- Review
- Open access
- Published:
The interplay of iron, oxidative stress, and α-synuclein in Parkinson’s disease progression
Molecular Medicine volume 31, Article number: 154 (2025)
Abstract
The irreversible degeneration of dopamine neurons induced by α-synuclein (α-syn) aggregation in the substantia nigra is the central pathological feature of Parkinson's disease (PD). Neuroimaging and pathological autopsy studies consistently confirm significant iron accumulation in the brain of PD patients, suggesting a critical role for iron in disease progression. Current research has established that iron overload induces ferroptosis in dopaminergic neurons, evidence indicates that the impact of iron on PD pathology extends beyond ferroptosis. Iron also plays a regulatory role in modulating α-syn, affecting its aggregation, spatial conformation, post-translational modifications, and mRNA stability. Iron-induced α-syn aggregation can contribute to dopaminergic neurodegeneration through additional mechanisms, potentially creating a feedback loop in which α-syn further enhances iron accumulation, thus perpetuating a vicious cycle of neurotoxicity. Given α-syn’s intrinsically disordered structure, targeting iron metabolism presents a promising therapeutic strategy for PD. Therefore, the development of iron chelators, alone or in combination with other therapeutic drugs, may offer a beneficial approach to alleviating PD symptoms and slowing disease progression.
Introduction
Parkinson's disease (PD) is a progressive neurodegenerative disorder primarily impacting motor function (Aubignat and Tir 2024). With an aging global population, the prevalence of PD continues to rise, making it an important public health issue worldwide. According to the Global Burden of Disease Study 2019, more than 8.5 million people globally are affected by PD (Xu et al. 2024). First described by Dr. James Parkinson in 1817, PD has remained challenging to treat due to its complex pathology and the difficulty in diagnosing it during its earliest stages. The main pathological feature of PD is the degeneration of neurons in the substantial nigra (SN), largely driven by the accumulation of abnormal α-synuclein (α-syn) (Calabresi et al. 2023). The protein buildup ultimately leads to cellular damage and neuronal death. Because the early clinical symptoms of PD are atypical and often go unrecognised, optimal treatment windows are frequently missed.
Under normal physiological conditions, α-syn is a small, soluble protein with no neurotoxic effect, predominantly found in neuronal synapses, where it is involved in neurotransmitter release and vesicular recycling (Gao et al. 2023). However, in PD, misfolded α-syn aggregates into prefibrillar structures, eventually forming fibrillar α-syn that constitutes the core of Lewy bodies, a hallmark of PD pathology, which ultimately results in neuronal death (Picca et al. 2021; Leitao et al. 2021; Li et al. 2024). α-syn is mainly expressed in presynaptic nerve terminals and is strongly associated with PD. Structurally, α-syn consists of 140 amino acids organised into three domains: the N-terminal (residues 1–60), the central non-amyloid component (NAC, residues 61–95), and the C-terminal (residues 96–140) (Sarchione et al. 2021) (Fig. 1). Various post-translational modifications (PTMs) such as—phosphorylation, ubiquitination, acetylation and nitration—alter α-syn’s biochemical properties (Yoo et al. 2022), affecting its tendency to aggregate, which is pivotal for the diagnosis and treatment of PD (Guhathakurta et al. 2017). In addition, changes in the spatial conformation of α-syn cause α-syn aggregation into Lewy bodies and is associated with neurotoxicity (Calabresi et al. 2023). Factors like oxidative stress and genetic mutations also influence the aggregation of α-syn, increasing its neurotoxicity in PD.
Epidemiological studies have found that long-term exposure to certain metal ions—such as iron, copper, manganese, and aluminium—is a risk factor for PD (Raj et al. 2021; Bisaglia and Bubacco 2020). Among these, iron is especially implicated as it reveals a higher iron deposition in the dopaminergic neurons of the SN in PD patients compared to healthy individuals (Mochizuki et al. 2020). Becker et al. reported that increased iron levels are detected early in PD, preceding the onset of clinical symptoms, as demonstrated by transcranial color-coded sonography (Becker et al. 1995). People with increased iron in the substantia nigra had a 17 times higher risk of developing PD than those with normal iron levels (Berg et al. 2011). In neurons, iron overload, generally in the form of Fe2+, can trigger the Fenton reaction, resulting in lipid peroxidation and neuronal ferroptosis (Jacquemyn et al. 2024), a form of iron dependent cell death. Iron overload is the result of synergistic action of multiple pathways, with the central mechanism being excessive iron uptake mediated by transferrin receptor (TfR) and divalent metal transporter 1 (DMT1) (Lane et al. 2015). While much research has largely focused on inhibiting iron-mediated ferroptosis to ameliorate PD, recent findings suggest that Fe2+ and Fe3+ may also modulate α-syn through ferroptosis—independent mechanisms (Moons et al. 2020). The interaction between Fe2+/Fe3+ and α-syn significantly affects the stability and aggregation of α-syn (Wang et al. 2024), influencing disease progression. More importantly, neuronal ferroptosis occurs before neuronal apoptosis in the development of PD (Zhang et al. 2020), which suggests that early improvement of iron accumulation is the key to treating PD.
Elucidating the mechanisms by which iron contribute to α-syn aggregation in a ferroptosis-independent manner is critical. This review explores the role of Fe2+ and Fe3+ in regulating α-syn aggregation, conformation and modification through alternative pathways unrelated to ferroptosis. By understanding these pathways we aim to provide new insights for early diagnosis and potential therapeutic strategies for PD.
The role of iron in regulating α-syn aggregation
The aggregation of α-syn in neurons is an important pathological feature of PD. Inhibition of α-syn aggregation is considered a promising therapeutic approach for PD management (Hideshima et al. 2022). Under normal physiological conditions, cellular mechanisms such as autophagy, the ubiquitin proteasome pathway, and the unfolded protein response help degrade misfolded proteins. However, excess Fe2+ and Fe3+ disrupts these protective pathways, promoting aggregation of α-syn and ultimately leading to neuronal death (Chen et al. 2019; Le 2014; Tang and Kroemer 2020). Beyond ferroptosis, exogenous Fe2+ and Fe3+ can independently promote aggregation of α-syn and increase its toxicity in a time- and concentration-dependent manner (Li et al. 2010).
Studies have shown that iron overload exacerbates α-syn pathology. For instance, in Drosophila models expressing the α-syn A53 T mutant, iron (ferric ammonium citrate, FAC) accumulation exacerbates motor dysfunction (Zhu et al. 2016), underscoring iron’s role in the α-syn aggregation and neurotoxicity. Similarly, in BE-M17 cells overexpressing α-syn, Fe2+ enhances its aggregation, leading to the formation of Lewy bodies (Ostrerova-Golts et al. 2000). Transmission electron microscopy (TEM) studies have revealed that, unlike copper (Cu2+), which induces amorphous aggregates in mutant α-syn with no fibril formation, Fe3+ triggers the formation of short, thick fibrils (Bharathi and Rao 2007). This suggests that aggregation of α-syn is metal-specific and iron (Fe2+ and Fe3+)-induced aggregation being particularly toxic compared to copper—induced aggregation (Li et al. 2022). Confocal single-molecule fluorescence analysis shows that Fe3+ induces aggregation of α-syn and induces the formation of large number of oligomers, which serve as seeds for amyloid fibril formation (Kostka et al. 2008). Notably, this process appears to be independent of the Fenton reaction and depends only on the direct interaction between Fe3+ and α-syn (Levin et al. 2011). Even Fe3+ at physiological concentrations still promotes the formation of α-syn oligomers (Kostka et al. 2008). Additionally, Fe2+ has been found to promote the aggregation of α-syn, while Fe3+ exhibits a concentration-dependent effect: at high concentrations, Fe3+ inhibits α-syn fibrillation, but at lower concentrations, it promotes α-syn fibrillation (Zhao et al. 2023). Cryo-electron microscopy (cryo-EM) study shows that low concentrations of Fe3+ bind to the negatively charged clefts of α-syn fibrils, promoting further aggregation. In contrast, TEM imaging reveals that high concentrations of Fe3+ leads to formation of spherical α-syn oligomers, which prevents further assembly of amyloid fibrils (Zhao et al. 2023). Thioflavin (ThT) kinetic trajectory and circular dichroism studies show that the binding of Fe3+ to α-syn increases the rate of fiber formation and leads to more secondary structures (β-sheets) (Uversky et al. 2001). These findings suggest that Fe3+ plays a more direct role in promoting the aggregation of α-syn (Uversky et al. 2001; Peng et al. 2010), whereas Fe2+ primarily contributes to α-syn aggregation by generating high levels of reactive oxygen species (ROS) through the Fenton reaction.
The role of iron in promoting α-syn aggregation by regulating its conformation
Altered α-syn conformation in neurons is a key factor in α-syn aggregation during PD. The N-terminal domain of α-syn, which is positively charged, interacts with the phospholipid bilayer to form α-helical structures (Musteikyte et al. 2021). The NAC domain regulates protein fibrillation (Gallardo et al. 2020), while negatively charged C-terminal domain also contributes to α-syn aggregation (Emamzadeh 2016; Zhang et al. 2022). The C-terminal domain of α-syn is rich in acidic amino acids, such as glutamate (Glu) and aspartate (Asp), giving it a negative charge under physiological conditions (Emamzadeh 2016). Metal ions interacting with the C-terminal of α-syn tend to expose the NAC region, accelerating fibril formation (Atarod et al. 2022). Conformational shifts in α-syn are crucial for synaptic transmission: while physiological shifts facilitate neurotransmitter release and vesicle recycling, pathological aggregation leads to synaptic dysfunction and promotes neuronal death. The extent and nature of α-syn’s conformational changes directly correlate with PD progression and symptom severity (So and Watts 2023; Tofaris 2022).
Research on iron’s role in promoting α-syn aggregation remains limited, but emerging evidence highlights key interactions. The nuclear magnetic resonance (NMR) analysis demonstrates that Fe2+ can bind to the C-terminal region of α-syn (Binolfi et al. 2006), altering the spatial conformation of α-syn and promoting its aggregation (Abeyawardhane et al. 2018). Similarly, Fe3+ induces a shift in α-syn structure from an α-helical to a β-sheet conformation, enhancing its propensity to aggregate (Kaindlstorfer et al. 2018) (Fig. 2). Cryo-electron microscopy studies show that Fe3+ binds to the negative cleft of α-syn fibrils, stabilizing the fibrillar structure and promoting further aggregation (Zhao et al. 2023). Beyond fibril formation, Fe3+ promotes the formation of α-syn oligomers, which can create pore-like structures in planar lipid bilayers (Kostka et al. 2008), altering neuronal membrane permeability and triggering neuronal death (Tsigelny et al. 2012) (Fig. 2). Both Fe3+ and Fe2+ binds to α-syn, inducing conformational changes that accelerate its aggregation (Moons et al. 2020; Peng et al. 2010). These findings suggest that Fe2+ and Fe3+ stabilises α-syn fibrils by binding to negatively charged residues via electrostatic interactions, promoting toxic aggregation and contributing to dopamine neuron degeneration in PD.
The role of iron in promoting α-syn aggregation by inhibiting autophagy
Dysfunction in autophagy is a key factor contributing to α-syn accumulation and the progression of PD. Previous studies have reported that iron overload can inhibit autophagy, thereby promoting α-syn accumulation in neurons (Wan et al. 2017; Wu et al. 2017). Specifically, Fe2+ overload activates the protein kinase B (AKT)/mechanistic target of rapamycin complex 1 (mTORC1) pathway, while also inhibiting the expression of transcription factor EB (TFEB), a master transcriptional regulator of autophagosome-lysosome fusion. This inhibition exacerbates α-syn aggregation and propagation (Xiao et al. 2018). Additionally, Fe3+ deposition suppresses insulin-like growth factor 2 (IGF2) and the transcription factor zinc finger protein 27 (ZFP27), which decreased LC3 expression and further impairs autophagy (Wang et al. 2023) (Fig. 3).
Iron overload activates the AKT/mTORC1 signalling pathway and inhibits TFEB expression, thereby inhibiting autophagy-mediated degradation of misfolded α-syn and leading to neuronal death. Iron overload suppresses IGF2 and ZFP27, which decreased LC3 expression and further impairs autophagy. Iron overload in lysosomes impairs autophagy
Furthermore, both Fe2+ and Fe3+ overload lead to iron accumulation within lysosomes, impairing their function and contributing to defective autophagy (Fernandez et al. 2016) (Fig. 3). These results illustrate that iron accumulation disrupts autophagic processes, preventing the clearance of α-syn and ultimately resulting in neuronal loss.
The role of iron in promoting α-syn aggregation by enhancing α-syn mRNA expression
Iron promotes α-syn aggregation by increasing its mRNA translation. Iron binds to iron regulatory protein (IRP), causing its dissociation from the iron response element (IRE), which enhances the translation of transcripts associated with α-syn (Zhou and Tan 2017). In SK-N-SH cells, IRP knockdown has been shown to upregulate α-syn mRNA expression, resulting in increased α-syn aggregation (Friedlich et al. 2007). Furthermore, the use of iron chelators reduces α-syn mRNA expression in HEK293 cells (Li et al. 2011).
Administration of iron chelators reduce the mRNA expression of α-syn in HEK293 cells (Febbraro et al. 2012). Research by Tong et al. identified Synucleozid-2.0, a small molecule that targets the IRE region of α-syn mRNA, promoting its degradation and reducing α-syn levels (Tong and Zhang 2024). These findings suggest that iron overload facilitates IRP dissociation from α-syn mRNA, enhancing α-syn translation and promoting its aggregation, which worsens PD pathology.
Additionally, oxidative stress in PD generates reactive oxygen species (ROS), which oxidise Fe2+ to Fe3+, further promoting α-syn aggregation (Levin et al. 2011). It's worth noting that α-syn functions as a ferrireductase to reduce Fe3+ to Fe2+ (Sian-Hulsmann and Riederer 2020). This forms a self-perpetuating cycle of iron-induced α-syn aggregation and neuronal damage, exacerbating PD progression (Fig. 4).
Iron overload binds to the iron regulatory proteins (IRP), causing their release from the iron-responsive element (IRE) on α-syn mRNA, which enhances α-syn translation and contributes to increased protein formation and aggregation. α-syn functions as a ferrireductase, reducing Fe3+ to Fe2+. Fe2+ catalyse H2O2 to produce Fe3+ via the Fenton reaction
The role of iron in regulating α-syn propagation
Research indicates that under pathological conditions, α-syn can be released from the cytoplasm into the extracellular matrix, where it is taken up by neighbouring neurons. This process is especially prevalent for “seed-toxic” oligomers of α-syn, which are more likely to be internalised by recipient neurons (Danzer et al. 2012). Studies have shown that Fe3+ promotes the transcellular propagation of α-syn in a concentration-dependent manner (Li et al. 2020). In addition, Fe3+ enhances the binding of α-syn to heparan sulfate proteoglycans (HSPGs) through its positive charge, altering the surface properties of α-syn and promoting fibril internalisation. This accelerates the prion-like propagation of α-syn (Li et al. 2020) (Fig. 5).
Although many studies link iron accumulation to increased α-syn levels, the direct mechanisms by which iron promotes α-syn propagation remains unmapped. Nonetheless, these findings suggest that iron plays a critical role in facilitating the spread of α-syn aggregates between cells, contributing to the progression of PD.
The role of iron in promoting α-synphosphorylation, exacerbating its aggregation
Post-translational modifications of α-syn is essential for its pathological aggregation in PD (Zhang et al. 2021). Among these, phosphorylation of α-syn, particularly at serine 129, is particularly significant. While only a small proportion of α-syn is phosphorylated in healthy brains, over 90% of the abnormally aggregated α-syn in PD patients is phosphorylated at serine 129 (Xu et al. 2015; Kawahata et al. 2022). Although phosphorylation of α-syn has been shown to induce cytotoxicity and promotes the formation of Lewy bodies, its exact role in PD pathology remains ambiguous (Ramalingam et al. 2024; Kontaxi and Edwards 2023).
Iron overload induces phosphorylation of α-syn at serine 129, correlating with increased expression of casein kinase 2 (CK2) and polo-like kinase 2 (PLK2), both of which regulate α-syn phosphorylation (Wang et al. 2019; Perfeito et al. 2014). Administration of antioxidants, such as N-acetyl-L-cysteine, protects neurons from the oxidative stress associated with iron overload, suggesting that ROS generated by iron may induce phosphorylation of α-syn thereby exacerbating its aggregation (Wang et al. 2019). The functional implication of α-syn phosphorylation at serine 129 is still being explored. Some studies indicate that this modification prevents α-syn oligomers from binding to the cell membranes, potentially protecting the cell membrane from neurotoxic effects (Sato et al. 2013). Others propose that phosphorylation of α-syn at serine 129 occurs during the initial stages of α-syn aggregation, which suppresses further aggregation (Ghanem et al. 2022). Given that iron accumulation occurs early in PD, we suggest that early iron-induced phosphorylation of α-syn is protective. However, as PD progresses, excessive iron accumulation exacerbates phosphorylation and induces α-syn aggregation. Therefore, the impact of serine 129 phosphorylation on PD pathology needs to be further investigated (Fig. 6). Furthermore, while iron’s role in α-syn phosphorylation is well documented, its effect on other post translational modifications, such as acylation and nitration remains unclear. Further research is needed to clarify their contributions to PD progression.
Iron overload induces phosphorylation of α-syn at serine 129, accompanied by elevated expression of CK2 and PLK2, which regulates α-syn phosphorylation at Ser129. Phosphorylation of α-syn at serine 129 prevents α-syn oligomers from binding to the cell membranes, potentially protecting the cell membrane from neurotoxic effects. The precise role of serine 129 phosphorylation in α-syn’s function and toxicity remains unclear
The role of iron in promoting α-syn aggregation by regulating ROS
Fe2+ can effectively catalyse H2O2 to produce Fe3+ and ROS via the Fenton reaction, contributing to oxidative stress (Yan et al. 2022). Interestingly, α-syn itself functions as a ferrireductase, reducing Fe3+ to Fe2+ (Jansen van Rensburg et al. 2021). This continuous cycle, combined with elevated expression of α-syn, leads to the accumulation of Fe2+, resulting in increased ROS production and neuronal toxicity. Davies et al. demonstrated that α-syn overexpression significantly increases ferrireductase activity, increasing the level of Fe2+ in SH-SY5Y cells lysates (Davies et al. 2011). Studies further show that the administration of metal chelators such as deferoxamine (DFO) reduces α-syn oligomer-induced ROS accumulation, mitigating oxidative damage and reducing the iron level (Shen et al. 2024; Deas et al. 2016).
While iron accumulation alone is not considered the primary cause of PD, early-life exposure to excess iron increases PD risk later in life (Hare et al. 2015; Zecca et al. 2004). Impaired iron metabolism is particularly detrimental to dopaminergic neurons (Hare and Double 2016), and increased iron levels in the substantia nigra region of the midbrain have been suggested as a potential early marker for PD (Martin et al. 2008). Abnormal aggregation of α-syn causes ROS accumulation, resulting in dopamine neuron death and contributing to PD progression. However, whether ROS accumulation directly causes α-syn aggregation in the early stages of PD is still unclear.
In animal models, long-term administration of MPTP, a neurotoxin used to mimic PD, causes accumulation of α-syn in the substantia nigra region of mice (Zhang et al. 2017). MPTP is metabolised to MPP+ by monoamine oxidase B, which inhibits mitochondrial complex I, leading to ROS accumulation and PD-like symptoms. In patients with asymptomatic PD, reduced glutathione levels in the substantia nigra mirror those in advanced PD, suggesting that oxidative stress may precede α-syn aggregation (Jenner et al. 1992).
Early accumulation of ROS may be an important cause of α-syn aggregation, and iron buildup could serve as an early marker of PD by inducing oxidative stress that promotes α-syn aggregation. We propose that ROS buildup plays a central role in the progression of PD by driving α-syn aggregation and exacerbating neuronal damage.
The role of iron in promoting dopaminergic neuronal death through apoptosis and necroptosis
Iron is essential for various physiological processes, including oxygen transport and energy metabolism. Excess iron leads to oxidative stress, high levels of ROS, and dopaminergic neuronal apoptosis. Intracerebroventricular injection of iron citrate exacerbated MPTP-induced Parkinsonism by promoting apoptosis via the Bcl-2/Bax and activated caspase-3 pathway (You et al. 2015). Ci et al. reported that knockdown of the iron regulatory protein 2 (IRP2) gene induces iron accumulation in the SN region, exacerbating neuronal apoptosis and PD-like symptoms (Ci et al. 2020). We suggest that ferroptosis precedes other iron-mediated neurotoxicity, as ferroptosis inhibitors can mitigate iron-induced apoptosis, whereas apoptosis inhibitors fail to prevent ferroptosis (Zhang et al. 2020). In conclusion, intracellular iron overload causes apoptosis and ferroptosis, and the use of iron chelators such as DFO protects neurons, effectively (Zeng et al. 2021).
In addition, necroptosis contributes to dopaminergic neuronal degeneration in the SN region of PD. Analysis of postmortem brain tissue of PD patients revealed elevated levels of the key necroptotic proteins, including receptor-interacting protein kinase 1 (RIPK1), receptor-interacting protein kinase 3 (RIPK3) and mixed lineage kinase domain-like (MLKL) (Iannielli et al. 2018).. Many studies have confirmed the presence of necroptotic proteins in animal models of PD (Leem et al. 2023, 2024). Necroptosis inhibitors, such as necrostatin-1, protects dopaminergic neurons from 6-hydroxydopamine -induced neurotoxicity (Wu et al. 2015; Onate et al. 2020). Iron overload is known to cause ROS accumulation and induce necroptosis. The use of the ferroptosis inhibitor ferrostatin-1 is more effective for iron overload toxicity induced by iron dextrose compared to necrosis inhibitors and apoptosis inhibitors (Kumfu et al. 2023).
In conclusion, elevated ROS induced by iron overload causes abnormal accumulation of α-syn, ultimately leading to neuronal death. Iron accumulation may precede the clinical onset of PD (Pyatigorskaya et al. 2020; Alushaj et al. 2024) with ferroptosis serving as the main mode of dopaminergic neuronal loss in early PD.
Discussion
PD is characterised by excessive iron accumulates in the brain, marked by increased levels of iron and iron-binding proteins such as ferritin (Ru et al. 2024). This iron overload, coupled with reduced glutathione levels, indicates disrupted redox homeostasis in patients with PD. Oxidative damage from iron accumulation is a major causes of PD pathogenesis (David et al. 2022), and neuronal ferroptosis due to iron overload is an important hallmark of PD. While current studies largely focuses on improving PD by inhibiting iron-induced toxicity, exploring alternative pathways of iron-related pathology may provide additional therapeutic insights. In particular, aggregation of α-syn and the resulting formation of Lewy bodies remain key pathological feature of PD. We suggest that Fe3+ is more effective in promoting the aggregation of α-syn (Uversky et al. 2001; Peng et al. 2010), whereas Fe2+ causes α-syn aggregation mainly by generating high levels of ROS through the Fenton reaction. However, Fe3+ produced by the Fenton reaction is subsequently reduced to Fe2+ by cellular reductase, creating a vicious cycle that further damages dopaminergic neurons.
Dysregulated iron homeostasis promotes oxidative stress and dopaminergic neuronal damage, both of which are central to the pathogenesis of PD (Holbein and Lehmann 2023). Iron accumulation, observed initially in the midbrain and gradually extending to the striatum, correlates with early PD clinical features and diagnosis (Yan et al. 2024). Inhibition or downregulation of transferrin in the central nervous system has been shown to reduce iron levels in the brain, which significantly ameliorates ROS in a 6-OHDA model, allocating oxidative stress (Xue et al. 2020). Elevated expression of divalent metal-ion transporter 1 (DMT1) and IRE in patients with PD significantly contribute to iron accumulation in the SN of patients of PD (Ndayisaba et al. 2019). Furthermore, activation of the mitogen-activated protein kinase (MAPK) pathway by α-syn inhibits ubiquitination of DMT1, promoting DMT1 expression and enhancing iron influx into neurons (Bi et al. 2020). This interplay suggests that aggregation of α-syn initially triggers iron deposition in the brain, exacerbating aggregation of α-syn, creating a detrimental feedback loop that accelerates PD progression. Disrupting this vicious cycle is critical for therapeutic intervention.
Iron overload can activate glial cells, leading to the release neuroinflammatory factors such as IL-1β, TNF-α, and IL-6, affecting neurons (Liu et al. 2022). The release of inflammatory factors promotes the transcription and accumulation of α-syn (Angelova and Brown 2018; Li et al. 2021). In rat neuron-microglia-astroglia cultures, microglia enhance iron-induced selective and progressive toxicity in dopamine neuronsl (Zhang et al. 2014). More importantly, Guo et al. found that iron is specifically deposited in microglia in the SN region, but not in dopaminergic neurons in the PD model of Macaca fascicularis induced by α-syn preformed fibrils (Guo et al. 2021). This suggests that iron deposition in microglia is an early response to induce inflammation in dopamine neurons. Additionally, microglia are the most prone to ferroptosis (Ryan et al. 2023). However, it remains unclear whether microglial activation is the primary cause of PD (Oleg and Weiner 2018; Hickman et al. 2018). In a mouse model of lentivirus-mediated accumulation of α-syn in microglia, dopaminergic neuronal damage occurred without endogenous α-syn accumulation (Bido et al. 2021). Since microglia are among the first cells to show iron deposition, addressing iron overload in microglia may be key to preventing PD.
Iron is an important regulator of mitochondrial function, but excess iron can lead to mitochondrial dysfunction. Fe2+ catalyses the generation of ROS via the Fenton reaction, leading to lipid peroxidation of mitochondrial membranes, which disrupts mitochondrial membrane integrity and results in mitochondrial dysfunction (Rochette et al. 2022). Iron accumulation interferes with the mitochondrial electron transport chain (ETC), leading to reduced ATP production and impaired energy metabolism, further exacerbating cellular damage (Guo et al. 2022). Lysosomes occupy a central position in iron homeostasis in addition to its role in degradation and recycling. Iron overload also affects lysosomal function and impairs mitophagy, which in turn exacerbates mitochondrial dysfunction. Fe3+ stored in lysosomes is reduced to Fe2+ in acidic environments, leading to lysosomal oxidative damage, releasing Fe2+ and proteases into the cytoplasm and triggering ferroptosis (Rizzollo et al. 2021; Kurz et al. 2011). In addition, lysosomal membranes are rich in polyunsaturated fatty acids, making them susceptible to Fe2+-mediated lipid peroxidation, which can directly disrupt membrane integrity. In summary, iron accumulation impairs both mitochondrial and lysosomal function, contributing to α-syn accumulation and inducing PD.
Currently, no approved drug effectively inhibits or degrades α-syn aggregation to treat PD. Due to α-syn’s intrinsically disordered structure, it remains challenging to target it directly with pharmacological agents (Bisi et al. 2021). Improving iron metabolism and limiting iron accumulation are promising alternative approaches to inhibit α-syn aggregation indirectly. Several studies have reported that iron chelators demonstrated positive results in PD models, by reducing α-syn aggregation and limiting iron induced neuronal damage (Jansen van Rensburg et al. 2021). For example, Huang et al. demonstrated that administration of deferoxamine mesylate ameliorated MPTP-induced Parkinsonism in mice (Mannan Thodukayil et al. 2019). A summary of iron chelators used in PD research is provided in Table 1.
Iron chelators, particularly when targeting both α-syn aggregation and ferroptosis, demonstrate a promising dual mechanism for PD treatment. Phase II clinical trials of deferiprone (DFP) show potential in improving motor symptoms, however, current evidence remains insufficient to support its routine use in patients with PD (Negida et al. 2024; Martin-Bastida et al. 2017). In addition, a significant limitation of existing iron chelators is their restricted ability to penetrate the blood–brain barrier, which limits their therapeutic efficacy in clinical PD applications. Therefore, developing next-generation iron chelators with enhanced CNS bioavailability or combining chelators with other therapeutic agents represents a crucial research direction for future treatments of PD.
Conclusion
This review has highlighted the role of iron in regulating α-syn aggregation through alternative pathological pathways.Regulation of iron metabolism not only inhibits α-syn aggregation but also effectively suppresses neuronal ferroptosis, both of which are central to PD pathology. Given the challenging nature of α-syn’s structure, designing drugs that promote its degradation has proven difficult. Targeting iron metabolism may offer a viable alternative and hold promise as an effective treatment for PD. Future research into iron-targeted therapies could pave the way for more successful treatment strategies for PD.
Data availability
No datasets were generated or analysed during the current study.
Abbreviations
- PD:
-
Parkinson's disease
- α-syn:
-
α-synuclein
- SN:
-
Substantial nigra
- PTMs:
-
Post-translational modifications
- AKT:
-
Protein kinase B
- mTORC1:
-
Mechanistic target of rapamycin complex 1
- TFEB:
-
Transcription factor EB
- IGF2:
-
Insulin-like growth factor 2
- ZFP27:
-
Transcription factor zinc finger protein 27
- IRP:
-
Iron regulatory protein
- IRE:
-
Iron response element
- ROS:
-
Reactive oxygen species
- HSPGs:
-
Heparan sulfate proteoglycans
- CK2:
-
Casein kinase 2
- PLK2:
-
Polo-like kinase 2
- DMT1:
-
Divalent metal-ion transporter 1
- MAPK:
-
Mitogen-activated protein kinase
- DFP:
-
Deferiprone
- TfR:
-
Transferrin receptor
- DMT1:
-
Divalent metal transporter 1
- FAC:
-
Ferric ammonium citrate
- TEM:
-
Transmission electron microscopy
- cryo-EM:
-
Cryo-electron microscopy
- ThT:
-
Thioflavin
- Glu:
-
Glutamate
- Asp:
-
Aspartate
- NMR:
-
Nuclear magnetic resonance
- DFO:
-
Deferoxamine
- RIPK1:
-
Receptor-interacting protein kinase 1
- RIPK3:
-
Receptor-interacting protein kinase 3
- MLKL:
-
Mixed lineage kinase domain-like
References
Abeyawardhane DL, et al. Iron redox chemistry promotes antiparallel oligomerization of alpha-synuclein. J Am Chem Soc. 2018;140:5028–32.
Alushaj E, et al. Increased iron in the substantia nigra pars compacta identifies patients with early Parkinson’s disease: a 3T and 7T MRI study. Neuroimage Clin. 2024;41: 103577.
Angelova DM, Brown DR. Model senescent microglia induce disease related changes in alpha-synuclein expression and activity. Biomolecules. 2018;8:67.
Atarod D, et al. Bivalent metal ions induce formation of alpha-synuclein fibril polymorphs with different cytotoxicities. Sci Rep. 2022;12:11898.
Aubignat M, Tir M. Continuous Subcutaneous foslevodopa-foscarbidopa in Parkinson’s disease: a mini-review of current scope and future outlook. Mov Disord Clin Pract. 2024;11:1188–94.
Bar-Am O, et al. Neuroprotective and neurorestorative activities of a novel iron chelator-brain selective monoamine oxidase-A/monoamine oxidase-B inhibitor in animal models of Parkinson’s disease and aging. Neurobiol Aging. 2015;36:1529–42.
Becker G, Seufert J, Bogdahn U, Reichmann H, Reiners K. Degeneration of substantia nigra in chronic Parkinson’s disease visualized by transcranial color-coded real-time sonography. Neurology. 1995;45:182–4.
Berg D, et al. Enlarged substantia nigra hyperechogenicity and risk for Parkinson disease: a 37-month 3-center study of 1847 older persons. Arch Neurol. 2011;68:932–7.
Bharathi ISS, Rao KS. Copper- and iron-induced differential fibril formation in alpha-synuclein: TEM study. Neurosci Lett. 2007;424:78–82.
Bi M, Du X, Jiao Q, Liu Z, Jiang H. alpha-synuclein regulates iron homeostasis via preventing Parkin-Mediated DMT1 ubiquitylation in Parkinson’s disease models. ACS Chem Neurosci. 2020;11:1682–91.
Bido S, et al. Microglia-specific overexpression of alpha-synuclein leads to severe dopaminergic neurodegeneration by phagocytic exhaustion and oxidative toxicity. Nat Commun. 2021;12:6237.
Binolfi A, et al. Interaction of alpha-synuclein with divalent metal ions reveals key differences: a link between structure, binding specificity and fibrillation enhancement. J Am Chem Soc. 2006;128:9893–901.
Bisaglia M, Bubacco L. Copper ions and Parkinson’s disease: why is homeostasis so relevant? Biomolecules. 2020;10:195.
Bisi N, et al. alpha-synuclein: an all-inclusive trip around its structure, influencing factors and applied techniques. Front Chem. 2021;9: 666585.
Calabresi P, et al. Alpha-synuclein in Parkinson’s disease and other synucleinopathies: from overt neurodegeneration back to early synaptic dysfunction. Cell Death Dis. 2023;14:176.
Chen LL, Huang YJ, Cui JT, Song N, Xie J. Iron dysregulation in Parkinson’s disease: focused on the autophagy-lysosome pathway. ACS Chem Neurosci. 2019;10:863–71.
Ci YZ, et al. Iron overload induced by IRP2 gene knockout aggravates symptoms of Parkinson’s disease. Neurochem Int. 2020;134: 104657.
Danzer KM, et al. Exosomal cell-to-cell transmission of alpha synuclein oligomers. Mol Neurodegener. 2012;7:42.
David S, Jhelum P, Ryan F, Jeong SY, Kroner A. Dysregulation of iron homeostasis in the central nervous system and the role of ferroptosis in neurodegenerative disorders. Antioxid Redox Signal. 2022;37:150–70.
Davies P, Moualla D, Brown DR. Alpha-synuclein is a cellular ferrireductase. PLoS ONE. 2011;6: e15814.
Deas E, et al. Alpha-synuclein oligomers interact with metal ions to induce oxidative stress and neuronal death in Parkinson’s disease. Antioxid Redox Signal. 2016;24:376–91.
Devos D, et al. Trial of deferiprone in Parkinson’s disease. N Engl J Med. 2022;387:2045–55.
Emamzadeh FN. Alpha-synuclein structure, functions, and interactions. J Res Med Sci. 2016;21:29.
Febbraro F, Giorgi M, Caldarola S, Loreni F, Romero-Ramos MJN. α-Synuclein expression is modulated at the translational level by iron. NeuroReport. 2012;23:576–80.
Fernandez B, et al. Iron overload causes endolysosomal deficits modulated by NAADP-regulated 2-pore channels and RAB7A. Autophagy. 2016;12:1487–506.
Friedlich AL, Tanzi RE, Rogers JT. The 5’-untranslated region of Parkinson’s disease alpha-synuclein messengerRNA contains a predicted iron responsive element. Mol Psychiatry. 2007;12:222–3.
Gallardo J, Escalona-Noguero C, Sot B. Role of alpha-synuclein regions in nucleation and elongation of amyloid fiber assembly. ACS Chem Neurosci. 2020;11:872–9.
Gao V, Briano JA, Komer LE, Burre J. Functional and pathological effects of alpha-synuclein on synaptic SNARE complexes. J Mol Biol. 2023;435: 167714.
Ghanem SS, et al. alpha-Synuclein phosphorylation at serine 129 occurs after initial protein deposition and inhibits seeded fibril formation and toxicity. Proc Natl Acad Sci USA. 2022;119: e2109617119.
Guhathakurta S, Bok E, Evangelista BA, Kim YS. Deregulation of alpha-synuclein in Parkinson’s disease: insight from epigenetic structure and transcriptional regulation of SNCA. Prog Neurobiol. 2017;154:21–36.
Guo JJ, et al. Intranasal administration of alpha-synuclein preformed fibrils triggers microglial iron deposition in the substantia nigra of Macaca fascicularis. Cell Death Dis. 2021;12:81.
Guo J, et al. Mitochondria as multifaceted regulators of ferroptosis. Life Metab. 2022;1:134–48.
Ham S, Kim JH, Kim H, Shin JY, Lee Y. ATF4-activated parkin induction contributes to deferasirox-mediated cytoprotection in Parkinson’s disease. Toxicol Res. 2023;39:191–9.
Hare DJ, Double KL. Iron and dopamine: a toxic couple. Brain. 2016;139:1026–35.
Hare DJ, et al. Is early-life iron exposure critical in neurodegeneration? Nat Rev Neurol. 2015;11:536–44.
Haskova P, et al. Examination of diverse iron-chelating agents for the protection of differentiated PC12 cells against oxidative injury induced by 6-hydroxydopamine and dopamine. Sci Rep. 2022;12:9765.
Hickman S, Izzy S, Sen P, Morsett L, El Khoury J. Microglia in neurodegeneration. Nat Neurosci. 2018;21:1359–69.
Hideshima M, et al. Two-step screening method to identify alpha-synuclein aggregation inhibitors for Parkinson’s disease. Sci Rep. 2022;12:351.
Holbein BE, Lehmann C. Dysregulated iron homeostasis as common disease etiology and promising therapeutic target. Antioxidants. 2023;12:671.
Iannielli A, et al. Pharmacological inhibition of necroptosis protects from dopaminergic neuronal cell death in Parkinson’s disease models. Cell Rep. 2018;22:2066–79.
Jacquemyn J, Ralhan I, Ioannou MS. Driving factors of neuronal ferroptosis. Trends Cell Biol. 2024;34:535–46.
Jansen van Rensburg Z, Abrahams S, Bardien S, Kenyon C. Toxic feedback loop involving iron, reactive oxygen species, alpha-synuclein and neuromelanin in Parkinson’s disease and intervention with turmeric. Mol Neurobiol. 2021;58:5920–36.
Jenner P, Dexter DT, Sian J, Schapira AH, Marsden CD. Oxidative stress as a cause of nigral cell death in Parkinson’s disease and incidental Lewy body disease. The Royal Kings and Queens Parkinson’s Disease Research Group. Ann Neurol. 1992;32:S82-87.
Kaindlstorfer C, et al. The relevance of iron in the pathogenesis of multiple system atrophy: a viewpoint. J Alzheimers Dis. 2018;61:1253–73.
Kalivendi SV, et al. 1-Methyl-4-phenylpyridinium (MPP+)-induced apoptosis and mitochondrial oxidant generation: role of transferrin-receptor-dependent iron and hydrogen peroxide. Biochem J. 2003;371:151–64.
Kawahata I, Finkelstein DI, Fukunaga K. Pathogenic impact of alpha-synuclein phosphorylation and its kinases in alpha-synucleinopathies. Int J Mol Sci. 2022;23:6216.
Kontaxi C, Edwards RH. Synuclein phosphorylation: pathogenic or physiologic? NPJ Parkinsons Dis. 2023;9:47.
Kostka M, et al. Single particle characterization of iron-induced pore-forming alpha-synuclein oligomers. J Biol Chem. 2008;283:10992–1003.
Kumfu S, et al. Ferroptosis inhibitor improves cardiac function more effectively than inhibitors of apoptosis and necroptosis through cardiac mitochondrial protection in rats with iron-overloaded cardiomyopathy. Toxicol Appl Pharmacol. 2023;479: 116727.
Kurz T, Eaton JW, Brunk UT. The role of lysosomes in iron metabolism and recycling. Int J Biochem Cell Biol. 2011;43:1686–97.
Lane DJ, et al. Cellular iron uptake, trafficking and metabolism: key molecules and mechanisms and their roles in disease. Biochim Biophys Acta. 2015;1853:1130–44.
Le W. Role of iron in UPS impairment model of Parkinson’s disease. Parkinsonism Relat Disord. 2014;20(Suppl 1):S158-161.
Leem YH, Kim DY, Park JE, Kim HS. Necrosulfonamide exerts neuroprotective effect by inhibiting necroptosis, neuroinflammation, and alpha-synuclein oligomerization in a subacute MPTP mouse model of Parkinson’s disease. Sci Rep. 2023;13:8783.
Leem YH, Park JS, Park JE, Kim DY, Kim HS. Creatine supplementation with exercise reduces alpha-synuclein oligomerization and necroptosis in Parkinson’s disease mouse model. J Nutr Biochem. 2024;126: 109586.
Lei L, et al. Targeting the labile iron pool with engineered DFO nanosheets to inhibit ferroptosis for Parkinson’s disease therapy. Adv Mater. 2024;36: e2409329.
Leitao ADG, et al. Selectivity of Lewy body protein interactions along the aggregation pathway of alpha-synuclein. Commun Biol. 2021;4:1124.
Levin J, et al. Generation of ferric iron links oxidative stress to alpha-synuclein oligomer formation. J Parkinsons Dis. 2011;1:205–16.
Li WJ, Jiang H, Song N, Xie JX. Dose- and time-dependent alpha-synuclein aggregation induced by ferric iron in SK-N-SH cells. Neurosci Bull. 2010;26:205–10.
Li W, Jiang H, Song N, Xie J. Oxidative stress partially contributes to iron-induced alpha-synuclein aggregation in SK-N-SH cells. Neurotox Res. 2011;19:435–42.
Li Y, et al. Copper and iron ions accelerate the prion-like propagation of alpha-synuclein: a vicious cycle in Parkinson’s disease. Int J Biol Macromol. 2020;163:562–73.
Li Y, et al. Targeting microglial alpha-synuclein/TLRs/NF-kappaB/NLRP3 inflammasome axis in Parkinson’s disease. Front Immunol. 2021;12: 719807.
Li Y, Yu Y, Ma G. Modulation effects of Fe(3+), Zn(2+), and Cu(2+) ions on the amyloid fibrillation of alpha-synuclein: insights from a FTIR investigation. Molecules. 2022;27:8383.
Li X, et al. Exploring heat shock proteins as therapeutic targets for Parkinson’s disease. Biochem Pharmacol. 2024;230: 116633.
Liu S, Gao X, Zhou S. New target for prevention and treatment of neuroinflammation: microglia iron accumulation and ferroptosis. ASN Neuro. 2022;14:17590914221133236.
Mannan Thodukayil N, et al. Desferrioxamine and dextromethorphan combination exhibited synergistic effect and reversed the catalepsy behaviour in 6-hydroxydopamine hydroydopamine administered rats through regulating brain glutamate levels. J Pharm Pharmacol. 2019;71:1271–81.
Martin WR, Wieler M, Gee M. Midbrain iron content in early Parkinson disease: a potential biomarker of disease status. Neurology. 2008;70:1411–7.
Martin-Bastida A, et al. Brain iron chelation by deferiprone in a phase 2 randomised double-blinded placebo controlled clinical trial in Parkinson’s disease. Sci Rep. 2017;7:1398.
Mochizuki H, Choong CJ, Baba K. Parkinson’s disease and iron. J Neural Transm (Vienna). 2020;127:181–7.
Moons R, et al. Metal ions shape alpha-synuclein. Sci Rep. 2020;10:16293.
Musteikyte G, et al. Interactions of alpha-synuclein oligomers with lipid membranes. Biochim Biophys Acta Biomembr. 2021;1863: 183536.
Ndayisaba A, Kaindlstorfer C, Wenning GK. Iron in neurodegeneration - cause or consequence? Front Neurosci. 2019;13:180.
Negida A, et al. Efficacy of the iron-chelating agent, deferiprone, in patients with Parkinson’s disease: a systematic review and meta-analysis. CNS Neurosci Ther. 2024;30: e14607.
Oleg B, Weiner HL. Microglial signatures and their role in health and disease. Nat Rev Neurosci. 2018;19:1.
Onate M, et al. The necroptosis machinery mediates axonal degeneration in a model of Parkinson disease. Cell Death Differ. 2020;27:1169–85.
Ostrerova-Golts N, et al. The A53T alpha-synuclein mutation increases iron-dependent aggregation and toxicity. J Neurosci. 2000;20:6048–54.
Peng Y, Wang C, Xu HH, Liu YN, Zhou F. Binding of alpha-synuclein with Fe(III) and with Fe(II) and biological implications of the resultant complexes. J Inorg Biochem. 2010;104:365–70.
Perfeito R, Lazaro DF, Outeiro TF, Rego AC. Linking alpha-synuclein phosphorylation to reactive oxygen species formation and mitochondrial dysfunction in SH-SY5Y cells. Mol Cell Neurosci. 2014;62:51–9.
Picca A, et al. Mitochondrial dysfunction, protein misfolding and neuroinflammation in Parkinson’s disease: roads to biomarker discovery. Biomolecules. 2021;11:1508.
Pyatigorskaya N, et al. Iron imaging as a diagnostic tool for Parkinson’s disease: a systematic review and meta-analysis. Front Neurol. 2020;11:366.
Raj K, Kaur P, Gupta GD, Singh S. Metals associated neurodegeneration in Parkinson’s disease: insight to physiological, pathological mechanisms and management. Neurosci Lett. 2021;753: 135873.
Ramalingam N, Haass C, Dettmer U. Physiological roles of alpha-synuclein serine-129 phosphorylation - not an oxymoron. Trends Neurosci. 2024;47:480–90.
Rizzollo F, More S, Vangheluwe P, Agostinis P. The lysosome as a master regulator of iron metabolism. Trends Biochem Sci. 2021;46:960–75.
Rochette L, et al. Lipid peroxidation and iron metabolism: two corner stones in the homeostasis control of ferroptosis. Int J Mol Sci. 2022;24:449.
Ru Q, et al. Iron homeostasis and ferroptosis in human diseases: mechanisms and therapeutic prospects. Signal Transduct Target Ther. 2024;9:271.
Ryan SK, et al. Microglia ferroptosis is regulated by SEC24B and contributes to neurodegeneration. Nat Neurosci. 2023;26:12–26.
Sarchione A, Marchand A, Taymans JM, Chartier-Harlin MC. Alpha-synuclein and lipids: the elephant in the room? Cells. 2021;10:2452.
Sato H, Kato T, Arawaka S. The role of Ser129 phosphorylation of alpha-synuclein in neurodegeneration of Parkinson’s disease: a review of in vivo models. Rev Neurosci. 2013;24:115–23.
Shen QQ, et al. Cell senescence induced by toxic interaction between alpha-synuclein and iron precedes nigral dopaminergic neuron loss in a mouse model of Parkinson’s disease. Acta Pharmacol Sin. 2024;45:268–81.
Sian-Hulsmann J, Riederer P. The role of alpha-synuclein as ferrireductase in neurodegeneration associated with Parkinson’s disease. J Neural Transm. 2020;127:749–54.
So RWL, Watts JC. alpha-synuclein conformational strains as drivers of phenotypic heterogeneity in neurodegenerative diseases. J Mol Biol. 2023;435: 168011.
Sun Y, Pham AN, Hider RC, Zheng H, Waite TD. Effectiveness of the iron chelator CN128 in mitigating the formation of dopamine oxidation products associated with the progression of Parkinson’s disease. ACS Chem Neurosci. 2020;11:3646–57.
Tang D, Kroemer G. Ferroptosis. Curr Biol. 2020;30:R1292–7.
Tofaris GK. Initiation and progression of alpha-synuclein pathology in Parkinson’s disease. Cell Mol Life Sci. 2022;79:210.
Tong Y, Zhang P, Yang X, Liu X, Zhang J, Grudniewska M, Jung I, Abegg D, Liu J, Childs-Disney JL, Gibaut QM. Decreasing the intrinsically disordered protein α-synuclein levels by targeting its structured mRNA with a ribonuclease-targeting chimera. Proc Natl Acad Sci. 2024;121(2): e2306682120.
Tsigelny IF, et al. Role of alpha-synuclein penetration into the membrane in the mechanisms of oligomer pore formation. FEBS J. 2012;279:1000–13.
Uversky VN, Li J, Fink AL. Metal-triggered structural transformations, aggregation, and fibrillation of human alpha-synuclein. A possible molecular NK between Parkinson’s disease and heavy metal exposure. J Biol Chem. 2001;276:44284–96.
van Schijndel MA, et al. Medical Psychiatric Units in the Netherlands: an investigation into distribution and quality. Ned Tijdschr Geneeskd. 2017;161:D890.
Wan W, et al. Iron deposition leads to neuronal alpha-synuclein pathology by inducing autophagy dysfunction. Front Neurol. 2017;8:1.
Wang R, et al. Iron-induced oxidative stress contributes to alpha-synuclein phosphorylation and up-regulation via polo-like kinase 2 and casein kinase 2. Neurochem Int. 2019;125:127–35.
Wang Y, et al. Iron-inhibited autophagy via transcription factor ZFP27 in Parkinson’s disease. J Cell Mol Med. 2023;27:3614–27.
Wang S, Guo Q, Zhou L, Xia X. Ferroptosis: a double-edged sword. Cell Death Discov. 2024;10:265.
Workman DG, et al. Protection from neurodegeneration in the 6-hydroxydopamine (6-OHDA) model of Parkinson’s with novel 1-hydroxypyridin-2-one metal chelators. Metallomics. 2015;7:867–76.
Wu JR, et al. Necrostatin-1 protection of dopaminergic neurons. Neural Regen Res. 2015;10:1120–4.
Wu KC, Liou HH, Kao YH, Lee CY, Lin CJ. The critical role of Nramp1 in degrading alpha-synuclein oligomers in microglia under iron overload condition. Neurobiol Dis. 2017;104:61–72.
Xiao Y, et al. Iron promotes alpha-synuclein aggregation and transmission by inhibiting TFEB-mediated autophagosome-lysosome fusion. J Neurochem. 2018;145:34–50.
Xu Y, Deng Y, Qing H. The phosphorylation of alpha-synuclein: development and implication for the mechanism and therapy of the Parkinson’s disease. J Neurochem. 2015;135:4–18.
Xu T, et al. Disease burden of Parkinson’s disease in China and its provinces from 1990 to 2021: findings from the global burden of disease study 2021. Lancet Reg Health West Pac. 2024;46: 101078.
Xue J, Wang HL, Xiao G. Transferrin1 modulates rotenone-induced Parkinson’s disease through affecting iron homeostasis in Drosophila melanogaster. Biochem Biophys Res Commun. 2020;531:305–11.
Yan F, et al. Role of iron-related oxidative stress and mitochondrial dysfunction in cardiovascular diseases. Oxid Med Cell Longev. 2022;2022:5124553.
Yan S, et al. Spatiotemporal patterns of brain iron-oxygen metabolism in patients with Parkinson’s disease. Eur Radiol. 2024;34:3074–83.
Yoo H, et al. Role of post-translational modifications on the alpha-synuclein aggregation-related pathogenesis of Parkinson’s disease. BMB Rep. 2022;55:323–35.
You LH, et al. Brain iron accumulation exacerbates the pathogenesis of MPTP-induced Parkinson’s disease. Neuroscience. 2015;284:234–46.
Youdim MB, Stephenson G, Ben SD. Ironing iron out in Parkinson’s disease and other neurodegenerative diseases with iron chelators: a lesson from 6-hydroxydopamine and iron chelators, desferal and VK-28. Ann N Y Acad Sci. 2004;1012:306–25.
Zecca L, et al. The role of iron and copper molecules in the neuronal vulnerability of locus coeruleus and substantia nigra during aging. Proc Natl Acad Sci USA. 2004;101:9843–8.
Zeng X, et al. Benefits of iron chelators in the treatment of Parkinson’s disease. Neurochem Res. 2021;46:1239–51.
Zhang W, et al. Role and mechanism of microglial activation in iron-induced selective and progressive dopaminergic neurodegeneration. Mol Neurobiol. 2014;49:1153–65.
Zhang QS, et al. Reassessment of subacute MPTP-treated mice as animal model of Parkinson’s disease. Acta Pharmacol Sin. 2017;38:1317–28.
Zhang P, et al. Ferroptosis was more initial in cell death caused by iron overload and its underlying mechanism in Parkinson’s disease. Free Radic Biol Med. 2020;152:227–34.
Zhang S, et al. Mechanistic basis for receptor-mediated pathological alpha-synuclein fibril cell-to-cell transmission in Parkinson’s disease. Proc Natl Acad Sci U S A. 2021;118: e2011196118.
Zhang C, et al. C-terminal truncation modulates alpha-Synuclein’s cytotoxicity and aggregation by promoting the interactions with membrane and chaperone. Commun Biol. 2022;5:798.
Zhao Q, et al. Structural insights of Fe(3+) induced alpha-synuclein fibrillation in Parkinson’s disease. J Mol Biol. 2023;435: 167680.
Zhou ZD, Tan EK. Iron regulatory protein (IRP)-iron responsive element (IRE) signaling pathway in human neurodegenerative diseases. Mol Neurodegener. 2017;12:75.
Zhu ZJ, Wu KC, Yung WH, Qian ZM, Ke Y. Differential interaction between iron and mutant alpha-synuclein causes distinctive Parkinsonian phenotypes in Drosophila. Biochim Biophys Acta. 2016;1862:518–25.
Acknowledgements
The authors acknowledge the use of Biorender that is used to create the figures.
Funding
This study was financially supported by grants from the Sichuan Science and Technology Program (No. 2024 NSFSC0610). This study was financially supported by grants from The Youth Innovation Project of Sichuan Provincial Medical Research (202407110270).
Author information
Authors and Affiliations
Contributions
Xiang Li and Wenjun Wang were responsible for the study concept and design. Yan Chen, Xixi Luo, and Yukun Yin drafted the manuscript and produced the figures. Elizabeth Rosalind Thomas, Kezhi Liu and Xiang Li provided critical revision of the manuscript for important intellectual content. All the authors read and approved the final version.
Corresponding authors
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare no competing interests.
Additional information
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
About this article
Cite this article
Chen, Y., Luo, X., Yin, Y. et al. The interplay of iron, oxidative stress, and α-synuclein in Parkinson’s disease progression. Mol Med 31, 154 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s10020-025-01208-3
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s10020-025-01208-3