Next Article in Journal
Novel Fluoridoaluminates from Ammonothermal Synthesis: Two Modifications of K2AlF5 and the Elpasolite Rb2KAlF6
Previous Article in Journal
Remedies to Avoid Failure Mechanisms of Lithium-Metal Anode in Li-Ion Batteries
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Probable Reasons for Neuron Copper Deficiency in the Brain of Patients with Alzheimer’s Disease: The Complex Role of Amyloid

1
Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6714415185, Iran
2
Institute of Biochemistry and Biophysics, University of Tehran, Tehran 1417614335, Iran
3
UR 1268 Biopolymeres Interactions Assemblages, Institute National de la Recherche Agronomique et Environementale, 44316 Nantes, France
4
Department of Laboratory Medicine, Research and Development Division, San Giovanni Calibita Fatebenefratelli Hospital, Isola Tiberina, 00186 Rome, Italy
5
Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University, 00185 Rome, Italy
*
Author to whom correspondence should be addressed.
Submission received: 9 November 2021 / Revised: 26 December 2021 / Accepted: 4 January 2022 / Published: 7 January 2022

Abstract

:
Alzheimer’s disease is a progressive neurodegenerative disorder that eventually leads the affected patients to die. The appearance of senile plaques in the brains of Alzheimer’s patients is known as a main symptom of this disease. The plaques consist of different components, and according to numerous reports, their main components include beta-amyloid peptide and transition metals such as copper. In this disease, metal dyshomeostasis leads the number of copper ions to simultaneously increase in the plaques and decrease in neurons. Copper ions are essential for proper brain functioning, and one of the possible mechanisms of neuronal death in Alzheimer’s disease is the copper depletion of neurons. However, the reason for the copper depletion is as yet unknown. Based on the available evidence, we suggest two possible reasons: the first is copper released from neurons (along with beta-amyloid peptides), which is deposited outside the neurons, and the second is the uptake of copper ions by activated microglia.

1. Introduction

Alzheimer’s disease (AD) is the most prevalent neurodegenerative disorder related to aging [1]. The number of Alzheimer’s patients in 2019 was estimated to be 50 million, and that figure is expected to exceed 152 million by 2050. It was approximated that about US$1 trillion was spent on AD in 2019 [2]. Despite the deadly nature of this disease and its high prevalence as well as the consequent economic burden, after more than a hundred years have passed since its discovery, no effective treatment has been found up to now. The complexity of the disease means its recognition and treatment development have been similar to the story of the “elephant in the dark”, with various researchers attributing various causes to the disease as well as studying wide-ranging and overlapping options for treatment of different aspects of the condition.
There are several hypotheses concerning the cause of AD based on the available experimental data [3]. Yet, the main accredited hypothesis of the ‘amyloid cascade’ has been challenged in the last decade [4]. Metal dyshomeostasis is among the relatively new proposed mechanisms for AD onset [5,6,7]. Considering that Aβ is produced in the brain under physiological conditions (as a soluble component), the “metal hypothesis” has also been put forward, in which Aβ binding to metal ions, especially copper and zinc ions, causes pathogenic Aβ deposits to form that eventually lead to AD [8].
In the past 10 years, a body of evidence has been collated suggesting that copper imbalance specifically affects a certain percentage of AD patients [9,10,11]. Copper imbalance appears to be typified by an increased level of copper in the general circulation, which can be attributed to expansion of the non-ceruloplasmin-bound (non-cp) copper fraction in serum as well as a decreased level of copper in the brain [12,13,14,15,16,17,18,19], though the proportion of labile copper (i.e., exchangeable copper relative to the copper content of the tissue) increases in the brain [20].
Though copper is highly concentrated in amyloid plaques, its concentration reduces in neurons [12,14,15,16,17,18,19,21]. A body of evidence suggests that intracellular copper deficiency in neurons could represent a possible cause of neuronal death in AD (reviewed in [22]). Beyond describing the processes of copper content dysregulation in the bloodstream (recently reviewed in [5]), it remains unclear why the concentration and distribution of copper in the brains of AD patients change, though a number of attempts have been made to interpret this complex phenomenon (reviewed in [9]). Determining how this happens may significantly contribute toward finding appropriate methods for intervening in the progression of the disease and identifying its starting point [23,24]. In this review, we compile and discuss the latest results available on one aspect of the complex puzzle of copper imbalance in AD—the brain copper deficiency.

2. Copper Physiology Focusing on the Brain

Copper in the gut is mainly absorbed by copper transporter 1 (CTR1) [25], and also by divalent metal transporter 1 (DMT1) to a lesser extent [26,27]. In the blood, approximately 75–95% of the absorbed copper ions bind to ceruloplasmin. Inside the cells, these are transported to different destinations using different chaperones [28]. It was found that essential metals for the body, such as copper and zinc, play major structural and catalytic roles in metalloenzymes; however, their high levels are fatal to cells [29]. Eukaryotes can activate certain mechanisms to store these metals inside intracellular organelles, and use them when needed. For instance, macrophages use Zn2+ and Cu+1 to attack bacteria, which is referred to as the brass dagger [30,31,32]. In fact, there is less than one free copper ion per cell under physiological conditions [33].
Copper, as a cofactor or structural component of several important enzymes, is known as an essential metal in brain functioning that plays key roles in various pathways, including neurotransmitter and neuropeptide synthesis, energy metabolism, antioxidative defense, and iron metabolism [34]. The brain barrier system (the blood-brain barrier (BBB) and the blood-cerebrospinal fluid barrier) plays an essential role in modulating copper homeostasis in the brain. The related evidence suggests that copper is principally transported to the brain through the BBB using both CTR1 and DMT1. Correspondingly, excess copper is transported into the CSF in the ventricles of the brain by the choroidal epithelial microvilli [35,36]. Some major changes to the cerebral vasculature, including increased permeability and weakened detoxification and repair functions, occur in people following aging. Subsequently, these changes can significantly affect the function of the brain barrier system [37]. Moreover, some studies have previously shown that exposure to other metals such as lead [38] or manganese [36,39], by altering the activity of copper transporters in the brain barrier system, can also alter the copper homeostasis in the brain.
The imported copper into the brain cells is likely to be detached by glutathione (GSH), stored as an metallothionein-copper complex, or transported by copper chaperones to some specific target cell locations [34]. Of note, the intracellular copper concentration in the brain is estimated to be 100–1000 times more than that of extracellular copper [40]. The approximate amount of copper in the human brain varies from 3.1 to 5.1 µg/g wet weight. In this regard, numerous studies have shown that copper is distributed unequally in the brain [41,42]. Overall, the copper concentration is higher in grey matter compared to white matter [43]. Notably, two-dimensional copper maps of human brain slices have shown that those areas heavily enriched in copper are found in the hippocampus [41]. Both the amount and distribution of copper in the brain also change with aging and in those affected by neurodegenerative diseases [44,45,46]. Brain copper levels naturally decrease with aging [47,48,49]. In addition, brain copper decreases in those with the neurological diseases, including early-onset familial AD [50], Menkes’ disease [51], Parkinson’s disease [52,53,54], transmissible spongiform encephalopathies, Lewy body dementia, Creutzfeldt–Jakob disease [53], and Huntington’s disease [54].

3. Connections between Copper and Aβ

Copper is an essential trace element in the nervous system’s development, and its dyshomeostasis causes neurodegenerative phenotypes of both Menkes’ and Wilson’s diseases [55]. As multivalent cations are highly concentrated in senile plaques in the brains of patients with AD, many studies have previously reported dyshomeostasis of copper and of zinc as the main causes of AD pathogenesis [22]. Both these metals were observed to affect the oxidative status of the brain. As well as this, the Cu(II)/Cu(I) cycle could primarily generate free radicals in Fenton-like reactions (reviewed in [9]) outside the cells, in the form of labile copper [20]. Furthermore, combined evidence suggested that a reduced copper content in the brain cells results in greater production of Aβ peptide, copper migration to lipid rafts, and copper-amyloid complex formation in lipid rafts [22]. It was demonstrated that binding to copper [56] and proximity to the cell membranes [57] change the folding of Aβ peptides, creating channels in the membrane [58]. It has been postulated that the created channels could lead to apoptosis [59,60]; however, the causative factor behind the copper deficiency of neurons remains unknown.
APP belongs to the mammalian APP gene family. APP family members have various biological functions, including nervous system development, synaptogenesis, axonal growth and guidance, and synaptic functions [61]. APP is a transmembrane protein strongly expressed in the brain and very rapidly metabolized by a series of proteases [62,63]. Several paths have been found for APP proteolysis, some of which end in Aβ production while others do not [62]. APP can be cleaved by α-secretase and then γ-secretase without leading to Aβ production; however, Aβ is produced if APP is cleaved by β-secretase (instead of α-secretase). Aβ is released into the extracellular space or degraded in lysosomes [62]. It has a number of significant functions in both synaptic physiology and regulation. In addition, Aβ accumulates in the brains of patients with AD for an unknown reason [64,65].
Copper transportation to the brain is complex and involves many factors, including APP, Aβ peptide, and cholesterol [66]. APP has two copper binding sites, one of which is included in the Aβ peptide sequence [67,68]. Though APP has no equivalent in yeast, overexpression of APP in yeast consequently causes copper efflux [69]. In transgenic mice, on the other hand, overexpression of APP induces reduced copper levels in the murine brain [70]. Therefore, based on this evidence, it can be proposed that APP may be a specific copper transporter in mammals [66] as it displays a high affinity for copper [71,72,73]. In addition, a recent study has indicated that Aβ scavenges copper from the synaptic space [74].

4. Copper’s Importance in the Hippocampus

Glutamate is the main excitatory neurotransmitter in the brain, acting on ionotropic and metabotropic receptors. The ionotropic glutamate receptors (iGluRs) are known to be responsible for rapid neural communication in excitatory synapses, which include the following three subfamilies: kainate receptors, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors, and N-methyl-d-aspartate (NMDA) receptors [75]. Nevertheless, extreme stimulation of glutamatergic signaling during excitotoxicity, causes neuronal cell death [76,77,78]. Excitotoxicity through NMDA receptors is primarily mediated by excessive entry of Ca2+ [79,80] since NMDA receptors are highly permeable to calcium ions compared to the other iGluRs [81].
The NMDA receptors play important roles in synaptic transmission and plasticity, which are known to form the basis for learning and memory processes [82]. A previous study conducted on older healthy adults showed that higher hippocampal activity is associated with increased Aβ deposition as well as decreased memory function [83]. Another study indicated that patients with mild cognitive impairment (MCI) and Aβ deposits had smaller hippocampal volume, higher hippocampal activity, and lower Mini-Mental State Examination (MMSE) scores compared to Aβ-negative patients. In addition, they showed higher rates of hippocampal atrophy and disease progression as well as a greater decrease in MMSE score over time, despite high levels of hippocampal activity [84]. Recently, a study showed that abnormal NMDA receptor activation is associated with synaptic dysfunction in AD [85]. Indeed, disruption of Ca2+ signaling results in a gradual loss of synaptic function, which eventually leads to neuronal cell death; this is clinically associated with a gradual decrease in cognition or memory [86].
The hippocampus, which is the brain part associated with neurogenesis and long-term memory, is one of the first affected brain areas in AD, and suffers from the most damage [87,88,89]. It was shown that about 15 μM of copper is released from post-synaptic vesicles into the glutamatergic synaptic cleft following neuronal depolarization through excitation of NMDA receptors in the hippocampal area [90,91]. This copper concentration (15 μM) has been obtained from two previously-performed independent studies, one that measured the emission of fluorescent dye (sensitive to copper binding) from synaptosomes derived from bovine chromaffin cells [91], and another that measured the 67Cu radioactivity of cells taken from a rat’s hypothalamus [90]. However, a study conducted on the synaptosomes of a rat’s cerebral cortex that used the atomic absorption technique calculated the amount of released copper to be at least 100 μM [92]. Thus, this process appears to be a protective mechanism for neurons against NMDA excitotoxicity.
High ATP7A expression can be observed in hippocampal neurons. Some studies on primary cultures of murine hippocampal neurons showed that after activation of the NMDA receptor, but not AMPA or kainate-type glutamate receptors, ATP7A trafficking occurs in the hippocampal neurons along with the rapid release of copper [93]. Copper chelation exacerbates cell death due to overactivation of the NMDA receptor, while the addition of copper protects the cell and then leads to a significant reduction in cytoplasmic Ca2+ levels after the NMDA receptor’s activation [94]. In this regard, further evidence showed that PrP interacts with the NMDA receptor, which then decreases its interaction with glycine (as a receptor activator), leading to reduced receptor activation [95]. PrP is involved in S-nitrosylation of the NMDA receptor, and to perform this activity, it is dependent on copper [96,97]. S-nitrosylation is a chemical post-translational modification process through which a nitric oxide group is added to the protein cysteine, playing a role in inhibiting the NMDA receptor [96]. The NMDA receptor is a highly calcium-permeable glutamate receptor and its hyperactivity leads to cell death [95]. It was shown that copper decreases the cytoplasmic Ca2+ concentration after the NMDA receptor’s activation and consequently attenuates NMDA-induced excitotoxicity [93,95,96,98,99].

5. Roles of Microglia in Both Healthy and AD Brains

Microglia are brain-specific macrophages known as the first line of defense in the central nervous system [100]. Increased microglia activation has been reported in dementia. Related data indicate that this is associated with white matter maintenance and preserved cognition in dementia with Lewy bodies; however, it is not associated with reduced grey matter volume [101]. While a study indicated that microglial activation is negatively correlated with hippocampal volume in patients with Parkinson’s disease dementia [102], another study reported that microglial activation is associated with white matter maintenance as well as preserved cognition in Parkinson’s disease dementia [103]. In a case with AD, microglia markers were repeatedly reported as being overexpressed in the brains of patients. Among the different regions of the brain, the white matter and cerebellum appear to be more resistant to this increase [104]. New findings suggest that microglial activation in MCI patients is correlated with greater grey matter and hippocampal volumes [105], whereas previously reported data indicated that microglia activation is inversely associated with the hippocampal volume in patients with AD [102]. In fact, microglia play a dual role in AD, either inducing nerve survival (by clearing Aβ deposits) or disturbing the neuronal function and then leading to cell death (by releasing cytotoxic mediators). The magnitude and context of microglial activation lead different molecules to be secreted with different effects [94,106,107] (Figure 1).

6. Interplay of Copper, Aβ, and Microglia Activation in Mediating NMDA Receptor-Induced Excitotoxicity

The ATP7A protein is a copper pump located in the Golgi network, which can transport cytoplasmic copper into secretory compartments to make it available for copper-dependent enzymes. ATP7A also exports copper from the cytoplasm to the post-Golgi vesicles or the plasma membrane via trafficking [108,109]. Overexpression of ATP7A has been reported in activated microglial cells observed around amyloid plaques in a TgCRND8 mouse model. In addition, after interferon gamma (IFN-γ) stimulation (but not tumor necrosis factor-α (TNF-α) or interleukin 1 beta), the cultured microglial cells showed a significant increase in both ATP7A and CTR1 expression [110]. It was shown that in IFN-γ-stimulated microglial cells, despite ATP7A trafficking into cytoplasmic vesicles, copper export did not happen. Copper accumulation was instead observed as a result of increased copper uptake due to the expression of copper importer CTR1 [110]. Murine macrophages accumulate copper in intracellular vesicles in response to infection with S. Typhimurium or treatment with lipopolysaccharides (LPS) [111]. Previously performed studies showed that both IFN-γ and TNF-α cause copper accumulation in phagosomes of infected macrophages [112]. The innate immune response uses copper to attack infectious microorganisms [113]. In the presence of copper, IFN-γ–activated macrophages have a higher bactericidal activity [114]. Moreover, it has been demonstrated that copper ions in culture medium induce the transformation of the macrophage phenotype to the proinflammatory M1 state by activating copper transport signaling [115]. Furthermore, a recent report shows that copper changes the microglia phenotype to a degenerative phenotype [116].
Findings on the NLRP3 inflammasome (NACHT, LRR, and PYD domains-containing protein 3) can be considered as additional evidence demonstrating the involvement of copper in the activity of microglia. Inflammasomes are multiprotein complexes mediating the proteolytic activation of caspase-1 and cytokines via a NOD-like receptor [117]. It was also indicated that the NLRP3 inflammasome is involved in various human disorders, including metabolic diseases, chronic inflammation, autoinflammatory syndromes, and infections [118]. Additionally, inflammasome activation is important in neuroinflammation induced by microglia [119], and it has been indicated that Aβ oligomers can activate NLRP3 inflammasomes in microglia [120,121]. The activation of NLRP3 inflammasome requires intracellular copper, and removing copper from superoxide dismutase-1 suspends the activity of inflammasomes. Notably, copper regulation of the NLRP3 inflammasome is only specific to macrophages [122]. Given that microglia are a type of macrophage, this mechanism may also be applied to them, as studies have previously confirmed [123].
High densities of the activated microglia are located around the Aβ plaques in the hippocampi of patients with AD [105,124], which contain Aβ deposits [125]. Aβ deposits can activate microglial cells (Figure 1a), which is regarded as an important pathological event in the brains of patients with AD [126,127,128], via various innate immune receptors expressed on their membranes [106]. Furthermore, it has been reported that Aβ deposits cause rapid ATP release as well as overexpression of the P2Y2 receptor gene, which consequently causes microglial activation to increase Aβ clearance [129]. It has been shown that ATP effectively induces pinocytosis in microglia with the involvement of P2Y4 receptors, which results in Aβ clearance by microglia [130]. Both phagosomes and pinosomes are membrane vesicles formed when phagocytes swallow extracellular material; phagosomes are formed when the solid material swallowing process occurs and pinosomes are formed when liquid is ingested [131]. Small soluble Aβ deposits are taken up nonspecifically by pinocytosis, and directed to late endosomes for degradation [132], while Aβ fibrils’ uptake is mediated by scavenger receptors, which results in localization to distinct subcellular compartments called phagosomes [133]. Aβ fibrils can increase the phagocytic capacity of microglia. Aβ oligomers cannot only reduce the phagocytosis of fluorescent microspheres by microglia, but can also significantly reduce Aβ fibril phagocytosis. The treatment of microglia with Aβ oligomers causes a rapid and temporary increase in interleukin-1β as well as production of more inflammatory mediators—including TNF-α, nitric oxide, prostaglandin E2 (PGE2), and intracellular superoxide anion—compared to Aβ fibrils. More data on this subject showed that inflammatory markers could reduce fibril-induced phagocytic potency. Altogether, these results indicated that oligomers disrupt microglia phagocytosis and fibril clearing by inducing a strong inflammatory response [134] (Figure 1b). High copper concentrations can also be found in amyloid plaques in the hippocampus [135]. Of note, copper ions play important roles in Aβ aggregation and neurotoxicity [136,137,138]. Aβ has been shown to be less toxic to neurons in the absence of copper ions, but these levels of copper ions (in control studies) in the absence of Aβ have not been found to be cytotoxic. In other words, copper ions can significantly increase Aβ’s toxicity to neurons [137]. Moreover, copper ions have been shown to induce toxic non-amyloidogenic aggregates of amyloid peptides [139]. In addition, the toxicity of Aβ is attributed to the production of reactive oxygen species, among which copper is known as one of the metals involved in this process [140]. Copper ions have also been shown to have the ability to increase the effect of Aβ on microglial activation as well as the subsequent neurotoxicity (Figure 2). Indeed, a copper-Aβ complex (but not copper or Aβ alone) at subneurotoxic concentrations can activate microglial cells and then induce both TNF-α and NO production in microglia that ultimately result in neuronal death [141]. It was demonstrated that copper could significantly decrease the phagocytic property of microglial cells activated by Aβ fibrils or LPS as well as reduce the intracellular degradation of Aβ, whereas it increases the secretion of proinflammatory cytokines [142,143].
Different concentrations of copper have the ability to increase the formation of fibrillar amyloids [137]. At substoichiometric levels of copper, fibril formation increases [137], and in contrast, when the ratio of copper to Aβ increases, fibril formation decreases [144]. Moreover, the addition of metal chelators to amorphous Aβ deposits converted them into fibrillar forms [140]. Furthermore, some previously-performed structural studies showed that at the ratio of 1:1 copper and Aβ, the β-sheet structure reaches a maximum and then decreases with an increased amount of copper [145], which is in line with other studies stating that copper at high concentrations reduces fibril formation [139]. In general, the role of copper in mediating Aβ between oligomeric and fibril forms mostly depends on the ratio of copper to Aβ. In this way, fibril formation up to a ratio of 1:1 of copper/Aβ can be facilitated; with an increasing copper/Aβ ratio, fibril formation decreases, while vice versa, the situation favors the generation of non-fibrillar forms.

7. Insights and Concluding Remarks

The grey matter and certain areas of the brain like the hippocampus that are the most damaged in AD have been reported to contain the highest levels of copper in a healthy brain [41,43]. The brains of patients with AD are copper deficient [12,14,15,16,17,18,19,146], and a dataset led to the hypothesis that this deficiency can consequently lead to channel creation in the neuron membrane, which results in apoptosis [22,146]; however, the cause of this deficiency is not yet known.
Recently, a critical, location-dependent copper dissociation constant (Kdc) was proposed as a new mechanism, featuring a shift from physiological bound metal ion pools to loose toxic pools in copper imbalance (reviewed in [9]). This hypothetical mechanism provided some clues on the key decreased copper enzymes and transporters in the AD brain that can majorly affect copper buffering and functioning in synapses during the glutamatergic transmission process. The concept proposed is applicable to Aβ and APP as well as other copper proteins relevant to the AD cascade, including the prion protein and α-synuclein [9].
Another putative mechanism of copper deficiency in neuronal cells lies in Aβ sorting and segregation within lipid rafts. Of note, Aβ is produced in lipid rafts [147,148] before entering the synaptic space or being digested inside the neuron [62]. In AD, it seems that Aβ production increases or its clearance decreases, and binding of copper ions to this peptide (with a high affinity for copper ions) or deposition outside the cell can lead to copper deficiency in neurons. The findings indicate that the Aβ is greatly deposited in areas of the brain with the most damage. As well as this, greater deposition may cause more intense apoptosis due to greater copper deficiency.
Certain evidence suggested that microglia activation can be considered as another possible cause of copper deficiency in neurons. Copper is unevenly distributed in the brain, and some brain areas contain greater amounts of copper. One such area is the hippocampus, which is related to memory and becomes severely damaged in AD [41,42]. Neurons of this area routinely use copper to prevent excitotoxicity by NMDA receptors, and also release copper at the micromolar level after each depolarization to synapses [90,91]. Aβ deposits have also been shown to activate microglia, and the activated microglia then clear amyloid from the environment [129]. Meanwhile, the activated microglia increase the expression of copper-related proteins, which consequently causes copper uptake into the microglia [110]. Also, a new study confirmed that microglia increase their intracellular copper in response to the inflammatory stimuli [149]. Therefore, it seems that if microglia are activated in the areas using copper to prevent excitotoxicity by NMDA receptors, by microglia absorbing copper from the synaptic space, copper re-uptake by neurons can be disrupted, which consequently causes a serious copper deficiency in neurons. Correspondingly, this phenomenon can cause overactivation of NMDA receptors and subsequently lead to neurodegeneration. Moreover, it was found that copper imbalance in the heart is dangerous. Loss of copper from the heart occurs in myocardial ischemia [150,151]. Recently, in an animal study, it was shown that upregulation of an intracellular copper exporter, such as copper metabolism MURR domain 1 (COMMD1), in the heart is key to exporting copper from the heart to the blood on ischemic insult [152]. Accordingly, this mechanism can also take place in the brain in AD, especially in the hippocampus. In addition, the upregulation of CTR1 in microglia can function to absorb the copper released from neurons.
NMDA receptors seem to regulate different processes in various brain regions [153,154]. Accordingly, they have different distributions in the central nervous system in terms of their type of subunit [155]. For instance, NR2A and NR2B are overexpressed in the cortex and hippocampal areas, respectively, while high NR2C expression is specific to the cerebellum area [155,156]. On the one hand, the earliest instance and highest level of damage were found to be related to the cortex and hippocampal areas, respectively, while the lowest was related to the cerebellum area [157,158]. On the other hand, the cortex and hippocampus have the highest levels of copper in the brain. Taken together, this evidence suggests that activation of microglia in the presence of copper-regulated NMDA receptors may be a significant factor in copper deficiency in neuronal cells.
In addition, the activation of microglia in areas using copper to prevent excitotoxicity by NMDA receptors can lead to copper deficiency through another mechanism. Copper reduces the phagocytic properties of microglia, which can consequently result in greater Aβ deposition [142,143]. Logically, a greater increase of Aβ deposition in this area would lead greater amounts of copper to be deposited out of reach of neurons. Otherwise, evidence has shown that copper intensifies Aβ-mediated microglia activation, and subsequently, highly activated microglia do not play a protective role, which leads to neuronal death [141] (Figure 2b). However, previous studies have shown that microglia form a barrier around small amyloid plaques, and slowing of the dystrophic neural process can be detected in areas with microglial coverage, suggesting peptide clearance by microglia protects neurons against Aβ toxicity [159].
In general, copper imbalance in AD, similar to the disease itself, is a complex phenomenon. In this review article, we attempted to specifically address the possible causes of copper depletion in neurons. We hypothesized that there may be two possible causes of copper depletion in neurons: first, the release of amyloid (as a copper transfer protein with a high affinity for copper) from neurons and its deposition outside neurons can trap copper outside neurons, which in turn, causes copper deficiency in neurons. Second, the uptake of copper by the activated microglia makes copper inaccessible to neurons.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

The authors would like to thank the Vice Chancellor for Research and Technology of the Kermanshah University of Medical Sciences for approving this research. The authors received no financial support for this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Zhang, Y.; Thompson, R.; Zhang, H.; Xu, H. APP processing in Alzheimer’s disease. Mol. Brain 2011, 4, 3. [Google Scholar] [CrossRef] [Green Version]
  2. Australia, D.; Baker, S.; Banerjee, S. Alzheimer’s Disease International World Alzheimer Report 2019: Attitudes to Dementia. Available online: https://www.alzint.org/resource/world-alzheimer-report-2019/ (accessed on 20 September 2019).
  3. Liu, P.P.; Xie, Y.; Meng, X.-Y.; Kang, J.-S. History and progress of hypotheses and clinical trials for Alzheimer’s disease. Signal Transduct. Target. Ther. 2019, 4, 29. [Google Scholar] [CrossRef] [PubMed]
  4. Kepp, K.P. Ten Challenges of the Amyloid Hypothesis of Alzheimer’s Disease. J. Alzheimer’s Dis. 2017, 55, 447–457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Sensi, S.L.; Granzotto, A.; Siotto, M.; Squitti, R. Copper and Zinc Dysregulation in Alzheimer’s Disease. Trends Pharmacol. Sci. 2018, 39, 1049–1063. [Google Scholar] [CrossRef]
  6. De Benedictis, C.A.; Vilella, A.; Grabrucker, A.M. The Role of Trace Metals in Alzheimer’s Disease. In Alzheimer’s Disease; Codon Publications: Brisbane, Australia, 2019; pp. 85–106. ISBN 9780646809687. [Google Scholar]
  7. Bagheri, S.; Saboury, A.A. What role do metals play in Alzheimer’s disease? J. Iran. Chem. Soc. 2021, 18, 2199–2213. [Google Scholar] [CrossRef]
  8. Bush, A.I.; Tanzi, R.E. Therapeutics for Alzheimer’s disease based on the metal hypothesis. Neurotherapeutics 2008, 5, 421–432. [Google Scholar] [CrossRef] [Green Version]
  9. Kepp, K.P.; Squitti, R. Copper imbalance in Alzheimer’s disease: Convergence of the chemistry and the clinic. Coord. Chem. Rev. 2019, 397, 168–187. [Google Scholar] [CrossRef]
  10. Squitti, R.; Simonelli, I.; Cassetta, E.; Ventriglia, M.; Lupoi, D.; Rongioletti, M.; Siotto, M. Patients with increased non-ceruloplasmin copper appear a distinct sub-group of alzheimer’s disease: A neuroimaging study. Curr. Alzheimer Res. 2017, 14, 1318–1326. [Google Scholar] [CrossRef] [PubMed]
  11. Squitti, R.; Ventriglia, M.; Gennarelli, M.; Colabufo, N.A.; El Idrissi, I.G.; Bucossi, S.; Mariani, S.; Rongioletti, M.; Zanetti, O.; Congiu, C.; et al. Non-Ceruloplasmin Copper Distincts Subtypes in Alzheimer’s Disease: A Genetic Study of ATP7B Frequency. Mol. Neurobiol. 2017, 54, 671–681. [Google Scholar] [CrossRef] [PubMed]
  12. Schrag, M.; Mueller, C.; Oyoyo, U.; Smith, M.A.; Kirsch, W.M. Iron, zinc and copper in the Alzheimer’s disease brain: A quantitative meta-analysis. Some insight on the influence of citation bias on scientific opinion. Prog. Neurobiol. 2011, 94, 296–306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Squitti, R.; Ghidoni, R.; Simonelli, I.; Ivanova, I.D.; Colabufo, N.A.; Zuin, M.; Benussi, L.; Binetti, G.; Cassetta, E.; Rongioletti, M.; et al. Copper dyshomeostasis in Wilson disease and Alzheimer’s disease as shown by serum and urine copper indicators. J. Trace Elem. Med. Biol. 2018, 45, 181–188. [Google Scholar] [CrossRef]
  14. Rembach, A.; Hare, D.J.; Lind, M.; Fowler, C.J.; Cherny, R.A.; Mclean, C.A.; Bush, A.I.; Masters, C.L.; Roberts, B.R. Decreased copper in Alzheimer’s disease brain is predominantly in the soluble extractable fraction. Int. J. Alzheimer’s Dis. 2013, 2013, 623241. [Google Scholar] [CrossRef]
  15. Akatsu, H.; Hori, A.; Yamamoto, T.; Yoshida, M.; Mimuro, M.; Hashizume, Y.; Tooyama, I.; Yezdimer, E.M. Transition metal abnormalities in progressive dementias. Biometals 2012, 25, 337–350. [Google Scholar] [CrossRef]
  16. Magaki, S.; Raghavan, R.; Mueller, C.; Oberg, K.C.; Vinters, H.V.; Kirsch, W.M. Iron, copper, and iron regulatory protein 2 in Alzheimer’s disease and related dementias. Neurosci. Lett. 2007, 418, 72–76. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Xu, J.; Begley, P.; Church, S.J.; Patassini, S.; McHarg, S.; Kureishy, N.; Hollywood, K.A.; Waldvogel, H.J.; Liu, H.; Zhang, S.; et al. Elevation of brain glucose and polyol-pathway intermediates with accompanying brain-copper deficiency in patients with Alzheimer’s disease: Metabolic basis for dementia. Sci. Rep. 2016, 6, 27524. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Deibel, M.A.; Ehmann, W.D.; Markesbery, W.R. Copper, iron, and zinc imbalances in severely degenerated brain regions in Alzheimer’s disease: Possible relation to oxidative stress. J. Neurol. Sci. 1996, 143, 137–142. [Google Scholar] [CrossRef]
  19. Scholefield, M.; Church, S.J.; Xu, J.; Patassini, S.; Roncaroli, F.; Hooper, N.M.; Unwin, R.D.; Cooper, G.J.S. Widespread Decreases in Cerebral Copper Are Common to Parkinson’s Disease Dementia and Alzheimer’s Disease Dementia. Front. Aging Neurosci. 2021, 13, 81. [Google Scholar] [CrossRef]
  20. James, S.A.; Volitakis, I.; Adlard, P.A.; Duce, J.A.; Masters, C.L.; Cherny, R.A.; Bush, A.I. Elevated labile Cu is associated with oxidative pathology in Alzheimer disease. Free Radic. Biol. Med. 2012, 52, 298–302. [Google Scholar] [CrossRef]
  21. 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]
  22. Bagheri, S.; Squitti, R.; Haertlé, T.; Siotto, M.; Saboury, A.A. Role of copper in the onset of Alzheimer’s disease compared to other metals. Front. Aging Neurosci. 2018, 9, 446. [Google Scholar] [CrossRef]
  23. 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]
  24. Malosio, M.L.; Tecchio, F.; Squitti, R. Molecular mechanisms underlying copper function and toxicity in neurons and their possible therapeutic exploitation for Alzheimer’s disease. Aging Clin. Exp. Res. 2021, 33, 2027–2030. [Google Scholar] [CrossRef]
  25. Nose, Y.; Kim, B.E.; Thiele, D.J. Ctr1 drives intestinal copper absorption and is essential for growth, iron metabolism, and neonatal cardiac function. Cell Metab. 2006, 4, 235–244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Kaler, S.G. ATP7A-related copper transport diseasesg-emerging concepts and future trends. Nat. Rev. Neurol. 2011, 7, 15–29. [Google Scholar] [CrossRef] [Green Version]
  27. Lutsenko, S. Human copper homeostasis: A network of interconnected pathways. Curr. Opin. Chem. Biol. 2010, 14, 211–217. [Google Scholar] [CrossRef]
  28. Hellman, N.E.; Gitlin, J.D. Ceruloplasmin metabolism and function. Annu. Rev. Nutr. 2002, 22, 439–458. [Google Scholar] [CrossRef] [PubMed]
  29. Lavado, L.K.; Zhang, M.; Patel, K.; Khan, S.; Patel, U.K. Biometals as Potential Predictors of the Neurodegenerative Decline in Alzheimer’s Disease. Cureus 2019, 11, e5573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. German, N.; Doyscher, D.; Rensing, C. Bacterial killing in macrophages and amoeba: Do they all use a brass dagger? Future Microbiol. 2013, 8, 1257–1264. [Google Scholar] [CrossRef] [PubMed]
  31. Hao, X.; Lüthje, F.L.; Qin, Y.; McDevitt, S.F.; Lutay, N.; Hobman, J.L.; Asiani, K.; Soncini, F.C.; German, N.; Zhang, S.; et al. Survival in amoeba—a major selection pressure on the presence of bacterial copper and zinc resistance determinants? Identification of a “copper pathogenicity island”. Appl. Microbiol. Biotechnol. 2015, 99, 5817–5824. [Google Scholar] [CrossRef] [PubMed]
  32. Hao, X.; Lüthje, F.; Rønn, R.; German, N.A.; Li, X.; Huang, F.; Kisaka, J.; Huffman, D.; Alwathnani, H.A.; Zhu, Y.G.; et al. A role for copper in protozoan grazing–two billion years selecting for bacterial copper resistance. Mol. Microbiol. 2016, 102, 628–641. [Google Scholar] [CrossRef]
  33. Rae, T.D.; Schmidt, P.J.; Pufahl, R.A.; Culotta, V.C.; O’Halloran, T.V. Undetectable intracellular free copper: The requirement of a copper chaperone for superoxide dismutase. Science 1999, 284, 805–808. [Google Scholar] [CrossRef] [Green Version]
  34. 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] [PubMed]
  35. Choi, B.S.; Zheng, W. Copper transport to the brain by the blood-brain barrier and blood-CSF barrier. Brain Res. 2009, 1248, 14–21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Zheng, G.; Chen, J.; Zheng, W. Relative contribution of CTR1 and DMT1 in copper transport by the blood-CSF barrier: Implication in manganese-induced neurotoxicity. Toxicol. Appl. Pharmacol. 2012, 260, 285–293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Zheng, W. Neurotoxicology of the brain barrier system: New implications. J. Toxicol.-Clin. Toxicol. 2001, 39, 711–719. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Zheng, G.; Zhang, J.; Xu, Y.; Shen, X.; Song, H.; Jing, J.; Luo, W.; Zheng, W.; Chen, J. Involvement of CTR1 and ATP7A in lead (Pb)-induced copper (Cu) accumulation in choroidal epithelial cells. Toxicol. Lett. 2014, 225, 110–118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Fu, X.; Zhang, Y.; Jiang, W.; Monnot, A.D.; Bates, C.A.; Zheng, W. Regulation of copper transport crossing brain barrier systems by CU-ATPases: Effect of manganese exposure. Toxicol. Sci. 2014, 139, 432–451. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Gaier, E.D.; Eipper, B.A.; Mains, R.E. Copper signaling in the mammalian nervous system: Synaptic effects. J. Neurosci. Res. 2013, 91, 2–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Dobrowolska, J.; Dehnhardt, M.; Matusch, A.; Zoriy, M.; Palomero-Gallagher, N.; Koscielniak, P.; Zilles, K.; Becker, J.S. Quantitative imaging of zinc, copper and lead in three distinct regions of the human brain by laser ablation inductively coupled plasma mass spectrometry. Talanta 2008, 74, 717–723. [Google Scholar] [CrossRef]
  42. Davies, K.M.; Hare, D.J.; Cottam, V.; Chen, N.; Hilgers, L.; Halliday, G.; Mercer, J.F.B.; Double, K.L. Localization of copper and copper transporters in the human brain. Metallomics 2013, 5, 43–51. [Google Scholar] [CrossRef] [Green Version]
  43. Bonilla, E.; Salazar, E.; Villasmil, J.J.; Villalobos, R.; Gonzalez, M.; Davila, J.O. Copper distribution in the normal human brain. Neurochem. Res. 1984, 9, 1543–1548. [Google Scholar] [CrossRef]
  44. Wang, L.M.; Becker, J.S.; Wu, Q.; Oliveira, M.F.; Bozza, F.A.; Schwager, A.L.; Hoffman, J.M.; Morton, K.A. Bioimaging of copper alterations in the aging mouse brain by autoradiography, laser ablation inductively coupled plasma mass spectrometry and immunohistochemistry. Metallomics 2010, 2, 348–353. [Google Scholar] [CrossRef]
  45. Serpa, R.F.B.; De Jesus, E.F.O.; Anjos, M.J.; De Oliveira, L.F.; Marins, L.A.; Do Carmo, M.G.T.; Corrêa, J.D.; Rocha, M.S.; Lopes, R.T.; Martinez, A.M.B. Topographic trace-elemental analysis in the brain of wistar rats by X-ray microfluorescence with synchrotron radiation. Anal. Sci. 2008, 24, 839–842. [Google Scholar] [CrossRef] [Green Version]
  46. Tarohda, T.; Yamamoto, M.; Amamo, R. Regional distribution of manganese, iron, copper, and zinc in the rat brain during development. Anal. Bioanal. Chem. 2004, 380, 240–246. [Google Scholar] [CrossRef] [PubMed]
  47. Ramos, P.; Santos, A.; Pinto, N.R.; Mendes, R.; Magalhães, T.; Almeida, A. Anatomical Region Differences and Age-Related Changes in Copper, Zinc, and Manganese Levels in the Human Brain. Biol. Trace Elem. Res. 2014, 161, 190–201. [Google Scholar] [CrossRef] [PubMed]
  48. 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. Alzheimer’s Dis. 2014, 42, 1407–1413. [Google Scholar] [CrossRef] [PubMed]
  49. Zecca, L.; Stroppolo, A.; Gatti, A.; Tampellini, D.; Toscani, M.; Gallorini, M.; Giaveri, G.; Arosio, P.; Santambrogio, P.; Fariello, R.G.; et al. The role of iron and copper molecules in the neuronal vulnerability of locus coeruleus and substantia nigra during aging. Proc. Natl. Acad. Sci. USA 2004, 101, 9843–9848. [Google Scholar] [CrossRef] [Green Version]
  50. Southon, A.; Greenough, M.A.; Ganio, G.; Bush, A.I.; Burke, R.; Camakaris, J. Presenilin Promotes Dietary Copper Uptake. PLoS ONE 2013, 8, e62811. [Google Scholar] [CrossRef] [Green Version]
  51. Tümer, Z.; Møller, L.B. Menkes disease. Eur. J. Hum. Genet. 2010, 18, 511–518. [Google Scholar] [CrossRef] [Green Version]
  52. Montes, S.; Rivera-Mancia, S.; Diaz-Ruiz, A.; Tristan-Lopez, L.; Rios, C. Copper and Copper Proteins in Parkinson’s Disease. Oxid. Med. Cell. Longev. 2014, 2014, 147251. [Google Scholar] [CrossRef] [Green Version]
  53. Deloncle, R.; Guillard, O. Is brain copper deficiency in Alzheimer’s, lewy body, and Creutzfeldt Jakob diseases the common key for a free radical mechanism and oxidative stress-induced damage? J. Alzheimer’s Dis. 2014, 43, 1149–1156. [Google Scholar] [CrossRef]
  54. Scholefield, M.; Unwin, R.D.; Cooper, G.J.S. Shared perturbations in the metallome and metabolome of Alzheimer’s, Parkinson’s, Huntington’s, and dementia with Lewy bodies: A systematic review. Ageing Res. Rev. 2020, 63, 101152. [Google Scholar] [CrossRef] [PubMed]
  55. Waggoner, D.J.; Bartnikas, T.B.; Gitlin, J.D. The role of copper in neurodegenerative disease. Neurobiol. Dis. 1999, 6, 221–230. [Google Scholar] [CrossRef] [Green Version]
  56. Curtain, C.C.; Ali, F.; Volitakis, I.; Cherny, R.A.; Norton, R.S.; Beyreuther, K.; Barrow, C.J.; Masters, C.L.; Bush, A.I.; Barnham, K.J. Alzheimer’s Disease Amyloid-β Binds Copper and Zinc to Generate an Allosterically Ordered Membrane-penetrating Structure Containing Superoxide Dismutase-like Subunits. J. Biol. Chem. 2001, 276, 20466–20473. [Google Scholar] [CrossRef] [Green Version]
  57. Ciccotosto, G.D.; Tew, D.J.; Drew, S.C.; Smith, D.G.; Johanssen, T.; Lal, V.; Lau, T.L.; Perez, K.; Curtain, C.C.; Wade, J.D.; et al. Stereospecific interactions are necessary for Alzheimer disease amyloid-β toxicity. Neurobiol. Aging 2011, 32, 235–248. [Google Scholar] [CrossRef]
  58. Di Scala, C.; Yahi, N.; Boutemeur, S.; Flores, A.; Rodriguez, L.; Chahinian, H.; Fantini, J. Common molecular mechanism of amyloid pore formation by Alzheimer’s β-amyloid peptide and α-synuclein. Sci. Rep. 2016, 6, 28781. [Google Scholar] [CrossRef] [Green Version]
  59. Jang, H.; Arce, F.T.; Ramachandran, S.; Capone, R.; Azimova, R.; Kagan, B.L.; Nussinov, R.; Lal, R. Truncated β-amyloid peptide channels provide an alternative mechanism for Alzheimer’s Disease and Down syndrome. Proc. Natl. Acad. Sci. USA 2010, 107, 6538–6543. [Google Scholar] [CrossRef] [Green Version]
  60. Arispe, N.; Rojas, E.; Pollard, H.B. Alzheimer disease amyloid β protein forms calcium channels in bilayer membranes: Blockade by tromethamine and aluminum. Proc. Natl. Acad. Sci. USA 1993, 90, 567–571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Müller, U.C.; Deller, T.; Korte, M. Not just amyloid: Physiological functions of the amyloid precursor protein family. Nat. Rev. Neurosci. 2017, 18, 281–298. [Google Scholar] [CrossRef]
  62. O’Brien, R.J.; Wong, P.C. Amyloid Precursor Protein Processing and Alzheimer’s Disease. Annu. Rev. Neurosci. 2011, 34, 185–204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Lee, J.; Retamal, C.; Cuitiño, L.; Caruano-Yzermans, A.; Shin, J.E.; Van Kerkhof, P.; Marzolo, M.P.; Bu, G. Adaptor protein sorting nexin 17 regulates amyloid precursor protein trafficking and processing in the early endosomes. J. Biol. Chem. 2008, 283, 11501–11508. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Abramov, E.; Dolev, I.; Fogel, H.; Ciccotosto, G.D.; Ruff, E.; Slutsky, I. Amyloid-Β as a positive endogenous regulator of release probability at hippocampal synapses. Nat. Neurosci. 2009, 12, 1567–1576. [Google Scholar] [CrossRef] [PubMed]
  65. Kamenetz, F.; Tomita, T.; Hsieh, H.; Seabrook, G.; Borchelt, D.; Iwatsubo, T.; Sisodia, S.; Malinow, R. APP Processing and Synaptic Function. Neuron 2003, 37, 925–937. [Google Scholar] [CrossRef] [Green Version]
  66. Macreadie, I. Copper transport and Alzheimer’s disease. Eur. Biophys. J. 2008, 37, 295–300. [Google Scholar] [CrossRef] [PubMed]
  67. Atwood, C.S.; Moir, R.D.; Huang, X.; Scarpa, R.C.; Bacarra, N.M.E.; Romano, D.M.; Hartshorn, M.A.; Tanzi, R.E.; Bush, A.I. Dramatic aggregation of alzheimer by Cu(II) is induced by conditions representing physiological acidosis. J. Biol. Chem. 1998, 273, 12817–12826. [Google Scholar] [CrossRef] [Green Version]
  68. Hesse, L.; Beher, D.; Masters, C.L.; Multhaup, G. The βA4 amyloid precursor protein binding to copper. FEBS Lett. 1994, 349, 109–116. [Google Scholar] [CrossRef] [Green Version]
  69. Treiber, C.; Simons, A.; Strauss, M.; Hafner, M.; Cappai, R.; Bayer, T.A.; Multhaup, G. Clioquinol mediates copper uptake and counteracts copper efflux activities of the amyloid precursor protein of Alzheimer’s disease. J. Biol. Chem. 2004, 279, 51958–51964. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Maynard, C.J.; Cappai, R.; Volitakis, I.; Cherny, R.A.; White, A.R.; Beyreuther, K.; Masters, C.L.; Bush, A.I.; Li, Q.X. Overexpression of Alzheimer’s disease amyloid-β opposes the age-dependent elevations of brain copper and iron. J. Biol. Chem. 2002, 277, 44670–44676. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Sarell, C.J.; Syme, C.D.; Rigby, S.E.J.; Viles, J.H. Copper(II) binding to amyloid-β fibrils of Alzheimer’s disease reveals a picomolar affinity: Stoichiometry and coordination geometry are independent of Aβ oligomeric form. Biochemistry 2009, 48, 4388–4402. [Google Scholar] [CrossRef]
  72. Barritt, J.D.; Viles, J.H. Truncated amyloid-β(11-40/42) from Alzheimer disease binds Cu2+ with a femtomolar affinity and influences fiber assembly. J. Biol. Chem. 2015, 290, 27791–27802. [Google Scholar] [CrossRef] [Green Version]
  73. Atwood, C.S.; Scarpa, R.C.; Huang, X.; Moir, R.D.; Jones, W.D.; Fairlie, D.P.; Tanzi, R.E.; Bush, A.I. Characterization of Copper Interactions with Alzheimer Amyloid β Peptides. J. Neurochem. 2008, 75, 1219–1233. [Google Scholar] [CrossRef]
  74. Wezynfeld, N.E.; Stefaniak, E.; Stachucy, K.; Drozd, A.; Płonka, D.; Drew, S.C.; Krężel, A.; Bal, W. Resistance of Cu(Aβ4-16) to Copper Capture by Metallothionein-3 Supports a Function for the Aβ4-42 Peptide as a Synaptic Cu II Scavenger. Angew. Chemie Int. Ed. 2016, 55, 8235–8238. [Google Scholar] [CrossRef]
  75. Traynelis, S.F.; Wollmuth, L.P.; McBain, C.J.; Menniti, F.S.; Vance, K.M.; Ogden, K.K.; Hansen, K.B.; Yuan, H.; Myers, S.J.; Dingledine, R. Glutamate receptor ion channels: Structure, regulation, and function. Pharmacol. Rev. 2010, 62, 405–496. [Google Scholar] [CrossRef] [Green Version]
  76. Rothman, S.M.; Olney, J.W. Glutamate and the pathophysiology of hypoxic–ischemic brain damage. Ann. Neurol. 1986, 19, 105–111. [Google Scholar] [CrossRef] [PubMed]
  77. Choi, D.W. Glutamate neurotoxicity and diseases of the nervous system. Neuron 1988, 1, 623–634. [Google Scholar] [CrossRef]
  78. Lipton, S.A.; Rosenberg, P.A. Excitatory amino acids as a final common pathway for neurologic disorders. N. Engl. J. Med. 1994, 330, 613–622. [Google Scholar]
  79. Tymianski, M.; Charlton, M.P.; Carlen, P.L.; Tator, C.H. Source specificity of early calcium neurotoxicity in cultured embryonic spinal neurons. J. Neurosci. 1993, 13, 2085–2104. [Google Scholar] [CrossRef]
  80. Choi, D.W. Ionic dependence of glutamate neurotoxicity. J. Neurosci. 1987, 7, 369–379. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Choi, D.W. Excitotoxic cell death. J. Neurobiol. 1992, 23, 1261–1276. [Google Scholar] [CrossRef] [PubMed]
  82. Paoletti, P.; Bellone, C.; Zhou, Q. NMDA receptor subunit diversity: Impact on receptor properties, synaptic plasticity and disease. Nat. Rev. Neurosci. 2013, 14, 383–400. [Google Scholar] [CrossRef] [PubMed]
  83. Leal, S.L.; Landau, S.M.; Bell, R.K.; Jagust, W.J. Hippocampal activation is associated with longitudinal amyloid accumulation and cognitive decline. Elife 2017, 6, e22978. [Google Scholar] [CrossRef] [PubMed]
  84. Huijbers, W.; Mormino, E.C.; Schultz, A.P.; Wigman, S.; Ward, A.M.; Larvie, M.; Amariglio, R.E.; Marshall, G.A.; Rentz, D.M.; Johnson, K.A.; et al. Amyloid-β deposition in mild cognitive impairment is associated with increased hippocampal activity, atrophy and clinical progression. Brain 2015, 138, 1023–1035. [Google Scholar] [CrossRef] [Green Version]
  85. Kodis, E.J.; Choi, S.; Swanson, E.; Ferreira, G.; Bloom, G.S. N-methyl-d-aspartate receptor–mediated calcium influx connects amyloid-β oligomers to ectopic neuronal cell cycle reentry in Alzheimer’s disease. Alzheimer’s Dement. 2018, 14, 1302–1312. [Google Scholar] [CrossRef]
  86. Berridge, M.J. Neuronal calcium signaling. Neuron 1998, 21, 13–26. [Google Scholar] [CrossRef] [Green Version]
  87. Dhikav, V.; Anand, K. Potential predictors of hippocampal atrophy in alzheimers disease. Drugs Aging 2011, 28, 1–11. [Google Scholar] [CrossRef]
  88. Gosche, K.M.; Mortimer, J.A.; Smith, C.D.; Markesbery, W.R.; Snowdon, D.A. Hippocampal volume as an index of Alzheimer neuropathology: Findings from the Nun study. Neurology 2002, 58, 1476–1482. [Google Scholar] [CrossRef] [PubMed]
  89. Scahill, R.I.; Schott, J.M.; Stevens, J.M.; Rossor, M.N.; Fox, N.C. Mapping the evolution of regional atrophy in Alzheimer’s disease: Unbiased analysis of fluid-registered serial MRI. Proc. Natl. Acad. Sci. USA 2002, 99, 4703–4707. [Google Scholar] [CrossRef] [Green Version]
  90. Hartter, D.E.; Barnea, A. Evidence for release of copper in the brain: Depolarization-induced release of newly taken-up 67copper. Synapse 1988, 2, 412–415. [Google Scholar] [CrossRef] [PubMed]
  91. Hopt, A.; Korte, S.; Fink, H.; Panne, U.; Niessner, R.; Jahn, R.; Kretzschmar, H.; Herms, J. Methods for studying synaptosomal copper release. J. Neurosci. Methods 2003, 128, 159–172. [Google Scholar] [CrossRef] [Green Version]
  92. Kardos, J.; Kovács, I.; Hajós, F.; Kálmán, M.; Simonyi, M. Nerve endings from rat brain tissue release copper upon depolarization. A possible role in regulating neuronal excitability. Neurosci. Lett. 1989, 103, 139–144. [Google Scholar] [CrossRef]
  93. Schlief, M.L.; Craig, A.M.; Gitlin, J.D. NMDA receptor activation mediates copper homeostasis in hippocampal neurons. J. Neurosci. 2005, 25, 239–246. [Google Scholar] [CrossRef] [Green Version]
  94. Gray, S.C.; Kinghorn, K.J.; Woodling, N.S. Shifting equilibriums in Alzheimer’s disease: The complex roles of microglia in neuroinflammation, neuronal survival and neurogenesis. Neural Regen. Res. 2020, 15, 1208–1219. [Google Scholar]
  95. You, H.; Tsutsui, S.; Hameed, S.; Kannanayakal, T.J.; Chen, L.; Xia, P.; Engbers, J.D.T.; Lipton, S.A.; Stys, P.K.; Zamponi, G.W. Aβ neurotoxicity depends on interactions between copper ions, prion protein, and N-methyl-d-aspartate receptors. Proc. Natl. Acad. Sci. USA 2012, 109, 1737–1742. [Google Scholar] [CrossRef] [Green Version]
  96. Gasperini, L.; Meneghetti, E.; Pastore, B.; Benetti, F.; Legname, G. Prion protein and copper cooperatively protect neurons by modulating NMDA receptor through S-nitrosylation. Antioxid. Redox Signal. 2015, 22, 772–784. [Google Scholar] [CrossRef] [Green Version]
  97. Tsiroulnikov, K.; Chobert, J.-M.; Haertlé, T. Copper-dependent degradation of recombinant ovine prion protein. FEBS J. 2006, 273, 1959–1965. [Google Scholar] [CrossRef] [PubMed]
  98. Schlief, M.L.; West, T.; Craig, A.M.; Holtzman, D.M.; Gitlin, J.D. Role of the Menkes copper-transporting ATPase in NMDA receptor-mediated neuronal toxicity. Proc. Natl. Acad. Sci. USA 2006, 103, 14919–14924. [Google Scholar] [CrossRef] [Green Version]
  99. Dodani, S.C.; Firl, A.; Chan, J.; Nam, C.I.; Aron, A.T.; Onak, C.S.; Ramos-Torres, K.M.; Paek, J.; Webster, C.M.; Feller, M.B.; et al. Copper is an endogenous modulator of neural circuit spontaneous activity. Proc. Natl. Acad. Sci. USA 2014, 111, 16280–16285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Filiano, A.J.; Gadani, S.P.; Kipnis, J. Interactions of innate and adaptive immunity in brain development and function. Brain Res. 2015, 1617, 18–27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Nicastro, N.; Mak, E.; Williams, G.B.; Surendranathan, A.; Bevan-Jones, W.R.; Passamonti, L.; Vàzquez Rodrìguez, P.; Su, L.; Arnold, R.; Fryer, T.D.; et al. Correlation of microglial activation with white matter changes in dementia with Lewy bodies. NeuroImage Clin. 2020, 25, 102200. [Google Scholar] [CrossRef] [PubMed]
  102. Femminella, G.D.; Ninan, S.; Atkinson, R.; Fan, Z.; Brooks, D.J.; Edison, P. Does microglial activation influence hippocampal volume and neuronal function in Alzheimer’s disease and Parkinson’s disease dementia? J. Alzheimer’s Dis. 2016, 51, 1275–1289. [Google Scholar] [CrossRef]
  103. Nicastro, N.; Surendranathan, A.; Mak, E.; Rowe, J.B.; O’Brien, J.T. 11C-PK11195 PET imaging and white matter changes in Parkinson’s disease dementia. Ann. Clin. Transl. Neurol. 2019, 6, 2133–2136. [Google Scholar] [CrossRef]
  104. Hopperton, K.E.; Mohammad, D.; Trépanier, M.O.; Giuliano, V.; Bazinet, R.P. Markers of microglia in post-mortem brain samples from patients with Alzheimer’s disease: A systematic review. Mol. Psychiatry 2018, 23, 177–198. [Google Scholar] [CrossRef] [PubMed]
  105. Femminella, G.D.; Dani, M.; Wood, M.; Fan, Z.; Calsolaro, V.; Atkinson, R.; Edginton, T.; Hinz, R.; Brooks, D.J.; Edison, P. Microglial activation in early Alzheimer trajectory is associated with higher gray matter volume. Neurology 2019, 92, 1331–1343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Doens, D.; Fernández, P.L. Microglia receptors and their implications in the response to amyloid β for Alzheimer’s disease pathogenesis. J. Neuroinflamm. 2014, 11, 48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Block, M.L.; Zecca, L.; Hong, J.-S. Microglia-mediated neurotoxicity: Uncovering the molecular mechanisms. Nat. Rev. Neurosci. 2007, 8, 57–69. [Google Scholar] [CrossRef]
  108. Petris, M.J.; Mercer, J.F.B.; Culvenor, J.G.; Lockhart, P.; Gleeson, P.A.; Camakaris, J. Ligand-regulated transport of the Menkes copper P-type ATPase efflux pump from the Golgi apparatus to the plasma membrane: A novel mechanism of regulated trafficking. EMBO J. 1996, 15, 6084–6095. [Google Scholar] [CrossRef]
  109. Yamaguchi, Y.; Heiny, M.E.; Suzuki, M.; Gitlin, J.D. Biochemical characterization and intracellular localization of the Menkes disease protein. Proc. Natl. Acad. Sci. USA 1996, 93, 14030–14035. [Google Scholar] [CrossRef] [Green Version]
  110. Zheng, Z.; White, C.; Lee, J.; Peterson, T.S.; Bush, A.I.; Sun, G.Y.; Weisman, G.A.; Petris, M.J. Altered microglial copper homeostasis in a mouse model of Alzheimer’s disease. J. Neurochem. 2010, 114, 1630–1638. [Google Scholar] [CrossRef] [Green Version]
  111. Achard, M.E.S.; Stafford, S.L.; Bokil, N.J.; Chartres, J.; Bernhardt, P.V.; Schembri, M.A.; Sweet, M.J.; Mcewan, A.G. Copper redistribution in murine macrophages in response to Salmonella infection. Biochem. J. 2012, 444, 51–57. [Google Scholar] [CrossRef] [Green Version]
  112. Wagner, D.; Maser, J.; Lai, B.; Cai, Z.; Barry, C.E.; Höner zu Bentrup, K.; Russell, D.G.; Bermudez, L.E. Elemental Analysis of Mycobacterium avium -, Mycobacterium tuberculosis -, and Mycobacterium smegmatis -Containing Phagosomes Indicates Pathogen-Induced Microenvironments within the Host Cell’s Endosomal System. J. Immunol. 2005, 174, 1491–1500. [Google Scholar] [CrossRef] [Green Version]
  113. Festa, R.A.; Thiele, D.J. Copper at the Front Line of the Host-Pathogen Battle. PLoS Pathog. 2012, 8, e1002887. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. White, C.; Lee, J.; Kambe, T.; Fritsche, K.; Petris, M.J. A role for the ATP7A copper-transporting ATPase in macrophage bactericidal activity. J. Biol. Chem. 2009, 284, 33949–33956. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Huang, Q.; Ouyang, Z.; Tan, Y.; Wu, H.; Liu, Y. Activating macrophages for enhanced osteogenic and bactericidal performance by Cu ion release from micro/nano-topographical coating on a titanium substrate. Acta Biomater. 2019, 100, 415–426. [Google Scholar] [CrossRef]
  116. Lim, S.L.; Rodriguez-Ortiz, C.J.; Hsu, H.W.; Wu, J.; Zumkehr, J.; Kilian, J.; Vidal, J.; Ayata, P.; Kitazawa, M. Chronic copper exposure directs microglia towards degenerative expression signatures in wild-type and J20 mouse model of Alzheimer’s disease. J. Trace Elem. Med. Biol. 2020, 62, 126578. [Google Scholar] [CrossRef]
  117. Martinon, F.; Burns, K.; Tschopp, J. The Inflammasome: A molecular platform triggering activation of inflammatory caspases and processing of proIL-β. Mol. Cell 2002, 10, 417–426. [Google Scholar] [CrossRef]
  118. Lamkanfi, M.; Dixit, V.M. Inflammasomes and Their Roles in Health and Disease. Annu. Rev. Cell Dev. Biol. 2012, 28, 137–161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. He, W.; Long, T.; Pan, Q.; Zhang, S.; Zhang, Y.; Zhang, D.; Qin, G.; Chen, L.; Zhou, J. Microglial NLRP3 inflammasome activation mediates IL-1β release and contributes to central sensitization in a recurrent nitroglycerin-induced migraine model. J. Neuroinflamm. 2019, 16, 78. [Google Scholar] [CrossRef] [Green Version]
  120. Halle, A.; Hornung, V.; Petzold, G.C.; Stewart, C.R.; Monks, B.G.; Reinheckel, T.; Fitzgerald, K.A.; Latz, E.; Moore, K.J.; Golenbock, D.T. The NALP3 inflammasome is involved in the innate immune response to amyloid-β. Nat. Immunol. 2008, 9, 857–865. [Google Scholar] [CrossRef] [Green Version]
  121. Salminen, A.; Ojala, J.; Suuronen, T.; Kaarniranta, K.; Kauppinen, A. Amyloid-β oligomers set fire to inflammasomes and induce Alzheimer’s pathology: Alzheimer Review Series. J. Cell. Mol. Med. 2008, 12, 2255–2262. [Google Scholar] [CrossRef]
  122. Deigendesch, N.; Zychlinsky, A.; Meissner, F. Copper Regulates the Canonical NLRP3 Inflammasome. J. Immunol. 2018, 200, 1607–1617. [Google Scholar] [CrossRef]
  123. Dong, J.; Wang, X.; Xu, C.; Gao, M.; Wang, S.; Zhang, J.; Tong, H.; Wang, L.; Han, Y.; Cheng, N.; et al. Inhibiting NLRP3 inflammasome activation prevents copper-induced neuropathology in a murine model of Wilson’s disease. Cell Death Dis. 2021, 12, 1–12. [Google Scholar] [CrossRef] [PubMed]
  124. McGeer, P.L.; Itagaki, S.; Tago, H.; McGeer, E.G. Reactive microglia in patients with senile dementia of the Alzheimer type are positive for the histocompatibility glycoprotein HLA-DR. Neurosci. Lett. 1987, 79, 195–200. [Google Scholar] [CrossRef]
  125. Haga, S.; Akai, K.; Ishii, T. Demonstration of microglial cells in and around senile (neuritic) plaques in the Alzheimer brain. An immunohistochemical study using a novel monoclonal antibody. Acta Neuropathol. 1989, 77, 569–575. [Google Scholar] [CrossRef] [PubMed]
  126. Maezawa, I.; Zimin, P.I.; Wulff, H.; Jin, L.W. Amyloid-β protein oligomer at low nanomolar concentrations activates microglia and induces microglial neurotoxicity. J. Biol. Chem. 2011, 286, 3693–3706. [Google Scholar] [CrossRef] [Green Version]
  127. Jekabsone, A.; Mander, P.K.; Tickler, A.; Sharpe, M.; Brown, G.C. Fibrillar beta-amyloid peptide Aβ1-40 activates microglial proliferation via stimulating TNF-α release and H2O2 derived from NADPH oxidase: A cell culture study. J. Neuroinflamm. 2006, 3, 24. [Google Scholar] [CrossRef] [Green Version]
  128. Li, Y.; Tan, M.-S.; Jiang, T.; Tan, L. Microglia in Alzheimer’s Disease. Biomed Res. Int. 2014, 2014, 437483. [Google Scholar] [CrossRef] [Green Version]
  129. Kim, H.J.; Ajit, D.; Peterson, T.S.; Wang, Y.; Camden, J.M.; Wood, W.G.; Sun, G.Y.; Erb, L.; Petris, M.; Weisman, G.A. Nucleotides released from Aβ 1-42-treated microglial cells increase cell migration and Aβ 1-42 uptake through P2Y 2 receptor activation. J. Neurochem. 2012, 121, 228–238. [Google Scholar] [CrossRef] [Green Version]
  130. Li, H.Q.; Chen, C.; Dou, Y.; Wu, H.-J.; Liu, Y.-J.; Lou, H.-F.; Zhang, J.-M.; Li, X.-M.; Wang, H.; Duan, S. P2Y4 Receptor-Mediated Pinocytosis Contributes to Amyloid Beta-Induced Self-Uptake by Microglia. Mol. Cell. Biol. 2013, 33, 4282–4293. [Google Scholar] [CrossRef] [Green Version]
  131. Swanson, J.A. Shaping cups into phagosomes and macropinosomes. Nat. Rev. Mol. Cell Biol. 2008, 9, 639–649. [Google Scholar] [CrossRef] [Green Version]
  132. Mandrekar, S.; Jiang, Q.; Lee, C.Y.D.; Koenigsknecht-Talboo, J.; Holtzman, D.M.; Landreth, G.E. Microglia mediate the clearance of soluble aβ through fluid phase macropinocytosis. J. Neurosci. 2009, 29, 4252–4262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. El Khoury, J.; Hickman, S.E.; Thomas, C.A.; Cao, L.; Silverstein, S.C.; Loike, J.D. Scavenger receptor-mediated adhesion of microglia to β-amyloid fibrils. Nature 1996, 382, 716–719. [Google Scholar] [CrossRef]
  134. Pan, X.D.; Zhu, Y.G.; Lin, N.; Zhang, J.; Ye, Q.Y.; Huang, H.P.; Chen, X.C. Microglial phagocytosis induced by fibrillar β-amyloid is attenuated by oligomeric β-amyloid: Implications for Alzheimer’s disease. Mol. Neurodegener. 2011, 6, 45. [Google Scholar] [CrossRef] [Green Version]
  135. James, S.A.; Churches, Q.I.; De Jonge, M.D.; Birchall, I.E.; Streltsov, V.; McColl, G.; Adlard, P.A.; Hare, D.J. Iron, Copper, and Zinc Concentration in Aβ Plaques in the APP/PS1 Mouse Model of Alzheimer’s Disease Correlates with Metal Levels in the Surrounding Neuropil. ACS Chem. Neurosci. 2017, 8, 629–637. [Google Scholar] [CrossRef]
  136. Barnham, K.J. Tyrosine gated electron transfer is key to the toxic mechanism of Alzheimer’s disease -amyloid. FASEB J. 2004, 18, 1427–1429. [Google Scholar] [CrossRef] [PubMed]
  137. Sarell, C.J.; Wilkinson, S.R.; Viles, J.H. Substoichiometric levels of Cu2+ ions accelerate the kinetics of fiber formation and promote cell toxicity of amyloid-β from Alzheimer disease. J. Biol. Chem. 2010, 285, 41533–41540. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Smith, D.P.; Ciccotosto, G.D.; Tew, D.J.; Fodero-Tavoletti, M.T.; Johanssen, T.; Masters, C.L.; Barnham, K.J.; Cappai, R. Concentration dependent Cu2+ induced aggregation and dityrosine formation of the Alzheimer’s disease amyloid-βpeptide. Biochemistry 2007, 46, 2881–2891. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Faller, P.; Hureau, C.; Berthoumieu, O. Role of Metal Ions in the Self-assembly of the Alzheimer ’s Amyloid β Peptide. Inorg. Chem. 2013, 52, 12193–12206. [Google Scholar] [CrossRef]
  140. Tõugu, V.; Karafin, A.; Zovo, K.; Chung, R.S.; Howells, C.; West, A.K.; Palumaa, P. Zn(II)- and Cu(II)-induced non-fibrillar aggregates of amyloid-β (1-42) peptide are transformed to amyloid fibrils, both spontaneously and under the influence of metal chelators. J. Neurochem. 2009, 110, 1784–1795. [Google Scholar] [CrossRef] [PubMed]
  141. Yu, F.; Gong, P.; Hu, Z.; Qiu, Y.; Cui, Y.; Gao, X.; Chen, H.; Li, J. Cu(II) enhances the effect of Alzheimer’s amyloid-β peptide on microglial activation. J. Neuroinflamm. 2015, 12, 122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Kitazawa, M.; Hsu, H.W.; Medeiros, R. Copper exposure perturbs brain inflammatory responses and impairs clearance of amyloid-beta. Toxicol. Sci. 2016, 152, 194–204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Tan, X.; Guan, H.; Yang, Y.; Luo, S.; Hou, L.; Chen, H.; Li, J. Cu(II) disrupts autophagy-mediated lysosomal degradation of oligomeric Aβ in microglia via mTOR-TFEB pathway. Toxicol. Appl. Pharmacol. 2020, 401, 115090. [Google Scholar] [CrossRef]
  144. Mayes, J.; Tinker-Mill, C.; Kolosov, O.; Zhang, H.; Tabner, B.J.; Allsop, D. β-Amyloid fibrils in alzheimer disease are not inert when bound to copper ions but can degrade hydrogen peroxide and generate reactive oxygen species. J. Biol. Chem. 2014, 289, 12052–12062. [Google Scholar] [CrossRef] [Green Version]
  145. Dai, X.L.; Sun, Y.X.; Jiang, Z.F. Cu(II) potentiation of Alzheimer Aβ1-40 cytotoxicity and transition on its secondary structure. Acta Biochim. Biophys. Sin. 2006, 38, 765–772. [Google Scholar] [CrossRef] [Green Version]
  146. Xu, J.; Church, S.J.; Patassini, S.; Begley, P.; Waldvogel, H.J.; Curtis, M.A.; Faull, R.L.M.; Unwin, R.D.; Cooper, G.J.S. Evidence for widespread, severe brain copper deficiency in Alzheimer’s dementia. Metallomics 2017, 9, 1106–1119. [Google Scholar] [CrossRef] [Green Version]
  147. Ehehalt, R.; Keller, P.; Haass, C.; Thiele, C.; Simons, K. Amyloidogenic processing of the Alzheimer β-amyloid precursor protein depends on lipid rafts. J. Cell Biol. 2003, 160, 113–123. [Google Scholar] [CrossRef]
  148. Abad-Rodriguez, J.; Ledesma, M.D.; Craessaerts, K.; Perga, S.; Medina, M.; Delacourte, A.; Dingwall, C.; De Strooper, B.; Dotti, C.G. Neuronal membrane cholesterol loss enhances amyloid peptide generation. J. Cell Biol. 2004, 167, 953–960. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Lee, S.; Chung, C.Y.S.; Liu, P.; Craciun, L.; Nishikawa, Y.; Bruemmer, K.J.; Hamachi, I.; Saijo, K.; Miller, E.W.; Chang, C.J. Activity-Based Sensing with a Metal-Directed Acyl Imidazole Strategy Reveals Cell Type-Dependent Pools of Labile Brain Copper. J. Am. Chem. Soc. 2020, 142, 14993–15003. [Google Scholar] [CrossRef] [PubMed]
  150. He, W.; James Kang, Y. Ischemia-induced copper loss and suppression of angiogenesis in the pathogenesis of myocardial infarction. Cardiovasc. Toxicol. 2013, 13, 1–8. [Google Scholar] [CrossRef] [PubMed]
  151. Berenshtein, E.; Mayer, B.; Goldberg, C.; Kitrossky, N.; Chevion, M. Patterns of mobilization of copper and iron following myocardial ischemia: Possible predictive criteria for tissue injury. J. Mol. Cell. Cardiol. 1997, 29, 3025–3034. [Google Scholar] [CrossRef] [PubMed]
  152. Li, K.; Li, C.; Xiao, Y.; Wang, T.; James Kang, Y. The loss of copper is associated with the increase in copper metabolism MURR domain 1 in ischemic hearts of mice. Exp. Biol. Med. 2018, 243, 780–785. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Sanchez-Perez, A.; Llansola, M.; Cauli, O.; Felipo, V. Modulation of NMDA receptors in the cerebellum. II. Signaling pathways and physiological modulators regulating NMDA receptor function. Cerebellum 2005, 4, 162–170. [Google Scholar] [CrossRef]
  154. Swanger, S.A.; Vance, K.M.; Pare, J.F.; Sotty, F.; Fog, K.; Smith, Y.; Traynelis, S.F. NMDA receptors containing the GluN2D subunit control neuronal function in the subthalamic nucleus. J. Neurosci. 2015, 35, 15971–15983. [Google Scholar] [CrossRef] [Green Version]
  155. Mullasseril, P.; Hansen, K.B.; Vance, K.M.; Ogden, K.K.; Yuan, H.; Kurtkaya, N.L.; Santangelo, R.; Orr, A.G.; Le, P.; Vellano, K.M.; et al. A subunit-selective potentiator of NR2C- and NR2D-containing NMDA receptors. Nat. Commun. 2010, 1, 90. [Google Scholar] [CrossRef] [Green Version]
  156. Llansola, M.; Sanchez-Perez, A.; Cauli, O.; Felipo, V. Modulation of NMDA receptors in the cerebellum. 1. Properties of the NMDA receptor that modulate its function. Cerebellum 2005, 4, 154–161. [Google Scholar] [CrossRef] [PubMed]
  157. Larner, A.J. The Cerebellum in alzheimer’s disease. Dement. Geriatr. Cogn. Disord. 1997, 8, 203–209. [Google Scholar] [CrossRef]
  158. Braak, H.; Braak, E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol. 1991, 82, 239–259. [Google Scholar] [CrossRef] [PubMed]
  159. Condello, C.; Yuan, P.; Schain, A.; Grutzendler, J. Microglia constitute a barrier that prevents neurotoxic protofibrillar Aβ42 hotspots around plaques. Nat. Commun. 2015, 6, 1–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Postulated differences in microglia activity with either the absence or presence of a high copper concentration. When high levels of amyloid are released into the synapse, amyloid fibrils are formed and the microglia are activated, clearing the fibrils by phagocytosis (a). Releasing high levels of copper into the synapse, as copper inhibits the NMDA receptor, results in soluble Aβ deposits, which are then taken up by microglia via pinocytosis, causing overactivation of the microglia and inflammation (b).
Figure 1. Postulated differences in microglia activity with either the absence or presence of a high copper concentration. When high levels of amyloid are released into the synapse, amyloid fibrils are formed and the microglia are activated, clearing the fibrils by phagocytosis (a). Releasing high levels of copper into the synapse, as copper inhibits the NMDA receptor, results in soluble Aβ deposits, which are then taken up by microglia via pinocytosis, causing overactivation of the microglia and inflammation (b).
Inorganics 10 00006 g001
Figure 2. Model of microglia’s dual role in AD. The activation of microglia is key to neuronal survival (a). The presence of large amounts of copper in the synapse changes the role of microglia from protective to lethal and also induces neurons’ copper deficiency (b).
Figure 2. Model of microglia’s dual role in AD. The activation of microglia is key to neuronal survival (a). The presence of large amounts of copper in the synapse changes the role of microglia from protective to lethal and also induces neurons’ copper deficiency (b).
Inorganics 10 00006 g002aInorganics 10 00006 g002b
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Bagheri, S.; Saboury, A.A.; Haertlé, T.; Rongioletti, M.; Saso, L. Probable Reasons for Neuron Copper Deficiency in the Brain of Patients with Alzheimer’s Disease: The Complex Role of Amyloid. Inorganics 2022, 10, 6. https://0-doi-org.brum.beds.ac.uk/10.3390/inorganics10010006

AMA Style

Bagheri S, Saboury AA, Haertlé T, Rongioletti M, Saso L. Probable Reasons for Neuron Copper Deficiency in the Brain of Patients with Alzheimer’s Disease: The Complex Role of Amyloid. Inorganics. 2022; 10(1):6. https://0-doi-org.brum.beds.ac.uk/10.3390/inorganics10010006

Chicago/Turabian Style

Bagheri, Soghra, Ali A. Saboury, Thomas Haertlé, Mauro Rongioletti, and Luciano Saso. 2022. "Probable Reasons for Neuron Copper Deficiency in the Brain of Patients with Alzheimer’s Disease: The Complex Role of Amyloid" Inorganics 10, no. 1: 6. https://0-doi-org.brum.beds.ac.uk/10.3390/inorganics10010006

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

Article Metrics

Back to TopTop