. Chin Med. 20:872025. View Article : Google Scholar : PubMed/NCBI | |
|
Dong X, Yang T and Jin Z: Lactobacillus Reuteri-derived γ-amino butyric acid alleviates MPTP-induced Parkinson's disease through inhibiting Ferroptosis via the AKT-GSK3β-GPX4 axis. NPJ Parkinsons Dis. 11:1492025. View Article : Google Scholar | |
|
Yusun A, Wan HM, Chen HX, Sun M, Zhang CN and Ding XD: Emodin ameliorates dopaminergic neuron loss in the MPP+ induced Parkinson's disease model: Significant inhibition of Ferroptosis by activating UQCRC1 protein. Nat Prod Res. 1–12. 2025. View Article : Google Scholar | |
|
Lei Y, Zhou J, Xu D, Chai S and Xiong N: Corilagin attenuates neuronal apoptosis and ferroptosis of Parkinson's disease through regulating the TLR4/src/NOX2 signaling pathway. ACS Chem Neurosci. 16:968–980. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Liu X, Zheng Z, Xue C, Wang X, Li J, Liu Z, Xin W, Xu X, Zhou D, Yao L and Lu G: LRRK2 mediates α-synuclein-induced neuroinflammation and ferroptosis through the p62-Keap1-Nrf2 pathway in Parkinson's disease. Inflammation. 48:3666–3691. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Huang L, Pi J, Gu L, Liao Z and Wang W: Teneligliptin, a DPP4 inhibitor protects dopaminergic neurons in PD models via inhibiting of oxidative stress and ferroptosis. Eur J Pharmacol. 1002:1777822025. View Article : Google Scholar : PubMed/NCBI | |
|
An JR, Su JN, Sun GY, Wang QF, Fan YD, Jiang N, Yang YF and Shi Y: Liraglutide alleviates cognitive deficit in db/db mice: Involvement in oxidative stress, iron overload, and ferroptosis. Neurochem Res. 47:279–294. 2022. View Article : Google Scholar | |
|
Chen J, Guo P, Han M, Chen K, Qin J and Yang F: Cognitive protection of sinomenine in type 2 diabetes mellitus through regulating the EGF/Nrf2/HO-1 signaling, the microbiota-gut-brain axis, and hippocampal neuron ferroptosis. Phytother Res. 37:3323–3341. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Wang B, Zhu S, Guo M, Ma RD, Tang YL, Nie YX and Gu HF: Artemisinin ameliorates cognitive decline by inhibiting hippocampal neuronal ferroptosis via Nrf2 activation in T2DM mice. Mol Med. 30:352024. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Z, Feng S, Li Q, Song Z, He J, Yang S, Yan C and Ling H: Dihydromyricetin alleviates hippocampal ferroptosis in type 2 diabetic cognitive impairment rats via inhibiting the JNK-inflammatory factor pathway. Neurosci Lett. 812:1374042023. View Article : Google Scholar : PubMed/NCBI | |
|
Hu Y, Xu J, Wang J, Wang J, Li Y, Chen W and Zhang Q: Resveratrol alleviates diabetic adipose Tissue-derived extracellular vesicles-induced hippocampal ferroptosis and cognitive dysfunction via inhibiting miR-9-3p/SLC7A11 axis. Mol Neurobiol. 62:12307–12330. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Chen X, Pang X, Yeo AJ, Xie S, Xiang M, Shi B, Yu G and Li C: The molecular mechanisms of ferroptosis and its role in blood-brain barrier dysfunction. Front Cell Neurosci. 16:8897652022. View Article : Google Scholar : PubMed/NCBI | |
|
Wang J, Cao Y, Lu Y, Zhu H, Zhang J, Che J, Zhuang R and Shao J: Recent progress and applications of small molecule inhibitors of Keap1-Nrf2 axis for neurodegenerative diseases. Eur J Med Chem. 264:1159982024. View Article : Google Scholar | |
|
Liu Q, Zhao Y, Zhou H and Chen C: Ferroptosis: Challenges and opportunities for nanomaterials in cancer therapy. Regen Biomater. 10:rbad0042023. View Article : Google Scholar : PubMed/NCBI | |
|
Delila L, Nebie O, Le NTN, Timmerman K, Lee DY, Wu YW, Chou ML, Buée L, Chou SY, Blum D, et al: Neuroprotective effects of intranasal extracellular vesicles from human platelet concentrates supernatants in traumatic brain injury and Parkinson's disease models. J Biomed Sci. 31:872024. View Article : Google Scholar : PubMed/NCBI | |
|
Liu J, Huang J, Zhang Z, Zhang R, Sun Q, Zhang Z, Liu Y and Ma B: Mesenchymal stem cell-derived exosomes ameliorate delayed neurocognitive recovery in aged mice by inhibiting hippocampus ferroptosis via activating SIRT1/Nrf2/HO-1 signaling pathway. Oxid Med Cell Longev. 2022:35932942022. View Article : Google Scholar : PubMed/NCBI | |
|
Xu Y, Jia B, Li J, Li Q and Luo C: The interplay between ferroptosis and neuroinflammation in central neurological disorders. Antioxidants (Basel). 13:3952024. View Article : Google Scholar : PubMed/NCBI | |
|
Cai Z, Wang S, Cao S, Chen Y, Penati S, Peng V, Yuede CM, Beatty WL, Lin K, Zhu Y, et al: Loss of ATG7 in microglia impairs UPR, triggers ferroptosis, and weakens amyloid pathology control. J Exp Med. 222:e202301732025. View Article : Google Scholar : PubMed/NCBI | |
|
Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS, et al: Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell. 149:1060–1072. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Li Y, Sun M, Cao F, Chen Y, Zhang L, Li H, Cao J, Song J, Ma Y, Mi W and Zhang X: The ferroptosis inhibitor liproxstatin-1 ameliorates LPS-induced cognitive impairment in mice. Nutrients. 14:45992022. View Article : Google Scholar : PubMed/NCBI | |
|
Long HZ, Cheng Y, Zhou ZW, Luo HY, Wen DD and Gao LC: The key roles of organelles and ferroptosis in alzheimer's disease. J Neurosci Res. 100:1257–1280. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Conrad M and Proneth B: Selenium: Tracing another essential element of ferroptotic cell death. Cell Chem Biol. 27:409–419. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Kalyanaraman B: NAC, NAC, Knockin' on Heaven's door: Interpreting the mechanism of action of N-acetylcysteine in tumor and immune cells. Redox Biol. 57:1024972022. View Article : Google Scholar : PubMed/NCBI | |
|
Xu J, Shen R, Qian M, Zhou Z, Xie B, Jiang Y, Yu Y and Dong W: Ferroptosis in Alzheimer's disease: The regulatory role of glial cells. J Integr Neurosci. 24:258452025. View Article : Google Scholar : PubMed/NCBI | |
|
Li Y, Zhang E, Yang H, Chen Y, Tao L, Xu Y, Chen T and Shen X: Gastrodin ameliorates cognitive dysfunction in vascular dementia rats by suppressing ferroptosis via the regulation of the Nrf2/Keap1-GPx4 signaling pathway. Molecules. 27:63112022. View Article : Google Scholar : PubMed/NCBI | |
|
Yang L and Nao J: Ferroptosis: A potential therapeutic target for Alzheimer's disease. Rev Neurosci. 34:573–598. 2023. View Article : Google Scholar | |
|
Mesa-Herrera F, Marín R, Torrealba E and Díaz M: Multivariate assessment of lipoxidative metabolites, trace biometals, and antioxidant and detoxifying activities in the cerebrospinal fluid define a fingerprint of preclinical stages of Alzheimer's disease. J Alzheimers Dis. 86:387–402. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Mayr E, Rotter J, Kuhrt H, Winter K, Stassart RM, Streit WJ and Bechmann I: Detection of molecular markers of ferroptosis in human Alzheimer's disease brains. J Alzheimers Dis. 102:1133–1154. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Ayton S, Wang Y, Diouf I, Schneider JA, Brockman J, Morris MC and Bush AI: Brain iron is associated with accelerated cognitive decline in people with Alzheimer pathology. Mol Psychiatry. 25:2932–2941. 2020. View Article : Google Scholar | |
|
Shi YS, Chen JC, Lin L, Cheng YZ, Zhao Y, Zhang Y and Pan XD: Dendrobine rescues cognitive dysfunction in diabetic encephalopathy by inhibiting ferroptosis via activating Nrf2/GPX4 axis. Phytomedicine. 119:1549932023. View Article : Google Scholar : PubMed/NCBI | |
|
Yang Q, Zhou L, Liu C, Liu D, Zhang Y, Li C, Shang Y, Wei X, Li C and Wang J: Brain iron deposition in type 2 diabetes mellitus with and without mild cognitive impairment-an in vivo susceptibility mapping study. Brain Imaging Behav. 12:1479–1487. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Hao L, Mi J, Song L, Guo Y, Li Y, Yin Y and Zhang C: SLC40A1 mediates ferroptosis and cognitive dysfunction in type 1 diabetes. Neuroscience. 463:216–226. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Clarke NC, McCabe E, Jensen LD, Creaven BS and Costello DA: Salicylaldehyde benzoylhydrazone protects against ferroptosis in models of neurotoxicity and behavioural dysfunction, in vitro and in vivo. J Mol Neurosci. 75:772025. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Y, Li P, Liang Y and Wang D: ANO6 targets TMEM30A to regulate endoplasmic reticulum Stress-induced lipid peroxidation and ferroptosis in Alzheimer's cells. Cell Biochem Biophys. 83:3707–3715. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Lu Z, Jiang Z, Huang X, Chen Y, Feng L, Mai J, Lao L, Li L, Chen WH and Hu J: Anti-Alzheimer effects of an HDAC6 inhibitor, WY118, alone and in combination of lithium chloride: Synergistic suppression of ferroptosis via the modulation of tau phosphorylation and MAPK signaling. Eur J Pharmacol. 997:1776052025. View Article : Google Scholar : PubMed/NCBI | |
|
Ramesh M and Govindaraju T: MiR-7a-Klf4 axis as a regulator and therapeutic target of neuroinflammation and ferroptosis in Alzheimer's disease. bioRxiv. Mar 25–2025.Epub ahead of print. View Article : Google Scholar | |
|
Zheng N, Zhang Z, Liu H, Zong S, Zhang L, Cui X, Liu Y, Wang C, Chen R and Lu Z: MK886 ameliorates Alzheimer's disease by activating the PRKCI/AKT signaling pathway. Eur J Pharmacol. 993:1773592025. View Article : Google Scholar : PubMed/NCBI | |
|
Feng Y, Wang H, Hu Y, Zhang X, Miao X, Li Z and Jia J: Hederagenin ameliorates ferroptosis-induced damage by regulating PPARα/Nrf2/GPX4 signaling pathway in HT22 cells: An in vitro and in silico study. Bioorg Chem. 155:1081192025. View Article : Google Scholar | |
|
Tian L, Li H, Xiong W, Li X, Duan S, Yang C and Shi C: Proteomic alteration in catalpol treatment of Alzheimer's disease by regulating HSPA5/GPX4. Eur J Pharmacol. 987:1770752025. View Article : Google Scholar | |
|
Gugliandolo A, Bramanti P and Mazzon E: Role of vitamin E in the treatment of Alzheimer's disease: Evidence from animal models. Int J Mol Sci. 18:25042017. View Article : Google Scholar : PubMed/NCBI | |
|
Xiao S, Liu L, Qin X, Xu L, Chai Z and Li Z: Astragenol alleviates neuroinflammation and improves Parkinson's symptoms through amino acid metabolism pathway and inhibition of ferroptosis. J Ethnopharmacol. 348:1198962025. View Article : Google Scholar : PubMed/NCBI | |
|
Wang J, Geng T, Yao X and Liu Y: GDF15 attenuates Parkinson's disease progression via suppressing the activation of cGAS-STING pathway. Mol Cell Biochem. 480:4449–4466. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao X, Kang Z, Han R, Wang M, Wang Y, Sun X, Wang C, Zhou J, Cao L and Lu M: JWA binding to NCOA4 alleviates degeneration in dopaminergic neurons through suppression of ferritinophagy in Parkinson's disease. Redox Biol. 73:1031902024. View Article : Google Scholar : PubMed/NCBI | |
|
Fu X, Qu L, Xu H and Xie J: Ndfip1 protected dopaminergic neurons via regulating mitochondrial function and ferroptosis in Parkinson's disease. Exp Neurol. 375:1147242024. View Article : Google Scholar : PubMed/NCBI | |
|
Wang T, Sobue A, Watanabe S, Komine O, Saido TC, Saito T and Yamanaka K: Dimethyl fumarate improves cognitive impairment and neuroinflammation in mice with Alzheimer's disease. J Neuroinflammation. 21:552024. View Article : Google Scholar : PubMed/NCBI |