Next Article in Journal
Self-Aggregation, Antimicrobial Activity and Cytotoxicity of Ester-Bonded Gemini Quaternary Ammonium Salts: The Role of the Spacer
Previous Article in Journal
Design and Synthesis of AMPK Activators and GDF15 Inducers
Previous Article in Special Issue
Chemical Modification of Auranofin Yields a New Family of Anticancer Drug Candidates: The Gold(I) Phosphite Analogues
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Bioinorganic Chemistry of Micronutrients Related to Alzheimer’s and Parkinson’s Diseases

Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
*
Author to whom correspondence should be addressed.
Molecules 2023, 28(14), 5467; https://doi.org/10.3390/molecules28145467
Submission received: 6 June 2023 / Revised: 10 July 2023 / Accepted: 13 July 2023 / Published: 17 July 2023
(This article belongs to the Special Issue The Role of Metal Ions in Bio-Inorganic Chemistry)

Abstract

:
Metal ions are fundamental to guarantee the regular physiological activity of the human organism. Similarly, vitamins play a key role in many biological functions of the metabolism, among which are coenzymes, redox mediators, and antioxidants. Due to their importance in the human organism, both metals and vitamins have been extensively studied for their involvement in neurodegenerative diseases (NDs). However, the full potential of the interaction between vitamins and metal ions has not been fully explored by researchers yet, and further investigation on this topic is needed. The aim of this review is to provide an overview of the scientific literature on the implications of vitamins and selected metal ions in two of the most common neurodegenerative diseases, Alzheimer’s and Parkinson’s disease. Furthermore, vitamin–metal ion interactions are discussed in detail focusing on their bioinorganic chemistry, with the perspective of arousing more interest in this fascinating bioinorganic field.

Graphical Abstract

1. Introduction

The human organism is primarily made up of water, fat, proteins, and minerals [1]. Metals are present in small quantities in the body. For example, an adult male body contains 3–4 g of iron [2]. Nevertheless, metals are necessary for the preservation of physiological functions of the organism. They are involved in several biological processes such as electron transfer, oxygen transport, the maintenance of osmotic pressure, and the regulation of DNA transcription [3]. Metals such as iron, cobalt, selenium, copper, zinc, and manganese are essential for human life and are usually required in trace amounts. On the other hand, aluminum, mercury, arsenic, and others are considered non-essential metals since they possess no biological function [4,5]. The importance of metals in the human organism is so fundamental that several pathologies, among which are neurodegenerative diseases (NDs), are related to a common phenomenon known as metal dyshomeostasis [6]. The scientific literature offers a large number of papers addressing the implications of metals in NDs [7,8,9].
Metal ions have been extensively studied for their interactions with important biomolecules such as amino acids [10,11], peptides [12,13], and proteins [14,15], which are involved in structural functions, cell signaling, cell expression, and hormone synthesis, to name a few [16,17]. Proteins often need to interact with metal ions to carry out their function [18].
As for metal ions, vitamins play a key and fundamental role in the healthy state of the human organism. This class of compounds, vital for existence, are essential micronutrients [19], although niacin [20] and vitamin D3 [21] can be synthesized by the organism. Vitamins are involved in many important biological functions in the human metabolism such as (i) coenzymes, (ii) redox mediators, (iii) antioxidants, (iv) hormones, and (v) regulators of gene transcription [22].
As with metals, the role of vitamins in neurodegeneration has been a hot research topic during recent decades [9,23]. However, the number of studies on the interaction between metal ions and vitamins is not comparable to the research concerning metal–amino acid, metal–peptide, and metal–protein interactions. This field is just at the beginning but possesses all the features to offer new insight to improve the knowledge about the interactions between vitamins and metal ions in biological systems.
In view of the above, the aim of this review was to contribute to this topic by giving an overview of the literature about the role of vitamins and selected metal ions in the two most common neurodegenerative diseases: Alzheimer’s and Parkinson’s diseases. In addition, the chemical interaction between vitamins and metal ions was addressed to evaluate the role of these associations and their impact on healthy homeostatic equilibria.

2. Vitamins in Parkinson’s and Alzheimer’s Diseases

Nowadays neurodegenerative diseases are recognized as the most severe brain disorders characterized by a loss of neuron structure and function in specific brain areas, which ultimately leads to neuronal death. Neurodegeneration greatly impacts ordinary life due to the gradual cognitive decline primarily associated with speech, motor, and memory faculties. The most common NDs are Alzheimer’s disease (AD) and Parkinson’s disease (PD), affecting millions of people worldwide [24,25]. Both diseases are strongly correlated with aging, considered the most impacting risk factor for the development of neurodegeneration [26,27]. Moreover, with the increasing life expectancy in the elderly population, the prevalence of both AD and PD has been growing year after year [28]. The major critical hallmarks of AD and PD are protein aggregation, synaptic and neuronal network dysfunction, abnormal protein homeostasis, cytoskeletal defects, mitochondrial dysfunction, altered energy metabolism, DNA and RNA defects, inflammation, and neuronal death [25,29,30].
In this scenario, many biomolecules are extensively impaired in both AD and PD. Among them, vitamins are known to be implicated in several cellular processes associated with NDs by acting as antioxidant and anti-inflammatory compounds [23,31,32]. Extensive research has been performed in this field, underlining the role of vitamins on healthy and neurodegenerative brains. Most of the investigations focused on understanding abnormal vitamin levels associated with AD and PD and evaluating the protective effects of vitamin administration. The most recent results are hereafter discussed and schematized in Table 1 and Table 2 and Figure 1.

2.1. Parkinson’s Disease

Several clinical and case-control studies have correlated PD with vitamin B deficiency, also associated with an increased risk of the disease’s onset (Table 1) [33,34,35,36,37,38,39,40]. Low serum levels of vitamins B6 and B12 have been detected in PD cases characterized by polyneuropathy, epilepsy, and cognitive impairments [33]. Concentrations of pantothenic acid (vitamin B5) were dramatically changed in PD post-mortem brains, showing an approximately 40% reduction in the substantia nigra and the cerebellum regions [41]. Moreover, the implication of vitamins B1, B2, B6, B9, and B12 in PD also emerged from case-control and population-based cohort studies, showing that (i) low levels of vitamin B6 correlate with higher PD risk [36,37]; (ii) vitamin B1 and vitamin B9 are involved in olfactory dysfunctions in the early phase of PD [34], and (iii) mild cognitive impairment (MCI) in PD is associated with plasma levels of thiamine (vitamin B1) and thiamine-monophosphate, which are differently influenced according to sex [35]. Finally, recent studies focusing on the comprehension of neurotoxic pathways induced by biotin (vitamin B7) deficiency show that the absence of said vitamin causes the extensive formation of vacuoles in the brain, locomotor deficits, reduced levels of biotin-dependent carboxylases, elongated mitochondrial conformation, and increased superoxide levels [38].
In addition to the low levels of B vitamins associated with PD, a plethora of studies has also supported the neuroprotective activity of B vitamins against PD and other neurodegenerative disorders. In most cases, B vitamin administration led to an improvement in PD symptoms as observed by population-based cohort studies and clinical and animal in vivo investigations (Table 1) [36,42,43,44,45]. In particular, long-term intramuscular administration of thiamin led to the significant and stable amelioration of motor and non-motor capacity in PD patients [42]. Similarly, oral intake of riboflavin (vitamin B2) in 19 PD cases resulted in improvements in motor functions [43]. Analogous results were obtained using Drosophila PD [44] and C. Elegans models [45] whose motor abilities were enhanced upon administration of vitamin B3 and B12, respectively.
In health status, B vitamins are involved in specific metabolic pathways, thus playing an essential role in regulating mitochondrial enzymes. The lower levels (Table 1 and Table 2) observed in both PD and AD (vide infra) correlate well with the compromised mitochondrial activity usually observed in neurodegeneration. For example, the reduction of α-ketoglutarate dehydrogenase complex (αKGDC) activity is implicated in several NDs [46,47]. αKGDC, requiring vitamin B1 as a cofactor, is involved in the Krebs cycle, regulating multiple cellular processes such as reactive oxygen species (ROS) generation and inhibition [48,49]. Vitamin B6 acts as a cofactor in a huge number of cellular biochemical reactions [50,51]. It is involved in the synthesis of neurotransmitters including dopamine whose levels are significantly reduced in PD brains. Vitamin B6 is also required for the conversion of homocysteine (Hcy) to cysteine and, together with vitamin B12 and folate (vitamin B9), plays a key role in maintaining physiological plasma levels of Hcy (10–20 mol/L) [52]. Interestingly, higher Hcy concentrations in the blood are considered a major risk factor for AD, PD, and vascular dementia [53,54,55].
The protective effects of vitamin supplementation on mitochondrial dysfunctions, oxidative stress, inflammation, and neurotoxicity are also supported by in vitro studies [38,44,45,56]. In particular, the anti-inflammatory activity exhibited by niacin (vitamin B3) was correlated with the inhibition of inflammatory cytokines and its G-protein-coupled receptor GPR109A [56], usually up-regulated in PD white patients [57]. On the other hand, research studies on the SH-SY5Y PD model have indicated that the antioxidant activity of vitamin B12 is dependent on the regulation of p-JNK and PGC-1α levels, which are usually altered in oxidative stress conditions [45].
Over the last few years, scientific research on vitamin D and its relationship with PD has exploded in terms of contributions. On this issue, a large number of reviews have been published in the last five years (see, for example, [58,59,60,61,62]). Several studies have shown low vitamin D levels in PD patients along with a connection between high vitamin D concentration and reduced risk of PD [63,64]. In a mouse model of PD, subcutaneous administration of vitamin D3 led to the attenuation of behavioral deficits induced by 6-hydroxydopamine [65]. The role played by vitamin D in brain development is well known, such that a daily intake of 600 UL of vitamin D is recommended for pregnant women [66]. Moreover, vitamin D supports those neuronal and glial functions necessary to preserve neurological development and protect the adult brain [67]. In fact, vitamin D is essential for the neuronal survival of hippocampal and cortical neurons by regulating the release of nerve growth factor [68] and it is involved in dopaminergic neurogenesis and differentiation [69].
Currently, the knowledge of the relationship between PD and vitamin K is quite limited, although recent findings have attracted the scientific community to the possible therapeutic use of vitamin K in NDs [70]. Vitamin K is implicated in neuronal development and survival by modulating sphingolipid metabolism whose alteration leads to neuroinflammation and neurodegeneration [71]. A case-control study reported lower serum levels of vitamin K2 in PD patients when compared to controls [72]. In a Drosophila PD model, feeding with vitamin K2 significantly ameliorated the survival rate of the flies. In addition, vitamin K2 rescued mitochondrial dysfunction by acting as an electron transporter in mitochondria and maintaining efficient adenosine triphosphate (ATP) production [73]. The neuroprotective effects exhibited by vitamin K are further supported by its ability to (i) regulate the mitochondrial membrane potential and (ii) inhibit dopamine neuron damage caused by 6-hydroxydopamine [74] and activate the GAS6 protein, fundamental for cellular growth, survival, and death [75]. Finally, in vitro experiments have shown that vitamin K can reduce α-synuclein (αSyn) fibrillization at a substoichiometric concentration as shown by ThT fluorescence and AFM experiments [76]. At the same time, NMR structural investigations provided evidence of vitamin K interactions with αSyn. The most perturbed chemical regions are Gly31, Lys32, and 1,4-naphthoquinones for α-synuclein and vitamins K, respectively [76]. Since the N-terminal repeat region of αSyn is critical for fibril formation and morphology, the binding of vitamin K to this region might lead to structural rearrangements capable of interfering with protein fibrilization.
Similar to vitamin K, vitamin A is able to reduce αSyn fibrilization as well [77]. However, in contrast to vitamin K, no specific and selective protein-vitamin association has been found for the investigated retinol derivatives so far. Despite this, in vivo animal studies have indicated that the intracerebroventricular treatment with retinoic acid had a protective effect against the degeneration of dopaminergic neurons [78]. On the other hand, oral administration of retinol did not protect 6-hydroxydopamine-induced dopaminergic denervation in Wistar rats [79].
Vitamins A, C, and E are powerful antioxidant compounds able to scavenge peroxyl radicals. Among them, vitamin E is the most effective one, being more active than glutathione (GSH) and ß-carotene [80,81]. Both vitamins C and E are normally highly concentrated in the brain where they exert their radical scavenging activity. In fact, vitamin C feeding was found to promote catalase activity and reduce protein oxidation and H2O2 production in a Drosophila model of PD [82]. Moreover, in a rotenone-induced model of PD, intramuscular vitamin E treatment showed neuroprotective effects by (i) improving motor functions, (ii) reducing lipid peroxidation, and (iii) ameliorating GSH and superoxide dismutase (SOD) levels [83].
By considering the antioxidant role played by vitamins A, C, and E, reduced levels of these vitamins are expected to be associated with PD, characterized by elevated oxidative stress conditions. However, no clear results or consensus are available so far. A clinical study conducted with 44 PD patients has indicated no difference in vitamins C and E levels between healthy and PD cases [84]. On the other hand, reduced levels of lymphocyte vitamin C were observed in patients with severe PD [85]. The same uncertainty is found between two recent studies reporting the results based on a population-based cohort [86] and a meta-analysis [87]. The former identified no association between dietary intake of A, C, and E vitamins and the risk of developing PD. The latter found protective effects of vitamin E supplementation. However, the same authors do not exclude that such results could be due to other contributions, such as lifestyle and behavioral factors that were not taken into account in these studies.
Table 1. Summary of the studies and the effects (↑ increase; ↓ decrease; ↔ no change) of vitamins in PD.
Table 1. Summary of the studies and the effects (↑ increase; ↓ decrease; ↔ no change) of vitamins in PD.
VitaminType of StudyInterventionResultsRef.
B1Case-controlPlasma levels↓ B1[35]
B6, B9, B12RotterdamDietary intake↑ B9, B12 ↔ PD risk; ↑ B6 ↓ PD risk[36]
B2, B6, B9, B12Case-controlDietary intake↑ B2, B9, B12 ↔ PD risk; ↓ B6 ↑ PD risk[37]
B7In vivoDrosophila↓ B7[38]
Rodents↓ Biotin carboxylase
In vitroNeurons↓ B7 ↑ Mitochondrial stress
B12ClinicalSerum levels↑ B12 ↓ PD risk[40]
B5Ex vivoHuman PD brains↓ B5[41]
B1ClinicalIntramuscular administration↑ B1 ↑ Motor and non-motor functions[42]
B2ClinicalOral administration↑ B2 ↑ Motor functions[43]
B3In vivoDrosophila↑ B3 ↑ Motor functions[44]
In vitroSK-N-MC neurons↑ B3 ↓ Cytotoxicity
B12In vivoC. elegans↑ B12 ↓ ROS ↑ Motor functions[45]
Rodents↑ B12 ↑ Motor functions;
In vitroSH-SY5Y neurons↑ B12 ↓ Ox. stress ↓ Apoptosis
B3In vitroRAW264.7 cells↑ B3 ↓ Neuroinflammation[56]
DCross-sectionalSerum levels↓ D[64]
D3In vivoRodents↑ D3 ↑ Motor and non-motor functions ↓ Ox. stress[65]
K2Case-controlSerum levels↓ K2[72]
K2In vivoDrosophila↑ K2 ↑ Survival rate[73]
In vitroMitochondria↑ K2 ↑ ATP
KIn vitroαSyn↑ K ↓ Fibrillization[76]
AIn vivoRodents↑ A ↑Neuroprotection[78]
In vitroE14–15 neurons↑ A ↑ Viability
AIn vivoRodents↑ A ↔ Neuroprotection[79]
C, EIn vivoDrosophila↑ C, E ↓ Ox. stress[82]
EIn vivoRodents↑ E ↑ Neuroprotection ↑ Motor functions[83]
A, C, EClinicalPlasma levels↔ A, C, E[84]
CClinicalLymphocyte levels↓ C[85]
A, C, EProspective cohortDietary intake↑ A, C, E ↔ PD risk[86]

2.2. Alzheimer’s Disease

Similar to PD, B vitamin deficiency was observed in AD cases as well (Table 2). Clinical studies on AD patients have indicated that lower concentrations of vitamins B9 and B12 correlate with high serum Hcy levels [88,89]. As previously mentioned, hyperhomocysteinemia is commonly observed in PD, AD, and MCI cases [53,54,55]. The correlation between elevated Hcy concentrations and vitamins B9 and B12 deficiency might be explained by taking into account that both vitamins are involved in the methionine/homocysteine cycle [90]. Vitamin B12 is the cofactor of methionine synthase that catalyzes the methylation of Hcy leading to the formation of methionine and S-adenosylmethionine. Methionine synthase also uses 5-methyltetrahydrofolate as a one-carbon donor. In addition to that, folate-bound one-carbon units are necessary for purine synthesis mediated by deoxythymidine monophosphate. In this regard, an interesting study has shown that three-year folate supplementation improved cognitive functions such as memory, information processing speed, and sensorimotor speed, usually associated with dementia [91].
Serum Hcy concentration has been reduced by administrating vitamins B6, B9, and B12 in AD patients but no improvements in cognitive decline were observed [92,93]. In a case-control study, higher levels of vitamins B9 and B12 were shown to act as protective factors against AD [94]. On the other hand, no correlation between Hcy, folate, and vitamin B12 was observed in other studies [95,96]. The observed discrepancy may rely on different factors such as the sample size, duration, and control of the variables applied to the study.
In addition to the impact on Hcy concentration, the supplementation of vitamins B6, B9, and B12 reduced tau hyperphosphorylation and ameliorated the cognitive functions of adult mice exposed to hypobaric hypoxia [97]. In fact, vitamin B12 is able to interfere with several mechanisms associated with AD, such as amyloid precursor protein (APP) processing, amyloid β (Aβ) fibrillization, Aβ-induced oxidative damage, and tau hyperphosphorylation and tau aggregation [98]. Interestingly, the inhibition of tau fibrillization is mediated by the binding of vitamin B12 to Cys residues of the tau protein [99].
In addition to vitamin B6, B12, and folate, the role of riboflavin in slowing the progression of cognitive decline and reducing the risk of depression in aging is well accepted [100]. Intragastric riboflavin administration to double transgenic APP/PS1 mice, used as an in vivo AD model, led to a significant reduction in ROS by increasing SOD activity [101]. Riboflavin is also required for the flavin adenin dinucleotide (FAD)-dependent flavoenzyme methylenetetrahydrofolate reductase (MTHFR), involved in the one-carbon metabolism of Hcy remethylation. Impaired activity of MTHFR and its variant 677T leads to high Hcy plasma levels attributed to cellular FAD deficiency [102,103].
Thiamin deficiency was observed in AD cases by measuring plasma vitamin levels and the activity of the transketolase, a thiamin pyrophosphate-dependent enzyme [104,105]. The interest in thiamin is primarily related to numerous similarities occurring between classical thiamine deficiency and AD in terms of both cognitive deficits and reductions in brain glucose metabolism [106].
There are different outcomes in terms of niacin integration. Dietary niacin intake was shown to protect against AD and cognitive decline development [107]. On the other hand, the administration of reduced nicotinamide adenine dinucleotide (NADH), the biologically active form of niacin, showed no cognitive improvements in patients with different types of dementia, among which is AD [108]. The two studies primarily differ in the number of cases, the former being approximately 40 times greater. In 3xTg-AD mice, oral supplementation of niacinamide improved cognitive dysfunctions [109]. The observed ameliorations were caused by the reduction of Thr231 phosphorylated tau, which, in turn, increased tau degradation [109]. Thr231 phosphorylated tau is well known for its implication in AD and is also used as a biomarker for AD in cerebrospinal fluid (CSF) [110]. Moreover, the same study demonstrated that nicotinamide treatment affects acetyl-α-tubulin brain levels, thus promoting increased microtubule stability [109]. However, the positive effects exhibited by nicotinamide were completely reversed in late-stage mice with an existing severe pathology suggesting that nicotinamide treatment might be effective at the early or mild AD stages only. By using a different AD rat model, obtained by the administration of Aβ42 peptide into the rodents’ brains, intraperitoneal niacinamide caused the reduction of oxidative stress, apoptosis, and poly(ADP-ribose) polymerase-1 (PARP-1) activity, which are well known to be associated with neuroinflammation and cell death [111].
In addition to what has been previously reported for mammalian tauopathies, the brains of postmortem AD subjects manifested reduced levels of biotin carboxylase [38]. Intraperitoneal biotin and oral coenzyme Q10 (CoQ10) supplementation, both alone and in combination, attenuated neuroinflammation and improved brain insulin signaling in rats with AD induced by oral administration of AlCl3 [112].
Finally, the analysis of 18 human brains with short post-mortem delay revealed the reduction of vitamin B5 levels in different brain regions (hippocampus, entorhinal cortex, middle temporal gyrus, cingulate gyrus, sensory cortex, and cerebellum) [113]. The involvement of pantothenic acid in AD is further supported by the reduction in tricarboxylic acid cycle protein concentrations [114].
Compared to PD, the role played by vitamins C and E in AD is less controversial. Most of the studies indicate that vitamin C and E supplements play a protective role in the risk of AD [115] and that AD patients have lower serum, brain, and CSF levels of vitamins C and E than controls [95,116,117,118]. However, a prospective cohort study showed that upon administration of vitamin E and C, alone or in combination, the risk of dementia or AD was not decreased [119].
In a mouse model of AD, a 6-month treatment with vitamin C in a drinking solution attenuated Aβ oligomerization and behavioral deficits and reduced brain oxidative damage and hyper-phosphorylated tau proteins [120]. Oral supplementation of vitamin C (200 and 400 mg/kg body weight) protected against neuroinflammation-mediated neurodegeneration and memory deficits in a colchicine-induced rat model of AD. However, a higher dose (600 mg/kg body weight) worsened oxidative stress, neuroinflammation, and cognitive impairments [121]. On the other hand, an open clinical trial reported that supplementation with vitamin C and E had no beneficial effect in AD patients, except a limited antioxidant activity in the CSF [122] and, in a Japanese cross-sectional study, lymphocyte and plasma vitamin C levels were weakly correlated with the Mini-Mental State Examination Japanese version (MMSE-J) scores in AD individuals [123]. The different outcomes obtained for vitamin C might be explained by considering the prooxidant role exhibited by ascorbic acid, which can generate a high oxidative environment in the presence of redox-active metal ions, such as Fe and Cu [124].
In a randomized trial, the administration of α-tocopherol (2000 IU/d) for over two years slowed the functional decline in mild to moderate AD cases compared to a placebo group [125]. In a rat model of AD, intraperitoneal injection of vitamins E and D3, alone or combined, led to improved cognitive and memory impairments along with reduced neuronal loss and oxidative stress [126]. Vitamin E prevented increased ROS formation, protein oxidation, and neurotoxicity in Aβ42-treated neuronal cultures [127]. However, in a Mendelian randomization study by Liu et al., the authors reported no significant correlation between circulating vitamin E levels and AD risk in individuals of European descent [128]. In addition, vitamin E administration (a total population of 57 AD subjects) did not prevent oxidative stress and was found to be detrimental to cognitive activities in some cases [129].
Lower serum and plasma vitamin A levels were also found in AD patients compared to controls [130,131]. The treatment of cells with retinoic acid prevented Aβ production by inhibiting γ-secretase-mediated cleavage of APP via retinoic acid receptor-α and retinoid X receptor-α [132]. In a streptozotocin-induced AD mouse model, β-carotene administration in the form of a suspension was found to improve cognitive functions, inhibit acetylcholinesterase (AChE), and reduce Aβ-protein fragments [133].
Several studies demonstrated the association between lower serum levels of vitamin D and an increased risk of AD [134,135]. In a randomized, double-blind, placebo-controlled trial, 12-month administration of vitamin D3 was found to ameliorate cognitive activities and reduce Aβ-related biomarkers in AD older adults [136]. A 7-year follow-up study by Annweiler and colleagues revealed a correlation between a higher vitamin D dietary assumption and reduced risk of developing AD in older women [137]. In a mouse model of intracerebroventricular streptozotocin-induced sporadic AD, oral vitamin D3 administration improved cognitive activities, attenuated neuroinflammation and oxidative stress, and ameliorated cholinergic functions [138]. On the other hand, a randomized controlled trial by Stein and coworkers reported that high-dose vitamin D2 supplementation does not improve cognition or disability with respect to a low dose in mild-moderate AD patients [139].
Finally, a lower vitamin K1 daily intake was found in early-stage AD patients compared to controls [140]. In post-mortem human brains, higher menaquinone-4 (vitamin K2) levels were associated with a lower risk of AD [141]. Pretreatment with vitamin K2 solution had a protective role against Aβ42-induced neurotoxicity by activating autophagy and ameliorating mitochondrial function in a Drosophila model of AD [142]. In a model of Alzheimer’s cell damage, the pretreatment of PC12 cells with vitamin K2 led to a significant decrease in Aβ42, H2O2, ROS cytotoxicity, and cell apoptosis via the inactivation of the p38 MAP kinase pathway [143].
Table 2. Summary of the studies and the effects (↑ increase; ↓ decrease; ↔ no change) of vitamins in AD.
Table 2. Summary of the studies and the effects (↑ increase; ↓ decrease; ↔ no change) of vitamins in AD.
VitaminType of StudyInterventionResultsRef.
B6, B9, B12ClinicalSerum levels↔ B6 ↓ B9, B12[88]
B9, B12Case-controlPlasma levels↓ B9, B12[89]
B6, B9, B12RCT *Supplementation↑ B6, B9, B12 ↔ Cognitive func.[37,92]
B9, B12Cross-sectionalSerum levels↑ B9, B12 ↓ AD risk[94]
B1, B2, B9, B12, C, ACross-sectionalPlasma levels↓ B2, C, A ↔ B1, B9, B12[95]
B6, B9, B12In vivoRodents↑ B6, B9, B12 ↑ Cognitive func.[97]
B12In vitroTau↑ B12 ↓ Fibrillization[99]
B2In vivoRodents↑ B2 ↑ Cognitive func. ↓ Ox. stress[101]
B3Prosp. cohortDietary intake↑ B3 ↑ Cognitive func.[107]
B3ClinicalSupplementation↑ B3 ↔ Cognitive func.[108]
B3In vivoRodents↑ B3 ↑ Cognitive func. ↑ Microtubule stability[109]
B3In vivoRodents↑ B3 ↓ Ox. stress[111]
B7In vivoDrosophila↓ B7[38]
Rodents↓ Biotin carboxylase
Ex vivoHuman AD brains↓ Biotin carboxylase
In vitroNeurons↓ B7 ↑ Mitochondrial stress
B7In vivoRodents↑ B7 ↓ Neuroinflammation[112]
B5Ex vivoHuman AD brains↓ B5[113]
C, EClinicalSupplementation↑ C, E ↓ AD risk[115]
EClinicalCSF, Plasma levels↓ E[117,118]
C, EProsp. cohortDietary intake↑ C, E ↔ AD risk[119]
CIn vivoRodents↑ C ↑ Cognitive func. ↓ Ox. stress ↓ Aβ oligomerization[120]
C, EClinicalSupplementation↑ C, E ↔ AD[122]
EClinicalSupplementation↑ E ↓ Functional decline[125]
EIn vitroE18 neurons↑ E ↓ Ox. stress[127]
A, ECase-controlSerum levels↓ A, E[130]
AIn vitroSeveral cell lines↑ A ↓ γ-secretase[132]
AIn vivoRodents↑ A ↓ AChE[133]
In silicoMolecular dockingA—A ChE[134]
DRotterdamSerum levels↓ D ↑ AD risk[135]
D3ClinicalSupplementation↑ D3 ↑ Neuroprotection[136]
DClinicalDietary intake↑ D ↓ AD risk[137]
D3In vivoRodents↑ D3 ↓ Ox.stress ↑ Cholinergic[138]
K2In vivoDrosophila↑ K2 ↑ Neuroprotection[142]
K2In vitroPC12↑ K2 ↓ Aβ42 cytotoxicity[143]
* Randomized controlled trial.

3. Metals in Parkinson’s and Alzheimer’s Diseases

Several transition metal ions are known to play key roles in AD and PD [8]. Altered homeostasis of biometals such as zinc, copper, iron, and manganese is associated with high neurotoxicity and oxidative stress conditions typically observed in AD and PD cases [144,145,146,147,148,149,150].
In particular, redox-active metals such as Cu(II)/Cu(I) and Fe(III)/Fe(II) can catalyze the Fenton reaction, producing cytotoxic hydroxyl radicals from hydrogen peroxide [151,152]. In addition, copper and zinc are normally released at the glutamatergic synapse in the cortex and hippocampus and, together with iron, are able to bind amyloidogenic proteins and other hallmark molecules associated with NDs (Figure 2) [153,154,155,156]. Mn is a co-factor of glutamine synthetase involved in the recycling of glutamate to glutamine and thus responsible for the glutamate clearance from the synapse [157]. Mn is also essential for MnSOD activity protecting mitochondria from oxidative stress [157]. Other metal ions such as aluminum and nickel may represent risk factors for neurodegenerative diseases leading to mitochondrial dysfunction, microglial activation, and neuroinflammation [158,159]. Ni is extensively distributed in the environment. It is an essential nutrient for some animals, plants, and microorganisms, while its functional role in humans has not been recognized yet [160]. In contrast to Ni, Al is not an essential element. It is the most abundant metal on the earth’s crust and is widely used in daily human and industrial activities. Both Ni and Al traces can be found in food, drinking water, and the air.
As for vitamins, the interplay between metal ions and neurodegenerative diseases has been extensively investigated over the last thirty years. Research in this field has exponentially grown since 1990, reaching more than 3400 publications in the last ten years (Pubmed source “metal” and “neurodegeneration”). The scientific community has made great efforts to identify the role played by metal ions in the molecular associations and cellular pathways related to AD and PD. While much progress has been made in this area, several points remain to be clarified yet. In this review, we have focused on the relationship between these six metal ions and the two most common NDs, PD and AD, by briefly highlighting the metal’s coordination chemistry properties and metal involvement in AD and PD states.

3.1. Zinc, Copper, and Iron

Zinc, copper, and iron levels in serum, hair, CSF, and the brain have been extensively measured trying to correlate their content with metal dyshomeostasis associated with AD and PD cases [161]. The most applied techniques are atomic absorption, inductively coupled plasma atomic emission spectroscopy (ICP-AES), ICP-mass spectrometry (MS), and ICP-optical emission spectrometry (OES). Serum zinc levels were generally found to be reduced in patients affected by both AD and PD [162,163,164,165,166,167,168]. Decreased Zn concentrations have also been determined in AD hair samples [169]. On the other hand, reduced and increased copper contents have been measured in both AD [164,166,168,170,171] and PD cases [165,172]. Finally, a different behavior is displayed by iron, whose levels are different according to the disease, usually lower [166,167,173] or higher [165,174] in AD and PD patients, respectively.
Altered zinc, iron, and copper concentrations have also been found in CSF and post-mortem brains [161,175,176,177,178]. In PD patients, zinc levels are higher in the substantia nigra, caudate nucleus, and lateral putamen [175]. Iron content is higher in the substantia nigra and lower in the globus pallidus [175]. In PD, copper is increased in the putamen and decreased in the substantia nigra [175], while it is decreased in AD brains [178].
The altered metal levels observed in AD also correlate with the presence of Fe and Cu, Zn, in the AD senile plaques, primarily constituted by the aggregated forms of Aβ [179,180,181,182]. Aβ is a well-known amyloidogenic protein associated with AD and it is able to bind copper, zinc, and iron by means of His imidazole, N-terminal amino, and Glu/Asp carboxylate groups [183,184,185,186,187,188,189,190]. In a similar way, the amyloidogenic proteins tau and alpha synuclein, associated with AD and PD, respectively, can steadily coordinate several transition metal ions [191,192,193,194,195] (Figure 2).
Metal ions such as Cu and Zn can impact the aggregation of amyloidogenic proteins by affecting the morphologies and kinetics of the aggregates. The scientific community put a lot of effort into understanding the influence of metal ions, primarily copper and zinc, in the aggregation of amyloidogenic proteins [196,197,198,199,200,201,202]. The obtained findings are quite heterogeneous primarily due to the intrinsic complexity of the systems and different experimental conditions and techniques. In general, it is evident that zinc promotes the formation of amorphous Aβ aggregates while copper favors the production of highly cytotoxic oligomers [203,204,205]. As for Aβ, metal ion binding impacts the aggregation of αSyn as well, either showing pro- or anti-aggregatory effects [206,207]. Among all the metal ions, iron and copper are able to influence αSyn aggregation by promoting the formation of multimeric species and αSyn assembly [208,209].
Zinc interaction with the third repeat unit of the microtubule-binding domain of tau (R3tau) leads to the formation of Zn(II)-R3tau aggregates [210]. Such complexes, compared to R3tau, possess higher toxicity towards Neuro-2A (N2A) cells by inducing higher ROS generation in N2A cells. Copper increases the aggregation propensity of tau through its capability to both bind tau and produce ROS [211]. Zn and Fe binding to tau-R1 and R4 was also investigated; Zn(II) and Fe(II) but not Fe(III) coordination was demonstrated by CD and ESI-MS. Both interactions induced conformational changes in R1 and R4 [212]. Copper binding to tau occurs via His residues present in R1, R2, R3, and R4 or at the N-terminal site [213,214,215]. Recent molecular dynamic studies have revealed the misfolding of R3tau upon Cu(II) binding [216]. In addition, the ability of the copper–R3tau complex to promote the oxidation of dopamine has been recently reported [215].
The involvement of zinc, iron, and copper in AD is also supported by in vivo animal studies showing the effects of metal deficiency and/or supplementation in AD mice models [217,218,219,220,221]. For example, a zinc-deficient diet in an APP/PS1 mouse model of AD accelerated memory deficits through the induction of the NLRP3-inflammasome complex [217]. Other studies show that treatment with low levels of Cu(II) in drinking water led to an increase in Aβ production in neuroinflammation [218] and promoted Aβ accumulation, reducing mice’s cognitive functions [219]. Finally, in hypercholesteremia-induced AD rabbits, the administration of Fe(III) chelator deferiprone in drinking water significantly reduced the levels of plasma iron and cholesterol and decreased tau phosphorylation, Aβ40, and Aβ42 but not ROS production in the hippocampus [220]. In contrast, the treatment of an AD mouse model with Fe(II)-containing water markedly reduced Aβ42 deposition, tau phosphorylation, and apoptotic neurons and led to an increase in Aβ40 and a reduction in the Aβ42/Aβ40 ratio [222].

3.2. Manganese, Nickel, and Aluminum

The relevance of Mn, Ni, and Al in both AD and PD is well documented in the literature even if to a lesser extent than the essential Zn, Cu, and Fe ions. In PD, the serum levels of Mn, Ni, and Al are generally higher compared to healthy controls [163,174,223].
Furthermore, acute exposure to Mn can result in manganism, a type of parkinsonism, considered part of the PD etiology [221]. Manganism may be caused by elevated Mn accumulation in the basal ganglia region of the brain [224].
The association between aluminum and PD was suggested by the detection of Al in the Lewy bodies of PD patients, while its value is below the limit of detection in control brains [225]. Such findings are further supported by the higher incidence of ulcer patients that make high use of Al(III)-containing antiacids in PD cases compared to controls [226]. In addition, Al(III) was found to increase monoamine oxidase B and SOD activities in a way similar to what was observed in PD patients [163,227].
As for PD, higher serum levels of Ni [223] and Al [166,228,229,230,231] have been found for AD cases. On the other hand, reduced [167,232] or increased [169,233] Mn serum levels are reported for AD and MCI subjects. Mn content was also lower in the hair and nails of AD cases compared to control subjects [233]. Nickel levels were higher in the post-mortem frontal cortex and ventricular fluid of AD subjects with respect to nondemented elderly controls [234]. At the same time, nickel supplementation in the forms of the NiCl2 and NiCl2-morpholine complex prevented tau aggregation and promoted its degradation with the formation of shorter aggregates [235].
Moreover, in vitro and in vivo investigations on APP/PS1 mice showed dose-dependent neurotoxicity and an increase in Aβ upon Mn(II) treatment [236].
Finally, Al(III) may be implicated in AD pathogenesis via the induction of APP overexpression and the subsequent increase in Aβ and plaque formation in the brain [237]. A laser microprobe mass analysis showed a primary accumulation of Al(III) in the neurofibrillary tangles (NFTs) of AD subjects [238]. A 15-year follow-up study revealed an association between the high consumption of aluminum from drinking water and an increased risk of AD [239]. APP/PS1 transgenic mice, treated with intracerebroventricular microinjections of AlCl3, presented more extensive worsening of cognitive abilities and increases in neural apoptotic rates than APP/PS1 alone and wild-type mice exposed to Al [240].

4. Interaction between Vitamins and Metal Ions

After dealing with the implications of vitamins and metals in AD and PD, this paragraph will review the literature concerning the chemical interaction between vitamins and the selected metal ions, i.e., Zn(II), Cu(II), Fe(II), Fe(III), Mn(II), Ni(II), and Al(III), with the final aim to evaluate if vitamins might interfere with the metal–protein associations usually dysregulated in AD and PD (vide Section 3), thus possibly explaining the positive effects observed for various vitamins (vide Section 2). In fact, vitamins may interfere with the metal binding modes and the protein structural rearrangements as previously observed for other natural compounds [241]. Several papers addressed the synthesis, characterization, and evaluation of the thermodynamic and structural features of vitamin–metal complexes in vitro. Figure 3, Figure 4 and Figure 5 show the metal ions and identified vitamin donor atoms as described in detail hereafter. Table 3 lists all the metal–vitamin interactions studied so far together with the experimental techniques and conditions, such as to provide a solid starting base for researchers involved in developing this intriguing bioinorganic field.
Research on the interaction between thiamin pyrophosphate and divalent metal cations reported different coordination modes by the ligand schematically represented in Figure 3. Ni(II) coordination occurs through the pyrophosphate group and a water molecule that bridges the metal to the ligand thanks to a hydrogen bond with N1′ [242,243]. On the other hand, Mn(II) is coordinated only with the pyrophosphate group, which is folded over the thiazolium ring [244].
The interaction between thiamin and Zn(II) was investigated by IR and NMR techniques revealing metal coordination at the N3′ position of the pyrimidine ring as shown in Figure 3 [245]. Indeed, a comparison of 1H NMR spectra of thiamin hydrochloride solutions with and without Zn(II) showed a significant upfield shift (0.28 ppm) and broadening of the amino group close to the binding nitrogen, while the 13C NMR revealed downfield chemical shifts of the carbons adjacent to the coordination site [245].
Extensive investigations on the riboflavin–metal ions interaction in an aqueous solution have indicated the ability of this vitamin to form 1:1 and 1:2 (ion:ligand) chelates with different metal cations, including Cu(II), Ni(II), Zn(II), Fe(III), and Mn(II) [246]. Metal coordination occurs via the carbonyl oxygen at the C4 position and N5 of the isoalloxazine moiety (Figure 3). A more recent study reported the synthesis and characterization of riboflavin metal complexes. In the case of Zn(II), the metal is bound through the oxygen at the C4 site and N3 of the ligand (Figure 3), with the addition of two water molecules to complete the metal coordination sphere [247].
The possibility for niacin to form complexes with divalent metal cations was investigated in several papers. Hernowo et al. studied the interaction of nicotinic acid with a number of metal ions, among them Cu(II) and Ni(II) [248]. Potentiometric measurements combined with software computations revealed higher stability for the copper complex, in line with the Irving–Williams series [249]. In addition, the authors predicted the molecular structure of both copper and nickel complexes in which metal coordination occurs via the pyridine nitrogen of nicotinic acid as shown in Figure 3 [248]. In a recent paper, nicotinamide was found to form coordination compounds with divalent metals such as Mn(II), Ni(II), Cu(II), and Zn(II) in a 2:1 ligand:metal ratio [250] (Figure 3). Furthermore, Sismanoglu reported the formation of 1:1 and 1:2 nicotinamide-Mn(II) complexes in which the pyridine N of the ligand binds Mn(II) ions [251].
Figure 3. Metal complexes with thiamin, riboflavin, niacin and vitamin C. Thiamin pyrophosphate: M1 = Ni(II) [242,243], M2 = Mn(II) [244]. Thiamin: M1 = Zn(II) [245]. Riboflavin: M1 = Zn(II), Cu(II), Fe(III), Mn(II), Ni(II) [246], M2 = Zn(II) [247]. Nicotinic acid: M1 = Cu(II), Ni(II) [248]. Nicotinamide: M1 = Zn(II), Cu(II), Ni (II) [250], Mn(II) [250,251]. L-Ascorbic acid: M1 = Zn(II), Mn(II) [252], Ni(II) [253], Al(III) [253,254], M2 = Zn(II), Mn(II) [252], Al(III) [254], M3 = Fe(III), Mn(II) [255], M4 = Cu(II) [256].
Figure 3. Metal complexes with thiamin, riboflavin, niacin and vitamin C. Thiamin pyrophosphate: M1 = Ni(II) [242,243], M2 = Mn(II) [244]. Thiamin: M1 = Zn(II) [245]. Riboflavin: M1 = Zn(II), Cu(II), Fe(III), Mn(II), Ni(II) [246], M2 = Zn(II) [247]. Nicotinic acid: M1 = Cu(II), Ni(II) [248]. Nicotinamide: M1 = Zn(II), Cu(II), Ni (II) [250], Mn(II) [250,251]. L-Ascorbic acid: M1 = Zn(II), Mn(II) [252], Ni(II) [253], Al(III) [253,254], M2 = Zn(II), Mn(II) [252], Al(III) [254], M3 = Fe(III), Mn(II) [255], M4 = Cu(II) [256].
Molecules 28 05467 g003
Vitamin B6 has been widely investigated for its ability to coordinate metal ions. Pyridoxal-5′-phosphate was found to form a 1:1 (ligand:metal) complex with Mn(II), coordinating the metal via the phosphate group and the aldehyde oxygen as shown in Figure 4 [257]. Thermodynamic and kinetic parameters for the complexation reaction of pyridoxine with Ni(II) indicated a positive value of the enthalpy of interaction, suggesting an endothermic process for the formation of a pyridoxine-Ni complex [258]. Using X-ray analysis, Thompson and colleagues determined the structure of the Zn(II)-pyridoxamine complex [259]. The compound presents an octahedral geometry with the zinc atom chelated to the 4-aminomethyl and phenolate groups of two pyridoxamines (Figure 4). Two water molecules complete the octahedral structure. In addition, two nitrate groups in the secondary coordination sphere of zinc form hydrogen bonds with the water molecules and the 4-aminomethyl group of the ligand. Another metal complex involving vitamin B6 was reported for pyridoxine and Zn(II) ions [260]. Again, the geometry is octahedral with metal coordination occurring via the 4-hydroxymethyl and phenolate oxygens of two pyridoxines and two water molecules (Figure 4). On the other hand, Zhu and coworkers showed the formation of ML chelates, where M = Ni(II), Cu(II), or Zn(II) and L = pyridoxine, in which the metal is bound through the phenolic group at the C3 position and the hydroxyl at the C4 site (Figure 4) [261].
Figure 4. Metal complexes with biotin, folate, vitamins B6 and D3. D-(+)-Biotin. M1 = Zn(II), Cu(II), Mn(II) [262,263]. Folic acid: M1 = Cu(II), Fe(III), Mn(II), Ni(II), Al(III) [264], M2 = Cu(II), Fe(III) [265]. PLP: M1 = Mn(II) [257]. Pyridoxine: M1 = Zn(II) [260,261], Cu(II) [261], Ni(II) [258,261]. Pyridoxamine: M1 = Zn(II) [259]. Cholecalciferol: M1 = Zn(II), Fe(II, III), Mn(II) [264], Cu(II), Ni(II) [266].
Figure 4. Metal complexes with biotin, folate, vitamins B6 and D3. D-(+)-Biotin. M1 = Zn(II), Cu(II), Mn(II) [262,263]. Folic acid: M1 = Cu(II), Fe(III), Mn(II), Ni(II), Al(III) [264], M2 = Cu(II), Fe(III) [265]. PLP: M1 = Mn(II) [257]. Pyridoxine: M1 = Zn(II) [260,261], Cu(II) [261], Ni(II) [258,261]. Pyridoxamine: M1 = Zn(II) [259]. Cholecalciferol: M1 = Zn(II), Fe(II, III), Mn(II) [264], Cu(II), Ni(II) [266].
Molecules 28 05467 g004
Studying the stability constants of binary 1:1 complexes of D(+)-biotin with Mn(II), Cu(II), and Zn(II) in various solvents, Sigel et al. determined the involvement of the carboxylic group in metal complexation [262]. In addition, for Mn(II) and Cu(II) complexes, NMR analysis revealed weak participation of the sulfur atom in the metal coordination, as suggested by line broadening of one of the two hydrogens of the methylene group next to the sulfur atom (Figure 4). Therefore, the authors proposed a stereospecific interaction between Mn(II) or Cu(II) and sulfur, where the metal binds the sulfur atom from below the plane [262]. Such a behavior of the thioether group was later confirmed in complexes involving D(+)-biotin and metal ions among which are Mn(II), Cu(II), and Zn(II). Again, metal coordination to sulfur was found to occur below the tetrahydrothiophene group, trans with respect to the imidazolidinone ring [263].
The complexing ability of folic acid with metal ions was largely studied by Yousef and coworkers. Potentiometric and conductometric studies showed the possibility of forming 1:1, 1:2, and 1:3 metal to ligand complexes for a series of metal ions, including Al(III), Fe(III), Cu(II), Mn(II), and Ni(II) [264]. The structure of the complexes is characterized by the presence of six-membered chelate rings. Folic acid binds the metal cation through the imino and the two carboxylic groups of the glutamic acid moiety (Figure 4). Other investigations pointed out the ability of folic acid to form two octahedral coordination compounds involving Cu(II) or Fe(III) as metal centers with a 1:2 metal-to-ligand ratio [265]. Folic acid was found to act as a bidentate ligand coordinating the metal ion through both carboxylic groups of the glutamate moiety and two water molecules in the coordination sphere (Figure 4). Polarimetric analysis revealed no angle of rotation for both complexes, suggesting a symmetric geometry.
NMR structural characterization of zincobalamin (Znbl), a Zn(II)-analogue of vitamin B12, revealed a structure similar to the Co(II) counterpart, i.e., Co(II)cobalamin (Cobl) (Figure 5) [267]. Using 1H-1H ROESY spectroscopy, the authors demonstrated that Znbl is isostructural to Cobl. In addition, the computationally generated structure of Znbl showed a downward movement of the 5,6-dimethylbenzimidazole moiety of the molecule with respect to Cobl. However, Znbl and Cobl were similar in overall architecture [267]. The cobalt ion in cobalamin was also replaced by Ni(II), generating nibalamin (Nibl). UV-Vis and NMR characterization suggested that Nibl is isoelectronically and roughly isostructurally analogous to Co(I)cobalamin [268].
Figure 5. Metal complexes with vitamin B12. Zincobalamin: M1 = Zn(II) [267]. Nibalamin: M1 = Ni(II) [268].
Figure 5. Metal complexes with vitamin B12. Zincobalamin: M1 = Zn(II) [267]. Nibalamin: M1 = Ni(II) [268].
Molecules 28 05467 g005
Researchers extensively studied the interaction between vitamin C and metal ions. Tajmir-Riahi focused on the interaction of L-ascorbic acid with several metal ions both in a solution and in a solid state (Figure 3). Solid salts with the stoichiometry M(L-ascorbate)2.2H2O, where M = Zn(II) or Mn(II), and Al(L-ascorbate)3 were isolated and studied by 13C NMR and Fourier-transform infrared (FT-IR) spectroscopy techniques. The author demonstrated that, in an aqueous solution, Zn(II) and Mn(II) ions form chelates where the ascorbate binds the metal via its O2 and O3 atoms. In the solid state, two ascorbate anions were involved in the metal coordination through O2 and O3 of the first ion and O1 and O3 of the second one, together with two water molecules, leading to a six-coordinated complex [252]. In an aqueous solution, Al(III) was chelated by the O2 and O3 atoms of the ligand, while in the solid state, Al is bound via O1, O3, and O2, O3 of the anions, and water molecules [254]. In a two-part research study on the synthesis and characterization of metal-ascorbate complexes, Obaleye and coworkers reported paramagnetic behavior and an octahedral structure for coordination compounds involving vitamin C and Mn(II) and Fe(III) (Figure 3). Such complexes, with the formula Mn(L-ascorbate)2(H2O)2 and [Fe(L-ascorbate)2(H2O)3]Cl, showed a bidentate complexation mode of the ascorbate anion via its carbonyl and O2 atom [255]. Recently the complexation of Al(III) and Ni(II) by L-ascorbic acid was investigated using potentiometry, 1H NMR spectroscopy, and DFT calculations [253]. Experimental and theoretical data indicated that vitamin C preferentially coordinated the metal ions via the deprotonated oxygens at C2 and C3 positions (Figure 3). DFT computations suggested that Al(III) complexes were formed with an ion:ligand stoichiometric ratio of 1:2, while for Ni(II) compounds, the 1:1, 1:2, and 1:3 stoichiometries were possible. All the computed structures exhibited water molecules in the coordination sphere along with a prevalence of octahedral geometry. Ünaleroğlu and colleagues prepared a stable Cu(II)-ascorbate complex in CH2Cl2. The compound was found to be paramagnetic, and susceptibility measurements undoubtedly indicated a +2 oxidation state of the metal. 1H NMR and IR analyses revealed Cu coordination by the enolic and side chain oxygens of the ascorbate (Figure 3). One ligand unit binds the metal via the O2 and O3 atoms, and another one through the side chain oxygens, leading to a four-fold coordinated copper; such a structural unit repeats to form a polymer [256].
Mercê et al. carried out potentiometric studies of vitamin D3 complexes with various metal ions, including Mn(II), Fe(II), Fe(III), Zn(II), Ni(II), and Cu(II) in a water ethanol-medium [266,269]. The authors showed the formation of ML (M = metal, L = OH deprotonated vitamin D3) and ML2 species for all the cations reported above, except for Cu(II) (Figure 4). The absence of this complex was partially attributed to the formation of insoluble compounds at pH~5. For Cu(II) and Fe(III), potentiometric analysis allowed the detection of MLH species, where the ligand is protonated at the OH site. Moreover, Fe(III) was found to form an ML3 complex by deprotonating and holding a third ligand molecule.
Table 3. Summary of the interactions between vitamins and metal ions.
Table 3. Summary of the interactions between vitamins and metal ions.
VitaminMetal
Ions
Experimental
Techniques
pHReferences
ThiaminZn(II)NMR, IR-[245]
Mn(II)NMR, EPR6.6[244]
Ni(II)NMR6.9[242,243]
RiboflavinZn(II)PT-[246]
FT-IR, LC-MS, AS-[247]
Cu(II)PT-[246]
Fe(III)PT-[246]
Mn(II)PT-[246]
Ni(II)PT-[246]
NiacinZn(II)UV-Vis, FT-IR, TGA, CV, MSM, MPD-[250]
Cu(II)PT, SP, UV-Vis5.0[248]
UV-Vis, FT-IR, TGA, CV, MSM, MPD-[250]
Mn(II)UV-Vis, FT-IR, TGA, CV, MSM, MPD-[250]
PT, TPD4.0[251]
Ni(II)PT, SP, UV-Vis5.0[248]
UV-Vis, FT-IR, TGA, CV, MSM, MPD-[250]
Vitamin B6Zn(II)X-ray-[259]
X-ray, DTA, FT-IR-[260]
PT, SP2.4–7.4 a[261]
Cu(II)PT, SP3.8–8.8 a[261]
Mn(II)NMR6.2, 7.0[257]
Ni(II)UV-Vis, MCV, TPD-[258]
PT, SP3.8–8.4 a[261]
BiotinZn(II)PT, NMR3.5–8.5 b[262]
NMR, UV-Vis2.0[263]
Cu(II)PT, NMR3.5–8.5 b[262]
NMR, UV-Vis2.0[263]
Mn(II)PT, NMR3.5–8.5 b[262]
NMR, UV-Vis2.0[263]
FolateCu(II)PT, conductometry>4.0[264]
EA, AA, polarimetry, FT-IR, DAEB7.6–7.8[265]
Fe(III)PT, conductometry>4.0[264]
EA, AA, polarimetry, FT-IR, DAEB7.6–7.8[265]
Mn(II)PT, conductometry>4.0[264]
Ni(II)PT, conductometry>4.0[264]
Al(III)PT, conductometry>4.0[264]
Vitamin B12Zn(II)NMR, UV-Vis, CD, F, MS, HPLC-DAD, X-ray, DFTC, SC6.0[267]
Ni(II)NMR, UV-Vis, CD, F, MS, HPLC-DAD, X-ray, DFTC, SC6.0[268]
Vitamin CZn(II)NMR, FT-IR6.0–7.0[252]
Cu(II)NMR, MS, IR, TGA, EA, SDCu, MSM-[256]
Fe(III)EA, MMD, UV-Vis, IR, AS8.0[255]
Mn(II)NMR, FT-IR6.0–7.0[252]
EA, MMD, UV-Vis, IR, AS8.0[255]
Ni(II)PT, NMR, DFTC>4.0[253]
Al(III)NMR, FT-IR6.0–7.0[254]
PT, NMR, DFTC>4.0[253]
Vitamin DZn(II)PT, SP>7.0[269]
Cu(II)PT, SP>2.0[266]
Fe(II)PT, SP>7.0[269]
Fe(III)PT, SP>2.0[269]
Mn(II)PT, SP>8.5[269]
Ni(II)PT, SP>8.0[266]
a pH range for the formation constant determination. b pH for the calculation of the stability constants. Abbreviations: AA, atomic absorption; AS, antimicrobial screening; CD, circular dichroism; CV, cyclic voltammetry; DAEB, determination of absorption efficiency in the blood; DFTC, density functional theory calculations; DTA, differential thermal analysis; EA, elemental analysis; EIS, enzyme inhibition studies; EPR, electron paramagnetic resonance; F, fluorescence; FT-IR, Fourier-transform infrared spectroscopy; HPLC-DAD, high-performance liquid chromatography with diode-array detection; IR, infrared; LC-MS, liquid chromatography-mass spectrometry; MCV, method of continuous variation; MMD, magnetic moment determination; MPD, melting point determination; MSM, magnetic susceptibility measurements; NMR, nuclear magnetic resonance; PT, potentiometric titration; SC, software computations; SDCu, spectrophotometrical determination of Cu; TGA, thermogravimetric analysis; TPD, thermodynamic parameters’ determination; UV-Vis, ultraviolet–visible.

5. Conclusions

Both vitamins and transition biometals such as copper, iron, and zinc are essential for brain health and development [66,270,271]. They are involved in a plethora of molecular processes guaranteeing cellular functioning. At the same time, they are also regulated by sophisticated homeostatic machinery preventing the accumulation of toxic species inside the cell.
For instance, copper is an essential metal ion for neural and glial function and it is necessary for the catalytic activity of antioxidant enzymes such as superoxide dismutase, and neurotransmitter-related enzymes, such as dopamine-β-monoxygenase [272]. Iron is implicated in oxygen delivery since it is an essential component of hemoglobin, and it is therefore crucial for brain metabolism. Zinc takes part in glutamatergic synaptic neurotransmission and is involved in the biological antioxidant system [273]. At the same time, copper and iron levels inside the cells need to be tightly controlled to avoid the overproduction of harmful ROS from copper/iron-catalyzed Haber–Weiss and Fenton reactions [274].
Water-soluble vitamins act as coenzymes capable of catalyzing a large number of chemical reactions essential for several cellular functions. For example, mitochondrial aerobic respiration and cellular energy production are dependent on the coenzymes derived from vitamins B1, B2, B3, and B5, which are directly involved in the citric acid cycle, the electron transport chain, and the resultant ATP formation. Indeed, vitamin B5 is a component of acetyl coenzyme A and is critical in the metabolism and synthesis of carbohydrates, proteins, and fats [271].
Whereas the brain is the most metabolically active organ in the body, B vitamins should have a particular impact on brain function and health. As for essential metal ions, their concentrations in the brain are tightly regulated by specific mechanisms ensuring vitamin transport across the blood–brain barrier and their correct distribution in the brain [275,276]. Moreover, as for copper, iron, and zinc ions, which are the three most abundant trace metals in the human brain [277], the vitamin concentration in the brain is usually higher than in the plasma [278].
The data reported in Table 3 have shown that all B vitamins share the ability to bind the selected metal ions, except vitamin B5. To our knowledge, no data have been collected so far on these associations; however, by considering the presence of carboxylic groups in their chemical structure, we would expect metal-vitamin B5 interactions comparable to the ones detected for vitamin B9.
Recently it has been shown that vitamin B9 can bind the tau protein, reducing its aggregation and β-sheet structure content [279]. Docking analysis has shown that folic acid interacts with Ser235, Gln244, Thr245, His268, and Gln269 in the proximity of the R1 region [279]. Interestingly, this region is also able to coordinate Zn(II), Fe(II), and Cu(II) [212,215], whose anchoring site is located at His268. By considering that folic acid can coordinate Cu(II) as well, in the future, it might be interesting to evaluate vitamins’ influence on the Cu(II)–R1tau interaction.
The two coenzymes derived from vitamin B2, flavin mononucleotide (FMN) and FAD, are involved in several biological processes: (i) The synthesis, conversion, and recycling of other B vitamins (B3, B6, and B9) and (ii) the synthesis of all heme proteins, among which are those involved in electron transfer and oxygen transport and storage [271]. Interestingly, riboflavin can bind the metal ions involved in both PD and AD and are discussed herein (vide supra). The reported outcomes point out the occurrence of stable metal complexes, which might reduce vitamin B2 bioavailability leading to riboflavin deficiency in the disease state, which, in turn, would result in severe mitochondrial dysfunctions. Although such a hypothesis needs to be further explored, it is well supported by previous contributions on metal-induced alterations in vitamins in birds [280]. The interaction of vitamin C with iron and copper has been widely investigated; nowadays, it is well-accepted that these reactions are catalytic and have unit stoichiometry. In the presence of these two metal ions, ascorbic acid loses its antioxidant properties, favoring the production of ROS species [124]. Moreover, ascorbic acid is able to tightly associate with Aβ and binds to Aβ42 oligomers at sites in the D23-K28 region [281]. In this respect, further investigations on ternary systems (Aβ, Cu(II), and vitamin C) may be useful to explain the chemical equilibria and the species forming in the solution.
Among the fat-soluble vitamins, only vitamin D has shown the ability to bind metal ions such as Zn(II), Cu(II), Fe(III), Fe(II), Mn(II), and Ni(II). In addition to the well-known role of vitamin D in calcium and bone metabolism, its neuroprotective effects have been recently described. Moreover, vitamin D is able to modulate the biosynthesis of neurotransmitters and neurotrophic factors, and its receptors are widespread in brain tissue [282,283]. Curiously, a recent study has investigated physical and chemical factors affecting vitamin D degradation, among which are metal ions such as Fe(II), Cu(I), and Cu(II) [284]. The obtained findings point out the occurrence of quick metal ion-mediated vitamin D3 degradation, regardless of the particular metal ion and its concentration. Metal-dependent degradation of vitamin D might be associated with the low levels of vitamin D found in both PD and AD (Figure 1).
In conclusion, this review points out the puzzling and fascinating role played by vitamins and metal ions in AD and PD. In this scenario, a new field focused on the bioinorganic chemistry of vitamins is emerging with the opportunity to illustrate the contribution of vitamin–biometal interactions to the well-known pathological implications associated with these two severe diseases.

Author Contributions

Conceptualization, A.K. and D.V.; methodology, A.K. and D.V.; validation, A.K. and D.V.; data curation, A.K., F.N. and D.V.; writing—original draft preparation, A.K. and F.N.; writing—review and editing, A.K., F.N. and D.V.; visualization, A.K. and F.N.; supervision, D.V. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

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

Acknowledgments

The “Consorzio Interuniversitario Risonanze Magnetiche di Metallo Proteine (CIRMMP)” and “Biosfera Nature” are kindly acknowledged.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Wang, Z.; Pierson, R.; Heymsfield, S. The Five-Level Model: A New Approach to Organizing Body-Composition Research. Am. J. Clin. Nutr. 1992, 56, 19–28. [Google Scholar] [CrossRef] [PubMed]
  2. Andrews, N.C. Disorders of Iron Metabolism. N. Engl. J. Med. 1999, 341, 1986–1995. [Google Scholar] [CrossRef] [PubMed]
  3. Moustakas, M. The Role of Metal Ions in Biology, Biochemistry and Medicine. Materials 2021, 14, 549. [Google Scholar] [CrossRef] [PubMed]
  4. Farina, M.; Avila, D.S.; da Rocha, J.B.T.; Aschner, M. Metals, Oxidative Stress and Neurodegeneration: A Focus on Iron, Manganese and Mercury. Neurochem. Int. 2013, 62, 575–594. [Google Scholar] [CrossRef] [Green Version]
  5. Slobodian, M.R.; Petahtegoose, J.D.; Wallis, A.L.; Levesque, D.C.; Merritt, T.J.S. The Effects of Essential and Non-Essential Metal Toxicity in the Drosophila Melanogaster Insect Model: A Review. Toxics 2021, 9, 269. [Google Scholar] [CrossRef]
  6. Lachowicz, J.I.; Lecca, L.I.; Meloni, F.; Campagna, M. Metals and Metal-Nanoparticles in Human Pathologies: From Exposure to Therapy. Molecules 2021, 26, 6639. [Google Scholar] [CrossRef]
  7. Chen, P.; Miah, M.R.; Aschner, M. Metals and Neurodegeneration. F1000Research 2016, 5, 366. [Google Scholar] [CrossRef] [Green Version]
  8. Cicero, C.E.; Mostile, G.; Vasta, R.; Rapisarda, V.; Signorelli, S.S.; Ferrante, M.; Zappia, M.; Nicoletti, A. Metals and Neurodegenerative Diseases. A Systematic Review. Environ. Res. 2017, 159, 82–94. [Google Scholar] [CrossRef]
  9. Shah, H.; Dehghani, F.; Ramezan, M.; Gannaban, R.B.; Haque, Z.F.; Rahimi, F.; Abbasi, S.; Shin, A.C. Revisiting the Role of Vitamins and Minerals in Alzheimer’s Disease. Antioxidants 2023, 12, 415. [Google Scholar] [CrossRef]
  10. Yamauchi, O.; Odani, A.; Takani, M. Metal–Amino Acid Chemistry. Weak Interactions and Related Functions of Side Chain Groups. J. Chem. Soc. Dalton Trans. 2002, 34, 3411–3421. [Google Scholar] [CrossRef]
  11. Liu, X.; Wu, M.; Li, C.; Yu, P.; Feng, S.; Li, Y.; Zhang, Q. Interaction Structure and Affinity of Zwitterionic Amino Acids with Important Metal Cations (Cd2+, Cu2+, Fe3+, Hg2+, Mn2+, Ni2+ and Zn2+) in Aqueous Solution: A Theoretical Study. Molecules 2022, 27, 2407. [Google Scholar] [CrossRef] [PubMed]
  12. Liu, Z.; Chen, S.; Qiao, F.; Zhang, X. Interaction of Peptide Backbones and Transition Metal Ions: 1. an IM-MS and DFT Study of the Binding Pattern, Structure and Fragmentation of Pd(II)/Ni(II)-Polyalanine Complexes. Int. J. Mass. Spectrom. 2019, 438, 87–96. [Google Scholar] [CrossRef]
  13. Di Natale, C.; De Benedictis, I.; De Benedictis, A.; Marasco, D. Metal–Peptide Complexes as Promising Antibiotics to Fight Emerging Drug Resistance: New Perspectives in Tuberculosis. Antibiotics 2020, 9, 337. [Google Scholar] [CrossRef] [PubMed]
  14. Witkowska, D.; Rowińska-Żyrek, M. Biophysical Approaches for the Study of Metal-Protein Interactions. J. Inorg. Biochem. 2019, 199, 110783. [Google Scholar] [CrossRef]
  15. Guo, C.; Cheng, M.; Gross, M.L. Protein-Metal-Ion Interactions Studied by Mass Spectrometry-Based Footprinting with Isotope-Encoded Benzhydrazide. Anal. Chem. 2019, 91, 1416–1423. [Google Scholar] [CrossRef]
  16. Wu, G. Amino Acids: Metabolism, Functions, and Nutrition. Amino Acids 2009, 37, 1–17. [Google Scholar] [CrossRef]
  17. Morris, R.; Black, K.A.; Stollar, E.J. Uncovering Protein Function: From Classification to Complexes. Essays Biochem. 2022, 66, 255–285. [Google Scholar] [CrossRef]
  18. Potocki, S.; Rowinska-Zyrek, M.; Witkowska, D.; Pyrkosz, M.; Szebesczyk, A.; Krzywoszynska, K.; Kozlowski, H. Metal Transport and Homeostasis within the Human Body: Toxicity Associated with Transport Abnormalities. Curr. Med. Chem. 2012, 19, 2738–2759. [Google Scholar] [CrossRef]
  19. Combs, G.F.; McClung, J.P. Chapter 1—What Is a Vitamin? In The Vitamins, 5th ed.; Combs, G.F., McClung, J.P., Eds.; Academic Press: Amsterdam, NL, USA; Boston, MA, USA, 2017; pp. 3–6. ISBN 978-0-12-802965-7. [Google Scholar]
  20. Fukuwatari, T.; Shibata, K. Nutritional Aspect of Tryptophan Metabolism. Int. J. Tryptophan Res. 2013, 6, 3–8. [Google Scholar] [CrossRef] [Green Version]
  21. Wacker, M.; Holick, M.F. Sunlight and Vitamin D: A Global Perspective for Health. Dermatoendocrinol 2013, 5, 51–108. [Google Scholar] [CrossRef] [Green Version]
  22. Combs, G.F.; McClung, J.P. Chapter 3—General Properties of Vitamins. In The Vitamins, 5th ed.; Combs, G.F., McClung, J.P., Eds.; Academic Press: Amsterdam, NL, USA; Boston, MA, USA, 2017; pp. 33–58. ISBN 978-0-12-802965-7. [Google Scholar]
  23. Rai, S.N.; Singh, P.; Steinbusch, H.W.M.; Vamanu, E.; Ashraf, G.; Singh, M.P. The Role of Vitamins in Neurodegenerative Disease: An Update. Biomedicines 2021, 9, 1284. [Google Scholar] [CrossRef] [PubMed]
  24. Przedborski, S.; Vila, M.; Jackson-Lewis, V. Series Introduction: Neurodegeneration: What Is It and Where Are We? J. Clin. Investig. 2003, 111, 3–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Wilson, D.M.; Cookson, M.R.; Van Den Bosch, L.; Zetterberg, H.; Holtzman, D.M.; Dewachter, I. Hallmarks of Neurodegenerative Diseases. Cell 2023, 186, 693–714. [Google Scholar] [CrossRef] [PubMed]
  26. Forrest, S.L.; Kovacs, G.G. Current Concepts of Mixed Pathologies in Neurodegenerative Diseases. Can. J. Neurol. Sci. 2023, 50, 329–345. [Google Scholar] [CrossRef] [PubMed]
  27. Lamptey, R.N.L.; Chaulagain, B.; Trivedi, R.; Gothwal, A.; Layek, B.; Singh, J. A Review of the Common Neurodegenerative Disorders: Current Therapeutic Approaches and the Potential Role of Nanotherapeutics. Int. J. Mol. Sci. 2022, 23, 1851. [Google Scholar] [CrossRef] [PubMed]
  28. Wyss-Coray, T. Ageing, Neurodegeneration and Brain Rejuvenation. Nature 2016, 539, 180–186. [Google Scholar] [CrossRef] [Green Version]
  29. López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The Hallmarks of Aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef] [Green Version]
  30. Hou, Y.; Dan, X.; Babbar, M.; Wei, Y.; Hasselbalch, S.G.; Croteau, D.L.; Bohr, V.A. Ageing as a Risk Factor for Neurodegenerative Disease. Nat. Rev. Neurol. 2019, 15, 565–581. [Google Scholar] [CrossRef]
  31. Sandeep; Sahu, M.R.; Rani, L.; Kharat, A.S.; Mondal, A.C. Could Vitamins Have a Positive Impact on the Treatment of Parkinson’s Disease? Brain Sci. 2023, 13, 272. [Google Scholar] [CrossRef]
  32. Kumar, R.R.; Singh, L.; Thakur, A.; Singh, S.; Kumar, B. Role of Vitamins in Neurodegenerative Diseases: A Review. CNS Neurol. Disord. Drug Targets 2022, 21, 766–773. [Google Scholar] [CrossRef]
  33. Modica, J.S.; Déry, C.; Canissario, R.; Logigian, E.; Bonno, D.; Stanton, M.; Dupré, N.; McDermott, M.P.; Bouchard, M.; Lang, A.E.; et al. A Systematic Review of the Potential Consequences of Abnormal Serum Levels of Vitamin B6 in People Living with Parkinson’s Disease. J. Neurol. Sci. 2023, 450, 120690. [Google Scholar] [CrossRef] [PubMed]
  34. Håglin, L.; Johansson, I.; Forsgren, L.; Bäckman, L. Intake of Vitamin B before Onset of Parkinson’s Disease and Atypical Parkinsonism and Olfactory Function at the Time of Diagnosis. Eur. J. Clin. Nutr. 2017, 71, 97–102. [Google Scholar] [CrossRef] [PubMed]
  35. Håglin, L.; Domellöf, M.; Bäckman, L.; Forsgren, L. Low Plasma Thiamine and Phosphate in Male Patients with Parkinson’s Disease Is Associated with Mild Cognitive Impairment. Clin. Nutr. ESPEN 2020, 37, 93–99. [Google Scholar] [CrossRef] [PubMed]
  36. De Lau, L.M.L.; Koudstaal, P.J.; Witteman, J.C.M.; Hofman, A.; Breteler, M.M.B. Dietary Folate, Vitamin B12, and Vitamin B6 and the Risk of Parkinson Disease. Neurology 2006, 67, 315–318. [Google Scholar] [CrossRef]
  37. Murakami, K.; Miyake, Y.; Sasaki, S.; Tanaka, K.; Fukushima, W.; Kiyohara, C.; Tsuboi, Y.; Yamada, T.; Oeda, T.; Miki, T.; et al. Dietary Intake of Folate, Vitamin B6, Vitamin B12 and Riboflavin and Risk of Parkinson’s Disease: A Case–Control Study in Japan. Br. J. Nutr. 2010, 104, 757–764. [Google Scholar] [CrossRef] [Green Version]
  38. Lohr, K.M.; Frost, B.; Scherzer, C.; Feany, M.B. Biotin Rescues Mitochondrial Dysfunction and Neurotoxicity in a Tauopathy Model. Proc. Natl. Acad. Sci. USA 2020, 117, 33608–33618. [Google Scholar] [CrossRef]
  39. Shen, L. Associations between B Vitamins and Parkinson’s Disease. Nutrients 2015, 7, 7197–7208. [Google Scholar] [CrossRef]
  40. McCarter, S.J.; Stang, C.; Turcano, P.; Mielke, M.M.; Ali, F.; Bower, J.H.; Savica, R. Higher Vitamin B12 Level at Parkinson’s Disease Diagnosis Is Associated with Lower Risk of Future Dementia. Park. Relat. Disord. 2020, 73, 19–22. [Google Scholar] [CrossRef]
  41. Scholefield, M.; Church, S.J.; Xu, J.; Patassini, S.; Hooper, N.M.; Unwin, R.D.; Cooper, G.J.S. Substantively Lowered Levels of Pantothenic Acid (Vitamin B5) in Several Regions of the Human Brain in Parkinson’s Disease Dementia. Metabolites 2021, 11, 569. [Google Scholar] [CrossRef]
  42. Costantini, A.; Fancellu, R. An Open-Label Pilot Study with High-Dose Thiamine in Parkinson’s Disease. Neural Regen. Res. 2016, 11, 406. [Google Scholar] [CrossRef]
  43. Coimbra, C.G.; Junqueira, V.B.C. High Doses of Riboflavin and the Elimination of Dietary Red Meat Promote the Recovery of Some Motor Functions in Parkinson’s Disease Patients. Braz. J. Med. Biol. Res. 2003, 36, 1409–1417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Jia, H.; Li, X.; Gao, H.; Feng, Z.; Li, X.; Zhao, L.; Jia, X.; Zhang, H.; Liu, J. High Doses of Nicotinamide Prevent Oxidative Mitochondrial Dysfunction in a Cellular Model and Improve Motor Deficit in a Drosophila Model of Parkinson’s Disease. J. Neurosci. Res. 2008, 86, 2083–2090. [Google Scholar] [CrossRef] [PubMed]
  45. Wu, Y.; Zhao, Z.; Yang, N.; Xin, C.; Li, Z.; Xu, J.; Ma, B.; Lim, K.-L.; Li, L.; Wu, Q.; et al. Vitamin B12 Ameliorates the Pathological Phenotypes of Multiple Parkinson’s Disease Models by Alleviating Oxidative Stress. Antioxidants 2023, 12, 153. [Google Scholar] [CrossRef] [PubMed]
  46. Kobayashi, T.; Matsumine, H.; Matuda, S.; Mizuno, Y. Association between the Gene Encoding the E2 Subunit of the Alpha-Ketoglutarate Dehydrogenase Complex and Parkinson’s Disease. Ann. Neurol. 1998, 43, 120–123. [Google Scholar] [CrossRef]
  47. Naseri, N.N.; Xu, H.; Bonica, J.; Vonsattel, J.P.G.; Cortes, E.P.; Park, L.C.; Arjomand, J.; Gibson, G.E. Abnormalities in the Tricarboxylic Acid Cycle in Huntington Disease and in a Huntington Disease Mouse Model. J. Neuropathol. Exp. Neurol. 2015, 74, 527–537. [Google Scholar] [CrossRef] [Green Version]
  48. Tretter, L.; Adam-Vizi, V. Alpha-Ketoglutarate Dehydrogenase: A Target and Generator of Oxidative Stress. Philos. Trans. R. Soc. Lond. B. Biol. Sci. 2005, 360, 2335–2345. [Google Scholar] [CrossRef] [Green Version]
  49. Hansen, G.E.; Gibson, G.E. The α-Ketoglutarate Dehydrogenase Complex as a Hub of Plasticity in Neurodegeneration and Regeneration. Int. J. Mol. Sci. 2022, 23, 12403. [Google Scholar] [CrossRef]
  50. Mooney, S.; Leuendorf, J.-E.; Hendrickson, C.; Hellmann, H. Vitamin B6: A Long Known Compound of Surprising Complexity. Molecules 2009, 14, 329–351. [Google Scholar] [CrossRef] [Green Version]
  51. Sato, K. Why Is Vitamin B6 Effective in Alleviating the Symptoms of Autism? Med. Hypotheses 2018, 115, 103–106. [Google Scholar] [CrossRef]
  52. Al-Kuraishy, H.M.; Al-Gareeb, A.I.; Elewa, Y.H.A.; Zahran, M.H.; Alexiou, A.; Papadakis, M.; Batiha, G.E.-S. Parkinson’s Disease Risk and Hyperhomocysteinemia: The Possible Link. Cell. Mol. Neurobiol. 2023, 43, 2743–2759. [Google Scholar] [CrossRef]
  53. Price, B.R.; Wilcock, D.M.; Weekman, E.M. Hyperhomocysteinemia as a Risk Factor for Vascular Contributions to Cognitive Impairment and Dementia. Front. Aging Neurosci. 2018, 10, 350. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Fan, X.; Zhang, L.; Li, H.; Chen, G.; Qi, G.; Ma, X.; Jin, Y. Role of Homocysteine in the Development and Progression of Parkinson’s Disease. Ann. Clin. Transl. Neurol. 2020, 7, 2332–2338. [Google Scholar] [CrossRef]
  55. Luzzi, S.; Cherubini, V.; Falsetti, L.; Viticchi, G.; Silvestrini, M.; Toraldo, A. Homocysteine, Cognitive Functions, and Degenerative Dementias: State of the Art. Biomedicines 2022, 10, 2741. [Google Scholar] [CrossRef]
  56. Giri, B.; Belanger, K.; Seamon, M.; Bradley, E.; Purohit, S.; Chong, R.; Morgan, J.C.; Baban, B.; Wakade, C. Niacin Ameliorates Neuro-Inflammation in Parkinson’s Disease via GPR109A. Int. J. Mol. Sci. 2019, 20, 4559. [Google Scholar] [CrossRef] [Green Version]
  57. Wakade, C.; Chong, R.; Bradley, E.; Thomas, B.; Morgan, J. Upregulation of GPR109A in Parkinson’s Disease. PLoS ONE 2014, 9, e109818. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Wang, W.; Li, Y.; Meng, X. Vitamin D and Neurodegenerative Diseases. Heliyon 2023, 9, e12877. [Google Scholar] [CrossRef] [PubMed]
  59. Palermo, S.; Stanziano, M.; Nigri, A.; Civilotti, C.; Celeghin, A. Parkinson’s Disease, SARS-CoV-2, and Frailty: Is There a Vicious Cycle Related to Hypovitaminosis D? Brain Sci. 2023, 13, 528. [Google Scholar] [CrossRef]
  60. Lasoń, W.; Jantas, D.; Leśkiewicz, M.; Regulska, M.; Basta-Kaim, A. The Vitamin D Receptor as a Potential Target for the Treatment of Age-Related Neurodegenerative Diseases Such as Alzheimer’s and Parkinson’s Diseases: A Narrative Review. Cells 2023, 12, 660. [Google Scholar] [CrossRef]
  61. Behl, T.; Arora, A.; Singla, R.K.; Sehgal, A.; Makeen, H.A.; Albratty, M.; Meraya, A.M.; Najmi, A.; Bungau, S.G. Understanding the Role of “Sunshine Vitamin D” in Parkinson’s Disease: A Review. Front. Pharmacol. 2022, 13, 993033. [Google Scholar] [CrossRef]
  62. Barichella, M.; Garrì, F.; Caronni, S.; Bolliri, C.; Zocchi, L.; Macchione, M.C.; Ferri, V.; Calandrella, D.; Pezzoli, G. Vitamin D Status and Parkinson’s Disease. Brain Sci. 2022, 12, 790. [Google Scholar] [CrossRef]
  63. Lv, L.; Tan, X.; Peng, X.; Bai, R.; Xiao, Q.; Zou, T.; Tan, J.; Zhang, H.; Wang, C. The Relationships of Vitamin D, Vitamin D Receptor Gene Polymorphisms, and Vitamin D Supplementation with Parkinson’s Disease. Transl. Neurodegener. 2020, 9, 34. [Google Scholar] [CrossRef] [PubMed]
  64. Zhang, H.-J.; Zhang, J.-R.; Mao, C.-J.; Li, K.; Wang, F.; Chen, J.; Liu, C.-F. Relationship between 25-Hydroxyvitamin D, Bone Density, and Parkinson’s Disease Symptoms. Acta Neurol. Scand. 2019, 140, 274–280. [Google Scholar] [CrossRef]
  65. Bayo-Olugbami, A.; Nafiu, A.B.; Amin, A.; Ogundele, O.M.; Lee, C.C.; Owoyele, B.V. Vitamin D Attenuated 6-OHDA-Induced Behavioural Deficits, Dopamine Dysmetabolism, Oxidative Stress, and Neuro-Inflammation in Mice. Nutr. Neurosci. 2022, 25, 823–834. [Google Scholar] [CrossRef] [PubMed]
  66. Cortés-Albornoz, M.C.; García-Guáqueta, D.P.; Velez-van-Meerbeke, A.; Talero-Gutiérrez, C. Maternal Nutrition and Neurodevelopment: A Scoping Review. Nutrients 2021, 13, 3530. [Google Scholar] [CrossRef] [PubMed]
  67. Anjum, I.; Jaffery, S.S.; Fayyaz, M.; Samoo, Z.; Anjum, S. The Role of Vitamin D in Brain Health: A Mini Literature Review. Cureus 2018, 10, e2960. [Google Scholar] [CrossRef] [Green Version]
  68. Gezen-Ak, D.; Dursun, E.; Yilmazer, S. The Effect of Vitamin D Treatment On Nerve Growth Factor (NGF) Release From Hippocampal Neurons. Noro Psikiyatr. Ars. 2014, 51, 157–162. [Google Scholar] [CrossRef] [Green Version]
  69. Pertile, R.A.N.; Brigden, R.; Raman, V.; Cui, X.; Du, Z.; Eyles, D. Vitamin D: A Potent Regulator of Dopaminergic Neuron Differentiation and Function. J. Neurochem. 2023. online ahead of print. [Google Scholar] [CrossRef]
  70. Emekli-Alturfan, E.; Alturfan, A.A. The Emerging Relationship between Vitamin K and Neurodegenerative Diseases: A Review of Current Evidence. Mol. Biol. Rep. 2023, 50, 815–828. [Google Scholar] [CrossRef]
  71. Tamadon-Nejad, S.; Ouliass, B.; Rochford, J.; Ferland, G. Vitamin K Deficiency Induced by Warfarin Is Associated With Cognitive and Behavioral Perturbations, and Alterations in Brain Sphingolipids in Rats. Front. Aging Neurosci. 2018, 10, 213. [Google Scholar] [CrossRef] [Green Version]
  72. Yu, Y.-X.; Yu, X.-D.; Cheng, Q.; Tang, L.; Shen, M.-Q. The Association of Serum Vitamin K2 Levels with Parkinson’s Disease: From Basic Case-Control Study to Big Data Mining Analysis. Aging 2020, 12, 16410–16419. [Google Scholar] [CrossRef]
  73. Vos, M.; Esposito, G.; Edirisinghe, J.N.; Vilain, S.; Haddad, D.M.; Slabbaert, J.R.; Van Meensel, S.; Schaap, O.; De Strooper, B.; Meganathan, R.; et al. Vitamin K2 Is a Mitochondrial Electron Carrier That Rescues Pink1 Deficiency. Science 2012, 336, 1306–1310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Tang, H.; Zheng, Z.; Wang, H.; Wang, L.; Zhao, G.; Wang, P. Vitamin K2 Modulates Mitochondrial Dysfunction Induced by 6-Hydroxydopamine in SH-SY5Y Cells via Mitochondrial Quality-Control Loop. Nutrients 2022, 14, 1504. [Google Scholar] [CrossRef] [PubMed]
  75. Ferland, G. Vitamin K, an Emerging Nutrient in Brain Function. BioFactors 2012, 38, 151–157. [Google Scholar] [CrossRef] [PubMed]
  76. da Silva, F.L.; Coelho Cerqueira, E.; de Freitas, M.S.; Gonçalves, D.L.; Costa, L.T.; Follmer, C. Vitamins K Interact with N-Terminus α-Synuclein and Modulate the Protein Fibrillization in Vitro. Exploring the Interaction between Quinones and α-Synuclein. Neurochem. Int. 2013, 62, 103–112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Ono, K.; Yamada, M. Vitamin A Potently Destabilizes Preformed α-Synuclein Fibrils in Vitro: Implications for Lewy Body Diseases. Neurobiol. Dis. 2007, 25, 446–454. [Google Scholar] [CrossRef]
  78. Yin, L.-H.; Shen, H.; Diaz-Ruiz, O.; Bäckman, C.M.; Bae, E.; Yu, S.-J.; Wang, Y. Early Post-Treatment with 9-Cis Retinoic Acid Reduces Neurodegeneration of Dopaminergic Neurons in a Rat Model of Parkinson’s Disease. BMC Neurosci. 2012, 13, 120. [Google Scholar] [CrossRef] [Green Version]
  79. Kunzler, A.; Ribeiro, C.T.; Gasparotto, J.; Petiz, L.L.; da Rosa Silva, H.T.; da Silva, J.D.; Bortolin, R.; de Souza, P.O.; Barreto, F.; Espitia-Perez, P.; et al. The Effects of Retinol Oral Supplementation in 6-Hydroxydopamine Dopaminergic Denervation Model in Wistar Rats. Neurochem. Int. 2019, 125, 25–34. [Google Scholar] [CrossRef]
  80. Rusu, M.E.; Fizesan, I.; Pop, A.; Mocan, A.; Gheldiu, A.-M.; Babota, M.; Vodnar, D.C.; Jurj, A.; Berindan-Neagoe, I.; Vlase, L.; et al. Walnut (Juglans Regia L.) Septum: Assessment of Bioactive Molecules and In Vitro Biological Effects. Molecules 2020, 25, 2187. [Google Scholar] [CrossRef]
  81. Lloret, A.; Esteve, D.; Monllor, P.; Cervera-Ferri, A.; Lloret, A. The Effectiveness of Vitamin E Treatment in Alzheimer’s Disease. Int. J. Mol. Sci. 2019, 20, 879. [Google Scholar] [CrossRef] [Green Version]
  82. Casani, S.; Gómez-Pastor, R.; Matallana, E.; Paricio, N. Antioxidant Compound Supplementation Prevents Oxidative Damage in a Drosophila Model of Parkinson’s Disease. Free Radic. Biol. Med. 2013, 61, 151–160. [Google Scholar] [CrossRef]
  83. Sharma, N.; Nehru, B. Beneficial Effect of Vitamin E in Rotenone Induced Model of PD: Behavioural, Neurochemical and Biochemical Study. Exp. Neurobiol. 2013, 22, 214–223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Paraskevas, G.P.; Kapaki, E.; Petropoulou, O.; Anagnostouli, M.; Vagenas, V.; Papageorgiou, C. Plasma Levels of Antioxidant Vitamins C and E Are Decreased in Vascular Parkinsonism. J. Neurol. Sci. 2003, 215, 51–55. [Google Scholar] [CrossRef] [PubMed]
  85. Ide, K.; Yamada, H.; Umegaki, K.; Mizuno, K.; Kawakami, N.; Hagiwara, Y.; Matsumoto, M.; Yoshida, H.; Kim, K.; Shiosaki, E.; et al. Lymphocyte Vitamin C Levels as Potential Biomarker for Progression of Parkinson’s Disease. Nutrition 2015, 31, 406–408. [Google Scholar] [CrossRef]
  86. Ying, A.F.; Khan, S.; Wu, Y.; Jin, A.; Wong, A.S.Y.; Tan, E.-K.; Yuan, J.-M.; Koh, W.-P.; Tan, L.C.S. Dietary Antioxidants and Risk of Parkinson’s Disease in the Singapore Chinese Health Study. Mov. Disord. 2020, 35, 1765–1773. [Google Scholar] [CrossRef] [PubMed]
  87. Chang, M.C.; Kwak, S.G.; Kwak, S. Effect of Dietary Vitamins C and E on the Risk of Parkinson’s Disease: A Meta-Analysis. Clin. Nutr. 2021, 40, 3922–3930. [Google Scholar] [CrossRef]
  88. Malaguarnera, M.; Ferri, R.; Bella, R.; Alagona, G.; Carnemolla, A.; Pennisi, G. Homocysteine, Vitamin B12 and Folate in Vascular Dementia and in Alzheimer Disease. Clin. Chem. Lab. Med. CCLM 2004, 42, 1032–1035. [Google Scholar] [CrossRef]
  89. Chen, H.; Liu, S.; Ji, L.; Wu, T.; Ma, F.; Ji, Y.; Zhou, Y.; Zheng, M.; Zhang, M.; Huang, G. Associations between Alzheimer’s Disease and Blood Homocysteine, Vitamin B12, and Folate: A Case-Control Study. Curr. Alzheimer Res. 2015, 12, 88–94. [Google Scholar] [CrossRef] [Green Version]
  90. Froese, D.S.; Fowler, B.; Baumgartner, M.R. Vitamin B12, Folate, and the Methionine Remethylation Cycle-Biochemistry, Pathways, and Regulation. J. Inherit. Metab. Dis. 2019, 42, 673–685. [Google Scholar] [CrossRef] [Green Version]
  91. Durga, J.; van Boxtel, M.P.J.; Schouten, E.G.; Kok, F.J.; Jolles, J.; Katan, M.B.; Verhoef, P. Effect of 3-Year Folic Acid Supplementation on Cognitive Function in Older Adults in the FACIT Trial: A Randomised, Double Blind, Controlled Trial. Lancet Lond. Engl. 2007, 369, 208–216. [Google Scholar] [CrossRef] [Green Version]
  92. Sun, Y.; Lu, C.-J.; Chien, K.-L.; Chen, S.-T.; Chen, R.-C. Efficacy of Multivitamin Supplementation Containing Vitamins B6 and B12 and Folic Acid as Adjunctive Treatment with a Cholinesterase Inhibitor in Alzheimer’s Disease: A 26-Week, Randomized, Double-Blind, Placebo-Controlled Study in Taiwanese Patients. Clin. Ther. 2007, 29, 2204–2214. [Google Scholar] [CrossRef]
  93. Aisen, P.S.; Schneider, L.S.; Sano, M.; Diaz-Arrastia, R.; van Dyck, C.H.; Weiner, M.F.; Bottiglieri, T.; Jin, S.; Stokes, K.T.; Thomas, R.G.; et al. High Dose B Vitamin Supplementation and Cognitive Decline in Alzheimer’s Disease: A Randomized Controlled Trial. JAMA J. Am. Med. Assoc. 2008, 300, 1774–1783. [Google Scholar] [CrossRef] [PubMed]
  94. Meng, H.; Li, Y.; Zhang, W.; Zhao, Y.; Niu, X.; Guo, J. The Relationship between Cognitive Impairment and Homocysteine in a B12 and Folate Deficient Population in China: A Cross-Sectional Study. Med. Baltim. 2019, 98, e17970. [Google Scholar] [CrossRef] [PubMed]
  95. Lanyau-Domínguez, Y.; Macías-Matos, C.; Llibre-Rodríguez, J.d.J.; Pita-Rodríguez, G.M.; Suárez-Medina, R.; Quintero-Alejo, M.E.; Noriega-Fernández, L.; Guerra-Hernández, M.; Calvo-Rodríguez, M.; Sánchez-Gil, Y.; et al. Levels of Vitamins and Homocysteine in Older Adults with Alzheimer Disease or Mild Cognitive Impairment in Cuba. MEDICC Rev. 2020, 22, 40–47. [Google Scholar] [PubMed]
  96. Ma, F.; Wu, T.; Zhao, J.; Ji, L.; Song, A.; Zhang, M.; Huang, G. Plasma Homocysteine and Serum Folate and Vitamin B12 Levels in Mild Cognitive Impairment and Alzheimer’s Disease: A Case-Control Study. Nutrients 2017, 9, 725. [Google Scholar] [CrossRef] [Green Version]
  97. Yu, L.; Chen, Y.; Wang, W.; Xiao, Z.; Hong, Y. Multi-Vitamin B Supplementation Reverses Hypoxia-Induced Tau Hyperphosphorylation and Improves Memory Function in Adult Mice. J. Alzheimers Dis. 2016, 54, 297–306. [Google Scholar] [CrossRef]
  98. Lauer, A.A.; Grimm, H.S.; Apel, B.; Golobrodska, N.; Kruse, L.; Ratanski, E.; Schulten, N.; Schwarze, L.; Slawik, T.; Sperlich, S.; et al. Mechanistic Link between Vitamin B12 and Alzheimer’s Disease. Biomolecules 2022, 12, 129. [Google Scholar] [CrossRef]
  99. Rafiee, S.; Asadollahi, K.; Riazi, G.; Ahmadian, S.; Saboury, A.A. Vitamin B12 Inhibits Tau Fibrillization via Binding to Cysteine Residues of Tau. ACS Chem. Neurosci. 2017, 8, 2676–2682. [Google Scholar] [CrossRef]
  100. Moore, K.; Hughes, C.F.; Ward, M.; Hoey, L.; McNulty, H. Diet, Nutrition and the Ageing Brain: Current Evidence and New Directions. Proc. Nutr. Soc. 2018, 77, 152–163. [Google Scholar] [CrossRef] [Green Version]
  101. Zhao, R.; Wang, H.; Qiao, C.; Zhao, K. Vitamin B2 Blocks Development of Alzheimer’s Disease in APP/PS1 Transgenic Mice via Anti-Oxidative Mechanism. Trop. J. Pharm. Res. 2018, 17, 1049. [Google Scholar] [CrossRef]
  102. Seshadri, S.; Beiser, A.; Selhub, J.; Jacques, P.F.; Rosenberg, I.H.; D’Agostino, R.B.; Wilson, P.W.F.; Wolf, P.A. Plasma Homocysteine as a Risk Factor for Dementia and Alzheimer’s Disease. N. Engl. J. Med. 2002, 346, 476–483. [Google Scholar] [CrossRef]
  103. Guenther, B.D.; Sheppard, C.A.; Tran, P.; Rozen, R.; Matthews, R.G.; Ludwig, M.L. The Structure and Properties of Methylenetetrahydrofolate Reductase from Escherichia Coli Suggest How Folate Ameliorates Human Hyperhomocysteinemia. Nat. Struct. Biol. 1999, 6, 359–365. [Google Scholar] [CrossRef] [PubMed]
  104. Gibson, G.E.; Sheu, K.F.; Blass, J.P.; Baker, A.; Carlson, K.C.; Harding, B.; Perrino, P. Reduced Activities of Thiamine-Dependent Enzymes in the Brains and Peripheral Tissues of Patients with Alzheimer’s Disease. Arch. Neurol. 1988, 45, 836–840. [Google Scholar] [CrossRef] [PubMed]
  105. Gold, M.; Hauser, R.A.; Chen, M.F. Plasma Thiamine Deficiency Associated with Alzheimer’s Disease but Not Parkinson’s Disease. Metab. Brain Dis. 1998, 13, 43–53. [Google Scholar] [CrossRef] [PubMed]
  106. Gibson, G.E.; Hirsch, J.A.; Fonzetti, P.; Jordon, B.D.; Cirio, R.T.; Elder, J. Vitamin B1 (Thiamine) and Dementia. Ann. N. Y. Acad. Sci. 2016, 1367, 21–30. [Google Scholar] [CrossRef] [Green Version]
  107. Morris, M.; Evans, D.; Bienias, J.; Scherr, P.; Tangney, C.; Hebert, L.; Bennett, D.; Wilson, R.; Aggarwal, N. Dietary Niacin and the Risk of Incident Alzheimer’s Disease and of Cognitive Decline. J. Neurol. Neurosurg. Psychiatry 2004, 75, 1093–1099. [Google Scholar] [CrossRef] [Green Version]
  108. Rainer, M.; Kraxberger, E.; Haushofer, M.; Mucke, H.A.M.; Jellinger, K.A. No Evidence for Cognitive Improvement from Oral Nicotinamide Adenine Dinucleotide (NADH) in Dementia. J. Neural Transm. 2000, 107, 1475–1481. [Google Scholar] [CrossRef]
  109. Green, K.N.; Steffan, J.S.; Martinez-Coria, H.; Sun, X.; Schreiber, S.S.; Thompson, L.M.; LaFerla, F.M. Nicotinamide Restores Cognition in Alzheimer’s Disease Transgenic Mice via a Mechanism Involving Sirtuin Inhibition and Selective Reduction of Thr231-Phosphotau. J. Neurosci. 2008, 28, 11500–11510. [Google Scholar] [CrossRef] [Green Version]
  110. Ewers, M.; Buerger, K.; Teipel, S.J.; Scheltens, P.; Schröder, J.; Zinkowski, R.P.; Bouwman, F.H.; Schönknecht, P.; Schoonenboom, N.S.M.; Andreasen, N.; et al. Multicenter Assessment of CSF-Phosphorylated Tau for the Prediction of Conversion of MCI. Neurology 2007, 69, 2205–2212. [Google Scholar] [CrossRef]
  111. Turunc Bayrakdar, E.; Uyanikgil, Y.; Kanit, L.; Koylu, E.; Yalcin, A. Nicotinamide Treatment Reduces the Levels of Oxidative Stress, Apoptosis, and PARP-1 Activity in Aβ(1–42)-Induced Rat Model of Alzheimer’s Disease. Free Radic. Res. 2014, 48, 146–158. [Google Scholar] [CrossRef]
  112. Attia, H.; Albuhayri, S.; Alaraidh, S.; Alotaibi, A.; Yacoub, H.; Mohamad, R.; Al-Amin, M. Biotin, Coenzyme Q10, and Their Combination Ameliorate Aluminium Chloride-Induced Alzheimer’s Disease via Attenuating Neuroinflammation and Improving Brain Insulin Signaling. J. Biochem. Mol. Toxicol. 2020, 34, e22519. [Google Scholar] [CrossRef]
  113. Xu, J.; Patassini, S.; Begley, P.; Church, S.; Waldvogel, H.J.; Faull, R.L.M.; Unwin, R.D.; Cooper, G.J.S. Cerebral Deficiency of Vitamin B5 (d-Pantothenic Acid; Pantothenate) as a Potentially-Reversible Cause of Neurodegeneration and Dementia in Sporadic Alzheimer’s Disease. Biochem. Biophys. Res. Commun. 2020, 527, 676–681. [Google Scholar] [CrossRef] [PubMed]
  114. Sang, C.; Philbert, S.A.; Hartland, D.; Unwin, R.D.; Dowsey, A.W.; Xu, J.; Cooper, G.J.S. Coenzyme A-Dependent Tricarboxylic Acid Cycle Enzymes Are Decreased in Alzheimer’s Disease Consistent With Cerebral Pantothenate Deficiency. Front. Aging Neurosci. 2022, 14, 893159. [Google Scholar] [CrossRef] [PubMed]
  115. Basambombo, L.L.; Carmichael, P.-H.; Côté, S.; Laurin, D. Use of Vitamin E and C Supplements for the Prevention of Cognitive Decline. Ann. Pharmacother. 2017, 51, 118–124. [Google Scholar] [CrossRef] [PubMed]
  116. de Wilde, M.C.; Vellas, B.; Girault, E.; Yavuz, A.C.; Sijben, J.W. Lower Brain and Blood Nutrient Status in Alzheimer’s Disease: Results from Meta-Analyses. Alzheimers Dement. Transl. Res. Clin. Interv. 2017, 3, 416–431. [Google Scholar] [CrossRef]
  117. Jiménez-Jiménez, F.J.; De Bustos, F.; Molina, J.A.; Benito-León, J.; Tallón-Barranco, A.; Gasalla, T.; Ortí-Pareja, M.; Guillamón, F.; Rubio, J.C.; Arenas, J.; et al. Cerebrospinal Fluid Levels of Alpha-Tocopherol (Vitamin E) in Alzheimer’s Disease. J. Neural Transm. 1997, 104, 703–710. [Google Scholar] [CrossRef]
  118. Casati, M.; Boccardi, V.; Ferri, E.; Bertagnoli, L.; Bastiani, P.; Ciccone, S.; Mansi, M.; Scamosci, M.; Rossi, P.D.; Mecocci, P.; et al. Vitamin E and Alzheimer’s Disease: The Mediating Role of Cellular Aging. Aging Clin. Exp. Res. 2020, 32, 459–464. [Google Scholar] [CrossRef]
  119. Gray, S.L.; Anderson, M.L.; Crane, P.K.; Breitner, J.C.S.; McCormick, W.; Bowen, J.D.; Teri, L.; Larson, E. Antioxidant Vitamin Supplement Use and Risk of Dementia or Alzheimer’s Disease in Older Adults. J. Am. Geriatr. Soc. 2008, 56, 291–295. [Google Scholar] [CrossRef]
  120. Murakami, K.; Murata, N.; Ozawa, Y.; Kinoshita, N.; Irie, K.; Shirasawa, T.; Shimizu, T. Vitamin C Restores Behavioral Deficits and Amyloid-β Oligomerization without Affecting Plaque Formation in a Mouse Model of Alzheimer’s Disease. J. Alzheimers Dis. 2011, 26, 7–18. [Google Scholar] [CrossRef]
  121. Sil, S.; Ghosh, T.; Gupta, P.; Ghosh, R.; Kabir, S.N.; Roy, A. Dual Role of Vitamin C on the Neuroinflammation Mediated Neurodegeneration and Memory Impairments in Colchicine Induced Rat Model of Alzheimer Disease. J. Mol. Neurosci. 2016, 60, 421–435. [Google Scholar] [CrossRef]
  122. Arlt, S.; Müller-Thomsen, T.; Beisiegel, U.; Kontush, A. Effect of One-Year Vitamin C-and E-Supplementation on Cerebrospinal Fluid Oxidation Parameters and Clinical Course in Alzheimer’s Disease. Neurochem. Res. 2012, 37, 2706–2714. [Google Scholar] [CrossRef]
  123. Ide, K.; Yamada, H.; Kawasaki, Y.; Yamanaka, M.; Kawakami, N.; Katsuyama, Y.; Yoshida, H.; Kim, K.; Shiosaki, E.; Sonoda, A.; et al. Peripheral Vitamin C Levels in Alzheimer’s Disease: A Cross-Sectional Study. J. Nutr. Sci. Vitaminol. 2016, 62, 432–436. [Google Scholar] [CrossRef] [Green Version]
  124. Shen, J.; Griffiths, P.T.; Campbell, S.J.; Utinger, B.; Kalberer, M.; Paulson, S.E. Ascorbate Oxidation by Iron, Copper and Reactive Oxygen Species: Review, Model Development, and Derivation of Key Rate Constants. Sci. Rep. 2021, 11, 7417. [Google Scholar] [CrossRef]
  125. Dysken, M.W.; Sano, M.; Asthana, S.; Vertrees, J.E.; Pallaki, M.; Llorente, M.; Love, S.; Schellenberg, G.D.; McCarten, J.R.; Malphurs, J.; et al. Effect of Vitamin E and Memantine on Functional Decline in Alzheimer Disease. JAMA J. Am. Med. Assoc. 2014, 311, 33–44. [Google Scholar] [CrossRef] [PubMed]
  126. Mehrabadi, S.; Sadr, S.S. Administration of Vitamin D3 and E Supplements Reduces Neuronal Loss and Oxidative Stress in a Model of Rats with Alzheimer’s Disease. Neurol. Res. 2020, 42, 862–868. [Google Scholar] [CrossRef] [PubMed]
  127. Yatin, S.M.; Varadarajan, S.; Butterfield, D.A. Vitamin E Prevents Alzheimer’s Amyloid Beta-Peptide (1-42)-Induced Neuronal Protein Oxidation and Reactive Oxygen Species Production. J. Alzheimers Dis. JAD 2000, 2, 123–131. [Google Scholar] [CrossRef] [PubMed]
  128. Liu, G.; Zhao, Y.; Jin, S.; Hu, Y.; Wang, T.; Tian, R.; Han, Z.; Xu, D.; Jiang, Q. Circulating Vitamin E Levels and Alzheimer’s Disease: A Mendelian Randomization Study. Neurobiol. Aging 2018, 72, 189.e1–189.e9. [Google Scholar] [CrossRef]
  129. Lloret, A.; Badía, M.-C.; Mora, N.J.; Pallardó, F.V.; Alonso, M.-D.; Viña, J. Vitamin E Paradox in Alzheimer’s Disease: It Does Not Prevent Loss of Cognition and May Even Be Detrimental. J. Alzheimers Dis. 2009, 17, 143–149. [Google Scholar] [CrossRef]
  130. Mullan, K.; Williams, M.A.; Cardwell, C.R.; McGuinness, B.; Passmore, P.; Silvestri, G.; Woodside, J.V.; McKay, G.J. Serum Concentrations of Vitamin E and Carotenoids Are Altered in Alzheimer’s Disease: A Case-Control Study. Alzheimers Dement. Transl. Res. Clin. Interv. 2017, 3, 432–439. [Google Scholar] [CrossRef] [Green Version]
  131. Lopes da Silva, S.; Vellas, B.; Elemans, S.; Luchsinger, J.; Kamphuis, P.; Yaffe, K.; Sijben, J.; Groenendijk, M.; Stijnen, T. Plasma Nutrient Status of Patients with Alzheimer’s Disease: Systematic Review and Meta-Analysis. Alzheimers Dement. 2014, 10, 485–502. [Google Scholar] [CrossRef] [Green Version]
  132. Kapoor, A.; Wang, B.-J.; Hsu, W.-M.; Chang, M.-Y.; Liang, S.-M.; Liao, Y.-F. Retinoic Acid-Elicited RARα/RXRα Signaling Attenuates Aβ Production by Directly Inhibiting γ-Secretase-Mediated Cleavage of Amyloid Precursor Protein. ACS Chem. Neurosci. 2013, 4, 1093–1100. [Google Scholar] [CrossRef] [Green Version]
  133. Hira, S.; Saleem, U.; Anwar, F.; Sohail, M.F.; Raza, Z.; Ahmad, B. β-Carotene: A Natural Compound Improves Cognitive Impairment and Oxidative Stress in a Mouse Model of Streptozotocin-Induced Alzheimer’s Disease. Biomolecules 2019, 9, 441. [Google Scholar] [CrossRef] [Green Version]
  134. Geng, T.; Lu, Q.; Wan, Z.; Guo, J.; Liu, L.; Pan, A.; Liu, G. Association of Serum 25-Hydroxyvitamin D Concentrations with Risk of Dementia among Individuals with Type 2 Diabetes: A Cohort Study in the UK Biobank. PLOS Med. 2022, 19, e1003906. [Google Scholar] [CrossRef] [PubMed]
  135. Licher, S.; De Bruijn, R.F.A.G.; Wolters, F.J.; Zillikens, M.C.; Ikram, M.A.; Ikram, M.K. Vitamin D and the Risk of Dementia: The Rotterdam Study. J. Alzheimers Dis. 2017, 60, 989–997. [Google Scholar] [CrossRef] [PubMed]
  136. Jia, J.; Hu, J.; Huo, X.; Miao, R.; Zhang, Y.; Ma, F. Effects of Vitamin D Supplementation on Cognitive Function and Blood Aβ-Related Biomarkers in Older Adults with Alzheimer’s Disease: A Randomised, Double-Blind, Placebo-Controlled Trial. J. Neurol. Neurosurg. Psychiatry 2019, 90, 1347–1352. [Google Scholar] [CrossRef] [PubMed]
  137. Annweiler, C.; Rolland, Y.; Schott, A.M.; Blain, H.; Vellas, B.; Herrmann, F.R.; Beauchet, O. Higher Vitamin D Dietary Intake Is Associated With Lower Risk of Alzheimer’s Disease: A 7-Year Follow-Up. J. Gerontol. Ser. A 2012, 67, 1205–1211. [Google Scholar] [CrossRef] [Green Version]
  138. Yamini, P.; Ray, R.S.; Chopra, K. Vitamin D3 Attenuates Cognitive Deficits and Neuroinflammatory Responses in ICV-STZ Induced Sporadic Alzheimer’s Disease. Inflammopharmacology 2018, 26, 39–55. [Google Scholar] [CrossRef]
  139. Stein, M.S.; Scherer, S.C.; Ladd, K.S.; Harrison, L.C. A Randomized Controlled Trial of High-Dose Vitamin D2 Followed by Intranasal Insulin in Alzheimer’s Disease. J. Alzheimers Dis. 2011, 26, 477–484. [Google Scholar] [CrossRef]
  140. Presse, N.; Shatenstein, B.; Kergoat, M.-J.; Ferland, G. Low Vitamin K Intakes in Community-Dwelling Elders at an Early Stage of Alzheimer’s Disease. J. Am. Diet. Assoc. 2008, 108, 2095–2099. [Google Scholar] [CrossRef]
  141. Booth, S.L.; Shea, M.K.; Barger, K.; Leurgans, S.E.; James, B.D.; Holland, T.M.; Agarwal, P.; Fu, X.; Wang, J.; Matuszek, G.; et al. Association of Vitamin K with Cognitive Decline and Neuropathology in Community-Dwelling Older Persons. Alzheimers Dement. Transl. Res. Clin. Interv. 2022, 8, e12255. [Google Scholar] [CrossRef]
  142. Lin, X.; Wen, X.; Wei, Z.; Guo, K.; Shi, F.; Huang, T.; Wang, W.; Zheng, J. Vitamin K2 Protects against Aβ42-Induced Neurotoxicity by Activating Autophagy and Improving Mitochondrial Function in Drosophila. Neuroreport 2021, 32, 431. [Google Scholar] [CrossRef]
  143. Hadipour, E.; Tayarani-Najaran, Z.; Fereidoni, M. Vitamin K2 Protects PC12 Cells against Aβ(1-42) and H2O2-Induced Apoptosis via P38 MAP Kinase Pathway. Nutr. Neurosci. 2018, 23, 343–352. [Google Scholar] [CrossRef] [PubMed]
  144. Gaggelli, E.; Kozlowski, H.; Valensin, D.; Valensin, G. Copper Homeostasis and Neurodegenerative Disorders (Alzheimer’s, Prion, and Parkinson’s Diseases and Amyotrophic Lateral Sclerosis). Chem. Rev. 2006, 106, 1995–2044. [Google Scholar] [CrossRef]
  145. Kozlowski, H.; Janicka-Klos, A.; Brasun, J.; Gaggelli, E.; Valensin, D.; Valensin, G. Copper, Iron, and Zinc Ions Homeostasis and Their Role in Neurodegenerative Disorders (Metal Uptake, Transport, Distribution and Regulation). Coord. Chem. Rev. 2009, 253, 2665–2685. [Google Scholar] [CrossRef]
  146. Kozlowski, H.; Luczkowski, M.; Remelli, M.; Valensin, D. Copper, Zinc and Iron in Neurodegenerative Diseases (Alzheimer’s, Parkinson’s and Prion Diseases). Coord. Chem. Rev. 2012, 256, 2129–2141. [Google Scholar] [CrossRef]
  147. Liu, Y.; Nguyen, M.; Robert, A.; Meunier, B. Metal Ions in Alzheimer’s Disease: A Key Role or Not? Acc. Chem. Res. 2019, 52, 2026–2035. [Google Scholar] [CrossRef]
  148. Wang, L.; Yin, Y.-L.; Liu, X.-Z.; Shen, P.; Zheng, Y.-G.; Lan, X.-R.; Lu, C.-B.; Wang, J.-Z. Current Understanding of Metal Ions in the Pathogenesis of Alzheimer’s Disease. Transl. Neurodegener. 2020, 9, 10. [Google Scholar] [CrossRef] [Green Version]
  149. Foley, P.B.; Hare, D.J.; Double, K.L. A Brief History of Brain Iron Accumulation in Parkinson Disease and Related Disorders. J. Neural Transm. Vienna Austria 1996 2022, 129, 505–520. [Google Scholar] [CrossRef]
  150. Balachandran, R.C.; Mukhopadhyay, S.; McBride, D.; Veevers, J.; Harrison, F.E.; Aschner, M.; Haynes, E.N.; Bowman, A.B. Brain Manganese and the Balance between Essential Roles and Neurotoxicity. J. Biol. Chem. 2020, 295, 6312–6329. [Google Scholar] [CrossRef] [Green Version]
  151. Cheignon, C.; Tomas, M.; Bonnefont-Rousselot, D.; Faller, P.; Hureau, C.; Collin, F. Oxidative Stress and the Amyloid Beta Peptide in Alzheimer’s Disease. Redox Biol. 2018, 14, 450–464. [Google Scholar] [CrossRef]
  152. Meneghini, R. Iron Homeostasis, Oxidative Stress, and DNA Damage. Free Radic. Biol. Med. 1997, 23, 783–792. [Google Scholar] [CrossRef]
  153. Barnham, K.J.; Bush, A.I. Metals in Alzheimer’s and Parkinson’s Diseases. Curr. Opin. Chem. Biol. 2008, 12, 222–228. [Google Scholar] [CrossRef] [PubMed]
  154. Swartz, H.M.; Sarna, T.; Zecca, L. Modulation by Neuromelanin of the Availability and Reactivity of Metal Ions. Ann. Neurol. 1992, 32, S69–S75. [Google Scholar] [CrossRef] [PubMed]
  155. Liu, Y.; Hong, L.; Kempf, V.R.; Wakamatsu, K.; Ito, S.; Simon, J.D. Ion-Exchange and Adsorption of Fe(III) by Sepia Melanin. Pigment. Cell Res. 2004, 17, 262–269. [Google Scholar] [CrossRef] [PubMed]
  156. Zecca, L.; Pietra, R.; Goj, C.; Mecacci, C.; Radice, D.; Sabbioni, E. Iron and Other Metals in Neuromelanin, Substantia Nigra, and Putamen of Human Brain. J. Neurochem. 1994, 62, 1097–1101. [Google Scholar] [CrossRef] [PubMed]
  157. Horning, K.J.; Caito, S.W.; Tipps, K.G.; Bowman, A.B.; Aschner, M. Manganese Is Essential for Neuronal Health. Annu. Rev. Nutr. 2015, 35, 71–108. [Google Scholar] [CrossRef]
  158. Myhre, O.; Utkilen, H.; Duale, N.; Brunborg, G.; Hofer, T. Metal Dyshomeostasis and Inflammation in Alzheimer’s and Parkinson’s Diseases: Possible Impact of Environmental Exposures. Oxid. Med. Cell. Longev. 2013, 2013, e726954. [Google Scholar] [CrossRef] [Green Version]
  159. 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]
  160. Genchi, G.; Carocci, A.; Lauria, G.; Sinicropi, M.S.; Catalano, A. Nickel: Human Health and Environmental Toxicology. Int. J. Environ. Res. Public. Health 2020, 17, 679. [Google Scholar] [CrossRef] [Green Version]
  161. Babić Leko, M.; Langer Horvat, L.; Španić Popovački, E.; Zubčić, K.; Hof, P.R.; Šimić, G. Metals in Alzheimer’s Disease. Biomedicines 2023, 11, 1161. [Google Scholar] [CrossRef]
  162. Brewer, G.J.; Kanzer, S.H.; Zimmerman, E.A.; Molho, E.S.; Celmins, D.F.; Heckman, S.M.; Dick, R. Subclinical Zinc Deficiency in Alzheimer’s Disease and Parkinson’s Disease. Am. J. Alzheimers Dis. Dementiasr 2010, 25, 572–575. [Google Scholar] [CrossRef]
  163. Ahmed, S.S.S.J.; Santosh, W. Metallomic Profiling and Linkage Map Analysis of Early Parkinson’s Disease: A New Insight to Aluminum Marker for the Possible Diagnosis. PLoS ONE 2010, 5, e11252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Giacoppo, S.; Galuppo, M.; Calabrò, R.S.; D’Aleo, G.; Marra, A.; Sessa, E.; Bua, D.G.; Potortì, A.G.; Dugo, G.; Bramanti, P.; et al. Heavy Metals and Neurodegenerative Diseases: An Observational Study. Biol. Trace Elem. Res. 2014, 161, 151–160. [Google Scholar] [CrossRef] [PubMed]
  165. Zhao, H.-W.; Lin, J.; Wang, X.-B.; Cheng, X.; Wang, J.-Y.; Hu, B.-L.; Zhang, Y.; Zhang, X.; Zhu, J.-H. Assessing Plasma Levels of Selenium, Copper, Iron and Zinc in Patients of Parkinson’s Disease. PLoS ONE 2013, 8, e83060. [Google Scholar] [CrossRef] [PubMed]
  166. Yadav, J.; Verma, A.K.; Ahmad, M.K.; Garg, R.K.; Shiuli; Mahdi, A.A.; Srivastava, S. Metals Toxicity and Its Correlation with the Gene Expression in Alzheimer’s Disease. Mol. Biol. Rep. 2021, 48, 3245–3252. [Google Scholar] [CrossRef]
  167. Paglia, G.; Miedico, O.; Cristofano, A.; Vitale, M.; Angiolillo, A.; Chiaravalle, A.E.; Corso, G.; Di Costanzo, A. Distinctive Pattern of Serum Elements During the Progression of Alzheimer’s Disease. Sci. Rep. 2016, 6, 22769. [Google Scholar] [CrossRef] [Green Version]
  168. Wang, Z.-X.; Tan, L.; Wang, H.-F.; Ma, J.; Liu, J.; Tan, M.-S.; Sun, J.-H.; Zhu, X.-C.; Jiang, T.; Yu, J.-T. Serum Iron, Zinc, and Copper Levels in Patients with Alzheimer’s Disease: A Replication Study and Meta-Analyses. J. Alzheimers Dis. 2015, 47, 565–581. [Google Scholar] [CrossRef]
  169. Koç, E.R.; Ilhan, A.; Zübeyde Aytürk, A.; Acar, B.; Gürler, M.; Altuntaş, A.; Karapirli, M.; Bodur, A.S. A Comparison of Hair and Serum Trace Elements in Patients with Alzheimer Disease and Healthy Participants. Turk. J. Med. Sci. 2015, 45, 1034–1039. [Google Scholar] [CrossRef]
  170. Rembach, A.; Doecke, J.D.; Roberts, B.R.; Watt, A.D.; Faux, N.G.; Volitakis, I.; Pertile, K.K.; Rumble, R.L.; Trounson, B.O.; Fowler, C.J.; et al. Longitudinal Analysis of Serum Copper and Ceruloplasmin in Alzheimer’s Disease. J. Alzheimers Dis. 2013, 34, 171–182. [Google Scholar] [CrossRef]
  171. Alsadany, M.A.; Shehata, H.H.; Mohamad, M.I.; Mahfouz, R.G. Histone Deacetylases Enzyme, Copper, and IL-8 Levels in Patients With Alzheimer’s Disease. Am. J. Alzheimers Dis. Other Demen. 2013, 28, 54–61. [Google Scholar] [CrossRef]
  172. Mariani, S.; Ventriglia, M.; Simonelli, I.; Donno, S.; Bucossi, S.; Vernieri, F.; Melgari, J.-M.; Pasqualetti, P.; Rossini, P.M.; Squitti, R. Fe and Cu Do Not Differ in Parkinson’s Disease: A Replication Study plus Meta-Analysis. Neurobiol. Aging 2013, 34, 632–633. [Google Scholar] [CrossRef]
  173. Crespo, Â.C.; Silva, B.; Marques, L.; Marcelino, E.; Maruta, C.; Costa, S.; Timóteo, Â.; Vilares, A.; Couto, F.S.; Faustino, P.; et al. Genetic and Biochemical Markers in Patients with Alzheimer’s Disease Support a Concerted Systemic Iron Homeostasis Dysregulation. Neurobiol. Aging 2014, 35, 777–785. [Google Scholar] [CrossRef] [PubMed]
  174. Fukushima, T.; Tan, X.; Luo, Y.; Kanda, H. Relationship between Blood Levels of Heavy Metals and Parkinson’s Disease in China. Neuroepidemiology 2010, 34, 18–24. [Google Scholar] [CrossRef] [PubMed]
  175. Dexter, D.T.; Carayon, A.; Javoy-Agid, F.; Agid, Y.; Wells, F.R.; Daniel, S.E.; Lees, A.J.; Jenner, P.; Marsden, C.D. Alterations in the Levels of Iron, Ferritin and Other Trace Metals in Parkinson’s Disease and Other Neurodegenerative Diseases Affecting the Basal Ganglia. Brain 1991, 114, 1953–1975. [Google Scholar] [CrossRef] [PubMed]
  176. Babić Leko, M.; Jurasović, J.; Nikolac Perković, M.; Španić, E.; Sekovanić, A.; Orct, T.; Lukinović Škudar, V.; Bačić Baronica, K.; Kiđemet-Piskač, S.; Vogrinc, Ž.; et al. The Association of Essential Metals with APOE Genotype in Alzheimer’s Disease. J. Alzheimers Dis. 2021, 82, 661–672. [Google Scholar] [CrossRef]
  177. 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]
  178. Squitti, R.; Ventriglia, M.; Simonelli, I.; Bonvicini, C.; Costa, A.; Perini, G.; Binetti, G.; Benussi, L.; Ghidoni, R.; Koch, G.; et al. Copper Imbalance in Alzheimer’s Disease: Meta-Analysis of Serum, Plasma, and Brain Specimens, and Replication Study Evaluating ATP7B Gene Variants. Biomolecules 2021, 11, 960. [Google Scholar] [CrossRef]
  179. Miller, L.M.; Wang, Q.; Telivala, T.P.; Smith, R.J.; Lanzirotti, A.; Miklossy, J. Synchrotron-Based Infrared and X-Ray Imaging Shows Focalized Accumulation of Cu and Zn Co-Localized with β-Amyloid Deposits in Alzheimer’s Disease. J. Struct. Biol. 2006, 155, 30–37. [Google Scholar] [CrossRef]
  180. Dong, J.; Atwood, C.S.; Anderson, V.E.; Siedlak, S.L.; Smith, M.A.; Perry, G.; Carey, P.R. Metal Binding and Oxidation of Amyloid-Beta within Isolated Senile Plaque Cores: Raman Microscopic Evidence. Biochemistry 2003, 42, 2768–2773. [Google Scholar] [CrossRef]
  181. Lovell, M.A.; Robertson, J.D.; Teesdale, W.J.; Campbell, J.L.; Markesbery, W.R. Copper, Iron and Zinc in Alzheimer’s Disease Senile Plaques. J. Neurol. Sci. 1998, 158, 47–52. [Google Scholar] [CrossRef]
  182. Smith, M.A.; Harris, P.L.R.; Sayre, L.M.; Perry, G. Iron Accumulation in Alzheimer Disease Is a Source of Redox-Generated Free Radicals. Proc. Natl. Acad. Sci. USA 1997, 94, 9866–9868. [Google Scholar] [CrossRef]
  183. Liu, B.; Moloney, A.; Meehan, S.; Morris, K.; Thomas, S.E.; Serpell, L.C.; Hider, R.; Marciniak, S.J.; Lomas, D.A.; Crowther, D.C. Iron Promotes the Toxicity of Amyloid β Peptide by Impeding Its Ordered Aggregation. J. Biol. Chem. 2011, 286, 4248–4256. [Google Scholar] [CrossRef] [Green Version]
  184. Sóvágó, I.; Várnagy, K.; Kállay, C.; Grenács, Á. Interactions of Copper(II) and Zinc(II) Ions with the Peptide Fragments of Proteins Related to Neurodegenerative Disorders: Similarities and Differences. Curr. Med. Chem. 2023, 30, 4050–4071. [Google Scholar] [CrossRef] [PubMed]
  185. Nath, A.K.; Dey, S.G. Simultaneous Binding of Heme and Cu with Amyloid β Peptides: Active Site and Reactivities. Dalton Trans. 2022, 51, 4986–4999. [Google Scholar] [CrossRef]
  186. Stefaniak, E.; Bal, W. CuII Binding Properties of N-Truncated Aβ Peptides: In Search of Biological Function. Inorg. Chem. 2019, 58, 13561–13577. [Google Scholar] [CrossRef] [Green Version]
  187. Arena, G.; Rizzarelli, E. Zn2+ Interaction with Amyloid-Β: Affinity and Speciation. Mol. Basel Switz. 2019, 24, 2796. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Atrián-Blasco, E.; Conte-Daban, A.; Hureau, C. Mutual Interference of Cu and Zn Ions in Alzheimer’s Disease: Perspectives at the Molecular Level. Dalton Trans. 2017, 46, 12750–12759. [Google Scholar] [CrossRef] [Green Version]
  189. Wärmländer, S.K.T.S.; Österlund, N.; Wallin, C.; Wu, J.; Luo, J.; Tiiman, A.; Jarvet, J.; Gräslund, A. Metal Binding to the Amyloid-β Peptides in the Presence of Biomembranes: Potential Mechanisms of Cell Toxicity. J. Biol. Inorg. Chem. JBIC Publ. Soc. Biol. Inorg. Chem. 2019, 24, 1189–1196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  190. De Gregorio, G.; Biasotto, F.; Hecel, A.; Luczkowski, M.; Kozlowski, H.; Valensin, D. Structural Analysis of Copper(I) Interaction with Amyloid β Peptide. J. Inorg. Biochem. 2019, 195, 31–38. [Google Scholar] [CrossRef] [PubMed]
  191. Trapani, G.; Satriano, C.; La Mendola, D. Peptides and Their Metal Complexes in Neurodegenerative Diseases: From Structural Studies to Nanomedicine Prospects. Curr. Med. Chem. 2018, 25, 715–747. [Google Scholar] [CrossRef]
  192. Ahmadi, S.; Zhu, S.; Sharma, R.; Wilson, D.J.; Kraatz, H.-B. Interaction of Metal Ions with Tau Protein. The Case for a Metal-Mediated Tau Aggregation. J. Inorg. Biochem. 2019, 194, 44–51. [Google Scholar] [CrossRef]
  193. Binolfi, A.; Quintanar, L.; Bertoncini, C.W.; Griesinger, C.; Fernández, C.O. Bioinorganic Chemistry of Copper Coordination to Alpha-Synuclein: Relevance to Parkinson’s Disease. Coord. Chem. Rev. 2012, 256, 2188–2201. [Google Scholar] [CrossRef] [Green Version]
  194. Valensin, D.; Dell’Acqua, S.; Kozlowski, H.; Casella, L. Coordination and Redox Properties of Copper Interaction with α-Synuclein. J. Inorg. Biochem. 2016, 163, 292–300. [Google Scholar] [CrossRef] [PubMed]
  195. González, N.; Arcos-López, T.; König, A.; Quintanar, L.; Menacho Márquez, M.; Outeiro, T.F.; Fernández, C.O. Effects of Alpha-Synuclein Post-Translational Modifications on Metal Binding. J. Neurochem. 2019, 150, 507–521. [Google Scholar] [CrossRef] [Green Version]
  196. Atrián-Blasco, E.; Gonzalez, P.; Santoro, A.; Alies, B.; Faller, P.; Hureau, C. Cu and Zn Coordination to Amyloid Peptides: From Fascinating Chemistry to Debated Pathological Relevance. Coord. Chem. Rev. 2018, 375, 38–55. [Google Scholar] [CrossRef] [PubMed]
  197. Leal, S.S.; Botelho, H.M.; Gomes, C.M. Metal Ions as Modulators of Protein Conformation and Misfolding in Neurodegeneration. Coord. Chem. Rev. 2012, 256, 2253–2270. [Google Scholar] [CrossRef]
  198. Gamez, P.; Caballero, A.B. Copper in Alzheimer’s Disease: Implications in Amyloid Aggregation and Neurotoxicity. AIP Adv. 2015, 5, 092503. [Google Scholar] [CrossRef]
  199. Faller, P.; Hureau, C.; La Penna, G. Metal Ions and Intrinsically Disordered Proteins and Peptides: From Cu/Zn Amyloid-β to General Principles. Acc. Chem. Res. 2014, 47, 2252–2259. [Google Scholar] [CrossRef]
  200. DeToma, A.S.; Salamekh, S.; Ramamoorthy, A.; Lim, M.H. Misfolded Proteins in Alzheimer’s Disease and Type II Diabetes. Chem. Soc. Rev. 2012, 41, 608–621. [Google Scholar] [CrossRef] [Green Version]
  201. Ke, P.C.; Sani, M.-A.; Ding, F.; Kakinen, A.; Javed, I.; Separovic, F.; Davis, T.P.; Mezzenga, R. Implications of Peptide Assemblies in Amyloid Diseases. Chem. Soc. Rev. 2017, 46, 6492–6531. [Google Scholar] [CrossRef]
  202. Viles, J.H. Metal Ions and Amyloid Fiber Formation in Neurodegenerative Diseases. Copper, Zinc and Iron in Alzheimer’s, Parkinson’s and Prion Diseases. Coord. Chem. Rev. 2012, 256, 2271–2284. [Google Scholar] [CrossRef]
  203. Miller, Y.; Ma, B.; Nussinov, R. Zinc Ions Promote Alzheimer Abeta Aggregation via Population Shift of Polymorphic States. Proc. Natl. Acad. Sci. USA 2010, 107, 9490–9495. [Google Scholar] [CrossRef]
  204. Sharma, A.K.; Pavlova, S.T.; Kim, J.; Kim, J.; Mirica, L.M. The Effect of Cu2+ and Zn2+ on the Aβ42 Peptide Aggregation and Cellular Toxicity. Metallomics 2013, 5, 1529–1536. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Bush, A.I.; Pettingell, W.H.; Multhaup, G.; d Paradis, M.; Vonsattel, J.P.; Gusella, J.F.; Beyreuther, K.; Masters, C.L.; Tanzi, R.E. Rapid Induction of Alzheimer A Beta Amyloid Formation by Zinc. Science 1994, 265, 1464–1467. [Google Scholar] [CrossRef] [PubMed]
  206. Carboni, E.; Lingor, P. Insights on the Interaction of Alpha-Synuclein and Metals in the Pathophysiology of Parkinson’s Disease. Metallomics 2015, 7, 395–404. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  207. Drew, S.C. The N Terminus of α-Synuclein Forms CuII-Bridged Oligomers. Chem. Eur. J. 2015, 21, 7111–7118. [Google Scholar] [CrossRef] [PubMed]
  208. Li, W.-J.; Jiang, H.; Song, N.; Xie, J.-X. Dose- and Time-Dependent α-Synuclein Aggregation Induced by Ferric Iron in SK-N-SH Cells. Neurosci. Bull. 2010, 26, 205–210. [Google Scholar] [CrossRef] [Green Version]
  209. Rasia, R.M.; Bertoncini, C.W.; Marsh, D.; Hoyer, W.; Cherny, D.; Zweckstetter, M.; Griesinger, C.; Jovin, T.M.; Fernández, C.O. Structural Characterization of Copper(II) Binding to α-Synuclein: Insights into the Bioinorganic Chemistry of Parkinson’s Disease. Proc. Natl. Acad. Sci. USA 2005, 102, 4294–4299. [Google Scholar] [CrossRef]
  210. Li, X.; Du, X.; Ni, J. Zn2+ Aggravates Tau Aggregation and Neurotoxicity. Int. J. Mol. Sci. 2019, 20, 487. [Google Scholar] [CrossRef] [Green Version]
  211. Zubčić, K.; Hof, P.R.; Šimić, G.; Jazvinšćak Jembrek, M. The Role of Copper in Tau-Related Pathology in Alzheimer’s Disease. Front. Mol. Neurosci. 2020, 13, 572308. [Google Scholar] [CrossRef]
  212. Ahmadi, S.; Wu, B.; Song, R.; Zhu, S.; Simpson, A.; Wilson, D.J.; Kraatz, H.-B. Exploring the Interactions of Iron and Zinc with the Microtubule Binding Repeats R1 and R4. J. Inorg. Biochem. 2020, 205, 110987. [Google Scholar] [CrossRef]
  213. Soragni, A.; Zambelli, B.; Mukrasch, M.D.; Biernat, J.; Jeganathan, S.; Griesinger, C.; Ciurli, S.; Mandelkow, E.; Zweckstetter, M. Structural Characterization of Binding of Cu(II) to Tau Protein. Biochemistry 2008, 47, 10841–10851. [Google Scholar] [CrossRef]
  214. Balogh, B.D.; Szakács, B.; Di Natale, G.; Tabbì, G.; Pappalardo, G.; Sóvágó, I.; Várnagy, K. Copper (II) Binding Properties of an Octapeptide Fragment from the R3 Region of Tau Protein: A Combined Potentiometric, Spectroscopic and Mass Spectrometric Study. J. Inorg. Biochem. 2021, 217, 111358. [Google Scholar] [CrossRef]
  215. Bacchella, C.; Gentili, S.; Bellotti, D.; Quartieri, E.; Draghi, S.; Baratto, M.C.; Remelli, M.; Valensin, D.; Monzani, E.; Nicolis, S.; et al. Binding and Reactivity of Copper to R1 and R3 Fragments of Tau Protein. Inorg. Chem. 2020, 59, 274–286. [Google Scholar] [CrossRef]
  216. Jing, J.; Tu, G.; Yu, H.; Huang, R.; Ming, X.; Zhan, H.; Zhan, F.; Xue, W. Copper (Cu2+) Ion-Induced Misfolding of Tau Protein R3 Peptide Revealed by Enhanced Molecular Dynamics Simulation. Phys. Chem. Chem. Phys. 2021, 23, 11717–11726. [Google Scholar] [CrossRef]
  217. Rivers-Auty, J.; Tapia, V.S.; White, C.S.; Daniels, M.J.D.; Drinkall, S.; Kennedy, P.T.; Spence, H.G.; Yu, S.; Green, J.P.; Hoyle, C.; et al. Zinc Status Alters Alzheimer’s Disease Progression through NLRP3-Dependent Inflammation. J. Neurosci. 2021, 41, 3025–3038. [Google Scholar] [CrossRef] [PubMed]
  218. Singh, I.; Sagare, A.P.; Coma, M.; Perlmutter, D.; Gelein, R.; Bell, R.D.; Deane, R.J.; Zhong, E.; Parisi, M.; Ciszewski, J.; et al. Low Levels of Copper Disrupt Brain Amyloid-β Homeostasis by Altering Its Production and Clearance. Proc. Natl. Acad. Sci. USA 2013, 110, 14771–14776. [Google Scholar] [CrossRef] [PubMed]
  219. Sparks, D.L.; Schreurs, B.G. Trace Amounts of Copper in Water Induce β-Amyloid Plaques and Learning Deficits in a Rabbit Model of Alzheimer’s Disease. Proc. Natl. Acad. Sci. USA 2003, 100, 11065–11069. [Google Scholar] [CrossRef] [PubMed]
  220. Prasanthi, J.R.P.; Schrag, M.; Dasari, B.; Marwarha, G.; Dickson, A.; Kirsch, W.M.; Ghribi, O. Deferiprone Reduces Amyloid-β and Tau Phosphorylation Levels but Not Reactive Oxygen Species Generation in Hippocampus of Rabbits Fed a Cholesterol-Enriched Diet. J. Alzheimers Dis. 2012, 30, 167–182. [Google Scholar] [CrossRef] [Green Version]
  221. Chen, P.; Chakraborty, S.; Mukhopadhyay, S.; Lee, E.; Paoliello, M.M.B.; Bowman, A.B.; Aschner, M. Manganese Homeostasis in the Nervous System. J. Neurochem. 2015, 134, 601–610. [Google Scholar] [CrossRef] [Green Version]
  222. Shen, X.; Liu, J.; Fujita, Y.; Liu, S.; Maeda, T.; Kikuchi, K.; Obara, T.; Takebe, A.; Sayama, R.; Takahashi, T.; et al. Iron Treatment Inhibits Aβ42 Deposition in Vivo and Reduces Aβ42/Aβ40 Ratio. Biochem. Biophys. Res. Commun. 2019, 512, 653–658. [Google Scholar] [CrossRef]
  223. Alimonti, A.; Ristori, G.; Giubilei, F.; Stazi, M.A.; Pino, A.; Visconti, A.; Brescianini, S.; Monti, M.S.; Forte, G.; Stanzione, P.; et al. Serum Chemical Elements and Oxidative Status in Alzheimer’s Disease, Parkinson Disease and Multiple Sclerosis. NeuroToxicology 2007, 28, 450–456. [Google Scholar] [CrossRef] [PubMed]
  224. Guilarte, T.R. Manganese Neurotoxicity: New Perspectives from Behavioral, Neuroimaging, and Neuropathological Studies in Humans and Non-Human Primates. Front. Aging Neurosci. 2013, 5, 23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Hirsch, E.C.; Brandel, J.-P.; Galle, P.; Javoy-Agid, F.; Agid, Y. Iron and Aluminum Increase in the Substantia Nigra of Patients with Parkinson’s Disease: An X-Ray Microanalysis. J. Neurochem. 1991, 56, 446–451. [Google Scholar] [CrossRef] [PubMed]
  226. Altschuler, E. Aluminum-Containing Antacids as a Cause of Idiopathic Parkinson’s Disease. Med. Hypotheses 1999, 53, 22–23. [Google Scholar] [CrossRef]
  227. Zatta, P.; Zambenedetti, P.; Milanese, M. Activation of Monoamine Oxidase Type-B by Aluminum in Rat Brain Homogenate. NeuroReport 1999, 10, 3645–3648. [Google Scholar] [CrossRef]
  228. Baum, L.; Chan, I.H.S.; Cheung, S.K.-K.; Goggins, W.B.; Mok, V.; Lam, L.; Leung, V.; Hui, E.; Ng, C.; Woo, J.; et al. Serum Zinc Is Decreased in Alzheimer’s Disease and Serum Arsenic Correlates Positively with Cognitive Ability. BioMetals 2010, 23, 173–179. [Google Scholar] [CrossRef]
  229. Bhattacharjee, S.; Zhao, Y.; Hill, J.M.; Culicchia, F.; Kruck, T.P.A.; Percy, M.E.; Pogue, A.I.; Walton, J.R.; Lukiw, W.J. Selective Accumulation of Aluminum in Cerebral Arteries in Alzheimer’s Disease (AD). J. Inorg. Biochem. 2013, 126, 35–37. [Google Scholar] [CrossRef] [Green Version]
  230. González-Domínguez, R.; García-Barrera, T.; Gómez-Ariza, J.L. Characterization of Metal Profiles in Serum during the Progression of Alzheimer’s Disease. Metallomics 2014, 6, 292–300. [Google Scholar] [CrossRef]
  231. Smorgon, C.; Mari, E.; Atti, A.R.; Dalla Nora, E.; Zamboni, P.F.; Calzoni, F.; Passaro, A.; Fellin, R. Trace Elements and Cognitive Impairment: An Elderly Cohort Study. Arch. Gerontol. Geriatr. 2004, 38, 393–402. [Google Scholar] [CrossRef]
  232. Du, K.; Liu, M.; Pan, Y.; Zhong, X.; Wei, M. Association of Serum Manganese Levels with Alzheimer’s Disease and Mild Cognitive Impairment: A Systematic Review and Meta-Analysis. Nutrients 2017, 9, 231. [Google Scholar] [CrossRef] [Green Version]
  233. Koseoglu, E.; Koseoglu, R.; Kendirci, M.; Saraymen, R.; Saraymen, B. Trace Metal Concentrations in Hair and Nails from Alzheimer’s Disease Patients: Relations with Clinical Severity. J. Trace Elem. Med. Biol. 2017, 39, 124–128. [Google Scholar] [CrossRef] [PubMed]
  234. Szabo, S.T.; Harry, G.J.; Hayden, K.M.; Szabo, D.T.; Birnbaum, L. Comparison of Metal Levels between Postmortem Brain and Ventricular Fluid in Alzheimer’s Disease and Nondemented Elderly Controls. Toxicol. Sci. 2016, 150, 292–300. [Google Scholar] [CrossRef] [PubMed]
  235. Gorantla, N.V.; Das, R.; Balaraman, E.; Chinnathambi, S. Transition Metal Nickel Prevents Tau Aggregation in Alzheimer’s Disease. Int. J. Biol. Macromol. 2020, 156, 1359–1365. [Google Scholar] [CrossRef] [PubMed]
  236. Tong, Y.; Yang, H.; Tian, X.; Wang, H.; Zhou, T.; Zhang, S.; Yu, J.; Zhang, T.; Fan, D.; Guo, X.; et al. High Manganese, A Risk for Alzheimer’s Disease: High Manganese Induces Amyloid-β Related Cognitive Impairment. J. Alzheimers Dis. 2014, 42, 865–878. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  237. Walton, J.R.; Wang, M.-X. APP Expression, Distribution and Accumulation Are Altered by Aluminum in a Rodent Model for Alzheimer’s Disease. J. Inorg. Biochem. 2009, 103, 1548–1554. [Google Scholar] [CrossRef]
  238. Bouras, C.; Giannakopoulos, P.; Good, P.F.; Hsu, A.; Hof, P.R.; Perl, D.P. A Laser Microprobe Mass Analysis of Brain Aluminum and Iron in Dementia Pugilistica: Comparison with Alzheimer’s Disease. Eur. Neurol. 2007, 38, 53–58. [Google Scholar] [CrossRef]
  239. Rondeau, V.; Jacqmin-Gadda, H.; Commenges, D.; Helmer, C.; Dartigues, J.-F. Aluminum and Silica in Drinking Water and the Risk of Alzheimer’s Disease or Cognitive Decline: Findings from 15-Year Follow-up of the PAQUID Cohort. Am. J. Epidemiol. 2009, 169, 489–496. [Google Scholar] [CrossRef] [Green Version]
  240. Zhang, Q.L.; Jia, L.; Jiao, X.; Guo, W.L.; Ji, J.W.; Yang, H.L.; Niu, Q. APP/PS1 Transgenic Mice Treated with Aluminum: An Update of Alzheimer’s Disease Model. Int. J. Immunopathol. Pharmacol. 2012, 25, 49–58. [Google Scholar] [CrossRef] [Green Version]
  241. Kola, A.; Dudek, D.; Valensin, D. Metal Complexation Mechanisms of Polyphenols Associated to Alzheimer’s Disease. Curr. Med. Chem. 2021, 28, 7278–7294. [Google Scholar] [CrossRef]
  242. Gallo, A.A.; Sable, H.Z. Conformation of Complexes of Thiamin Pyrophosphate with Divalent Cations as Studied by Nuclear Magnetic Resonance Spectroscopy. J. Biol. Chem. 1975, 250, 4986–4991. [Google Scholar] [CrossRef]
  243. Gallo, A.A.; Hansen, I.L.; Sable, H.Z.; Swift, T.J. Coenzyme Interactions—Vii. Proton Magnetic Resonance Studies of Complexes of Thiamine Pyrophosphate with Divalent Cations. J. Biol. Chem. 1972, 247, 5913–5920. [Google Scholar] [CrossRef]
  244. Grande, H.J.; Veeger, C.; Houghton, R.L. A Nuclear-Magnetic-Resonance Study of the Manganese · Thiamine-Pyrophosphate Complex in Solution. Eur. J. Biochem. 1973, 37, 563–569. [Google Scholar] [CrossRef]
  245. Gary, J.; Adeyemo, A. Interaction of Vitamin B1 (Thiamine Hydrochloride) with Zn(II), Cd(II) and Hg(II) in Deuterated Dimethyl Sulfoxide. Inorganica Chim. Acta 1981, 55, 93–98. [Google Scholar] [CrossRef]
  246. Albert, A. Quantitative Studies of the Avidity of Naturally Occurring Substances for Trace Metals. 3. Pteridines, Riboflavin and Purines. Biochem. J. 1953, 54, 646–654. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  247. Aruna Kumari, S.; Kishore Babu, B.; Muralikrishna, N.; Mohana Rao, K.; Sinduri, V.L.; Praveen, K.; Padma Rao, C.V.; Swarna Latha, B.; Ramarao, K.; Veeraiah, V.; et al. Riboflavin Metal Complexes. Pharma Chem. 2015, 7, 307–315. [Google Scholar]
  248. Hernowo, E.; Angkawijaya, A.E.; Fazary, A.E.; Ismadji, S.; Ju, Y.-H. Complex Stability and Molecular Structure Studies of Divalent Metal Ion with l-Norleucine and Vitamin B3. J. Chem. Eng. Data 2011, 56, 4549–4555. [Google Scholar] [CrossRef]
  249. Irving, H.; Williams, R.J.P. The Stability of Transition-Metal Complexes. J. Chem. Soc. Resumed 1953, 3192–3210. [Google Scholar] [CrossRef]
  250. Hasan, M.M.; Hossain, M.E.; Halim, M.E.; Ehsan, M.Q. Preparation and Characterisation of Some Transition Metal Complexes of Niacinamide (Vitamin B3). Pak. J. Sci. Ind. Res. Ser. Phys. Sci. 2015, 58, 59–65. [Google Scholar] [CrossRef]
  251. Sismanoglu, T. Thermodynamics of Stability Constant of Binary Complex of Nicotinamide with Mn++. Chin. Chem. Lett. 2003, 14, 1207–1210. [Google Scholar]
  252. Tajmir-Riahi, H.A. Coordination Chemistry of Vitamin C. Part II. Interaction of L-Ascorbic Acid with Zn(II), Cd(II), Hg(II), and Mn(II) Ions in the Solid State and in Aqueous Solution. J. Inorg. Biochem. 1991, 42, 47–55. [Google Scholar] [CrossRef]
  253. Cesario, D.; Furia, E.; Mazzone, G.; Beneduci, A.; De Luca, G.; Sicilia, E. Complexation of Al3+ and Ni2+ by l-Ascorbic Acid: An Experimental and Theoretical Investigation. J. Phys. Chem. A 2017, 121, 9773–9781. [Google Scholar] [CrossRef] [PubMed]
  254. Tajmir-Riahi, H.A. Coordination Chemistry of Vitamin C. Part III. Interaction of L-Ascorbic Acid with Al(III), La(III), and Pb(II) Ions. Evidence for Metal Chelate Formation in the Solid and Aqueous Solution. J. Inorg. Biochem. 1991, 44, 39–45. [Google Scholar] [CrossRef]
  255. Obaleye, J.A.; Orjiekwe, C.L. Synthesis and Characterization of Some Metal Complexes of Vitamin C. Part 21 —Ascorbate Complexes of Mn(II), Fe(III) and Co(II). Synth. React. Inorg. Met.-Org. Chem. 1992, 22, 1015–1029. [Google Scholar] [CrossRef]
  256. Ünaleroğlu, C.; Mert, Y.; Zümreoğlu-Karan, B. Synthesis and Characterization of Copper Ascorbate. Synth. React. Inorg. Met.-Org. Chem. 2001, 31, 1531–1543. [Google Scholar] [CrossRef]
  257. Viswanathan, T.S.; Swift, T.J. A Nuclear Magnetic Resonance Study of Pyridoxal Phosphate—Metal Ion Interactions. II. Binding of Manganese(II). Can. J. Chem. 1979, 57, 1050–1055. [Google Scholar] [CrossRef]
  258. Jasim, M.A.; Al-Ani, H.N. Evaluating Spectra, Thermodynamic and Kinetic Parameters of the Complexation Reaction of Organic Compound or Chelation Therapy Drug with Some Heavy Metal Pollutants. Eurasian Chem. Commun. 2022, 4, 41–51. [Google Scholar] [CrossRef]
  259. Thompson, D.M.; Balenovich, W.; Hornich, L.H.M.; Richardson, M.F. Reactions of Metal Ions with Vitamins. IV. The Crystal Structure of a Zinc Complex of Pyridoxamine (Vitamin B6). Inorganica Chim. Acta 1980, 46, 199–203. [Google Scholar] [CrossRef]
  260. Furmanova, N.G.; Berdalieva, Z.I.; Chernaya, T.S.; Resnyanskiĭ, V.F.; Shiitieva, N.K.; Sulaĭmankulov, K.S. Synthesis and Crystal Structures of Coordination Compounds of Pyridoxine with Zinc and Cadmium Sulfates. Crystallogr. Rep. 2009, 54, 228–235. [Google Scholar] [CrossRef]
  261. Zhu, Y.C.; Zhang, M.Q.; Wu, J.G.; Deng, R.W. The Study of Equilibrium and Formation Constants of Some Transition Metal Complexes with Vitamin B6 in Solution by Potentiometry. Chem. Pap. 2001, 55, 229–232. [Google Scholar]
  262. Sigel, H.; McCormick, D.B.; Griesser, R.; Prijs, B.; Wright, L.D. Metal Ion Complexes with Biotin and Biotin Derivatives. Participation of Sulfur in the Orientation of Divalent Cations. Biochemistry 1969, 8, 2687–2695. [Google Scholar] [CrossRef]
  263. Sigel, H.; Scheller, K.H. Metal Ion Complexes of D-Biotin in Solution. Stability of the Stereoselective Thioether Coordination. J. Inorg. Biochem. 1982, 16, 297–310. [Google Scholar] [CrossRef]
  264. Yousef, W. Potentiometric and Conductometric Studies on Complexes of Folic Acid with Some Metal Ions. Int. J. Electrochem. Sci. 2017, 12, 1146–1156. [Google Scholar] [CrossRef] [PubMed]
  265. Hamed, E.; Attia, M.S.; Bassiouny, K. Synthesis, Spectroscopic and Thermal Characterization of Copper(II) and Iron(III) Complexes of Folic Acid and Their Absorption Efficiency in the Blood. Bioinorg. Chem. Appl. 2009, 2009, e979680. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  266. Mercê, A.L.R.; Szpoganicz, B.; Dutra, R.C.; Khan, M.A.; Thanh, X.D.; Bouet, G. Potentiometric Study of Vitamin D3 Complexes with Cobalt (II), Nickel (II) and Copper (II) in Water–Ethanol Medium. J. Inorg. Biochem. 1998, 71, 87–91. [Google Scholar] [CrossRef]
  267. Kieninger, C.; Baker, J.A.; Podewitz, M.; Wurst, K.; Jockusch, S.; Lawrence, A.D.; Deery, E.; Gruber, K.; Liedl, K.R.; Warren, M.J.; et al. Zinc Substitution of Cobalt in Vitamin B12: Zincobyric Acid and Zincobalamin as Luminescent Structural B12-Mimics. Angew. Chem. Int. Ed. 2019, 58, 14568–14572. [Google Scholar] [CrossRef] [Green Version]
  268. Kieninger, C.; Wurst, K.; Podewitz, M.; Stanley, M.; Deery, E.; Lawrence, A.D.; Liedl, K.R.; Warren, M.J.; Kräutler, B. Replacement of the Cobalt Center of Vitamin B12 by Nickel: Nibalamin and Nibyric Acid Prepared from Metal-Free B12 Ligands Hydrogenobalamin and Hydrogenobyric Acid. Angew. Chem. 2020, 132, 20304–20311. [Google Scholar] [CrossRef]
  269. Mercê, A.L.R.; Szpoganicz, B.; Khan, M.A.; Do Thanh, X.; Bouet, G. Potentiometric Study of Vitamin D3 Complexes with Manganese(II), Iron(II), Iron(III) and Zinc(II) in Water-Ethanol Medium. J. Inorg. Biochem. 1999, 73, 167–172. [Google Scholar] [CrossRef]
  270. Das, N.; Raymick, J.; Sarkar, S. Role of Metals in Alzheimer’s Disease. Metab. Brain Dis. 2021, 36, 1627–1639. [Google Scholar] [CrossRef]
  271. Kennedy, D. B Vitamins and the Brain: Mechanisms, Dose and Efficacy—A Review. Nutrients 2016, 8, 68. [Google Scholar] [CrossRef] [Green Version]
  272. Scheiber, I.F.; Mercer, J.F.B.; Dringen, R. Metabolism and Functions of Copper in Brain. Prog. Neurobiol. 2014, 116, 33–57. [Google Scholar] [CrossRef]
  273. Oteiza, P.I. Zinc and the Modulation of Redox Homeostasis. Free Radic. Biol. Med. 2012, 53, 1748–1759. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  274. Tassone, G.; Kola, A.; Valensin, D.; Pozzi, C. Dynamic Interplay between Copper Toxicity and Mitochondrial Dysfunction in Alzheimer’s Disease. Life 2021, 11, 386. [Google Scholar] [CrossRef] [PubMed]
  275. Spector, R. Vitamin Transport Diseases of Brain: Focus on Folates, Thiamine and Riboflavin. Brain Disord. Ther. 2014, 3, 2. [Google Scholar] [CrossRef] [Green Version]
  276. Spector, R.; Johanson, C.E. Vitamin Transport and Homeostasis in Mammalian Brain: Focus on Vitamins B and E. J. Neurochem. 2007, 103, 425–438. [Google Scholar] [CrossRef]
  277. Graham, S.F.; Nasaruddin, M.B.; Carey, M.; Holscher, C.; McGuinness, B.; Kehoe, P.G.; Love, S.; Passmore, P.; Elliott, C.T.; Meharg, A.A.; et al. Age-Associated Changes of Brain Copper, Iron, and Zinc in Alzheimer’s Disease and Dementia with Lewy Bodies. J. Alzheimers Dis. 2014, 42, 1407–1413. [Google Scholar] [CrossRef]
  278. Uchida, Y.; Ito, K.; Ohtsuki, S.; Kubo, Y.; Suzuki, T.; Terasaki, T. Major Involvement of Na+-Dependent Multivitamin Transporter (SLC5A6/SMVT) in Uptake of Biotin and Pantothenic Acid by Human Brain Capillary Endothelial Cells. J. Neurochem. 2015, 134, 97–112. [Google Scholar] [CrossRef]
  279. Ghasemzadeh, S.; Riazi, G.H. Inhibition of Tau Amyloid Fibril Formation by Folic Acid: In-Vitro and Theoretical Studies. Int. J. Biol. Macromol. 2020, 154, 1505–1516. [Google Scholar] [CrossRef]
  280. Espín, S.; Sánchez-Virosta, P. A Review of Metal-Induced Effects on Vitamins A, E and D3 in Birds. Ecotoxicology 2021, 30, 1–16. [Google Scholar] [CrossRef]
  281. Sampaio, I.; Quatroni, F.D.; Pincela Lins, P.M.; Nascimento, A.S.; Zucolotto, V. Modulation of Beta-Amyloid Aggregation Using Ascorbic Acid. Biochimie 2022, 200, 36–43. [Google Scholar] [CrossRef]
  282. Cannell, J.J.; Grant, W.B. What Is the Role of Vitamin D in Autism? Dermatoendocrinol. 2013, 5, 199–204. [Google Scholar] [CrossRef] [Green Version]
  283. Máčová, L.; Bičíková, M.; Ostatníková, D.; Hill, M.; Stárka, L. Vitamin D, Neurosteroids and Autism. Physiol. Res. 2017, 66, S333–S340. [Google Scholar] [CrossRef]
  284. Temova Rakuša, Ž.; Pišlar, M.; Kristl, A.; Roškar, R. Comprehensive Stability Study of Vitamin D3 in Aqueous Solutions and Liquid Commercial Products. Pharmaceutics 2021, 13, 617. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of vitamin impairment in PD and AD. Water- and fat-soluble vitamins are highlighted in white and yellow, respectively. “=” indicates no significant difference between vitamin levels of sick subjects and healthy controls, while “−” refers to lower vitamin levels in patients with PD or AD.
Figure 1. Schematic representation of vitamin impairment in PD and AD. Water- and fat-soluble vitamins are highlighted in white and yellow, respectively. “=” indicates no significant difference between vitamin levels of sick subjects and healthy controls, while “−” refers to lower vitamin levels in patients with PD or AD.
Molecules 28 05467 g001
Figure 2. Schematic representation of the amyloidogenic proteins and their metal ion interactions in AD and PD.
Figure 2. Schematic representation of the amyloidogenic proteins and their metal ion interactions in AD and PD.
Molecules 28 05467 g002
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

Kola, A.; Nencioni, F.; Valensin, D. Bioinorganic Chemistry of Micronutrients Related to Alzheimer’s and Parkinson’s Diseases. Molecules 2023, 28, 5467. https://doi.org/10.3390/molecules28145467

AMA Style

Kola A, Nencioni F, Valensin D. Bioinorganic Chemistry of Micronutrients Related to Alzheimer’s and Parkinson’s Diseases. Molecules. 2023; 28(14):5467. https://doi.org/10.3390/molecules28145467

Chicago/Turabian Style

Kola, Arian, Federico Nencioni, and Daniela Valensin. 2023. "Bioinorganic Chemistry of Micronutrients Related to Alzheimer’s and Parkinson’s Diseases" Molecules 28, no. 14: 5467. https://doi.org/10.3390/molecules28145467

Article Metrics

Back to TopTop