You are on page 1of 17

Pharmacology & Therapeutics 111 (2006) 567 – 583

www.elsevier.com/locate/pharmthera

Associate editor: A.G. Ramage

Zinc and copper: Pharmacological probes and endogenous modulators of


neuronal excitability
Alistair Mathie a,*, Gemma L. Sutton a, Catherine E. Clarke b, Emma L. Veale a
a
Biophysics Section, Blackett Laboratory, Division of Cell and Molecular Biology, Imperial College London, Exhibition Road, London SW7 2AZ, United Kingdom
b
St. Vincent’s Clinical School, University of New South Wales, and Victor Chang Cardiac Research Institute, Sydney, NSW 2010, Australia

Abstract

As well as being key structural components of many proteins, increasing evidence suggests that zinc and copper ions function as signaling
molecules in the nervous system and are released from the synaptic terminals of certain neurons. In this review, we consider the actions of these
two ions on proteins that regulate neuronal excitability.
In addition to the established actions of zinc, and to a lesser degree copper, on excitatory and inhibitory ligand-gated ion channels, we show
that both ions have a number of actions on selected members of the voltage-gated-like ion channel superfamily. For example, zinc is a much more
effective blocker of one subtype of tetrodotoxin (TTX)-insensitive sodium (Na+) channel (NaV1.5) than other Na+ channels, whereas a certain T-
type calcium (Ca2+) channel subunit (CaV3.2) is particularly sensitive to zinc. For potassium (K+) channels, zinc can have profound effects on the
gating of certain KV channels whereas zinc and copper have distinct actions on closely related members of the 2 pore domain potassium channel
(K2P) channel family. In addition to direct actions on these proteins, zinc is able to permeate a number of membrane proteins such as (S)-alpha-
amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA)/kainate receptors, Ca2+ channels and some transient receptor potential (trp)
channels.
There are a number of important physiological and pathophysiological consequences of these many actions of zinc and copper on membrane
proteins, in terms of regulation of neuronal excitability and neurotoxicity. Furthermore, the concentration of free zinc and copper either in the
synaptic cleft or neuronal cytoplasm may contribute to the etiology of certain disease states such as Alzheimer’s disease (AD) and epilepsy.
D 2005 Elsevier Inc. All rights reserved.

Keywords: Zinc; Copper; Voltage-gated-like ion channels; Neuronal excitability; Epilepsy; Alzheimer’s disease

Abbreviations: Ah, amyloid-h protein; AD, Alzheimer’s disease; AMPA, (S)-alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid; Ca2+, calcium; CNS,
central nervous system; CSF, cerebrospinal fluid; GABA, g-aminobutyric acid; IA, transient potassium current; K+, potassium; K2P, 2 pore domain potassium
channel; Na+, sodium; NMDA, N-methyl-d-aspartate; SOCC, store-operated calcium channel; SOD, superoxide dismutase; trp, transient receptor potential; TTX,
tetrodotoxin; ZEN, zinc-enriched neurons.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 568
2. Zinc and copper in the brain. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 568
3. Protein targets . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 570
3.1. How do zinc and copper alter the function of proteins? . . . . . . . . . . . . . . . . 570
3.2. Ligand-gated ion channels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 570
3.2.1. Glutamate receptors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 570
3.2.2. g-Aminobutyric acidA receptors. . . . . . . . . . . . . . . . . . . . . . . . 571
3.2.3. P2X receptors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 571

* Corresponding author. Tel.: +44 20 7594 7691; fax: +44 20 7589 0191.
E-mail address: a.mathie@imperial.ac.uk (A. Mathie).

0163-7258/$ - see front matter D 2005 Elsevier Inc. All rights reserved.
doi:10.1016/j.pharmthera.2005.11.004
568 A. Mathie et al. / Pharmacology & Therapeutics 111 (2006) 567 – 583

3.3. Voltage-gated-like ion channels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 571


3.3.1. Sodium channels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 571
3.3.2. Potassium channels. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 573
3.3.3. Calcium channels. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 575
3.3.4. Store-operated calcium channels and transient receptor potential channels . . 576
4. Physiological consequences of zinc and copper actions . . . . . . . . . . . . . . . . . . . . 577
4.1. Neuronal excitability . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 577
4.2. Neurotoxicity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 578
5. Clinical consequences . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 578
5.1. Alzheimer’s disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 578
5.2. Epilepsy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 579
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 579
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 579

1. Introduction remains protein bound. The binding of zinc to l-histidine in


the plasma and cerebrospinal fluid (CSF) promotes transport
Zinc and copper ions are of key physiological importance in of zinc to target sites, from where its uptake across the
mammalian tissue. Zinc is a vital nutrient and with the blood – brain barrier is tightly regulated. Following uptake,
exception of iron, it is the most abundant trace element in passage of zinc through the CSF and brain extracellular fluid
the body (see Takeda, 2001). Copper is also a vital trace compartments is unrestricted. Entry routes into glia and
element, the third most abundant in humans, and is present at neurons are still to be clarified, although several zinc
low levels in a variety of cells and tissues with the highest transporters have been identified (see below; Mocchegiani et
concentrations in the liver (Gaetke & Chow, 2003). Both ions al., 2005). Even within the brain, 90% of total brain zinc is
have a range of physiologically important roles in humans. bound to zinc metalloproteins, with much of the remaining
They are a key structural component of many proteins and act 10% found in presynaptic vesicles, either loosely bound or
as co-factors for the activity of many enzymes that are critical free (and therefore, histochemically reactive) (see Takeda,
for brain function. These include enzymes involved in 2001). Indeed, it is known that free ionic zinc is a potent
antioxidant defense (superoxide dismutase; SOD) cellular killer of neurons and glia, with prolonged exposure to growth
respiration (cytochrome c oxidase) and catecholamine synthe- media containing in excess of 100 nM leading to cell death
sis (dopamine-h-hydroxylase) and a plethora of other enzymes (see Frederickson et al., 2005).
involved in multiple biological processes required for growth, Like zinc, copper accumulates in the brain. The average
development, and maintenance of the nervous system (Gaetke copper concentration in the CSF has been estimated to be
& Chow, 2003; Barnes et al., 2005, see Frederickson et al., around 70 AM. While, like zinc, most of this is protein bound,
2005). loosely bound copper is estimated at around 0.1 –0.8 AM,
There are increasingly compelling arguments that both whereas the normal extracellular copper concentration in the
ions can also function as signaling molecules, with evidence brain is of the order of 0.2 –1.7 AM (Gutteridge, 1984; Kardos
of release from synaptic terminals and measured actions on a et al., 1989; Linder & Hazegh-Azam, 1996; Stuerenberg, 2000;
wide range of membrane proteins (see Frederickson et al., Schumann et al., 2002; White et al., 2004). However, these
2005). As such, in the nervous system, both ions might be values are frequently exceeded in the synaptic cleft and during
predicted to have modulatory roles in regulating neuronal neurodegenerative disease where concentrations of copper may
excitability. In this review, we will consider the roles of zinc reach 200 AM and 400 AM, respectively (White & Cappai,
and copper ions in the nervous system, both in terms of their 2003).
physiological actions and in terms of their potential use as Zinc is not uniformly distributed about the brain. Higher
pharmacological mediators of proteins that regulate neuronal concentrations are present in the grey than white matter, while
excitability. the highest concentrations are located in specific forebrain
regions including the hippocampus, amygdala and neocortex
2. Zinc and copper in the brain (Slomianka et al., 1990, see Frederickson & Moncrieff, 1994;
Takeda, 2000). Histochemically stainable zinc (free plus
In the plasma, zinc and copper are present at concentra- loosely bound zinc) is found in particularly high concentrations
tions of around 15 AM. For zinc, almost all of this is bound to in certain synaptic vesicles. Neurons that contain such vesicles
proteins such as albumin, hence free, ionic, zinc is in the have been termed zinc-enriched neurons (ZEN). It is clear that
nanomolar range (see Takeda, 2001). The brain has the these neurons are not associated with a single ‘‘primary’’
highest zinc content compared to other organs (see Mocche- synaptic neurotransmitter. For example, GABAergic ZEN
giani et al., 2005) about 10-fold higher than that found in terminals have been found in the cerebellum (Wang et al.,
plasma (around 100– 150 AM). This has been localized to 2002), whereas in the cerebral cortex, amygdalar nuclei,
several specific brain regions; however, the vast majority olfactory bulb, and hippocampal formation, ZEN terminals
A. Mathie et al. / Pharmacology & Therapeutics 111 (2006) 567 – 583 569

are glutamatergic (see Frederickson & Bush, 2001). This has Of the 3 methods, it is clear that the third method is the most
given rise to the term ‘‘gluzinergic’’ neurons, used by some. direct, the fastest, and the most informative. For adult
The hippocampus is particularly rich in gluzinergic neurons in preparations at least, fluorescent zinc probes reveal clear
regions such as the dentate gyrus, granule cell mossy fibers, increases in synaptic zinc following nerve stimulation and
and in CA3 and CA1 neurons. Zinc-containing fibers from estimates of concentration in the range 10 – 30 AM (e.g.
hippocampal region innervate the cerebral cortex, amygdala, Thompson et al., 2002; Li et al., 2003). Not all studies,
striatum, or limbic regions (see Mocchegiani et al., 2005). however, have been able to demonstrate clear evidence of
Again, just like zinc, copper is found to have a differential synaptic release of zinc (Kay, 2003). So, at present, this issue is
distribution in the central nervous system (CNS) with certain not fully resolved.
synaptic vesicles showing particularly high levels of copper. In addition to synaptic release from neurons, there are a
Copper is also primarily associated with glutaminergic or number of other zinc-secreting cells in the CNS. Zinc can be
adrenergic neurons, particularly in regions such as the hippo- secreted from brain capillary endothelial cells and choroidal
campus, olfactory bulb, and locus coeruleus (Sato et al., 1994; epithelial cells to brain extracellular fluid and the CSF,
Ono & Cherian, 1999; see also Kardos et al., 1989). For now, at although the mechanisms underlying this are unclear at present
least, we have been spared the term ‘‘glucupergic’’ neurons. (see Takeda, 2001). Both zinc and copper are essential for
Giant boutons of hippocampal mossy fibers contain ¨ 300 – correct development and functioning of the brain. Furthermore,
350 AM of zinc (see Takeda, 2001). Release of zinc occurs both zinc and copper levels in the immature brain increase with
from presynaptic, small clear round vesicles within neurons age until adulthood when a constant concentration is main-
(see Frederickson et al., 2000). Pools of zinc and copper can be tained (Tarohda et al., 2004, see Fig. 1).
released following membrane depolarization or neural activity When zinc concentrations rise inside either neurons or glial
in a calcium (Ca2+)-dependent manner (Assaf & Chung, 1984; cells, the ions can be expelled from the cell or else concentrated
Howell et al., 1984; Hartter & Barnea, 1988; Kardos et al., in synaptic vesicles by ZnT transporters, taken up by
1989; Horning & Trombley, 2001). Atomic absorption mitochondria or incorporated into zinc binding proteins. A
spectroscopy has also demonstrated the evoked release of zinc family of zinc transporters are involved in transporting zinc
from hippocampal mossy fibers (Charton et al., 1985). across cell membranes (Colvin et al., 2003). In terms of the
A recent review by Frederickson et al. (2005) has illustrated nervous system, perhaps the most interesting of these are ZnT3
how 3 primary methods have been used to show synaptic and ZnT4, which are required for the transport of zinc into
release of zinc. These are imaging of zinc in boutons to show synaptic vesicles. The distribution of ZnT3, in particular, has
depletion following nerve stimulation; detection of zinc in the been widely studied and its presence is taken to be evidence in
perfusate following nerve stimulation; and direct imaging of favor of synaptic release of zinc in such neurons (see Harris,
released zinc using fluorescent probes. 2002).

30 mm
Copper

Zinc

1 3 5 7 14 21 42 77 147 days

Low High
Fig. 1. Regional distribution map of copper and zinc in the brain of rats at various developmental stages. For each ion the maximum concentration observed is in red
and the minimum (0 ppm) in blue. For zinc the maximum level observed was 41.9 Ag g 1, whereas for copper it was 3.95 Ag g 1. The different regions of the brain
(top panel) are separated as follows: (A) visual cortex, (B) somatosensory cortex, (C) motor cortex, (D) granular insular cortex, (E) piriform cortex, (F) olfactory
bulb, (G) putamen (striatum), (H) olfactory tubercle, (I) corpus callosum, (J) external cortex of inferior colic, (K) hippocampus, (L) substantia nigra, (M) internal
capsule cerebral peduncle, (N) thalamic nucleus, (O) amygdaloid area, (P) globus pallidus, (Q) thalamic nucleus, (R) mesencephalic nucleus. Adapted from Tarohda
et al. (2004) with kind permission of Springer Science and Business Media.
570 A. Mathie et al. / Pharmacology & Therapeutics 111 (2006) 567 – 583

While relatively little is currently known about copper because much work has suggested that members of these
transport in the CNS, copper-transporting ATPases (ATP7A protein families are significant synaptic targets for zinc and
and ATP7B) play a central role in distribution of copper (Puig copper, with resultant consequences for synaptic transmission.
& Thiele, 2002). Genetic mutations in ATP7A and 7B lead to
severe neurodegenerative disorders, Menkes and Wilson 3.2. Ligand-gated ion channels
diseases, respectively (Strausak et al., 2001). These 2 proteins
are distributed in a cell-specific manner in accordance with The most well-studied membrane proteins regulated by zinc
their distinct functional properties. Furthermore, a recent (and to a lesser extent copper) are the receptors for fast
study by Barnes et al. (2005) demonstrated that cerebellar excitatory (glutamate) and fast inhibitory (g-aminobutyric acid;
expression of the 2 copper ATPases is regulated individually GABA) transmission in the CNS. Many of the details of these
during development. ATP7B is constantly localized to regulations have been worked out over the last few years (see
Purkinje neurons, allowing steady transport of copper to Smart et al., 2004) to the extent that, for some receptor
ceruloplasmin. However, the expression profile of ATP7A is subunits, the exact amino acids zinc binds to, have been
variable such that during early development expression occurs identified (e.g., Hosie et al., 2003). There is strong evidence
in Purkinje neurons, while during late development and into from studies in hippocampal mossy fibers that synaptically
adult life expression switches to Bergmann glia. Current data released zinc can modulate both excitatory and inhibitory
indicate that ATP7A is responsible for the overall supply of synaptic transmission under physiological conditions (Vogt et
copper to the brain due to its presence in the choroid plexus al., 2000; Ruiz et al., 2004).
(Barnes et al., 2005). Intracellular copper homeostasis is
mediated by copper-induced trafficking to the plasma 3.2.1. Glutamate receptors
membrane (ATP7A) or to vesicles (ATP7B) followed by Excitatory amino acid receptors are the mediators of synaptic
expulsion of excess copper from cells (Llanos & Mercer, transmission at many synapses that can undergo use-dependent
2002). modifications of synaptic efficiency. Ionotropic excitatory
amino acid receptors can be divided into 2 large families, the
3. Protein targets N-methyl-d-aspartate (NMDA) and the (S)-alpha-amino-3-
hydroxy-5-methyl-4-isoxazole propionic acid (AMPA)/kainate
3.1. How do zinc and copper alter the function of proteins? receptor family.
With regard to NMDA receptors, it is well established that
Zinc binds directly to proteins to alter their function. It both zinc and copper are potent inhibitors (Peters et al., 1987;
particularly targets histidine, cysteine, aspartate, and glutamic Mayer et al., 1989; Trombley & Shepherd, 1996; Vlachova et
acid residues. Unlike the situation for zinc, there are several al., 1996; Trombley et al., 1998). While the copper binding site
ways in which copper can modify the activity of a protein. has not yet been elucidated, the inhibition of NMDA receptor
Copper is therefore potentially much more complicated than currents by zinc has been shown to be mediated by 2 separate
zinc and perhaps much more interesting, at least from a mechanisms, a highly sensitive (low nanomolar range) voltage-
mechanistic perspective. Like zinc, it may bind directly to an independent site on the NR2A subunit and a less sensitive
amino acid (again most likely cysteine, histidine or glutamic (AM) voltage-dependent site on the NR2B subunit (Paoletti et
acid residues) to alter protein function. However, because it is a al., 1997; Rachline et al., 2005).
redox metal a second action may be to bind to cysteine residues AMPA/kainate receptors are also blocked by copper in the
and oxidize them. This may catalyse the formation of low AM range, with binding again proposed to occur at 2
disulphide bonds between physically adjacent cysteine residues separate binding sites (Weiser & Wienrich, 1996). Unlike
thereby changing protein function. A third more indirect way copper, zinc elicits biphasic current responses from AMPA/
that copper can modulate protein function is through the kainate receptors that are characterized by potentiation at low
generation of free radicals, which can profoundly alter protein concentrations (50 AM) and inhibition at high concentrations
and cell function (see Section 4). (1 mM). However, a recent study by Blakemore and Trombley
It is beyond the scope of this review to detail all the effects (2004) has highlighted the existence of a zinc-insensitive
of these 2 ions on all membrane proteins. Instead we will focus population of AMPA/kainate receptors, in addition to the well-
on the actions of these ions on proteins that are of particular documented zinc-sensitive AMPA/kainate receptor population
relevance for the control of neuronal excitability and/or where within the rat olfactory bulb. Application of zinc to the
zinc and copper may serve as useful pharmacological probes in insensitive population of AMPA/kainate receptors produced
the absence of other more selective compounds. As such, we uniphasic, inhibitory responses or occasionally had no effect at
will focus on the actions of these compounds on neuronal ion all. The differential effects of zinc on AMPA/kainate receptors
channels; members of the voltage-gated-like ion channel have primarily been attributed to the varying subunit composi-
superfamily (see Yu & Catterall, 2004); in particular sodium tions expressed by individual receptors.
(Na+), potassium (K+) and calcium channels. Before doing so, Copper reduces the efficacy of kainate at the AMPA
however, it is important to summarize the actions of these ions receptor (Weiser & Wienrich, 1996) whereas zinc significantly
on members of the ligand-gated ion channel superfamily, in increases the kinetics and specific binding of AMPA (Bresink
particular glutamate, GABAA, and purinergic P2X receptors et al., 1996). This ability of both copper and zinc to change
A. Mathie et al. / Pharmacology & Therapeutics 111 (2006) 567 – 583 571

AMPA receptor properties may be relevant to neurotoxicity Trace metals have been known to enhance the cationic
associated with AMPA receptor activation. currents elicited by most extracellular excitatory ATP receptors
As well as agonist and antagonist actions on excitatory in native tissues, including those in rat superior cervical
amino acid receptors, it has been demonstrated that zinc is ganglion (Cloues et al., 1993; Cloues, 1995) nodose and
permeable through these receptors (Sensi et al., 1997; Marin et coeliac ganglion neurons (Li et al., 1993) and PC12 cells
al., 2000; Jia et al., 2002, see also Fig. 5). The Ca2+-permeable (Koizumi et al., 1995). However, not all P2X receptors respond
subtype of AMPA/kainate receptors is a primary route of zinc to zinc in the same manner. While the activity of the P2X2 and
entry/uptake into neurons (Sensi et al., 1997) with maximum P2X3 receptors is potentiated by zinc (Nakazawa & Ohno,
translocation expected during intense neuronal activity. Related 1997; Wildman et al., 1998; Wildman et al., 1999) as expected
to this, NMDA receptor activation has been proposed to from studies in native tissues, 1– 10 AM copper or zinc has
regulate copper homeostasis in hippocampal neurons through been found to inhibit the activity of homomeric P2X7 receptors
the release of copper via translocation of the copper transporter (Virginio et al., 1997; Coddou et al., 2002) or P2X1 receptors
ATP7A to hippocampal neuronal processes (Schlief et al., (Wildman et al., 1999).
2005). P2X2 receptor currents are also potentiated with equal
affinity by copper (Xiong et al., 1999), whereas zinc and
3.2.2. c-Aminobutyric acidA receptors copper differentially modulate the P2X4 receptor. While zinc
After glutamate receptors, GABAA receptors have been potentiates P2X4 receptor ATP-gated currents, copper inhibits
studied most extensively for zinc and (to a lesser extent) copper them in a time- and concentration-dependent manner (Soto et
sensitivity (Smart et al., 1994; Trombley & Shepherd, 1996; al., 1996; Xiong et al., 1999; Acuna-Castillo et al., 2000,
Hosie et al., 2003). While normal synaptic GABAA receptors Coddou et al., 2003).
are thought to be relatively zinc-insensitive, tonic GABAA Although relatively little is known about the P2X6 receptor
receptors have a much higher sensitivity to block. This occurs (see North, 2002), increased expression of the P2X6 purinergic
because different GABAA receptor subunit combinations have receptor has been demonstrated in the hippocampus of zinc
different zinc sensitivities. For example, the a1h3 splice diet-restricted rats (Chu et al., 2003). Although the role the
variant of the GABAA receptor is most sensitive to block by P2X6 receptor plays in the physiological response of the
zinc, with other a and h variants having lower sensitivity. hippocampus to zinc depletion remains to be determined, it is
GABAA receptors that contain g subunits have greatly reduced known that the P2X4 and the P2X6 receptors co-assemble in
sensitivity due to the interposition of the g subunit and vitro, forming functional heteromeric channels (Le et al., 1998)
structural changes at the a –h interface site (Hosie et al., and that this heteromer is also potentiated by micromolar zinc.
2003). GABAA receptors containing g subunits are by far the
most prevalent subtypes found at GABAergic synapses. The 3.3. Voltage-gated-like ion channels
effects of endogenous zinc on GABAA receptors has also been
shown and modulation of GABAA receptors by zinc is 3.3.1. Sodium channels
probably a vital factor in normal brain function (e.g., Xie & Voltage-gated sodium (Na+) channels are present in the
Smart 1991), but this probably occurs through extrasynaptic membrane of most excitable cells. There are 9 primary a
rather than synaptic GABAA receptors and/or following subunits (NaV1.1 – NaV1.9) which show differential expression
changes in GABAA receptor composition in disease states throughout the body (see Alexander et al., 2004). The majority
such as epilepsy (Dudek, 2001). Again, much less information of Na+ channels are highly sensitive to block by tetrodotoxin
is available for copper effects. At least in some cases, copper is (TTX) but some are much less sensitive (see Goldin, 2001).
thought to block GABAA receptors through the same mech- For example, NaV1.5, the so-called ‘‘cardiac’’ Na+ channel, and
anism as zinc (Narahashi et al., 1994; Trombley & Shepherd, NaV1.8 and NaV1.9 (expressed particularly highly in peripheral
1996; Sharanova et al., 2000). nociceptive neurons) are much less sensitive to TTX than other
Inhibitory glycine receptors are also modulated by zinc; Na+ channels.
however, zinc has a biphasic effect on these receptors Modulation of Na+ channels by zinc has been widely
producing a potentiation of response at low concentrations studied. Gilly and Armstrong (1982a) showed that millimolar
(Laube et al., 2000) but an inhibition at higher concentrations concentrations of external zinc modified the kinetics of squid
(Laube et al., 1995). giant axon Na+ currents. Despite the low potency of this effect,
it has been suggested to arise from zinc binding to a specific
3.2.3. P2X receptors site within the channel rather than from a screening of negative
The P2X receptors are a family of ionotropic receptors that surface charges (e.g., Tanguy & Yeh, 1988; Hank & Sheets,
are widely distributed in the brain, peripheral nerves, and many 1992).
other cell types, existing as both homomers and heteromers. Thus, initially, zinc was envisaged as being a rather weak
There are currently 7 members of the P2X family classed as modulator of Na+ channel activity. However Frelin et al. (1986)
P2X1 – P2X7 (see review by North, 2002). The co-localization compared the sensitivity of TTX-sensitive and TTX-resistant
of zinc with P2X receptors in the nervous system suggests a (cardiac) Na+ channels to zinc. TTX-resistant channels were
physiological role in zinc modulation of ATP-evoked currents potently inhibited by zinc with an IC50 of 50 AM, compared to
(Nicke et al., 1998; Kanjhan et al., 1999). TTX-sensitive channels with an IC50 of 2 mM. Similar effects
572 A. Mathie et al. / Pharmacology & Therapeutics 111 (2006) 567 – 583

on cardiac Na+ channels were seen by others (Baumgarten & Although NaV1.5 channels are often described as ‘‘cardi-
Fozzard, 1989; Ravindran et al., 1991; Schild & Moczy- ac’’ sodium channels, they are also expressed in neurons.
dlowski, 1991; Schild et al., 1991). It was suggested that a site White et al. (1993) identified both TTX-sensitive and TTX-
related to the TTX binding site may be important and that resistant Na+ channels in acutely dissociated neurons from
cysteine residues played a role (Schild et al., 1991; also Kurata the medial entorhinal cortex of rat. The TTX-resistant
et al., 1998). channel, like cardiac Na+ channels, was highly sensitive to
Following elucidation of the sequences of voltage-gated Na+ zinc, with an IC50 of around 9 AM (Fig. 2B). This high
channels, Satin et al. (1992) identified a key cysteine residue sensitivity to zinc may be of physiological importance
(C374) in TTX-resistant cardiac Na+ channels (see Fig. 2A). considering that zinc is highly localized in the synaptic
When this cysteine residue was mutated, TTX sensitivity was terminals of the entorhinal cortex (see Harrison & Gibbons,
induced. The mutation also lowered sensitivity to cadmium 1994).
(suggesting a lowered sensitivity to zinc also). The converse However, not all TTX-resistant Na+ channels are highly
experiments on TTX-sensitive channels confirmed these sensitive to zinc. Kuo et al. (2004) have shown that zinc can
observations. Mutation of the corresponding residue of the block TTX-resistant Na+ channels (NaV1.8 and NaV1.9) in
brain Na+ channel to a cysteine (as is present in the cardiac Na+ dorsal root ganglion neurons but that this occurs at relatively
channel) reduced the sensitivity of the brain Na+ channel to high concentrations (IC50 of around 300 AM). This may reflect
TTX (Backx et al., 1992; Heinemann et al., 1992) and the fact that these channels (NaV1.8 and NaV1.9) have a serine
increased its sensitivity to zinc (Heinemann et al., 1992). residue in place of the key cysteine residue present in cardiac

Na channel Gene Region Sequence TTX Zinc

“Brain” NaV1.1 374-389 S L F R L M T Q D F W E N LY Q √ X

“Cardiac” NaV1.5 365-380 A L F R L M T Q D C W E R LY Q X √

“PNS” NaV1.8 347-362 S L F R L M T Q D S W E R LY Q X X

B
0

-500 zinc
I (pA)

-1000 wash

control
-1500
-40 0 40
V (mV)

IC50 = 9.1 μM
I (norm)

0.5 zinc

control 1 nA
0 5 ms

0.1 1 10 100
zinc (μM)

Fig. 2. (A) Potent zinc block of NaV channels depends on a critical cysteine residue found exclusively in the ‘‘cardiac’’ NaV1.5 channel. This residue is not present in
‘‘brain’’ Na+ channels such as NaV1.1 or peripheral nervous system ‘‘PNS’’ Na+ channels such as NaV1.8. (B) These ‘‘cardiac’’ channels are present in certain neurons
and the currents are potently blocked by zinc (at a concentration of 100 AM in the examples shown). (B) Adapted from Neuron, volume 11, White, J.A., Alonso, A.,
and Kay, A.R., A heart-like Na+ current in the medial entorhinal cortex, pp. 1037 – 1047, 1993, with permission from Elsevier.
A. Mathie et al. / Pharmacology & Therapeutics 111 (2006) 567 – 583 573

NaV1.5 channels (or phenylalanine in TTX-sensitive channels) and activation as seen in these studies can lead to either an
giving reduced sensitivity to both TTX and zinc (Fig. 2A). apparent increase or decrease in IA, depending on the holding
Taken together, it seems that only NaV1.5 channels are highly potential of the cell. Similar effects have been seen on IA with
sensitive to block by zinc. other metal ions (e.g., Mayer & Sugiyama, 1988; Watkins &
Mathie, 1994). Physiologically, IA modulates action potentials
3.3.2. Potassium channels by increasing both the rate of action potential repolarization
Potassium (K+) channels play a key role in a number of and accommodation. An apparent increase in IA as a result of
different aspects of the electrical responses of the nervous zinc action would likely decrease action potential frequency.
system. K+ channel activity determines neuronal action Horning and Trombley (2001) considered the excitatory
potential frequency and waveform and regulates the excitability effect of both zinc and copper on rat olfactory bulb neurons.
of individual neurons (see Hille, 2001). This physiological They found that zinc (100 AM) produced modest but biphasic
importance coupled with their diversity (with well over 70 effects on voltage-gated K+ currents, potentiating peak current
different a subunits expressed in the mammalian nervous amplitudes of an IA current by 17% (presumably by shifting the
system) (see Coetzee et al., 1999; Alexander et al., 2004) voltage dependence of inactivation as above) but inhibiting
makes them fundamental regulators of neuronal excitability. It steady-state current amplitudes of delayed rectifier-type cur-
is important then to understand the nature and consequences of rents by 15%. Copper (30 AM), however, inhibited both peak
actions of compounds such as zinc and copper on these and steady-state amplitudes of delayed rectifier-type currents
channels. by an average of 20% and 17%, respectively. Thus, copper and
zinc can differentially influence neuronal excitability and
3.3.2.1. Voltage-gated potassium channels. As for Na+ synaptic transmission in the rat olfactory bulb.
channels, modulatory effects of divalent metal ions such as For many K+ channels, the site(s) of action of zinc and
zinc and copper on the gating of K+ channels are well copper remains to be determined. However, Kehl et al. (2002)
characterized (Gilly & Armstrong, 1982b; Spires & Begen- have shown that inhibition of human KV1.5 channels expressed
isich, 1990; Davidson & Kehl, 1995). In a number of studies, in HEK293 cells by both hydrogen ions and zinc was
divalent cation effects have been shown to cause equal shifts in substantially reduced by a mutation of histidine 463 (H) or
the voltage-dependent kinetics of K+ channels, explained by arginine 487 (R), amino acids located either in the channel
surface charge effects (see Hille, 2001; Elinder & Arhem, turret (H) or near the mouth of the pore (R) of the channel.
2003). Again, as for Na+ channels, these effects were seen to A particularly interesting set of experiments was carried out
occur at millimolar concentrations and were originally envis- recently by Cusimano et al. (2004) who found that human
aged as having little physiological relevance. KV1.1 channels were inhibited by zinc with an IC50 of around 3
More recent studies, however, on subtypes of voltage-gated mM. In other words, zinc was a rather poor blocker of these
K+ channels have revealed responses that occur at much lower channels. However, on mutating amino acid F184 to a cysteine,
ion concentrations. Furthermore, there are examples of inhibition by zinc increased, with an IC50 for the mutated
particular K+ channel subtypes that are expressed in neurons, channel of 0.75 mM. Furthermore, a slowing of the activation
showing notable sensitivity to these ions (e.g., Harrison et al., kinetics and a substantial shift in the voltage dependence of
1992; Poling et al., 1996; Horning & Trombley, 2001). For activation was seen in the presence of 30 AM zinc, which was
example, Harrison et al. (1992) looked at the effect of zinc on much larger for the mutant channel compared to wild type.
mouse fibroblasts, transfected with cloned rat KV1.1, human Because this mutation of KV1.1 is associated with a rare
KV1.5, and human KV1.4. They found shifts in the inactivation autosomal dominant neurological disorder known as episodic
and activation curves of each current, at concentrations of zinc ataxia type-1 (EA1), this increased sensitivity to zinc in the
less than 200 AM. At higher concentration of zinc (> 200 AM), mutated channel could have a profound effect on KV1.1
a block of these channels was seen. Related to these function and contribute to the pathogenesis of the disease.
observations, Huang et al. (1993) looked at the effect of zinc
on the voltage-dependent transient potassium current (IA) in 3.3.2.2. Background or ‘‘leak’’ potassium conductances.
acutely dissociated neurons from the suprachiasmatic nucleus. Background K+ currents regulate the resting membrane
They found that zinc caused a shift in the steady-state potential of neurons and are fundamental regulators of neuronal
inactivation of IA, with 30 AM zinc causing a shift in the half excitability (Mathie et al., 2003). As such, compounds that
inactivation by 20 mV. In contrast, copper at 200 AM had little regulate the activity of these channels will have a huge
effect on IA. influence on neuronal excitability. Two pore domain potassium
Subsequent studies support these observations (e.g., Easaw channel (K2P) is thought to underlie such leak conductances in
et al., 1999; Kuo & Chen, 1999). For example, Kuo and Chen many neurons (see Goldstein et al., 2001; Lesage, 2003).
(1999) found that zinc caused a shift in the inactivation curve Leonoudakis et al. (1998) found that the K2P channel
of IA currents in rat hippocampal neurons by 40 mV. TASK-1 was sensitive to zinc with an IC50 of 175 AM. While
Depending on the experimental protocol they employed, zinc this is not a particularly potent effect, initial experiments
was seen to apparently inhibit the channel at hyperpolarized suggested that the related K2P channel, TASK-3, was
potentials yet enhance current at depolarized potentials. Thus, a insensitive to zinc. Because very few pharmacological agents
differential effect on the voltage dependence of inactivation are available to distinguish TASK-1 from TASK-3 channels,
574 A. Mathie et al. / Pharmacology & Therapeutics 111 (2006) 567 – 583

zinc was seen as a useful diagnostic tool to distinguish the two. and has been implicated in neuronal apoptosis (Lauritzen et al.,
A number of subsequent studies identified TASK-like con- 2003). Thus, modulation by a compound such as zinc could be
ductances in native cells and attributed the current to TASK-1 a useful pharmacological tool in the treatment of neurodegen-
channels at least in part on the basis of zinc sensitivity erative and proliferative diseases.
(Hartness et al., 2001; Barbuti et al., 2002; Gurney et al., 2003; In contrast, Kim et al. (2005) found that a different K2P
Cooper et al., 2004; Johnson et al., 2004). channel, TREK-2 was enhanced rather than inhibited by zinc
In fact more recent experiments from our laboratory and with an EC50 of around 90 AM. Related to this, Gruss et al.
elsewhere suggest that TASK-3 channels are much more (2004) had previously found that the K2P channel, TREK-1,
sensitive to block by zinc than TASK-1 with an IC50 of was activated by copper with an EC50 of 3 AM. As for
around 10 – 20 AM depending on the recording conditions TASK-3, zinc inhibited TREK-1, with an IC50 of 3 AM. K2P
used (see Fig. 3; Clarke et al., 2004; Gruss et al., 2004; Kim channels are widely distributed throughout brain and are
et al., 2005). involved in setting the resting membrane potential and
We have found that mutation of a glutamate at position 70 modulating neuronal excitability. The differential effects of
and a histidine at position 98, of TASK-3, left the channel with zinc and copper on different K2P channels are useful
a reduced sensitivity to zinc block. Conversely, mutation of a diagnostically. Furthermore, the influence of these ions on
lysine at position 70 in TASK-1 to a glutamate (as is present in neuronal excitability could be either increased or decreased
TASK-3) induced zinc sensitivity on the TASK-1 channel dependent upon the K2P channel expressed in the neuron of
(Clarke et al., 2004). TASK-3 is highly expressed in the brain interest.

Fig. 3. Zinc sensitivity of TASK K2P K+ channels. (A – C) Zinc is a comparatively selective blocker of TASK-3 potassium channels (at pH 7.4) with little effect on
TASK-2 and TASK-1 at comparable concentrations. (D – F) Block of TASK-3 channels by zinc shows little voltage dependence. From Clarke et al. (2004) with
permission.
A. Mathie et al. / Pharmacology & Therapeutics 111 (2006) 567 – 583 575

3.3.2.3. Other potassium currents. There is much less effect of copper and zinc on recombinant T-type Ca2+ channels
information available about the effects of zinc and copper on (CaV3.1, CaV3.2, CaV3.3). Copper and zinc blocked all 3 T-
other K+ channels, such as inward rectifier K+ channels or type channels but showed a high affinity for CaV3.2 channels
Ca2+-activated K+ channels. Coulter et al. (1995) showed that (IC50 = 0.9 AM and 2.3 AM for copper and zinc, respectively).
the inward rectifier K+ channel HIR (Kir2.3) was weakly Much higher concentrations were required to block CaV3.1
sensitive to block by external zinc, having an IC50 between 100 and CaV3.3 channels (IC50  200 AM, see Fig. 4).
and 200 AM at physiological pH. Another strongly rectifying Utilizing this relatively selective block of CaV3.2 channels
channel IRK1 (Kir2.1), however, was completely insensitive to by zinc, Nikonenko et al. (2005) looked at the neuroprotective
zinc at similar concentrations. Morera et al. (2003) looked at effect of various LVA Ca2+ channel antagonists in a model of in
the effect of copper on the activity of the large conductance vitro ischemia on rat organotypic hippocampal cultures. They
calcium- and voltage-sensitive potassium channels (BKCa). found 1 AM zinc was sufficient to inhibit more than 80% of the
They found that copper concentrations of 20 AM and above T-type current in these neurons, which in turn prevented the
induced a concentration- and time-dependent decrease in the increase in intracellular calcium associated with ischemia and
channel open probability. Zinc, at concentrations up to 100 AM delayed neuronal death.
had no effect on these channels. Copper was shown to act via
oxidation of extracellular cysteine residues, involved in gating 3.3.3.2. L-, N-, P-, and Q-type Ca2+ channels. The L-, N-, P-,
of the channel (Morera et al., 2003). and Q-type (high voltage activated, HVA) voltage-gated Ca2+
channels are, for the most part, as sensitive as T-type Ca2+
3.3.3. Calcium channels channels to inhibition by copper but are generally less sensitive
Voltage-gated calcium (Ca2+) channels can be divided into 3 to zinc inhibition (Nachshen, 1984; Busselberg et al., 1992;
families based on their structural and functional characteristics Vega et al., 1994; Easaw et al., 1999), at least compared to
(Ertel et al., 2000; Alexander et al., 2004) and within each recombinant CaV3.2 channels (Jeong et al., 2003). Kasai and
family there are several different subunits. Functionally, these Neher (1992) showed that N- and L-type Ca2+ channels in
differences are reflected in distinct inactivation kinetics; mouse neuroblastoma and rat glioma hybrid cell line (NG108-
activation and inactivation gating; and single channel con- 15) were particularly sensitive to block by copper with IC50s of
ductances. This diversity allows each type to play a different 7 AM and 14 AM, respectively.
but critical role in aspects of neuronal function. Horning and Trombley (2001) looked at the effect of zinc
and copper on the rat olfactory bulb neurons in primary
3.3.3.1. T-type Ca2+ channels. In terms of block by zinc and cultures. These studies are of particular interest because the
copper ions, T-type or low voltage-activated Ca2+ channels mammalian olfactory bulb has one of the highest concentra-
(LVAs) are arguably the most important group. They constitute tions of zinc and copper in the CNS (Donaldson et al., 1973;
a family of 3 channel isoforms, CaV3.1, CaV3.2, and CaV3.3. Ono & Cherian, 1999). In these neurons, inhibition by zinc
Most brain regions express more than 1 isoform and some (100 AM) of 63% and by copper (30 AM) of 52% was
neurons, such as olfactory granule cells and hippocampal observed.
pyramidal neurons, express all 3 genes (Craig et al., 1999; Kase Similarly, in pyramidal neurons from rat piriform cortex,
et al., 1999; Talley et al., 1999). The activation and inactivation 20 AM zinc inhibited each of the 4 components of HVA
curves of T-type Ca2+ channels overlap and cross at ¨ 60 mV, current (corresponding to L-, N-, P-, and Q-type currents) by
allowing T-type Ca2+ channels to sustain a continuous Ca2+ around 35– 57% whereas copper had an IC50 of less than
influx in neurons and glia due to a small number of channels 1 AM for each component of the HVA current (Castelli et al.,
being continuously open. T-type Ca2+ channels are generally 2003; Magistretti et al., 2003). In the study with zinc, a higher
thought of as a generator of pacemaker activity and/or degree of block was observed when the concentration of the
regulators of hormone and neurotransmitter secretion. They permeant ion (either Ca2+ or barium) was lowered. Thus, the
are also known to contribute to pathophysiological conditions action of zinc is consistent with competitive binding of zinc
such as cardiac hypertrophy and absence epilepsy. and the permeant ion to an extracellular binding site, the
A number of papers have been published on the LVA T-type occupancy of which by zinc results in Ca2+ channel block. It
Ca2+ channel, demonstrating inhibition by micromolar zinc. is worth noting that many studies on zinc block of Ca2+
Akaike et al. (1989) showed that the T-type Ca2+ channel channels may underestimate the effectiveness of the ion
current in rat aorta smooth muscle cells in primary culture because they are done in artificially high concentrations of
was reversibly inhibited by zinc with an IC50 of 30 AM, permeating ion (barium or Ca2+) to allow reliable measure-
while Takahashi and Akaike (1990) showed in CA1 pyrami- ment of current through the channels.
dal cells from acutely isolated rat hippocampal neurons that
zinc inhibited the T-type Ca2+ channel with an IC50 of around 3.3.3.3. Permeation of zinc through voltage-gated Ca chan-
20 AM. Similarly, Busselberg et al. (1992) showed that zinc nels. As well as being blocked by zinc, Ca2+ channels can
was highly specific for T-type Ca2+ channels in rat dorsal root mediate the entry of zinc into neurons, at least under certain
ganglion cells, with 20 AM zinc producing >80% block. N- and recording conditions. For example, Oyama et al. (1982) looked
L-type channels were less potently inhibited by zinc, with an at the effect of zinc on the neurones isolated from the
IC50 of 69 AM. More recently, Jeong et al. (2003) looked at the subesophageal ganglia of Helix aspersa. In magnesium and
576 A. Mathie et al. / Pharmacology & Therapeutics 111 (2006) 567 – 583

Fig. 4. Differential block by copper and zinc of cloned T-type Ca2+ channels. Panel A shows block of individual current traces by copper. Both ions are much more
effective at blocking CaV3.2 channels compared to CaV3.1 or CaV3.3 channels (panels A – C). From Jeong et al. (2003) with permission from Lippincott, Williams &
Wilkins.

Ca2+-free media, 25 mM zinc generated all-or-none action This so-called ‘‘capacitive calcium influx’’ plays an impor-
potentials. Ca2+ antagonists such as verapamil and cobalt tant role in shaping the Ca2+ response of various tissues and
reduced these action potentials. Thus, the voltage-gated Ca2+ cell types. Inhibition by heavy metals is a hallmark of SOCC
channel identified in Helix nerve cell bodies is permeable to activity. The first demonstration of a Ca2+ current that
zinc. Similar zinc permeation through Ca2+ channels has been corresponded with SOCC activity was recorded in rat
observed for mammalian neurons (Sensi et al., 1997; Kerchner peritoneal mast cells and was termed ICRAC (Hoth & Penner,
et al., 2000; Sheline et al., 2002). 1993). One characteristic of ICRAC was inhibition by 1 mM
The fact that zinc both blocks Ca2+ channel current and zinc and this has been borne out by subsequent studies (e.g.,
carries charge through the channel suggests that its interaction Parekh & Penner, 1993; Foskett & Wong, 1994; Koizumi et al.,
with the channel pore may be similar to that of Ca2+, which 1995; Gore et al., 2004). Prothero et al. (1998, 2000) showed
also blocks current through the channel carried by divalent or that a rise in intracellular Ca2+ via SOCCs following activation
monovalent cations. Furthermore, this carrying of zinc by of metabotropic receptors in rat cortical glial cells was
voltage-gated Ca2+ channels supports the notion that blockade powerfully inhibited by 100 AM zinc. More recently, Kresse
of these channels may have therapeutic utility in pathological et al. (2005) showed a similar zinc inhibition of capacitive Ca2+
conditions, such as cardiac arrest or sustained seizures, where entry in mouse hippocampal astrocytes, following activation of
excessive zinc influx may contribute to neuronal death (Choi & the metabotropic receptors, and suggested that the site of action
Koh, 1998; see Fig. 5). of zinc may involve a change in the redox potential, possibly
through an action on cysteine residues in the SOCCs (see also
3.3.4. Store-operated calcium channels Gore et al., 2004).
and transient receptor potential channels Transient receptor potential (trp) channels have been
In addition to voltage-gated Ca2+ channels, another major identified as major pathways for cation movement in non-
route for Ca2+ entry into cells is through store-operated calcium excitable cells (Clapham et al., 2001; Montell et al., 2002;
channels (SOCCs). Ca2+ entry and the subsequent refilling of Vennekens et al., 2002). Cloning of the Drosophila trp channel
intracellular stores in many cells, particularly cells that lack or (Montell & Rubin, 1989) revealed homology to voltage-gated
express small numbers of voltage-gated Ca2+ channels, has Ca2+ channels and it was suggested that this gene encodes a
now been established to occur through such SOCCs. This may SOCC. Interestingly, Petersen et al. (1995) showed that the
be of particular relevance in the CNS when considering the role putative capacitative Ca2+ influx evoked in Xenopus oocytes
of glial cells. by the expression of a trp homologue was blocked by 1 mM
A. Mathie et al. / Pharmacology & Therapeutics 111 (2006) 567 – 583 577

Fig. 5. Zinc release from zinc containing neuron terminals. (A) During normal stimulation, zinc (red circles) is released from presynaptic terminals. It can act on
postsynaptic channel proteins such as GABAA receptors, NMDA receptors, or many other different ion channels to alter their activity. In addition, it may permeate
through certain channel proteins (such as voltage-gated Ca2+ channels, trp channels, or AMPA/kainate receptors as indicated in the schematic) to enter the
postsynaptic cell. (B) Following excessive excitation, zinc is depleted from presynaptic vesicles so less is available for release; however, the intracellular
concentration of zinc in the postsynaptic cell is increased, which may be protective or detrimental to the neuron depending on the particular neuron considered (see
text for further details, developed from an idea of Takeda, 2000).

zinc. More recently, however, there has been much debate over sensitive to zinc modulation than all other glutamate receptors
the exact correlation between functional SOCCs and recombi- (see Chen et al., 1997; Tovar et al., 2000), whereas, for
nant trp channels (see Clapham, 2003; Nilius, 2003). It is GABAA receptors, it is those that lack the g subunit; that is,
possible that block by zinc could provide a useful diagnostic those expressed primarily extra-synaptically, that are most
tool to help in this debate (see Gore et al., 2004). sensitive to zinc. Similarly for Na+ channels, zinc is a much
Like voltage-gated Ca2+ channels, at least some trp more effective blocker of 1 subtype of TTX-insensitive Na+
channels are permeable to zinc. For example, 2 channels of channel (NaV1.5) than other Na+ channels, whereas for Ca2+
the TRPM subfamily, TRPM6 and TRPM7, have been shown channels a certain T-type channel subunit (CaV3.2) is partic-
to be permeable to various divalent cations, including zinc ularly sensitive. For K+ channels, zinc can have profound
(Hermosura et al., 2002; Monteilh-Zoller et al., 2003; Schmitz effects on the gating of certain K V channel subtypes
et al., 2003; Voets et al., 2004). Indeed, Monteilh-Zoller et al. (particularly those that show fast inactivation such as KV1.4),
(2003) showed that TRPM7 was 2-fold more permeant to zinc whereas it displays rather selective blocking actions on closely
than to Ca2+. This suggests that TRPM7, in a similar manner to related members of the K2P channel family (compare the
voltage-gated Ca2+ channels or zinc-permeable AMPA/kainate effects on TASK-3 and TASK-1).
receptors, could potentially play a major role in mediating the There are 2 main consequences of such differential effects
severe neurotoxic effects associated with high levels of zinc in of zinc. Firstly, zinc can provide a useful diagnostic tool to
the brain during ischemia. identify channels in mammalian neurons that underlie ob-
served currents, when other pharmacological tools are lacking.
4. Physiological consequences of zinc and copper actions This has been exemplified recently by the use of zinc to
identify TASK-3 channels as underlying background K+
4.1. Neuronal excitability currents in cerebellar granule cells (see Clarke et al., 2004;
Aller et al., 2005). In a more physiological sense, these
As shown in the previous section, physiologically relevant differences are useful because they allow the possibility of
concentrations of zinc and copper can modulate several ligand- differential responses of neurons to either exogenously applied
gated ion channels including glutamate receptors, GABAA zinc or synaptically released zinc (see Fig. 5), depending on
receptors, and P2X receptors, as well as affecting many the particular ion channels that neuron expresses at a given
members of the voltage-gated-like ion channel family includ- time and where they are localized. KV1.4, for example, has
ing K+, Na+, and Ca2+ channels. With such a wide range of been found highly localized in axons and terminals, with a
actions, it is difficult to predict (and indeed measure) what the possible role in modulating the excitability of nerve terminals.
net effects on neuronal excitability of these ions might be. In the mossy fiber terminals of the hippocampus, high
Nevertheless, a number of striking features emerge from concentrations of zinc are also found and thus zinc may play
studies of these receptors and channels, principally the a physiological role in hippocampal transmission through
differential sensitivity of certain members of each family to regulation of KV1.4. In terms of changes with time, a good
zinc and, although much less thoroughly studied, occasionally example is the altered sensitivity of GABAA receptors to
to copper too. For ligand-gated ion channels, for example, released zinc that can occur in temporal lobe epilepsy (see
NMDA receptors that lack the NR2B subunit are much more below and Dudek, 2001).
578 A. Mathie et al. / Pharmacology & Therapeutics 111 (2006) 567 – 583

4.2. Neurotoxicity mortality rate of these CA3 pyramidal cells (Cote et al., 2005).
Similarly, if the free intracellular zinc is decreased in some
Neurotoxicity can be observed following unwanted rises in neurons, for example, through the use of zinc chelators such as
extracellular copper or zinc concentration. Extracellular levels TPEN, this can trigger apoptosis (Frederickson et al., 2005).
of copper are significantly elevated during aging and some Thus, the intracellular concentration of these ions also needs to
neurodegenerative disorders. The ability of copper to undertake be tightly regulated by neurons.
redox cycling to activate molecular oxygen is used by a variety
of enzymes. Although normally bound to proteins, excess 5. Clinical consequences
copper may be released and become free to catalyze the
formation of highly reactive hydroxyl radicals, implicated in In the previous section, a number of potential physiological
disorders associated with abnormal copper metabolism and and pathophysiological consequences of zinc and copper
neurodegenerative changes (Barnham et al., 2004; Valko et al., actions were considered. It is of interest, finally, to consider
2005). Ironically, one of the most important roles of copper, these ideas in terms of known disease states, specifically
physiologically, is in controlling free radical reactions partic- Alzheimer’s disease (AD) and certain forms of epilepsy.
ularly as part of the enzyme, copper/zinc superoxide dismutase
(SOD1). So, for example, a dietary deficiency of copper 5.1. Alzheimer’s disease
increases cellular susceptibility to oxidative damage. Thus,
either too little or too much copper in the brain can lead to an Alzheimer’s disease (AD) is a progressive neurodegener-
increased vulnerability to oxidative damage. ative disorder characterized by amyloidal plaques and
The brain is particularly susceptible to oxidative stress due neurofibrillary tangles in conjunction with neuronal cell loss
to its high energetic requirement (utilizing 20% basal oxygen or dysfunction. The primary constituent of amyloid deposits
consumption), in addition to the high levels of transition metals associated with all cases of AD is amyloid-h protein (Ah),
and reduced antioxidant defenses compared to other organs derived from the proteolytic cleavage of amyloid precursor
(Maynard et al., 2005). Within the brain, neurons are the most protein. In healthy individuals, Ah is predominantly mem-
metabolically active cells having the greatest oxygen require- brane associated; however, in the AD brain there is a
ment. Thus, SOD expression is most prominent amongst marked augmentation in the proportion of aggregated
neuronal populations consisting of large neurons that are (diffuse and plaque amyloid) and soluble Ah peptides (see
continually vulnerable to oxidative damage (Peluffo et al., Bush, 2003).
2005). In the adult CNS, SOD1 is expressed in numerous A number of recent studies have demonstrated that the
regions, the most notable being hippocampal pyramidal toxicity and aggregation of Ah during AD is promoted by
neurons, granule neurons of the dentate gyrus, cortical neurons aberrant interactions with metals, in particular copper and zinc
(especially pyramidal cells), neurons of the substantia nigra, as (see Maynard et al., 2005). Ah contains selective high and low
well as distinctly high expression in motor neurons of the affinity copper and zinc binding sites that facilitate its
spinal cord (Peluffo et al., 2005). precipitation via interactions with these metal ions. The AD-
Although not a redox-active metal-like copper, zinc too is affected brain contains increased concentrations of copper and
required to allow optimal antioxidant responses and for DNA zinc within the core and peripheral regions of amyloid plaques
repair. Zinc is a functional and structural component of several (Lovell et al., 1998; Zecca et al., 2004; Maynard et al., 2005).
enzymes and transcription factors involved in the antioxidant Interaction between Ah and oxidized metal ions renders the Ah
response and DNA integrity. Inadequate levels of zinc in the peptide toxic to neurons in cell culture (see Maynard et al.,
brain lead to abnormal functioning of SOD, which in addition 2005), an effect that is abolished under copper-free conditions
to its dismutase activity, acquires peroxidase activity resulting (see Bush, 2003). If copper has a primary role in the toxicity of
in generation of peroxynitrite and neuronal death (Mocchegiani Ah peptide actions, zinc seems to play a key role in
et al., 2005). aggregation of amyloid deposits. At neutral pH, interactions
The physiological intracellular concentration of free zinc in between zinc and Ah result in the formation of insoluble
eukaryotic cells lies in the low picomolar range. Increases in aggregates, whereas binding of copper is competitive resulting
free intracellular zinc (nanomolar concentrations), following in soluble Ah complexes. In contrast, within the slightly acidic
ischemia, for example, can lead to neurotoxicity (see Fig. 5; environment of an elderly or inflamed brain, copper causes
Frederickson et al., 2005). insolubilization and aggregation of Ah (see Maynard et al.,
However, intracellularly accumulated zinc may be neuro- 2005).
toxic or neuroprotective depending on its concentration and the Neuronal death in AD occurs selectively in the hippocam-
particular neuron of interest (Cote et al., 2005). For example, pus and neocortex as well as in particular subcortical regions.
within the strata oriens and lucidum of the CA3 region of the Because zinc-releasing neurons are also seen in these regions
hippocampus, high intracellular concentrations of zinc resulted high concentrations of free, ionic zinc may be released during
in cell death, an effect that could be ameliorated by reducing synaptic transmission. Under conditions of reduced metabolism
zinc levels. Conversely, moderate intracellular concentrations in the aged or AD-affected brain, pools of extracellular zinc
of zinc in neurons of the CA3 pyramidal layer resulted in cell could accumulate, promoting aggregation of Ah. In support of
survival. Surprisingly, zinc chelation led to an increase in the this idea, studies using mice that are deficient in the synaptic
A. Mathie et al. / Pharmacology & Therapeutics 111 (2006) 567 – 583 579

zinc transporter, ZnT3 showed an ¨ 50% reduction in amyloid Acknowledgments


load compared to wild-type animals (see Maynard et al., 2005).
Our work is supported by the MRC and ARC. CEC is an
5.2. Epilepsy Australian Research Council Postdoctoral Fellow.

Several of the ion channel proteins (such as Na+ channels, References


T-type Ca2+ channels, and GABAA receptor channels), which
zinc and copper act on, have been implicated in certain forms Acuna-Castillo, C., Morales, B., & Huidobro-Toro, J. P. (2000). Zinc and
of epilepsy, thus it is difficult to make simple predictions about copper modulate differentially the P2X4 receptor. J Neurochem 74(4),
1529 – 1537.
the pro- or anti-convulsive actions of these ions. This is Akaike, N., Kostyuk, P. G., & Osipchuk, Y. V. (1989). Dihydropyridine-
compounded by experimental observations where zinc has sensitive low-threshold calcium channels in isolated rat hypothalamic
been documented to act as either an anticonvulsant (William- neurones. J Physiol 412, 181 – 195.
son & Spencer, 1995) or a pro-convulsant (Pei et al., 1983). Alexander, S. P. H., Mathie, A., & Peters, J. A. (2004). Guide to receptors and
Seizure activity in human epileptics and model animals channels. Br J Pharmacol 141, S1 – S126.
Aller, M. I., Veale, E. L., Linden, A. -M., Sandu, C., Schwaninger, M., Evans,
influences the distribution of essential trace elements, including L. J., et al. (2005). Modifying the subunit composition of TASK channels
copper and zinc, in the brain and peripheral tissues (Papava- alters the modulation of a leak conductance in cerebellar granule neurons. J
siliou & Miller, 1983; Carl et al., 1990; Hirate et al., 2002). A Neurosci 25, 11455 – 11467.
number of studies suggest that altered zinc homeostasis in the Assaf, S. Y., & Chung, S. -H. (1984). Release of endogenous Zn2+ from brain
brain contributes to the occurrence of epileptic seizures (e.g., tissue during activity. Nature 308, 734 – 736.
Backx, P. H., Yue, D. T., Lawrence, J. H., Marban, E., & Tomaselli, G. F.
Buhl et al., 1996; Takeda et al., 2003). Prior to seizure activity, (1992). Molecular localization of an ion-binding site within the pore of
zinc concentrations are higher in the brain of epileptic (EL) mammalian sodium channels. Science 257(5067), 248 – 251.
mice than control mice. The enhanced uptake of zinc into the Barbuti, A., Ishii, S., Shimizu, T., Robinson, R. B., & Feinmark, S. J. (2002).
brain may reflect a compensatory mechanism for maintaining Block of the background K(+) channel TASK-1 contributes to arrhythmo-
genic effects of platelet-activating factor. Am J Physiol Heart Circ Physiol
correct brain function in EL mice (Hirate et al., 2002; Takeda et
282(6), H2024 – H2030.
al., 2003). Conversely, postseizure activity, the zinc concen- Barnes, N., Tsivkovskii, R., Tsivkovskii, N., & Lutsenko, S. (2005). The
tration in the brain of EL mice is significantly lower than that in copper-transporting ATPases, Menkes and Wilson disease proteins,
control mice (Hirate et al., 2002). The likelihood of seizure have distinct roles in adult and developing cerebellum. J Biol Chem 280,
activity in EL mice is diminished by zinc supplementation, 9640 – 9645.
whereas it is augmented by zinc deprivation (Fukahori & Itoh, Barnham, K. J., Masters, C. L., & Bush, A. I. (2004). Neurodegenerative
diseases and oxidative stress. Nat Rev Drug Discov 3, 205 – 214.
1990; Hirate et al., 2002; Takeda et al., 2003). Kainate-induced Baumgarten, C. M., & Fozzard, H. A. (1989). Cd2+ and Zn2+ block unitary Na+
seizures in EL mice trigger a substantial, but tissue-specific currents in Purkinje and ventricular cells. Biophys J 55, 313a.
loss of zinc from the brain. A reduction of zinc occurs in the Blakemore, L. J., & Trombley, P. Q. (2004). Diverse modulation of olfactory
hippocampus, amygdala, and cerebral cortex where there is an bulb AMPA receptors by zinc. NeuroReport 15(5), 919 – 923.
abundance of gluzinergic neuron terminals but not in the Bresink, I., Ebert, B., Parsons, C. G., & Mutschler, E. (1996). Zinc changes
AMPA receptor properties: results of binding studies and patch clamp
cerebellum (Sloviter, 1985; Frederickson et al., 1988; Takeda et recordings. Neuropharmacology 35(4), 503 – 509.
al., 2003). Buhl, E. H., Otis, T. S., & Mody, I. (1996). Zinc-induced collapse of
Perhaps the single most interesting observation to date augmented inhibition by GABA in a temporal lobe epilepsy model. Science
concerning the importance of zinc in epilepsy arises from 271, 369 – 373.
Bush, A. I. (2003). The metallobiology of Alzheimer’s disease. TINS 26,
observed structural and receptor distribution changes that occur
207 – 214.
in the hippocampus of patients with temporal lobe epilepsy or Busselberg, D., Michael, D., Evans, M. L., Carpenter, D. O., & Haas, H. L.
in animal models of this condition (Dudek, 2001). Two quite (1992). Zinc (Zn2+) blocks voltage-gated calcium channels in cultured
distinct changes occur, but the relationship between the two has dorsal root ganglion cells. Brain Res 593, 77 – 81.
clear functional consequences. Firstly, the composition of the Carl, G., Critchfield, J. W., Thompson, J. L., Holmes, G. L., Gallagher,
GABAA receptors in dentate granule cells changes from B. B., & Keen, C. L. (1990). Genetically epilepsy prone rats are
characterized by altered tissue trace element concentrations. Epilepsia
relatively zinc-insensitive receptors (incorporating g subunits) 31, 247 – 252.
to receptors comprised, primarily, of zinc-sensitive subunits Castelli, L., Tanzi, F., Taglietti, V., & Magistretti, J. (2003). Cu2+, Co2+, and
(see Section 3.2.2). Secondly, mossy fiber terminals (which Mn2+ modify the gating kinetics of high-voltage-activated Ca2+ channels
release zinc) are reorganized to innervate the dentate granule in rat palaeocortical neurons. J Membr Biol 195, 121 – 136.
cells. It has been proposed that zinc released from mossy fibers Charton, G., Rovira, C., Ben-Ari, Y., & Leviel, V. (1985). Spontaneous and
evoked release of endogenous Zn2+ in the hippocampal mossy fiber zone of
during intense stimulation will block these novel GABAA the rat in situ. Exp Brain Res 58, 202 – 205.
receptors, decrease inhibition, and contribute to the generation Chen, N., Moshaver, A., & Raymond, L. A. (1997). Differential sensitivity of
of seizures in the dentate gyrus (Buhl et al., 1996; Hamed & recombinant N-methyl-d-aspartate receptor subtypes to zinc inhibition. Mol
Abdellah, 2004). This represents an excellent example of the Pharmacol 51, 1015 – 1023.
functional consequences that arise from the combination of an Choi, D. W., & Koh, J. Y. (1998). Zinc and brain injury. Annu Rev Neurosci 21,
347 – 375.
alteration of zinc release from synaptic terminals and the Chu, Y., Mouat, M. F., Coffield, J. A., Orlando, R., & Grider, A. (2003).
selective sensitivity of particular protein subunits to modulation Expression of P2X6, a purinergic receptor subunit, is affected by dietary
by zinc. zinc deficiency in rat hippocampus. Biol Trace Elem Res 91(1), 77 – 87.
580 A. Mathie et al. / Pharmacology & Therapeutics 111 (2006) 567 – 583

Clapham, D. E. (2003). TRP channels as cellular sensors. Nature 426(6966), kainic acid-induced seizures: a histo-fluorescence study. Brain Res 446,
517 – 524. 383 – 386.
Clapham, D. E., Runnels, L. W., & Strubing, C. (2001). The TRP ion channel Frederickson, C. J., Suh, S. W., Silva, D., Frederickson, C. J., & Thompson,
family. Nat Rev Neurosci 2(6), 387 – 396. R. B. (2000). Importance of zinc in the central nervous system: the zinc-
Clarke, C. E., Veale, E. L., Green, P. J., Meadows, H. J., & Mathie, A. (2004). containing neuron. J Nutr 130, 1471S – 1483S.
Selective block of the human 2-P domain potassium channel, TASK-3, and Frederickson, C. J., Koh, J. -Y., & Bush, A. I. (2005). The neurobiology of zinc
the native leak potassium current, IKSO, by zinc. J Physiol 560(1), 51 – 62. in health and disease. Nat Rev Neurosci 6, 449 – 462.
Cloues, R. (1995). Properties of ATP-gated channels recorded from rat Frelin, C., Cognard, C., Vigne, P., & Lazdunski, M. (1986). Tetrodotoxin-
sympathetic neurons: voltage dependence and regulation by Zn2+ ions. sensitive and tetrodotoxin-resistant Na+ channels differ in their sensitivity to
J Neurophysiol 73, 312 – 319. Cd2+ and Zn2+. Eur J Pharmacol 122, 245 – 250.
Cloues, R., Jones, S., & Brown, D. A. (1993). Zn2+ potentiates ATP-activated Fukahori, M., & Itoh, M. (1990). Effects of dietary zinc status on seizure
currents in rat sympathetic neurons. Pflügers Arch 424(2), 152 – 158. susceptibility and hippocampal zinc content in the El (epilepsy) mouse.
Coddou, C., Villalobos, C., Gonzalez, J., Acuna-Castillo, C., Loeb, B., & Brain Res 529, 16 – 22.
Huidobro-Toro, J. P. (2002). Formation of carnosine-Cu (II) complexes Gaetke, L. M., & Chow, C. K. (2003). Copper toxicity, oxidative stress, and
prevents and reverts the inhibitory action of copper in P2X4 and P2X7 antioxidant nutrients. Toxicology 189, 147 – 163.
receptors. J Neurochem 80(4), 626 – 633. Gilly, F. A. R., & Armstrong, C. M. (1982a). Slowing of sodium channel
Coddou, C., Morales, B., & Huidobro-Toro, J. P. (2003). Neuromodulator role opening kinetics in squid axon by extracellular zinc. J Gen Physiol 79,
of zinc and copper during prolonged ATP applications to P2X4 purinocep- 935 – 964.
tors. Eur J Pharmacol 472(1 – 2), 49 – 56. Gilly, F. A. R., & Armstrong, C. M. (1982b). Divalent cations and the
Coetzee, W. A., Amarillo, Y., Chiu, J., Chow, A., Lau, D., McCormack, T., et activation kinetics of potassium channels in squid giant axons. J Gen
al. (1999). Molecular diversity of K+ channels. Ann N Y Acad Sci 868, Physiol 79, 965 – 996.
233 – 285. Goldin, A. L. (2001). Resurgence of sodium channel research. Ann Rev Physiol
Colvin, R. A., Fontaine, C. P., Laskowski, M., & Thomas, D. (2003). Zn2+ 63, 871 – 894.
transporters and Zn2+ homeostasis in neurons. Eur J Pharmacol 479(1 – 3), Goldstein, S. A. N., Bockenhauer, D., O’Kelly, I., & Zilberberg, N. (2001).
171 – 185. Potassium leak channels and the KCNK family of two-P-domain subunits.
Cooper, B. Y., Johnson, R. D., & Rau, K. K. (2004). Characterization and Nat Rev Neurosci 2, 175 – 184.
function of TWIK-related acid sensing K+ channels in a rat nociceptive cell. Gore, A., Moran, A., Hershfinkel, M., & Sekler, I. (2004). Inhibitory
Neuroscience 129, 209 – 224. mechanism of store-operated Ca2+ channels by zinc. J Biol Chem 279,
Cote, A., Chiasson, M., Peralta, M. R., Lafortune, K., Pellegrini, L., & Toth, K. 11106 – 11111.
(2005). Cell-type specific action of seizure-induced intracellular zinc Gruss, M., Mathie, A., Lieb, W. R., & Franks, N. P. (2004). The two-pore-
accumulation. J Physiol 566(3), 821 – 837. domain K+ channels TREK-1 and TASK-3 are differentially modulated by
Coulter, K. L., Perier, F., Radeke, C. M., & Vandenberg, C. A. (1995). copper and zinc. Mol Pharmacol 66, 530 – 537.
Identification and molecular localization of a pH-sensing domain for the Gurney, A. M., Osipenko, O. N., MacMillan, D., McFurlane, K. M., Tate, R. J.,
inward rectifier potassium channel HIR. Neuron 15, 1157 – 1168. & Kempsill, F. E. J. (2003). Two-pore domain K channel, TASK-1 in
Craig, P. J., Beattie, R. E., Folly, E. A., Banerjee, M. D., Reeves, M. B., pulmonary artery smooth muscle cells. Circ Res 83, 957 – 964.
Preistley, J. V., et al. (1999). Distribution of the voltage-dependent calcium Gutteridge, J. M. (1984). Copper-phenanthroline-induced site-specific oxygen-
channel a1G subunit mRNA and protein throughout the mature rat brain. radical damage to DNA. Detection of loosely bound trace copper in
Eur J Neurosci 11(8), 2949 – 2964. biological fluids. Biochem J 218(3), 983 – 985.
Cusimano, A., D’Adamo, M. C., & Pessia, M. (2004). An episodic ataxia type- Hamed, S. A., & Abdellah, M. M. (2004). Trace elements and
1 mutation in the S1 segment sensitises the hKv1.1 potassium channel to electrolytes homeostasis and their relation to antioxidant enzyme
extracellular Zn2+. FEBS Letters 576, 237 – 244. activity in brain hyperexcitability of epileptic patients. J Pharmacol
Davidson, J. L., & Kehl, S. J. (1995). Changes of activation and inactivation Sci 96, 349 – 359.
gating of the transient potassium current of rat pituitary melanotrophs Hank, D. A., & Sheets, M. F. (1992). Extracellular divalent and trivalent cation
caused by micromolar Cd2+ and Zn2+. Can J Physiol Pharmacol 73(1), effects on sodium current kinetics in single canine cardiac Purkinje cells.
36 – 42. J Physiol 454, 267 – 298.
Donaldson, J., St Pierre, T., Minnich, J. L., & Barbeau, A. (1973). Harris, E. D. (2002). Cellular transporters for zinc. Nutr Rev 60(4), 121 – 124.
Determination of Na+, K+, Mg2+, Cu2+, Zn2+, and Mn2+ in rat brain Harrison, N. L., & Gibbons, S. J. (1994). Zn2+: an endogenous modulator
regions. Can J Biochem 51, 87 – 92. of ligand- and voltage-gated ion channels. Neuropharmacology 33(8),
Dudek, F. E. (2001). Zinc and epileptogenesis. Epilepsy Curr 1, 66 – 70. 935 – 952.
Easaw, J. C., Jassar, B. S., Harris, K. H., & Jhamandas, J. H. (1999). Zinc Harrison, N. L., Radke, H. K., Tamkun, M. M., & Lovinger, D. M. (1992).
modulation of ionic currents in the horizontal limb of the diagonal band of Modulation of gating of cloned rat and human K+ channels by micromolar
Broca. Neuroscience 94(3), 785 – 795. Zn2+. Mol Pharmacol 43, 482 – 486.
Elinder, F., & Arhem, P. (2003). Metal ion effects on ion channel gating. Q Rev Hartness, M. E., Lewis, A., Searle, G. J., O’Kelly, I., Peers, C., & Kemp, P. J.
Biophys 36, 373 – 427. (2001). Combined antisense and pharmacological approaches implicate
Ertel, E. A., Campbell, K. P., Harpold, M. M., Hofamnn, F., Mori, Y., Perez- hTASK as an airway O(2) sensing K(+) channel. J Biol Chem 276(28),
Reyes, E., et al. (2000). Nomenclature of voltage-gated calcium channels. 26499 – 26508.
Neuron 25(3), 533 – 535. Hartter, D. E., & Barnea, A. (1988). Brain tissue accumulates 67copper by two
Foskett, J. K., & Wong, D. C. P. (1994). [Ca2+]i inhibition of Ca2+ ligand-dependent saturable processes. A high affinity, low capacity and a
release-activated Ca2+ influx underlies agonist- and thapsigargin- low affinity, high capacity process. J Biol Chem 263(2), 799 – 805.
induced [Ca2+]i oscillations in salivary acinar cells. J Biol Chem Heinemann, S. H., Terlau, H., & Imoto, K. (1992). Molecular basis for
269, 31525 – 31532. pharmacological differences between brain and cardiac sodium channels.
Frederickson, C. J., & Bush, A. I. (2001). Synaptically released zinc: Eur J Physiol 422, 90 – 92.
physiological functions and pathological effects. BioMetals 14(3 – 4), Hermosura, M. C., Monteilh-Zoller, M. K., Scharenberg, A. M., Penner, R., &
353 – 366. Fleig, A. (2002). Dissociation of the store-operated calcium current
Fredrickson, C. J., & Moncrieff, D. W. (1994). Zinc-containing neurons. Biol I(CRAC) and the Mg-nucleotide-regulated metal ion current MagNuM.
Signals 3(3), 127 – 139. J Physiol 539(2), 445 – 458.
Frederickson, C. J., Hernandez, M. D., Goik, S. A., Morton, J. D., & McGinty, Hille, B. (2001). Ionic channel of excitable membranes (3rd edR). USA’ Sinauer
J. F. (1988). Loss of zinc staining from hippocampal mossy fibers during Associates, Inc.
A. Mathie et al. / Pharmacology & Therapeutics 111 (2006) 567 – 583 581

Hirate, M., Takeda, A., Tamano, H., Enomoto, S., & Oku, N. (2002). Kurata, Y., Hisatome, I., Tsuboi, M., Uenishi, H., Zhang, G., Oyaizu, M., et al.
Distribution of trace elements in the brain of EL (epilepsy) mice. Epilepsy (1998). Effect of sulfhydryl oxidoreduction on permeability of cardiac
Res 51, 109 – 116. tetrodotoxin-insensitive sodium channel. Life Sci 63(12), 1023 – 1035.
Horning, M. S., & Trombley, P. Q. (2001). Zinc and copper influence Llanos, R. M., & Mercer, J. F. B. (2002). The molecular basis of copper
excitability of rat olfactory bulb neurons by multiple mechanisms. homeostasis and copper-related disorders. DNA Cell Biol 21(4), 259 – 270.
J Neurophysiol 86, 1652 – 1660. Laube, B., Kuhse, J., & Betz, H. (2000). Kinetic and mutational analysis
Hosie, A. M., Dunne, E. L., Harvey, R. J., & Smart, T. G. (2003). Zinc- of Zn2+ modulation of recombinant human inhibitory glycine receptors.
mediated inhibition of GABA(A) receptors: discrete binding sites underlie J Physiol 522(2), 215 – 230.
subtype specificity. Nat Neurosci 6(4), 362 – 369. Laube, B., Kuhse, J., Rundstrom, N., Kirsch, J., Schmieden, V., & Betz, H.
Hoth, M., & Penner, R. (1993). Calcium release-activated calcium current in rat (1995). Modulation by zinc ions of native rat and recombinant human
mast cells. J Physiol 465, 359 – 386. inhibitory glycine receptors. J Physiol 483(3), 613 – 619.
Howell, G. A., Welch, M. G., & Frederickson, C. J. (1984). Stimulation- Lauritzen, I., Zanzouri, M., Honore, E., Duprat, F., Ehrengruber, M. U.,
induced uptake and release of zinc in hippocampal slices. Nature Lazdunski, M., & Patel, A. J. (2003). K+-dependent cerebellar granule
308(5961), 736 – 738. neuron apoptosis. Role of TASK leak K+ channels. J Biol Chem 278(34),
Huang, R. -C., Peng, Y. -W., & Yau, K. -W. (1993). Zinc modulation of 32068 – 32076.
a transient potassium current and histochemical localization of the Le, K. T., Babinski, K., & Seguela, P. (1998). Central P2X4 and P2X6 channel
metal in neurons of the suprachiasmatic nucleus. Proc Natl Acad Sci 90, subunits co-assemble into a novel heteromeric ATP receptor. J Neurosci
11806 – 11810. 18(18), 7152 – 7159.
Jeong, S. -W., Park, B. -G., Park, J. -Y., Lee, J. -W., & Lee, J. -H. (2003). Leonoudakis, D., Gray, A. T., Winegar, B. D., Kindler, C. H., Harada, M.,
Divalent metals differentially block cloned T-type calcium channels. Taylor, D. M., et al. (1998). An open rectifier potassium channel with two
NeuroReport 14, 1537 – 1540. pore domains in tandem cloned rat cerebellum. J Neurosci 18(3), 868 – 877.
Jia, Y., Jeng, J. M., Sensi, S. L., & Weiss, J. H. (2002). Zn2+ currents are Lesage, F. (2003). Pharmacology of neuronal background potassium channels.
mediated by calcium-permeable AMPA/kainate channels in cultured murine Neuropharmacology 44, 1 – 7.
hippocampal neurones. J Physiol 543(1), 35 – 48. Li, C., Peoples, R. W., Li, Z., & Weight, F. F. (1993). Zn2+ potentiates
Johnson, R. P., O’Kelly, I. M., & Fearon, I. M. (2004). System-specific O2 excitatory action of ATP on mammalian neurons. Proc Natl Acad Sci
sensitivity of the tandem pore domain K+ channel TASK-1. Am J Physiol 90(17), 8264 – 8267.
Cell Physiol 286(2), C391 – C397. Li, Y. V., Hough, C. J., & Sarvey, J. M. (2003). Do we need zinc to think? Sci
Kanjhan, R., Housley, G. D., Burton, L. D., Christie, D. L., Kippenberger, A., Signal Transduct Knowl Environ 182, 19.
Thorne, P. R., et al. (1999). Distribution of the P2X2 receptor subunit of the Linder, M. C., & Hazegh-Azam, M. (1996). Copper biochemistry and
ATP-gated ion channels in the rat central nervous system. J Comp Neurol molecular biology. Am J Clin Nutr 63(5), 797S – 811S.
407, 11 – 32. Lovell, M. A., Robertson, J. D., Teesdale, W. J., Campbell, J. L., &
Kardos, J., Kovacs, I., Hajos, F., Kalman, M., & Simonyi, M. (1989). Markesbery, W. R. (1998). Copper, iron and zinc in Alzheimer’s disease
Nerve endings from rat brain tissue release copper upon depolarisation. senile plaques. J Neurol Sci 158, 47 – 52.
A possible role in regulating neuronal excitability. Neurosci Lett 103(2), Magistretti, J., Castelli, L., Taglietti, V., & Tanzi, F. (2003). Dual effect of Zn2+
139 – 144. on multiple types of voltage-dependent Ca2+ currents in rat palaeocortical
Kasai, H., & Neher, E. (1992). Dihydropyridine-sensitive and N-conotoxin- neurons. Neuroscience 117, 249 – 264.
sensitive calcium channels in mammalian neuroblastoma-glioma cell line. Marin, P., Israel, M., Glowinski, J., & Premont, J. (2000). Routes of zinc entry
J Physiol 448, 161 – 188. in mouse cortical neurons: role in zinc-induced neurotoxicity. Eur
Kase, M., Kakimoto, S., Sakuma, S., Houtani, T., Ohishi, H., Ueyama, T., et al. J Neurosci 12(1), 8 – 18.
(1999). Distribution of neurons expressing a1G subunit mRNA of T-type Mathie, A., Clarke, C. E., Ranatunga, K. M., & Veale, E. (2003). What are the
voltage-dependent calcium channel in adult rat central nervous system. roles of the many different types of potassium channel expressed in
Neurosci Lett 268, 77 – 80. cerebellar granule cells? Cerebellum 2(1), 11 – 25.
Kay, A. R. (2003). Evidence for chelatable zinc in the extracellular space of the Mayer, M. L., & Sugiyama, K. (1988). A modulatory action of divalent cations
hippocampus, but little evidence for synaptic release of Zn. J Neurosci on transient outward current in cultured rat sensory neurons. J Physiol 396,
23(17), 6847 – 6855. 417 – 433.
Kehl, S. J., Eduljee, C., Kwan, D. C. H., Zhang, S., & Fedida, D. (2002). Mayer, M. L., Vyklicky Jr, L., & Westbrook, G. L. (1989). Modulation of
Molecular determinants of the inhibition of human Kv1.5 potassium excitatory amino acid receptors by group IIB metal cations in cultured
currents by external protons and Zn2+. J Physiol 541(1), 9 – 24. mouse hippocampal neurones. J Physiol 415(1), 329 – 350.
Kerchner, G. A., Canzoniero, L. M. T., Yu, S. P., Ling, C., & Choi, D. W. Maynard, C. J., Bush, A. I., Masters, C. L., Cappai, R., & Li, Q. (2005).
(2000). Zn2+ current is mediated by voltage-gated Ca2+ channels and Metals and amyloid-h in Alzheimer’s disease. Int J Exp Path 86,
enhanced by extracellular acidity in mouse cortical neurones. J Physiol 147 – 159.
528(1), 39 – 52. Mocchegiani, E., Bertoni-Freddari, C., Marcellini, F., & Malavolta, M. (2005).
Kim, J. -S., Park, J. -Y., Kang, H. -W., Lee, E. -J., Bang, H., & Lee, J. -H. Brain, aging and neurodegeneration: role of zinc ion availability. Prog
(2005). Zinc activates TREK-2 potassium channel activity. J Pharm Exp Neurobiol 75, 367 – 390.
Ther 314, 618 – 625. Monteilh-Zoller, M. K., Hermosura, M. C., Nadler, M. J. S., Scharenberg,
Koizumi, S., Nakazawa, K., & Inoue, K. (1995). Inhibition by Zn2+ of uridine A. M., Penner, R., & Fleig, A. (2003). TRPM7 provides an ion
5V-triphosphate-induced Ca(2+)-influx but not Ca(2+)-mobilization in rat channel mechanism for cellular entry of trace metal ions. J Gen Physiol
phaeochromocytoma cells. Br J Pharmacol 115(8), 1502 – 1508. 121, 49 – 60.
Kresse, W., Sekler, I., Hoffmann, A., Peters, O., Nolte, C., Moran, A., & Montell, C., Birnbaumer, L., & Flockerzi, V. (2002). The TRP channels, a
Kettenmann, H. (2005). Zinc ions are endogenous modulators of neuro- remarkably functional family. Cell 108(5), 595 – 598.
transmitter-stimulated capacitative Ca2+ entry in both cultures and in situ Montell, C., & Rubin, G. M. (1989). Molecular characterization of the
mouse astrocytes. Eur J Neurosci 21, 1626 – 1634. Drosophila trp locus: a putative integral membrane protein required for
Kuo, C. -C., & Chen, F. -P. (1999). Zn2+ modulation of neuronal transient K+ phototransduction. Neuron 2(4), 1313 – 1323.
current: fast and selective binding to the deactivated channels. Biophys J 77, Morera, F. J., Wolff, D., & Vergara, C. (2003). External copper inhibits the
2552 – 2562. activity of the large-conductance calcium- and voltage-sensitive potassium
Kuo, C. -C., Chen, W. -Y., & Yang, Y. -C. (2004). Block of tetrodotoxin- channel from skeletal muscle. J Membr Biol 192, 65 – 72.
resistant Na+ channel pore by multivalent cations: gating modification and Nachshen, D. A. (1984). Selectivity of the Ca binding site in synaptosome Ca
Na+ flow dependence. J Gen Physiol 124, 27 – 42. Channels. J Gen Physiol 83, 941 – 967.
582 A. Mathie et al. / Pharmacology & Therapeutics 111 (2006) 567 – 583

Nakazawa, K., & Ohno, Y. (1997). Effects of neuroamines and divalent to mitochondrial copper in cerebellum. J Histochem Cytochem 42(12),
cations on cloned and mutated ATP-gated channels. Eur J Pharmacol 1585 – 1591.
325(1), 101 – 108. Schild, L., & Moczydlowski, E. (1991). Competitive binding interaction
Narahashi, T., Ma, J. Y., Arakawa, O., Reuveny, E., & Nakahiro, M. (1994). between Zn2+ and saxitoxin in cardiac Na+ channels. Biophys J 59,
GABA receptor-channel complex as a target site of mercury, copper, zinc, 523 – 537.
and lanthanides. Cell Mol Neurobiol 14(6), 599 – 621. Schild, L., Ravindran, A., & Moczydlowski, E. (1991). Zn2+-induced
Nicke, A., Baumert, H. G., Rettinger, J., Eichele, A., Lambrecht, G., subconductance events in cardiac Na+ channels prolonged by batracho-
Mutschler, E., et al. (1998). P2X1 and P2X3 receptors form stable toxin. J Gen Physiol 97, 117 – 142.
trimers: a novel structural motif of ligand-gated ion channels. EMBO J Schlief, M. L., Craig, A. M., & Gitlin, J. D. (2005). NMDA receptor activation
17, 3016 – 3028. mediates copper homeostasis in hippocampal neurons. J Neurosci 25(1),
Nikonenko, I., Bancila, M., Bloc, A., Muller, D., & Bijlenga, P. (2005). 239 – 246.
Inhibition of T-type calcium channels protects neurons from delayed Schmitz, C., Perraud, A., Johnson, C., Inabe, K., Smith, M., Penner, R., et al.
ischemia-induced damage. Mol Pharmacol 68, 84 – 89. (2003). Regulation of vertebrate cellular Mg2+ homeostasis by TRPM7.
Nilius, B. (2003). From TRPs to SOCs, CCEs, and CRACs: consensus and Cell 114, 191 – 200.
controversies. Cell Calcium 33(5 – 6), 293 – 298. Schumann, K., Classen, H. G., Dieter, H. H., Konig, J., Multhaup, G.,
North, R. A. (2002). Molecular physiology of P2X receptors. Physiol Rev 82, Rukgauer, M., Summer, K. H., Bernhardt, J., & Biesalski, H. K.
1013 – 1067. (2002). Hohenheim consensus workshop: copper. Eur J Clin Nutr 56,
Ono, S., & Cherian, M. G. (1999). Regional distribution of metallothionein, 469 – 483.
zinc and copper in the brain of different strains of rats. Biol Trace Elem Res Sensi, S. L., Canzoniero, L. M., Yu, S. P., Ying, H. S., Koh, J. Y., Kerchner,
69(2), 151 – 159. G. A., et al. (1997). Measurement of intracellular free zinc in living
Oyama, Y., Nishi, K., Yatani, A., & Akaike, N. (1982). Zinc current in Helix cortical neurons: routes of entry. J Neurosci 17(24), 9554 – 9564.
soma membrane. Comp Biochem Physiol 72C, 403 – 410. Sharanova, I. N., Vorobjev, V. S., & Haas, H. L. (2000). Interactions between
Paoletti, P., Ascher, P., & Neyton, J. (1997). High-affinity zinc inhibition of copper and zinc at GABAA receptors in acutely isolated cerebellar Purkinje
NMDA NR1-NR2A receptors. J Neurosci 17(15), 5711 – 5725. cells of the rat. Br J Pharmacol 130, 851 – 856.
Papavasiliou, P. S., & Miller, T. (1983). Generalized seizures alter the cerebral Sheline, C. T., Ying, H. S., Ling, C. S., Canzoniero, L. M. T., & Choi, D. W.
and peripheral metabolism of essential metals in mice. Exp Neurol 82, (2002). Depolarization-induced zinc influx into cultured cortical neurons.
223 – 226. Neurobiol Dis 10, 41 – 53.
Parekh, A. B., & Penner, R. (1993). Store depletion and calcium influx. Physiol Slomianka, L., Danscher, G., & Frederickson, C. J. (1990). Labelling of the
Rev 77, 901 – 930. neurons of origin of zinc-containing pathways by intraperitoneal injections
Pei, Y., Zhao, D., Huang, J., & Cao, L. (1983). Zinc-induced seizures: a new of sodium selenite. Neuroscience 38(3), 843 – 854.
experimental model of epilepsy. Epilepsy 24, 169 – 176. Sloviter, R. (1985). A selective loss of hippocampal mossy fiber Timm stain
Peluffo, H., Acarin, L., Faiz, M., Castellano, B., & Gonzalez, B. (2005). Cu/Zn accompanies granule cell seizure activity induced by perforant path
superoxide dismutase expression in the postnatal rat brain following an stimulation. Brain Res 330, 150 – 153.
excitotoxic injury. J Neuroinflammation 2(1), 12 – 25. Smart, T. G., Xie, X., & Krishek, B. J. (1994). Modulation of inhibitory and
Peters, S., Koh, J., & Choi, D. W. (1987). Zinc selectively blocks the excitatory amino acid receptor ion channels by zinc. Prog Neurobiol 42(3),
action of N-methyl-d-aspartate on cortical neurons. Science 236(4801), 341 – 393.
589 – 593. Smart, T. G., Hosie, A. M., & Miller, P. S. (2004). Zn2+ ions: modulators
Petersen, C. C., Berridge, M. J., Borgese, M. F., & Bennett, D. L. (1995). of excitatory and inhibitory synaptic activity. Neuroscientist 10(5),
Putative capacitative calcium entry channels: expression of Drosophila trp 432 – 442.
and evidence for the existence of vertebrate homologues. Biochem J 311, Soto, F., Garcia-Guzman, M., Gomez-Hernandez, J. M., Hollmann, M.,
41 – 44. Karschin, C., & Stuhmer, W. (1996). P2X4: an ATP-activated ionotropic
Poling, J. S., Vicini, S., Rogawski, M. A., & Salem, N. (1996). Docosahex- receptor cloned from rat brain. Proc Natl Acad Sci 93(8), 3684 – 3688.
aenoic acid block of neuronal voltage-gated K+ channels: subunit selective Spires, S., & Begenisich, T. (1990). Modification of potassium channel kinetics
antagonism by zinc. Neuropharmacology 35, 969 – 982. by histidine-specific reagents. J Gen Physiol 96(4), 757 – 775.
Prothero, L. S., Richards, C. D., & Mathie, A. (1998). Inhibition by inorganic Strausak, D., Mercer, J. F. B., Dieter, H. H., Stremmel, W., & Multhaup, G.
ions of a sustained calcium signal evoked by activation of mGlu5 receptors (2001). Copper in disorders with neurological symptoms: Alzheimer’s,
in rat cortical neurons and glia. Br J Pharmacol 125, 1551 – 1561. Menkes, and Wilson diseases. Brain Res Bull 55, 175 – 185.
Prothero, L. S., Mathie, A., & Richards, C. D. (2000). Purinergic and Stuerenberg, H. J. (2000). CSF copper concentrations, blood – brain barrier
muscarinic receptor activation activates a common calcium entry function, and ceruloplasmin synthesis during the treatment of Wilson’s
pathway in rat neocortical neurons and glial cells. Neuropharmacology disease. J Neural Transm 107, 321 – 329.
39, 1768 – 1778. Takahashi, K., & Akaike, N. (1990). Calcium antagonist effects on low-
Puig, S., & Thiele, D. J. (2002). Molecular mechanisms of copper uptake and threshold (T-type) calcium current in rat isolated hippocampal CA1
distribution. Curr Opinion Chem Biol 6(2), 171 – 180. pyramidal neurons. J Pharmacol Exp Ther 256, 169 – 175.
Rachline, J., Perin-Dureau, F., Le Goff, A., Neyton, J., & Paoletti, P. (2005). Takeda, A. (2000). Movement of zinc and its functional significance in the
The micromolar zinc-binding domain on the NMDA receptor subunit brain. Brain Res Rev 34, 137 – 148.
NR2B. J Neurosci 25(2), 308 – 317. Takeda, A. (2001). Zinc homeostasis and functions of zinc in the brain.
Ravindran, A., Schild, L., & Moczydlowski, E. (1991). Divalent cation BioMetals 14, 343 – 351.
selectivity for external block of voltage-dependent Na+ channels prolonged Takeda, A., Hirate, M., Tamano, H., & Oku, N. (2003). Zinc movement in the
by batrachotoxin: Zn2+ induces discrete substates in cardiac Na+ channels. brain under kainate-induced seizures. Epilepsy Res 54, 123 – 129.
J Gen Physiol 97, 89 – 115. Talley, E. M., Cribbs, L. L., Lee, J. H., Daud, A., Perez-Reyes, E., & Bayliss,
Ruiz, A., Walker, M. C., Fabian-Fine, R., & Kullmann, D. M. (2004). D. A. (1999). Differential distribution of three members of a gene family
Endogenous zinc inhibits GABA(A) receptors in a hippocampal pathway. encoding low voltage-activated (T-type) calcium channels. J Neurosci
J Neurophysiol 91(2), 1091 – 1096. 19(6), 1895 – 1911.
Satin, J., Kyle, J. W., Chen, M., Bell, P., Cribbs, L. L., Fozzard, H. A., & Tanguy, J., & Yeh, J. Z. (1988). Divalent cation block of normal and BTX-
Rogart, R. B. (1992). A mutant of TTX-resistant cardiac sodium channels modified sodium channels in squid axons. Biophys J 53, 229a.
with TTX-sensitive properties. Science 256(5060), 1202 – 1205. Tarohda, T., Yamamoto, M., & Amano, R. (2004). Regional distribution of
Sato, M., Ohtomo, K., Daimon, T., Sugiyama, T., & Iijima, K. (1994). manganese, iron, copper, and zinc in the rat brain during development. Anal
Localization of copper to afferent terminals in rat locus ceruleus, in contrast Bioanal Chem 380, 240 – 246.
A. Mathie et al. / Pharmacology & Therapeutics 111 (2006) 567 – 583 583

Thompson, R. B., Peterson, D., Mahoney, W., Cramer, M., Maliwal, B. P., Won immunohistochemistry for zinc transporter 3 and glutamate decarboxylase.
Suh, S., et al. (2002). Fluorescent zinc indicators for neurobiology. Neurosci Lett 321(1 – 2), 37 – 40.
J Neurosci Methods 118(1), 63 – 75. Watkins, C. S., & Mathie, A. (1994). Modulation of the gating of the transient
Tovar, K. R., Sprouffske, K., & Westbrook, G. L. (2000). Fast NMDA receptor- outward potassium current of rat isolated cerebellar granule neurons by
mediated synaptic currents in neurons from mice lacking the NR2B subunit. lanthanum. Pflügers Arch 428, 209 – 216.
J Neurophysiol 83(1), 616 – 620. Weiser, T., & Wienrich, M. (1996). The effects of copper ions on glutamate
Trombley, P. Q., Horning, M. S., & Blakemore, L. J. (1998). Carnosine receptors in cultured rat cortical neurons. Brain Res 742(1 – 2), 211 – 218.
modulates zinc and copper effects on amino acid receptors and synaptic White, A. R., Barnham, K. J., Huang, X., Voltakis, I., Beyreuther, K., Masters,
transmission. NeuroReport 9(15), 3503 – 3507. C. L., et al. (2004). Iron inhibits neurotoxicity induced by trace copper and
Trombley, P. Q., & Shepherd, G. M. (1996). Differential modulation by zinc biological reductants. J Biol Inorg Chem 9, 269 – 280.
and copper of amino acid receptors from rat olfactory bulb neurons. White, A. R., & Cappai, R. (2003). Neurotoxicity from glutathione depletion is
J Neurophysiol 76(4), 2536 – 2546. dependent on extracellular trace copper. J Neurosci Res 71, 889 – 897.
Valko, M., Morris, H., & Cronin, M. T. D. (2005). Metals, toxicity and White, J. A., Alonso, A., & Kay, A. R. (1993). A heart-like Na+ current in the
oxidative stress. Curr Med Chem 12, 1161 – 1208. medial entorhinal cortex. Neuron 11, 1037 – 1047.
Vega, M. T., Villalobos, C., Garrido, B., Gandia, L., Bulbena, O., Garcia- Wildman, S. S., King, B. F., & Burnstock, G. (1998). Zn2+ modulation of ATP-
Sancho, J., et al. (1994). Permeation by zinc of bovine chromaffin cell responses at recombinant P2X2 receptors and its dependence on extracel-
calcium channels: relevance to secretion. Pflügers Arch 429, 231 – 239. lular pH. Br J Pharmacol 123(6), 1214 – 1220.
Vennekens, R., Voets, T., Bindels, R. J., Droogmans, G., & Nilius, B. (2002). Wildman, S. S., King, B. F., & Burnstock, G. (1999). Modulatory activity of
Current understanding of mammalian TRP homologues. Cell Calcium extracellular H+ and Zn2+ on ATP-responses at rP2X1 and rP2X3 receptors.
31(6), 253 – 264. Br J Pharmacol 128, 486 – 492.
Virginio, C., Church, D., North, R. A., & Surprenant, A. (1997). Effects of Williamson, A., & Spencer, D. (1995). Zinc reduces dentate granule cell
divalent cations, protons and calmidazolium at the rat P2X7 receptor. hyperexcitability in epileptic humans. NeuroReport 6, 1562 – 1564.
Neuropharmacology 36(9), 1285 – 1294. Xie, X. M., & Smart, T. G. (1991). A physiological role for endogenous zinc in
Vlachova, V., Zemkova, H., & Vyklicky, L., Jr. (1996). Copper modulation of rat hippocampal synaptic neurotransmission. Nature 349(6309), 521 – 524.
NMDA responses in mouse and rat cultured hippocampal neurons. Eur J Xiong, K., Peoples, R. W., Montgomery, J. P., Chiang, Y., Stewart, R. R.,
Neurosci 8(11), 2257 – 2264. Weight, F. F., et al. (1999). Differential modulation by copper and zinc of
Voets, T., Nilius, B., Hoefs, S., vanderKemp, A. W., Droogmans, G., Bindels, P2X2 and P2X4 receptor function. J Neurophysiol 81(5), 2088 – 2089.
R. J., et al. (2004). TRPM6 forms the Mg2+ influx channel involved in Yu, F. H., & Catterall, W. A. (2004). The VGL-chanome: a protein superfamily
intestinal and renal Mg2+ absorption. J Biol Chem 279, 19 – 25. specialized for electrical signalling and ionic homeostasis. Sci Signal
Vogt, K., Mellor, J., Tong, G., & Nicoll, R. (2000). The actions of Transduct Knowl Environ 253, 15.
synaptically released zinc at hippocampal mossy fiber synapses. Neuron Zecca, L., Stroppolo, A., Gatti, A., Tampellini, D., Toscani, M., Gallorini, M.,
26(1), 187 – 196. et al. (2004). The role of iron and copper molecules in the neuronal
Wang, Z., Danscher, G., Kim, Y. K., Dahlstrom, A., & Mook Jo, S. (2002). vulnerability of locus caeruleus and substantia nigra during aging. Proc
Inhibitory zinc-enriched terminals in the mouse cerebellum: double- Natl Acad Sci 101, 9843 – 9848.

You might also like