You are on page 1of 7

Archives of Biochemistry and Biophysics 611 (2016) 93e99

Contents lists available at ScienceDirect

Archives of Biochemistry and Biophysics


journal homepage: www.elsevier.com/locate/yabbi

Insight into cognitive decline from Zn2þ dynamics through


extracellular signaling of glutamate and glucocorticoids
Atsushi Takeda*, Hanuna Tamano
Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan

a r t i c l e i n f o a b s t r a c t

Article history: Glutamatergic neuron activity and/or the modification of the activity with glucocorticoids are closely
Received 1 April 2016 linked to synaptic Zn2þ dynamics as well as synaptic Ca2þ dynamics. The dynamic crosstalk of synaptic
Received in revised form Zn2þ signaling to intracellular Ca2þ signaling via calcium channels is involved in synaptic plasticity such
23 June 2016
as long-term potentiation (LTP) and cognitive activity. The influx of extracellular Zn2þ into postsynaptic
Accepted 30 June 2016
neurons, which is closely linked to glutamate signaling in the synaptic cleft, is critical for cognitive ac-
Available online 5 July 2016
tivity. However, excess intracellular Zn2þ signaling induced by excess glutamatergic neuron activity is
involved in not only cognitive decline in neurological disorders but also stress-induced cognitive decline.
Keywords:
Zinc
On the other hand, it has been recognized that excess Ca2þ influx into postsynaptic neurons induces
Glutamate neuronal death, while the involvement of excess intracellular Ca2þ signaling in cognitive decline is poorly
Glucocorticoid understood. Understanding of synaptic Zn2þ dynamics, which are modified by glutamate and gluco-
Hippocampus corticoid signaling, may be meaningful to prevent Zn2þ-mediated cognitive decline. This paper sum-
Cognition marizes the current knowledge on Zn2þ dynamics under changing synaptic environment and its impact
Calcium on cognitive decline.
© 2016 Elsevier Inc. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 93
2. Brain zinc homeostasis through the brain barrier system and its impact on cognition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 94
3. Impact on cognition from extracellular and intracellular Zn2þ homeostasis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 94
4. Impact on cognition from neuronal death via excess synaptic Zn2þ signaling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 95
5. Impact on cognition from synaptic Zn2þ signaling under diverse synaptic environment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 96
6. Perspective . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 98
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 98

1. Introduction responsible for the transport of Zn2þ out of the cytoplasm, are
involved in the absorption and excretion of Zn2þ to maintain zinc
Plasma zinc is thought to be a major pool of zinc in the living homeostasis in the living body [2e4].
body to transfer zinc to the tissues and organs including the brain. Zinc concentration in the plasma is approximately 15 mM. Zinc
Zinc homeostasis in the living body is tightly controlled by both transport from the plasma to the brain extracellular fluid and the
intestinal absorption and intestinal and renal excretions [1]. Zrt-Irt- cerebrospinal fluid (CSF) is regulated by the brain barrier system,
like proteins (ZIP), which are responsible for the movement of Zn2þ i.e., the bloodebrain and bloodeCSF barriers. Zinc concentration in
into the cytoplasm, and the zinc transporter (ZnT) family, which are the CSF is approximately 0.15e0.38 mM [5e7] and extracellular zinc
concentration in the brain is estimated to be less than 1 mM [8]. Zinc
concentration in the brain increases along with the development
* Corresponding author. and reaches 13.3 ± 0.3 mg/g wet brain in the human adult brain
E-mail address: takedaa@u-shizuoka-ken.ac.jp (A. Takeda). (infant brain, 8.2 ± 0.8 mg/g wet brain) [9]. Brain zinc homeostasis is

http://dx.doi.org/10.1016/j.abb.2016.06.021
0003-9861/© 2016 Elsevier Inc. All rights reserved.
94 A. Takeda, H. Tamano / Archives of Biochemistry and Biophysics 611 (2016) 93e99

strictly controlled through the brain barrier system under healthy Zn2þ homeostasis is critical for brain function such as cognitive
condition [10,11], while synaptic Zn2þ movement is changed by activity and seems to be modified along with brain aging [29e31].
synaptic activity, especially glutamatergic synapse activity and is Brain aging is characterized by neuronal loss, cognitive decline,
involved in synaptic function [12]. Approximately 80% of the total and susceptibility to neurological disorders [32]. The changes
brain zinc functions as zinc metalloproteins and the remaining part occurring in the brain during aging may be often related to the
is histochemically reactive as revealed by Timm’s sulfide-silver modification of Zn2þ homeostasis. Zinc transporter-3 (ZnT3) pro-
staining method [13,14] and serves as a signal factor (in the form tein is responsible for loading zinc into presynaptic vesicles [33]
of free Zn2þ) in the cytosolic compartment as well as the extra- and consequently controls the availability of Zn2þ at zincergic
cellular compartment. synapses, which is critical for hippocampus-dependent memory as
The hippocampus and amygdala are involved in cognitive and described later [34]. Because ZnT3 protein decreases along with
emotional behavior and strongly stained by Timm’s method aging [35], it may be involved in age-related cognitive decline.
[15e17]. To process the information on memory, glutamatergic Adlard et al. [36,37] report that metal chaperones i.e., clioquinol
neurons compose the principal neural circuits in the hippocampus and PBT2, are transported into the brain through the brain barrier
and amygdala. The role of the neural circuits has been extensively system, can increase the availability of Zn2þ, and prevent normal
studied in the hippocampus. Memory are linked to strengthening of age-related cognitive decline. They demonstrate that metal chap-
synaptic connections between neurons, i.e., synaptic plasticity such erones are effective for preventing Zn2þ-mediated cognitive decline
as long-term potentiation (LTP). Glutamatergic neuron activity is that might be increased with aging. Metal chaperones transported
closely linked to synaptic Zn2þ dynamics as well as synaptic Ca2þ into the brain may be also effective as a therapeutic strategy for
dynamics [15,16]. The dynamic crosstalk of synaptic Zn2þ signaling neurodegenerative diseases associated with brain Zn2þ dysho-
to intracellular Ca2þ signaling via calcium channels is involved in meostasis [38,39] Brain Zn2þ dyshomeostasis may be involved in
cognitive activity based on synaptic plasticity. Because a subclass of cognitive decline under both pathological and physiological
glutamatergic (zincergic) neurons releases Zn2þ into the synaptic conditions.
cleft [13], synaptic (extracellular) Zn2þ movement is closely linked Zinc concentration in the brain is hardly affected after exposure
to glutamate signaling in the synaptic cleft and modifies intracel- to zinc. The brain barrier system protects brain parenchyma cells by
lular Zn2þ signaling [18]. Even at non-zincergic synapses, intracel- blocking excess Zn2þ influx. In a bloodebrain barrier model, the
lular Zn2þ signaling, which mainly originates in the internal stores expressions of zinc transporters such as ZnT1 and ZnT2, and also
containing Zn2þ, is critical for cognitive activity. metallothioneins, zinc-binding proteins are changed to respond to
The hypothalamic-pituitary-adrenal (HPA) axis serves to main- moderately excessive zinc environment [40]. The brain barrier
tain homeostasis in the living body and the HPA axis activity, i.e., system protects brain parenchyma cells from neurotoxicity of
glucocorticoid secretion, is enhanced after exposure to stress. metals including zinc. The bloodebrain barrier breakdown that
Glucocorticoids can potentiate glutamatergic neuron activity under occurs during normal aging is associated with inflammation and
both pathological and physiological conditions [19,20] and modify disruption of tight junction complex assembly but not through
synaptic Zn2þ signaling [21]. Excess intracellular Zn2þ signaling leukocyte trafficking [41]. Montagne et al. report that the bloode-
induced by excess glutamatergic neuron activity is involved in not brain barrier breakdown is an early event in the aged human brain
only cognitive decline in neurological disorders but also stress- that begins in the hippocampus and may contribute to cognitive
induced cognitive decline [22]. Thus, diverse Zn2þ dynamics decline [42]. Furthermore, the brain capillary endothelial cells play
induced by glutamate and glucocorticoid signaling may modify an active part in the process of neurological disorders, in which
cognitive activity. To prevent Zn2þ-mediated cognitive decline, this cognitive decline is involved. The bloodebrain barrier is modified
paper summarizes the current knowledge on Zn2þ dynamics under and/or disrupted in the process of neurological disorders such as
changing synaptic environment and its impact on cognitive decline. neoplasia, stroke/ischemia, epilepsy, and dementia [43]. The in-
The present topics propose that the influx of extracellular Zn2þ into crease in the bloodebrain barrier permeability leads to the dys-
postsynaptic neurons may be critical for cognitive decline in com- homeostasis of essential metals such as zinc and iron, which has
parison with that of extracellular Ca2þ and probably involved in been implicated in many neurodegenerative disorders, followed by
age-related cognitive decline. cognitive decline [44].

2. Brain zinc homeostasis through the brain barrier system 3. Impact on cognition from extracellular and intracellular
and its impact on cognition Zn2þ homeostasis

Healthy brain maintains a very stable environment via the brain The basal (static) concentration of Zn2þ in the brain extracellular
barrier system, which is critical for brain function including fluid is estimated to be 10 nM [45]. Fluorescence intensity of
cognition. Zn2þ is slowly transported into the brain through the extracellular ZnAF-2, an extracellular Zn2þ indicator [46], varies
bloodebrain and the bloodeCSF barriers [23,24]. The cells forming among areas in the hippocampus [47,48], where the stratum luci-
the brain barrier system express ZIP and ZnT, and play key roles for dum and the hilus are strongly stained with ZnAF-2. At zincergic
Zn2þ transport into the brain parenchyma and also brain zinc ho- synapses such as mossy fiber and Schaffer collateral synapses,
meostasis [25]. However, the mechanism on Zn2þ transport across extracellular Zn2þ levels are increased by cognitive activity and LTP
the brain barrier system is poorly understood and the mechanism induction. However, the optimal range (level) of Zn2þ in the
on brain zinc homeostasis is unclear [26,27]. On the other hand, extracellular compartment remains to be clarified [49]. In vivo CA1
chronic zinc deficiency suppresses the increase in zinc concentra- LTP is significantly attenuated under local perfusion of the
tion in the rat hippocampus, indicating that hippocampal zinc is recording region with 100 nM ZnCl2 by using a recording electrode
relatively susceptible to zinc deficiency in the brain [28]. Chronic attached to a microdialysis probe [50]. In the microdialysis exper-
zinc deficiency also suppresses the increase in vesicular zinc. Brain iment, it is estimated that extracellular Zn2þ level in the recording
Zn2þ including hippocampal Zn2þ seems to be susceptible to region is less than 100 nM, because the perfusate reaches the
chronic zinc deficiency. The decrease in plasma zinc elicited by equilibrium with extracellular fluid. When the basal level of
dietary zinc deficiency may lead to the decrease in CSF zinc, which extracellular Zn2þ rises to several times by zincergic excitation in
is important for Zn2þ transport into the hippocampus [24]. Brain the CA1 and the rise is maintained for a period of time, CA1 LTP can
A. Takeda, H. Tamano / Archives of Biochemistry and Biophysics 611 (2016) 93e99 95

be affected. On the other hand, extracellular Zn2þ concentration 4. Impact on cognition from neuronal death via excess
during tetanic stimulation to induce LTP is unclear at zincergic synaptic Zn2þ signaling
synapses. It is possible that the concentration reaches micromolar
levels based on the estimated concentration of vesicular zinc Glutamatergic neuron activity plays a key role for cognition
[51,52]. under physiological and pathological conditions. Glutamate re-
The basal Zn2þ concentrations are extremely low in the intra- ceptor activation by excess extracellular glutamate leads to a
cellular (cytosol) compartment and estimated to be less than 1 nM number of deleterious consequences, including impairment of
[53,54]. Intracellular Zn2þ homeostasis is closely linked to extra- calcium buffering, generation of free radicals, activation of the
cellular Zn2þ dynamics even at non-zincergic synapses because the mitochondrial permeability transition and secondary excitotoxicity
basal concentration of intracellular Zn2þ is much lower than that of [70,71] and is known as glutamate excitotoxicity. It is a final com-
extracellular Zn2þ. Fluorescence intensity of intracellular ZnAF-2 mon pathway for neuronal death and is observed in numerous
after addition of ZnAF-2DA, an intracellular Zn2þ indicator [46], pathological processes such as stroke/ischemia, temporal lobe ep-
also varies among areas in the hippocampus [47,48], and stratum ilepsy, Alzheimer’s disease and amyotrophic lateral sclerosis
lucidum and the hilus, which contain mossy fibers, are strongly [72,73], which are often accompanied by cognitive impairment.
stained with ZnAF-2. It is likely that the basal levels of intracellular Neuronal death is induced by excess Zn2þ influx into postsynaptic
Zn2þ are correlated with zinc concentration in the synaptic vesicle neurons, in addition to excess Ca2þ influx, at zincergic synapses and
[33]. There is the close correlation between vesicular Zn2þ level and is a cause for cognitive decline (Fig. 1) [74,75]. At non-zincergic
ZnT3 protein expression [55]. Zn2þ level other than Zn2þ released synapses, on the other hand, there is no report on Zn2þ-mediated
from the synaptic vesicle is low and estimated to be less than 5% of neuronal death. It is possible that Zn2þ influx into non-zincergic
the total amount of Zn2þ in the hippocampus and cerebral cortex postsynaptic neurons, in addition to Ca2þ influx, is involved in
[55]. At zincergic synapses, transsynaptic influx of Zn2þ released neuronal death because extracellular Zn2þ can pass through
from neuron terminals is involved in hippocampus-dependent calcium-permeable channels during synaptic excitation induced
memory [50,56,57]. with glutamate. Bell et al. demonstrates a striking pathology-
ZnT proteins such as ZnT1, ZnT3 and ZnT10, and ZIP such as dependent pattern of glutamatergic synaptic remodeling with
ZIP4 and ZIP6 are involved in the control of Zn2þ level in the disease progression. Subjects with mild cognitive impairment
cytosolic compartment, especially under the basal circumstance display a paradoxical elevation of glutamatergic presynaptic bou-
[58]. Some of these transporters transport cytosolic Zn2þ into ton density [76]. Because glutamate excitotoxicity more readily
different subcellular organelles, e.g., mitochondria, lysosomes, may occur in elderly people, it is likely that the excitotoxicity is a
endosomes, and Golgi apparatus, probably to maintain the basal cause for age-dependent cognitive decline [77].
Zn2þ level in the cytosolic compartment [59e61]. On the other Hippocampal CA1 and CA3 are susceptible to neuronal death
hand, Zn2þ release from the subcellular organelles is involved in induced by glutamate excitotoxicity [78,79] and attention has
intracellular Zn2þ signaling [62]. Intracellular Ca2þ signaling via been paid to Zn2þ release from hippocampal zincergic neuron
glutamate receptor activation may play a key role for intracellular terminals in neurodegeneration. Brief forebrain ischemia in ro-
Zn2þ signaling, although the mechanism is unknown. At non- dents induces selective and delayed neuronal death, particularly
zincergic synapses, intracellular Zn2þ signaling via glutamate re- in hippocampal CA1 pyramidal neurons mainly via Zn2þ accu-
ceptor activation is involved in hippocampus-dependent memory mulation [80e82]. Epileptic seizure induces neuronal death in
[63]. the hippocampal CA1 and CA3, but not in the dentate gyrus
ZnT1 is a major Zn2þ transporter on the plasma membrane [83,84]. GluR2-lacking calcium-permeable AMPA receptors may
and involved in cytosolic Zn2þ homeostasis in neurons by play a key role for Zn2þ-mediated neurodegeneration [81,85,86].
transporting Zn2þ from the somata to the extracellular space [64]. Therefore, Zn2þ-chelating strategies, which block Zn2þ-mediated
It has been reported that ZnT1 restricts excessive accumulation of neurodegeneration, may be effective for protecting CA1 and CA3
Zn2þ in the cytosolic compartment [65], resulting in protecting neurons in neurological disorders associated with glutamate
neurons from Zn2þ toxicity in neurological disorders such as excitotoxicity. On the other hand, dentate granule cells, which
transient forebrain ischemia [66]. Furthermore, ZnT1 concen- were scarcely innervated by zincergic neurons, seem to be less
trates at the postsynaptic density of hippocampal synapses susceptible to Zn2þ neurotoxicity in the hippocampal formation
regardless of the presence of vesicular zinc, suggesting that ZnT1 [87,88]. The levels of messenger RNA encoding the AMPA-
is involved in hippocampus-dependent memory via maintaining selective glutamate receptor subunit-1, GluR1 and GluR2 are
intracellular Zn2þ homeostasis [67]. ZIP1 and ZIP3 may provide the highest in the dentate gyrus, followed by the CA1 and CA3
selective therapeutic targets to protect hippocampal neurons hippocampal subfields [77], suggesting that the dentate gyrus is
from early zinc-induced neurodegeneration following injury [68]. susceptible for the influx of Ca2þ and Zn2þ. The GluR1/GluR2
Tissue plasminogen activator, a secreted serine protease is exci- messenger RNA ratios, an index of calcium-permeable AMPA re-
totoxic and increases lysosomal sequestration of the increased ceptors, are increased in the aged hippocampus, suggesting that
Zn2þ in the cytosolic compartment through interaction with ZIP4, intracellular Zn2þ dyshomeostasis, in addition to intracellular
which might also lead to protecting neurons from Zn2þ toxicity Ca2þ dyshomeostasis, may contribute to age-related neuronal
[58]. The spatiotemporal control of intracellular Zn2þ via ZIP and death in the hippocampus including the dentate gyrus [77],
ZnT is involved in the steady environment of Zn2þ in the cytosolic which may be linked to cognitive decline in elderly people
compartment, which is critical for hippocampus-dependent (Fig. 1). The social behavioral deficits are accompanied by a
memory [29]. The hippocampus, especially the dentate gyrus is change in AMPA receptor composition, which is regulated by the
the most susceptible to aging in the brain [69]. If one of the brain-enriched microRNA-124, leading to an imbalance between
causes is the bloodebrain barrier breakdown in the hippocampus calcium-permeable and calcium-impermeable AMPA receptors
[42], the control of ZIP and ZnT protein expression might be a key [89]. The evidence suggests a potential therapeutic avenue for
for the strategy preventing age-related hippocampus-dependent regulating social behavior in neurodegenerative disorders such as
cognitive decline. frontotemporal dementia.
96 A. Takeda, H. Tamano / Archives of Biochemistry and Biophysics 611 (2016) 93e99

Fig. 1. Neuronal death induced by glutamate excitotoxicity often leads to cognitive decline. It is well known that Ca2þ influx through calcium channels (CC) such as NMDA receptors
and calcium-permeable AMPA receptors is involved in postsynaptic neuronal death, while Zn2þ influx at zincergic synapses is also involved in the death.

5. Impact on cognition from synaptic Zn2þ signaling under cognition [90]. Interestingly, the extracellular zinc is decreased in
diverse synaptic environment the hippocampus in spite of the increase in extracellular glutamate
after exposure to a novel environment [101]. When the decrease is
The HPA axis activity, i.e., glucocorticoid secretion, is increased blocked with CaEDTA, an extracellular Zn2þ chelator, exploratory
after exposure to stress and buffers stress. The HPA axis activity is activity is simultaneously reduced, suggesting that Zn2þ influx into
linked to hippocampal function and also involved in cognitive and hippocampal neurons is linked to exploratory activity [101]. It is
emotional behavior [90]. It is well known that the HPA axis activity possible that the increase in glucocorticoid secretion induced by
is enhanced in aging [91,92] and neurological disorders [93]. Hip- the novelty stress is involved in Zn2þ influx into hippocampal
pocampal neuronal impairment is associated with the continuous neurons. Glucocorticoids require intracellular Zn2þ signaling for the
activation of the HPA axis, which is involved in the occurrence and genomic effect [102], suggesting that glucocorticoids modify syn-
progression of cognitive disorders in elderly subjects [94]. aptic Zn2þ signaling under stressful environment and that the
Furthermore, correlations have been reported between increases in modification is involved in cognitive activity.
HPA axis activity and dementia severity or hippocampal volume In contrast, restraint stress and foot-shock stress increases
loss in individuals with probable Alzheimer’s disease [95]. Corre- glutamate and zinc in the extracellular compartment. Both in-
lations are also reported between increases in HPA axis activity and creases may be mediated by the action of glucocorticoids [103,104].
depression severity or hippocampal volume loss in human de- In amygdala perfusion experiment, psychological stress decreases
pressives [96]. zinc in the extracellular fluid with no significant change in extra-
The hippocampus is a major target of glucocorticoids [97] and cellular glutamate [104]. The diverse dynamics of Zn2þ and gluta-
enriched with corticosteroid receptors [98]. The hippocampus is mate might be linked to stress-induced glucocorticoid signaling. A
involved in the negative feedback mechanism in glucocorticoid high dose of corticosterone increases Zn2þ release probability from
secretion. Mineralocorticoid receptors are extensively occupied zincergic neuron terminals through the rapid non-genomic effect
with low levels of corticosterone and glucocorticoid receptors are and affects CA1 LTP induction via excess influx of extracellular Zn2þ
particularly activated after exposure to stress [90,99]. An increase in into CA1 pyramidal neurons (Fig. 2) [21]. The evidence suggests
plasma corticosterone level leads to a rapid increase in corticoste- that glucocorticoid-mediated excess Zn2þ influx can affect learning
rone level in the hippocampus, which is in parallel with an increase and memory after exposure to stress. It is likely that the increase in
in extracellular glutamate level [100]. In the hippocampus, glucocorticoid secretion in neurological disorders is linked to Zn2þ-
corticosterone-mediated increase in extracellular glutamate levels mediated cognitive decline. On the other hand, glucocorticoids also
may occur through the action of membrane-associated mineralo- increase Ca2þ influx into hippocampal neurons [105,106].
corticoid receptors and/or glucocorticoid receptors, which in- Glucocorticoid-mediated impairment of CA1 LTP induction is
creases glutamate release probability in the non-genomic manner rescued in the presence of CaEDTA [21], suggesting that the
[19,20]. The rapid effects of corticosterone on glutamatergic impairment of LTP induction is due to Zn2þ influx, but not Ca2þ
neuron activity seem to be linked to the diverse effects on synaptic influx. It has been recognized that excess Ca2þ influx into post-
plasticity and memory processes in the hippocampus. Corticoste- synaptic neurons induces neuronal death. However, the involve-
rone contributes to increasing the efficacy of glutamatergic trans- ment of excess intracellular Ca2þ signaling in cognitive decline is
mission by AMPA receptor insertion at synaptic sites through both poorly understood.
the rapid and the delayed (genomic) effects. These effects are of The disease rate of stroke/ischemia is increased with aging. The
advantage to the process of synaptic plasticity such as LTP and concentration of extracellular Kþ is 3.5e5.5 mM under
A. Takeda, H. Tamano / Archives of Biochemistry and Biophysics 611 (2016) 93e99 97

Fig. 2. Attenuated LTP induction is linked to cognitive decline. Excess Zn2þ influx through calcium channels (CC) such as calcium-permeable AMPA receptors is induced by glu-
tamatergic synapse excitation. The Zn2þ influx subsequently induces cognitive decline via attenuated LTP induction.

physiological condition and reaches approximately 75 mM in brain cognitive decline [120,121]. Metal chaperone, i.e., PBT2, ameliorates
ischemia followed by glutamate excitotoxicity [107]. Glutamate- age-related cognitive decline via increases in dendritic spine den-
mediated neuronal death results in the efflux of intracellular Kþ sity, hippocampal neuron number, and markers of neurogenesis
[108]. Thus it is likely that the increase in extracellular Kþ, which [36], suggesting that the decrease in bioavailable Zn2þ is involved in
non-specifically induces neuronal excitation, leads to Zn2þ-medi- age-related cognitive decline.
ated cognitive decline. Excess glutamatergic neuron activity after In conclusion, extracellular Zn2þ dynamics are diversely modi-
injection of high Kþ into the hippocampal CA1 transiently impairs fied in response to changing synapse environment and involved in
object recognition (Fig. 2) [109]. The impairment is linked to excess cognitive activity in the normal brain. The modification is poten-
Zn2þ release from Schaffer collaterals and the influx of the Zn2þ into tially facilitated with normal aging and critical for cognitive activity
CA1 pyramidal neurons. Even in the dentate gyrus where is scarcely under pathological condition. Extracellular Zn2þ homeostasis is
innervated by zincergic neurons, excess influx of Zn2þ into dentate affected by biologically active substances such as glutamate and
granule cells affects object recognition via attenuated LTP induction glucocorticoids, followed by alteration of intracellular Zn2þ ho-
(Fig. 2) [110]. The findings suggest that the increase in extracellular meostasis (Fig. 3). This process induces Zn2þ-mediated cognitive
Kþ, which induces glutamatergic excitation, occurs in a probability
in the brain of elderly people and is a cause of age-related cognitive
decline.
Numerous studies have demonstrated that traumatic brain
injury and epileptic seizures increase hippocampal neurogenesis in
the rodent and human brain [111]. Newly born dentate granule cells
that arise as a result of traumatic brain injury and epileptic seizures
are integrated into existing hippocampal circuit and may provide
network plasticity for hippocampus-dependent learning and
memory [112,113]. Intracellular Zn2þ signaling is required for hip-
pocampal neurogenesis and has an essential role for hippocampal
neurogenesis after traumatic brain injury and seizure [114e116]. On
the other hand, stress and glucocorticoid strongly inhibit adult
hippocampal neurogenesis [117] and decreased neurogenesis has
been implicated in the pathogenesis of anxiety and depression
[118,119]. Aging is associated with decreased neurogenesis in the Fig. 3. Is Zn2þ-mediated cognitive decline increased along with aging? Glutamatergic
hippocampus, which is linked to hippocampus-dependent synapse excitation and calcium-permeable AMPA receptor expression are increased
with aging as described in the text and critical for Zn2þ influx.
98 A. Takeda, H. Tamano / Archives of Biochemistry and Biophysics 611 (2016) 93e99

decline and may be linked to the changes in Zn2þ-transport protein A.I. Bush, D.I. Finkelstein, R.A. Cherny, Aging Cell. 13 (2014) 351e359.
[37] P.A. Adlard, J. Parncutt, V. Lal, S. James, D. Hare, P. Doble, D.I. Finkelstein,
expression, e.g., increase in calcium-permeable AMPA receptors. To
A.I. Bush, Neurobiol. Dis. 81 (2015) 196e202.
understand not only Zn2þ-mediated cognitive decline but also age- [38] A.I. Bush, J. Alzheimers Dis. 33 (2013) S277eS281.
related cognitive decline, it is important to analyze in vivo extra- [39] S. Ayton, P. Lei, A.I. Bush, Neurotherapeutics 12 (2015) 109e120.
cellular Zn2þ dynamics. [40] D.J. Bobilya, N.A. Gauthier, S. Karki, B.J. Olley, W.K. Thomas, J. Nutr. Biochem.
19 (2008) 129e137.
[41] P. Ballabh, A. Braun, M. Nedergaard, Neurobiol. Dis. 16 (2004) 1e13.
6. Perspective [42] M. Elahy, C. Jackaman, J.C. Mamo, V. Lam, S.S. Dhaliwal, C. Giles, D. Nelson,
R. Takechi, Immun. Ageing 12 (2015) 2.
[43] A. Montagne, S.R. Barnes, M.D. Sweeney, M.R. Halliday, A.P. Sagare, Z. Zhao,
In the last century, cognitive decline in elderly people was A.W. Toga, R.E. Jacobs, C.Y. Liu, L. Amezcua, M.G. Harrington, H.C. Chui,
considered as the consequence of neuronal death (Fig. 1). However, M. Law, B.W. Zlokovic, Blood-brain barrier breakdown in the aging human
hippocampus, Neuron 85 (2015) 296e302.
the later analyses suggest that the changes in synaptic function, i.e., [44] R.A. Yokel, J. Alzheimers Dis. 10 (2006) 223e253.
progressive decrease in neural plasticity, are critical for cognitive [45] C.J. Frederickson, L.J. Giblin, A. Krezel, D.J. McAdoo, R.N. Muelle, Y. Zeng,
decline and that the changes generally precede loss of neurons in R.V. Balaji, R. Masalha, R.B. Thompson, C.A. Fierke, J.M. Sarvey,
M. Valdenebro, D.S. Prough, M.H. Zornow, Exp. Neurol. 198 (2006) 285e293.
normal aging (Fig. 2) [122,123]. The changes in synaptic function,
[46] S. Ueno, M. Tsukamoto, T. Hirano, K. Kikuchi, M.K. Yamada, N. Nishiyama,
which lead to cognitive decline, may be dynamically linked to T. Nagano, N. Matsuki, Y. Ikegaya, J. Cell Biol. 158 (2002) 215e220.
intracellular Zn2þ dyshomeostasis. It is important to analyze Zn2þ- [47] A. Minami, N. Sakurada, S. Fuke, K. Kikuchi, T. Nagano, N. Oku, A. Takeda,
J. Neurosci. Res. 83 (2006) 167e176.
mediated cognitive decline in the process of aging, based on evi-
[48] A. Takeda, H. Tamano, T. Ogawa, S. Takada, M. Ando, N. Oku, M. Watanabe,
dence of the possible involvement of Zn2þ in age-related cognitive Behav. Brain Res. 226 (2012) 259e264.
decline [32,36,124] (Fig. 3). A primary important issue that remains [49] A. Takeda, H. Tamano, Mol. Neurobiol. (2016, Mar 16).
to be addressed is how homeostasis of extracellular and intracel- [50] A. Takeda, M. Suzuki, M. Tempaku, K. Ohashi, H. Tamano, Neuroscience 304
(2015) 209e216.
lular Zn2þ is controlled in the hippocampus and how it is altered [51] P. Paoletti, A.M. Vergnano, B. Barbour, M. Casado, Neuroscience 158 (2009)
along with aging. 126e136.
[52] M. Khan, C.R. Goldsmith, Z. Huang, J. Georgiou, T.T. Luyben, J.C. Roder,
S.J. Lippard, K. Okamoto, Proc. Natl. Acad. Sci. U. S. A. 111 (2014) 6786e6791.
References [53] S.L. Sensi, L.M.T. Canzoniero, S.P. Yu, H.S. Ying, J.Y. Koh, G.A. Kerchner,
D.W. Choi, J. Neurosci. 15 (1997) 9554e9564.
[1] M.Y. Jou, A.G. Hall, A.F. Philipps, S.L. Kelleher, B.J. Lo €nnerdal, Nutrition 139 [54] R.A. Colvin, A.I. Bush, I. Volitakis, C.P. Fontaine, D. Thomas, K. Kikuchi,
(2009) 835e841. W.R. Holmes, Am. J. Physiol. Cell Physiol. 294 (2008) C726eC742.
[2] J. Dufner-Beattie, Y.M. Kuo, J. Gitschier, G.K. Andrews, J. Biol. Chem. 279 [55] J.Y. Lee, J.S. Kim, H.R. Byun, R.D. Palmiter, J.Y. Koh, Brain Res. 1418 (2011)
(2004) 49082e49090. 12e22.
[3] R.J. Cousins, J.P. Liuzzi, L.A. Lichten, J. Biol. Chem. 281 (2006) 24085e24089. [56] Y. Li, C.J. Hough, C.J. Frederickson, J.M. Sarvey, J. Neurosci. 21 (2001)
[4] T. Fukada, T. Kambe, Metallomics 3 (2011) 662e674. 8015e8025.
[5] C.O. Hershey, L.A. Hershey, A. Varnes, S.D. Vibhakar, P. Lavin, W.H. Strain, [57] E. Pan, X.A. Zhang, Z. Huang, A. Krezel, M. Zhao, C.E. Tinberg, S.J. Lippard,
Neurology 33 (1983) 1350e1353. J.O. McNamara, Neuron 71 (2011) 1116e1126.
[6] K. Gellein, J.H. Skogholt, J. Aaseth, G.B. Thoresen, S. Lierhagen, E. Steinnes, [58] J. Emmetsberger, M.M. Mirrione, C. Zhou, M. Fernandez-Monreal,
T. Syversen, T.P. Flaten, J. Neurol. Sci. 266 (2008) 70e78. M.M. Siddiq, K. Ji, S.E. Tsirka, J. Neurosci. 30 (2010) 6538e6547.
[7] B. Michalke, V. Nischwitz, Anal. Chim. Acta 682 (2010) 23e36. [59] S.L. Sensi, D. Ton-That, P.G. Sullivan, E.A. Jonas, K.R. Gee, L.K. Kaczmarek,
[8] J.H. Weiss, S.L. Sensi, J.Y. Koh, Trends Pharmacol. Sci. 21 (2000) 395e401. J.H. Weiss, Proc. Natl. Acad. Sci. U. S. A. 100 (2003) 6157e6162.
[9] W.R. Markesbery, W.D. Ehmann, M. Alauddin, T.I.M. Hossain, Neurobiol. [60] G. Danscher, M. Stoltenberg, J. Histochem. Cytochem 53 (2005) 141e153.
Aging 5 (1984) 19e28. [61] R.A. Colvin, M. Laskowski, C.P. Fontaine, Brain Res. 1085 (2006) 1e10.
[10] A. Takeda, Brain Res. Rev. 34 (2000) 137e148. [62] C.J. Stork, Y.V. Li, J. Mol. Signal 5 (2010) 5.
[11] A. Takeda, BioMetals 14 (2001) 343e352. [63] A. Takeda, H. Tamano, T. Ogawa, S. Takada, M. Nakamura, H. Fujii, M. Ando,
[12] A. Takeda, M. Nakamura, H. Fujii, H. Tamano, Metallomics 5 (2013) 417e423. Hippocampus 24 (2014) 1404e1412.
[13] C.J. Frederickson, Int. Rev. Neurobiol. 31 (1989) 145e238. [64] I. Sekler, A. Moran, M. Hershfinkel, A. Dori, A. Margulis, N. Birenzweig,
[14] C.J. Frederickson, G. Danscher, Prog. Brain Res. 83 (1990) 71e84. Y. Nitzan, W.F. Silverman, J. Comp. Neurol. 447 (2002) 201e209.
[15] A. Takeda, H. Tamano, Brain Res. Rev. 62 (2009) 33e34. [65] C. Nolte, A. Gore, I. Sekler, W. Kresse, M. Hershfinkel, A. Hoffmann,
[16] A. Takeda, Mol. Neurobiol. 44 (2011) 167e174. H. Kettenmann, A. Moran, Glia 48 (2004) 145e155.
[17] C.B. Sindreu, H. Varoqui, J.D. Erickson, J. Pe rez-Clausell, Cereb. Cortex 13 [66] P. Aguilar-Alonso, D. Martinez-Fong, N.G. Pazos-Salazar, E. Brambila,
(2003) 823e829. J.A. Gonzalez-Barrios, A. Mejorada, G. Flores, L. Millan-Perezpen ~ a, H. Rubio,
[18] A. Takeda, H. Tamano, Significance of the degree of synaptic Zn2þ signaling in B.A. Leon-Chavez, Brain Res. 1200 (2008) 89e98.
cognition, BioMetals 29 (2016) 177e185. 
[67] C. Sindreu, A. Baye s, X. Altafaj, J. Pe
rez-Clausell, Zinc transporter-1 concen-
[19] H. Karst, S. Berger, M. Turiault, F. Tronche, G. Schütz, M. Joe €ls, Proc. Natl. trates at the postsynaptic density of hippocampal synapses, Mol. Brain 7
Acad. Sci. U. S. A. 102 (2005) 19204e19207. (2014) 16.
[20] L. Musazzi, M. Milanese, P. Farisello, S. Zappettini, D. Tardito, V.S. Barbiero, [68] J. Qian, K. Xu, J. Yoo, T.T. Chen, G. Andrews, J.E. Noebels, Knockout of Zn
T. Bonifacino, A. Mallei, P. Baldelli, G. Racagni, M. Raiteri, F. Benfenati, transporters Zip-1 and Zip-3 attenuates seizure-induced CA1 neuro-
G. Bonanno, M. Popoli, PLoS One 5 (2010) e8566. degeneration, J. Neurosci. 31 (2011) 97e104.
[21] A. Takeda, M. Suzuki, H. Tamano, S. Takada, K. Ide, N. Oku, Neurochem. Int. 60 [69] S.A. Small, S.A. Schobel, R.B. Buxton, M.P. Witter, C.A. Barnes, Nat. Rev.
(2012) 394e399. Neurosci. 12 (2011) 585e601.
[22] A. Takeda, H. Tamano, Front. Aging Neurosci. 6 (2014) 26. [70] N.C. Danbolt, Prog. Neurobiol. 65 (2001) 1e105.
[23] A. Takeda, T. Akiyama, J. Sawashita, S. Okada, Brain Res. 640 (1994) 341e344. [71] X.X. Dong, Y. Wang, Z.H. Qin, Acta Pharmacol. Sin. 30 (2009) 379e387.
[24] A. Takeda, J. Sawashita, S. Okada, Brain Res. 658 (1994) 252e254. [72] T.W. Lai, S. Zhang, Y.T. Wang, Excitotoxicity and stroke: identifying novel
[25] L. Belloni-Olivi, C. Marshall, B. Laal, G.K. Andrews, J. Bressler, J. Neurosci. Res. targets for neuroprotection, Prog. Neurobiol. 115 (2014) 157e188.
87 (2009) 3221e3230. [73] J. Lewerenz, P. Maher, Front. Neurosci. 9 (2015) 469.
[26] W. Chowanadisai, S.L. Kelleher, B. Lo €nnerdal, J. Nutr. 135 (2005) 1002e1007. [74] C.J. Frederickson, J.Y. Koh, A.I. Bush, Nat. Rev. Neurosci. 6 (2005) 449e462.
[27] A.S. Nakashima, R.H. Dyck, Brain Res. Rev. 59 (2009) 347e373. [75] S.L. Sensi, P. Paoletti, J.Y. Koh, E. Aizenman, A.I. Bush, M. Hershfinkel, The
[28] A. Takeda, A. Minami, S. Takefuta, M. Tochigi, N. Oku, J. Neurosci. Res. 63 neurophysiology and pathology of brain zinc, J. Neurosci. 31 (2011)
(2001) 447e452. 16076e16085.
[29] B. Szewczyk, Front. Aging Neurosci. 5 (2013) 33. [76] K.F. Bell, D.A. Bennett, A.C. Cuello, Paradoxical upregulation of glutamatergic
[30] S. Sharma, M. Ebadi, Neurochem. Int. 65 (2014) 40e48. presynaptic boutons during mild cognitive impairment, J. Neurosci. 27
[31] J.R. Nuttall, P.I. Oteiza, Genes Nutr. 9 (2014) 379. (2007) 10810e10817.
[32] E. Mocchegiani, C. Bertoni-Freddari, F. Marcellini, M. Malavolta, Prog. Neu- [77] S.R. Pagliusi, P. Gerrard, M. Abdallah, D. Talabot, S. Catsicas, Neuroscience 61
robiol. 75 (2005) 367e390. (1994) 429e433.
[33] T.B. Cole, H.J. Wenzel, K.E. Kafer, P.A. Schwartzkroin, R.D. Palmiter, Proc. Natl. [78] Y.V. Medvedeva, J.H. Weiss, Neurobiol. Dis. 68 (2014) 137e144.
Acad. Sci. U. S. A. 96 (1999) 1716e1721. [79] U. Malairaman, K. Dandapani, A. Katyal, PLoS One 9 (2014) e110253.
[34] J. Qian, J.L. Noebels, J. Physiol. 566 (2005) 747e758. [80] F. Colbourne, S.Y. Grooms, R.S. Zukin, A.M. Buchan, M.V. Bennett, Proc. Natl.
[35] P.A. Adlard, J.M. Parncutt, D.I. Finkelstein, A.I. Bush, J. Neurosci. 30 (2010) Acad. Sci. U. S. A. 100 (2003) 2906e2910.
1631e1636. [81] K.M. Noh, H. Yokota, T. Mashiko, P.E. Castillo, R.S. Zukin, M.V. Bennett, Proc.
[36] P.A. Adlard, A. Sedjahtera, L. Gunawan, L. Bray, D. Hare, J. Lear, P. Doble, Natl. Acad. Sci. U. S. A. 102 (2005) 12230e12235.
A. Takeda, H. Tamano / Archives of Biochemistry and Biophysics 611 (2016) 93e99 99

[82] C.J. Stork, Y.V. Li, J. Cereb, Blood Flow. Metab. 29 (2009) 1399e1408. [104] A. Takeda, H. Tamano, S. Imano, N. Oku, Neuroscience 168 (2010) 715e722.
[83] A. Co ^ te
, M. Chiasson, M.R. Peralta 3rd, K. Lafortune, L. Pellegrini, K. To  th, [105] D.S. Kerr, L.W. Campbell, O. Thibault, P.W. Landfield, Proc. Natl. Acad. Sci. U.
J. Physiol. 566 (2005) 821e837. S. A. 89 (1992) 8527e8531.
[84] A. Takeda, H. Tamano, A. Nagayoshi, K. Yamada, N. Oku, Neurochem. Int. 47 [106] A. Bhargava, O.C. Meijer, M.F. Dallman, D. Pearce, J. Neurosci. 20 (2000)
(2005) 539e544. 3129e3138.
[85] S. Liu, L. Lau, J. Wei, D. Zhu, S. Zou, H.S. Sun, Y. Fu, F. Liu, Y. Lu, Neuron 43 [107] A.Z. Hansen, T. Zeuthen, Acta Physiol. Scand. 113 (1981) 437e445.
(2004) 43e55. [108] D. Liu, J.R. Slevin, C. Lu, S.L. Chan, M. Hansson, E. Elme r, M.P. Mattson,
[86] J.H. Weiss, Front. Mol. Neurosci. 4 (2011) 42. J. Neurochem. 86 (2003) 966e979.
[87] R. Schmidt-Kastner, K.A. Hossmann, Acta Neuropathol. 76 (1988) 411e421. [109] A. Takeda, S. Takada, M. Nakamura, M. Suzuki, H. Tamano, M. Ando, N. Oku,
[88] J.H. Kim, B.G. Jang, B.Y. Choi, L.M. Kwon, M. Sohn, H.K. Song, S.W. Suh, PLoS PLoS One 6 (2011) e28615.
One 7 (2012) e48543. [110] M. Suzuki, Y. Fujise, Y. Tsuchiya, H. Tamano, A. Takeda, Neurochem. Int. 87
[89] E. Gascon, K. Lynch, H. Ruan, S. Almeida, J.M. Verheyden, W.W. Seeley, (2015) 60e65.
D.W. Dickson, L. Petrucelli, D. Sun, J. Jiao, H. Zhou, M. Jakovcevski, [111] J. Bengzon, Z. Kokaia, E. Elmer, A. Nanobashvili, M. Kokaia, O. Lindvall, Proc.
S. Akbarian, W.D. Yao, F.B. Gao, Nat. Med. 20 (2014) 1444e1451. Natl. Acad. Sci. U. S. A. 94 (1997) 10432e10437.
[90] C. Sandi, Trends Neurosci. 34 (2011) 165e176. [112] B.Y. Choi, J.H. Kim, H.J. Kim, B.E. Lee, I.Y. Kim, M. Sohn, S.W. Suh, J. Trace Elem.
[91] P.W. Landfield, J.C. Eldridge, Neurobiol. Aging 15 (1994) 579e588. Med. Biol. 28 (2014) 474e481.
[92] E. Ferrari, D. Casarotti, B. Muzzoni, N. Albertelli, L. Cravello, M. Fioravanti, [113] J.H. Kim, B.G. Jang, B.Y. Choi, L.M. Kwon, M. Sohn, H.K. Song, S.W. Suh, PLoS
S.B. Solerte, F. Magri, Brain Res. Rev. 37 (2001) 294e300. One 7 (2012) e48543.
[93] N.C. Nicolaides, E. Kyratzi, A. Lamprokostopoulou, G.P. Chrousos, [114] S.W. Suh, S.J. Won, A.M. Hamby, Y. Fan, C.T. Sheline, H. Tamano, A. Takeda,
E. Charmandari, Neuroimmunomodulation 22 (2015) 6e19. J. Liu, J. Cereb. Blood Flow. Metab. 29 (2009) 1579e1588.
[94] F. Magri, L. Cravello, L. Barili, S. Sarra, W. Cinchetti, F. Salmoiraghi, G. Micale, [115] E.C. Cope, D.R. Morris, S.D. Gower-Winter, N.C. Brownstein, C.W. Levenson,
E. Ferrari, Stress and dementia: the role of the hypothalamicpituitary- Exp. Neurol. 279 (2016) 96e103.
adrenal axis, Aging Clin. Exp. Res. 18 (2006) 167e170. [116] J.R. Nuttall, S. Supasai, J. Kha, B.M. Vaeth, G.G. Mackenzie, A.M. Adamo,
[95] J.G. Csernansky, H. Dong, A.M. Fagan, L. Wang, C. Xiong, D.M. Holtzman, P.I. Oteiza, J. Nutr. Biochem. 26 (2015) 1116e1123.
J.C. Morris, Am. J. Psychiatry 163 (2006) 2164e2169. [117] C. Mirescu, E. Gould, Hippocampus 16 (2016) 233e238.
[96] J.T. O’Brien, D. Ames, I. Schweitzer, P. Colman, P. Desmond, B. Tress, Br. J. [118] J.S. Snyder, A. Soumier, M. Brewer, J. Pickel, H.A. Cameron, Nature 476 (2011)
Psychiatry 168 (1996) 679e687. 458e461.
[97] M. Kawata, K. Yuri, H. Ozawa, M. Nishi, T. Ito, Z. Hu, H. Lu, M. Yoshida, [119] H.K. Kim, P.V. Nunes, K.C. Oliveira, L.T. Young, B. Lafer, Prog. Neuro-
J. Steroid Biochem. Mol. Biol. 65 (1998) 273e280. psychopharmacol. Biol. Psychiatry 67 (2016) 51e57.
[98] M. Joe €ls, Eur. J. Pharmacol. 583 (2008) 312e321. [120] E.A. Meyers, K.T. Gobeske, A.M. Bond, J.C. Jarrett, C.Y. Peng, J.A. Kessler,
[99] M. Joe €ls, H. Karst, R. DeRijk, E.R. de Kloet, Trends Neurosci. 31 (2008) 1e7. Neurobiol. Aging 38 (2016) 164e175.
[100] C. Venero, J. Borrell, Eur. J. Neurosci. 11 (1999) 2465e2473. [121] E. Pannese, Brain Struct. Funct. 216 (2011) 85e89.
[101] A. Takeda, N. Sakurada, S. Kanno, A. Minami, N. Oku, J. Neurochem. 99 (2006) [122] C. Freitas, F. Farzan, A. Pascual-Leone, Front. Neurosci. 7 (2013) 42.
670e676. [123] S. Jinno, J. Chem. Neuroanat. (2015) pii: S0891-0618(15) 00094e0.
[102] A. Takeda, H. Tamano, Metallomics 4 (2012) 614e618. [124] M. Warthon-Medina, V.H. Moran, A.L. Stammers, S. Dillon, P. Qualter,
[103] T. Itoh, T. Saito, M. Fujimura, S. Watanabe, K. Saito, Brain Res. 618 (1993) M. Nissensohn, L. Serra-Majem, N.M. Lowe, Eur. J. Clin. Nutr. 69 (2015)
318e322. 649e661.

You might also like