Next Article in Journal
Analysis of Changes in the Expression of Selected Genes from the ABC Family in Patients with Triple-Negative Breast Cancer
Next Article in Special Issue
Mechanical Regulation of Redox Balance via the Induction of the PIN1/NRF2/ARE Axis in Pancreatic Cancer
Previous Article in Journal
Metal Complexes as Promising Matrix Metalloproteinases Regulators
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Unscrambling the Role of Redox-Active Biometals in Dopaminergic Neuronal Death and Promising Metal Chelation-Based Therapy for Parkinson’s Disease

by
Alfredo Gonzalez-Alcocer
1,
Ana Patricia Duarte-Jurado
1,
Adolfo Soto-Dominguez
1,
Maria de Jesus Loera-Arias
1,
Eliud Enrique Villarreal-Silva
2,
Odila Saucedo-Cardenas
1,
Roberto Montes de Oca-Luna
1,
Aracely Garcia-Garcia
1,* and
Humberto Rodriguez-Rocha
1,*
1
Departamento de Histologia, Facultad de Medicina, Universidad Autonoma de Nuevo Leon, Francisco I. Madero S/N, Mitras Centro, Monterrey 64460, Mexico
2
Servicio de Neurocirugía y Terapia Endovascular Neurológica, Hospital Universitario, Dr. Jose Eleuterio Gonzalez, Monterrey 64460, Mexico
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(2), 1256; https://doi.org/10.3390/ijms24021256
Submission received: 29 November 2022 / Revised: 28 December 2022 / Accepted: 30 December 2022 / Published: 9 January 2023
(This article belongs to the Special Issue Programmed Cell Death and Oxidative Stress)

Abstract

:
Biometals are all metal ions that are essential for all living organisms. About 40% of all enzymes with known structures require biometals to function correctly. The main target of damage by biometals is the central nervous system (CNS). Biometal dysregulation (metal deficiency or overload) is related to pathological processes. Chronic occupational and environmental exposure to biometals, including iron and copper, is related to an increased risk of developing Parkinson’s disease (PD). Indeed, biometals have been shown to induce a dopaminergic neuronal loss in the substantia nigra. Although the etiology of PD is still unknown, oxidative stress dysregulation, mitochondrial dysfunction, and inhibition of both the ubiquitin–proteasome system (UPS) and autophagy are related to dopaminergic neuronal death. Herein, we addressed the involvement of redox-active biometals, iron, and copper, as oxidative stress and neuronal death inducers, as well as the current metal chelation-based therapy in PD.

1. Introduction

Metals play essential biological functions in all living organisms. The human body requires minimal amounts of metallic elements to maintain a healthy development of physiological functions. However, metal levels outside the normal range may lead to the development of pathologies. An organism’s deficiency or excess of essential metals leads to severe biological alterations. However, not all metals are essential or beneficial to organisms; some nonessential and potentially toxic metals might be capable of causing undesirable effects on the genome, glycolysis, Krebs cycle, oxidative phosphorylation, and carbohydrate, lipid, protein, and amino acid metabolism [1]. Only biometals, including iron, copper, zinc, manganese, molybdenum, sodium, potassium, calcium, chromium, and cobalt, are indispensable for life. About 40% of all enzymes with known structures require biometals to function correctly [2]. These biometals are not limited to enzymatic activities but also play structural, electrostatic, energetic, and transport functions. Biometals directly affect by targeting biomolecules (DNA, lipids, proteins) (Figure 1), organelles, cells, tissues, organs, and the biological micro- and macro-environment, as they become part of it [3,4]. Living cells possess a redox metabolism where oxidation-reduction (redox) reactions occur in fundamental processes of redox regulation, collectively termed “redox signaling” and “redox control” [5]. Several studies performed on biological systems have shown that redox-active metals, including iron, copper, cobalt, chromium, and manganese, can undergo redox cycling reactions and produce reactive free radicals, also termed reactive oxygen species (ROS) or reactive nitrogen species (RNS) by upregulating Haber–Weiss and Fenton reactions and generating damage to cells [6].
The brain contains some of the highest iron, copper, zinc, and manganese concentrations in the human body [7]. These metals participate in synaptic transmission, myelinogenesis, energy production, and regulation of oxidative stress. Many biochemical processes rely on metals to transfer electrons via redox chemistry, neuronal excitation, protein structure, and enzymatic function [8].
The average human young brain consumes 20% of the oxygen taken in through respiration [9]. Because of high oxygen demand and cell complexity, high metal levels diffuse to the central nervous system (CNS) [10]. Therefore, the CNS is susceptible to metal damage [11]. Alterations in Fe, Cu, Zn, and Mn levels and distribution are associated with Parkinson’s Disease (PD) [12].
This review discusses the role of biologically essential redox-active biometals, iron, and copper, as oxidative stress and neuronal death inducers and the current metal chelation-based therapy in PD.

2. Metals, Parkinson’s Disease, and Oxidative Stress

Humans are regularly exposed to electromagnetic radiation, pollutants, and cellular metabolism byproducts that generate free radicals. Free radicals have an unpaired electron in their outer orbit [13,14], and oxygen radicals are involved in many cellular biochemical activities, such as signal transduction and gene transcription [15]. The most common cellular free radicals are hydroxyl (OH), superoxide anion (O2•–), and nitric monoxide (NO). Even some other species, such as hydrogen peroxide (H2O2) and peroxynitrite (ONOO), are highly reactive but are not free radicals; however, they can generate free radicals [16]. Mammalian cells produce free radicals and ROS as byproducts through physiological and biochemical processes, primarily due to aerobic metabolism [17]. Because of this, cells have an effective antioxidant defense involving glutathione, arginine, vitamins E, C, and A, and antioxidant enzymes to regulate ROS generation [18].
Overproduction of reactive species can cause oxidative damage to biomolecules (lipids, proteins, DNA), leading to chronic diseases such as atherosclerosis, cancer, diabetes, rheumatoid arthritis, myocardial infarction, chronic inflammation, and cardiovascular and neurodegenerative diseases in humans [19].
Oxidative and nitrative stress in the mesencephalon, where dopaminergic neurons are located, is one of the main factors related to PD pathogenesis [20,21]. Dopamine is susceptible to auto-oxidization, producing toxic semiquinone species, H2O2, and a small amount of the neurotoxin 6-hydroxydopamine [22,23]. A study performed in post-mortem idiopathic PD brains and neurologically healthy adult brains matched by age showed that glutathione peroxidase activity was slightly but significantly reduced in several brain areas, including substantia nigra in PD brain samples [24]. A decrease in mitochondrial complex I activity has been reported in the substantia nigra of PD patients, which ultimately increases oxidative stress [25].
Chronic occupational and environmental exposure to metals, including iron and copper, increases the risk of developing PD [26]. Abnormally high iron and zinc levels have been detected in the substantia nigra of PD patients’ post-mortem samples. A deficiency or an overload of metals may influence the appearance of this disorder [27]. Metal ions and byproducts of the electron transport chain play a crucial role in forming intracellular free radicals leading to oxidative stress, where the imbalance of free radicals, antioxidants, and detoxifying enzymes occurs [28]. As a result, oxidatively modified molecules such as nucleotides, proteins, and lipids accumulate in the cellular compartment provoking dysfunction [29]. Therefore, the lack of control of the defense system, especially in sensitive cells such as neurons, eventually will lead to cell death [28,30].
Lewy bodies (LB) are abnormal protein deposits containing α-synuclein associated with ubiquitin and tau, among other proteins (Figure 2). Multivalent metal ions such as iron, copper, and manganese increase α-synuclein fibril formation by inducing conformational changes [31,32,33]. Oxidative modifications and phosphorylation may engage both protein activity and half-life. Phosphorylated proteins strongly bind to certain metals [34,35,36].
Although non-enzymatic antioxidants have shown neuroprotective effects in PD experimental models, they have failed to reproduce this protection in clinical trials [37]. Therefore, it is imperative to understand the mechanisms involved in PD to explore diverse potential therapeutics more efficiently.

3. How Cells Die: Classical Mechanisms of Cell Death

Before discussing the role of biometals in neuronal death, it is worth summarizing the classical mechanisms of cell death. According to morphological, biochemical, and genetic characteristics, cell death is classified into three major types: apoptosis, necrosis, and autophagy. However, another section will discuss the latter, and additional cell death mechanisms will briefly be described.
Apoptosis is well-characterized and known as programmed cell death type I (PCD type I). Its morphological changes include cell shrinkage, chromatin condensation (pyknosis), nuclear fragmentation (karyorrhexis), loss of plasma membrane integrity, and plasma membrane blebbing forming apoptotic bodies [38].
Necrosis, usually described as an accidental and uncontrollable mechanism, shows a substantial gain in cell volume (oncosis), swollen organelles, and disruption of the plasma membrane with the subsequent intracellular content release [39]. However, according to recent findings and following the guidelines of the Nomenclature Committee on Cell Death 2018, this classification has been updated, and mitochondrial permeability transition (MPT)-driven necrosis and necroptosis are now included, among other cell death subroutines [38]. MPT-driven necrosis is a regulated cell death induced by cell microenvironment disturbances that alter the inner mitochondrial membrane impermeability and is cyclophilin D (CYPD)-dependent [40,41,42]. Necroptosis is a regulated process activated by the recognition of extracellular and intracellular triggers through death receptors, and mixed lineage kinase domain-like pseudokinase (MLKL), receptor-interacting protein kinase 1 (RIPK1), and RIPK3 are crucial signaling molecules [43]. Ferroptosis is an intracellular iron- and ROS-dependent cell death mechanism that involves strong lipid peroxidation, glutathione peroxidase-4 depletion, glutathione imbalance, and mitochondria morphological alterations, including increased mitochondria membrane electron density, decreased or loss of cristae, and outer mitochondrial membrane rupture [44].

4. Redox-Active Metals’ Role in Dopaminergic Neuronal Death

4.1. Iron

Iron is the most abundant metal on Earth, and almost all organisms have evolved to use this ubiquitous transition metal [45]. This metal is essential for the human body’s proper functioning. Iron is vital for oxygen transport (bound to hemoglobin), oxidative phosphorylation (bound to cytochrome C), neurotransmitter synthesis, myelin formation, and regulation of the biosynthesis of proteins such as ferritin and transferrin receptor (to store or mobilize iron) through iron-response proteins binding to iron-responsive elements at mRNA level [46,47,48].
In contrast, unbound iron causes cell toxicity as it can trigger a series of highly oxidative and toxic reactions [49]; this occurs when the iron concentrations exceed the binding capacity of transferrin [50]. Iron has a wide range of oxidative states, Fe2+ (ferrous) and Fe3+ (ferric) being the most common in biological environments [51] (Figure 3). Fe2+ binding to proteins is very unstable [52]. Iron’s neurotoxic effect has been related to the divalent metal ion transporter 1 (DMT1) overexpression, which imports iron into the cell, and can also be inhibited by H-ferritin [53]. Moreover, S-nitrosylation (SNO) of DMT1 cysteine thiol enhances Mn2+ and Fe2+ uptake [54]. Additionally, SNO-DMT1 has been detected in the post-mortem substantia nigra of PD patients [54]. The redox state of iron determines its role in cytotoxic reactions [55].
In a healthy brain, iron is distributed in a specific pattern by region and cell type. It is abundant in the substantia nigra and the basal ganglia, which are rich in dopaminergic neurons [56].
For over four decades, changes in iron and ferritin levels have been described in the brain of PD patients [27]. Several mechanisms are associated with iron-induced dopaminergic cell death, including (1) Fenton redox-reactions producing hydroxyl radicals [57]; (2) DA oxidative deamination, which is catalyzed by monoamine oxidase B (MAO-B) and regulated by Fe2+ and Fe3+ [58,59]; (3) 6-hydroxydopamine neurotoxic formation through DA metabolites reaction with iron and H2O2 [60]; and (4) increased rate of iron-induced α-synuclein fibril formation [33]. Iron takes part in the Fenton reaction producing free radicals; Fe2+ reacts with H2O2 or lipid peroxides to generate Fe3+, hydroxyl ion (OH), and OH or lipid radicals, which may lead to oxidative damage of macromolecules [47,61]. The Haber–Weiss reaction is where hydroxyl ion and hydroxyl radical are generated from the reaction of H2O2 and O2•– catalyzed by iron [62]. Additionally, the formation and accumulation of OH lead to activation of the mitochondrial permeability transition pore (mPTP), which temporarily opens and increases ROS, provoking long-lasting activation and cell death. The latter is triggered by a decrease of ATP production, mitochondrial swelling, and rupture of the outer mitochondrial membrane, with subsequent release of mitochondrial death factors such as cytochrome C to the cytosolic compartment activating cell death by apoptosis [63]. Moreover, p53 is involved in mitochondrial dysfunction and oxidative stress mediated by Fe2+ in neuronal synaptic terminals [64].
Interestingly, lysosomes contain a redox-active iron pool derived from iron-rich macromolecules and cellular organelles, such as ferritin and mitochondria [65,66]. Most iron is found in a non-redox active form bound to ferritin. Ferritin degradation inside lysosomes during autophagy may be an intracellular redox-active iron source [67]. Next, H2O2 diffuses into lysosomes and reacts with the iron species through the Fenton and Fenton-like reactions, resulting in hydroxyl radical generation [65]. Intriguingly, defective mitochondria and lysosomes may promote RIPK1 activation, making cells susceptible to necroptosis [68]. Recently, necroptosis’s partial contribution to iron-mediated toxicity was demonstrated by using iron chelator deferoxamine (DFO) and the necroptosis inhibitor necrostatin 1 (NEC-1), significantly reducing cell death rates in the glutamate-induced model in vitro [69].
Iron accumulation in the brain is age-dependent, having the lowest levels at birth and a marked increase with age [70]. Brain regions associated with motor function have a high concentration of iron [70], and its levels in the whole brain are around 35.6–54.2 µg/g [71]. Its accumulation in the brain affects neurons. Exposure of neurons to iron induces oxidative stress, causing lipid peroxidation and DNA damage, which leads to caspase-dependent apoptotic cell death [72].
Several key mediators of ferroptosis have previously been implicated in PD pathogenesis. The SNpc is an iron-rich, dopamine (DA)-producing midbrain nucleus, which probably explains why it has a high risk of suffering neuronal death [73,74], mainly when iron accumulates, representing a PD feature [27,75]. Iron produces hydroxyl radicals with subsequent dopamine oxidation, likely contributing to an oxidative environment that increases the loss of nigral dopaminergic neurons in PD patients [76]. Moreover, genetic disorders that result in brain iron dyshomeostasis often cause Parkinsonism [77,78,79], demonstrating increased iron’s potential to contribute to PD pathogenesis. Indeed, mutations in several proteins involved in iron transport, increasing iron uptake and decreasing its export, are linked to PD. Mutant forms of transferrin, a critical protein for neuronal iron uptake, are associated with increased susceptibility to PD [80,81]. These data suggest that the iron uptake mechanism is overactive in these patients resulting in increased neuronal iron accumulation.
Conversely, mutations in transferrin receptor 2 (TfR2) [81] are associated with a protective effect in PD, potentially due to reduced iron uptake. Neuronal iron export occurs via a transmembrane ion channel, ferroportin [82], and the Alzheimer’s disease (AD)-implicated amyloid precursor protein (APP) stabilizes ferroportin expression on the membrane to promote iron efflux [83]. In contrast, loss of APP membrane function results in impaired iron efflux and consequent neuronal iron retention [84]. Indeed, several rare variants of APP predispose individuals to PD, and several studies of familial AD indicate APP mutations are associated with Parkinsonism and LB formation [85,86,87,88]. Deficits in iron export in PD were further identified in the substantia nigra, with a significant depletion in APP expression levels independently of cell loss and an 80% decrease in ceruloplasmin (CP) activity [74,89]. Ceruloplasmin also has a ferrous oxidase activity and enables iron export by converting Fe2+ to Fe3+ [90], which is then bound to and removed by transferrin. Several point mutations in the CP-encoding gene are significantly associated with PD [91] and Parkinsonism [78,92], indicating that CP-mediated iron homeostasis is also likely involved in PD pathogenesis.
Vitamin C, or ascorbic acid, has been shown to improve the absorption of Levodopa in some PD patients with poor Levodopa bioavailability [93]. However, it might be toxic as it loses one electron and forms an ascorbate radical (Asc•–). The electron can reduce metal ions such as iron and copper. The acidic extracellular environment favors the reduction of protein-centered metal, represented as Fe3+ reduction to Fe2+. Subsequently, Fe2+ donates an electron to O2 forming O2•– with subsequent dismutation to H2O2 [94]. Moreover, vitamin C administration may aggravate PD progression due to the possible peroxidation of Fe2+ bound to Asc•–. Therefore, combined vitamin C therapy for ROS scavenging and an iron chelator to sequester the metal may be a promising PD treatment option, reducing the toxicity induced by DA-derived quinones [95].

Current Status of Iron Chelation Therapeutic Effect on PD Patients

Iron chelation is a successful treatment for iron accumulation-based systemic pathologies, such as cardiomyopathy associated with hemochromatosis [96] and thalassemia [97]. Since iron accumulation in the brain has been linked to PD development, this metal chelation emerges as a promising therapeutic target [90,91,92].
Studies in animal PD models have shown that iron chelation reduces and stops the pathological accumulation of α-synuclein [98] and decreases oxidative stress [99,100] when administered focally, intranasally, and even orally. In addition, some sophisticated translational studies (Table 1) demonstrated that iron chelation therapy decreases labile iron and oxidative stress in vitro and in vivo, ending with a pilot clinical study that reported symptomatic improvement in PD patients [101].
These promising results justify using iron chelating agents in clinical trials. In these studies, deferiprone (DFP) doses of 20 mg/kg/day and 30 mg/kg/day were well-tolerated by patients. In addition, decreasing iron levels in the dentate and caudate nuclei were detected by MRI, with a consequent improvement in the Unified Parkinson Disease Rating Scale (UPDRS) scores [102]. These improvements were observed more markedly in patients with low CP activity [103]. However, these promising results were not reproduced when a more extensive study was carried out in patients without dopaminergic treatment, where the disability increased over 36 weeks, suggesting that iron accumulation is only an early temporary compensatory mechanism to increase dopamine synthesis; however, in the long term, it worsens cell death [104].
Despite this, the initial results are still promising. Nevertheless, doubt remains regarding the effect of long-term iron chelation at the systemic level, as it could affect the circulating white blood cell number and iron homeostasis in cerebral glial cells, which is essential for processes such as myelin production by oligodendrocytes [105,106]. This controversy confronts us with the challenge of finding ways to modulate iron, not affecting other cells. Interestingly, lactoferrin, a cationic iron-binding glycoprotein, can cross the blood–brain barrier through transferrin receptor 1-mediated transcytosis on the surface of the brain capillary endothelial cells [107]. Lactoferrin protected from dopaminergic neuronal loss in a PD model induced with the neurotoxins MPP+ (1-methyl-4-phenylpyridinium) /MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) by upregulation of brain-derived neurotrophic factor (BDNF), hypoxia-inducible factor 1α (HIF-1α), along with extracellular regulated protein kinases (ERK) and cAMP response element-binding protein (CREB) activation, and decreased phosphorylation of c-Jun N-terminal kinase (JNK) and P38 kinase [108]. Moreover, pretreatment with human lactoferrin positively affected the nigrostriatal system recovery after acute exposure to MPTP [109]. These results propose a new strategy for the regulation of cerebral iron homeostasis.
Table 1. Summary of deferiprone iron chelation effect on PD clinical trials.
Table 1. Summary of deferiprone iron chelation effect on PD clinical trials.
Study DesignClinical TrialSubjects
Male:Female
(m:f)
OutcomesReference
Randomized, double-blinded, placebo-controlled clinical trialPhase 222 subjects:
•8 placebo
(m:f) 3:5
•7 DFP 20 mg/kg/day
(m:f) 4:3
•7 DFP 30 mg/kg/day
(m:f) 5:2
Brain iron chelation by DFP therapy was well-tolerated; there was an associated reduced dentate and caudate nucleus iron content with a trend for improvement in motor-UPDRS scores and quality of life, not statistical significance.[102]
Randomized, placebo-controlled clinical trial Phase 140 subjects:
•21 early start
DFP 30 mg/kg/day
(m:f) 12:9
•19 delayed start
DFP 30 mg/kg/day
(m:f) 13:6
Most DFP-treated patients displayed clinical and radiological improvements. Those with lower CP activity appeared to respond better to iron chelation.[103]
Randomized, double-blind, placebo-controlled, parallel-group, single-center trialPhase 240 subjects:
•21 early start
DFP 30 mg/kg/day
(m:f) 12:9
•19 delayed start
DFP 30 mg/kg/day
(m:f) 13:6
SN iron levels and UPDRS motor scores were reduced in patients with higher CP-ferroxidase activity in serum and CSF. [101,110]
A multicentric, parallel-group, placebo-controlled, randomized clinical trialPhase 2372 subjects:
•186 placebo
•186 DFP
30 mg/kg/day
DFP without dopaminergic treatment worsened the handicap at the PD diagnosis time compared with placebo over 36 weeks. This finding provides evidence that the iron accumulation in the nigrostriatal pathway is a powerful short-term compensatory mechanism for increasing dopamine synthesis but possibly at the expense of long-term worsening iron-related cell death.[104]

4.2. Copper

Copper is a trace element that constitutes 70 parts per million of the Earth’s crust. However, it is an essential micronutrient found in small amounts in tissues and cells, with a high concentration in the kidney, liver, and brain [111].
This metal functions as an essential cofactor and is required for structural and catalytic proprieties of more than 30 necessary enzymes; among them are ceruloplasmin, cytochrome oxidase, lysine oxidase, dopamine-hydroxylase, ascorbate oxidase, tyrosinase, and Cu/Zn SOD [112]. In living organisms, copper is mainly found oxidized (Cu2+) and reduced (Cu+) [113].
After the liver, the brain is the organ that accumulates the most significant amount of copper, reaching contents between 2.9 to 10 µg/g wet weight [114], and it is distributed differently in each region. The regions with the highest concentrations of copper are the substantia nigra, cerebellum, hippocampus, and hypothalamus [115]. Copper plays a crucial role in essential processes in CNS, such as brain development [116], antioxidant defense, synaptic transmission [117], and acting as an enzyme cofactor with oxidoreductase activity [112].
Notwithstanding, like any redox-active metal, copper becomes toxic when its intracellular accumulation is excessive, facilitating the formation of ROS and apoptotic processes [118] (Figure 4). When the cell is exposed to oxidative stress or copper, DNA damage and p53 expression are induced [119,120]. Furthermore, p53 undergoes oligomerization and phosphorylation to be translocated into the nucleus to induce genes such as BAX (BCL2 Associated X) and PUMA (p53 upregulated modulator of apoptosis) and subsequent release of cytochrome C into the cytosol to initiate apoptosis [121,122]. Therefore, copper plays a vital role in many diseases, such as Menkes disease, where copper is abnormally low in the brain. Conversely, in Wilson’s disease, the damage is caused by an excess of copper stored in brain tissue. Moreover, some neurodegenerative disorders such as AD, amyotrophic lateral sclerosis (ALS), prion disease, and PD have been linked to copper dyshomeostasis [123]. Increased copper levels have been reported in the cerebrospinal fluid and blood of PD patients [124,125]. However, a recent meta-analysis reported decreased copper levels in the substantia nigra of PD patients compared to healthy age-matched subjects [126]. Nevertheless, it has been demonstrated that chronic occupational exposure to copper increases the risk of developing PD [127,128,129].
Copper toxicity affects the basal ganglia and frontal cortex inducing Parkinson-like symptoms and cognitive deficits. The mechanism of copper-mediated toxicity includes cell cycle arrest via the upregulation of p21 (Cyclin Dependent Kinase Inhibitor 1A), reprimo (involved in regulating p53-dependent G2 arrest of the cell cycle and coded by RPRM gene), stathmin (microtubule destabilizing protein coded by STMN1 gene), and Tp53INP1 (Tumor Protein P53 Inducible Nuclear Protein 1). Additionally, stat-3 (Signal Transducer and Activator of Transcription 3), hsp70 (Heat Shock Protein 70), and hsp27 (Heat Shock Protein 27) are increased in an attempt to survive. Finally, p53-dependent and independent apoptosis are triggered, where IGFBP-6 (insulin-like growth factor binding protein-6), glutathione peroxidase, BCL-2, RB-1, PUMA, and several members of the redox-active PIG family of proteins, play a role [130]. Moreover, copper binding to α-synuclein increases oxidative stress and α-synuclein phosphorylation and accelerates the protein aggregation process [34,131,132]. In the neuroblastoma cell line SK-N-SH with dopaminergic phenotype, copper transporter protein 1 (Ctr1) overexpression led to intracellular glutathione depletion and potentiated the caspase-3-dependent-cell death induced by copper, indicating that copper’s toxicity is due to alterations in its intracellular homeostasis. In addition, copper-induced oxidative stress was primarily localized in the cytosol, and Nrf2 was upregulated to mediate an antioxidant response. In addition, copper increased protein ubiquitination, AMPK-Ulk1 signaling, p62, and Atg5-dependent autophagy as a protective mechanism [133]. The release of redox-active copper ions from copper-binding proteins and its binding to thiol or amine groups of cysteinyl and histidinyl residues of globular proteins, including enzymes, may result in conformational changes leading to its inactivation [134,135].
Paradoxically, some clinical trials report decreased circulating copper levels in PD patients compared to healthy controls [136], which may occur, because by binding to ceruloplasmin, copper stimulates ferroxidase activity and participates in iron homeostasis. Therefore, low levels of copper can indirectly generate toxicity by altering iron concentrations [137].
Preclinical in vivo PD models showed that the chelation of heavy metals such as copper improved motor and non-motor deficits after MPTP intoxication [138], which has also been reproduced in other models of neurodegenerative diseases such as AD [139]. However, there is no evidence of any effects of copper chelators in PD patients in clinical trials.
Since the strong chelation of metals can have systemic effects, alternative therapies targeting metal dyshomeostasis are critical. Recently, metal-protein attenuating compounds (MPACs) have emerged as promising therapeutic strategies. MPACs are moderate chelators that disrupt specific, abnormal metal-protein interactions [140] (Table 2). Under physiological conditions, MPACs bind to metal ions with a high affinity by competing with the metal-binding proteins to avoid their oligomerization and prevent the formation of metal-catalyzed ROS [141].
A novel ligand, 1-methyl-1H-imidazole-2-carboxaldehyde isonicotinoyl hydrazone (X1INH), attenuated abnormal copper+/copper2+-α-synuclein interactions and affected protein aggregation in a cellular model of synucleinopathy where inclusions were smaller and less compact [142]. Moreover, a moderate metal-binding compound, 8-hydroxyquinoline-2-carboxaldehyde isonicotinoyl hydrazine (INHHQ), was non-toxic to human neuroglioma H4 cells and was able to disrupt anomalous copper-α-synuclein interactions, probably by sequestering the metal ions. Importantly, INHHQ crosses the BBB and can be detected in rats’ brains as late as 24 h after its IP administration. After 48 h, brain clearance is complete, but INHHQ remains in the liver even 72 h after acute exposure. [143]. The effect of D-penicillamine, a relatively specific copper chelator, was assessed in the MPTP-induced PD mice model, showing a modest effect in preventing MPTP-induced striatal dopamine depletion [144,145]. In contrast, another study in the MPTP model detected a decrease in copper content in the striatum and midbrain, suggesting that its neurotoxicity is independent of copper [146]. Clioquinol (CQ, 5-chloro-7-iodo-8-quinolinol) can bind to the metal ions Fe3+, Cu2+, and Zn2+, which is why it plays a critical role in PD. Likewise, CQ remarkably improved the motor and non-motor deficits based on reduced iron content and ROS level in the SN [138]. HPCIH, HPCFur (pyridine-2-carboxaldehyde isonicotinoyl hydrazone, pyridine-2-car-boxaldehyde 2-furoyl hydrazone) has the ability to bind to Cu2+, which is why it has been linked to neurodegenerative diseases derived from misfolded prion proteins. HPCFur has a protective effect on methionine and histidine oxidation, which is related to physiological and pathological aging [147]. Therefore, physiopathologically relevant PD models reproducing the disease as in humans are urgent as the current models may not reproduce all characteristics of the disease and may lead to it through different mechanisms.
Table 2. Summary of metal-protein attenuating compounds (MPACs) effects on neurodegenerative diseases.
Table 2. Summary of metal-protein attenuating compounds (MPACs) effects on neurodegenerative diseases.
Metal-Protein Attenuating CompoundMetal Ions BindingNeurodegenerative DiseaseOutcomesReference
X1INH
1-methyl-1H-imidazole-2-carboxaldehyde isonicotinoyl hydrazone
Cu+
Cu2+
Parkinson’s diseaseX1INH increased the number of smaller, less compact inclusions in a well-established model of α-Syn aggregation.[142]
INHHQ
8-hydroxyquinoline-2-carboxaldehyde isonicotinoyl hydrazone INHHQ
Cu2+ Zn2+Alzheimer’s disease, Parkinson’s diseaseINHHQ can disrupt, in vitro, anomalous copper-α-Syn interactions through a mechanism probably involving metal ions sequestering.[141]
Clioquinol (CQ)
5-chloro-7-iodo-8-quinolinol
Fe3+, Cu2+ Zn2+Parkinson’s diseaseCQ remarkably improved the motor and non-motor deficits based on reduced iron content and ROS level in the SN. [138]
HPCIH, HPCFur
pyridine-2-carboxaldehyde isonicotinoyl hydrazone
pyridine-2-car-boxaldehyde 2-furoyl hydrazone
Cu2+Misfolded prion proteinHPCFur has a protective effect on methionine and histidine oxidation, which is related to physiological and pathological aging.[147]

5. Concluding Remarks

Nearly 40% of our proteins need a biometal as a cofactor to fulfill their function. Therefore, it is crucial to understand the transition metals’ role in health and disease because their dyshomeostasis (deficiency or overloading) is closely related to different disorders and mainly to neurodegeneration. Research in this field has found that biometals are tightly regulated because a narrow unbalance provokes diseases such as PD. Therefore, understanding the complexity of the interaction between transition metals and proteins can shed light on possible neurodegeneration biomarkers for preventing neuronal cell death. Hopefully, neurodegeneration may be achieved by either supplementing transition metals when there is a deficiency or using chelating agents to avoid metal overload that induces neuronal cell death in PD.
The lack of success of metal-chelating agents in PD clinical trials is partly due to the lack of suitable models for its preclinical testing. So far, there is no animal model combining chronic exposure to metal ions emulating environmental and occupational exposure and aging, which may reflect how humans develop PD, as it is a multifactor disorder. PD animal models, like many others, are short-term, which has enormously contributed to our understanding of the mechanism implicated. However, it is time to combine the aging process with other risk factors, including environmental and occupational exposure, to develop more accurate PD animal models so that their translation into clinical trials leads to a higher probability of success. Why have we not used long-term models? The answer is simple; the main limitation is the time, followed by the increased resources required to feed, treat, and take care of mice. However, this approach may be fundamental to improving our chances of success in clinical trials.

Funding

This research was funded by Programa de Apoyo a la Investigacion Cientifica y Tecnologica (PAICyT) 254-CS-2022 (G.-G.A.) and 215-CS-2022 (R.-R.H.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

G.-A.A. (No. CVU: 791892) and D.-J.A.P. (No. CVU: 856246) received a scholarship from the National Council of Science and Technology (Consejo Nacional de Ciencia y Tecnologia, CONACYT).

Conflicts of Interest

The authors declare that there is no conflict of interest regarding the publication of this paper.

References

  1. Ortega, R.; Carmona, A. Neurotoxicity of Environmental Metal Toxicants: Special Issue. Toxics 2022, 10, 382. [Google Scholar] [CrossRef]
  2. Andreini, C.; Bertini, I.; Cavallaro, G.; Holliday, G.L.; Thornton, J.M. Metal ions in biological catalysis: From enzyme databases to general principles. J. Biol. Inorg. Chem. 2008, 13, 1205–1218. [Google Scholar] [CrossRef]
  3. Shanker, A.K. Mode of Action and Toxicity of Trace Elements. In Trace Elements as Contaminants and Nutrients: Consequences in Ecosystems and Human Health; John Wiley & Sons, Inc.: Hoboken, NJ, USA, 2008. [Google Scholar]
  4. Pospíšil, P.; Prasad, A.; Rác, M. Mechanism of the Formation of Electronically Excited Species by Oxidative Metabolic Processes: Role of Reactive Oxygen Species. Biomolecules 2019, 9, 258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Lennicke, C.; Cochemé, H.M. Redox metabolism: ROS as specific molecular regulators of cell signaling and function. Mol. Cell. 2021, 81, 3691–3707. [Google Scholar] [CrossRef] [PubMed]
  6. Jomova, K.; Baros, S.; Valko, M. Redox active metal-induced oxidative stress in biological systems. Transit. Met. Chem. 2012, 37, 127–134. [Google Scholar] [CrossRef]
  7. DeBenedictis, C.A.; Raab, A.; Ducie, E.; Howley, S.; Feldmann, J.; Grabrucker, A.M. Concentrations of Essential Trace Metals in the Brain of Animal Species-A Comparative Study. Brain Sci. 2020, 10, 460. [Google Scholar] [CrossRef]
  8. Chang, C.J. Searching for harmony in transition-metal signaling. Nat. Chem. Biol. 2015, 11, 744–747. [Google Scholar] [CrossRef]
  9. Watts, M.E.; Pocock, R.; Claudianos, C. Brain Energy and Oxygen Metabolism: Emerging Role in Normal Function and Disease. Front. Mol. Neurosci. 2018, 11, 216. [Google Scholar] [CrossRef] [Green Version]
  10. Que, E.L.; Domaille, D.W.; Chang, C.J. Metals in neurobiology: Probing their chemistry and biology with molecular imaging. Chem. Rev. 2008, 108, 1517–1549. [Google Scholar] [CrossRef]
  11. Borisova, T. Nervous System Injury in Response to Contact With Environmental, Engineered and Planetary Micro- and Nano-Sized Particles. Front. Physiol. 2018, 9, 728. [Google Scholar] [CrossRef]
  12. Ullah, I.; Zhao, L.; Hai, Y.; Fahim, M.; Alwayli, D.; Wang, X.; Li, H. Metal elements and pesticides as risk factors for Parkinson’s disease—A review. Toxicol. Rep. 2021, 8, 607–616. [Google Scholar] [CrossRef] [PubMed]
  13. McCord, J.M. The evolution of free radicals and oxidative stress. Am. J. Med. 2000, 108, 652–659. [Google Scholar] [CrossRef] [PubMed]
  14. Martemucci, G.; Costagliola, C.; Mariano, M.; D’Andrea, L.; Napolitano, P.; D’Alessandro, A.G. Free Radical Properties, Source and Targets, Antioxidant Consumption and Health. Oxygen 2022, 2, 48–78. [Google Scholar] [CrossRef]
  15. Pizzino, G.; Irrera, N.; Cucinotta, M.; Pallio, G.; Mannino, F.; Arcoraci, V.; Squadrito, F.; Altavilla, D.; Bitto, A. Oxidative Stress: Harms and Benefits for Human Health. Oxidative Med. Cell. Longev. 2017, 2017, 8416763. [Google Scholar] [CrossRef] [Green Version]
  16. Gilgun-Sherki, Y.; Melamed, E.; Offen, D. Oxidative stress induced-neurodegenerative diseases: The need for antioxidants that penetrate the blood brain barrier. Neuropharmacology 2001, 40, 959–975. [Google Scholar] [CrossRef]
  17. Navarro-Yepes, J.; Burns, M.; Anandhan, A.; Khalimonchuk, O.; del Razo, L.M.; Quintanilla-Vega, B.; Pappa, A.; Panayiotidis, M.; Franco, R. Oxidative stress, redox signaling, and autophagy: Cell death versus survival. Antioxid. Redox Signal. 2014, 21, 66–85. [Google Scholar] [CrossRef] [Green Version]
  18. Kurutas, E.B. The importance of antioxidants which play the role in cellular response against oxidative/nitrosative stress: Current state. Nutr. J. 2016, 15, 71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Forman, H.J.; Zhang, H. Targeting oxidative stress in disease: Promise and limitations of antioxidant therapy. Nat. Rev. Drug Discov. 2021, 20, 689–709. [Google Scholar] [CrossRef] [PubMed]
  20. Stykel, M.G.; Ryan, S.D. Nitrosative stress in Parkinson’s disease. NPJ Parkinsons Dis. 2022, 8, 104. [Google Scholar] [CrossRef] [PubMed]
  21. Hartmann, A. Postmortem studies in Parkinson’s disease. Dialogues Clin. Neurosci. 2004, 6, 281–293. [Google Scholar] [CrossRef]
  22. Farzam, A.; Chohan, K.; Strmiskova, M.; Hewitt, S.J.; Park, D.S.; Pezacki, J.P.; Özcelik, D. A functionalized hydroxydopamine quinone links thiol modification to neuronal cell death. Redox Biol. 2020, 28, 101377. [Google Scholar] [CrossRef] [PubMed]
  23. Cobley, J.N.; Fiorello, M.L.; Bailey, D.M. 13 reasons why the brain is susceptible to oxidative stress. Redox Biol. 2018, 15, 490–503. [Google Scholar] [CrossRef]
  24. Kish, S.J.; Morito, C.; Hornykiewicz, O. Glutathione peroxidase activity in Parkinson’s disease brain. Neurosci. Lett. 1985, 58, 343–346. [Google Scholar] [CrossRef] [PubMed]
  25. Chen, C.; Turnbull, D.M.; Reeve, A.K. Mitochondrial Dysfunction in Parkinson’s Disease-Cause or Consequence? Biology 2019, 8, 38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Ball, N.; Teo, W.-P.; Chandra, S.; Chapman, J. Parkinson’s Disease and the Environment. Front. Neurol. 2019, 10, 218. [Google Scholar] [CrossRef] [Green Version]
  27. Dexter, D.T.; Wells, F.R.; Lees, A.J.; Agid, F.; Agid, Y.; Jenner, P.; Marsden, C.D. Increased nigral iron content and alterations in other metal ions occurring in brain in Parkinson’s disease. J. Neurochem. 1989, 52, 1830–1836. [Google Scholar] [CrossRef]
  28. Nakamura, K.; Bindokas, V.P.; Marks, J.D.; Wright, D.A.; Frim, D.M.; Miller, R.J.; Kang, U.J. The selective toxicity of 1-methyl-4-phenylpyridinium to dopaminergic neurons: The role of mitochondrial complex I and reactive oxygen species revisited. Mol. Pharm. 2000, 58, 271–278. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Obata, T. Dopamine efflux by MPTP and hydroxyl radical generation. J. Neural Transm. 2002, 109, 1159–1180. [Google Scholar] [CrossRef]
  30. Hoffman, C.; Aballay, A. Role of neurons in the control of immune defense. Curr. Opin. Immunol. 2019, 60, 30–36. [Google Scholar] [CrossRef]
  31. Lesage, S.; Brice, A. Parkinson’s disease: From monogenic forms to genetic susceptibility factors. Hum. Mol. Genet. 2009, 18, R48–R59. [Google Scholar] [CrossRef]
  32. Shults, C.W. Lewy bodies. Proc. Natl. Acad. Sci. USA 2006, 103, 1661–1668. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Uversky, V.N.; Li, J.; Fink, A.L. 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, 44284–44296. [Google Scholar] [CrossRef] [Green Version]
  34. Liu, L.L.; Franz, K.J. Phosphorylation-dependent metal binding by alpha-synuclein peptide fragments. J. Biol. Inorg. Chem. 2007, 12, 234–247. [Google Scholar] [CrossRef]
  35. Liu, L.L.; Franz, K.J. Phosphorylation of an alpha-synuclein peptide fragment enhances metal binding. J. Am. Chem. Soc. 2005, 127, 9662–9663. [Google Scholar] [CrossRef]
  36. Garcia-Garcia, A.; Rodriguez-Rocha, H.; Madayiputhiya, N.; Pappa, A.; Panayiotidis, M.I.; Franco, R. Biomarkers of protein oxidation in human disease. Curr. Mol. Med. 2012, 12, 681–697. [Google Scholar] [CrossRef]
  37. Duarte-Jurado, A.P.; Gopar-Cuevas, Y.; Saucedo-Cardenas, O.; de Jesus Loera-Arias, M.; Montes-de-Oca-Luna, R.; Garcia-Garcia, A.; Rodriguez-Rocha, H. Antioxidant Therapeutics in Parkinson’s Disease: Current Challenges and Opportunities. Antioxidants 2021, 10, 453. [Google Scholar] [CrossRef] [PubMed]
  38. Galluzzi, L.; Vitale, I.; Aaronson, S.A.; Abrams, J.M.; Adam, D.; Agostinis, P.; Alnemri, E.S.; Altucci, L.; Amelio, I.; Andrews, D.W.; et al. Molecular mechanisms of cell death: Recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 2018, 25, 486–541. [Google Scholar]
  39. D’Arcy, M.S. Cell death: A review of the major forms of apoptosis, necrosis and autophagy. Cell Biol. Int. 2019, 43, 582–592. [Google Scholar] [CrossRef] [PubMed]
  40. Izzo, V.; Bravo San Pedro, J.M.; Sica, V.; Kroemer, G.; Galluzzi, L. Mitochondrial Permeability Transition: New Findings and Persisting Uncertainties. Trends Cell Biol. 2016, 26, 655–667. [Google Scholar] [CrossRef] [PubMed]
  41. Baines, C.P.; Kaiser, R.A.; Purcell, N.H.; Blair, N.S.; Osinska, H.; Hambleton, M.A.; Brunskill, E.W.; Sayen, M.R.; Gottlieb, R.A.; Dorn, G.W., II.; et al. Loss of cyclophilin D reveals a critical role for mitochondrial permeability transition in cell death. Nature 2005, 434, 658–662. [Google Scholar] [CrossRef] [PubMed]
  42. Nakagawa, T.; Shimizu, S.; Watanabe, T.; Yamaguchi, O.; Otsu, K.; Yamagata, H.; Inohara, H.; Kubo, T.; Tsujimoto, Y. Cyclophilin D-dependent mitochondrial permeability transition regulates some necrotic but not apoptotic cell death. Nature 2005, 434, 652–658. [Google Scholar] [CrossRef] [PubMed]
  43. Bertheloot, D.; Latz, E.; Franklin, B.S. Necroptosis, pyroptosis and apoptosis: An intricate game of cell death. Cell. Mol. Immunol. 2021, 18, 1106–1121. [Google Scholar] [CrossRef] [PubMed]
  44. Čepelak, I.; Dodig, S.; Dodig, D. Ferroptosis: Regulated cell death. Arh. Za Hig. Rada I Toksikol. 2020, 71, 99–109. [Google Scholar] [CrossRef]
  45. Dlouhy, A.C.; Outten, C.E. The iron metallome in eukaryotic organisms. Met. Ions Life Sci. 2013, 12, 241–278. [Google Scholar] [PubMed] [Green Version]
  46. Casey, J.L.; Hentze, M.W.; Koeller, D.M.; Caughman, S.W.; Rouault, T.A.; Klausner, R.D.; Harford, J.B. Iron-responsive elements: Regulatory RNA sequences that control mRNA levels and translation. Science 1988, 240, 924–928. [Google Scholar] [CrossRef] [PubMed]
  47. Hentze, M.W.; Muckenthaler, M.U.; Andrews, N.C. Balancing acts: Molecular control of mammalian iron metabolism. Cell 2004, 117, 285–297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Arosio, P.; Elia, L.; Poli, M. Ferritin, cellular iron storage and regulation. IUBMB Life 2017, 69, 414–422. [Google Scholar] [CrossRef] [Green Version]
  49. Bresgen, N.; Eckl, P.M. Oxidative stress and the homeodynamics of iron metabolism. Biomolecules 2015, 5, 808–847. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Siah, C.W.; Ombiga, J.; Adams, L.A.; Trinder, D.; Olynyk, J.K. Normal iron metabolism and the pathophysiology of iron overload disorders. Clin. Biochem. Rev. 2006, 27, 5–16. [Google Scholar]
  51. Núñez, M.T.; Urrutia, P.; Mena, N.; Aguirre, P.; Tapia, V.; Salazar, J. Iron toxicity in neurodegeneration. Biometals 2012, 25, 761–776. [Google Scholar] [CrossRef]
  52. Cotruvo, J.J.A.; Stubbe, J. Metallation and mismetallation of iron and manganese proteins in vitro and in vivo: The class I ribonucleotide reductases as a case study. Metallomics 2012, 4, 1020–1036. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Zhu, W.; Li, X.; Xie, W.; Luo, F.; Kaur, D.; Andersen, J.K.; Jankovic, J.; Le, W. Genetic iron chelation protects against proteasome inhibition-induced dopamine neuron degeneration. Neurobiol. Dis. 2010, 37, 307–313. [Google Scholar] [CrossRef] [PubMed]
  54. Liu, C.; Zhang, C.-W.; Lo, S.Q.; Ang, S.T.; Chew, K.C.M.; Yu, D.; Chai, B.H.; Tan, B.; Tsang, F.; Tai, Y.K.; et al. S-Nitrosylation of Divalent Metal Transporter 1 Enhances Iron Uptake to Mediate Loss of Dopaminergic Neurons and Motoric Deficit. J. Neurosci. 2018, 38, 8364–8377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Yarjanli, Z.; Ghaedi, K.; Esmaeili, A.; Rahgozar, S.; Zarrabi, A. Iron oxide nanoparticles may damage to the neural tissue through iron accumulation, oxidative stress, and protein aggregation. BMC Neurosci. 2017, 18, 51. [Google Scholar] [CrossRef] [Green Version]
  56. Ramos, P.; Santos, A.; Pinto, N.R.; Mendes, R.; Magalhães, T.; Almeida, A. Iron levels in the human brain: A post-mortem study of anatomical region differences and age-related changes. J. Trace Elem. Med. Biol. 2014, 28, 13–17. [Google Scholar] [CrossRef]
  57. Linert, W.; Jameson, G.N. Redox reactions of neurotransmitters possibly involved in the progression of Parkinson’s Disease. J. Inorg. Biochem. 2000, 79, 319–326. [Google Scholar] [CrossRef]
  58. Youdim, M.B.; Riederer, P.F. A review of the mechanisms and role of monoamine oxidase inhibitors in Parkinson’s disease. Neurology 2004, 63 (Suppl. 2), S32-5. [Google Scholar] [CrossRef]
  59. Lu, H.; Chen, J.; Huang, H.; Zhou, M.; Zhu, Q.; Yao, S.Q.; Chai, Z.; Hu, Y. Iron modulates the activity of monoamine oxidase B in SH-SY5Y cells. Biometals 2017, 30, 599–607. [Google Scholar] [CrossRef]
  60. Napolitano, A.; Manini, P.; d’Ischia, M. Oxidation chemistry of catecholamines and neuronal degeneration: An update. Curr. Med. Chem. 2011, 18, 1832–1845. [Google Scholar] [CrossRef] [Green Version]
  61. Barbusinski, K. Toxicity of Industrial Wastewater Treated by Fenton’s Reagent. Pol. J. Environ. Stud. 2005, 14, 11–16. [Google Scholar]
  62. Kehrer, J.P. The Haber-Weiss reaction and mechanisms of toxicity. Toxicology 2000, 149, 43–50. [Google Scholar] [CrossRef] [PubMed]
  63. Zorov, D.B.; Juhaszova, M.; Sollott, S.J. Mitochondrial reactive oxygen species (ROS) and ROS-induced ROS release. Physiol. Rev. 2014, 94, 909–950. [Google Scholar] [CrossRef] [Green Version]
  64. Gilman, C.P.; Chan, S.L.; Guo, Z.; Zhu, X.; Greig, N.; Mattson, M.P. p53 is present in synapses where it mediates mitochondrial dysfunction and synaptic degeneration in response to DNA damage, and oxidative and excitotoxic insults. Neuromolecular Med. 2003, 3, 159–172. [Google Scholar] [CrossRef]
  65. Koskenkorva-Frank, T.S.; Weiss, G.; Koppenol, W.H.; Burckhardt, S. The complex interplay of iron metabolism, reactive oxygen species, and reactive nitrogen species: Insights into the potential of various iron therapies to induce oxidative and nitrosative stress. Free Radic. Biol. Med. 2013, 65, 1174–1194. [Google Scholar] [CrossRef] [PubMed]
  66. Dixon, S.J.; Stockwell, B.R. The role of iron and reactive oxygen species in cell death. Nat. Chem. Biol. 2014, 10, 9–17. [Google Scholar] [CrossRef] [PubMed]
  67. Latunde-Dada, G.O. Ferroptosis: Role of lipid peroxidation, iron and ferritinophagy. Biochim. Et Biophys. Acta Gen. Subj. 2017, 1861, 1893–1900. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Iannielli, A.; Bido, S.; Folladori, L.; Segnali, A.; Cancellieri, C.; Maresca, A.; Massimino, L.; Rubio, A.; Morabito, G.; Caporali, L.; et al. Pharmacological Inhibition of Necroptosis Protects from Dopaminergic Neuronal Cell Death in Parkinson’s Disease Models. Cell Rep. 2018, 22, 2066–2079. [Google Scholar] [CrossRef] [Green Version]
  69. Gonzalez, G.; Grúz, J.; D’Acunto, C.W.; Kaňovský, P.; Strnad, M. Cytokinin Plant Hormones Have Neuroprotective Activity in In Vitro Models of Parkinson’s Disease. Molecules 2021, 26, 361. [Google Scholar] [CrossRef]
  70. Ashraf, A.; Clark, M.; So, P.-W. The Aging of Iron Man. Front. Aging Neurosci. 2018, 10, 65. [Google Scholar] [CrossRef] [Green Version]
  71. McAllum, E.J.; Hare, D.J.; Volitakis, I.; McLean, C.A.; Bush, A.I.; Finkelstein, D.; Roberts, B.R. Regional iron distribution and soluble ferroprotein profiles in the healthy human brain. Prog. Neurobiol. 2020, 186, 101744. [Google Scholar] [CrossRef]
  72. Zhang, Z.; Wei, T.; Hou, J.; Li, G.; Yu, S.; Xin, W. Iron-induced oxidative damage and apoptosis in cerebellar granule cells: Attenuation by tetramethylpyrazine and ferulic acid. Eur. J. Pharm. 2003, 467, 41–47. [Google Scholar] [CrossRef]
  73. Hare, D.; Aror, M.; Jenkins, N.; Finkelstein, D.; Doble, P.; Bush, A. Is early-life iron exposure critical in neurodegeneration? Nat. Rev. Neurol. 2015, 11, 536–544. [Google Scholar] [CrossRef] [PubMed]
  74. Ayton, S.; Lei, P.; Hare, D.; Duce, J.; George, J.; Adlard, P.; McLean, C.; Rogers, J.; Cherny, R.; Finkelstein, D.; et al. Parkinson’s disease iron deposition caused by nitric oxide-induced loss of β-amyloid precursor protein. J. Neurosci. 2015, 35, 3591–3597. [Google Scholar] [CrossRef] [PubMed]
  75. Ayton, S.; Lei, P. Nigral iron elevation is an invariable feature of Parkinson’s disease and is a sufficient cause of neurodegeneration. Biomed Res. Int. 2014, 2014, 581256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Jenner, P.; Dexter, D.T.; Sian, J.; Schapira, A.H.; Marsden, C.D. 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, S82-7. [Google Scholar] [CrossRef]
  77. Costello, D.J.; Walsh, S.L.; Harrington, H.J.; Walsh, C.H. Concurrent hereditary haemochromatosis and idiopathic Parkinson’s disease: A case report series. J. Neurol. Neurosurg. Psychiatry 2004, 75, 631–633. [Google Scholar] [CrossRef]
  78. Miyajima, H.; Takahashi, Y.; Kono, S. Aceruloplasminemia, an inherited disorder of iron metabolism. Biometals 2003, 16, 205–213. [Google Scholar] [CrossRef]
  79. Nielsen, J.E.; Jensen, L.N.; Krabbe, K. Hereditary haemochromatosis: A case of iron accumulation in the basal ganglia associated with a parkinsonian syndrome. J. Neurol. Neurosurg. Psychiatry 1995, 59, 318–321. [Google Scholar] [CrossRef] [Green Version]
  80. Borie, C.; Gasparini, F.; Verpillat, P.; Bonnet, A.-M.; Agid, Y.; Hetet, G.; Brice, A.; Dürr, A.; Grandchamp, B. French Parkinson’s disease genetic study group. Association study between iron-related genes polymorphisms and Parkinson’s disease. J. Neurol. 2002, 249, 801–804. [Google Scholar] [CrossRef]
  81. Rhodes, S.; Buchanan, D.; Ahmed, I.; Taylor, K.; Loriot, M.-A.; Sinsheimer, J.; Bronstein, J.; Elbaz, A.; Mellick, G.; Rotter, J.; et al. Pooled analysis of iron-related genes in Parkinson’s disease: Association with transferrin. Neurobiol. Dis. 2014, 62, 172–178. [Google Scholar] [CrossRef] [Green Version]
  82. Donovan, A.; Lima, C.A.; Pinkus, J.L.; Pinkus, G.S.; Zon, L.I.; Robine, S.; Andrews, N.C. The iron exporter ferroportin/Slc40a1 is essential for iron homeostasis. Cell Metab. 2005, 1, 191–200. [Google Scholar] [CrossRef] [Green Version]
  83. Duce, J.A.; Tsatsanis, A.; Cater, M.A.; James, S.A.; Robb, E.; Wikhe, K.; Leong, S.L.; Perez, K.; Johanssen, T.; Greenough, M.A.; et al. Iron-export ferroxidase activity of β-amyloid precursor protein is inhibited by zinc in Alzheimer’s disease. Cell 2010, 142, 857–867. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Lei, P.; Ayton, S.; I Finkelstein, D.; Spoerri, L.; Ciccotosto, G.D.; Wright, D.K.; Wong, B.X.W.; A Adlard, P.; A Cherny, R.; Lam, L.Q.; et al. Tau deficiency induces parkinsonism with dementia by impairing APP-mediated iron export. Nat. Med. 2012, 18, 291–295. [Google Scholar] [CrossRef] [PubMed]
  85. Schulte, E.C.; Fukumori, A.; Mollenhauer, B.; Hor, H.; Arzberger, T.; Perneczky, R.; Kurz, A.; Diehl-Schmid, J.; Hüll, M.; Lichtner, P.; et al. Rare variants in β-Amyloid precursor protein (APP) and Parkinson’s disease. Eur. J. Hum. Genet. 2015, 23, 1328–1333. [Google Scholar] [CrossRef] [Green Version]
  86. Edwards-Lee, T.; Ringman, J.M.; Chung, J.; Werner, J.; Morgan, A.; Hyslop, P.S.G.; Thompson, P.; Dutton, R.; Mlikotic, A.; Rogaeva, E.; et al. An African American family with early-onset Alzheimer disease and an APP (T714I) mutation. Neurology 2005, 64, 377–379. [Google Scholar] [CrossRef] [PubMed]
  87. Halliday, G.; Brooks, W.; Arthur, H.; Creasey, H.; Broe, G. Further evidence for an association between a mutation in the APP gene and Lewy body formation. Neurosci. Lett. 1997, 227, 49–52. [Google Scholar] [CrossRef]
  88. Rosenberg, C.K.; Pericak-Vance, M.A.; Saunders, A.M.; Gilbert, J.R.; Gaskell, P.C.; Hulette, C.M. Lewy body and Alzheimer pathology in a family with the amyloid-beta precursor protein APP717 gene mutation. Acta Neuropathol. 2000, 100, 145–152. [Google Scholar] [CrossRef]
  89. Ayton, S.; Lei, P.; Duce, J.A.; Wong, B.X.; Sedjahtera, A.; Adlard, P.A.; Bush, A.I.; Finkelstein, D.I. Ceruloplasmin dysfunction and therapeutic potential for Parkinson’s disease. Ann. Neurol. 2013, 73, 554–559. [Google Scholar] [CrossRef]
  90. Osaki, S.; Johnson, D.A.; Frieden, E. The possible significance of the ferrous oxidase activity of ceruloplasmin in normal human serum. J. Biol. Chem. 1966, 241, 2746–2751. [Google Scholar] [CrossRef]
  91. Hochstrasser, H.; Bauer, P.; Walter, U.; Behnke, S.; Spiegel, J.; Csoti, I.; Zeiler, B.; Bornemann, A.; Pahnke, J.; Becker, G.; et al. Ceruloplasmin gene variations and substantia nigra hyperechogenicity in Parkinson’s disease. Neurology 2004, 63, 1912–1917. [Google Scholar] [CrossRef] [PubMed]
  92. Kohno, S.; Miyajima, H.; Takahashi, Y.; Inoue, Y. Aceruloplasminemia with a novel mutation associated with parkinsonism. Neurogenetics 2000, 2, 237–238. [Google Scholar] [CrossRef]
  93. Nagayama, H.; Hamamoto, M.; Ueda, M.; Nito, C.; Yamaguchi, H.; Katayama, Y. The effect of ascorbic acid on the pharmacokinetics of levodopa in elderly patients with Parkinson disease. Clin. Neuropharmacol. 2004, 27, 270–273. [Google Scholar] [CrossRef] [PubMed]
  94. Chen, Q.; Espey, M.G.; Sun, A.Y.; Lee, J.-H.; Krishna, M.C.; Shacter, E.; Choyke, P.L.; Pooput, C.; Kirk, K.L.; Buettner, G.R.; et al. Ascorbate in pharmacologic concentrations selectively generates ascorbate radical and hydrogen peroxide in extracellular fluid in vivo. Proc. Natl. Acad. Sci. USA 2007, 104, 8749–8754. [Google Scholar] [CrossRef]
  95. Sun, Y.; Pham, A.N.; Waite, T.D. The effect of vitamin C and iron on dopamine-mediated free radical generation: Implications to Parkinson’s disease. Dalton Trans 2018, 47, 4059–4069. [Google Scholar] [CrossRef] [PubMed]
  96. Aronow, W.S. Management of cardiac hemochromatosis. Arch. Med. Sci. 2018, 14, 560–568. [Google Scholar] [CrossRef]
  97. Taher, A.; Hershko, C.; Cappellini, M.D. Iron overload in thalassaemia intermedia: Reassessment of iron chelation strategies. Br. J. Haematol. 2009, 147, 634–640. [Google Scholar] [CrossRef]
  98. Febbraro, F.; Andersen, K.J.; Sanchez-Guajardo, V.; Tentillier, N.; Romero-Ramos, M. Chronic intranasal deferoxamine ameliorates motor defects and pathology in the α-synuclein rAAV Parkinson’s model. Exp. Neurol. 2013, 247, 45–58. [Google Scholar] [CrossRef] [PubMed]
  99. Dexter, D.T.; Statton, S.A.; Whitmore, C.; Freinbichler, W.; Weinberger, P.; Tipton, K.F.; Della Corte, L.; Ward, R.J.; Crichton, R.R. Clinically available iron chelators induce neuroprotection in the 6-OHDA model of Parkinson’s disease after peripheral administration. J. Neural. Transm. Vienna 2011, 118, 223–231. [Google Scholar] [CrossRef]
  100. Kaur, D.; Yantiri, F.; Rajagopalan, S.; Kumar, J.; Mo, J.Q.; Boonplueang, R.; Viswanath, V.; Jacobs, R.; Yang, L.; Beal, M.F.; et al. Genetic or pharmacological iron chelation prevents MPTP-induced neurotoxicity in vivo: A novel therapy for Parkinson’s disease. Neuron 2003, 37, 899–909. [Google Scholar] [CrossRef] [Green Version]
  101. Devos, D.; Moreau, C.; Devedjian, J.C.; Kluza, J.; Petrault, M.; Laloux, C.; Jonneaux, A.; Ryckewaert, G.; Garçon, G.; Rouaix, N. Targeting chelatable iron as a therapeutic modality in Parkinson’s disease. Antioxid. Redox Signal 2014, 21, 195–210. [Google Scholar] [CrossRef] [Green Version]
  102. Martin-Bastida, A.; Ward, R.J.; Newbould, R.; Piccini, P.; Sharp, D.; Kabba, C.; Patel, M.C.; Spino, M.; Connelly, J.; Tricta, F. 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. [Google Scholar] [CrossRef] [Green Version]
  103. Grolez, G.; Moreau, C.; Sablonnière, B.; Garçon, G.; Devedjian, J.C.; Meguig, S.; Gelé, P.; Delmaire, C.; Bordet, R.; Defebvre, L.; et al. Ceruloplasmin activity and iron chelation treatment of patients with Parkinson’s disease. BMC Neurol. 2015, 15, 74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. University Hospital, Lille; European Commission; ApoPharma. Conservative Iron Chelation as a Disease-Modifying Strategy in Parkinson’s Disease. 2016. Available online: https://ClinicalTrials.gov/show/NCT02655315 (accessed on 17 October 2022).
  105. 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]
  106. Ballas, S.K.; Zeidan, A.M.; Duong, V.H.; DeVeaux, M.; Heeney, M.M. A Systematic Review of the Effect of Iron Chelation Therapy on Survival in Patients with Sickle Cell Disease and β-Thalassemia. Blood 2017, 130, 3743. [Google Scholar]
  107. Li, Y.Q.; Guo, C. A Review on Lactoferrin and Central Nervous System Diseases. Cells 2021, 10, 1810. [Google Scholar] [CrossRef]
  108. Xu, S.-F.; Zhang, Y.-H.; Wang, S.; Pang, Z.-Q.; Fan, Y.-G.; Li, J.-Y.; Wang, Z.-Y.; Guo, C. Lactoferrin ameliorates dopaminergic neurodegeneration and motor deficits in MPTP-treated mice. Redox Biol. 2019, 21, 101090. [Google Scholar] [CrossRef]
  109. Kopaeva, M.Y.; Cherepov, A.B.; Nesterenko, M.V.; Zarayskaya, I.Y. Pretreatment with Human Lactoferrin Had a Positive Effect on the Dynamics of Mouse Nigrostriatal System Recovery after Acute MPTP Exposure. Biology 2021, 10, 24. [Google Scholar] [CrossRef] [PubMed]
  110. University Hospital, Lille. Efficacy and Safety of the Iron Chelator Deferiprone in Parkinson’s Disease. 2009. Available online: https://ClinicalTrials.gov/show/NCT00943748 (accessed on 17 October 2022).
  111. Focarelli, F.; Giachino, A.; Waldron, K.J. Copper microenvironments in the human body define patterns of copper adaptation in pathogenic bacteria. PLoS Pathog. 2022, 18, e1010617. [Google Scholar] [CrossRef] [PubMed]
  112. Tsang, T.; Davis, C.I.; Brady, D.C. Copper biology. Curr. Biol. 2021, 31, R421–R427. [Google Scholar] [CrossRef]
  113. Tapiero, H.; Townsend, D.M.; Tew, K.D. Trace elements in human physiology and pathology. Copper. Biomed Pharm. 2003, 57, 386–398. [Google Scholar] [CrossRef]
  114. Lutsenko, S.; Bhattacharjee, A.; Hubbard, A.L. Copper handling machinery of the brain. Metallomics 2010, 2, 596–608. [Google Scholar] [CrossRef] [PubMed]
  115. Tarnacka, B.; Jopowicz, A.; Maślińska, M. Copper, Iron, and Manganese Toxicity in Neuropsychiatric Conditions. Int. J. Mol. Sci. 2021, 22, 7820. [Google Scholar] [CrossRef] [PubMed]
  116. Amorós, R.; Murcia, M.; González, L.; Soler-Blasco, R.; Rebagliato, M.; Iñiguez, C.; Carrasco, P.; Vioque, J.; Broberg, K.; Levi, M.; et al. Maternal copper status and neuropsychological development in infants and preschool children. Int. J. Hyg. Environ. Health 2019, 222, 503–512. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Gaier, E.D.; Eipper, B.A.; Mains, R.E. Copper signaling in the mammalian nervous system: Synaptic effects. J. Neurosci. Res. 2013, 91, 2–19. [Google Scholar] [CrossRef] [PubMed]
  118. Gaetke, L.M.; Chow-Johnson, H.S.; Chow, C.K. Copper: Toxicological relevance and mechanisms. Arch. Toxicol. 2014, 88, 1929–1938. [Google Scholar] [CrossRef] [Green Version]
  119. Da, S.; Rashed, L. Oxidative stress and genotoxicity among workers exposed to copper in a factory for non-ferrous industry in Egypt. Egypt. J. Occup. Med. 2019, 43, 1–15. [Google Scholar] [CrossRef]
  120. Santos, S.; Silva, A.M.; Matos, M.; Monteiro, S.M.; Álvaro, A.R. Copper induced apoptosis in Caco-2 and Hep-G2 cells: Expression of caspases 3, 8 and 9, AIF and p53. Comp. Biochem. Physiol. Part C Toxicol. Pharm. 2016, 185–186, 138–146. [Google Scholar] [CrossRef]
  121. Macip, S.; Igarashi, M.; Berggren, P.; Yu, J.; Lee, S.W.; Aaronson, S.A. Influence of induced reactive oxygen species in p53-mediated cell fate decisions. Mol. Cell. Biol. 2003, 23, 8576–8585. [Google Scholar] [CrossRef] [Green Version]
  122. Narayanan, V.S.; Fitch, C.A.; Levenson, C.W. Tumor suppressor protein p53 mRNA and subcellular localization are altered by changes in cellular copper in human Hep G2 cells. J. Nutr. 2001, 131, 1427–1432. [Google Scholar] [CrossRef] [Green Version]
  123. Gromadzka, G.; Tarnacka, B.; Flaga, A.; Adamczyk, A. Copper Dyshomeostasis in Neurodegenerative Diseases-Therapeutic Implications. Int. J. Mol. Sci. 2020, 21, 9259. [Google Scholar] [CrossRef]
  124. Hozumi, I.; Hasegawa, T.; Honda, A.; Ozawa, K.; Hayashi, Y.; Hashimoto, K.; Yamada, M.; Koumura, A.; Sakurai, T.; Kimura, A.; et al. Patterns of levels of biological metals in CSF differ among neurodegenerative diseases. J. Neurol. Sci. 2011, 303, 95–99. [Google Scholar] [CrossRef] [PubMed]
  125. Pall, H.S.; Williams, A.C.; Blake, D.R.; Lunec, J.; Gutteridge, J.M.; Hall, M.; Taylor, A. Raised cerebrospinal-fluid copper concentration in Parkinson’s disease. Lancet 1987, 2, 238–241. [Google Scholar] [CrossRef] [PubMed]
  126. Genoud, S.; Senior, A.M.; Hare, D.J.; Double, K.L. Meta-Analysis of Copper and Iron in Parkinson’s Disease Brain and Biofluids. Mov. Disord. 2020, 35, 662–671. [Google Scholar] [CrossRef] [PubMed]
  127. Santner, A.; Uversky, V.N. Metalloproteomics and metal toxicology of α-synuclein. Metallomics 2010, 2, 378–392. [Google Scholar] [CrossRef]
  128. Gorell, J.M.; Johnson, C.C.; Rybicki, B.A.; Peterson, E.L.; Kortsha, G.X.; Brown, G.G.; Richardson, R.J. Occupational exposure to manganese, copper, lead, iron, mercury and zinc and the risk of Parkinson’s disease. Neurotoxicology 1999, 20, 239–247. [Google Scholar]
  129. Rybicki, B.A.; Johnson, C.C.; Uman, J.; Gorell, J.M. Parkinson’s disease mortality and the industrial use of heavy metals in Michigan. Mov. Disord. 1993, 8, 87–92. [Google Scholar] [CrossRef]
  130. VanLandingham, J.W.; Tassabehji, N.M.; Somers, R.C.; Levenson, C.W. Expression profiling of p53-target genes in copper-mediated neuronal apoptosis. Neuromolecular Med. 2005, 7, 311–324. [Google Scholar] [CrossRef]
  131. Lu, Y.; Prudent, M.; Fauvet, B.; Lashuel, H.A.; Girault, H.H. Phosphorylation of α-Synuclein at Y125 and S129 alters its metal binding properties: Implications for understanding the role of α-Synuclein in the pathogenesis of Parkinson’s Disease and related disorders. ACS Chem. Neurosci. 2011, 2, 667–675. [Google Scholar] [CrossRef] [Green Version]
  132. Miotto, M.C.; Rodriguez, E.E.; Valiente-Gabioud, A.A.; Torres-Monserrat, V.; Binolfi, A.; Quintanar, L.; Zweckstetter, M.; Griesinger, C.; Fernández, C.O. Site-specific copper-catalyzed oxidation of α-synuclein: Tightening the link between metal binding and protein oxidative damage in Parkinson’s disease. Inorg. Chem. 2014, 53, 4350–4358. [Google Scholar] [CrossRef]
  133. Anandhan, A.; Rodriguez-Rocha, H.; Bohovych, I.; Griggs, A.M.; Zavala-Flores, L.; Reyes-Reyes, E.M.; Seravalli, J.; Stanciu, L.A.; Lee, J.; Rochet, J.-C.; et al. Overexpression of alpha-synuclein at non-toxic levels increases dopaminergic cell death induced by copper exposure via modulation of protein degradation pathways. Neurobiol. Dis. 2015, 81, 76–92. [Google Scholar] [CrossRef] [Green Version]
  134. Letelier, M.E.; FaúnNdez, M.; Jara-Sandoval, J.; Molina-Berrã os, A.; Cortãés-Troncoso, J.; Aracena-Parks, P.; Marín-Catalán, R. Mechanisms underlying the inhibition of the cytochrome P450 system by copper ions. J. Appl. Toxicol. 2009, 29, 695–702. [Google Scholar] [CrossRef]
  135. Letelier, M.E.; Lepe, A.M.; Faúndez, M.; Salazar, J.; Marín, R.; Aracena, P.; Speisky, H. Possible mechanisms underlying copper-induced damage in biological membranes leading to cellular toxicity. Chem. Biol. Interact 2005, 151, 71–82. [Google Scholar] [CrossRef] [PubMed]
  136. Kim, M.J.; Oh, S.B.; Kim, J.; Kim, K.; Ryu, H.S.; Kim, M.S.; Ayton, S.; Bush, A.I.; Lee, J.Y.; Chung, S.J. Association of metals with the risk and clinical characteristics of Parkinson’s disease. Park. Relat Disord 2018, 55, 117–121. [Google Scholar] [CrossRef]
  137. Doguer, C.; Ha, J.H.; Collins, J.F. Intersection of Iron and Copper Metabolism in the Mammalian Intestine and Liver. Compr. Physiol. 2018, 8, 1433–1461. [Google Scholar]
  138. Shi, L.; Huang, C.; Luo, Q.; Xia, Y.; Liu, W.; Zeng, W.; Cheng, A.; Shi, R.; Zhengli, C. Clioquinol improves motor and non-motor deficits in MPTP-induced monkey model of Parkinson’s disease through AKT/mTOR pathway. Aging Albany NY 2020, 12, 9515–9533. [Google Scholar] [CrossRef]
  139. Zhao, J.; Shi, Q.; Tian, H.; Li, Y.; Liu, Y.; Xu, Z.; Robert, A.; Liu, Q.; Meunier, B. TDMQ20, a Specific Copper Chelator, Reduces Memory Impairments in Alzheimer’s Disease Mouse Models. ACS Chem. Neurosci. 2021, 12, 140–149. [Google Scholar] [CrossRef]
  140. Barnham, K.J.; Cherny, R.A.; Cappai, R.; Melov, S.; Masters, C.L.; Bush, A.I. Metal-Protein Attenuating Compounds (MPACs) for the Treatment of Alzheimer’s Disease. Drug Des. Rev.-Online Discontin. 2004, 1, 75–82. [Google Scholar]
  141. Deas, E.; Cremades, N.; Angelova, P.R.; Ludtmann, M.H.; Yao, Z.; Chen, S.; Horrocks, M.H.; Banushi, B.; Little, D.; Devine, M.J.; 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, 376–391. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Cukierman, D.S.; Lázaro, D.F.; Sacco, P.; Ferreira, P.R.; Diniz, R.; Fernández, C.O.; Outeirobef, T.F.; Nicolás, A. ReyX1INH, an improved next-generation affinity-optimized hydrazonic ligand, attenuates abnormal copper(I)/copper(II)-α-Syn interactions and affects protein aggregation in a cellular model of synucleinopathy. Dalton Trans. 2020, 49, 16252–16267. [Google Scholar] [CrossRef]
  143. Cukierman, D.S.; Pinheiro, A.B.; Castiñeiras-Filho, S.L.; da Silva, A.S.; Miotto, M.C.; De Falco, A.; de PRibeiro, T.; Maisonette, S.; da Cunha, A.L.; Hauser-Davis, R.A.; et al. A moderate metal-binding hydrazone meets the criteria for a bioinorganic approach towards Parkinson’s disease: Therapeutic potential, blood-brain barrier crossing evaluation and preliminary toxicological studies. J. Inorg. Biochem. 2017, 170, 160–168. [Google Scholar] [CrossRef]
  144. Youdim, M.B.; Grünblatt, E.; Mandel, S. The copper chelator, D-penicillamine, does not attenuate MPTP induced dopamine depletion in mice. J. Neural. Transm. Vienna 2007, 114, 205–209. [Google Scholar] [CrossRef] [PubMed]
  145. Bisaglia, M.; Bubacco, L. Copper Ions and Parkinson’s Disease: Why Is Homeostasis So Relevant? Biomolecules 2020, 10, 195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Rios, C.; Alvarez-Vega, R.; Rojas, P. Depletion of copper and manganese in brain after MPTP treatment of mice. Pharm. Toxicol. 1995, 76, 348–352. [Google Scholar] [CrossRef] [PubMed]
  147. Cukierman, D.S.; Bodnár, N.; Evangelista, B.N.; Nagy, L.; Kállay, C.; Rey, N.A. Impact of pyridine-2-carboxaldehyde-derived aroylhydrazones on the copper-catalyzed oxidation of the M112A PrP103–112 mutant fragment. J. Biol. Inorg. Chem. 2019, 24, 1231–1244. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Biometals are essential for the proper function and maintenance of neuronal cells. Iron is mainly found in the erythrocytes, bound to hemoglobin, and is essential for oxygen transfer to all tissues, including neurons. Iron also takes place in the Fenton reaction producing free radicals. Zinc and copper, and manganese are cofactors required for the antioxidant enzymes SOD1 and SOD2, respectively. These enzymes are scavengers of superoxide anion, and their activity is critical in regulating oxidative stress in the cytoplasm and the mitochondria. Oxidative stress may ultimately induce DNA damage, protein oxidation, and lipid peroxidation with deleterious consequences.
Figure 1. Biometals are essential for the proper function and maintenance of neuronal cells. Iron is mainly found in the erythrocytes, bound to hemoglobin, and is essential for oxygen transfer to all tissues, including neurons. Iron also takes place in the Fenton reaction producing free radicals. Zinc and copper, and manganese are cofactors required for the antioxidant enzymes SOD1 and SOD2, respectively. These enzymes are scavengers of superoxide anion, and their activity is critical in regulating oxidative stress in the cytoplasm and the mitochondria. Oxidative stress may ultimately induce DNA damage, protein oxidation, and lipid peroxidation with deleterious consequences.
Ijms 24 01256 g001
Figure 2. Interrelated events involved in the neurodegeneration process. Redox-active metals, including iron and copper, are recognized as inducers of oxidative stress. The latter relates to mitochondria dysfunction, lysosomal, and proteasomal degradation pathways inhibition, and ulterior protein accumulation and aggregation. Both protein degradation pathways may compensate for each other’s disruption and are affected in Parkinson’s Disease (PD) patients, where a more advanced state is characterized by Lewy body appearance and dopaminergic neuronal loss.
Figure 2. Interrelated events involved in the neurodegeneration process. Redox-active metals, including iron and copper, are recognized as inducers of oxidative stress. The latter relates to mitochondria dysfunction, lysosomal, and proteasomal degradation pathways inhibition, and ulterior protein accumulation and aggregation. Both protein degradation pathways may compensate for each other’s disruption and are affected in Parkinson’s Disease (PD) patients, where a more advanced state is characterized by Lewy body appearance and dopaminergic neuronal loss.
Ijms 24 01256 g002
Figure 3. Iron toxicity in neuronal cells is mediated by oxidative stress. Fe3+ is reduced to Fe2+ to enter the cell, and DMT1 is the major iron importer in neurons. Iron toxicity is induced by a sequence of highly oxidative and toxic reactions as it takes place in the Fenton reaction producing free radicals. Fe2+ reacts with H2O2 or lipid peroxides to generate Fe3+, OH, and hydroxyl radical (OH) or lipid radicals, which may lead to oxidative damage of macromolecules. Accumulation of OH leads to activation of the mitochondrial permeability transition pore (mPTP), which temporarily opens and increases reactive oxygen species (ROS). It provokes apoptotic cell death triggered by decreased ATP production, mitochondrial swelling, and rupture of the outer mitochondrial membrane, with subsequent release of cytochrome C to the cytosolic compartment.
Figure 3. Iron toxicity in neuronal cells is mediated by oxidative stress. Fe3+ is reduced to Fe2+ to enter the cell, and DMT1 is the major iron importer in neurons. Iron toxicity is induced by a sequence of highly oxidative and toxic reactions as it takes place in the Fenton reaction producing free radicals. Fe2+ reacts with H2O2 or lipid peroxides to generate Fe3+, OH, and hydroxyl radical (OH) or lipid radicals, which may lead to oxidative damage of macromolecules. Accumulation of OH leads to activation of the mitochondrial permeability transition pore (mPTP), which temporarily opens and increases reactive oxygen species (ROS). It provokes apoptotic cell death triggered by decreased ATP production, mitochondrial swelling, and rupture of the outer mitochondrial membrane, with subsequent release of cytochrome C to the cytosolic compartment.
Ijms 24 01256 g003
Figure 4. Copper-mediated neuronal cell death. Copper can be found in its oxidized (Cu2+) and reduced (Cu+) forms within the cells. The oxidized form of copper is bound to proteins, and when reduced by a reductase enzyme, it enters the cell through the transporters CTR1 and CTR2. Once inside and in high concentration, it induces DNA damage and p53 expression. p53 undergoes oligomerization/phosphorylation and is translocated into the nucleus to induce BAX and PUMA, with the consequent release of cytochrome C into the cytosol to initiate apoptosis. p53-dependent and independent apoptosis is triggered, where insulin-like growth factor binding protein-6 (IGFBP-6) and PIG proteins (prooxidant proteins), among other players, are involved.
Figure 4. Copper-mediated neuronal cell death. Copper can be found in its oxidized (Cu2+) and reduced (Cu+) forms within the cells. The oxidized form of copper is bound to proteins, and when reduced by a reductase enzyme, it enters the cell through the transporters CTR1 and CTR2. Once inside and in high concentration, it induces DNA damage and p53 expression. p53 undergoes oligomerization/phosphorylation and is translocated into the nucleus to induce BAX and PUMA, with the consequent release of cytochrome C into the cytosol to initiate apoptosis. p53-dependent and independent apoptosis is triggered, where insulin-like growth factor binding protein-6 (IGFBP-6) and PIG proteins (prooxidant proteins), among other players, are involved.
Ijms 24 01256 g004
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

Gonzalez-Alcocer, A.; Duarte-Jurado, A.P.; Soto-Dominguez, A.; Loera-Arias, M.d.J.; Villarreal-Silva, E.E.; Saucedo-Cardenas, O.; de Oca-Luna, R.M.; Garcia-Garcia, A.; Rodriguez-Rocha, H. Unscrambling the Role of Redox-Active Biometals in Dopaminergic Neuronal Death and Promising Metal Chelation-Based Therapy for Parkinson’s Disease. Int. J. Mol. Sci. 2023, 24, 1256. https://doi.org/10.3390/ijms24021256

AMA Style

Gonzalez-Alcocer A, Duarte-Jurado AP, Soto-Dominguez A, Loera-Arias MdJ, Villarreal-Silva EE, Saucedo-Cardenas O, de Oca-Luna RM, Garcia-Garcia A, Rodriguez-Rocha H. Unscrambling the Role of Redox-Active Biometals in Dopaminergic Neuronal Death and Promising Metal Chelation-Based Therapy for Parkinson’s Disease. International Journal of Molecular Sciences. 2023; 24(2):1256. https://doi.org/10.3390/ijms24021256

Chicago/Turabian Style

Gonzalez-Alcocer, Alfredo, Ana Patricia Duarte-Jurado, Adolfo Soto-Dominguez, Maria de Jesus Loera-Arias, Eliud Enrique Villarreal-Silva, Odila Saucedo-Cardenas, Roberto Montes de Oca-Luna, Aracely Garcia-Garcia, and Humberto Rodriguez-Rocha. 2023. "Unscrambling the Role of Redox-Active Biometals in Dopaminergic Neuronal Death and Promising Metal Chelation-Based Therapy for Parkinson’s Disease" International Journal of Molecular Sciences 24, no. 2: 1256. https://doi.org/10.3390/ijms24021256

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