Next Article in Journal
Interleukin-11/IL-11 Receptor Promotes Glioblastoma Cell Proliferation, Epithelial–Mesenchymal Transition, and Invasion
Next Article in Special Issue
Akkermansia muciniphila Is Beneficial to a Mouse Model of Parkinson’s Disease, via Alleviated Neuroinflammation and Promoted Neurogenesis, with Involvement of SCFAs
Previous Article in Journal
Balancing Act: Acute and Contextual Vestibular Sensations of Cranial Electrotherapy Stimulation Using Survey and Sensor Outcomes in a Non-Clinical Sample
Previous Article in Special Issue
The Cannabigerol Derivative VCE-003.2 Exerts Therapeutic Effects in 6-Hydroxydopamine-Lesioned Mice: Comparison with The Classic Dopaminergic Replacement Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Calcium and Iron Homeostasis in Parkinson’s Disease

by
Ji Wang
1,2,†,
Jindong Zhao
2,†,
Kunying Zhao
2,
Shangpeng Wu
2,
Xinglong Chen
1,* and
Weiyan Hu
2,*
1
School of Chinese Materia Medica & Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming 650500, China
2
School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Brain Sci. 2024, 14(1), 88; https://doi.org/10.3390/brainsci14010088
Submission received: 11 December 2023 / Revised: 4 January 2024 / Accepted: 9 January 2024 / Published: 17 January 2024
(This article belongs to the Special Issue Molecular Mechanism and Pathology of Parkinson's Disease)

Abstract

:
Calcium and iron are essential elements that regulate many important processes of eukaryotic cells. Failure to maintain homeostasis of calcium and iron causes cell dysfunction or even death. PD (Parkinson’s disease) is the second most common neurological disorder in humans, for which there are currently no viable treatment options or effective strategies to cure and delay progression. Pathological hallmarks of PD, such as dopaminergic neuronal death and intracellular α-synuclein deposition, are closely involved in perturbations of iron and calcium homeostasis and accumulation. Here, we summarize the mechanisms by which Ca2+ signaling influences or promotes PD progression and the main mechanisms involved in ferroptosis in Parkinson’s disease. Understanding the mechanisms by which calcium and iron imbalances contribute to the progression of this disease is critical to developing effective treatments to combat this devastating neurological disorder.

1. Introduction

PD (Parkinson’s disease), the second most common multifactorial progressive neurodegenerative disease after Alzheimer’s disease, attracts widespread attention due to the rising prevalence as the world population ages and lives longer [1,2].
Though the cause of PD is currently unknown, it is now generally accepted that aging is the most influential risk factor and various environmental and genetic factors have also been shown to influence Parkinson’s risk [3,4,5]. However, age and genetic and environmental factors can only explain a limited part of PD [6]. The causes of most cases of PD can be complex and could be the result of the interplay of age, genetic susceptibility, and environmental factors.
Iron and calcium are found in low concentrations in the body, but they play an essential and irreplaceable role. Excessive metal accumulation in the central nervous system may induce oxidative stress, disrupt mitochondrial function, and damage different types of proteins and receptors [7,8]. Various studies have shown that changes in metal ion homeostasis may lead to damage to the CNS (central nervous system) with neurodegeneration, disability, and neuroinflammation, contributing to the development of several neurodegenerative diseases [9]. Growing evidence indicates that calcium overload and iron deposition play a vital role in the pathogenesis of Parkinson’s disease [10,11,12].
In this paper, we focus on the pathophysiological role of calcium and iron ions and their homeostatic imbalance in pathogenesis, exploring the therapeutic potential of regulating calcium and iron homeostasis in Parkinson’s disease.

2. Calcium and PD

Ca2+ is the second messenger of all living things [13]. When the cells are in a resting state, the calcium ion concentration in the cytoplasm is 100 nmol·L−1, the calcium ion concentration in the endoplasmic reticulum is about 0.5–1 mmol·L−1, and the extracellular calcium ion concentration is 1–2 mmol·L−1. The concentration of Ca2+ ions in the cytoplasm is 20,000 times lower than the concentration in the extracellular space [14]. This gradient allows cells to use Ca2+ as an effective intracellular signal to respond to and adapt to rapidly changing extracellular and intracellular environments. Cells can alter various physiological functions by activating or inhibiting Ca2+-dependent signal transduction pathways. In neurons, Ca2+ motion can occur across the plasma membrane, either through electrical activity or through agonists like the DHP (dihydropyridine channel) derivative BAY K8644, which increases calcium influx through L-type calcium channels [15]. Ca2+ signaling affects all aspects of neuronal cell biology, so calcium homeostasis must be tightly controlled [16,17].
Intracellular Ca2+ levels, Ca2+ microdomains, Ca2+ buffering, and Ca2+ entry patterns are tightly controlled in SN (Substantia nigra) DA(dopaminergic) neurons because Ca2+ regulates multiple cellular functions such as excitability, neurotransmitter release, ATP production [18], apoptosis, and the general regulation of enzymes and gene expression [19]. Defects in calcium processing play an essential role in aging and neurodegeneration, with neurons relying on calcium ions to regulate key processes such as neurogenesis, neurotransmission, synaptic plasticity, and gene transcription [16]. In the investigation of individuals with PD and healthy controls, it was observed that PD patients exhibit a higher average calcium level compared to their healthy counterparts. This elevated calcium level has been associated with an increased susceptibility to Parkinson’s disease due to its role in stimulating excessive dopamine biosynthesis, thereby leading to dopaminergic neurotoxicity [20].
Ca2+ is regulated by various pathways [21]. α-synaptic nucleoprotein aggregation, an important pathologic feature of Parkinson’s disease, leads to disruption of calcium homeostasis [17]. The Ca2+ of SN DA neurons can pass through channels in the plasma membrane (voltage-gated calcium channels, VGCC) and transporters (glutamate receptors, NMDA (N-methyl-D-aspartic acid receptor)-R) or AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid receptor)-R) from extracellular to intracellular [22]. In the cell, Ca2+ is also released from mitochondria (mitochondrial calcium transfer protein, MCU) and ER (Endoplasmic reticulum) through RyR (Ryanodine receptor) and IP3 (Inositol 1,4,5-trisphosphate) receptors), or through storage-operated calcium channels such as STIM (Stromal interaction molecule)/Orai (Calcium release-activated calcium modulator) and transient receptors potential channel complexes [23,24] (Figure 1).
Ten types of VGCCs are distinguished based on their distinct pharmacological and functional properties, each containing a different pore-forming α1 subunit. The subcellular expression, activity, gating behavior, and pharmacological properties of VGCCs are tuned by extensive alternative splicing and their association with modulatory accessory subunits (β1–β4, α2δ1–α2δ4) [25]. Based on their α1 subunit sequence homology as well as their functional and pharmacological properties, three VGCC families have been defined: Cav1, Cav2, and Cav3. The Cav1 family is composed of four LTCC members (Cav1.1–Cav1.4), which are characterized by their sensitivity to low nanomolar concentrations of DHPs [26]. Cav1.2 and Cav1.3 in the L-type voltage-gated Ca2+ channels are associated with PD [27] and Cav1.2 is prevalent in juvenile SNc (Substantia Nigra) DA neurons, but in senescent SNc DA neurons, Cav1.3 is preferentially used for Ca2+ inflow, allowing Ca2+ to enter through an oscillatory pathway that contributes to the membrane potential threshold, which is the basis of autonomous pacing [28]. Autonomic pacing is a distinctive active state displayed by dopaminergic neurons, characterized by the presence of a spontaneous, slowly depolarizing membrane current that sustains their basal activity level. This mechanism ensures an uninterrupted supply of dopamine to interconnected brain regions, which is crucial for maintaining baseline dopamine levels in the striatum. Unlike Cav1.2, the working range of Cav1.3 does not allow for complete closure of Cav1.3 channels during pacing, which results in elevated intracellular Ca2+ levels, and continuous Ca2+ influx is essential for regulating physiologic DA release from SNc DA neurons [29]. It is necessary, however, that the chronic and excessive presence of Ca2+ may synergize with several risk factors (i.e., aging, mitochondrial toxins, mutations) and produce metabolic stress and mitochondrial damage. The increased expression of Cav1.3 in the cerebral cortex of patients with early Parkinson’s disease occurs before pathological changes, suggesting that calcium homeostasis disorders are not only a compensatory consequence of neurodegenerative processes [30], but also an early feature of Parkinson’s disease. DHP antihypertensive drugs appear to be attractive agents for neuroprotective and preventive treatment of PD [31,32]. Epidemiological studies have shown that administration of DHP-type Ca2+ channel blockers reduces the risk of PD in late life by 20 to 30% [33,34]. In contrast, members of the Cav2 family (Cav2.1–Cav2.3), mediating P/Q-, N-, and R-type voltage-gated Ca2+ currents, are located presynaptically and are required for fast neurotransmitter release [35]. Low voltage-activated Cav3 channels (Cav3.1–Cav3.3) compose the family of T-type Ca2+ channels (TTCCs). TTCCs activate and inactivate at more negative potentials than Cav1 and Cav2 channels. This negative operation range allows them to be active at subthreshold voltages and provides them with a prominent role in the control of neuronal firing patterns [36].
The activity of dopaminergic neurons is regulated by Ca2+ carriers and influenced by Ca2+ homeostasis [37]. About 80% of calcium is stored in organelles (such as mitochondria, sarcoplasmic reticulum, endoplasmic reticulum, etc.). Mitochondria and the endoplasmic reticulum are the main intracellular organelles that regulate calcium, but evidence suggests that other organelles, such as lysosomes and the Golgi apparatus, also act as important intracellular stores of Ca2+ [38].
Among them, ER (the endoplasmic reticulum) is considered to be the main reservoir of intracellular Ca2+, so the calcium signaling conveyed by ER is crucial in cell conduction. For example, SOCE (store-operated calcium entry) is one of the crucial channels that mediate extracellular Ca2+ into the cell, which is due to a large reduction in calcium ions in the lumen of the endoplasmic reticulum, which, in turn, triggers extracellular Ca2+ to flow into the cytoplasm and then supplements Ca2+ back to ER through the SERCA (ATPase sarco-ER Ca2+-ATPase) [39]. The occurrence of this process is mediated by the STIM1 protein in the endoplasmic reticulum matrix and the Orai calcium channel on the plasma membrane. Genetic mutations in Orai1 or STIM1 often lead to SOCE channel defects, which are closely related to the occurrence of various diseases [40,41]. There are two subtypes of STIM proteins, STIM1 and STIM2, whose calcium affinity has been debated in the field for many years. STIM1 is thought to regulate the classical SOCE process [42], while STIM2, due to its lower Ca2+ affinity, can sense more subtle changes in Ca2+ concentration and play a fine-tuning role [43]. STIM1, a type of transmembrane protein mainly located in the endoplasmic reticulum, is the calcium sensor in the endoplasmic reticulum, mainly promoting Ca2+ transport [44]. Significant reductions in STIM1 protein levels have been found in brain tissue in AD (Alzheimer’s disease) patients, while in PD patients, complexes formed by STIM1 and TRPC1 (transient receptor potential channel 1) can inhibit CaV1.3 channels, leading to disruption of neuronal Ca2+ homeostasis [45], ultimately leading to the development of PD symptoms. STIM2 is more widely distributed in the hippocampus and is involved in dendrite production [46], maturation, and stabilization through the important calcium/calmodulin-dependent protein kinase or calmodulin kinase pathways [47]. Syt7 (synaptotagmin 7) is a calcium ion detector that dynamically increases neurotransmitter release [48]. It has been demonstrated that STIM2 is mostly present at the presynaptic end, and SOCE can enhance spontaneous neurotransmission at excitatory synapses by facilitating the release of syt7-mediated neurotransmitters through STIM2 [24,49]. Long-term increased self-release can lead to severe synaptic dysfunction and apoptosis [50]. Endoplasmic reticulum stress has been found in a variety of neurodegenerative diseases, and it is likely that the presynaptic SOCE process also secretly contributes its own dark power to these diseases [51,52,53]. This sheds light on the possible impact of a mechanism of neurotransmitter release regulated by endoplasmic reticulum calcium ions on Parkinson’s disease and opens new possibilities for the treatment of neurodegenerative diseases [54].
The Ca2+ concentration in mitochondria mainly depends on the ER-mitochondria-associated membrane (MAM) and mitochondrial pathways [55]. Mitochondrial Ca2+ levels are strongly correlated with ER, as 20% of mitochondria are closely associated with the endoplasmic reticulum [56]. This region is called MAM, and the two are usually about 10–25 nm apart, which regulates the influx of calcium ions into the mitochondrial matrix [57]. The specific mechanism is through the IP3-VDAC1 (voltage-dependent anion channel 1) pathway, calcium ions in the endoplasmic reticulum can be directly released through IP3R (IP 3 receptor), enter the cytoplasm from the endoplasmic reticulum, and then enter mitochondria through the calcium ion channel VDAC1 on the outer mitochondrial membrane [58,59]. VDAC1 and IP3R control the permeability of the endoplasmic reticulum and mitochondria to calcium ions, respectively, so the abnormal expression of the two will have a great influence on the distribution of calcium ions. Previous studies have shown that when VDAC1 expression is increased, oligomerization forms large pores, opening MCU (mitochondrial calcium uniporter) and coupling MUC to IP3Rs, which leads to mitochondrial calcium uptake and mitochondrial calcium overload. It can promote the oligomerization of Bax/Bak to form a pore or open the mitochondrial permeability transition pore or Mitochondrial permeability transition pore (mPTP) [60]. Bax and Bak are two key pro-apoptotic proteins of the Bcl2 family. In healthy cells, Bak is distributed in the outer mitochondrial membrane, and Bax is distributed in the cytoplasm. After Cyt C enters the cytoplasm, it binds to Apaf1 and activates Caspase-9, which, in turn, activates Caspase-3, thus initiating the apoptotic cascade and causing apoptosis [61].
Interestingly, excess calcium levels trigger mitochondrial stress, mitochondrial dysfunction, and even neuronal death. Low mitochondrial calcium levels can also affect the viability of nerve cells. For example, in Parkinson’s disease, α-synapsin has been confirmed to be localized to MAM [62,63]. Studies have shown that the overexpression of α-synapsin in MAM will destroy the VAPB (ER protein capsule) [64]. VAPB (vesicle-associated membrane protein-associated protein B)—PTPIP51 (mitochondrial outer membrane protein, protein tyrosine phosphatase interacting protein 51) tethers affected ER-mitochondrial association, thereby affecting calcium signaling between mitochondria and endoplasmic reticulum and ATP production [65,66,67]. Studies have shown that no matter what kind of α-synapsin mutation, such as familial mutation or wild-type changes, can affect the VAPB–PTPIP51 tether; the result is the amount of calcium delivered by the ER to the mitochondria, which is reduced, and thus the calcium conduction is delayed, and because the mitochondrial tricarboxylic acid cycle is also regulated by Ca2+, resulting in reduced mitochondrial ATP production, which poses a serious threat to the viability of neurons [68,69].
Given the ubiquity of Ca2+ signaling in biology and its involvement in the etiology of PD and other neurodegenerative diseases, a growing body of theory now supports the idea that Ca2+ imbalance may be a key factor in aging. As discussed in this review, Ca2+ homeostasis regulation involves plasma membrane and intracellular calcium storage and the complex coordination within organelle networks; ultimately, the realization of metabolic interactions, intracellular signaling, cell maintenance, and cell survival regulation all require Ca2+ involvement. Understanding the mechanisms by which Ca2+ signaling promotes PD progression is therefore critical for the development of effective therapies for the disease.

3. Ferrum and PD

Iron is an indispensable trace element in cellular metabolism and is particularly important in the central nervous system [70]. Iron is involved in the development of brain synapses [71], myelination, and the generation and operation of neurotransmitters [72], which can affect the development of children’s IQ [73], cognition, movement, and social functions [74]. Studies have shown that ferritin levels in the brain increase with age and may be responsible for brain iron overload and neurodegenerative diseases in older adults [75,76]. Excess iron is linked to oxidative stress, inflammation, and cell death [77,78], and elevated iron levels have been detected in neurodegenerative conditions such as Parkinson’s disease and Alzheimer’s disease [79]. Cellular metabolism in the central nervous system requires iron as a redox metal to generate energy, primarily ATP, but neural tissue is susceptible to oxidative damage due to excess iron deposition, ultimately resulting in fibrinolysis [80].
Interestingly, iron deposition in the central nervous system is not solely attributed to aging; it is also influenced by iron metabolism-related diseases such as HH (hereditary hemochromatosis) [81]. HH is an autosomal recessive disorder of iron metabolism from mutations in the HFE (homeostatic iron regulator) gene (most notably C282Y and H63D) that regulate hepcidin function [81]. Iron in the brain is majorly concentrated in the basal ganglia and SN. In systemic iron imbalance, the blood–brain barrier protects the brain from the effects. However, in the case of iron overload, the BBB can disrupt the facilitation of the excess entry of iron into the brain. Patients with HH may be at an increased risk of PD due to their dysregulation of iron storage and metabolism [82].
Ferroptosis is an emerging caspase-independent RCD (regulated cell death) form proposed by Dr. Brent R. Stockwell, which is an iron-dependent form of cell death distinct from apoptosis, cell necrosis, and autophagy. There is no significant plasma membrane rupture and bioenergetic depletion in ferroptosis, and unlike other cell death morphologies, ferroptosis has smaller mitochondria than normal mitochondria and is characterized by reduced mitochondrial density [83], reduced or missing mitochondrial crest, and rupture of the outer mitochondrial membrane. In neurodegenerative diseases, ferroptosis triggers a series of events, including inflammatory activation, neurotransmitter oxidation, neuronal communication failure, myelin degeneration, astrocyte dysregulation, and nerve cell death [84,85].
It has been shown that accumulation of glutamate, iron, and polyunsaturated fatty acids, or depletion of GSH (Glutathione), NAD(P)H (Nicotinamide adenine dinucleotide phosphate), and GPX4 (Glutathione peroxidase 4) can trigger ferroptosis [86]. Iron homeostasis regulation is particularly important in the process of ferroptosis, and mutations in genes involved in iron metabolism may lead to increased intracellular iron input or decreased iron output [87]. The import of iron begins with the binding of TFR1 (Transferrin receptor 1) and subsequent endocytosis in endosomes. In acidic endosomes, Fe3+ is reduced to Fe2+ by STEAP3 (six-transmembrane epithelial antigen of prostate 3) and transported to the cytoplasm by DMT1 (divalent metal transporter 1) [88]. Iron in the form of Fe2+ can then be stored in ferritin or retained in the cytoplasm as a LIP (labile iron pool). Excess intracellular Fe2+ can directly catalyze the generation of lipid-free radicals through the Fenton reaction. PUFA (polyunsaturated fatty acid) is the main substrate for lipid peroxidation, and the plasma membrane contains a large amount of PUFAs and lipid-free radicals. PUFAs undergo peroxidation with lipid radicals to form PL-OOH (lipid peroxides). PL-OOH then cleaves, producing highly electrophilic secondary oxidation products [89], including epoxy, oxo, or aldehyde groups, which are highly reactive and toxic to membranes and cells. Eventually, nanometer-sized pores form on the cell-surface membrane, mediating the cell break-up that triggers ferroptosis [90]. Peroxidation-induced signals propagate from cell to cell in a unique wave-like diffusion pattern before cell breakdown as iron and lipid peroxidation persist, repeatedly creating a vicious cycle leading to cell death [91].
At present, there are three ways to prevent ferroptosis: the GSH/GPX4 pathway, the recently discovered FSP1/CoQ10/NAD(P)H pathway, and the GCH1 (GTP cyclohydrolase 1)/BH4 (Tetrahydrobiopterin)/DHFR (Dihydrofolate reductase) pathway (Figure 2) [92].
GPX4 (glutathione peroxidase 4), the star molecule for the study of ferroptosis, is an indicator of ferroptosis. Under physiological conditions, GPX4 oxidizes glutathione and reduces lipid peroxide (PL-OOH) associated with iron death to equivalent alcohol (PL-OH); thus, GPX4 activity is crucial in ferroptosis [93]. In GPX4’s fight against ferroptosis, glutathione is an essential substrate of GPX4. GSH is a linear tripeptide composed of glutamic acid, cysteine, and glycine, of which the thiol group on cysteine is considered its active site [94]. Its synthesis is influenced by the system XC-(cystine/glutamateantiporter), which regulates intracellular cystine levels [95]. Cystine enters the cell via system XC- and is then reduced to cysteine under the action of GSH or TRR1 (the enzyme thioredoxin reductase 1) [96]. Then, in the presence of ATP (adenosine triphosphate), GCL (glutamate cysteine ligase) catalyzes the combination of glutamate and cysteine to form γ-glutamylcysteine molecules. Finally, γ-glutamylcysteine is catalyzed by GS (glutathione synthetase) to connect with glycine to form GSH. In the biosynthesis of GSH, several transcriptases play a great role, such as GCL and GS. The synthesis of these enzymes is related to Nrf2 (nuclear factor-E2-related factor 2), and Nrf2 also exhibits strong antioxidant activity, which mainly relies on the expression of downstream gene proteins such as HO-1 (Heme Oxygenase-1), NQO1 (NAD(P)H quinone dehydrogenase 1) [97,98], GSTs (Glutathione-S-transferses), and GCLC (Glutamate cysteine ligase catalytic). Modulating Nrf2 expression may offer a new therapeutic approach to neurodegenerative diseases such as PD.
The FSP1-CoQ10-NAD(P)H pathway exists as an independent parallel system that cooperates with GPX4 and glutathione to inhibit phospholipid peroxidation and ferroptosis [99]. FSP1 (ferroptosis suppressor protein 1), discovered to be a glutathione-independent ferroptosis suppressor, contains an N-myristoylation signal and a flavoprotein oxidoreductase domain, which are thought to be critical sites for the inhibition of ferroptosis. CoQ10 (Coenzyme Q10) has long been shown to be a mobile lipophilic electron carrier, an endogenous synthetic fat-soluble antioxidant that plays a critical role in the mitochondrial electron transport chain [100,101]. Recent studies have demonstrated that CoQ10 can also act as a lipophilic radical scavenger antioxidant in plasma membranes [102,103]. FSP1 can prevent lipid oxidation by reducing CoQ10 with the assistance of NAD(P)H, and CoQ10 and its reduced form CoQ10-H2 can scavenge free radicals on the membrane surface and protect the integrity of lipoprotein and phospholipid molecules on the cell membrane [104]. In addition, NQO1 is a well-known NAD(P)H-dependent oxidoreductase that plays a role in ferroptosis protection by acting synergistically with FSP1 and regulating the reduction of ubiquinone to ubiquinol [105]. Interestingly, Nrf2 is also the central control factor for NQO1 expression [106]. The FSP1-CoQ10-NAD(P)H pathway still needs to be studied as a treatment for ferroptosis, but the FSP1-CoQ10-NAD(P)H pathway is second only to the GPX4/GSH pathway in the prevention of iron death to some extent and has great potential.
At present, there is minor research on combating ferroptosis through the GCH1/BH4/DHFR pathway in neurodegenerative diseases [107]. BH4 is a central factor in this pathway, playing a key role in biological processes and pathological states associated with the formation of certain neurotransmitters, immune responses, and pain sensitivity, and is an important cofactor in dopamine synthesis. BH4 is an antioxidant that traps lipid peroxidation radicals, while also promoting CoQ10 synthesis by converting phenylalanine to tyrosine, another way it exerts antioxidant effects. GCH1 is a rate-limiting enzyme for the synthesis of BH4, and the recovery and reuse of BH4 requires the participation of DHFR [108]. So, the DHFR inhibition can work together with GPX4 inhibitors to induce ferroptosis [109]. At present, the potential physiological and pathological role of GCH1/BH4/DHFR system in iron death and its application in Parkinson’s disease remain debatable.
In view of the mechanism of ferroptosis, we summarized the main therapeutic approaches for ferroptosis. We suggest that iron chelators, antioxidants, GPX4 activators, and NAD(P)H therapeutic agents may be more effective. We have collected the drugs targeting ferroptosis in PD in the past five years, and the specific content is in Table 1.

4. Calcium and Iron Crosstalk

The aforementioned statement highlights the role of iron in facilitating the generation of reactive oxygen species (ROS), which play a crucial physiological function in neuronal cells. ROS participates in the functional and structural modifications required for synaptic plasticity. Moreover, ROS levels regulate the function of the neuronal NMDA-R, Ca2+ channels, K+ channels, and CaMKII (calcium calmodulin dependent protein kinase II), among other cellular signaling enzymes. In particular, ROS can facilitate redox modification of RyR in hippocampal neurons, leading to activation of RyR-mediated Ca2+ release, thereby inducing phosphorylation of two enzymes involved in synaptic plasticity. Similarly, iron-induced ROS production can also trigger RyR-mediated Ca2+ signaling that promotes ERK1/2 phosphorylation in primary hippocampal cultures under Ca2+-free conditions.
However, it is indisputable that high levels of ROS exert deleterious effects on neuronal cells. If the iron-induced release of Ca2+ mediated by RyR is enhanced, subsequent mitochondrial fission may also contribute to neuronal dysfunction. Excessive iron-induced peroxidation damages mitochondria, resulting in elevated levels of mitochondrial Ca2+, which subsequently stimulate the Ca2+-dependent phosphatase calcineurin and leads to neuronal cell death. It should be noted that the interaction between iron and Ca2+ signaling is bidirectional, as numerous proteins involved in cellular antioxidant defense and ROS production are dependent on Ca2+. In addition, lipid peroxidation products such as 4-HNE stimulate the opening of plasma membrane Ca2+ channels, including VGCCs in hippocampal neurons. Additionally, elevated 4-HNE levels can provoke energy dyshomeostasis and neuronal death by diminishing the functionality of Na+- and Ca2+-pumps and by altering Ca2+ channel permeability. It is important to note that the crosstalk between iron and Ca2+ signaling is bidirectional, as many proteins involved in cellular antioxidant defense and ROS production are Ca2+-dependent. An abnormal increase in intracellular calcium or an increased influx of calcium within mitochondria leads to more ROS production, which can lead to a disorder of the iron pool. Moreover, an increase of Ca2+ in the cytoplasm stimulates the neuronal nitric oxide synthase and the NAD(P)H oxidase, two enzymes involved in ROS generation and synaptic plasticity.
The above description points to a significant crosstalk between Ca2+ and iron. Thus, when one of the components is dysregulated, the other component is also affected, resulting in neuronal death.

5. Conclusions and Future Perspectives

PD is a common neurodegenerative disease, the true cause of which remains unknown. Metal elements’ ions may play an essential and irreplaceable role in the occurrence and development of Parkinson’s disease, of which the role of calcium and iron is particularly prominent. As we describe in this review, calcium, regulated by the plasma membrane and intracellular stored organelles (endoplasmic reticulum, mitochondria, etc.) is an indispensable signaling molecule in biological activity. Ca2+ homeostasis regulation is crucial for nerve cell activity, and the anti-neurodegenerative effects of calcium-channel blockers have brought light to us that microscopic exploration of Ca2+ signaling is crucial for the development of effective therapies for PD. Parkinson’s disease patients have a large amount of iron deposits in the substantia nigra, and ferroptosis is also an important factor influencing the occurrence and development of Parkinson’s disease. In the study of ferroptosis, most studies focus on the GPX/GSH pathway, and most of the studies in the past five years are Nrf2 agonists. While this is a truly effective and classical pathway, the therapeutic potential of the FSP1/CoQ10/NAD (P) H pathway is also of interest, but the key question here should be how to address the problem of CoQ10 synthesis at the cell membrane. BH4 has been described in dopamine synthesis and may also be valuable for the study of the significance of ferroptosis as well as the effect of PD. In our review, we have identified a crosstalk between calcium and iron signaling pathways, indicating that the imbalance of one pathway can significantly impact the balance of the other. Therefore, it is imperative to develop multifunctional drugs that target both Ca2+- and iron-dependent mechanisms.

Author Contributions

Writing—original draft preparation, J.W. and J.Z.; Visualization, K.Z. and S.W.; Writing—review and editing, X.C. and W.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by National Natural Science Foundation of China (81960666, W.Y.H.), Yunnan Province Young Academic and Technical Leaders Project (202105AC160078, W.Y.H.), and the joint Program of Yunnan Province and Kunming Medical University (202101AY070001-009, W.Y.H.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

The authors would like to thank Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University for the support.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Appel-Cresswell, S.; Vilarino-Guell, C.; Encarnacion, M.; Sherman, H.; Yu, I.; Shah, B.; Weir, D.; Thompson, C.; Szu-Tu, C.; Trinh, J.; et al. Alpha-synuclein p.H50Q, a novel pathogenic mutation for Parkinson’s disease. Mov. Disord. 2013, 28, 811–813. [Google Scholar] [CrossRef] [PubMed]
  2. Darweesh, S.K.L.; Raphael, K.G.; Brundin, P.; Matthews, H.; Wyse, R.K.; Chen, H.; Bloem, B.R. Parkinson Matters. J. Parkinsons. Dis. 2018, 8, 495–498. [Google Scholar] [CrossRef] [PubMed]
  3. Bezard, E.; Yue, Z.; Kirik, D.; Spillantini, M.G. Animal models of Parkinson’s disease: Limits and relevance to neuroprotection studies. Mov. Disord. 2013, 28, 61–70. [Google Scholar] [CrossRef] [PubMed]
  4. Blandini, F.; Armentero, M.T. Animal models of Parkinson’s disease. FEBS J. 2012, 279, 1156–1166. [Google Scholar] [CrossRef]
  5. Breckenridge, C.B.; Berry, C.; Chang, E.T.; Sielken, R.L., Jr.; Mandel, J.S. Association between Parkinson’s Disease and Cigarette Smoking, Rural Living, Well-Water Consumption, Farming and Pesticide Use: Systematic Review and Meta-Analysis. PLoS ONE 2016, 11, e0151841. [Google Scholar] [CrossRef]
  6. Ascherio, A.; Schwarzschild, M.A. The epidemiology of Parkinson’s disease: Risk factors and prevention. Lancet Neurol. 2016, 15, 1257–1272. [Google Scholar] [CrossRef]
  7. Das, N.; Raymick, J.; Sarkar, S. Role of metals in Alzheimer’s disease. Metab. Brain Dis. 2021, 36, 1627–1639. [Google Scholar] [CrossRef]
  8. Li, B.; Xia, M.; Zorec, R.; Parpura, V.; Verkhratsky, A. Astrocytes in heavy metal neurotoxicity and neurodegeneration. Brain Res. 2021, 1752, 147234. [Google Scholar] [CrossRef]
  9. Mezzaroba, L.; Alfieri, D.F.; Simao, A.N.C.; Reiche, E.M.V. The role of zinc, copper, manganese and iron in neurodegenerative diseases. Neurotoxicology 2019, 74, 230–241. [Google Scholar] [CrossRef]
  10. Kadala, A.; Verdier, D.; Morquette, P.; Kolta, A. Ion Homeostasis in Rhythmogenesis: The Interplay Between Neurons and Astroglia. Physiology 2015, 30, 371–388. [Google Scholar] [CrossRef]
  11. Li, L.B.; Chai, R.; Zhang, S.; Xu, S.F.; Zhang, Y.H.; Li, H.L.; Fan, Y.G.; Guo, C. Iron Exposure and the Cellular Mechanisms Linked to Neuron Degeneration in Adult Mice. Cells 2019, 8, 198. [Google Scholar] [CrossRef] [PubMed]
  12. Ureshino, R.P.; Erustes, A.G.; Bassani, T.B.; Wachilewski, P.; Guarache, G.C.; Nascimento, A.C.; Costa, A.J.; Smaili, S.S.; Pereira, G. The Interplay between Ca2+ Signaling Pathways and Neurodegeneration. Int. J. Mol. Sci. 2019, 20, 6004. [Google Scholar] [CrossRef] [PubMed]
  13. Raffaello, A.; Mammucari, C.; Gherardi, G.; Rizzuto, R. Calcium at the Center of Cell Signaling: Interplay between Endoplasmic Reticulum, Mitochondria, and Lysosomes. Trends Biochem. Sci. 2016, 41, 1035–1049. [Google Scholar] [CrossRef] [PubMed]
  14. Raiteri, L.; Raiteri, M. Multiple functions of neuronal plasma membrane neurotransmitter transporters. Prog. Neurobiol. 2015, 134, 1–16. [Google Scholar] [CrossRef] [PubMed]
  15. Cronin, M.J.; Anderson, J.M.; Rogol, A.D.; Koritnik, D.R.; Thorner, M.O.; Evans, W.S. Calcium channel agonist BAY k8644 enhances anterior pituitary secretion in rat and monkey. Am. J. Physiol. 1985, 249, E326–E329. [Google Scholar] [CrossRef] [PubMed]
  16. Thiel, G.; Schmidt, T.; Rossler, O.G. Ca2+ Microdomains, Calcineurin and the Regulation of Gene Transcription. Cells 2021, 10, 875. [Google Scholar] [CrossRef] [PubMed]
  17. Buttner, S.; Faes, L.; Reichelt, W.N.; Broeskamp, F.; Habernig, L.; Benke, S.; Kourtis, N.; Ruli, D.; Carmona-Gutierrez, D.; Eisenberg, T.; et al. The Ca2+/Mn2+ ion-pump PMR1 links elevation of cytosolic Ca2+ levels to alpha-synuclein toxicity in Parkinson’s disease models. Cell Death Differ. 2013, 20, 465–477. [Google Scholar] [CrossRef]
  18. Vance, J.E. Phospholipid synthesis and transport in mammalian cells. Traffic 2015, 16, 1–18. [Google Scholar] [CrossRef]
  19. Verkhratsky, A.; Trebak, M.; Perocchi, F.; Khananshvili, D.; Sekler, I. Crosslink between calcium and sodium signalling. Exp. Physiol. 2018, 103, 157–169. [Google Scholar] [CrossRef]
  20. Samavarchi Tehrani, S.; Sarfi, M.; Yousefi, T.; Ahangar, A.A.; Gholinia, H.; Ahangar, R.M.; Maniati, M.; Saadat, P. Comparison of the calcium-related factors in Parkinson’s disease patients with healthy individuals. Caspian J. Intern. Med. 2020, 11, 28–33. [Google Scholar]
  21. Llorente-Folch, I.; Rueda, C.B.; Pardo, B.; Szabadkai, G.; Duchen, M.R.; Satrustegui, J. The regulation of neuronal mitochondrial metabolism by calcium. J. Physiol. 2015, 593, 3447–3462. [Google Scholar] [CrossRef] [PubMed]
  22. Mira, R.G.; Cerpa, W. Building a Bridge Between NMDAR-Mediated Excitotoxicity and Mitochondrial Dysfunction in Chronic and Acute Diseases. Cell Mol. Neurobiol. 2021, 41, 1413–1430. [Google Scholar] [CrossRef] [PubMed]
  23. Bohush, A.; Lesniak, W.; Weis, S.; Filipek, A. Calmodulin and Its Binding Proteins in Parkinson’s Disease. Int. J. Mol. Sci. 2021, 22, 3016. [Google Scholar] [CrossRef] [PubMed]
  24. Chanaday, N.L.; Nosyreva, E.; Shin, O.H.; Zhang, H.; Aklan, I.; Atasoy, D.; Bezprozvanny, I.; Kavalali, E.T. Presynaptic store-operated Ca2+ entry drives excitatory spontaneous neurotransmission and augments endoplasmic reticulum stress. Neuron 2021, 109, 1314–1332.e5. [Google Scholar] [CrossRef] [PubMed]
  25. Elyasi, L.; Jahanshahi, M.; Jameie, S.B.; Abadi, H.G.H.; Nikmahzar, E.; Khalili, M.; Jameie, M.; Jameie, M. 6-OHDA mediated neurotoxicity in SH-SY5Y cellular model of Parkinson disease suppressed by pretreatment with hesperidin through activating L-type calcium channels. J. Basic. Clin. Physiol. Pharmacol. 2020, 32, 11–17. [Google Scholar] [CrossRef]
  26. Jefri, M.; Bell, S.; Peng, H.; Hettige, N.; Maussion, G.; Soubannier, V.; Wu, H.; Silveira, H.; Theroux, J.F.; Moquin, L.; et al. Stimulation of L-type calcium channels increases tyrosine hydroxylase and dopamine in ventral midbrain cells induced from somatic cells. Stem Cells Transl. Med. 2020, 9, 697–712. [Google Scholar] [CrossRef] [PubMed]
  27. Liss, B.; Striessnig, J. The Potential of L-Type Calcium Channels as a Drug Target for Neuroprotective Therapy in Parkinson’s Disease. Annu. Rev. Pharmacol. Toxicol. 2019, 59, 263–289. [Google Scholar] [CrossRef]
  28. Berger, S.M.; Bartsch, D. The role of L-type voltage-gated calcium channels Cav1.2 and Cav1.3 in normal and pathological brain function. Cell Tissue Res. 2014, 357, 463–476. [Google Scholar] [CrossRef]
  29. Samak, G.; Narayanan, D.; Jaggar, J.H.; Rao, R. CaV1.3 channels and intracellular calcium mediate osmotic stress-induced N-terminal c-Jun kinase activation and disruption of tight junctions in Caco-2 CELL MONOLAYERS. J. Biol. Chem. 2011, 286, 30232–30243. [Google Scholar] [CrossRef]
  30. Kang, S.; Cooper, G.; Dunne, S.F.; Luan, C.H.; Surmeier, D.J.; Silverman, R.B. Antagonism of L-type Ca2+ channels CaV1.3 and CaV1.2 by 1,4-dihydropyrimidines and 4H-pyrans as dihydropyridine mimics. Bioorg Med. Chem. 2013, 21, 4365–4373. [Google Scholar] [CrossRef]
  31. Hasreiter, J.; Goldnagl, L.; Bohm, S.; Kubista, H. Cav1.2 and Cav1.3 L-type calcium channels operate in a similar voltage range but show different coupling to Ca(2+)-dependent conductances in hippocampal neurons. Am. J. Physiol. Cell Physiol. 2014, 306, C1200–C1213. [Google Scholar] [CrossRef] [PubMed]
  32. Jung, J.H.; Na, H.K.; Jeong, S.H.; Chung, S.J.; Yoo, H.S.; Lee, Y.H.; Baik, K.; Kim, S.J.; Sohn, Y.H.; Lee, P.H. Effects of Dihydropyridines on the Motor and Cognitive Outcomes of Patients with Parkinson’s Disease. Mov. Disord. 2023, 38, 843–853. [Google Scholar] [CrossRef]
  33. Guzman, J.N.; Ilijic, E.; Yang, B.; Sanchez-Padilla, J.; Wokosin, D.; Galtieri, D.; Kondapalli, J.; Schumacker, P.T.; Surmeier, D.J. Systemic isradipine treatment diminishes calcium-dependent mitochondrial oxidant stress. J. Clin. Investig. 2018, 128, 2266–2280. [Google Scholar] [CrossRef] [PubMed]
  34. Ilijic, E.; Guzman, J.N.; Surmeier, D.J. The L-type channel antagonist isradipine is neuroprotective in a mouse model of Parkinson’s disease. Neurobiol. Dis. 2011, 43, 364–371. [Google Scholar] [CrossRef] [PubMed]
  35. Zamponi, G.W.; Striessnig, J.; Koschak, A.; Dolphin, A.C. The Physiology, Pathology, and Pharmacology of Voltage-Gated Calcium Channels and Their Future Therapeutic Potential. Pharmacol. Rev. 2015, 67, 821–870. [Google Scholar] [CrossRef]
  36. Alexander, S.P.; Peters, J.A.; Kelly, E.; Marrion, N.V.; Faccenda, E.; Harding, S.D.; Pawson, A.J.; Sharman, J.L.; Southan, C.; Davies, J.A. The concise guide to pharmacology 2017/18: Ligand-gated ion channels. Br. J. Pharmacol. 2017, 174 (Suppl. S1), S130–S159. [Google Scholar] [CrossRef]
  37. Surmeier, D.J. Determinants of dopaminergic neuron loss in Parkinson’s disease. FEBS J. 2018, 285, 3657–3668. [Google Scholar] [CrossRef] [PubMed]
  38. Minakaki, G.; Krainc, D.; Burbulla, L.F. The Convergence of Alpha-Synuclein, Mitochondrial, and Lysosomal Pathways in Vulnerability of Midbrain Dopaminergic Neurons in Parkinson’s Disease. Front. Cell Dev. Biol. 2020, 8, 580634. [Google Scholar] [CrossRef]
  39. Erhardt, B.; Marcora, M.S.; Frenkel, L.; Bochicchio, P.A.; Bodin, D.H.; Silva, B.A.; Farias, M.I.; Allo, M.A.; Hocht, C.; Ferrari, C.C.; et al. Plasma membrane calcium ATPase downregulation in dopaminergic neurons alters cellular physiology and motor behaviour in Drosophila melanogaster. Eur. J. Neurosci. 2021, 54, 5915–5931. [Google Scholar] [CrossRef]
  40. Collins, H.E.; Zhang, D.; Chatham, J.C. STIM and Orai Mediated Regulation of Calcium Signaling in Age-Related Diseases. Front. Aging 2022, 3, 876785. [Google Scholar] [CrossRef]
  41. Nwokonko, R.M.; Cai, X.; Loktionova, N.A.; Wang, Y.; Zhou, Y.; Gill, D.L. The STIM-Orai Pathway: Conformational Coupling Between STIM and Orai in the Activation of Store-Operated Ca2+ Entry. Adv. Exp. Med. Biol. 2017, 993, 83–98. [Google Scholar] [PubMed]
  42. Prakriya, M.; Lewis, R.S. Store-Operated Calcium Channels. Physiol. Rev. 2015, 95, 1383–1436. [Google Scholar] [CrossRef]
  43. Serwach, K.; Gruszczynska-Biegala, J. Target Molecules of STIM Proteins in the Central Nervous System. Front. Mol. Neurosci. 2020, 13, 617422. [Google Scholar] [CrossRef] [PubMed]
  44. Nguyen, N.T.; Han, W.; Cao, W.M.; Wang, Y.; Wen, S.; Huang, Y.; Li, M.; Du, L.; Zhou, Y. Store-Operated Calcium Entry Mediated by ORAI and STIM. Compr. Physiol. 2018, 8, 981–1002. [Google Scholar] [PubMed]
  45. Meng, F.; Fleming, B.A.; Jia, X.; Rousek, A.A.; Mulvey, M.A.; Ward, D.M. Lysosomal iron recycling in mouse macrophages is dependent upon both LcytB and Steap3 reductases. Blood Adv. 2022, 6, 1692–1707. [Google Scholar] [CrossRef] [PubMed]
  46. Gruszczynska-Biegala, J.; Kuznicki, J. Native STIM2 and ORAI1 proteins form a calcium-sensitive and thapsigargin-insensitive complex in cortical neurons. J. Neurochem. 2013, 126, 727–738. [Google Scholar] [CrossRef] [PubMed]
  47. Gruszczynska-Biegala, J.; Pomorski, P.; Wisniewska, M.B.; Kuznicki, J. Differential roles for STIM1 and STIM2 in store-operated calcium entry in rat neurons. PLoS ONE 2011, 6, e19285. [Google Scholar] [CrossRef]
  48. Xiao, B.; Li, J.; Fan, Y.; Ye, M.; Lv, S.; Xu, B.; Chai, Y.; Zhou, Z.; Wu, M.; Zhu, X. Downregulation of SYT7 inhibits glioblastoma growth by promoting cellular apoptosis. Mol. Med. Rep. 2017, 16, 9017–9022. [Google Scholar] [CrossRef]
  49. Ahmad, M.; Ong, H.L.; Saadi, H.; Son, G.Y.; Shokatian, Z.; Terry, L.E.; Trebak, M.; Yule, D.I.; Ambudkar, I. Functional communication between IP3R and STIM2 at subthreshold stimuli is a critical checkpoint for initiation of SOCE. Proc. Natl. Acad. Sci. USA 2022, 119, e2114928118. [Google Scholar] [CrossRef]
  50. Skopin, A.Y.; Grigoryev, A.D.; Glushankova, L.N.; Shalygin, A.V.; Wang, G.; Kartzev, V.G.; Kaznacheyeva, E.V. A Novel Modulator of STIM2-Dependent Store-Operated Ca2+ Channel Activity. Acta Naturae 2021, 13, 140–146. [Google Scholar] [CrossRef]
  51. Silva-Rojas, R.; Laporte, J.; Bohm, J. STIM1/ORAI1 Loss-of-Function and Gain-of-Function Mutations Inversely Impact on SOCE and Calcium Homeostasis and Cause Multi-Systemic Mirror Diseases. Front. Physiol. 2020, 11, 604941. [Google Scholar] [CrossRef]
  52. Sukumaran, P.; Da Conceicao, V.N.; Sun, Y.; Ahamad, N.; Saraiva, L.R.; Selvaraj, S.; Singh, B.B. Calcium Signaling Regulates Autophagy and Apoptosis. Cells 2021, 10, 2125. [Google Scholar] [CrossRef] [PubMed]
  53. Scremin, E.; Agostini, M.; Leparulo, A.; Pozzan, T.; Greotti, E.; Fasolato, C. ORAI2 Down-Regulation Potentiates SOCE and Decreases Abeta42 Accumulation in Human Neuroglioma Cells. Int. J. Mol. Sci. 2020, 21, 5288. [Google Scholar] [CrossRef] [PubMed]
  54. Sun, Y.; Zhang, H.; Selvaraj, S.; Sukumaran, P.; Lei, S.; Birnbaumer, L.; Singh, B.B. Inhibition of L-Type Ca2+ Channels by TRPC1-STIM1 Complex Is Essential for the Protection of Dopaminergic Neurons. J. Neurosci. 2017, 37, 3364–3377. [Google Scholar] [CrossRef]
  55. Veeresh, P.; Kaur, H.; Sarmah, D.; Mounica, L.; Verma, G.; Kotian, V.; Kesharwani, R.; Kalia, K.; Borah, A.; Wang, X.; et al. Endoplasmic reticulum-mitochondria crosstalk: From junction to function across neurological disorders. Ann. N. Y Acad. Sci. 2019, 1457, 41–60. [Google Scholar] [CrossRef] [PubMed]
  56. Barazzuol, L.; Giamogante, F.; Brini, M.; Cali, T. PINK1/Parkin Mediated Mitophagy, Ca2+ Signalling, and ER-Mitochondria Contacts in Parkinson’s Disease. Int. J. Mol. Sci. 2020, 21, 1772. [Google Scholar] [CrossRef] [PubMed]
  57. Szymanski, J.; Janikiewicz, J.; Michalska, B.; Patalas-Krawczyk, P.; Perrone, M.; Ziolkowski, W.; Duszynski, J.; Pinton, P.; Dobrzyn, A.; Wieckowski, M.R. Interaction of Mitochondria with the Endoplasmic Reticulum and Plasma Membrane in Calcium Homeostasis, Lipid Trafficking and Mitochondrial Structure. Int. J. Mol. Sci. 2017, 18, 1576. [Google Scholar] [CrossRef] [PubMed]
  58. Yamamoto, K.; Izumi, Y.; Arifuku, M.; Kume, T.; Sawada, H. α-Synuclein oligomers mediate the aberrant form of spike-induced calcium release from IP3 receptor. Sci. Rep. 2019, 9, 15977. [Google Scholar] [CrossRef]
  59. Chung, K.M.; Jeong, E.J.; Park, H.; An, H.K.; Yu, S.W. Mediation of Autophagic Cell Death by Type 3 Ryanodine Receptor (RyR3) in Adult Hippocampal Neural Stem Cells. Front. Cell. Neurosci. 2016, 10, 116. [Google Scholar] [CrossRef]
  60. Huo, H.; Zhou, Z.; Qin, J.; Liu, W.; Wang, B.; Gu, Y. Erastin Disrupts Mitochondrial Permeability Transition Pore (mPTP) and Induces Apoptotic Death of Colorectal Cancer Cells. PLoS ONE 2016, 11, e0154605. [Google Scholar] [CrossRef]
  61. Li, X.; Fang, F.; Gao, Y.; Tang, G.; Xu, W.; Wang, Y.; Kong, R.; Tuyihong, A.; Wang, Z. ROS Induced by KillerRed Targeting Mitochondria (mtKR) Enhances Apoptosis Caused by Radiation via Cyt c/Caspase-3 Pathway. Oxidative Med. Cell. Longev. 2019, 2019, 4528616. [Google Scholar] [CrossRef] [PubMed]
  62. Lee, H.J.; Jung, Y.H.; Choi, G.E.; Kim, J.S.; Chae, C.W.; Lim, J.R.; Kim, S.Y.; Yoon, J.H.; Cho, J.H.; Lee, S.J.; et al. Urolithin A suppresses high glucose-induced neuronal amyloidogenesis by modulating TGM2-dependent ER-mitochondria contacts and calcium homeostasis. Cell Death Differ. 2021, 28, 184–202. [Google Scholar] [CrossRef] [PubMed]
  63. Liu, Y.; Ma, X.; Fujioka, H.; Liu, J.; Chen, S.; Zhu, X. DJ-1 regulates the integrity and function of ER-mitochondria association through interaction with IP3R3-Grp75-VDAC1. Proc. Natl. Acad. Sci. USA 2019, 116, 25322–25328. [Google Scholar] [CrossRef] [PubMed]
  64. Apicco, D.J.; Shlevkov, E.; Nezich, C.L.; Tran, D.T.; Guilmette, E.; Nicholatos, J.W.; Bantle, C.M.; Chen, Y.; Glajch, K.E.; Abraham, N.A.; et al. The Parkinson’s disease-associated gene ITPKB protects against α-synuclein aggregation by regulating ER-to-mitochondria calcium release. Proc. Natl. Acad. Sci. USA 2021, 118, e2006476118. [Google Scholar] [CrossRef] [PubMed]
  65. Huttlin, E.L.; Ting, L.; Bruckner, R.J.; Gebreab, F.; Gygi, M.P.; Szpyt, J.; Tam, S.; Zarraga, G.; Colby, G.; Baltier, K.; et al. The BioPlex Network: A Systematic Exploration of the Human Interactome. Cell 2015, 162, 425–440. [Google Scholar] [CrossRef]
  66. Paillusson, S.; Gomez-Suaga, P.; Stoica, R.; Little, D.; Gissen, P.; Devine, M.J.; Noble, W.; Hanger, D.P.; Miller, C.C.J. alpha-Synuclein binds to the ER-mitochondria tethering protein VAPB to disrupt Ca(2+) homeostasis and mitochondrial ATP production. Acta Neuropathol. 2017, 134, 129–149. [Google Scholar] [CrossRef]
  67. Stoica, R.; Paillusson, S.; Gomez-Suaga, P.; Mitchell, J.C.; Lau, D.H.; Gray, E.H.; Sancho, R.M.; Vizcay-Barrena, G.; De Vos, K.J.; Shaw, C.E.; et al. ALS/FTD-associated FUS activates GSK-3beta to disrupt the VAPB–PTPIP51 interaction and ER-mitochondria associations. EMBO Rep. 2016, 17, 1326–1342. [Google Scholar] [CrossRef]
  68. Stoica, R.; De Vos, K.J.; Paillusson, S.; Mueller, S.; Sancho, R.M.; Lau, K.F.; Vizcay-Barrena, G.; Lin, W.L.; Xu, Y.F.; Lewis, J.; et al. ER-mitochondria associations are regulated by the VAPB-PTPIP51 interaction and are disrupted by ALS/FTD-associated TDP-43. Nat. Commun. 2014, 5, 3996. [Google Scholar] [CrossRef]
  69. van Vliet, A.R.; Verfaillie, T.; Agostinis, P. New functions of mitochondria associated membranes in cellular signaling. Biochim. Biophys. Acta (BBA)—Mol. Cell Res. 2014, 1843, 2253–2262. [Google Scholar] [CrossRef]
  70. Ward, R.J.; Zucca, F.A.; Duyn, J.H.; Crichton, R.R.; Zecca, L. The role of iron in brain ageing and neurodegenerative disorders. Lancet Neurol. 2014, 13, 1045–1060. [Google Scholar] [CrossRef]
  71. Angelova, P.R.; Choi, M.L.; Berezhnov, A.V.; Horrocks, M.H.; Hughes, C.D.; De, S.; Rodrigues, M.; Yapom, R.; Little, D.; Dolt, K.S.; et al. Alpha synuclein aggregation drives ferroptosis: An interplay of iron, calcium and lipid peroxidation. Cell Death Differ. 2020, 27, 2781–2796. [Google Scholar] [CrossRef]
  72. Piao, Y.S.; Lian, T.H.; Hu, Y.; Zuo, L.J.; Guo, P.; Yu, S.Y.; Liu, L.; Jin, Z.; Zhao, H.; Li, L.X.; et al. Restless legs syndrome in Parkinson disease: Clinical characteristics, abnormal iron metabolism and altered neurotransmitters. Sci. Rep. 2017, 7, 10547. [Google Scholar] [CrossRef]
  73. Hect, J.L.; Daugherty, A.M.; Hermez, K.M.; Thomason, M.E. Developmental variation in regional brain iron and its relation to cognitive functions in childhood. Dev. Cogn. Neurosci. 2018, 34, 18–26. [Google Scholar] [CrossRef] [PubMed]
  74. Cheli, V.T.; Correale, J.; Paez, P.M.; Pasquini, J.M. Iron Metabolism in Oligodendrocytes and Astrocytes, Implications for Myelination and Remyelination. ASN Neuro 2020, 12, 1759091420962681. [Google Scholar] [CrossRef]
  75. Masaldan, S.; Clatworthy, S.A.S.; Gamell, C.; Meggyesy, P.M.; Rigopoulos, A.T.; Haupt, S.; Haupt, Y.; Denoyer, D.; Adlard, P.A.; Bush, A.I.; et al. Iron accumulation in senescent cells is coupled with impaired ferritinophagy and inhibition of ferroptosis. Redox Biol. 2018, 14, 100–115. [Google Scholar] [CrossRef]
  76. Mehrpouya, S.; Nahavandi, A.; Khojasteh, F.; Soleimani, M.; Ahmadi, M.; Barati, M. Iron administration prevents BDNF decrease and depressive-like behavior following chronic stress. Brain Res. 2015, 1596, 79–87. [Google Scholar] [CrossRef] [PubMed]
  77. Cui, Y.; Zhang, Z.; Zhou, X.; Zhao, Z.; Zhao, R.; Xu, X.; Kong, X.; Ren, J.; Yao, X.; Wen, Q.; et al. Microglia and macrophage exhibit attenuated inflammatory response and ferroptosis resistance after RSL3 stimulation via increasing Nrf2 expression. J. Neuroinflammation 2021, 18, 249. [Google Scholar] [CrossRef] [PubMed]
  78. Wang, C.; Chen, S.; Guo, H.; Jiang, H.; Liu, H.; Fu, H.; Wang, D. Forsythoside A Mitigates Alzheimer’s-like Pathology by Inhibiting Ferroptosis-mediated Neuroinflammation via Nrf2/GPX4 Axis Activation. Int. J. Biol. Sci. 2022, 18, 2075–2090. [Google Scholar] [CrossRef]
  79. Thomas, G.E.C.; Zarkali, A.; Ryten, M.; Shmueli, K.; Gil-Martinez, A.L.; Leyland, L.A.; McColgan, P.; Acosta-Cabronero, J.; Lees, A.J.; Weil, R.S. Regional brain iron and gene expression provide insights into neurodegeneration in Parkinson’s disease. Brain 2021, 144, 1787–1798. [Google Scholar] [CrossRef]
  80. Park, M.W.; Cha, H.W.; Kim, J.; Kim, J.H.; Yang, H.; Yoon, S.; Boonpraman, N.; Yi, S.S.; Yoo, I.D.; Moon, J.S. NOX4 promotes ferroptosis of astrocytes by oxidative stress-induced lipid peroxidation via the impairment of mitochondrial metabolism in Alzheimer’s diseases. Redox Biol. 2021, 41, 101947. [Google Scholar] [CrossRef]
  81. Bagwe-Parab, S.; Kaur, G. Molecular targets and therapeutic interventions for iron induced neurodegeneration. Brain Res. Bull. 2020, 156, 101947. [Google Scholar] [CrossRef] [PubMed]
  82. Li, Y.; Huang, X.; Wang, J.; Huang, R.; Wan, D. Regulation of Iron Homeostasis and Related Diseases. Mediat. Inflamm. 2020, 2020, 6062094. [Google Scholar] [CrossRef]
  83. Gao, M.; Yi, J.; Zhu, J.; Minikes, A.M.; Monian, P.; Thompson, C.B.; Jiang, X. Role of Mitochondria in Ferroptosis. Mol. Cell 2019, 73, 354–363.e3. [Google Scholar] [CrossRef]
  84. Liu, Y.; Wang, Z.; Cao, C.; Xu, Z.; Lu, J.; Shen, H.; Li, X.; Li, H.; Wu, J.; Chen, G. Aquaporin 4 Depolarization-Enhanced Transferrin Infiltration Leads to Neuronal Ferroptosis after Subarachnoid Hemorrhage in Mice. Oxidative Med. Cell. Longev. 2022, 2022, 8808677. [Google Scholar] [CrossRef]
  85. Wu, H.; Wang, F.; Ta, N.; Zhang, T.; Gao, W. The Multifaceted Regulation of Mitochondria in Ferroptosis. Life 2021, 11, 222. [Google Scholar] [CrossRef] [PubMed]
  86. Tang, D.; Chen, X.; Kang, R.; Kroemer, G. Ferroptosis: Molecular mechanisms and health implications. Cell Res. 2021, 31, 107–125. [Google Scholar] [CrossRef] [PubMed]
  87. Duck, K.A.; Connor, J.R. Iron uptake and transport across physiological barriers. Biometals 2016, 29, 573–591. [Google Scholar] [CrossRef] [PubMed]
  88. Lu, J.; Xu, F.; Lu, H. LncRNA PVT1 regulates ferroptosis through miR-214-mediated TFR1 and p53. Life Sci. 2020, 260, 118305. [Google Scholar] [CrossRef]
  89. 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, 5920–5936. [Google Scholar] [CrossRef]
  90. Skouta, R.; Dixon, S.J.; Wang, J.; Dunn, D.E.; Orman, M.; Shimada, K.; Rosenberg, P.A.; Lo, D.C.; Weinberg, J.M.; Linkermann, A.; et al. Ferrostatins inhibit oxidative lipid damage and cell death in diverse disease models. J. Am. Chem. Soc. 2014, 136, 4551–4556. [Google Scholar] [CrossRef]
  91. Bhat, A.H.; Dar, K.B.; Anees, S.; Zargar, M.A.; Masood, A.; Sofi, M.A.; Ganie, S.A. Oxidative stress, mitochondrial dysfunction and neurodegenerative diseases; a mechanistic insight. Biomed. Pharmacother. 2015, 74, 101–110. [Google Scholar] [CrossRef]
  92. Wang, X.; Wang, Y.; Li, Z.; Qin, J.; Wang, P. Regulation of Ferroptosis Pathway by Ubiquitination. Front. Cell Dev. Biol. 2021, 9, 699304. [Google Scholar] [CrossRef]
  93. Friedmann Angeli, J.P.; Schneider, M.; Proneth, B.; Tyurina, Y.Y.; Tyurin, V.A.; Hammond, V.J.; Herbach, N.; Aichler, M.; Walch, A.; Eggenhofer, E.; et al. Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nat. Cell Biol. 2014, 16, 1180–1191. [Google Scholar] [CrossRef] [PubMed]
  94. Wang, Y.; Chen, M.; Zhang, Y.; Huo, T.; Fang, Y.; Jiao, X.; Yuan, M.; Jiang, H. Effects of realgar on GSH synthesis in the mouse hippocampus: Involvement of system XAG, system XC, MRP-1 and Nrf2. Toxicol. Appl. Pharmacol. 2016, 308, 91–101. [Google Scholar] [CrossRef] [PubMed]
  95. Parker, J.L.; Deme, J.C.; Kolokouris, D.; Kuteyi, G.; Biggin, P.C.; Lea, S.M.; Newstead, S. Molecular basis for redox control by the human cystine/glutamate antiporter system xc. Nat. Commun. 2021, 12, 7147. [Google Scholar] [CrossRef]
  96. Floros, K.V.; Cai, J.; Jacob, S.; Kurupi, R.; Fairchild, C.K.; Shende, M.; Coon, C.M.; Powell, K.M.; Belvin, B.R.; Hu, B.; et al. MYCN-Amplified Neuroblastoma Is Addicted to Iron and Vulnerable to Inhibition of the System Xc/Glutathione Axis. Cancer Res. 2021, 81, 1896–1908. [Google Scholar] [CrossRef]
  97. Liu, Q.; Jin, Z.; Xu, Z.; Yang, H.; Li, L.; Li, G.; Li, F.; Gu, S.; Zong, S.; Zhou, J.; et al. Antioxidant effects of ginkgolides and bilobalide against cerebral ischemia injury by activating the Akt/Nrf2 pathway in vitro and in vivo. Cell Stress Chaperones 2019, 24, 441–452. [Google Scholar] [CrossRef]
  98. Ross, D.; Siegel, D. The diverse functionality of NQO1 and its roles in redox control. Redox Biol. 2021, 41, 101950. [Google Scholar] [CrossRef]
  99. Bersuker, K.; Hendricks, J.M.; Li, Z.; Magtanong, L.; Ford, B.; Tang, P.H.; Roberts, M.A.; Tong, B.; Maimone, T.J.; Zoncu, R.; et al. The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature 2019, 575, 688–692. [Google Scholar] [CrossRef]
  100. Doll, S.; Freitas, F.P.; Shah, R.; Aldrovandi, M.; da Silva, M.C.; Ingold, I.; Grocin, A.G.; da Silva, T.N.X.; Panzilius, E.; Scheel, C.H.; et al. FSP1 is a glutathione-independent ferroptosis suppressor. Nature 2019, 575, 693–698. [Google Scholar] [CrossRef]
  101. Avcı, B.; Günaydın, C.; Güvenç, T.; Yavuz, C.K.; Kuruca, N.; Bilge, S.S. Idebenone Ameliorates Rotenone-Induced Parkinson’s Disease in Rats Through Decreasing Lipid Peroxidation. Neurochem. Res. 2021, 46, 513–522. [Google Scholar] [CrossRef] [PubMed]
  102. Yan, A.; Liu, Z.; Song, L.; Wang, X.; Zhang, Y.; Wu, N.; Lin, J.; Liu, Y.; Liu, Z. Idebenone Alleviates Neuroinflammation and Modulates Microglial Polarization in LPS-Stimulated BV2 Cells and MPTP-Induced Parkinson’s Disease Mice. Front. Cell Neurosci. 2018, 12, 529. [Google Scholar] [CrossRef]
  103. Cleren, C.; Yang, L.; Lorenzo, B.; Calingasan, N.Y.; Schomer, A.; Sireci, A.; Wille, E.J.; Beal, M.F. Therapeutic effects of coenzyme Q10 (CoQ10) and reduced CoQ10 in the MPTP model of Parkinsonism. J. Neurochem. 2008, 104, 1613–1621. [Google Scholar] [CrossRef]
  104. Mischley, L.K.; Allen, J.; Bradley, R. Coenzyme Q10 deficiency in patients with Parkinson’s disease. J. Neurol. Sci. 2012, 318, 72–75. [Google Scholar] [CrossRef]
  105. Bian, Y.; Chen, Y.; Wang, X.; Cui, G.; Ung, C.O.L.; Lu, J.H.; Cong, W.; Tang, B.; Lee, S.M. Oxyphylla A ameliorates cognitive deficits and alleviates neuropathology via the Akt-GSK3beta and Nrf2-Keap1-HO-1 pathways in vitro and in vivo murine models of Alzheimer’s disease. J. Adv. Res. 2021, 34, 1–12. [Google Scholar] [CrossRef] [PubMed]
  106. Wang, L.; Cai, X.; Shi, M.; Xue, L.; Kuang, S.; Xu, R.; Qi, W.; Li, Y.; Ma, X.; Zhang, R.; et al. Identification and optimization of piperine analogues as neuroprotective agents for the treatment of Parkinson’s disease via the activation of Nrf2/keap1 pathway. Eur. J. Med. Chem. 2020, 199, 112385. [Google Scholar] [CrossRef] [PubMed]
  107. Kraft, V.A.N.; Bezjian, C.T.; Pfeiffer, S.; Ringelstetter, L.; Muller, C.; Zandkarimi, F.; Merl-Pham, J.; Bao, X.; Anastasov, N.; Kossl, J.; et al. GTP Cyclohydrolase 1/Tetrahydrobiopterin Counteract Ferroptosis through Lipid Remodeling. ACS Cent. Sci. 2020, 6, 41–53. [Google Scholar] [CrossRef]
  108. Nasser, A.; Moller, L.B. GCH1 variants, tetrahydrobiopterin and their effects on pain sensitivity. Scand. J. Pain. 2014, 5, 121–128. [Google Scholar] [CrossRef]
  109. Ding, X.-S.; Gao, L.; Han, Z.; Eleuteri, S.; Shi, W.; Shen, Y.; Song, Z.-Y.; Su, M.; Yang, Q.; Qu, Y.; et al. Ferroptosis in Parkinson’s disease: Molecular mechanisms and therapeutic potential. Ageing Res. Rev. 2023, 91, 102077. [Google Scholar] [CrossRef]
  110. Zheng, Q.; Ma, P.; Yang, P.; Zhai, S.; He, M.; Zhang, X.; Tu, Q.; Jiao, L.; Ye, L.; Feng, Z.; et al. Alpha lipoic acid ameliorates motor deficits by inhibiting ferroptosis in Parkinson’s disease. Neurosci. Lett. 2023, 810, 137346. [Google Scholar] [CrossRef]
  111. Li, Q.M.; Xu, T.; Zha, X.Q.; Feng, X.W.; Zhang, F.Y.; Luo, J.P. Buddlejasaponin IVb ameliorates ferroptosis of dopaminergic neuron by suppressing IRP2-mediated iron overload in Parkinson’s disease. J. Ethnopharmacol. 2024, 319, 117196. [Google Scholar] [CrossRef]
  112. Devos, D.; Labreuche, J.; Rascol, O.; Corvol, J.C.; Duhamel, A.; Delannoy, P.G.; Poewe, W.; Compta, Y.; Pavese, N.; Růžička, E.; et al. Trial of Deferiprone in Parkinson’s Disease. N. Engl. J. Med. 2022, 387, 2045–2055. [Google Scholar] [CrossRef]
  113. Zeng, X.; An, H.; Yu, F.; Wang, K.; Zheng, L.; Zhou, W.; Bao, Y.; Yang, J.; Shen, N.; Huang, D. Benefits of Iron Chelators in the Treatment of Parkinson’s Disease. Neurochem. Res. 2021, 46, 1239–1251. [Google Scholar] [CrossRef] [PubMed]
  114. Ye, Z.; Li, C.; Liu, S.; Liang, H.; Feng, J.; Lin, D.; Chen, Y.; Peng, S.; Bu, L.; Tao, E.; et al. Dl-3-n-butylphthalide activates Nrf2, inhibits ferritinophagy, and protects MES23.5 dopaminergic neurons from ferroptosis. Chem.-Biol. Interact. 2023, 382, 110604. [Google Scholar] [CrossRef] [PubMed]
  115. Tourville, A.; Viguier, S.; González-Lizárraga, F.; Tomas-Grau, R.H.; Ramirez, P.; Brunel, J.M.; Pereira, M.D.S.; Del-Bel, E.; Chehin, R.; Ferrié, L.; et al. Rescue of Dopamine Neurons from Iron-Dependent Ferroptosis by Doxycycline and Demeclocycline and Their Non-Antibiotic Derivatives. Antioxidants 2023, 12, 575. [Google Scholar] [CrossRef] [PubMed]
  116. Jiang, T.; Chu, J.; Chen, H.; Cheng, H.; Su, J.; Wang, X.; Cao, Y.; Tian, S.; Li, Q. Gastrodin Inhibits H2O2-Induced Ferroptosis through Its Antioxidative Effect in Rat Glioma Cell Line C6. Biol. Pharm. Bull. 2020, 43, 480–487. [Google Scholar] [CrossRef]
  117. Xi, J.; Zhang, Z.; Wang, Z.; Wu, Q.; He, Y.; Xu, Y.; Ding, Z.; Zhao, H.; Da, H.; Zhang, F.; et al. Hinokitiol functions as a ferroptosis inhibitor to confer neuroprotection. Free Radic. Biol. Med. 2022, 190, 202–215. [Google Scholar] [CrossRef]
  118. Fan, W.; Zhou, J. Icariside II suppresses ferroptosis to protect against MPP+-Induced Parkinson’s disease through Keap1/Nrf2/GPX4 signaling. Chin. J. Physiol. 2023, 66, 437–445. [Google Scholar] [CrossRef]
  119. Mansour, H.M.; Mohamed, A.F.; Khattab, M.M.; El-Khatib, A.S. Lapatinib ditosylate rescues motor deficits in rotenone-intoxicated rats: Potential repurposing of anti-cancer drug as a disease-modifying agent in Parkinson’s disease. Eur. J. Pharmacol. 2023, 954, 175875. [Google Scholar] [CrossRef]
  120. Li, M.; Zhang, J.; Jiang, L.; Wang, W.; Feng, X.; Liu, M.; Yang, D. Neuroprotective effects of morroniside from Cornus officinalis sieb. Et zucc against Parkinson’s disease via inhibiting oxidative stress and ferroptosis. BMC Complement. Med. Ther. 2023, 23, 218. [Google Scholar] [CrossRef]
  121. Wang, L.; An, H.; Yu, F.; Yang, J.; Ding, H.; Bao, Y.; Xie, H.; Huang, D. The neuroprotective effects of paeoniflorin against MPP+-induced damage to dopaminergic neurons via the Akt/Nrf2/GPX4 pathway. J. Chem. Neuroanat. 2022, 122, 102103. [Google Scholar] [CrossRef] [PubMed]
  122. Mansour, H.M.; Mohamed, A.F.; Khattab, M.M.; El-Khatib, A.S. Pazopanib ameliorates rotenone-induced Parkinsonism in rats by suppressing multiple regulated cell death mechanisms. Food Chem. Toxicol. 2023, 181, 114069. [Google Scholar] [CrossRef] [PubMed]
  123. Yue, M.; Wei, J.; Chen, W.; Hong, D.; Chen, T.; Fang, X. Neurotrophic Role of the Next-Generation Probiotic Strain L. lactis MG1363-pMG36e-GLP-1 on Parkinson’s Disease via Inhibiting Ferroptosis. Nutrients 2022, 14, 4886. [Google Scholar] [CrossRef] [PubMed]
  124. Lin, Z.H.; Liu, Y.; Xue, N.J.; Zheng, R.; Yan, Y.Q.; Wang, Z.X.; Li, Y.L.; Ying, C.Z.; Song, Z.; Tian, J.; et al. Quercetin Protects against MPP+/MPTP-Induced Dopaminergic Neuron Death in Parkinson’s Disease by Inhibiting Ferroptosis. Oxidative Med. Cell. Longev. 2022, 2022, 7769355. [Google Scholar] [CrossRef]
  125. Liu, T.; Wang, P.; Yin, H.; Wang, X.; Lv, J.; Yuan, J.; Zhu, J.; Wang, Y. Rapamycin reverses ferroptosis by increasing autophagy in MPTP/MPP+-induced models of Parkinson’s disease. Neural Regen. Res. 2023, 18, 2514–2519. [Google Scholar] [CrossRef]
  126. Yu, X.; Yang, Y.; Zhang, B.; Han, G.; Yu, J.; Yu, Q.; Zhang, L. Ketone Body β-Hydroxybutyric Acid Ameliorates Dopaminergic Neuron Injury Through Modulating Zinc Finger Protein 36/Acyl-CoA Synthetase Long-Chain Family Member Four Signaling Axis-Mediated Ferroptosis. Neuroscience 2023, 509, 157–172. [Google Scholar] [CrossRef]
Figure 1. Ca2+ signaling and homeostasis in neuron. Schematic diagram of a neuron illustrating several related proteins and pathways affecting Ca2+. Ca2+ enters cells via Cav1.2, Cav1.3, α-synuclein-permeable pore, NMDA-R, and Orai1. The endoplasmic reticulum is the main intracellular Ca2+ reservoir, and its calcium regulation is affected by Orai1 and STIM. Calcium exchange between endoplasmic reticulum and mitochondria is mainly through MAM. Calcium ions in the ER are released directly by IP3R from the ER into the cytoplasm, and then into the mitochondria via VDAC1, a calcium channel on the outer mitochondrial membrane. Calcium overload caused by VDAC1 oligomerization and MCU opening opens mPTP and leads to cell apoptosis.
Figure 1. Ca2+ signaling and homeostasis in neuron. Schematic diagram of a neuron illustrating several related proteins and pathways affecting Ca2+. Ca2+ enters cells via Cav1.2, Cav1.3, α-synuclein-permeable pore, NMDA-R, and Orai1. The endoplasmic reticulum is the main intracellular Ca2+ reservoir, and its calcium regulation is affected by Orai1 and STIM. Calcium exchange between endoplasmic reticulum and mitochondria is mainly through MAM. Calcium ions in the ER are released directly by IP3R from the ER into the cytoplasm, and then into the mitochondria via VDAC1, a calcium channel on the outer mitochondrial membrane. Calcium overload caused by VDAC1 oligomerization and MCU opening opens mPTP and leads to cell apoptosis.
Brainsci 14 00088 g001
Figure 2. Ferroptosis-suppressing pathways. The input of iron begins with TFR1, and Fe3+ is reduced to Fe2+ by STEAP3. Excess Fe2+ directly catalyze PUFA to generate lipid-free radicals through Fenton reaction, resulting in cell membrane destruction. Glutathione synthesis begins when cystine enters the cell via system XC- and is then synthesized in the presence of GSH, TRR1, GCL, and GS. GPX4 oxidizes glutathione and reduces PL-OOH associated with iron death to an PL-OH. FSP1 reduces CoQ10 under the action of NAD(P)H. CoQ10 and its reduced form Coq10-H2 remove free radicals on the surface of the membrane. BH4 traps lipid peroxidation free radicals and promotes the synthesis of CoQ10. The synthesis of BH4 requires the participation of GCH1, while the recovery and reuse of BH4 requires the participation of DHFR.
Figure 2. Ferroptosis-suppressing pathways. The input of iron begins with TFR1, and Fe3+ is reduced to Fe2+ by STEAP3. Excess Fe2+ directly catalyze PUFA to generate lipid-free radicals through Fenton reaction, resulting in cell membrane destruction. Glutathione synthesis begins when cystine enters the cell via system XC- and is then synthesized in the presence of GSH, TRR1, GCL, and GS. GPX4 oxidizes glutathione and reduces PL-OOH associated with iron death to an PL-OH. FSP1 reduces CoQ10 under the action of NAD(P)H. CoQ10 and its reduced form Coq10-H2 remove free radicals on the surface of the membrane. BH4 traps lipid peroxidation free radicals and promotes the synthesis of CoQ10. The synthesis of BH4 requires the participation of GCH1, while the recovery and reuse of BH4 requires the participation of DHFR.
Brainsci 14 00088 g002
Table 1. Medicine targeting ferroptosis and its mechanism in the last five years.
Table 1. Medicine targeting ferroptosis and its mechanism in the last five years.
MedicineMechanism and Function
Alpha lipoic acidAntioxidant and iron chelator; regulates iron metabolism and mitigating ferroptosis through the SIRT1/Nrf2 signaling pathway [110].
Buddlejasaponin IvbSuppressed IRP2 (iron responsive element binding protein 2)-mediated iron overload [111].
DeferiproneIron chelator; inhibits pathological toxicity of α-syn in a mouse model of sporadic PD [112].
DesferrioxamineIron chelator; chelates irons [113].
Dl-3-n-butylphthalideRegulates FTH (ferritin) expression, promotes Nrf2 nuclear translocation, and inhibits NCOA4-mediated ferritinophagy [114].
Doxycycline and DemeclocyclinePrevent intracellular oxidative stress and mitochondrial membrane depolarization [115].
GastrodinAntioxidant; increases the protein expression of Nrf2, GPX4, ferroportin-1 (FPN1), and HO-1 [116].
HinokitiolAntioxidant and iron chelator; chelates irons and activates cytoprotective transcription factor Nrf2 [117].
Icariside IIAntioxidant; activates Keap1/Nrf2/GPX4 signaling [118].
IdebenoneInhibits the decrease of expression of NAD(P)H dehydrogenase, decreases the levels of the lipid peroxidation products, and increases the expression of GPx-4 [101].
LapatinibActivates GPX4/GSH/NRF2 axis; inhibits oxidative markers, including iron, TfR1, PTGS2, and 4-HNE; and suppresses p-EGFR/c-SRC/PKCβII/PLC-γ/ACSL-4 pathway [119].
MorronisideAntioxidant; activates the Nrf2/ARE signaling pathway to protect dopaminergic neurons from ferroptosis in PD [120].
PaeoniflorinAntioxidant; activates the Akt/Nrf2/Gpx4 pathway [121].
PazopanibTargets HSP90/CDC37 and its multiple RCD mechanisms [122].
Probiotic Strain L. lactis MG1363-pMG36e-GLP-1Antioxidants and FSP1; activate the Keap1/Nrf2/GPX4 signaling pathway to down-regulate ACSL4 and up-regulate FSP1 to suppress ferroptosis [123].
QuercetinAntioxidant; inhibits ferroptosis by activating the Nrf2 protein [124].
RapamycinAutophagy inducer; inhibits ferroptosis by activating autophagy [125].
β-hydroxybutyrateAlleviates oxidative stress and ferroptosis via modulating ZFP36/ACSL4 axis [126].
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wang, J.; Zhao, J.; Zhao, K.; Wu, S.; Chen, X.; Hu, W. The Role of Calcium and Iron Homeostasis in Parkinson’s Disease. Brain Sci. 2024, 14, 88. https://doi.org/10.3390/brainsci14010088

AMA Style

Wang J, Zhao J, Zhao K, Wu S, Chen X, Hu W. The Role of Calcium and Iron Homeostasis in Parkinson’s Disease. Brain Sciences. 2024; 14(1):88. https://doi.org/10.3390/brainsci14010088

Chicago/Turabian Style

Wang, Ji, Jindong Zhao, Kunying Zhao, Shangpeng Wu, Xinglong Chen, and Weiyan Hu. 2024. "The Role of Calcium and Iron Homeostasis in Parkinson’s Disease" Brain Sciences 14, no. 1: 88. https://doi.org/10.3390/brainsci14010088

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop