You are on page 1of 10

Critical Review

Copper Metallothioneins Jenifer Calvo


Hunmin Jung
Gabriele Meloni*

Department of Chemistry and Biochemistry, University of Texas at Dallas,


Richardson, TX, USA

Abstract
Metallothioneins (MTs) are a class of low molecular weight metabolic status, Cu(I)-bound MT isoforms have been isolated,
and cysteine-rich metal binding proteins present in all the and their chemistry and biology characterized. Besides the rec-
branches of the tree of life. MTs efficiently bind with high ognized role in the biochemistry of divalent metals, it is
affinity several essential and toxic divalent and monovalent becoming evident that unique biological functions in selective-
transition metals by forming characteristic polynuclear metal- ly controlling copper levels, its reactivity as well as copper-
thiolate clusters within their structure. MTs fulfil multiple bio- mediated biochemical processes have evolved in some mem-
logical functions related to their metal binding properties, with bers of the MT superfamily. Selected examples are reviewed
essential roles in both Zn(II) and Cu(I) homeostasis as well as to highlight the peculiar chemical properties and biological
metal detoxification. Depending on the organism considered, functions of copper MTs. V C 2017 IUBMB Life, 69(4):236–245,

the primary sequence, and the specific physiological and 2017

Keywords: Metallothioneins; Copper; Zinc; metal-thiolate clusters;


metal homeostasis

Metallothioneins (MTs) constitute a superfamily of cysteine- Binz and Ka € gi (2), MTs are classified into 15 families based on
rich metal-binding proteins and peptides characterized by the taxonomic parameters and the patterns of distribution of Cys
presence of single or multiple metal-thiolate clusters in their residues in their sequence.
three dimensional structure (1). Metal clusters in MTs are usu- The recognized fundamental functions of MTs arise from
ally formed through the preferential coordination of d10 metal their capabilities to bind transition metals with high affinity,
ions (Zn(II), Cd(II), and Cu(I)), by an array of closely spaced and their primary biological roles include homeostasis of
cysteine thiolate ligands present in their primary sequence. essential trace metals zinc and copper, and sequestration and
The presence of nonsulfur coordination residues (e.g., histi- protection from environmental toxic metals such as cadmium,
dine) in some members of the superfamily, such as bacterial mercury, and lead (1). In addition, in light of the reactivity of
and plant MTs, has broken the dogma of the exclusive metal the metal-coordinating thiolate ligands, MTs play fundamental
coordination by cysteine thiolate residues in MTs. According to roles in protection against oxidative stress including reactive
oxygen and nitrogen species and other free radicals (1,3–5).
However, additional specific MT functions arise from specific
Abbreviations: Ab, amyloid-b; AD, Alzheimer’s disease; a-Syn, a-synu-
clein; CNS, central nervous system ; ESI-MS, electrospray ionization mass
biological needs and complexity of the organisms in which
spectrometry; MymT, mycobacterial metallothionein; MT, metallothionein; they are expressed. Since 60 years from their discovery, it
NMR, nuclear magnetic resonance ; ORF, open reading frame; ROS, reac- emerged that metallothoineins indeed possess complex pleio-
tive oxygen species; 3D, three-dimensional tropic functions. This is exemplified by the better-studied
C 2017 International Union of Biochemistry and Molecular Biology
V mammalian MTs for which additional specialized roles in
Volume 69, Number 4, April 2017, Pages 236–245 adaptation to stress, protection against brain injury, regulation
*Address correspondence to: Gabriele Meloni, Chemistry and Biochemis- of neuronal outgrowth, antiapoptotic effects, and reactivity
try, University of Texas at Dallas, Richardson, TX 75080, USA. Tel: 972-
and inactivation of metal-based chemotherapeutics leading to
883-4207. Fax: 972-883-2925.
E-mail: gabriele.meloni@utdallas.edu resistance have been demonstrated (4).
J.C. and H.J. contributed equally to the article. As a result of the high number of thiolate coordinating res-
Received 16 January 2017; Accepted 16 February 2017 idues in MTs and the lack of defined three-dimensional (3D)
DOI 10.1002/iub.1618 structures in the metal depleted apo forms, MTs can bind a
Published online 13 March 2017 in Wiley Online Library number of different monovalent and divalent metals both in
(wileyonlinelibrary.com) vitro and in vivo. The affinity of the metal ions for the binding

236 IUBMB Life


sites in MTs follows the order found for inorganic thiolates: SmtA is directly involved in Cu(I) binding in vivo and no native
HgII > AgI  CuI > CdII > ZnII (6). The replacement of one metal Cu(I)7-SmtA have been isolated. Based on these studies, it has
by another based on the relative affinities leads to the forma- been assumed that prokaryotic MTs are exclusively involved in
tion of mixed metal-MT complexes. As a result, homogeneous Zn(II) homeostasis (11).
or mixed species can be generated in vitro as a function of Surprisingly, in 2008, the discovery of a copper-binding
metal concentrations and relative ratios (7). In vivo, the spe- MT in the pathogen Mycobacterium tuberculosis demonstrated
cific metal composition and the functional roles of isolated MT the existence of prokaryotic MTs specifically evolved to control
appear to depend on the source organism, on the previous copper metabolism (12). This key copper-binding protein
exposure to metals, as well as on the primary sequence of MT involved in M. tuberculosis copper homeostasis has been
chain which can impose partial selectivity toward specific met- named mycobacterial MT (MymT) based on the presence of
als. Thus, homometallic or heterometallic M(II)MTs (with M(II) characteristic Cys-X-Cys motifs in its primary sequence and its
indicating divalent transition metals such as Zn(II) and Cd(II)) capability in efficiently coordinating transition metal ions.
are predominantly isolated in physiological conditions or upon MymT is the first MT from gram-positive bacteria for which a
cadmium exposure. The biochemistry and structural biology of defined physiological function has been discovered, which is to
MTs bound to divalent metal ions has been extensively investi- partially protect M. tuberculosis from copper toxicity. M. tuber-
gated and reviewed (4,5,8). In specific organisms, or under culosis, the pathogen that causes tuberculosis, shows a
specific physiological or pathological situations selected MT remarkable capability of evading the immune system response
isoforms are instead isolated as homometallic Cu(I)-MTs or by being capable of surviving in the phagosome compartment
heterometallic Cu(I),Zn(II) species. Thus, an important role for of hosts’ macrophages. Its virulence is strictly correlated to the
MTs in copper physiology has been revealed. Base on the capability of controlling metal homeostasis within this com-
structural/functional relationships, a classification of MTs as partment. During the M. tuberculosis infection, the host mac-
zinc–thioneins and copper–thioneins has been proposed in the rophage activates key immune responses including metal over-
efforts to identify evolutionary features defining copper- loading of the phagosome compartment. Copper is mobilized
specific roles for MTs (9). However, many challenges remain toward site of infection to aid in bacterial killing, imparting
in understanding the structural and functional diversity the toxicity by catalyzing the metal-dependent production of reac-
copper MTs within the entire MT superfamily. This review tive oxygen species (ROS) and disrupting iron–sulfur clusters in
focuses on aspects concerning structural and biochemical fea- the infecting pathogen. However, M. tuberculosis counters this
tures in four classes of copper-binding MTs to exemplify their copper toxicity by activation of copper resistance mechanisms
unique chemical and biological properties in copper such as upregulation of copper export, copper sequestration,
biochemistry. and oxidation of Cu (I) to less toxic Cu (II) (12–14) (Fig. 1).
MymT is a 4.9-KDa polypeptide of 53 amino acids possess-
ing metal binding properties (with the first 5 amino acids pos-
Bacterial Copper MTS sibly absent in the mature form) containing 7 Cys and 2 His
Bacterial MTs (Family 15) have been first identified in the residues and conserved in other Mycobacterium homologue
marine cyanobacterium Synechococcus sp. (strain RRIMP N1) proteins (12). MymT show poor amino acid sequence homology
(10). Until recently, the only bacterial MT family identified with other known eukaryotic or prokaryotic MTs. Although the
and characterized was the BmtA family. The prototypical bio- Cys-X-His-X-X-Cys-X-Cys motif of MymT is conserved in SmtA,
chemical and biophysical properties of BmtA family members the second metal binding motif, Cys-His-Cys-(X)3-Gly-(X)2-Tyr-
have emerged by detailed spectroscopic, biophysical, and Arg-Cys-Thr-Cys, has no known counterpart in other MT
structural investigation of the SmtA protein from Synechococcus sequences (12). Biophysical studies on purified MymT obtained
PCC 7942. The expression of MTs in Synechococcus sp. is upre- by recombinant expression in Escherichia coli revealed that
gulated by exposure to increased levels of Cd(II) and Zn(II) and Cu(I)-binding results in the formation of stable copper-bound
only to a lesser extent by Cu(II). SmtA has been demonstrated to species while, differently from SmtA, stable zinc species could
be involved in Zn(II) and Cd(II) sequestration and detoxification not be formed (12). MymT generates stable polynuclear Cu(I)-
in vivo. The three-dimensional structure of zinc- and cadmium- thiolate cluster as demonstrated by characteristic emissive
loaded Zn4SmtA revealed a single domain protein in which four luminescence spectra typical of proteins containing solvent-
M(II) ions are coordinated in distorted tehtrahedral geometry by shielded polynuclear Cu(I)-thiolate cores (12). Cu(I) titration
nine terminal and l2-bridging cysteine thiolates sulfurs and two studies coupled to metal–protein complexes analysis by nano
histidine imidazole nitrogen ligands (11). Another important Electrospray Ionization Mass Spectrometry (ESI-MS) revealed
feature is the presence of an inert Zn(II)Cys4 site surrounded by that the predominant species formed contains 4–6 copper ions.
b-strands and a-helix that form a zinc finger (11). SmtA has Exposure to nitric oxide resulted in cluster disruption and met-
been demonstrated to be capable of binding Cu(I) in vitro via al release in accordance to S-nitrosylation of the coordinating
zinc displacement from Zn4SmtA to generate monomeric homo- residues with concomitant metal dissociation (12). MymT
metallic Cu(I)7-SmtA in which significant conformational expression is induced by Cu, Cd, Co, Ni, and Zn. Nevertheless,
changes occurs. However, no evidence has been obtained that the existence of the Cu(I) bound form in vivo has been

Calvo et al. 237


IUBMB LIFE

Copper homeostasis and MymT in M. tuberculosis. Copper is pumped into the macrophage, phagosome and transported into
FIG 1 the cytoplasm of M. tuberculosis via an unknown import mechanism. Elevated copper levels are sensed by the copper-
responsive transcriptional repressor CsoR which, by losing affinity to its DNA cognate sequence upon copper binding, triggers
operon activation, and expression of CtpV, a P-type ATPase of the inner membrane (IM) likely involved in copper efflux. In the
outer membrane (OM), Mctb prevents the accumulation of copper within the mycobacterial cell possibly by copper export. In
the cytoplasm, the copper-binding MT, Mymt, binds and sequesters copper, enabling growth at elevated copper levels. Copper
release from MymT occurs in the presence of reactive nitrogen species (RNS), further triggering the copper operon activation
via formation of Cu-CsoR (12–14).

demonstrated by comparing the characteristic Cu(I)-MymT concentrations thereby preventing copper toxicity when copper
luminescence in vivo for copper-exposed wild-type M. tubercu- levels increase (16). Despite their sequential diversity, the
losis with the one in mutants in which MymT has been mechanisms of their copper-induced regulation follow shared
knocked-out and therefore absent. A significant decrease in biochemical principles. When intracellular copper concentra-
luminescence was attributed to the absence of Cu(I)-MymT tions rise, specific copper-sensing transcription factors are
species in those cells. In agreement to its specific role in cop- activated by Cu(I)-binding to their metal binding domains
per homeostasis, MymT knock-out mutants are more sensitive through conserved cysteine thiolates (16,17). Cu(I)-binding
to high copper concentrations but not to other metals or reac- triggers structural changes within their three-dimensional
tive oxygen and nitrogen species, further supporting the Cu(I) structure, thus enabling them to bind to specific DNA motifs
binding nature of MymT in vivo. Thus, by binding and seques- and to induce the transcription of the downstream MT genes
tration of Cu(I), the role of MymT is crucial in virulence and (16,17).
resistance in M. tuberculosis (12). The best characterized MT isolated from yeast is the Sac-
Although the identification of prokaryotic copper MTs has charomyces Cerevisiae MT. S. cerevisiae MT is encoded by the
so far been restricted to MymT homologues exclusively present CUP1 gene and the mature protein is 53 amino acids long, with
in Mycobacterium species, it cannot be excluded that copper the first 8 amino acids encoded by the Open Reading Frame
MTs present in other bacterial classes remain to be discov- (ORF) cleaved off and absent in the mature protein (18,19). MT
ered. MT sequences are short and often display a low level of expression in S. cerevisiae is exclusively induced by increased
complexity; thus, they can be easily overlooked in sequenced intracellular Cu(I) and Ag(I) levels, while Zn(II) and Cd(II) fail to
bacterial genomes (15). induce CUP1 transcription (18,19). Increased Cu(I) concentra-
tions result in Cu(I) binding (forming a Cu(I)-thiolate cluster) and
activation of the ACE1 transcription factor, which in turn binds
Yeast and Fungal Copper MTS to the promoter region upstream of the CUP1 gene thereby trig-
In yeasts and fungi, a large number of MT proteins have been gering MT gene transcription (17). According to the activation of
identified and studied. Most of the these yeast and fungal MTs expression profile under physiological conditions, yeast MT is
(belonging to families 8–13) are isolated exclusively as Cu-MTs isolated exclusively as Cu(I)-bound form.
and their expression is induced by Cu(I) (16). A number of The nuclear magnetic resolution (NMR) solution structure
studies have established their functional importance as major was initially determined for the native copper-containing form
intracellular copper chelators responsible for controlling intra- and for the NMR-active 109Ag(I) derivative, both showing that
cellular copper pools. As a consequence of their Cu(I)-binding 10 of the 12 available cysteines bind 7 Cu(I) or Ag(I) ions in a
nature, MTs play key roles in regulating available copper single cluster (20). However, as it was uncertain that Cu/Ag

238 Copper Metallothioneins


Yeast MT. (A) Crystal structure of S. cerevisiae Cu(I)8-MT (PDB ID: 1RJU) (22). Cu(I) ions are shown as orange spheres, and
FIG 2 CysS-Cu(I) connectivities are presented as dashed lines. (B) Structure of the Cu(I)8–thiolate cluster in S. cerevisiae Cu(I)8-MT.
Thiolate sulfur atoms from Cys10 and Cys22 coordinate 3 Cu(I) and connect the two distorted Cu(I)- tetrahedron structures (22).
The figure was generated with Chimera.

isomorphous replacement can be assumed, it was realized copper sequestering molecule in vivo similarly to S. cerevisiae
that the structure of the Cu7-cluster derived from the 109Ag(I)7- MT, despite in vitro also divalent metal ions can be bound. The
MT derivative might have not been necessarily correct as the sequence of N. crassa MT is 25 amino acids long and contains 7
coordination number for Cu(I) tends to be larger than that for Cys residues all involved in metal coordination. Circular dichro-
Ag(I) (21). ism, electronic absorption and luminescence spectroscopic
Structural determination by X-ray crystallography of the characterization revealed the presence of a Cu(I)6S-thiolate
Cu(I)8-MT has provided definitive details on the structural cluster in which all the Cys residues are involved in metal coor-
properties of the metal-thiolate cluster in yeast Cu(I)-MT (22) dination. The fold of the polypetide chain has been determined
(Fig. 2A). The yeast MT sequence contains 12 cysteine resi- by solution NMR but the direct Cys-Cu(I) connectivity could not
dues. However, only 10 of the 12 residues are involved in Cu(I) be determined due to the NMR-insensitive nature of the Cu(I)
coordination. The two Cys residues located closest in the quadrupolar nuclei (23). Interestingly, the native-like Cu(I)6-MT
sequence the C-terminus are not involved in metal binding. could be obtained in vitro only by metal substitution from the
The structure revealed a single Cu(I)8-thiolate cluster core sur- Zn(II)3-MT. Contrarily, only Cu(I)4-MT species could be obtained
rounded by the polypeptide chain in which defined secondary from the direct Cu(I) incorporation into the apo-MT, suggesting
structure elements are lacking. Six of the eight bound Cu(I) diverse pathway for the cluster assembly in the two cases. The
are trigonally coordinated each by three cysteine thiolate sul- structure revealed the lack of classic secondary structural ele-
fur atoms, while the two remaining Cu(I) are coordinated digo- ments with the N-terminal and C-terminal halves wound around
nally by two cysteine thiolate sulfurs (Fig. 2B). The Cu(I) atoms the Cu(I)-thiolate core in a left- and right-handed fashion,
are arranged in two groups of four, each assuming two con- respectively. A similar Cu-MT containing a Cu(I)6-thiolate core
nected distorted tetrahedron structures. In one of these tetra- has been isolated from the mushroom Agaricus bisporus, and
hedron, the Cu–Cu distances range between 2.59-2-91 Å, while spectroscopic analysis suggested a similar cluster organization
in the other longer distances (2.78–3.12 Å) are observed. The as in N. crassa MT (24).
overall structure and the short Cu–Cu distances indicate the Additonal insights about the physiological roles of Cu(I) MTs
presence of a compact Cu(I)-thiolate core shielded from solvent in fungi has been obtained in Podospora anserina. P. anserina
access. Accordingly, strong luminescence emissive features possesses a Cu(I)-inducible MT whose expression progressively
are observed in agreement with the shielded nature of copper- increases during aging. It has been demonstrated that alteration
thiolate cluster. Of the 10 coordinating Cys thiolate sulfurs, in the turnover of mitochondrial respiratory complex proteins
8 coordinate 2 Cu(I) in a l2-bridging fashion while the remain- during aging is responsible for the release of copper ions bound
ing 2 Cys sulfurs bind 3 Cu(I) and connect the 2 distorted tetra- to cytochrome c oxidase. Increasing copper concentration due to
hedron structures. these processes in turn causes the induction of P. anserina MT
Despite the structure of the yeast MT is the only structure expression, further supporting its role in copper sequestration
available for a copper MT other Cu(I)-MTs from yeasts and fungi and detoxification in yeast and fungal MTs (25).
have been identified and characterized. The expression of Neu- Other Cu(I) MTs have been identified in yeasts or fungi,
rospora crassa MT is exclusively induced by Cu(II) and not by but detailed characterization of their structure, reactivity, and
Zn(II), Cd(II), Co(II), or Ni(II), supporting its role as an exclusive physiological roles remain more elusive.

Calvo et al. 239


IUBMB LIFE

Snail MTs. Sequence alignment of the three MT isoforms from the Roman snail Helix Pomatia. The conserved Cys residues are
FIG 3 highlighted in yellow and the metal selectivity for each isoform is indicated on the left.

Copper MTS in Molluscs Overall, it appears that cell-type specific expression com-
bined with evolution of metal selectivity preferences achieved
Studies of MTs from pulmonate gasteropods have proven
by modulation of noncysteine amino acid positions result in
instrumental to investigate how metal specificity can be
the generation of specific homometallic species, at least in
achieved in MTs. In the snail Helix Pomatia, two different MTs
snail. As a result, functional HpCuMT and HpCdMT species
showing selective binding of cadmium and copper, respective-
show a metal composition tailored to satisfy their specialized
ly, have been identified. Interestingly, these two MT isoforms
localization and cellular function (i.e., HpCuMT in rhogocytes).
possess specific tissue expression (26). A third MT isoform,
recovered as a mixed Cd21- and Cu1-containing complex, also
exists, but considering its low abundance, it probably is less Mammalian Copper MTs
important to the snail’s metal metabolism. The first isoform In mammals, four distinct MT isoforms have been identified.
isolated from Cd-exposed Roman snails showed binding of They are designated MT-1 through MT-4.
6 Cd(II) ions (27), while the second isoform from the same spe- In certain species, including Homo sapiens, genetic poly-
cies was isolated with 12 Cu(I) equivalents, and they have morphism occurs with at least seven MT-1 isoforms expressed,
been named HpCdMT and HpCuMT, respectively (28). These while in others, such as Mus musculus, single MT gene exists
two isoform are 66 and 64 amino acids long and possess a (MT-1, 22, 23, 24). Mammalian MTs are single polypetide
conserved array of 18 Cys residues in corresponding positions chains of 61–68 amino acids characterized by a conserved
(Fig. 3). Studies of metal binding to the two isoforms expressed array of 20 Cys residues, with no aromatic or histidine resi-
in E. coli in the presence of the cognate metals confirmed the dues present in the sequence. Cysteine residues are clustered
homothallic nature of the isolated products. In HpCd6MT, in Cys–X–Cys, Cys–Cys and Cys–X–X–Cys motifs characteristic
Cd(II) ions are bound in tetrahedral coordination each by four of the vertebrate MT family (Family 1, according to Binz and
thiolate ligands in the form oligonuclear metal thiolate clus- Ka€ gi) (2).
ters, for which the structure remains to be determined. In MT-1 and MT-2 are inducible proteins and ubiquitously
Cu12-HpCuMT, spectroscopic analysis indicate that Cu(I) is expressed in several organs including liver, kidney, intestine,
organized in the form of oligonuclear Cu(I) thiolate clusters and brain. MT-3 and MT-4 expression is instead restricted
with features very similar to mammalian Cu12-MT in which mainly to the central nervous system (CNS) and stratified epi-
metals are bound predominantly in trigonal coordination thelia, respectively, and their expression is not inducible by
(29,30). Stability of the expressed Cd6-HpCdMT and Cu12- the classic stimuli of MT-1/-2 (4,32,33). MT-1 and MT-2 are
HpCuMT complexes was demonstrated by their metal major zinc-binding proteins in the cells and they are overex-
exchange inertness. Contrarily, expression in the presence of pressed in response to heavy metal exposure. A large number
the noncognate metal partners resulted in multiple heteroge- of toxicological studies have revealed that cadmium binding to
neous heterometallic species containing both Cu(I) and Zn(II)/ MT-1 and MT-2 isoforms represent a fundamental protective
Cd(II) ions (29,30). In light of the fact that heterothallic species effect for heavy metal cellular detoxification. In agreement to
can be generated when exposed to noncognate metals, abso- this role, increased MT content in liver, kidney, and intestines
lute selectivity cannot result exclusively from difference in the occurs following the parenteral or dietary administration of
noncysteine residues in their primary sequence (29,30). cadmium, zinc, or copper to experimental animals (1,4). Cad-
HpCuMT is constitutively expressed in a specialized molluscan mium detoxification is a property of MTs rather than its evolu-
cell type, the rhogocyte, which is the site of synthesis of the tionary function. As MTs constitute a major fraction of zinc-
copper protein hemocyanin. It is likely that specific cellular binding proteins in the cell under philological conditions,
localization exposes each synthesized isoforms to environ- mammalian MTs can potentially modulate many important
ments with unique and diverse metal buffering capabilities. It biological processes that involve zinc-requiring enzymes. It has
is expected that HpCuMT has an affinity for copper high been demonstrated that MT-1/-2 can directly activate zinc
enough to efficiently compete with other copper buffering mol- enzymes by direct zinc transfer, inactivate zinc-finger tran-
ecules in rhogocytes cytosol (31). On the other hand, expres- scription factors via zinc removal, and indirectly modulate
sion of HpCdMT appears to be heavily regulated by exposure zinc-dependent processes by controlling zinc availability in the
to increasing concentrations of cadmium in the snail midgut cell (1).
gland and epithelial cells of foot, gut, and kidney, thus playing As a consequence of their fundamental properties in regu-
a major role in metal detoxification (31). lating physiological Zn(II) concentrations and their role in

240 Copper Metallothioneins


Mammalian MTs. (A) Overview of the crystal structure of Cd5,Zn2-MT-2 isolated from rat (PDB ID: 4MT2). Two Zn(II) and one
FIG 4 Cd(II) are located in the N-terminus b-domain, and the other four Cd(II) in the C-terminus a-domain. The figure was generated
with Chimera. (B) Spectroscopic model of Cu(I)4CysS6-7 clusters in Cu(I)8-MT (40). (C) Sequence alignment of human MT-1, 22,
23, and 24 along with rat MT-2. The conserved 20 Cys residues are highlighted in yellow.

detoxification of divalent toxic metals (e.g., Cd(II)), a wealth of coordination. Differently from M(II) which are each coordinat-
information is available regarding the structural properties of ed in tetrahedral geometry by four Cys ligands, Cu(I) ions are
M(II)-bound mammalian MTs. The 3D structures have been coordinated digonally and trigonally by 2–3 sulfurs from bridg-
obtained for rat Cd5, Zn2MT-2 (34), and rat (35), rabbit (36), ing and terminal cysteine ligands. Evidence for the existence
and human (37) Cd7MT-2, as well as the mouse (38) and of a distinct stable MT complexes with 8 Cu(I) equivalents
human (39) a-domain of Cd4-MT-3. These structures showed bound (Cu(I)8-MT) has been obtained, indicating that two sepa-
identical metal–thiolate clusters organization and very similar rated Cu(I)4-thiolate clusters are formed in the pathway of
protein folds. Thus, mammalian MTs show a dumbbell-like cluster assembly. In these clusters, Cu(I)–Cu(I) distances are
topology with two separated domains linked by flexible hinge shorter than 2.8 Å allowing d10–d10 orbital overlap resulting in
region each encompassing a Metal-thiolate cluster. A a certain degree of metal–metal bonding character in the cop-
M(II)3CysS9 is present in the N-terminal b-domain and a per core (Fig. 4B) . In the presence of increasing Cu(I)-to-pro-
M(II)4CysS11 in its C-terminal domain (with M(II) 5 Cd(II) or tein ratios, the Cu(I)4S(6-7) cores can be expanded to Cu(I)6-
Zn(II)) with all the 20 conserved cysteines involved in metal Cys9 clusters, resulting in an increase in Cu–Cu distance to
coordination. All metals in the clusters show tetrahedral coor- >2.8Å (40). Thus, cluster structures and metal coordination
dination by l2-bridging and terminal cysteine thiolates (Fig. geometries are different in Cu(I)-MT and Zn(II)-MT forms. As a
4A). consequence, a different polypeptide arrangement around the
When mammalian MTs are isolated from livers of various metal core of Cu(I)-MT and Zn(II)-MT occurs (41).
species MT-1 and MT-2 usually contain seven Zn(II) ions. MT- In the human brain, besides MT-1/MT-2, the MT-3 isoform
3 and MT-4 are instead isolated from brain and epithelia, is also present. When isolated from human, bovine, and equine
respectively, as mixed species containing Zn(II) and Cu(I) (4). brains, MT-3 contains four Cu(I) ions and 3–4 Zn(II). MT-3 has
However, in situations of cellular copper overload, such as been discovered by Uchida et al. and named neuronal growth
Wilson’s disease, MT plays major role in Cu(I) binding and inhibitory factor, as MT-3 possess distinct biological functions
detoxification, and MT-1/-2 species containing predominantly that are not shared by other MT isoforms (32). MT-3 antago-
Cu(I) ions can be isolated from the liver (1). nizes the ability of Alzheimer’s disease brain extract to stimu-
Presently, no 3D structure of the Cu(I)-containing mamma- late survival and neuritic sprouting of cultured neurons, pos-
lian MT-1 and MT-2 isoforms is available. However, spectro- sesses extracellular growth inhibitory activity in neuronal
scopic and biophysical investigations have revealed that Cu(I)- primary cultures, and protects neuronal cells from the toxic
loaded MT can bind up to 12 Cu(I) ions organized in two sepa- effect of amyloid-b (Ab) peptide Ab1–40, suggesting that MT-3
rated Cu(I)6-thiolate clusters, with even higher stoichiometries may be involved in protection from pathogenic processes lead-
reported. These clusters are located in the b- and a-domains ing to Alzheimer’s disease (AD) (4). Besides the CNS, lower
with all the conserved 20 Cys residues involved in metal MT-3 expression has been subsequently reported also in renal

Calvo et al. 241


IUBMB LIFE

epithelial cells (42) and human prostate (43) as well as in sev- protein misfolding, aggregation into soluble oligomers and
eral cancers cells including urinary bladder (44), breast (45), insoluble amyloid aggregates, and copper-catalyzed ROS pro-
and prostate(46), suggesting that MT-3 could represent a bio- duction via Haber-Weiss and Fenton-type chemistry. In these
marker for cell growth and/or proliferation. neurodegenerative diseases, the expression of MT-3 is found
The differences in amino acid sequences are responsible to be altered (4).
for the observed bioactivity in the CNS. MT-3 sequence con- In light of its reactivity toward copper, Zn7MT-3 can effi-
tains two unique features when compared with MT-1/-2 iso- ciently scavenge Cu(II) from soluble and aggregated Ab-Cu(II)
forms (Fig. 4C): (1) a threonine in position 5 in the N-terminal complexes as well as from soluble oligomers via a metal-swap
b-domain followed by a conserved C6PCP9 motif and (2) an reaction generating the described redox-inert Cu(I)4Zn4MT-3
acidic hexapeptide in the C-terminal domain. Native monomer- species and Ab-Zn(II) (50). These reaction underlie the abolish-
ic Cu(I)4Zn3-4MT-3 contains two homometallic clusters, a Cu(I)- ment of copper redox cycling and concomitant radical produc-
thiolate cluster and a Zn(II) cluster. By direct reconstitution of tion, providing a protective effect against the toxicity of Ab-
Cu(I) and Zn(II) in the recombinantly expressed apoMT-3, the Cu(II). Accordingly, the toxic effect of Ab1–40-Cu(II) in cell cul-
Cu(I)4-thiolate cluster was demonstrated to be located in the ture was abolished thereby providing a possible molecular
N-terminal b-domain, while the Zn(II)-cluster in the C-terminal mechanism explaining the protective effect by MT-3 against
domain. While the Cu(I)4-thiolate cluster shows a remarkable Ab toxicity shown in previous studies. Similar metal-swapping
stability toward oxidation, the C-terminal Zn(II)4-thiolate clus- reactions between a number of different PrP-Cu(II) complexes
ter is susceptible to oxidation with consequent release of 1 as well as a-synuclein-Cu(II) complexes resulting in copper
Zn(II) ion and concomitant formation of Cu(I)4Zn3MT-3. scavenging and formation of Cu(I)4Zn4MT-3 substantiate a
Details on Cu(I) binding to isolated MT-3 domains have putative and more general role played by MT-3 in controlling
been characterized. Binding of Cu(I) to the N-terminal b- aberrant protein-Cu(II) interaction in the brain (51,52).
domain revealed the initial cooperative formation of a The expression of the last mammalian isoform discovered
Cu(I)4S(6-7) cluster which could be subsequently expanded to a by Quaife and Palmiter et al., MT-4, is restricted to cornified
Cu(I)6S9 core by further addition of Cu(I) (47). Contrarily, in and stratified epithelia, where a role in metal metabolism dur-
the a-domain, a Cu(I)4S7-8 cluster is formed that can be further ing differentiation has been proposed (33). When isolated from
expanded to generate a Cu(I)6S11(48). Characterization of the tongue epithelium, MT-4 is purified as a mixed copper and
low-temperature luminescence properties confirmed that, as zinc form, substantiating its role in the homeostasis of both
observed for MT-1/-2, the expansion of the Cu(I)4-thiolate metals (33,53). MT-4 expression in E. coli with media supple-
cores to Cu(I)6-thiolate cores is responsible for increased Cu– mented with zinc, cadmium, or copper indicated that MT-4
Cu distances and disruption of d10–d10 orbital overlap which has a stronger preference for Cu(I) over divalent metal ions
might be pivotal in the air-stability of the Cu(I)4 cluster toward Zn(II) and Cd(II) when compared with MT-1, resulting in the
oxidation (47,48). formation of heterometallic mixed species. By in vitro titration,
Besides being a neuronal intracellular protein, MT-3 can it could be demonstrated that full-length MT-4 could bind up
be efficiently secreted in the brain extracellular space. A pro- to 10 Cu(I) to generate Cu(I)10-MT-4, while Cu(I)5-aMT-4 and
tective role of extracellular Zn7MT-3 from Cu(II) toxicity has Cu(I)7-bMT-4 species could be formed and isolated in the a-
been suggested. Zn7MT-3 thiolates can efficiently reduce Cu(II) and b- domains, respectively (54). Based on these results and
to Cu(I) and bind it to form Cu(I)4Zn4MT-3 via cooperative for- in silico protein sequence analysis, MT-4 was classified as a
mation of an air stable Cu(I)4-thiolate cluster in its b-domain novel copper–thionein, made up of two copper–thionein
(49). In this process, the three Zn(II) ions are released and two domains (54). The discovery of the participation of sulfide
disulfide bonds, likely located in the a-domain, are generated. anions as additional ligands in the metal clusters of Zn/Cd-MT-
The formed Cu(I)4Zn4MT-3 species are redox inert, and the 4 forms, generated in vivo via recombinant expression, has
observed quenching of the ascorbate-driven and Cu(II)-cata- pioneered the discovery of S2- as additional metal ligands in a
lyzed production of ROS highlight the stability of the protein series of other MTs from different species (recombinant and
and its capability to act as a protective factor from Cu(II) toxic- native) (55). Despite S2- incorporation does not commonly
ity. Overall, these results indicate that MT-3 b-domain possess occur in Cu-bound MT forms, the discovery that the higher sul-
higher reactivity and affinity toward copper ions over Zn(II) fide content in the cadmium-loaded MTs is obtained in MT-
compared with the a-domain (49). isoforms possessing the higher the copper–thionein character
In light of the reactivity of MT-3 toward Cu(II), a major has provided to the community further criterion for the classi-
role played by MT-3 in controlling aberrant copper–protein fication of MTs (56).
interactions in neurodegenerative disorders has been pro-
posed. Cu(II) binding to Ab peptides in AD (the major compo-
nent of extracellular amyloid plaques in Alzheimer’s diseases), Other Characterized MTS
to prion protein in disorders like Creutzfeldt–Jakob disease, A number of other MT isoforms from different species have
and to a-synuclein (a-Syn) in Parkinson’s disease has been been characterized and, for some, their Cu(I) binding ability
implicated as disease-potentiating factor contributing to demonstrated, including MTs belonging to protozoan (57),

242 Copper Metallothioneins


arthropoda (58), crustacea (59), molluscs (26,60), echinoderms [2] Binz, P. A. and Ka €gi, J. H. R. (1999) Metallothionein: molecular evolution and
classification. In Metallothioenein IV (Klaassen, C., ed.). pp. 7 – 13, Birkha €user
(60), nematodes (61), and plants (some of which show Cu(I)-
Verlag, Basel.
thionein character) (62). Complete 3D structures are available [3] Thornalley, P. J. and Va sak, M. (1985) Possible role for metallothionein in
from the cadmium-bound forms of some of these MTs includ- protection against radiation-induced oxidative stress. Kinetics and mecha-
ing crustacean blue crab Cd6-MT-1 (Callinectes sapidus) (63), nism of its reaction with superoxide and hydroxyl radicals. Biochim. Biophys.
lobster Cd6MT-1 (Homarus americanus) (64), sea urchin Acta 827, 36 – 44.
[4] Va k, M. and Meloni, G. (2011) Chemistry and biology of mammalian metal-
sa
Cd7MTA (Strongylocentrotus purpuratus) (65), and fish
lothioneins. J. Biol. Inorg. Chem. 16, 1067 – 1078.
Cd7MT_nc (Notothenia coriiceps) (66) (all of which are mono- [5] Stillman, M. J. (1995) Metallothioneins. Coord. Chem. Rev. 144, 461 – 511.
meric proteins with two globular domains each encompassing [6] Romero-Isart, N. and Va k, M. (2002) Advances in the structure and chemis-
sa
a metal thiolate cluster), as well as wheat MT Ec-1 (67,68). try of metallothioneins. J. Inorg. Biochem. 88, 388 – 396.
Although some investigations have been undertaken to under- [7] Good, M., Hollenstein, R., and Va sak, M. (1991) Metal selectivity of clusters in
rabbit liver metallothionein. Eur. J. Biochem. 197, 655 – 659.
stand Cu(I) binding to some of these MTs, no other 3D struc-
[8] Stillman, M. J. (1992) Metallothionein. In Metallothioneins (Stillman, M. J.,
tures of Cu(I)-bound MT has been obtained so far, limiting our Shaw, C. F. I., and Suzuki, K. T., eds.). pp. 55 – 127, VCH, New York.
understanding on conserved and divergent features in the [9] Valls, M., Bofill, R., Gonzalez-Duarte, R., Gonzalez-Duarte, P., Capdevila, M.,
structure, reactivity, and functional properties of copper MTs. et al. (2001) A new insight into metallothionein (MT) classification and evolu-
tion—the in vivo and in vitro metal binding features of Homarus americanus
recombinant MT. J. Biol. Chem. 276, 32835 – 32843.
Conclusions [10] Olafson, R. W., Abel, K., and Sim, R. G. (1979) Prokaryotic metallothionein:
preliminary characterization of a blue-green alga heavy metal-binding pro-
Since their discovery, MTs emerged as a central and conserved tein. Biochem. Biophys. Res. Commun. 89, 36 – 43.
protein class involved in both essential metal homeostasis and [11] Blindauer, C. A., Harrison, M. D., Parkinson, J. A., Robinson, A. K., Cavet, J.
toxic metal detoxification in all the kingdoms of life. Character- S., et al. (2001) A metallothionein containing a zinc finger within a four-
metal cluster protects a bacterium from zinc toxicity, Proc. Natl. Acad. Sci.
ization of MTs isolated from a wide variety of organisms and
USA 98, 9593 – 9598.
tissues, together with in silico protein sequence analysis sug- [12] Gold, B., Deng, H., Bryk, R., Vargas, D., Eliezer, D., et al. (2008) Identification
gest that specific MT isoforms evolved to play preferential of a copper-binding metallothionein in pathogenic mycobacteria. Nat.
roles in controlling copper homeostasis over other transition Chem. Biol. 4, 609 – 616.
metals. In recent years, it has been demonstrated that the [13] Robinson, N. J. (2008) A bacterial copper metallothionein. Nat. Chem. Biol.
4, 582 – 583.
metal-selectivity and metal-composition in MTs builds on com-
[14] Rowland, J. L. and Niederweis, M. (2012) Resistance mechanisms of Myco-
plex factors arising from a strict interplay between the prima- bacterium tuberculosis against phagosomal copper overload. Tuberculosis
ry sequence, the pattern of the conserved coordinating cys- (Edinb) 92, 202 – 210.
teines, the nature of the neighboring non-coordinating amino [15] Blindauer, C. A. (2011) Bacterial metallothioneins: past, present, and ques-
acids, the metal composition of the media/cellular milieu tions for the future. J. Biol. Inorg. Chem. 16, 1011 – 1024.
[16] Dolderer, B., Hartmann, H. J., and Weser, U. (2009) Metallothioneins in
where the MTs are expressed, and the nature of metals induc-
yeast and fungi. Met. Ions Life Sci. 5, 83 – 105.
ing MT expression in vivo. The functional classification of MTs [17] Furst, P., Hu, S., Hackett, R., and Hamer, D. (1988) Copper activates metallo-
as zinc- or copper-thioneins taking into account several of thionein gene transcription by altering the conformation of a specific DNA
these aspects has significantly contributed to our understand- binding protein. Cell 55, 705 – 717.
ing the pleiotropic chemistry of MTs (56). However, the a pri- [18] Butt, T. R., Sternberg, E. J., Gorman, J. A., Clark, P., Hamer, D., et al. (1984)
Copper metallothionein of yeast, structure of the gene, and regulation of
ori prediction of the metal binding character and properties
expression. Proc. Natl. Acad. Sci. USA 81, 3332 – 3336.
still remains challenging when sequence and functional homol- [19] Karin, M., Najarian, R., Haslinger, A., Valenzuela, P., Welch, J., et al. (1984)
ogy among different MTs are divergent. Presently, the under- Primary structure and transcription of an amplified genetic locus: the CUP1
standing of the chemistry and biology of Cu-MTs has not yet locus of yeast. Proc. Natl. Acad. Sci. USA 81, 337 – 341.
reached the level obtained for Zn(II)-MTs. Several questions [20] Peterson, C. W., Narula, S. S., and Armitage, I. M. (1996) 3D solution struc-
ture of copper and silver-substituted yeast metallothioneins. FEBS Lett. 379,
such as how protein sequence and structure control preferen-
85 – 93.
tial metal selectivity in Cu-MTs, how metal buffering and avail- [21] Bertini, I., Hartmann, H. J., Klein, T., Liu, G., Luchinat, C., et al. (2000) High
ability in distinct cellular and tissue localizations determines resolution solution structure of the protein part of Cu7 metallothionein. Eur.
the generation of Cu-MTs, as well as specific and unique physi- J. Biochem. 267, 1008 – 1018.
ological function of Cu-MTs remain to be answered. More [22] Calderone, V., Dolderer, B., Hartmann, H. J., Echner, H., Luchinat, C., et al.
(2005) The crystal structure of yeast copper thionein: the solution of a long-
work is yet to be carried out to understand the chemistry and
lasting enigma. Proc. Natl. Acad. Sci. USA 102, 51 – 56.
biology of Cu-MTs and intriguing new findings are likely to [23] Cobine, P. A., McKay, R. T., Zangger, K., Dameron, C. T., and Armitage, I.
emerge. M. (2004) Solution structure of Cu6 metallothionein from the fungus Neu-
rospora crassa. Eur. J. Biochem. 271, 4213 – 4221.
[24] Munger, K. and Lerch, K. (1985) Copper Metallothionein from the fungus
References Agaricus-Bisporus—chemical and Spectroscopic Properties. Biochemistry
[1] Vasa
k, M. and Romero-Isart, N. (2005) Metallothioneins. In Encyclopedia of 24, 6751 – 6756.
Inorganic Chemistry, 2nd ed. (King, R. B. ed.). pp. 3208 – 3221, Wiley, New [25] Averbeck, N. B., Borghouts, C., Hamann, A., Specke, V., and Osiewacz, H. D.
York. (2001) Molecular control of copper homeostasis in filamentous fungi:

Calvo et al. 243


IUBMB LIFE

increased expression of a metallothionein gene during aging of Podospora [45] Sens, M. A., Somji, S., Garrett, S. H., Beall, C. L., and Sens, D. A. (2001)
anserina. Mol. Gen. Genet. 264, 604 – 612. Metallothionein isoform 3 overexpression is associated with breast cancers
[26] Dallinger, R., Berger, B., Hunziker, P., and Ka €gi, J. H. (1997) Metallothionein having a poor prognosis. Am. J. Pathol. 159, 21 – 26.
in snail Cd and Cu metabolism Nature 388, 237 – 238. [46] Juang, H. H., Chung, L. C., Sung, H. C., Feng, T. H., Lee, Y. H., et al. (2013)
[27] Dallinger, R., Wang, Y. J., Berger, B., Mackay, E. A., and Ka €gi, J. H. R. (2001) Metallothionein 3: an androgen-upregulated gene enhances cell invasion
Spectroscopic characterization of metallothionein from the terrestrial snail, and tumorigenesis of prostate carcinoma cells. Prostate 73, 1495 – 1506.
Helix pomatia. Eur. J. Biochem. 268, 4126 – 4133. [47] Faller, P., and Va k, M. (1997) Distinct metal-thiolate clusters in the N-
sa
[28] Gehrig, P. M., You, C., Dallinger, R., Gruber, C., Brouwer, M., et al. (2000) terminal domain of neuronal growth inhibitory factor. Biochemistry 36,
Electrospray ionization mass spectrometry of zinc, cadmium, and copper 13341 – 13348.
metallothioneins: evidence for metal-binding cooperativity. Protein Sci. 9, [48] Hasler, D. W., Faller, P., and Va k, M. (1998) Metal-thiolate clusters in the
sa
395 – 402. C-terminal domain of human neuronal growth inhibitory factor (GIF). Bio-
[29] Palacios, O., Pagani, A., Perez-Rafael, S., Egg, M., Hockner, M., et al. (2011) chemistry 37, 14966 – 14973.
Shaping mechanisms of metal specificity in a family of metazoan metallo- [49] Meloni, G., Faller, P., and Va k, M. (2007) Redox silencing of copper in
sa
thioneins: evolutionary differentiation of mollusc metallothioneins. BMC metal-linked neurodegenerative disorders: reaction of Zn7metallothionein-3
Biol. 9, 4. with Cu21 ions. J. Biol. Chem. 282, 16068 – 16078.
[30] Palacios, O., Perez-Rafael, S., Pagani, A., Dallinger, R., Atrian, S., et al. [50] Meloni, G., Sonois, V., Delaine, T., Guilloreau, L., Gillet, A., et al. (2008) Met-
(2014) Cognate and noncognate metal ion coordination in metal-specific al swap between Zn7-metallothionein-3 and amyloid-b-Cu protects against
metallothioneins: the Helix pomatia system as a model. J. Biol. Inorg. amyloid-b toxicity. Nat. Chem. Biol. 4, 366 – 372. [PMC][10.1038/nchem-
Chem. 19, 923 – 935. bio.89] [18454142]-
[31] Foster, A. W. and Robinson, N. J. (2011) Promiscuity and preferences of [51] Meloni, G., Crameri, A., Fritz, G., Davies, P., Brown, D. R., et al. (2012) The
metallothioneins: the cell rules. BMC Biol. 9, 25. catalytic redox activity of prion protein-Cu(II) is controlled by metal
[32] Uchida, Y., Takio, K., Titani, K., Ihara, Y., and Tomonaga, M. (1991) The exchange with the Zn(II)-thiolate clusters of Zn(7) metallothionein-3. Chem.
growth inhibitory factor that is deficient in the Alzheimer’s disease brain is Bio. Chem. 13, 1261 – 1265.
a 68 amino acid metallothionein-like protein. Neuron 7, 337 – 347. [52] Meloni, G., and Va k, M. (2011) Redox activity of alpha-synuclein-Cu is
sa
[33] Quaife, C. J., Findley, S. D., Erickson, J. C., Froelick, G. J., Kelly, E. J., et al. silenced by Zn(7)-metallothionein-3. Free Radic. Biol. Med. 50, 1471 – 1479.
(1994) Induction of a new metallothionein isoform (MT-IV) occurs during [53] Meloni, G., Zovo, K., Kazantseva, J., Palumaa, P., and Vasak, M. (2006)
differentiation of stratified squamous epithelia. Biochemistry 33, 7250 – Organization and assembly of metal-thiolate clusters in epithelium-specific
7259. metallothionein-4. J. Biol. Chem. 281, 14588 – 14595.
[34] Braun, W., Vasak, M., Robbins, A. H., Stout, C. D., Wagner, G., et al. (1992) [54] Tio, L., Villarreal, L., Atrian, S., and Capdevila, M. (2004) Functional differen-
Comparison of the NMR solution structure and the x-ray crystal structure of tiation in the mammalian metallothionein gene family: metal binding fea-
rat metallothionein-2. Proc. Natl. Acad. Sci. USA 89, 10124 – 10128. tures of mouse MT4 and comparison with its paralog MT1. J. Biol. Chem.
[35] Schultze, P., Wo € rgo
€ tter, E., Braun, W., Wagner, G., Va k, M., et al. (1988)
sa 279, 24403 – 24413.
Conformation of [Cd7]-metallothionein-2 from rat liver in aqueous solution [55] Capdevila, M., Domenech, J., Pagani, A., Tio, L., Villarreal, L., et al. (2005)
determined by nuclear magnetic resonance spectroscopy. J. Mol. Biol. 203, Zn- and Cd-metallothionein recombinant species from the most diverse
251 – 268. phyla may contain sulfide (S2-) ligands. Angew. Chem. Int. Ed. Engl. 44,
[36] Arseniev, A., Schultze, P., Wo € rgo
€ tter, E., Braun, W., Wagner, G., et al. (1988) 4618 – 4622.
Three-dimensional structure of rabbit liver Cd7metallothionein-2a in aqueous [56] Palacios, O., Atrian, S., and Capdevila, M. (2011) Zn- and Cu-thioneins: a
solution determined by nuclear magnetic resonance. J. Mol. Biol. 201, 637 – functional classification for metallothioneins? J. Biol. Inorg. Chem. 16, 991 –
657. 1009.
[37] Messerle, B. A., Scha €ffer, A., Va
sak, M., Ka €gi, J. H., and W€ uthrich, K. (1990) [57] Santovito, G., Irato, P., Palermo, S., Boldrin, F., Sack, R., et al. (2001) Identifi-
Three-dimensional structure of human [113Cd7]metallothionein-2 in solution cation, cloning and characterisation of a novel copper-metallothionein in tet-
determined by nuclear magnetic resonance spectroscopy. J. Mol. Biol. 214, rahymena pigmentosa. Sequencing of cDNA and expression. Protist 152,
765 – 779. 219 – 229.
[38] € G., Zangger, K., and Armitage, I. M. (2001) Three-dimensional structure
Oz, [58] Valls, M., Bofill, R., Romero-Isart, N., Gonzalez-Duarte, R., Abian, J., et al.
and dynamics of a brain specific growth inhibitory factor: metallothionein-3. (2000) Drosophila MTN: a metazoan copper-thionein related to fungal forms.
Biochemistry 40, 11433 – 11441. FEBS Lett. 467, 189 – 194.
[39] Wang, H., Zhang, Q., Cai, B., Li, H., Sze, K. H., et al. (2006) Solution structure [59] Brouwer, M., Syring, R., and Hoexum Brouwer, T. (2002) Role of a copper-
and dynamics of human metallothionein-3 (MT-3). FEBS Lett. 580, 795 – specific metallothionein of the blue crab, Callinectes sapidus, in copper
800. metabolism associated with degradation and synthesis of hemocyanin. J.
[40] Pountney, D. L., Schauwecker, I., Zarn, J., and Va k, M. (1994) Formation
sa Inorg. Biochem. 88, 228 – 239.
of mammalian Cu8-metallothionein in vitro: evidence for the existence of [60] Vergani, L. (2009) Metallothioneins in aquatic organisms: fish, crustaceans,
two Cu(I)4-thiolate clusters. Biochemistry 33, 9699 – 9705. molluscs, and echinoderms. Met Ions Life Sci 5, 199 – 237.
[41] Dolderer, B., Echner, H., Beck, A., Hartmann, H. J., Weser, U., et al. (2007) [61] Stuerzenbaum, S. (2009) Earthworm and nematode metallothioneins. Met.
Coordination of three and four Cu(I) to the a- and b-domain of vertebrate Ions Life Sci 5, 183 – 197.
Zn-metallothionein-1, respectively, induces significant structural changes. [62] Tarasava, K., Loebus, J., and Freisinger, E. (2016) Localization and spectro-
FEBS J. 274, 2349 scopic analysis of the Cu(I) binding site in wheat metallothionein Ec-1. Int.
[42] Hoey, J. G., Garrett, S. H., Sens, M. A., Todd, J. H., and Sens, D. A. (1997) J. Mol. Sci. 17, 371.
Expression of MT-3 mRNA in human kidney, proximal tubule cell cultures, [63] Narula, S. S., Brouwer, M., Hua, Y., and Armitage, I. M. (1995) Three-dimen-
and renal cell carcinoma. Toxicol. Lett. 92, 149 – 160. sional solution structure of Callinectes sapidus metallothionein-1 deter-
[43] Garrett, S. H., Sens, M. A., Shukla, D., Nestor, S., Somji, S., et al. (1999) mined by homonuclear and heteronuclear magnetic resonance
Metallothionein isoform 3 expression in the human prostate and cancer- spectroscopy. Biochemistry 34, 620 – 631.
derived cell lines. Prostate 41, 196 – 202. [64] Munoz, A., Forsterling, F. H., Shaw, C. F. III, and Petering, D. H. (2002) Struc-
[44] Sens, M. A., Somji, S., Lamm, D. L., Garrett, S. H., Slovinsky, F., et al. (2000) ture of the (113)Cd(3)beta domains from Homarus americanus metallothio-
Metallothionein isoform 3 as a potential biomarker for human bladder can- nein-1: hydrogen bonding and solvent accessibility of sulfur atoms. J. Biol.
cer. Environ. Health Perspect. 108, 413 – 418. Inorg. Chem. 7, 713 – 724.

244 Copper Metallothioneins


[65] Riek, R., Precheur, B., Wang, Y., Mackay, E. A., Wider, G., et al. (1999) NMR [67] Loebus, J., Peroza, E. A., Bluthgen, N., Fox, T., Meyer-Klaucke, W., et al. (2011)
structure of the sea urchin (Strongylocentrotus purpuratus) metallothionein Protein and metal cluster structure of the wheat metallothionein domain gam-
MTA. J. Mol. Biol. 291, 417 – 428. ma-E(c)21: the second part of the puzzle. J. Biol. Inorg. Chem. 16, 683 – 694.
[66] Capasso, C., Carginale, V., Crescenzi, O., Di Maro, D., Parisi, E., et al. (2003) [68] Peroza, E. A., Schmucki, R., Guntert, P., Freisinger, E., and Zerbe, O. (2009)
Solution structure of MT_nc, a novel metallothionein from the antarctic fish The beta(E)-domain of wheat E(c)-1 metallothionein: a metal-binding
Notothenia coriiceps. Structure 11, 435 – 443. domain with a distinctive structure. J. Mol. Biol. 387, 207 – 218.

Calvo et al. 245

You might also like