HHS Public Access: Metal Toxicity Links To Alzheimer's Disease and Neuroinflammation
HHS Public Access: Metal Toxicity Links To Alzheimer's Disease and Neuroinflammation
HHS Public Access: Metal Toxicity Links To Alzheimer's Disease and Neuroinflammation
Author manuscript
J Mol Biol. Author manuscript; available in PMC 2020 April 19.
Author Manuscript
2Centre for Stem Cell Ageing and Regenerative Engineering, The University of Queensland,
Australia.
3Queensland Brain Institute, The University of Queensland, Australia.
4Center for Occupational and Environmental Health, Department of Medicine, University of
California, Irvine, CA, USA.
Abstract
As the median age of the population increases, the number of individuals with Alzheimer’s disease
(AD) and the associated socio-economic burden are predicted to worsen. While aging and inherent
genetic predisposition play major roles in the onset of AD, lifestyle, physical fitness, medical
condition, and social environment have emerged as relevant disease modifiers. These
Author Manuscript
environmental risk factors can play a key role in accelerating or decelerating disease onset and
progression. Among known environmental risk factors, chronic exposure to various metals has
become more common among the public as the aggressive pace of anthropogenic activities
releases excess amount of metals into the environment. As a result, we are exposed not only to
essential metals, such as iron, copper, zinc and manganese, but also to toxic metals including lead,
aluminum, and cadmium, which perturb metal homeostasis at the cellular and organismal levels.
Herein, we review how these metals affect brain physiology and immunity, as well as their roles in
the accumulation of toxic AD proteinaceous species (i.e., β-amyloid and tau). We also discuss
studies that validate the disruption of immune-related pathways as an important mechanism of
toxicity by which metals can contribute to AD. Our goal is to increase the awareness of metals as
players in the onset and progression of AD.
Author Manuscript
Keywords
Environment; dementia; β-amyloid; tau; neurodegeneration
*
Corresponding Authors: Rodrigo Medeiros, Ph.D., rodrigo.medeiros@neurula.org or r.medeiros@uq.edu.au. Tee Jong Huat, Ph.D.,
t.tee@uq.edu.au, Address: Queensland Brain Institute, The University of Queensland, Brisbane, 4072, Australia. Phone:
+61734431104.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our
customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of
the resulting proof before it is published in its final citable form. Please note that during the production process errors may be
discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Huat et al. Page 2
Introduction
Author Manuscript
individual’s risk of developing AD, many of which are potentially modifiable, including
dietary habits and exposure to environmental and occupational hazards [2, 3].
production and misfolding occur prior to any of the other pathologies associated with AD,
the trigger(s) for this remains to be fully identified [5]. The production of Aβ has been shown
to lead to the hyperphosphorylation of the microtubule-associated tau protein, stimulated by
the phosphorylation of the tau kinases glycogen synthase kinase-3β (GSK-3β), cyclin-
dependent kinase 5 (CDK5) and extracellular signal-regulated kinase 1/2 (ERK1/2) [6, 7].
The hyperphosphorylation of tau can cause microtubule destabilization and breakdown,
perturb axonal transport, and make tau far more prone to aggregation into NFTs [8–10]. NFT
load correlates with cognitive impairment and neurodegeneration, leading to the suggestion
that reducing tau hyperphosphorylation and NFT formation are key to preventing AD [11, 12].
The brain has mechanisms to clear toxic proteins, including degradation pathways and the
immune system. Inflammation poses a key risk for AD with respect to both genetic (e.g.,
Author Manuscript
apoe, trem2, cd33) and lifestyle factors (e.g., diabetes mellitus, obesity, traumatic brain
injury) [3]. With respect to the proteins involved in AD, both Aβ and tau can be altered by
neuroinflammation. Accumulation of Aβ triggers a proinflammatory response from the
brain’s resident immune cells, microglia and astrocytes, leading to the phagocytosis of
plaques as well as their proteolytic degradation. Receptors on microglia, such as triggering
receptor expressed on myeloid cells 2 (TREM2) and toll-like receptors (TLRs), can
recognize Aβ and trigger a phagocytic pathway [13, 14]. Aβ degrading enzymes, including
neprilysin and insulin degrading enzyme, can also be released to remove Aβ extracellularly,
although the activity of these enzymes is reduced in AD [15, 16]. Moreover, the exacerbated
Author Manuscript
proinflammatory state that occurs during this period of the disease can trigger the
hyperphosphorylation of tau. Several of the kinases responsible for tau phosphorylation are
activated by proinflammatory mediators and have been shown to worsen tau pathology
[17, 18]. Interestingly, microglial uptake of hyperphosphorylated tau is linked to the
Among the chemical elements of relevance to humans, metals play a significant role in both
Author Manuscript
health and disease. Metals are natural constituents of the Earth’s crust and are disseminated
into the biosphere through human activities [24, 25]. These compounds display high stability,
solubility in atmospheric precipitation, and ability to be absorbed by soil and living
organisms, with human exposure soaring because of an exponential increase in their use in
several industrial, agricultural, domestic and technological applications. Common sources of
metals are mining, tailings, industrial waste, agricultural runoff, paints, treated timber, aging
water supply infrastructure, vehicle emissions, lead-acid batteries, fertilizers and
microplastics. The main routes of human exposure include ingestion, inhalation and dermal
contact [25]. Physiologically, some metals are either essential nutrients (e.g., iron and zinc)
or relatively harmless (e.g., ruthenium, silver, and indium), but even these can be toxic in
larger amounts or certain forms (Fig. 2). This is because metals are usually essential
components of larger biological molecules that can interact with or regulate the levels of
Author Manuscript
relatively large numbers of other molecules [26]. This means that the optimal physiological
concentration range between deficiency and toxicity of metals is relatively small and needs
to be tightly controlled. Importantly, tiny amounts of non-essential metals also promote
severe toxicity as they inadvertently disrupt the physiological activity of essential metals.
Because of their high degree of toxicity, cadmium, lead and aluminum rank among the
priority metals that are of public health significance (Fig. 2). These metallic elements are
considered systemic toxicants that are known to induce multiple organ damage, even at
lower levels of exposure. Notably, evidence suggests that dysregulation in the homeostasis of
essential metals (Fig. 3) and exposure to non-essential metals (Fig. 4) have significant
impact on the pathogenesis of AD. In the next part of this review we will discuss the impact
that some of these trace metals exert in the brain, and how they contribute to AD and the
dysregulation of the immune system.
Author Manuscript
of the spinal cord and white matter, and in the synthesis, packaging, uptake and degradation
Author Manuscript
have been linked to several diseases. While brain iron deficiency results in unfavorable
pregnancy outcomes and cognitive developmental defects in children, such as attention
deficit hyperactivity disorder [31], its excessive accumulation in adults has been linked to
neurodegenerative diseases, including AD [32–34]. Changes in iron levels in AD can be
detected as early as the mild cognitive impairment stage of the disease [35]. For this reason,
iron-based magnetic resonance imaging contrast, which can detect iron content in the brain,
has been investigated as a potential imaging biomarker due to its capacity to discriminate
pathological changes related to AD [36–39]. Recent evidence has demonstrated that
cerebrospinal fluid ferritin levels predict brain hypometabolism in people with underlying
AD pathology [40]. Together, these studies indicate that changes in iron metabolism may
facilitate disease progression in the prodromal stages, which could be used as a biomarker of
AD.
Author Manuscript
Over the years, many studies have helped to mechanistically link iron metabolism and AD.
For instance, APP mRNA encodes a functional IRE in its 5′-untranslated region that binds
with IRP-1, and its translation is down-regulated in response to intracellular iron chelation
[41, 42]. At the protein level, APP acts as a modulator of iron function, as it oxidizes Fe2+,
loads Fe3+ into transferrin, and interacts with the iron transporter ferroportin. In vitro studies
have shown that ablation of APP induces marked iron retention in cells, whereas APP
overexpression promotes iron export. Likewise, genetic deletion of APP in mice causes iron
accumulation and oxidative stress in cortical neurons, making animals vulnerable to dietary
iron exposure compared to normal mice [43]. Moreover, several of the biological
abnormalities seen in AD are consistent with an excessive action of oxygenic free radicals
caused by impairment in iron homeostasis, and Aβ plaques and NFTs are major sites for
Author Manuscript
catalytic redox reactivity [44, 45]. Iron and its regulatory proteins ferritin (iron storage) and
transferrin (iron mobilization) are largely found in oligodendrocytes in physiological
conditions. In AD, however, substantial amounts of iron and ferritin are detected within
plaques, NFTs and blood vessels [46, 47, 45]. Although iron- and ferritin-positive
oligodendrocytes are present, most of the iron- and ferritin-containing cells associated with
the markers of AD are microglia [48–51]. Likewise, transferrin is distributed extracellularly
around plaques and in astrocytes in the AD brain rather than its normal distribution in
oligodendrocytes [48]. It has been suggested that microglia and astrocytes accumulate higher
Author Manuscript
iron content in AD because they are more resistant to oxidative stress than neuronal cells
[52]. Despite this, the accumulation of iron in these glial cells does seem to compromise their
function [53]. Other components of the iron network are also altered in AD, including
lactotransferrin [54, 55], melanotransferrin [56], and IRP-2 [57]. Changes in iron metabolism
are therefore prominent in AD at the cellular level. However, how these changes affect the
function of the distinct brain cells and their communication, remain to be fully elucidated.
A large part of iron’s detrimental effect in the AD is related to its cytotoxic redox properties,
which are further impaired during the progression of the disease. In a mouse model, the
neurological impairment induced by the infusion of Aβ peptide was exacerbated by the
genetic deletion of mitochondrial ferritin, a process that was associated with an
accumulation of intracellular iron and increased levels of oxidative stress [58]. In vitro
studies have demonstrated that the affinity of Aβ for iron increases following its
Author Manuscript
aggregation, and that this binding potentiates Aβ neuronal toxicity [59–62]. This occurs
because the binding between iron and Aβ modulates the redox potential to a level at which
iron’s redox cycling occurs, resulting not only in the generation of oxidative species, but also
the depletion of essential oxygen and biological reductants. By binding to iron, Aβ also
competes against other essential iron-containing proteins. For example, the affinity of Aβ for
iron is eight orders of magnitude stronger than that of transferrin, and its accumulation can
therefore change the iron homeostasis [63]. In agreement with this idea, it has been shown
that Aβ deposition in the APP/PS1 mouse model is accompanied by changes in iron-related
proteins, DMT1 and ferroportin 1 (FPN1), whose levels in the brain are increased and
reduced, respectively [64]. Of note, DMT1 relocates to cellular and endosomal membranes,
where it is a key player in non-transferrin bound iron uptake and transferrin-bound iron
uptake, respectively [65]. FPN1, on the other hand, plays an essential role in the export of
Author Manuscript
iron from cells to the blood [66]. Interestingly, in vitro studies have demonstrated that
silencing of endogenous DMT1 not only reduces iron influx but also leads to reductions in
APP expression and Aβ production [67–69]. Pharmacological inhibition of DMT1 also seems
to reduce iron-induced tau pathology in human neuroblastoma SH-SY5Y cells, through the
inhibition of the tau kinases CDK5 and GSK-3β [70]. Importantly, other studies have
demonstrated the capacity of iron to interfere with markers of AD pathology, as in vitro and
in vivo data show that iron overload results in higher Aβ production, neuronal toxicity and
cognitive impairment [71, 72]. Another relevant study in the APP/PS1 mouse model revealed
that the iron chelating agent deferoxamine inhibits Aβ accumulation and improves cognitive
function. It was proposed that this effect is dependent on the alternative activation of
microglia, which become more prone to clear Aβ via phagocytosis [73]. This hypothesis,
however, needs further validation as many agents that reduce Aβ deposition in mice can
Author Manuscript
With regards to changes in the brain immune system, microglial dystrophy in the aged and
AD brains is associated with ferritin immunoreactivity [53]. Disruption of iron homeostasis
through brain hemorrhage also enhances microgliosis and astrogliosis, and results in
elevated levels of the proinflammatory cytokines tumor necrosis factor-α (TNF-α) and
interleukin-1β (IL-1β) [74]. This is particularly interesting since traumatic brain injury is a
potential risk factor for AD [3]. Excess iron has also been shown to activate microglia by
and IL-6, triggers the upregulation of DMT1 and downregulation of ferroportin, suggesting
that the mechanism for iron accumulation into neurons and microglia during inflammation is
in part due to changes in the levels of iron transporters [77]. Iron accumulation in
macrophages and microglia following CNS damage has also been linked to a shift in cell
activation towards a proinflammatory phenotype, which results in lower phagocytic
efficiency [78]. Although these studies suggest that a positive feedback loop between iron
accumulation and excessive proinflammation progressively contributes to neurotoxicity, they
failed to provide a clear molecular mechanism by which iron contributes to the function of
glial cells. Particularly, it would be important to determine whether iron metabolism directly
regulates functions of microglia and astrocytes, like immune surveillance and synaptic
pruning, during homeostasis and disease states.
Author Manuscript
Copper (Cu)
Like iron, copper is a highly desirable chemical element in cell biology due to its ability to
gain and donate electrons. Once taken up and distributed in an organism, copper ions cycle
between cupric Cu2+ (oxidized) and cuprous Cu+ (reduced) states, mostly bound to
cuproenzymes with only a small portion available as labile copper, also known as free or
unbound copper [79]. This redox property is essential for catalysis by many enzymes whose
activities control a broad range of cellular biochemical and regulatory functions. The
relevance of copper for human health is easily emphasized by two life-threatening disorders
caused by mutations in P-type Cu+-transporting ATPase pumps: Menkes and Wilson’s
diseases [80]. Menkes disease is an X-linked lethal disorder of intestinal copper
hyperaccumulation, and severe copper deficiency in peripheral and central tissues. It is
Author Manuscript
many roles in processes essential for normal brain function, including the synthesis of
catecholamines, activation of neuropeptides and hormones, antioxidant defense, connective
tissue production, immune function and synaptic transmission [82–84]. Wilson’s disease, on
the other hand, is an autosomal recessive disease caused by mutations in the ATP7B protein,
a transporter that loads Cu+ onto newly synthesized cuproenzymes in the trans-Golgi
network (TGN) and exports excess copper out of cells by trafficking from the TGN to the
plasma membrane. The disease is characterized by striking hepatic and neuronal copper
overload, hepatotoxicity, and neuropsychological and other defects that require chronic
therapy to enhance copper excretion or reduce its absorption [85]. Remarkably, this disease
Author Manuscript
highlights the fact that copper’s greatest strength is also its major weakness as excess causes
it to participate in redox reactions that generate ROS, leading to catastrophic damage to
lipids, proteins and DNA. Therefore, intracellular levels of copper must be tightly regulated
as excessive free copper is highly toxic [79].
Considering the robust evidence for copper’s essential roles in the brain, it is not surprising
that many studies have proposed that an imbalance in its homeostasis is associated with
neurodegenerative disorders. Besides AD, copper has been linked to amyotrophic lateral
In AD, it has been suggested that individuals suffering from the disease have higher
serum levels of copper than healthy controls, with higher levels of labile copper in the serum
correlating with poor cognitive performance and increased rates of conversion from mild
cognitive impairment to AD [91–93]. Analyses of post-mortem human brains have revealed
that the overall soluble levels of copper are reduced, whereas its presence within insoluble
neuritic plaques is increased, in AD versus non-demented individuals [94–96]. Notably,
despite a lower total copper content, AD brains possess a higher proportion of redox-active
exchangeable copper, which positively correlates with increased oxidative damage and AD
neuropathology [97].
Biochemical analyses have identified two binding sites for copper in full-length APP,
including one within the Aβ sequence. Binding of Cu2+ to the N-terminal domain of APP
results in its reduction to Cu+ [98]. Genetic studies in mouse models have demonstrated that
Author Manuscript
the APP-induced conversion of Cu2+ to Cu+ favors copper removal from the brain, which
could explain the fact that AD patients have lower levels of copper in their brain and higher
levels in their serum [99, 96, 91–93, 100]. Binding of copper to the N-terminus of APP may also
control other aspects of this protein, including its synaptogenic function, stability and
metabolism [101–105]. Interestingly, reduction in the level of brain copper increases the ratio
of APP endocytosis and processing, and the production of Aβ, perhaps as a protective
mechanism to reverse the excessive loss of copper [101, 103]. However, newly synthesized
intracellular Aβ can sequester copper, which possibly serves to initially exacerbate the
imbalance in copper levels and then later enable the aggregation of Aβ into plaques. Such a
process is highly relevant in the formation of neuritic plaques in AD because copper can
potentiate Aβ aggregation and cell damage due to the generation of ROS [106–108]. The
protein tau has also been shown to bind copper, which facilitates the formation of NFTs
Author Manuscript
[109–111]. Likewise, tau demonstrates redox activity when bound to copper, further
contributing to the oxidative damage in the brain [112]. Considering the significance of tau in
AD, it is surprising that only a small proportion of studies has investigated how changes in
copper homeostasis affect its function. Future studies assessing whether copper regulates tau
kinases and phosphatases as well as tau-mediated cognitive impairment are needed, and will
greatly increase our understanding of the neuropathology of AD.
Interestingly, the effects produced by these complexes are not observed upon treatment with
either copper or Aβ alone [113, 114]. Recently, we have demonstrated that exposure to copper-
Aβ reduces the phagocytic phenotype of BV2 microglia and increases TNF-α and IL-1β
release, followed by significant down-regulation of lipoprotein receptor-related protein-1
(LRP-1) expression [115]. Reduced levels of LRP-1 further impair the transcytotic clearance
of Aβ and exacerbate neuroinflammation [3]. It has been reported that trace copper
potentiates the Aβ-induced inflammatory response in cholesterol-fed mice; however, no
inflammatory effects were observed upon treatment with copper or cholesterol alone [116].
These studies highlight the role of copper as a cofactor in increasing the potency of Aβ
Author Manuscript
Different strategies have been used to prevent the pathogenic effects caused by an imbalance
in copper homeostasis, including administration of metal chelators that redistribute brain
metal pools and reverse Aβ plaque aggregation [121–126]. Alternatively, reestablishing the
proper intracellular bioavailable copper reduces secreted Aβ levels via a mechanism that is
dependent on the activation of phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) and c-
Jun N-terminal kinase [127]. Likewise, it has recently been shown that increased intracellular
copper inhibits Aβ production by directly targeting the subunits PSEN and nicastrin in the
γ-secretase complex [128]. Increasing intracellular copper bioavailability can also restore
cognitive function by inhibiting the accumulation of neurotoxic Aβ and phosphorylated tau
[129]. Furthermore, there is evidence that the immunomodulatory potential of systemic
Author Manuscript
copper bis(thiosemicarbazones), which are stable, lipophilic neutral Cu2+ complexes that are
capable of crossing cell membranes and the blood-brain barrier. This compound can inhibit
microglial and astrocytic activation, and reducing the acute cerebrovascular inflammation
caused by bacterial lipopolysaccharide [130]. Based on the evidence above, it would be
expected that increasing copper levels in the body would restore its homeostasis in the brain.
Unfortunately, systemic administration of copper promotes detrimental effects in AD by
reducing Aβ clearance. For instance, trace levels of copper in drinking water has been
associated with higher Aβ levels in the brain of distinct animal models [131, 132], a process
that seems to be related to the dysfunction of LRP-1-mediated Aβ clearance through the
vasculature [133]. In addition to its effect on Aβ, chronic systemic copper exposure
exacerbates tau pathology, promotes cognitive impairment, and dysregulates the tau-related
kinase CDK5 and the synaptic-related proteins complexin-1 and complexin-2, in a mouse
Author Manuscript
model of AD [134, 135]. Taken altogether, these studies indicate that systemic copper
promotes AD and therefore controlled copper diets should be considered. Despite the
challenges, mitigating dysregulation in copper homeostasis in the brain clearly has great
benefits and should be further explored as a therapeutic strategy for AD.
Zinc (Zn)
Author Manuscript
The brain contains the highest zinc concentrations of any organ in the body. Seventy percent
of proteins present in the brain contain zinc as a structural or catalytic component,
contributing to the efficient performance of over 2,000 transcription factors and more than
300 enzymes [136]. The transport of zinc into the brain parenchyma occurs via the blood-
brain and blood-cerebrospinal fluid barriers [137]. Its binding with L-histidine, in both
plasma and the cerebrospinal fluid, is involved in transferring zinc to target sites, regulating
its uptake across the brain barrier systems [137]. Following its uptake, zinc can be transferred
freely through the cerebrospinal and the brain extracellular fluid compartments [137]. Zinc
homeostasis in the brain is tightly regulated, primarily via three families of proteins: the
metallothioneins, which are involved in the regulation and maintenance of intracellular zinc
homeostasis [138]; the zinc- and iron-like regulatory proteins, which are responsible for zinc
uptake from extracellular fluids into both neurons and glia [139], and the zinc transporters,
Author Manuscript
which are associated with cellular zinc efflux [140]. Interestingly, many of these zinc
regulatory proteins also regulate other metal ions. In the brain, zinc is also present in its free
ionic form (Zn2+) and enriched within synaptic vesicles at glutamatergic nerve terminals
from where it is synaptically released during neuronal activity [141–143]. Zinc released in the
synaptic cleft affects the expression and activity of N-methyl-Daspartate (NMDA) and α-
amino-3-hydroxy-5-methyl-4-isoxazole propionate (AMPA) glutamatergic receptors, and
glycine ionotropic and γ-aminobutyric acid (GABAA) receptors [144]. Therefore, zinc is
intimately linked to the balance of excitation and inhibition signaling in the brain and is
essential for memory function and behavior [145].
in AD began with the observation that it can precipitate Aβ into plaques above a
concentration of 300 nM [151]. Interestingly, the extracellular concentration of zinc during
synaptic transmission rises to 300 μM, and it is therefore possible that synaptic transmission
contributes to Aβ deposition in the diseased brain [152–154]. Aβ itself is a metalloprotein that
contains binding sites for zinc, which is enriched in plaques and in the cerebral amyloid
angiopathy surrounding diseased blood vessels [155–157]. Studies using Tg2576 transgenic
mice crossed with zinc transporter 3 knockout mice, which is the transporter responsible for
the accumulation of Zn2+ in presynaptic vesicles, have reported reduced plaque load,
indicating that synaptic zinc does indeed contribute to Aβ deposition [158]. Moreover, zinc
has a high affinity for Aβ, and when trapped by accumulated Aβ, it inhibits APP ferroxidase
activity, thereby increasing the levels of iron and ROS [43, 159].
Author Manuscript
copper chelators reduce Aβ accumulation, it has recently been demonstrated that this
strategy can result in potentially detrimental effects in the healthy brain [121–126]. Depletion
Author Manuscript
of zinc levels in the brain of young mice (2.5 months old) using the zinc/copper chelator
clioquinol impaired short- and long-term memory performance. Mechanistically, treatment
with clioquinol reduced levels of brain derived neurotrophic factor (BDNF), synaptic
plasticity-related proteins and dendritic spine density in vivo [163]. These changes were
regionally restricted to the hippocampus, cortex, and striatum, without having any effects on
the cerebellum, an area devoid of pools of cheatable zinc [163, 164]. These results support the
notion that zinc is an important modulator of synaptic plasticity, neurotransmission, neuronal
function and cognitive processes in the brain, and highlights the potential detrimental
consequences of reducing the availability of zinc in the brains of healthy individuals or in
the early stages of AD. Notably, some studies that raised zinc levels via supplementation
showed an increase in plaque number and size in mouse models of AD [165–167]. In Tg2576
Author Manuscript
mice, the intake of zinc acetate caused a reduction of insoluble Aβ in the brain, despite the
absence of any significant changes in cognition and behavior [168]. The discrepancies related
to the effect of zinc intake on plaque load might be due to its diverse roles in the regulation
of Aβ, as zinc has been shown to prevent the proteolytic degradation of Aβ by matrix
metalloprotease 2, and to modulate the activity of the α-, γ- and β-secretases [169, 170].
Contradictory results are also present when comparing post-mortem analyses of zinc levels
in AD brains, as there are studies showing increased, decreased or unchanged zinc levels
[171–173, 94, 174, 175].
Zinc not only affects Aβ aggregation, but also the level of hyperphosphorylated tau and the
formation of NFTs. Like the effects of zinc on Aβ, low micromolar zinc concentrations can
cause the aggregation of tau [176–178]. Zinc is also able to promote tau hyperphosphorylation
indirectly via the inactivation of major tau phosphatases, such as protein phosphatase 2A
Author Manuscript
(PP2A) [179, 180]. Importantly, zinc chelators or blockade of synaptic zinc signaling can
abolish zinc-mediated tau hyperphosphorylation [181]. Accordingly, zinc supplementation in
a tau mouse model intensified the cognitive deficit, in association with an increase in tau
phosphorylation, and the number of NFTs in the hippocampus, and a decrease in free zinc
ion levels [182]. The influence of zinc on tau pathology was further confirmed when mice
with advanced pathology were treated with a copper/zinc chaperone. This caused increased
PP2A activity, and was sufficient to improve memory, to decrease tau pathology and to
prevent neurodegeneration [183]. This highlights the potential value of targeting zinc in
pathological conditions in which tau pathology is present. However, additional investigation
regarding the impact of zinc on the activity of tau-related kinases would be welcomed to
further validate the protective role of this metal on tau pathology. Oxidative stress is another
factor that contributes to the progression of AD, and ROS or exogenous oxidants can
Author Manuscript
promote harmful zinc release from metallothioneins [184–186]. Zinc accumulation can, in
turn, induce mitochondrial dysfunction and further oxidative stress, being particularly high
in AD neurons expressing mutant APP, PSEN1 and tau [187, 188].
In terms of immunity, zinc seems to be essential for immune cell proliferation, antioxidant
response, acquired and innate responses [189, 190]. Interestingly, the molecular mechanisms
by which zinc improves immune function are unknown. Studies in mice have shown that
dietary zinc is an important nutritional factor for a proper immune response [191, 192].
Accordingly, low zinc status is associated with increased susceptibility to infection and
Author Manuscript
altered zinc homeostasis, as only 40% of elderly people have a sufficient zinc intake
[147, 195]. Impaired zinc homeostasis promotes immune dysfunction and has been associated
The above findings, even though at times inconsistent, strongly support the hypothesis that
disturbed zinc homeostasis plays an important role in the pathogenesis of AD. Giving that
zinc contributes to the aggregation of Aβ into plaques as well as the formation of NFTs,
inflammation and oxidative stress, research has focused on the development of compounds
to neutralize its toxicity in AD. Administration of a metal-protein attenuating compound that
affects copper- and zinc-mediated toxic Aβ oligomerization lowered the cerebrospinal fluid
levels of Aβ and improved cognition in AD patients [201–203]. Other studies in mouse models
of AD have reported comparable results. Supplementation with L-carnosine, a compound
with chelating properties, in 3xTg-AD mice reduced the intraneuronal accumulation of Aβ,
and completely rescued the mitochondrial dysfunction [204]. Zinc supplementation in the
same mouse model delayed hippocampal-dependent memory deficits and strongly reduced
Author Manuscript
both Aβ and tau pathology [205]. The results of these studies highlight the integral role of
zinc in the pathogenesis of AD and support the hypothesis that restoring zinc homeostasis is
a potential strategy to treat AD. However, it is also important to emphasize the need for
mechanistic studies demonstrating how zinc promotes its effects, particularly at cellular and
molecular levels in the brain.
Manganese (Mn)
Manganese is a naturally occurring trace element that is essential for human development
and brain function. Excessive manganese is neurotoxic and has been linked to
developmental disorders and neurodegenerative disorders associated with basal ganglia
dysfunction, such as Parkinson’s disease and Huntington’s disease [206–208]. Moreover, the
relevance of manganese for the regulation of brain functions has been further emphasized by
Author Manuscript
the discovery of loss-of-function mutations in genes related to its transport, which lead to
neurotoxicity [209]. The neurotoxicity induced by manganese overexposure includes the
disruption of mitochondrial function, disruption of neurotransmitter metabolism, alteration
of iron homeostasis and induction of oxidative stress [210–214]. Manganese has also been
linked to the regulation of brain immunity, and it can have profound effects on microglia and
astrocytes, regulating the activation of proinflammatory responses which contribute to its
neurotoxic effects [215–217]. The link between manganese and AD, however, is still very
limited. It has been demonstrated that the concentration of manganese does not change in
response to human aging and AD [218]. In the periphery, conflicting data have been
Author Manuscript
presented, although a systematic review and meta-analysis has indicated that the manganese
level in the serum of AD subjects is reduced compared to that in healthy controls [219].
Although manganese can bind to Aβ, it does so with a weak binding affinity in the
millimolar to micromolar range, suggesting that it does not have a large effect on plaque
formation [220]. However, studies in non-human primates have shown that chronic
manganese exposure produces a cellular stress response that leads to neurodegenerative
changes, diffuse Aβ plaques in the frontal cortex, and impairments in visuospatial
associative learning [221, 222]. Mechanistically, manganese neurotoxicity seems to be related
to excessive iron accumulation via translational repression of APP ferritin [223]. Manganese
has also been shown to reduce the glial glutamate transporter-1 (GLT1), which may cause
sustained glutamate neurotransmission and excitotoxicity [224]. Likewise, manganese
Author Manuscript
and considered a heavy metal. Although lead toxicity has been known for many centuries, it
was only in 1892 that it was recognized as a serious threat to health following the report that
white lead paint on the porches and rails in houses in Brisbane, Australia was the cause of
severe neurological disorders in children [226]. Environmental lead absorbed into the
bloodstream has a half-life of 30 days. Lead binds to circulating erythrocytes and is
distributed throughout the body, eventually accumulating in bone. The half-life of bone-
deposited lead can span 20–30 years. Blood lead levels tend to increase during pregnancy,
menopause, lactation and aging due to an increase bone demineralization, which causes the
release of stored lead [227–229]. The presence of lead in the blood interferes with many
organs and functions of the body, but the CNS is by far the most vulnerable. In the brain, the
effect of lead can be classified as either morphological or pharmacological. Morphological
effects alter neuronal differentiation, myelination, and synaptogenesis [230–232].
Author Manuscript
Pharmacologically, lead competes with biometals, particularly calcium and to a lesser extent
zinc, for their binding sites, thereby disrupting the corresponding essential biometal-
dependent mechanisms [233]. Due to its ability to substitute for calcium ions, lead rapidly
crosses the blood-brain barrier and causes severe damage to the brain [234]. Lead also
interferes with neurotransmitter release, disrupting the function of the GABAergic,
dopaminergic and cholinergic systems as well as inhibiting NMDA receptors [235, 236].
Furthermore, lead is involved in the inactivation of glutathione, an important antioxidant
found in cells, by binding to sulfhydryl groups [237]. It is now well known that lead exposure
during childhood is associated with cognitive deficits and behavioral disturbances [238, 239].
Author Manuscript
It has been reported that juvenile lead exposure inhibits NMDA and AMPA receptors,
impairing LTP and promoting detrimental synaptic morphological changes in hippocampal
CA1 pyramidal neurons, thereby leading to a decline in learning and memory [240].
Similarly, animals exposed to lead either prenatally or postnatally develop memory
impairment and cognitive decline later in life [241, 242]. Recently, it has been reported that
low-level gestational lead exposure results in dendritic spine alterations in the hippocampus
by down-regulating neuroligin-1 protein levels, which in turn results in learning and memory
impairment [243]. Given that the effects of early life exposure to lead can persist in
adulthood, it is possible that this contributes to the development of AD. In fact, a
longitudinal study in former organolead manufacturing workers has shown that past lead
exposure is associated with a longitudinal decline in cognitive function and persistent brain
lesions [244, 245].
Author Manuscript
Numerous studies have reported that either developmental or acute lead exposure contributes
to the hallmarks of AD, including Aβ accumulation, tau pathology and inflammation. Early
lead exposure in young rats increased the expression of APP and β-secretase 1 (BACE1)
which subsequently induced AD-like pathology by inducing Aβ accumulation and plaque
formation in the hippocampus and cortex [246]. Another study also reported that the
expression of APP and BACE1 were increased in the aging rat brain in response to lead
exposure during the fetal stage [247]. Likewise, lead exposure during infancy increased the
expression of APP, BACE1, transcription factor specific protein 1 (Sp1) and promoted Aβ
deposition in aged monkeys [248]. Synergistic exposure to lead, arsenic and cadmium further
enhanced APP and BACE1 expression, followed by maximum induction of Aβ production
[249]. Developmental lead exposure has also been shown to activate the sterol regulatory
Author Manuscript
Lead poisoning is also accompanied by inflammatory events that lead to neuronal death.
Individuals exposed to lead present with higher serum TNF-α and granulocyte-colony
stimulating factor levels than non-exposed people [255]. Administration of lead to a rat model
results in chronic glial activation, together with inflammatory and neurodegenerative
features [256]. Likewise, it has been demonstrated that lead exposure results in the activation
of microglia and the overproduction of proinflammatory proteins such as inducible nitric
oxide synthase (iNOS), IL-1β and TNF-α [257]. These factors are known to contribute to the
brain neurotoxicity in AD [3]. Increased microglial activation due to lead exposure is also
accompanied by impaired LTP [257]. The mechanism of lead-induced microglial activation
Author Manuscript
Aluminum (Al)
Author Manuscript
Aluminum is not essential for life but is a well-established neurotoxin. Exposure to high
aluminum content in drinking water causes lifelong cerebral impairments, such as loss of
concentration and short-term memory deficits [261]. Mass spectrometry studies have
demonstrated that aluminum crosses the blood-brain barrier and accumulates in a
semipermanent manner [262, 263]. Although no biological process is dependent on aluminum,
it can influence more than 200 biologically-relevant reactions and cause various adverse
effects on the mammalian brain. These include essential brain processes such as axonal
transport, neurotransmitter synthesis, synaptic transmission, phosphorylation or
dephosphorylation of proteins, protein degradation, gene expression, and inflammatory
responses [264]. Aluminum exhibits one oxidation state, Al3+, which has affinity for
negatively charged oxygen-donor ligands. Some of the ligands which form strong bonds
Author Manuscript
with aluminum are inorganic and organic phosphates, carboxylate, and deprotonated
hydroxyl groups, thereby making DNA, RNA and ATP perfect targets, affecting gene
expression, energy metabolism and the action of several kinases and phosphatases [265–267].
Aluminum can also cause the oligomerization of proteins, inducing conformational changes
that can inhibit their degradation by proteases, and thus affect their turnover. For instance,
strong binding of aluminum to phosphorylated amino acids promotes the self-aggregation
and accumulation of highly phosphorylated cytoskeleton proteins, such as neurofilament and
microtubule-associated proteins [268]. These properties make the presence of aluminum in
the brain toxic, causing the apoptotic death of neurons and glial cells. Aluminum affects
LTP, the function of enzymes, including those involved in neurotransmitter synthesis
[269, 270]. It also affects voltage-gated calcium channels and neurotransmitter receptors,
a tau mouse model showing slow progressive tau accumulation, higher tau aggregation,
apoptosis and neurological dysfunction were observed in animals that already had a
pathological process causing tau aggregation, but not in the controls [285], thereby suggesting
an exacerbating effect of aluminum on tau pathology. Aluminum achieves these effects by
enhancing the activity of the tau kinases CDK5 and GSK-3β, inhibiting the
dephosphorylation of tau, and enhancing its aggregation [286–290]. Interestingly, aluminum is
preferentially taken up by glial cells, which induces the production of inflammatory
cytokines, including IL-6 [291, 52, 292]. IL-6 in turn has been reported to induce
phosphorylation of tau by dysregulating the CDK5/p35 cascade [293, 294]. Increased glial
activation and an inflammatory response have been described upon aluminum treatment in
rats [295]; however, whether glial activation due to aluminum exposure plays a role in the
Author Manuscript
Although the effects of aluminum on AD pathology were first attributed to its interaction
with tau, it was later demonstrated that it also affects Aβ by promoting its production,
aggregation, and by inhibiting its degradation [296–299]. Oral administration of aluminum to
AD mice induced an increase in the amount of Aβ, both in its secreted and accumulated
forms, and increased deposition in plaques [300]. In addition, Aβ coupled with aluminum is
more toxic than Aβ itself as it causes membrane disruption and perturbation of neural
calcium homeostasis and mitochondrial respiration [301–303]. Aluminum can also influence
the expression of iron-binding proteins expression with IRE/IRP sequences in their mRNA,
causing an increase in iron concentration [41, 304, 305]. The presence of aluminum in the brain
can therefore modulate the expression, distribution and accumulation of APP and induce the
Author Manuscript
causes oxidative damage [308–310]. Despite its non-redox status, several studies have
suggested that aluminum has strong oxidative activity [311, 312]. The interaction of aluminum
with iron generates labile iron from iron-containing enzymes and proteins, thereby
increasing the intracellular pool of free iron, which in turn leads to the formation of ROS
[313]. Aluminum also decreases the activity of some antioxidant enzymes such as catalase,
superoxide dismutase, and glutathione peroxidase, thus exacerbating the neuronal damage
induced by oxidative stress in neurodegenerative disease such as AD [314, 315].
Aluminum has also been reported to affect neurotransmission. Due to its ability to block Aβ-
mediated formation of calcium permeable ion channel, aluminum can inhibit the increase in
Author Manuscript
calcium levels induced by neurotrophic factors such as BDNF [316–318]. The level of other
neurotransmitters, such as serotonin, dopamine, glutamate and aspartate, have also been
reported to decrease upon aluminum exposure [319, 320]. A lower availability of glutamate
induced by aluminum has been attributed to the induction of glutamine synthetase and
inhibition of glutaminase activity in astrocytes [292]. Moreover, it has been reported that
aluminum affects the cholinergic system, which has been shown to degenerate in AD
pathogenesis [321, 322]. Thus, like the therapeutic approaches used to block the neurotoxic
effects of other metals, aluminum chelation has been studied as a potential therapy for AD
[323].The use of deferoxamine, a chelator of aluminum and iron, as well as silicates, which
Author Manuscript
couples with aluminum and reduces its toxicity, has been shown to attenuate cognitive
decline in AD patients [324, 325]. Despite not being preventative, aluminum chelators could
potentially minimize the neurodegenerative effects of aluminum in patients with known
exposure throughout their lives.
Cadmium (Cd)
Cadmium is a carcinogenic heavy metal that is present in the environment. Unlike many
heavy metals, due to its water-soluble property, cadmium can be transported from soil to
plants and concentrated in the food chain [326]. Although the effect of cadmium on the plant
can be detrimental, some plants, such as tobacco, show cadmium tolerance [327, 328].
Therefore, consumption of tobacco products or inhalation of tobacco smoke increases the
risk of cadmium-related morbidities in the general population [329]. Once taken into the
Author Manuscript
body, cadmium accumulates in the kidney and liver and has an extremely long half-life of
20–40 years [330–332]. Chronic cadmium exposure is associated with hypertension, kidney
dysfunction, bone demineralization, and neurological diseases [333–337]. Cadmium is known
to cross the blood-brain barrier and eventually accumulate in the brain, leading to
neurotoxicity [338]. In the brain, cadmium induces activation of various signaling pathways
involved in inflammation, oxidative stress, and neuronal apoptosis [339, 340, 337].
Recent epidemiological studies reported that blood cadmium levels were significantly
associated with AD-related mortality among older adults [341, 342]. In the AD brain, there is
increasing evidence that cadmium is involved in the aggregation of Aβ plaques [343–345]. In
an in vivo study, APP/PS1 mice administered cadmium in their drinking water exhibited an
increase in the number and size of plaques [343]. Cadmium ions can interact with the Aβ,
subsequently promoting formation of plaques [344]. Furthermore, it has been hypothesized
Author Manuscript
that cadmium treatment downregulates the expression of α-secretase (ADAM10) and neutral
endopeptidase which play essential roles in reducing Aβ levels in the brain [346, 343].
Interestingly, a recent study has reported that the synergistic effect of cadmium, lead, and
arsenic further enhance amyloidogenic processing by increasing APP, BACE1 and PSEN1
expression, suggesting an interactive effect of cadmium with other metals in AD [249]. In
addition to its effects on Aβ, cadmium is also involved in the conformation and self-
aggregation of tau in the AD brain [347, 348]. Cadmium has been reported to bind to the third
repeat (R3) of the microtubule-binding domain of tau. As a result, the R3 domain partially
loses its random coil conformation and gains an α-helix structure which promotes the self-
aggregation of tau. Moreover, cadmium treatment selectively blocks muscarinic M1
receptors which are known to regulate GSK-3β negatively and subsequently increase both
total and phosphorylated tau protein [349, 347, 350]. These data support the notion that
Author Manuscript
been shown to induce astrocyte cytotoxicity by increasing intracellular calcium ions via the
MAPK and PI3K/AKT signaling pathways [353]. These data suggest that the regulation of
Author Manuscript
cadmium-induced Ca2+ homeostasis may be a good strategy for the prevention of related
diseases in the CNS. However, there is still a lack of in vivo studies showing the effect of
cadmium on neuroinflammation in AD mouse models.
Concluding remarks
Altogether, evidence strongly supports that disruption in the homeostasis of essential metals
and the accumulation of non-essential metals disturb the cellular metabolism, antioxidant
defense, and immune responses, leading to the onset and progression of AD. Not
surprisingly, greater emphasis has been given to the interaction of metals and Aβ, in which it
has been shown that biometals interfere with APP function and facilitate aggregation of Aβ
into plaques. The relationship between biometals and tau, however, has only recently
Author Manuscript
emerged, and these studies have shown that changes in the metabolism of metals are
detrimental to tau function, resulting in its loss-of-function and aggregation; however, the
mechanism of how biometals affect tau function remains elusive. Likewise, most findings
regarding to the interaction between brain immunity and biometals have been limited to
evidence of the overproduction of inflammatory mediators (i.e., cytokines) in response to
changes in metal metabolism, with a clear cellular and molecular mechanism still to be
established. Nevertheless, studies using metal chelators have shown promising disease-
modifying properties in vitro and in AD mouse models, emphasizing the potential
therapeutic value of this approach, as well as the important role of metals in AD.
In moving forward in the understanding of the complex links among biometals, AD, and
immunity, studies will need to provide greater mechanistic evidence at cellular and
Author Manuscript
molecular levels for how essential and non-essential metals affect the brain, with respect to
the chronic accumulation of Aβ, tau, and immune mediators. In this context, genetic
approaches, like CRISPR gene editing, targeting specific molecular components of essential
metals metabolism (e.g., point mutations in binding sites, enzymes deletion) in cell culture
models are likely to provide better defined data on how biometals regulate cell function and
signaling, before moving to more complex in vivo models. At cellular level, there is great
need for research on how biometals modulate the functions and interactions of microglia and
astrocytes during homeostasis and disease states, particularly regarding their roles in
immune surveillance and response, and synaptic pruning. Such studies will demonstrate
whether the imbalance of biometals affect the capacity of glial cells to build an immune
response to properly recognize and clear pathological forms of Aβ and tau. Moreover, it is
also not clear whether the potential loss-of-function of glial cells caused by changes in the
Author Manuscript
Acknowledgements
Author Manuscript
We appreciate the editing support from Ms. Rowan Tweedale. This work was funded by the Australian National
Health and Medical Research Council [GNT1128436, GNT1129192, GNT1139469 (RM)] and the National
Institutes of Health [R01ES024331 (MK)].
Abbreviations used
Aβ β-amyloid
AD Alzheimer’s disease
APOE apolipoprotein E
BACE1 β-secretase-1
CD cluster of differentiation
FPN1 ferroportin 1
IL interleukini
LPS lipopolysaccharide
NMDA N-methyl-D-aspartate
PSEN1 presenilin-1
PSEN2 presenilin-2
References
[1]. United Nations, Department of Economic and Social Affairs, Population Division (2017). World
Population Prospects: The 2017 Revision, Key Findings and Advance Tables.
Author Manuscript
[2]. Livingston G, Sommerlad A, Orgeta V, Costafreda SG, Huntley J, Ames D, Ballard C, Banerjee S,
Burns A, Cohen-Mansfield J, Cooper C, Fox N, Gitlin LN, Howard R, Kales HC, Larson EB,
Ritchie K, Rockwood K, Sampson EL, Samus Q, Schneider LS, Selbaek G, Teri L & Mukadam
N (2017). Dementia prevention, intervention, and care. Lancet. 390,2673–734. [PubMed:
28735855]
[3]. Newcombe EA, Camats-Perna J, Silva ML, Valmas N, Huat TJ & Medeiros R (2018).
Inflammation: The link between comorbidities, genetics, and alzheimer’s disease. J
Neuroinflammation. 15,276. [PubMed: 30249283]
[4]. Plummer S, Van Den Heuvel C, Thornton E, Corrigan F & Cappai R (2016). The neuroprotective
properties of the amyloid precursor protein following traumatic brain injury. Aging Dis. 7,163–
79. [PubMed: 27114849]
[5]. Bateman RJ, Xiong C, Benzinger TL, Fagan AM, Goate A, Fox NC, Marcus DS, Cairns NJ, Xie
X, Blazey TM, Holtzman DM, Santacruz A, Buckles V, Oliver A, Moulder K, Aisen PS, Ghetti
B, Klunk WE, Mcdade E, Martins RN, Masters CL, Mayeux R, Ringman JM, Rossor MN,
Schofield PR, Sperling RA, Salloway S & Morris JC (2012). Clinical and biomarker changes in
Author Manuscript
[8]. Cowan CM, Bossing T, Page A, Shepherd D & Mudher A (2010). Soluble hyper-phosphorylated
tau causes microtubule breakdown and functionally compromises normal tau in vivo. Acta
Author Manuscript
[13]. Liu S, Liu Y, Hao W, Wolf L, Kiliaan AJ, Penke B, Rube CE, Walter J, Heneka MT, Hartmann T,
Menger MD & Fassbender K (2012). Tlr2 is a primary receptor for alzheimer’s amyloid beta
peptide to trigger neuroinflammatory activation. J Immunol. 188,1098–107. [PubMed:
22198949]
[14]. Zhao Y, Wu X, Li X, Jiang LL, Gui X, Liu Y, Sun Y, Zhu B, Pina-Crespo JC, Zhang M, Zhang N,
Chen X, Bu G, An Z, Huang TY & Xu H (2018). Trem2 is a receptor for beta-amyloid that
mediates microglial function. Neuron. 97,1023–31.e7. [PubMed: 29518356]
[15]. Hellstrom-Lindahl E, Ravid R & Nordberg A (2008). Age-dependent decline of neprilysin in
alzheimer’s disease and normal brain: Inverse correlation with a beta levels. Neurobiol Aging.
29,210–21. [PubMed: 17098332]
[16]. Zhao Z, Xiang Z, Haroutunian V, Buxbaum JD, Stetka B & Pasinetti GM (2007). Insulin
degrading enzyme activity selectively decreases in the hippocampal formation of cases at high
risk to develop alzheimer’s disease. Neurobiol Aging. 28,824–30. [PubMed: 16769157]
[17]. Kitazawa M, Oddo S, Yamasaki TR, Green KN & Laferla FM (2005). Lipopolysaccharide-
induced inflammation exacerbates tau pathology by a cyclin-dependent kinase 5-mediated
Author Manuscript
[22]. Castillo E, Leon J, Mazzei G, Abolhassani N, Haruyama N, Saito T, Saido T, Hokama M, Iwaki
T, Ohara T, Ninomiya T, Kiyohara Y, Sakumi K, Laferla FM & Nakabeppu Y (2017).
Comparative profiling of cortical gene expression in alzheimer’s disease patients and mouse
models demonstrates a link between amyloidosis and neuroinflammation. Sci Rep. 7,17762.
[PubMed: 29259249]
[23]. Hong S, Beja-Glasser VF, Nfonoyim BM, Frouin A, Li S, Ramakrishnan S, Merry KM, Shi Q,
Rosenthal A, Barres BA, Lemere CA, Selkoe DJ & Stevens B (2016). Complement and microglia
mediate early synapse loss in alzheimer mouse models. Science. 352,712–6. [PubMed:
27033548]
[24]. Programme UNE, Nations U & Publications UN Unep year book 2014: Emerging issues in our
global environment: UNITED NATIONS PUBN; 2015.
Author Manuscript
[25]. Prüss-Üstün A & Neira M Preventing disease through healthy environments: A global assessment
of the burden of disease from environmental risks: World Health Organization; 2016.
[26]. Que EL, Domaille DW & Chang CJ (2008). Metals in neurobiology: Probing their chemistry and
biology with molecular imaging. Chem Rev. 108,1517–49. [PubMed: 18426241]
[27]. Torti SV, Manz DH, Paul BT, Blanchette-Farra N & Torti FM (2018). Iron and cancer. Annu Rev
Nutr. 38,97–125. [PubMed: 30130469]
[28]. Hower V, Mendes P, Torti FM, Laubenbacher R, Akman S, Shulaev V & Torti SV (2009). A
general map of iron metabolism and tissue-specific subnetworks. Mol Biosyst. 5,422–43.
[PubMed: 19381358]
[29]. Liu JL, Fan YG, Yang ZS, Wang ZY & Guo C (2018). Iron and alzheimer’s disease: From
pathogenesis to therapeutic implications. Front Neurosci. 12,632. [PubMed: 30250423]
[30]. Hentze MW, Muckenthaler MU, Galy B & Camaschella C (2010). Two to tango: Regulation of
mammalian iron metabolism. Cell. 142,24–38. [PubMed: 20603012]
[31]. Camaschella C (2015). Iron-deficiency anemia. N Engl J Med. 373,485–6.
Author Manuscript
[32]. Mesquita SD, Ferreira AC, Sousa JC, Santos NC, Correia-Neves M, Sousa N, Palha JA &
Marques F (2012). Modulation of iron metabolism in aging and in alzheimer’s disease:
Relevance of the choroid plexus. Front Cell Neurosci. 6,25. [PubMed: 22661928]
[33]. Pirpamer L, Hofer E, Gesierich B, De Guio F, Freudenberger P, Seiler S, Duering M, Jouvent E,
Duchesnay E, Dichgans M, Ropele S & Schmidt R (2016). Determinants of iron accumulation in
the normal aging brain. Neurobiol Aging. 43,149–55. [PubMed: 27255824]
[34]. Xu J, Jia Z, Knutson MD & Leeuwenburgh C (2012). Impaired iron status in aging research. Int J
Mol Sci. 13,2368–86. [PubMed: 22408459]
[35]. Smith MA, Zhu X, Tabaton M, Liu G, Mckeel DW Jr., Cohen ML, Wang X, Siedlak SL, Dwyer
BE, Hayashi T, Nakamura M, Nunomura A & Perry G (2010). Increased iron and free radical
generation in preclinical alzheimer disease and mild cognitive impairment. J Alzheimers Dis.
19,363–72. [PubMed: 20061651]
[36]. El Tannir El Tayara N, Delatour B, Le Cudennec C, Guegan M, Volk A & Dhenain M (2006).
Age-related evolution of amyloid burden, iron load, and mr relaxation times in a transgenic
Author Manuscript
Selective translational control of the alzheimer amyloid precursor protein transcript by iron
regulatory protein-1. J Biol Chem. 285,31217–32. [PubMed: 20558735]
[42]. Rogers JT, Randall JD, Cahill CM, Eder PS, Huang X, Gunshin H, Leiter L, Mcphee J, Sarang
SS, Utsuki T, Greig NH, Lahiri DK, Tanzi RE, Bush AI, Giordano T & Gullans SR (2002). An
iron-responsive element type ii in the 5’-untranslated region of the alzheimer’s amyloid precursor
protein transcript. J Biol Chem. 277,45518–28. [PubMed: 12198135]
[43]. Duce JA, Tsatsanis A, Cater MA, James SA, Robb E, Wikhe K, Leong SL, Perez K, Johanssen T,
Greenough MA, Cho HH, Galatis D, Moir RD, Masters CL, Mclean C, Tanzi RE, Cappai R,
Barnham KJ, Ciccotosto GD, Rogers JT & Bush AI (2010). Iron-export ferroxidase activity of
[44]. Sayre LM, Perry G, Harris PL, Liu Y, Schubert KA & Smith MA (2000). In situ oxidative
catalysis by neurofibrillary tangles and senile plaques in alzheimer’s disease: A central role for
bound transition metals. J Neurochem. 74,270–9. [PubMed: 10617129]
[45]. Smith MA, Harris PL, Sayre LM & Perry G (1997). Iron accumulation in alzheimer disease is a
source of redox-generated free radicals. Proc Natl Acad Sci U S A. 94,9866–8. [PubMed:
9275217]
[46]. Good PF, Perl DP, Bierer LM & Schmeidler J (1992). Selective accumulation of aluminum and
iron in the neurofibrillary tangles of alzheimer’s disease: A laser microprobe (lamma) study. Ann
Neurol. 31,286–92. [PubMed: 1637136]
[47]. Perez M, Valpuesta JM, De Garcini EM, Quintana C, Arrasate M, Lopez Carrascosa JL, Rabano
A, Garcia De Yebenes J & Avila J (1998). Ferritin is associated with the aberrant tau filaments
present in progressive supranuclear palsy. Am J Pathol. 152,1531–9. [PubMed: 9626057]
[48]. Connor JR, Menzies SL, St Martin SM & Mufson EJ (1992). A histochemical study of iron,
transferrin, and ferritin in alzheimer’s diseased brains. J Neurosci Res. 31,75–83. [PubMed:
Author Manuscript
1613823]
[49]. Grundke-Iqbal I, Fleming J, Tung YC, Lassmann H, Iqbal K & Joshi JG (1990). Ferritin is a
component of the neuritic (senile) plaque in alzheimer dementia. Acta Neuropathol. 81,105–10.
[PubMed: 2082650]
[50]. Jellinger K, Paulus W, Grundke-Iqbal I, Riederer P & Youdim MB (1990). Brain iron and ferritin
in parkinson’s and alzheimer’s diseases. J Neural Transm Park Dis Dement Sect. 2,327–40.
[PubMed: 2078310]
[51]. Van Duijn S, Bulk M, Van Duinen SG, Nabuurs RJA, Van Buchem MA, Van Der Weerd L &
Natte R (2017). Cortical iron reflects severity of alzheimer’s disease. J Alzheimers Dis. 60,1533–
45. [PubMed: 29081415]
[52]. Oshiro S, Kawahara M, Kuroda Y, Zhang C, Cai Y, Kitajima S & Shirao M (2000). Glial cells
contribute more to iron and aluminum accumulation but are more resistant to oxidative stress
than neuronal cells. Biochim Biophys Acta. 1502,405–14. [PubMed: 11068183]
[53]. Lopes KO, Sparks DL & Streit WJ (2008). Microglial dystrophy in the aged and alzheimer’s
disease brain is associated with ferritin immunoreactivity. Glia. 56,1048–60. [PubMed:
Author Manuscript
18442088]
[54]. Kawamata T, Tooyama I, Yamada T, Walker DG & Mcgeer PL (1993). Lactotransferrin
immunocytochemistry in alzheimer and normal human brain. Am J Pathol. 142,1574–85.
[PubMed: 8494052]
[55]. Leveugle B, Spik G, Perl DP, Bouras C, Fillit HM & Hof PR (1994). The iron-binding protein
lactotransferrin is present in pathologic lesions in a variety of neurodegenerative disorders: A
comparative immunohistochemical analysis. Brain Res. 650,20–31. [PubMed: 7953673]
[56]. Jefferies WA, Food MR, Gabathuler R, Rothenberger S, Yamada T, Yasuhara O & Mcgeer PL
(1996). Reactive microglia specifically associated with amyloid plaques in alzheimer’s disease
brain tissue express melanotransferrin. Brain Res. 712,122–6. [PubMed: 8705294]
[57]. Smith MA, Wehr K, Harris PL, Siedlak SL, Connor JR & Perry G (1998). Abnormal localization
of iron regulatory protein in alzheimer’s disease. Brain Res. 788,232–6. [PubMed: 9555030]
[58]. Wang P, Wu Q, Wu W, Li H, Guo Y, Yu P, Gao G, Shi Z, Zhao B & Chang YZ (2017).
Mitochondrial ferritin deletion exacerbates beta-amyloid-induced neurotoxicity in mice. Oxid
Author Manuscript
[62]. Schubert D & Chevion M (1995). The role of iron in beta amyloid toxicity. Biochem Biophys
Res Commun. 216,702–7. [PubMed: 7488167]
Author Manuscript
[63]. Jiang D, Li X, Williams R, Patel S, Men L, Wang Y & Zhou F (2009). Ternary complexes of iron,
amyloid-beta, and nitrilotriacetic acid: Binding affinities, redox properties, and relevance to iron-
induced oxidative stress in alzheimer’s disease. Biochemistry. 48,7939–47. [PubMed: 19601593]
[64]. Xian-Hui D, Wei-Juan G, Tie-Mei S, Hong-Lin X, Jiang-Tao B, Jing-Yi Z & Xi-Qing C (2015).
Age-related changes of brain iron load changes in the frontal cortex in appswe/ps1deltae9
transgenic mouse model of alzheimer’s disease. J Trace Elem Med Biol. 30,118–23. [PubMed:
25575693]
[65]. Skjorringe T, Burkhart A, Johnsen KB & Moos T (2015). Divalent metal transporter 1 (dmt1) in
the brain: Implications for a role in iron transport at the blood-brain barrier, and neuronal and
glial pathology. Front Mol Neurosci. 8,19. [PubMed: 26106291]
[66]. Ward DM & Kaplan J (2012). Ferroportin-mediated iron transport: Expression and regulation.
Biochim Biophys Acta. 1823,1426–33. [PubMed: 22440327]
[67]. Mita H, Yui Y, Taniguchi N, Yasueda H & Shida T (1985). Increased activity of 5-lipoxygenase
in polymorphonuclear leukocytes from asthmatic patients. Life Sci. 37,907–14. [PubMed:
Author Manuscript
2993772]
[68]. Tian J, Zheng W, Li XL, Cui YH & Wang ZY (2018). Lower expression of ndfip1 is associated
with alzheimer disease pathogenesis through decreasing dmt1 degradation and increasing iron
influx. Front Aging Neurosci. 10,165. [PubMed: 29937728]
[69]. Zheng W, Xin N, Chi ZH, Zhao BL, Zhang J, Li JY & Wang ZY (2009). Divalent metal
transporter 1 is involved in amyloid precursor protein processing and abeta generation. Faseb j.
23,4207–17. [PubMed: 19679638]
[70]. Xie L, Zheng W, Xin N, Xie JW, Wang T & Wang ZY (2012). Ebselen inhibits iron-induced tau
phosphorylation by attenuating dmt1 up-regulation and cellular iron uptake. Neurochem Int.
61,334–40. [PubMed: 22634399]
[71]. Becerril-Ortega J, Bordji K, Freret T, Rush T & Buisson A (2014). Iron overload accelerates
neuronal amyloid-beta production and cognitive impairment in transgenic mice model of
alzheimer’s disease. Neurobiol Aging. 35,2288–301. [PubMed: 24863668]
[72]. Peters DG, Pollack AN, Cheng KC, Sun D, Saido T, Haaf MP, Yang QX, Connor JR &
Meadowcroft MD (2018). Dietary lipophilic iron alters amyloidogenesis and microglial
Author Manuscript
MT (2013). Inflammation alters the expression of dmt1, fpn1 and hepcidin, and it causes iron
accumulation in central nervous system cells. J Neurochem. 126,541–9. [PubMed: 23506423]
[78]. Kroner A, Greenhalgh AD, Zarruk JG, Passos Dos Santos R, Gaestel M & David S (2014). Tnf
and increased intracellular iron alter macrophage polarization to a detrimental m1 phenotype in
the injured spinal cord. Neuron. 83,1098–116. [PubMed: 25132469]
[79]. Kim BE, Nevitt T & Thiele DJ (2008). Mechanisms for copper acquisition, distribution and
regulation. Nat Chem Biol. 4,176–85. [PubMed: 18277979]
[80]. Madsen E & Gitlin JD (2007). Copper and iron disorders of the brain. Annu Rev Neurosci.
30,317–37. [PubMed: 17367269]
[81]. Kaler SG (2011). Atp7a-related copper transport diseases-emerging concepts and future trends.
Nat Rev Neurol. 7,15–29. [PubMed: 21221114]
Author Manuscript
[82]. Gaier ED, Eipper BA & Mains RE (2013). Copper signaling in the mammalian nervous system:
Synaptic effects. J Neurosci Res. 91,2–19. [PubMed: 23115049]
[83]. Lutsenko S, Bhattacharjee A & Hubbard AL (2010). Copper handling machinery of the brain.
Metallomics. 2,596–608. [PubMed: 21072351]
[84]. Opazo CM, Greenough MA & Bush AI (2014). Copper: From neurotransmission to
neuroproteostasis. Front Aging Neurosci. 6,143. [PubMed: 25071552]
[85]. Bandmann O, Weiss KH & Kaler SG (2015). Wilson’s disease and other neurological copper
disorders. Lancet Neurol. 14,103–13. [PubMed: 25496901]
[86]. Brown DR, Qin K, Herms JW, Madlung A, Manson J, Strome R, Fraser PE, Kruck T, Von Bohlen
A, Schulz-Schaeffer W, Giese A, Westaway D & Kretzschmar H (1997). The cellular prion
protein binds copper in vivo. Nature. 390,684–7. [PubMed: 9414160]
[87]. Rose F, Hodak M & Bernholc J (2011). Mechanism of copper(ii)-induced misfolding of
parkinson’s disease protein. Sci Rep. 1,11. [PubMed: 22355530]
[88]. Siggs OM, Cruite JT, Du X, Rutschmann S, Masliah E, Beutler B & Oldstone MB (2012).
Author Manuscript
Disruption of copper homeostasis due to a mutation of atp7a delays the onset of prion disease.
Proc Natl Acad Sci U S A. 109,13733–8. [PubMed: 22869751]
[89]. Valentine JS & Hart PJ (2003). Misfolded cuznsod and amyotrophic lateral sclerosis. Proc Natl
Acad Sci U S A. 100,3617–22. [PubMed: 12655070]
[90]. Xiao G, Fan Q, Wang X & Zhou B (2013). Huntington disease arises from a combinatory toxicity
of polyglutamine and copper binding. Proc Natl Acad Sci U S A. 110,14995–5000. [PubMed:
23980182]
[91]. Squitti R, Ghidoni R, Siotto M, Ventriglia M, Benussi L, Paterlini A, Magri M, Binetti G,
Cassetta E, Caprara D, Vernieri F, Rossini PM & Pasqualetti P (2014). Value of serum
nonceruloplasmin copper for prediction of mild cognitive impairment conversion to alzheimer
disease. Ann Neurol. 75,574–80. [PubMed: 24623259]
[92]. Ventriglia M, Bucossi S, Panetta V & Squitti R (2012). Copper in alzheimer’s disease: A meta-
analysis of serum, plasma, and cerebrospinal fluid studies. J Alzheimers Dis. 30,981–4.
[PubMed: 22475798]
Author Manuscript
[93]. Wang ZX, Tan L, Wang HF, Ma J, Liu J, Tan MS, Sun JH, Zhu XC, Jiang T & Yu JT (2015).
Serum iron, zinc, and copper levels in patients with alzheimer’s disease: A replication study and
meta-analyses. J Alzheimers Dis. 47,565–81. [PubMed: 26401693]
[94]. Lovell MA, Robertson JD, Teesdale WJ, Campbell JL & Markesbery WR (1998). Copper, iron
and zinc in alzheimer’s disease senile plaques. J Neurol Sci. 158,47–52. [PubMed: 9667777]
[95]. Miller LM, Wang Q, Telivala TP, Smith RJ, Lanzirotti A & Miklossy J (2006). Synchrotron-
based infrared and x-ray imaging shows focalized accumulation of cu and zn co-localized with
beta-amyloid deposits in alzheimer’s disease. J Struct Biol. 155,30–7. [PubMed: 16325427]
[96]. Rembach A, Hare DJ, Lind M, Fowler CJ, Cherny RA, Mclean C, Bush AI, Masters CL &
Roberts BR (2013). Decreased copper in alzheimer’s disease brain is predominantly in the
soluble extractable fraction. Int J Alzheimers Dis. 2013,623241. [PubMed: 24228186]
[97]. James SA, Volitakis I, Adlard PA, Duce JA, Masters CL, Cherny RA & Bush AI (2012). Elevated
labile cu is associated with oxidative pathology in alzheimer disease. Free Radic Biol Med.
52,298–302. [PubMed: 22080049]
[98]. Multhaup G, Schlicksupp A, Hesse L, Beher D, Ruppert T, Masters CL & Beyreuther K (1996).
Author Manuscript
The amyloid precursor protein of alzheimer’s disease in the reduction of copper(ii) to copper(i).
Science. 271,1406–9. [PubMed: 8596911]
[99]. Maynard CJ, Cappai R, Volitakis I, Cherny RA, White AR, Beyreuther K, Masters CL, Bush AI
& Li QX (2002). Overexpression of alzheimer’s disease amyloid-beta opposes the age-dependent
elevations of brain copper and iron. J Biol Chem. 277,44670–6. [PubMed: 12215434]
[100]. White AR, Reyes R, Mercer JF, Camakaris J, Zheng H, Bush AI, Multhaup G, Beyreuther K,
Masters CL & Cappai R (1999). Copper levels are increased in the cerebral cortex and liver of
app and aplp2 knockout mice. Brain Res. 842,439–44. [PubMed: 10526140]
[101]. Acevedo KM, Hung YH, Dalziel AH, Li QX, Laughton K, Wikhe K, Rembach A, Roberts B,
Masters CL, Bush AI & Camakaris J (2011). Copper promotes the trafficking of the amyloid
Author Manuscript
[117]. Zheng Z, White C, Lee J, Peterson TS, Bush AI, Sun GY, Weisman GA & Petris MJ (2010).
Altered microglial copper homeostasis in a mouse model of alzheimer’s disease. J Neurochem.
Author Manuscript
[122]. Cherny RA, Atwood CS, Xilinas ME, Gray DN, Jones WD, Mclean CA, Barnham KJ, Volitakis
I, Fraser FW, Kim Y, Huang X, Goldstein LE, Moir RD, Lim JT, Beyreuther K, Zheng H, Tanzi
RE, Masters CL & Bush AI (2001). Treatment with a copper-zinc chelator markedly and rapidly
inhibits beta-amyloid accumulation in alzheimer’s disease transgenic mice. Neuron. 30,665–76.
[PubMed: 11430801]
[123]. Cherny RA, Legg JT, Mclean CA, Fairlie DP, Huang X, Atwood CS, Beyreuther K, Tanzi RE,
Masters CL & Bush AI (1999). Aqueous dissolution of alzheimer’s disease abeta amyloid
deposits by biometal depletion. J Biol Chem. 274,23223–8. [PubMed: 10438495]
[124]. Grossi C, Francese S, Casini A, Rosi MC, Luccarini I, Fiorentini A, Gabbiani C, Messori L,
Moneti G & Casamenti F (2009). Clioquinol decreases amyloid-beta burden and reduces working
memory impairment in a transgenic mouse model of alzheimer’s disease. J Alzheimers Dis.
17,423–40. [PubMed: 19363260]
[125]. Matlack KE, Tardiff DF, Narayan P, Hamamichi S, Caldwell KA, Caldwell GA & Lindquist S
(2014). Clioquinol promotes the degradation of metal-dependent amyloid-beta (abeta) oligomers
to restore endocytosis and ameliorate abeta toxicity. Proc Natl Acad Sci USA. 111,4013–8.
Author Manuscript
[PubMed: 24591589]
[126]. Segal-Gavish H, Danino O, Barhum Y, Ben-Zur T, Shai E, Varon D, Offen D & Fischer B
(2017). A multifunctional biocompatible drug candidate is highly effective in delaying
pathological signs of alzheimer’s disease in 5xfad mice. J Alzheimers Dis. 58,389–400.
[PubMed: 28453480]
[127]. Donnelly PS, Caragounis A, Du T, Laughton KM, Volitakis I, Cherny RA, Sharples RA, Hill
AF, Li QX, Masters CL, Barnham KJ & White AR (2008). Selective intracellular release of
copper and zinc ions from bis(thiosemicarbazonato) complexes reduces levels of alzheimer
disease amyloid-beta peptide. J Biol Chem. 283,4568–77. [PubMed: 18086681]
[128]. Gerber H, Wu F, Dimitrov M, Garcia Osuna GM & Fraering PC (2017). Zinc and copper
differentially modulate amyloid precursor protein processing by gamma-secretase and amyloid-
beta peptide production. J Biol Chem. 292,3751–67. [PubMed: 28096459]
[129]. Crouch PJ, Hung LW, Adlard PA, Cortes M, Lal V, Filiz G, Perez KA, Nurjono M, Caragounis
A, Du T, Laughton K, Volitakis I, Bush AI, Li QX, Masters CL, Cappai R, Cherny RA, Donnelly
PS, White AR & Barnham KJ (2009). Increasing cu bioavailability inhibits abeta oligomers and
Author Manuscript
tau phosphorylation. Proc Natl Acad Sci USA. 106,381–6. [PubMed: 19122148]
[130]. Choo XY, Liddell JR, Huuskonen MT, Grubman A, Moujalled D, Roberts J, Kysenius K, Patten
L, Quek H, Oikari LE, Duncan C, James SA, Mcinnes LE, Hayne DJ, Donnelly PS, Pollari E,
Vahatalo S, Lejavova K, Kettunen MI, Malm T, Koistinaho J, White AR & Kanninen KM (2018).
Cu(ii)(atsm) attenuates neuroinflammation. Front Neurosci. 12,668. [PubMed: 30319344]
[131]. Sparks DL, Friedland R, Petanceska S, Schreurs BG, Shi J, Perry G, Smith MA, Sharma A,
Derosa S, Ziolkowski C & Stankovic G (2006). Trace copper levels in the drinking water, but not
zinc or aluminum influence cns alzheimer-like pathology. J Nutr Health Aging. 10,247–54.
[PubMed: 16886094]
[132]. Sparks DL & Schreurs BG (2003). Trace amounts of copper in water induce beta-amyloid
plaques and learning deficits in a rabbit model of alzheimer’s disease. Proc Natl Acad Sci U S A.
Author Manuscript
[137]. Takeda A (2001). Zinc homeostasis and functions of zinc in the brain. Biometals. 14,343–51.
[PubMed: 11831464]
[138]. Mocchegiani E, Giacconi R, Cipriano C, Muzzioli M, Fattoretti P, Bertoni-Freddari C, Isani G,
Zambenedetti P & Zatta P (2001). Zinc-bound metallothioneins as potential biological markers of
ageing. Brain Res Bull. 55,147–53. [PubMed: 11470310]
[139]. Liuzzi JP & Cousins RJ (2004). Mammalian zinc transporters. Annu Rev Nutr. 24,151–72.
[PubMed: 15189117]
[140]. Kambe T, Tsuji T, Hashimoto A & Itsumura N (2015). The physiological, biochemical, and
molecular roles of zinc transporters in zinc homeostasis and metabolism. Physiol Rev. 95,749–
84. [PubMed: 26084690]
[141]. Frederickson CJ, Suh SW, Silva D, Frederickson CJ & Thompson RB (2000). Importance of
zinc in the central nervous system: The zinc-containing neuron. J Nutr. 130,1471s–83s. [PubMed:
10801962]
[142]. Paoletti P, Vergnano AM, Barbour B & Casado M (2009). Zinc at glutamatergic synapses.
Neuroscience. 158,126–36. [PubMed: 18353558]
Author Manuscript
[143]. Sensi SL, Paoletti P, Koh JY, Aizenman E, Bush AI & Hershfinkel M (2011). The
neurophysiology and pathology of brain zinc. J Neurosci. 31,16076–85. [PubMed: 22072659]
[144]. Smart TG, Hosie AM & Miller PS (2004). Zn2+ ions: Modulators of excitatory and inhibitory
synaptic activity. Neuroscientist. 10,432–42. [PubMed: 15359010]
[145]. Frederickson CJ & Danscher G (1990). Zinc-containing neurons in hippocampus and related cns
structures. Prog Brain Res. 83,71–84. [PubMed: 2203108]
[146]. Doraiswamy PM & Finefrock AE (2004). Metals in our minds: Therapeutic implications for
neurodegenerative disorders. Lancet Neurol. 3,431–4. [PubMed: 15207800]
[147]. Mocchegiani E, Bertoni-Freddari C, Marcellini F & Malavolta M (2005). Brain, aging and
neurodegeneration: Role of zinc ion availability. Prog Neurobiol. 75,367–90. [PubMed:
15927345]
[148]. Cuajungco MP & Lees GJ (1997). Zinc metabolism in the brain: Relevance to human
neurodegenerative disorders. Neurobiol Dis. 4,137–69. [PubMed: 9361293]
[149]. Frederickson CJ, Koh JY & Bush AI (2005). The neurobiology of zinc in health and disease.
Author Manuscript
[153]. Bush AI & Tanzi RE (2002). The galvanization of beta-amyloid in alzheimer’s disease. Proc
Natl Acad Sci U S A. 99,7317–9. [PubMed: 12032279]
Author Manuscript
[154]. Deshpande A, Kawai H, Metherate R, Glabe CG & Busciglio J (2009). A role for synaptic zinc
in activity-dependent abeta oligomer formation and accumulation at excitatory synapses. J
Neurosci. 29,4004–15. [PubMed: 19339596]
[155]. Bush AI, Multhaup G, Moir RD, Williamson TG, Small DH, Rumble B, Pollwein P, Beyreuther
K & Masters CL (1993). A novel zinc(ii) binding site modulates the function of the beta a4
amyloid protein precursor of alzheimer’s disease. J Biol Chem. 268,16109–12. [PubMed:
8344894]
[156]. Maynard CJ, Bush AI, Masters CL, Cappai R & Li QX (2005). Metals and amyloid-beta in
alzheimer’s disease. Int J Exp Pathol. 86,147–59. [PubMed: 15910549]
[157]. Suh SW, Jensen KB, Jensen MS, Silva DS, Kesslak PJ, Danscher G & Frederickson CJ (2000).
Histochemically-reactive zinc in amyloid plaques, angiopathy, and degenerating neurons of
alzheimer’s diseased brains. Brain Res. 852,274–8. [PubMed: 10678753]
[158]. Lee JY, Cole TB, Palmiter RD, Suh SW & Koh JY (2002). Contribution by synaptic zinc to the
gender-disparate plaque formation in human swedish mutant app transgenic mice. Proc Natl
Author Manuscript
[PubMed: 25549871]
[163]. Frazzini V, Granzotto A, Bomba M, Massetti N, Castelli V, D’aurora M, Punzi M, Iorio M,
Mosca A, Delli Pizzi S, Gatta V, Cimini A & Sensi SL (2018). The pharmacological perturbation
of brain zinc impairs bdnf-related signaling and the cognitive performances of young mice. Sci
Rep. 8,9768. [PubMed: 29950603]
[164]. Wall MJ (2005). A role for zinc in cerebellar synaptic transmission? Cerebellum. 4,224–9.
[PubMed: 16321877]
[165]. Railey AM, Groeber CM & Flinn JM (2011). The effect of metals on spatial memory in a
transgenic mouse model of alzheimer’s disease. J Alzheimers Dis. 24,375–81. [PubMed:
21239856]
[166]. Stoltenberg M, Bush AI, Bach G, Smidt K, Larsen A, Rungby J, Lund S, Doering P & Danscher
G (2007). Amyloid plaques arise from zinc-enriched cortical layers in app/ps1 transgenic mice
and are paradoxically enlarged with dietary zinc deficiency. Neuroscience. 150,357–69.
[PubMed: 17949919]
Author Manuscript
[167]. Wang CY, Wang T, Zheng W, Zhao BL, Danscher G, Chen YH & Wang ZY (2010). Zinc
overload enhances app cleavage and abeta deposition in the alzheimer mouse brain. PLoS One.
5,e15349. [PubMed: 21179415]
[168]. Harris CJ, Voss K, Murchison C, Ralle M, Frahler K, Carter R, Rhoads A, Lind B, Robinson E
& Quinn JF (2014). Oral zinc reduces amyloid burden in tg2576 mice. J Alzheimers Dis. 41,179–
92. [PubMed: 24595193]
[169]. Capasso M, Jeng JM, Malavolta M, Mocchegiani E & Sensi SL (2005). Zinc dyshomeostasis: A
key modulator of neuronal injury. J Alzheimers Dis. 8,93–108; discussion 209–15. [PubMed:
16308478]
[170]. Crouch PJ, Savva MS, Hung LW, Donnelly PS, Mot AI, Parker SJ, Greenough MA, Volitakis I,
Adlard PA, Cherny RA, Masters CL, Bush AI, Barnham KJ & White AR (2011). The alzheimer’s
Author Manuscript
therapeutic pbt2 promotes amyloid-beta degradation and gsk3 phosphorylation via a metal
chaperone activity. J Neurochem. 119,220–30. [PubMed: 21797865]
[171]. Andrasi E, Farkas E, Gawlik D, Rosick U & Bratter P (2000). Brain iron and zinc contents of
german patients with alzheimer disease. J Alzheimers Dis. 2,17–26. [PubMed: 12214107]
[172]. Corrigan FM, Reynolds GP & Ward NI (1993). Hippocampal tin, aluminum and zinc in
alzheimer’s disease. Biometals. 6,149–54. [PubMed: 8400761]
[173]. Danscher G, Jensen KB, Frederickson CJ, Kemp K, Andreasen A, Juhl S, Stoltenberg M &
Ravid R (1997). Increased amount of zinc in the hippocampus and amygdala of alzheimer’s
diseased brains: A proton-induced x-ray emission spectroscopic analysis of cryostat sections
from autopsy material. J Neurosci Methods. 76,53–9. [PubMed: 9334939]
[174]. Panayi AE, Spyrou NM, Iversen BS, White MA & Part P (2002). Determination of cadmium
and zinc in alzheimer’s brain tissue using inductively coupled plasma mass spectrometry. J
Neurol Sci. 195,1–10. [PubMed: 11867068]
[175]. Rulon LL, Robertson JD, Lovell MA, Deibel MA, Ehmann WD & Markesber WR (2000).
Author Manuscript
Serum zinc levels and alzheimer’s disease. Biol Trace Elem Res. 75,79–85. [PubMed: 11051598]
[176]. An WL, Bjorkdahl C, Liu R, Cowburn RF, Winblad B & Pei JJ (2005). Mechanism of zinc-
induced phosphorylation of p70 s6 kinase and glycogen synthase kinase 3beta in sh-sy5y
neuroblastoma cells. J Neurochem. 92,1104–15. [PubMed: 15715661]
[177]. Mo ZY, Zhu YZ, Zhu HL, Fan JB, Chen J & Liang Y (2009). Low micromolar zinc accelerates
the fibrillization of human tau via bridging of cys-291 and cys-322. J Biol Chem. 284,34648–57.
[PubMed: 19826005]
[178]. Pei JJ, An WL, Zhou XW, Nishimura T, Norberg J, Benedikz E, Gotz J & Winblad B (2006).
P70 s6 kinase mediates tau phosphorylation and synthesis. FEBS Lett. 580,107–14. [PubMed:
16364302]
[179]. Sun XY, Wei YP, Xiong Y, Wang XC, Xie AJ, Wang XL, Yang Y, Wang Q, Lu YM, Liu R &
Wang JZ (2012). Synaptic released zinc promotes tau hyperphosphorylation by inhibition of
protein phosphatase 2a (pp2a). J Biol Chem. 287,11174–82. [PubMed: 22334661]
[180]. Xiong Y, Jing XP, Zhou XW, Wang XL, Yang Y, Sun XY, Qiu M, Cao FY, Lu YM, Liu R &
Wang JZ (2013). Zinc induces protein phosphatase 2a inactivation and tau hyperphosphorylation
Author Manuscript
through src dependent pp2a (tyrosine 307) phosphorylation. Neurobiol Aging. 34,745–56.
[PubMed: 22892311]
[181]. Huang Y, Wu Z, Cao Y, Lang M, Lu B & Zhou B (2014). Zinc binding directly regulates tau
toxicity independent of tau hyperphosphorylation. Cell Rep. 8,831–42. [PubMed: 25066125]
[182]. Craven KM, Kochen WR, Hernandez CM & Flinn JM (2018). Zinc exacerbates tau pathology in
a tau mouse model. J Alzheimers Dis. 64,617–30. [PubMed: 29914030]
[183]. Sedjahtera A, Gunawan L, Bray L, Hung LW, Parsons J, Okamura N, Villemagne VL, Yanai K,
Liu XM, Chan J, Bush AI, Finkelstein DI, Barnham KJ, Cherny RA & Adlard PA (2018).
Targeting metals rescues the phenotype in an animal model of tauopathy. Metallomics. 10,1339–
47. [PubMed: 30168573]
[184]. Aizenman E, Stout AK, Hartnett KA, Dineley KE, Mclaughlin B & Reynolds IJ (2000).
Induction of neuronal apoptosis by thiol oxidation: Putative role of intracellular zinc release. J
Neurochem. 75,1878–88. [PubMed: 11032877]
[185]. Bossy-Wetzel E, Talantova MV, Lee WD, Scholzke MN, Harrop A, Mathews E, Gotz T, Han J,
Author Manuscript
Ellisman MH, Perkins GA & Lipton SA (2004). Crosstalk between nitric oxide and zinc
pathways to neuronal cell death involving mitochondrial dysfunction and p38-activated k+
channels. Neuron. 41,351–65. [PubMed: 14766175]
[186]. Jomova K, Vondrakova D, Lawson M & Valko M (2010). Metals, oxidative stress and
neurodegenerative disorders. Mol Cell Biochem. 345,91–104. [PubMed: 20730621]
[187]. Sensi SL, Rapposelli IG, Frazzini V & Mascetra N (2008). Altered oxidant-mediated
intraneuronal zinc mobilization in a triple transgenic mouse model of alzheimer’s disease. Exp
Gerontol. 43,488–92. [PubMed: 18068923]
[188]. Sensi SL, Ton-That D, Sullivan PG, Jonas EA, Gee KR, Kaczmarek LK & Weiss JH (2003).
Modulation of mitochondrial function by endogenous zn2+ pools. Proc Natl Acad Sci U S A.
Author Manuscript
[199]. Brewer GJ (2012). Copper excess, zinc deficiency, and cognition loss in alzheimer’s disease.
Biofactors. 38,107–13. [PubMed: 22438177]
[200]. Mocchegiani E, Romeo J, Malavolta M, Costarelli L, Giacconi R, Diaz LE & Marcos A (2013).
Zinc: Dietary intake and impact of supplementation on immune function in elderly. Age (Dordr).
35,839–60. [PubMed: 22222917]
[201]. Lannfelt L, Blennow K, Zetterberg H, Batsman S, Ames D, Harrison J, Masters CL, Targum S,
Bush AI, Murdoch R, Wilson J & Ritchie CW (2008). Safety, efficacy, and biomarker findings of
pbt2 in targeting abeta as a modifying therapy for alzheimer’s disease: A phase iia, double-blind,
randomised, placebo-controlled trial. Lancet Neurol. 7,779–86. [PubMed: 18672400]
[202]. Parikh SR, Hurwitz RA, Hubbard JE, Brown JW, King H & Girod DA (1991). Preoperative and
postoperative “aneurysm” associated with coarctation of the aorta. J Am Coll Cardiol. 17,1367–
72. [PubMed: 2016456]
[203]. Ritchie CW, Bush AI, Mackinnon A, Macfarlane S, Mastwyk M, Macgregor L, Kiers L, Cherny
R, Li QX, Tammer A, Carrington D, Mavros C, Volitakis I, Xilinas M, Ames D, Davis S,
Beyreuther K, Tanzi RE & Masters CL (2003). Metal-protein attenuation with
Author Manuscript
ad mice increases bdnf levels and prevents cognitive deficits as well as mitochondrial
dysfunction. Cell Death Dis. 1,e91. [PubMed: 21368864]
Author Manuscript
[206]. Bryan MR & Bowman AB (2017). Manganese and the insulin-igf signaling network in
huntington’s disease and other neurodegenerative disorders. Adv Neurobiol. 18,113–42.
[PubMed: 28889265]
[207]. Caudle WM (2017). Occupational metal exposure and parkinsonism. Adv Neurobiol. 18,143–
58. [PubMed: 28889266]
[208]. Lucchini R, Placidi D, Cagna G, Fedrighi C, Oppini M, Peli M & Zoni S (2017). Manganese
and developmental neurotoxicity. Adv Neurobiol. 18,13–34. [PubMed: 28889261]
[209]. Zogzas CE & Mukhopadhyay S (2017). Inherited disorders of manganese metabolism. Adv
Neurobiol. 18,35–49. [PubMed: 28889262]
[210]. Chtourou Y, Trabelsi K, Fetoui H, Mkannez G, Kallel H & Zeghal N (2011). Manganese
induces oxidative stress, redox state unbalance and disrupts membrane bound atpases on murine
neuroblastoma cells in vitro: Protective role of silymarin. Neurochem Res. 36,1546–57.
[PubMed: 21533646]
[211]. Fitsanakis VA, Au C, Erikson KM & Aschner M (2006). The effects of manganese on
Author Manuscript
[217]. Wang D, Zhang J, Jiang W, Cao Z, Zhao F, Cai T, Aschner M & Luo W (2017). The role of
nlrp3-casp1 in inflammasome-mediated neuroinflammation and autophagy dysfunction in
manganese-induced, hippocampal-dependent impairment of learning and memory ability.
Autophagy. 13,914–27. [PubMed: 28318352]
[218]. Markesbery WR, Ehmann WD, Hossain TI & Alauddin M (1984). Brain manganese
concentrations in human aging and alzheimer’s disease. Neurotoxicology. 5,49–57. [PubMed:
6538949]
[219]. Du K, Liu M, Pan Y, Zhong X & Wei M (2017). Association of serum manganese levels with
alzheimer’s disease and mild cognitive impairment: A systematic review and meta-analysis.
Nutrients. 9.
[220]. Wallin C, Kulkarni YS, Abelein A, Jarvet J, Liao Q, Strodel B, Olsson L, Luo J, Abrahams JP,
Sholts SB, Roos PM, Kamerlin SC, Graslund A & Warmlander SK (2016). Characterization of
mn(ii) ion binding to the amyloid-beta peptide in alzheimer’s disease. J Trace Elem Med Biol.
38,183–93. [PubMed: 27085215]
[221]. Guilarte TR, Burton NC, Verina T, Prabhu VV, Becker KG, Syversen T & Schneider JS (2008).
Author Manuscript
Schwartz J & Hu H (2002). Impact of breastfeeding on the mobilization of lead from bone. Am J
Epidemiol. 155,420–8. [PubMed: 11867353]
[230]. Brubaker CJ, Schmithorst VJ, Haynes EN, Dietrich KN, Egelhoff JC, Lindquist DM, Lanphear
BP & Cecil KM (2009). Altered myelination and axonal integrity in adults with childhood lead
exposure: A diffusion tensor imaging study. Neurotoxicology. 30,867–75.
[231]. Hu F, Xu L, Liu ZH, Ge MM, Ruan DY & Wang HL (2014). Developmental lead exposure
alters synaptogenesis through inhibiting canonical wnt pathway in vivo and in vitro. PLoS One.
9,e101894. [PubMed: 24999626]
[232]. Senut MC, Sen A, Cingolani P, Shaik A, Land SJ & Ruden DM (2014). Lead exposure disrupts
global DNA methylation in human embryonic stem cells and alters their neuronal differentiation.
Toxicol Sci. 139,142–61. [PubMed: 24519525]
[233]. Li S, Liu XL, Zhou XL, Jiang SJ & Yuan H (2015). Expression of calmodulin-related genes in
lead-exposed mice. Interdiscip Toxicol. 8,155–8. [PubMed: 27486376]
[234]. Sanders T, Liu Y, Buchner V & Tchounwou PB (2009). Neurotoxic effects and biomarkers of
lead exposure: A review. Rev Environ Health. 24,15–45. [PubMed: 19476290]
Author Manuscript
[235]. Bijoor AR, Sudha S & Venkatesh T (2012). Neurochemical and neurobehavioral effects of low
lead exposure on the developing brain. Indian J Clin Biochem. 27,147–51. [PubMed: 23543765]
[236]. Stansfield KH, Ruby KN, Soares BD, Mcglothan JL, Liu X & Guilarte TR (2015). Early-life
lead exposure recapitulates the selective loss of parvalbumin-positive gabaergic interneurons and
subcortical dopamine system hyperactivity present in schizophrenia. Transl Psychiatry. 5,e522.
[PubMed: 25756805]
[237]. Flora G, Gupta D & Tiwari A (2012). Toxicity of lead: A review with recent updates. Interdiscip
Toxicol. 5,47–58. [PubMed: 23118587]
[238]. Mazumdar M, Bellinger DC, Gregas M, Abanilla K, Bacic J & Needleman HL (2011). Low-
level environmental lead exposure in childhood and adult intellectual function: A follow-up
study. Environ Health. 10,24. [PubMed: 21450073]
[239]. Reuben A, Caspi A, Belsky DW, Broadbent J, Harrington H, Sugden K, Houts RM, Ramrakha
S, Poulton R & Moffitt TE (2017). Association of childhood blood lead levels with cognitive
function and socioeconomic status at age 38 years and with iq change and socioeconomic
Author Manuscript
[242]. Hu Q, Fu H, Ren T, Wang S, Zhou W, Song H, Han Y & Dong S (2008). Maternal low-level
lead exposure reduces the expression of psa-ncam and the activity of sialyltransferase in the
Author Manuscript
Zawia NH (2005). Lead (pb) exposure and its effect on app proteolysis and abeta aggregation.
Faseb j. 19,2083–4. [PubMed: 16230335]
[248]. Wu J, Basha MR, Brock B, Cox DP, Cardozo-Pelaez F, Mcpherson CA, Harry J, Rice DC,
Maloney B, Chen D, Lahiri DK & Zawia NH (2008). Alzheimer’s disease (ad)-like pathology in
aged monkeys after infantile exposure to environmental metal lead (pb): Evidence for a
developmental origin and environmental link for ad. J Neurosci. 28,3–9. [PubMed: 18171917]
[249]. Ashok A, Rai NK, Tripathi S & Bandyopadhyay S (2015). Exposure to as-, cd-, and pb-mixture
induces abeta, amyloidogenic app processing and cognitive impairments via oxidative stress-
dependent neuroinflammation in young rats. Toxicol Sci. 143,64–80. [PubMed: 25288670]
[250]. Giudetti AM, Romano A, Lavecchia AM & Gaetani S (2016). The role of brain cholesterol and
its oxidized products in alzheimer’s disease. Curr Alzheimer Res. 13,198–205. [PubMed:
26391039]
[251]. Gu H, Wei X, Monnot AD, Fontanilla CV, Behl M, Farlow MR, Zheng W & Du Y (2011). Lead
exposure increases levels of beta-amyloid in the brain and csf and inhibits lrp1 expression in app
transgenic mice. Neurosci Lett. 490,16–20. [PubMed: 21167913]
Author Manuscript
[252]. Bihaqi SW, Eid A & Zawia NH (2017). Lead exposure and tau hyperphosphorylation: An in
vitro study. Neurotoxicology. 62,218–23. [PubMed: 28765091]
[253]. Bihaqi SW & Zawia NH (2013). Enhanced taupathy and ad-like pathology in aged primate
brains decades after infantile exposure to lead (pb). Neurotoxicology. 39,95–101. [PubMed:
23973560]
[254]. Bihaqi SW, Alansi B, Masoud AM, Mushtaq F, Subaiea GM & Zawia NH (2018). Influence of
early life lead (pb) exposure on alpha-synuclein, gsk-3beta and caspase-3 mediated tauopathy:
Implications on alzheimer’s disease. Curr Alzheimer Res. 15,1114–22. [PubMed: 30068273]
[255]. Di Lorenzo L, Vacca A, Corfiati M, Lovreglio P & Soleo L (2007). Evaluation of tumor necrosis
factor-alpha and granulocyte colony-stimulating factor serum levels in lead-exposed smoker
workers. Int J Immunopathol Pharmacol. 20,239–47. [PubMed: 17624258]
[256]. Petrini M, Azzara A, Polidori R, Vatteroni ML, Caracciolo F, Carulli G & Ambrogi F (1982).
Serum factors inhibiting some leukocytic functions in hodgkin’s disease. Clin Immunol
Immunopathol. 23,124–32. [PubMed: 7047031]
Author Manuscript
[257]. Liu MC, Liu XQ, Wang W, Shen XF, Che HL, Guo YY, Zhao MG, Chen JY & Luo WJ (2012).
Involvement of microglia activation in the lead induced long-term potentiation impairment. PLoS
One. 7,e43924. [PubMed: 22952811]
[258]. Kumawat KL, Kaushik DK, Goswami P & Basu A (2014). Acute exposure to lead acetate
activates microglia and induces subsequent bystander neuronal death via caspase-3 activation.
Neurotoxicology. 41,143–53. [PubMed: 24530660]
[259]. Liu JT, Chen BY, Zhang JQ, Kuang F & Chen LW (2015). Lead exposure induced microgliosis
and astrogliosis in hippocampus of young mice potentially by triggering tlr4-myd88-nfkappab
signaling cascades. Toxicol Lett. 239,97–107. [PubMed: 26386401]
[260]. Majewska M & Szczepanik M (2006). [the role of toll-like receptors (tlr) in innate and adaptive
immune responses and their function in immune response regulation]. Postepy Hig Med Dosw
Author Manuscript
[265]. Colomina MT & Peris-Sampedro F (2017). Aluminum and alzheimer’s disease. Adv Neurobiol.
18,183–97. [PubMed: 28889268]
[266]. Singla N & Dhawan DK (2012). Regulatory role of zinc during aluminium-induced altered
carbohydrate metabolism in rat brain. J Neurosci Res. 90,698–705. [PubMed: 22108899]
[267]. Yokel RA & Mcnamara PJ (2001). Aluminium toxicokinetics: An updated minireview.
Pharmacol Toxicol. 88,159–67. [PubMed: 11322172]
[268]. Muma NA & Singer SM (1996). Aluminum-induced neuropathology: Transient changes in
microtubule-associated proteins. Neurotoxicol Teratol. 18,679–90. [PubMed: 8947945]
[269]. Colomina MT, Roig JL, Sanchez DJ & Domingo JL (2002). Influence of age on aluminum-
induced neurobehavioral effects and morphological changes in rat brain. Neurotoxicology.
23,775–81. [PubMed: 12520767]
[270]. Shafer TJ & Mundy WR (1995). Effects of aluminum on neuronal signal transduction:
Mechanisms underlying disruption of phosphoinositide hydrolysis. Gen Pharmacol. 26,889–95.
[PubMed: 7557263]
Author Manuscript
[271]. Walton JR (2012). Aluminum disruption of calcium homeostasis and signal transduction
resembles change that occurs in aging and alzheimer’s disease. J Alzheimers Dis. 29,255–73.
[PubMed: 22330830]
[272]. Kaji R, Izumi Y, Adachi Y & Kuzuhara S (2012). Als-parkinsonism-dementia complex of kii
and other related diseases in japan. Parkinsonism Relat Disord. 18 Suppl 1,S190–1. [PubMed:
22166431]
[273]. Flaten TP (2001). Aluminium as a risk factor in alzheimer’s disease, with emphasis on drinking
water. Brain Res Bull. 55,187–96. [PubMed: 11470314]
[274]. Forbes WF & Mclachlan DR (1996). Further thoughts on the aluminum-alzheimer’s disease
link. J Epidemiol Community Health. 50,401–3. [PubMed: 8882222]
[275]. Martyn CN, Barker DJ, Osmond C, Harris EC, Edwardson JA & Lacey RF (1989).
Geographical relation between alzheimer’s disease and aluminum in drinking water. Lancet.
1,59–62. [PubMed: 2562879]
[276]. Neri LC & Hewitt D (1991). Aluminium, alzheimer’s disease, and drinking water. Lancet.
338,390.
Author Manuscript
[277]. O’farrell EK (1975). Write for the reader, he may need to know what you have to say. Can
Nurse. 71,24–7.
[278]. Boni UD, Otvos A, Scott JW & Crapper DR (1976). Neurofibrillary degeneration induced by
systemic aluminum. Acta Neuropathol. 35,285–94. [PubMed: 961381]
[279]. Kihira T, Yoshida S, Yase Y, Ono S & Kondo T (2002). Chronic low-ca/mg high-al diet induces
neuronal loss. Neuropathology. 22,171–9. [PubMed: 12416556]
[280]. Klatzo I, Wisniewski H & Streicher E (1965). Experimental production of neurofibrillary
degeneration. I. Light microscopic observations. J Neuropathol Exp Neurol. 24,187–99.
[PubMed: 14280496]
[281]. Savory J, Huang Y, Herman MM, Reyes MR & Wills MR (1995). Tau immunoreactivity
associated with aluminum maltolate-induced neurofibrillary degeneration in rabbits. Brain Res.
Author Manuscript
[PubMed: 15113600]
[287]. Huang W, Cheng P, Yu K, Han Y, Song M & Li Y (2017). Hyperforin attenuates aluminum-
induced abeta production and tau phosphorylation via regulating akt/gsk-3beta signaling pathway
in pc12 cells. Biomed Pharmacother. 96,1–6. [PubMed: 28961505]
[288]. Nubling G, Bader B, Levin J, Hildebrandt J, Kretzschmar H & Giese A (2012). Synergistic
influence of phosphorylation and metal ions on tau oligomer formation and coaggregation with
alpha-synuclein at the single molecule level. Mol Neurodegener. 7,35. [PubMed: 22824345]
[289]. Scott CW, Fieles A, Sygowski LA & Caputo CB (1993). Aggregation of tau protein by
aluminum. Brain Res. 628,77–84. [PubMed: 8313173]
[290]. Yamamoto H, Saitoh Y, Yasugawa S & Miyamoto E (1990). Dephosphorylation of tau factor by
protein phosphatase 2a in synaptosomal cytosol fractions, and inhibition by aluminum. J
Neurochem. 55,683–90. [PubMed: 2164575]
[291]. Akiyama H, Hosokawa M, Kametani F, Kondo H, Chiba M, Fukushima M & Tabira T (2012).
Long-term oral intake of aluminium or zinc does not accelerate alzheimer pathology in abetapp
and abetapp/tau transgenic mice. Neuropathology. 32,390–7. [PubMed: 22118300]
Author Manuscript
[292]. Zielke HR, Jackson MJ, Tildon JT & Max SR (1993). A glutamatergic mechanism for
aluminum toxicity in astrocytes. Mol Chem Neuropathol. 19,219–33. [PubMed: 8104402]
[293]. Helber A, Dvorak K, Winkelmann W, Meurer KA, Wurz H & Dickmans A (1975). [131i-
cholesterol scanning of the adrenals: Results in various adrenal diseases, especially unilateral
adrenal turmours (author’s transl)]. Dtsch Med Wochenschr. 100,2524–7. [PubMed: 1192963]
[294]. Quintanilla RA, Orellana DI, Gonzalez-Billault C & Maccioni RB (2004). Interleukin-6 induces
alzheimer-type phosphorylation of tau protein by deregulating the cdk5/p35 pathway. Exp Cell
Res. 295,245–57. [PubMed: 15051507]
[295]. Akinrinade ID, Memudu AE, Ogundele OM & Ajetunmobi OI (2015). Interplay of glia
activation and oxidative stress formation in fluoride and aluminium exposure. Pathophysiology.
22,39–48. [PubMed: 25577494]
[296]. Exley C, Price NC, Kelly SM & Birchall JD (1993). An interaction of beta-amyloid with
aluminium in vitro. FEBS Lett. 324,293–5. [PubMed: 8405368]
[297]. Luo Y, Niu F, Sun Z, Cao W, Zhang X, Guan D, Lv Z, Zhang B & Xu Y (2009). Altered
Author Manuscript
[300]. Pratico D, Uryu K, Sung S, Tang S, Trojanowski JQ & Lee VM (2002). Aluminum modulates
brain amyloidosis through oxidative stress in app transgenic mice. Faseb j. 16,1138–40.
Author Manuscript
[PubMed: 12039845]
[301]. Drago D, Bolognin S & Zatta P (2008). Role of metal ions in the abeta oligomerization in
alzheimer’s disease and in other neurological disorders. Curr Alzheimer Res. 5,500–7. [PubMed:
19075576]
[302]. Drago D, Cavaliere A, Mascetra N, Ciavardelli D, Di Ilio C, Zatta P & Sensi SL (2008).
Aluminum modulates effects of beta amyloid(1–42) on neuronal calcium homeostasis and
mitochondria functioning and is altered in a triple transgenic mouse model of alzheimer’s
disease. Rejuvenation Res. 11,861–71. [PubMed: 18788899]
[303]. Ricchelli F, Drago D, Filippi B, Tognon G & Zatta P (2005). Aluminum-triggered structural
modifications and aggregation of beta-amyloids. Cell Mol Life Sci. 62,1724–33. [PubMed:
15990957]
[304]. Oshiro S, Kawahara M, Mika S, Muramoto K, Kobayashi K, Ishige R, Nozawa K, Hori M,
Yung C, Kitajima S & Kuroda Y (1998). Aluminum taken up by transferrin-independent iron
uptake affects the iron metabolism in rat cortical cells. J Biochem. 123,42–6. [PubMed: 9504407]
Author Manuscript
[305]. Yamanaka K, Minato N & Iwai K (1999). Stabilization of iron regulatory protein 2, irp2, by
aluminum. FEBS Lett. 462,216–20. [PubMed: 10580122]
[306]. Lin R, Chen X, Li W, Han Y, Liu P & Pi R (2008). Exposure to metal ions regulates mrna levels
of app and bace1 in pc12 cells: Blockage by curcumin. Neurosci Lett. 440,344–7. [PubMed:
18583042]
[307]. Walton JR & Wang MX (2009). App expression, distribution and accumulation are altered by
aluminum in a rodent model for alzheimer’s disease. J Inorg Biochem. 103,1548–54. [PubMed:
19818510]
[308]. Kaneko N, Sugioka T & Sakurai H (2007). Aluminum compounds enhance lipid peroxidation in
liposomes: Insight into cellular damage caused by oxidative stress. J Inorg Biochem. 101,967–75.
[PubMed: 17467804]
[309]. Kim Y, Olivi L, Cheong JH, Maertens A & Bressler JP (2007). Aluminum stimulates uptake of
non-transferrin bound iron and transferrin bound iron in human glial cells. Toxicol Appl
Pharmacol. 220,349–56. [PubMed: 17376497]
[310]. Oteiza PI (1994). A mechanism for the stimulatory effect of aluminum on iron-induced lipid
Author Manuscript
[319]. Abu-Taweel GM, Ajarem JS & Ahmad M (2012). Neurobehavioral toxic effects of perinatal
oral exposure to aluminum on the developmental motor reflexes, learning, memory and brain
neurotransmitters of mice offspring. Pharmacol Biochem Behav. 101,49–56. [PubMed:
22115621]
[320]. Liu YQ, Xin TR, Liang JJ, Wang WM & Zhang YY (2010). Memory performance, brain
excitatory amino acid and acetylcholinesterase activity of chronically aluminum exposed mice in
response to soy isoflavones treatment. Phytother Res. 24,1451–6. [PubMed: 20878693]
[321]. Hefti F & Weiner WJ (1986). Nerve growth factor and alzheimer’s disease. Ann Neurol.
20,275–81. [PubMed: 3532929]
[322]. Yellamma K, Saraswathamma S & Kumari BN (2010). Cholinergic system under aluminium
toxicity in rat brain. Toxicol Int. 17,106–12. [PubMed: 21170257]
[323]. Hegde ML, Bharathi P, Suram A, Venugopal C, Jagannathan R, Poddar P, Srinivas P,
Sambamurti K, Rao KJ, Scancar J, Messori L, Zecca L & Zatta P (2009). Challenges associated
with metal chelation therapy in alzheimer’s disease. J Alzheimers Dis. 17,457–68. [PubMed:
Author Manuscript
19363258]
[324]. Exley C (2007). Organosilicon therapy in alzheimer’s disease? J Alzheimers Dis. 11,301–2;
discussion 3–4. [PubMed: 17851179]
[325]. Yokel RA (1994). Aluminum chelation: Chemistry, clinical, and experimental studies and the
search for alternatives to desferrioxamine. J Toxicol Environ Health. 41,131–74. [PubMed:
8301696]
[326]. Qadir S, Jamshieed S, Rasool S, Ashraf M, Akram NA & Ahmad P (2014). Modulation of plant
growth and metabolism in cadmium-enriched environments. Rev Environ Contam Toxicol.
229,51–88. [PubMed: 24515810]
[327]. Huang B & Goldsbrough PB (1988). Cadmium tolerance in tobacco cell culture and its
relevance to temperature stress. Plant Cell Rep. 7,119–22. [PubMed: 24241547]
[328]. Zhang M, Mo H, Sun W, Guo Y & Li J (2016). Systematic isolation and characterization of
cadmium tolerant genes in tobacco: A cdna library construction and screening approach. PLoS
One. 11,e0161147. [PubMed: 27579677]
Author Manuscript
[329]. Richter P, Faroon O & Pappas RS (2017). Cadmium and cadmium/zinc ratios and tobacco-
related morbidities. Int J Environ Res Public Health. 14.
[330]. Fransson MN, Barregard L, Sallsten G, Akerstrom M & Johanson G (2014). Physiologically-
based toxicokinetic model for cadmium using markov-chain monte carlo analysis of
concentrations in blood, urine, and kidney cortex from living kidney donors. Toxicol Sci.
141,365–76. [PubMed: 25015660]
[331]. Satarug S (2018). Dietary cadmium intake and its effects on kidneys. Toxics. 6.
[332]. Suwazono Y, Kido T, Nakagawa H, Nishijo M, Honda R, Kobayashi E, Dochi M & Nogawa K
(2009). Biological half-life of cadmium in the urine of inhabitants after cessation of cadmium
exposure. Biomarkers. 14,77–81. [PubMed: 19330585]
[333]. Akesson A, Bjellerup P, Lundh T, Lidfeldt J, Nerbrand C, Samsioe G, Skerfving S & Vahter M
(2006). Cadmium-induced effects on bone in a population-based study of women. Environ Health
Perspect. 114,830–4. [PubMed: 16759980]
[334]. Del Pino J, Zeballos G, Anadon MJ, Capo MA, Diaz MJ, Garcia J & Frejo MT (2014). Higher
sensitivity to cadmium induced cell death of basal forebrain cholinergic neurons: A
Author Manuscript
[338]. Wang B & Du Y (2013). Cadmium and its neurotoxic effects. Oxid Med Cell Longev.
2013,898034. [PubMed: 23997854]
Author Manuscript
[339]. Chen L, Liu L & Huang S (2008). Cadmium activates the mitogen-activated protein kinase
(mapk) pathway via induction of reactive oxygen species and inhibition of protein phosphatases
2a and 5. Free Radic Biol Med. 45,1035–44. [PubMed: 18703135]
[340]. Xu B, Chen S, Luo Y, Chen Z, Liu L, Zhou H, Chen W, Shen T, Han X, Chen L & Huang S
(2011). Calcium signaling is involved in cadmium-induced neuronal apoptosis via induction of
reactive oxygen species and activation of mapk/mtor network. PLoS One. 6,e19052. [PubMed:
21544200]
[341]. Min JY & Min KB (2016). Blood cadmium levels and alzheimer’s disease mortality risk in
older us adults. Environ Health. 15,69. [PubMed: 27301955]
[342]. Peng Q, Bakulski KM, Nan B & Park SK (2017). Cadmium and alzheimer’s disease mortality in
u.S. Adults: Updated evidence with a urinary biomarker and extended follow-up time. Environ
Res. 157,44–51. [PubMed: 28511080]
[343]. Li X, Lv Y, Yu S, Zhao H & Yao L (2012). The effect of cadmium on abeta levels in app/ps1
transgenic mice. Exp Ther Med. 4,125–30. [PubMed: 23060935]
Author Manuscript
[344]. Notarachille G, Arnesano F, Calo V & Meleleo D (2014). Heavy metals toxicity: Effect of
cadmium ions on amyloid beta protein 1–42. Possible implications for alzheimer’s disease.
Biometals. 27,371–88. [PubMed: 24557150]
[345]. Syme CD & Viles JH (2006). Solution 1h nmr investigation of zn2+ and cd2+ binding to
amyloid-beta peptide (abeta) of alzheimer’s disease. Biochim Biophys Acta. 1764,246–56.
[PubMed: 16266835]
[346]. Endres K & Fahrenholz F (2012). The role of the anti-amyloidogenic secretase adam10 in
shedding the app-like proteins. Curr Alzheimer Res. 9,157–64. [PubMed: 21605036]
[347]. Del Pino J, Zeballos G, Anadon MJ, Moyano P, Diaz MJ, Garcia JM & Frejo MT (2016).
Cadmium-induced cell death of basal forebrain cholinergic neurons mediated by muscarinic m1
receptor blockade, increase in gsk-3beta enzyme, beta-amyloid and tau protein levels. Arch
Toxicol. 90,1081–92. [PubMed: 26026611]
[348]. Jiang LF, Yao TM, Zhu ZL, Wang C & Ji LN (2007). Impacts of cd(ii) on the conformation and
self-aggregation of alzheimer’s tau fragment corresponding to the third repeat of microtubule-
binding domain. Biochim Biophys Acta. 1774,1414–21. [PubMed: 17920001]
Author Manuscript
[349]. Del Pino J, Zeballos G, Anadon MJ, Diaz MJ, Moyano P, Diaz GG, Garcia J, Lobo M & Frejo
MT (2016). Muscarinic m1 receptor partially modulates higher sensitivity to cadmium-induced
cell death in primary basal forebrain cholinergic neurons: A cholinesterase variants dependent
mechanism. Toxicology. 361–362,1–11.
[350]. Medeiros R, Kitazawa M, Caccamo A, Baglietto-Vargas D, Estrada-Hernandez T, Cribbs DH,
Fisher A & Laferla FM (2011). Loss of muscarinic m1 receptor exacerbates alzheimer’s disease-
like pathology and cognitive decline. Am J Pathol. 179,980–91. [PubMed: 21704011]
[351]. Phuagkhaopong S, Ospondpant D, Kasemsuk T, Sibmooh N, Soodvilai S, Power C &
Vivithanaporn P (2017). Cadmium-induced il-6 and il-8 expression and release from astrocytes
are mediated by mapk and nf-kappab pathways. Neurotoxicology. 60,82–91. [PubMed:
28288823]
[352]. Liu C, Cui G, Zhu M, Kang X & Guo H (2014). Neuroinflammation in alzheimer’s disease:
Chemokines produced by astrocytes and chemokine receptors. Int J Clin Exp Pathol. 7,8342–55.
[PubMed: 25674199]
Author Manuscript
[353]. Jiang JH, Ge G, Gao K, Pang Y, Chai RC, Jia XH, Kong JG & Yu AC (2015). Calcium signaling
involvement in cadmium-induced astrocyte cytotoxicity and cell death through activation of
mapk and pi3k/akt signaling pathways. Neurochem Res. 40,1929–44. [PubMed: 26248512]
brain. The production of Aβ occurs due to aberrant processing of the APP, whereby it is
cleaved by β- and γ-secretases, instead of α-secretase. The Aβ peptide is prone to
misfolding and aggregation, leading to eventual oligomerization and formation of Aβ
plaques, which trigger a proinflammatory response from microglia and astrocytes. Aβ also
causes the hyperphosphorylation of tau, leading to its dissociation from microtubules and
their eventual destabilization within neurons. Hyperphosphorylated tau is also prone to
aggregation, forming NFTs, which correlates with neuronal loss and neurodegeneration.
Author Manuscript
Figure 2 ‒. The molecular composition of the body: essential and non-essential elements.
In human biology, the constituents of the body are classified into four groups according to
their increasing complexity: atomic, molecular, cellular and tissue-system. In this system,
the more complex components are built by combining the more basic ones. At the atomic
level, only four of the 118 chemical elements currently known (i.e., oxygen, carbon,
hydrogen and nitrogen) are needed to make about 96% of the mass of the human body.
Author Manuscript
aluminum, lead and cadmium. Non-essential elements can cause cellular dysfunction at low
Author Manuscript
removal from the brain. Zinc and manganese are also present in trace amounts which are
finely regulated and essential to maintain brain function. (Right panel) In the AD brain,
dyshomeostasis of essential metals seems to be linked with AD pathogenesis. Impairment of
APP function, present in AD, can trigger an increased level of both intracellular and
extracellular Fe2+ and Cu2+, and a reduction of extracellular Cu+, thus promoting its
accumulation. Excessive Fe2+ and Cu2+ increases oxidative stress via production of ROS. In
addition, iron, copper and zinc have higher binding affinity to Aβ and can promote its
aggregation. Increased neuronal iron, copper and zinc also bind to tau protein and facilitate
Author Manuscript
the formation of NFTs. Consequently, excessive amounts of heavy metals are found within
plaques and NFT. Dyshomeostasis of essential metals in the extracellular space induce
microglial and astrocytic activation, followed by the overproduction of proinflammatory
cytokines such as, IL-1β and TNF-α.
Author Manuscript
Author Manuscript
Author Manuscript