Você está na página 1de 7

Journal of Inorganic Biochemistry 117 (2012) 140–146

Contents lists available at SciVerse ScienceDirect

Journal of Inorganic Biochemistry


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

Bis(L-cysteinato)zincate(lI) as a coordination compound that induces


metallothionein gene transcription without inducing cell-stress-related
gene transcription
Tomoki Kimura a,⁎, Kengo Yoshida b, c, Chika Yamamoto d, Minako Suzuki a, Tomoko Uno a, Masakazu Isobe a,
Hiroshi Naka e, Shuji Yasuike f, Masahiko Satoh g, Toshiyuki Kaji h, Masanobu Uchiyama b, c
a
Department of Toxicology, Faculty of Pharmaceutical Sciences, Setsunan University, 45–1, Nagaotoge-cho, Hirakata, Osaka 573–0101, Japan
b
Advanced Elements Chemistry Laboratory, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113–0033, Japan
c
Advanced Elements Chemistry Laboratory, the Institute of Physical and Chemical Research (RIKEN), Wako-shi, Saitama 351–0198, Japan
d
Department of Environmental Health, Faculty of Pharmaceutical Sciences, Hokuriku University, Ho-3 Kanagawa-machi, Kanazawa, Ishikawa 920–1181, Japan
e
Research Center for Materials Science, Nagoya University, Chikusa, Nagoya 464–8602, Japan
f
Faculty of Pharmaceutical Sciences, Hokuriku University, Ho-3 Kanagawa-machi, Kanazawa, Ishikawa 920–1181, Japan
g
Laboratory of Pharmaceutical Health Sciences, School of Pharmacy, Aichi Gakuin University, 1–100 Kusumotocho, Chikusaku, Nagoya 464–8650, Japan
h
Department of Environmental Health, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278–8510, Japan

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

Article history: Zinc is an essential micronutrient, deficiency of which results in growth retardation, immunodeficiency, and
Received 27 February 2012 neurological diseases such as dysgeusia. Several zinc coordination compounds are used for zinc supplemen-
Received in revised form 25 July 2012 tation; however, supplemented zinc ions have no specificity and interact with various groups of molecules.
Accepted 31 July 2012
Here, we found that, from a library of 30 zinc coordination compounds, bis(L-cysteinato)zincate(II), designat-
Available online 29 August 2012
ed Z01, functioned as a metallothionein (MT) inducer. Z01 induced MT expression mediated by the transcrip-
Keywords:
tion factor MTF-1, without inducing cell-stress-related heme oxygenase-1 gene expression at specific
Zn coordination compound concentration. The zinc ion was necessary for the MT induction. 65Zn incorporation following treatment
Metallothionein with 65Zn-labeled Z01 suggested that Z01 did not act as zinc ionophore despite its hydrophilicity. Electropho-
MTF-1 retic mobility shift assays revealed that Z01 facilitates MTF-1–MRE complex formation, and, by inference,
Cell-stress transfer of zinc from Z01 to MTF-1. Phosphorylated ERK levels were increased by ZnSO4 treatment but not
Transcription by Z01. Although our data do not definitely prove that Z01 is an MTF-1-specific activator, our observations
suggest that zinc coordination compounds can regulate zinc distribution and act as zinc donors for specific
molecules.
© 2012 Elsevier Inc. All rights reserved.

1. Introduction metal response element (MRE)-binding transcription factor-1 (MTF-1)


has six C2H2-type zinc fingers [6]. Zinc binding to all six fingers serves
Zinc is an essential micronutrient and plays a diverse role in many DNA-binding ability [7].
biological processes by serving as a structural component of proteins, Zinc has a variety of physiological activities. It suppresses tran-
an essential cofactor in enzymes, and a modulator of signal transduc- scription factor STAT3 activation in Th17 cells [8], by blocking the
tion cascades within the cell [1–3]. The intracellular zinc concentra- association of STAT3 with JAK kinase and gp 130. The hydrolase
tion is tightly regulated by various sets of proteins, including zinc (phosphatase) activity of protein tyrosine phosphatase 1B (PTP1B)
efflux and influx transporter protein families, such as the ZnT proteins is also inhibited by zinc [9]. Zinc increases phosphorylation of recep-
(SLC30A) and ZIP proteins (SLC39A), as well as zinc-binding proteins tor tyrosine kinases and triggers growth factor signals. Zinc also
and metallothioneins (MTs) [4]. Furthermore, at least three thousand inhibits adenylate cyclase activity and thereby inhibits cAMP signal-
proteins have one or more zinc-binding sites [5]. The functions of ing in N18TG2 neuroblastoma cells [10,11]. These facts suggest that
some proteins are directly modulated by zinc ions; for example, the function of zinc proteins may be modulated by intracellular zinc
trafficking in many cases. We hypothesize that zinc compounds are
useful for zinc delivery to modulate zinc protein function(s).
Specific changes in metal distribution by chemicals (coordination
⁎ Corresponding author at: Department of Toxicology, Faculty of Pharmaceutical
compounds and metal chelators) control cellular processes. Mutation
Sciences, Setsunan University, Japan. Tel./fax: +81 72 866 3107. of the ATP7B gene, which encodes a copper transporter that is
E-mail address: tomoki@pharm.setsunan.ac.jp (T. Kimura). expressed in hepatocytes and effluxes excess copper into the bile,

0162-0134/$ – see front matter © 2012 Elsevier Inc. All rights reserved.
http://dx.doi.org/10.1016/j.jinorgbio.2012.07.021
T. Kimura et al. / Journal of Inorganic Biochemistry 117 (2012) 140–146 141

leads to Wilson's disease, which is characterized by hyper-accumulation 2.2. Cell culture


of copper in the liver [12]. The copper chelator D-penicillamine, which
sequesters excess hepatic copper, thereby lowering the concentration, Mouse embryo fibroblasts (MEFs) prepared from wild-type (WT)
is an effective treatment for Wilson's disease. Water-solubilized zinc mice and MTF-1 knockout (MTF-KO) mice were transformed and se-
ionophores are reported to have potential as anti-cancer agents lected as described previously [41]. MEFs were cultured in Dulbecco's
[13,14]. Yoshikawa et al. found that zinc compounds could function as modified Eagle's medium (DMEM) — 4.5 g/L glucose supplemented
a new type of orally active anti-diabetic medicine [15]. Polaprezinc with 10% fetal bovine serum (FBS). BALB/3 T3 A31-1-1 cells, a
(catena-(S)-[μ-[Nα-(3-aminopropionyl) histidinato(2-)-N1, N2, O: Nτ]- mouse embryonic fibroblast cell line, were provided by the JCRB,
zinc]), a coordination compound consisting of zinc and L-carnosine, Japan, JCRB1356. Hepa 1–6 cells (mouse hepatoma cells), HepG2
improves hypogeusia, a symptom of zinc deficiency, without serious cells (human hepatoma cells), and HEK293 cells (human embryonic
side effects [16,17]. Polaprezinc also has an anti-ulcer effect and is used kidney cells) were provided by the RIKEN BRC (Tsukuba, Japan)
clinically to treat gastric ulcers in Japan. These examples suggest that through the National Bio-Resource Project of the MEXT, Japan.
regulation of the intracellular distribution of metals can effectively modu- These cells were cultured in DMEM — 1.0 g/L glucose supplemented
late cellular processes. However, the targets of these chemicals, for exam- with 10% FBS at 37 °C in 5% CO2/95% air. During experiment, cells
ple the copper donor of D-penicillamine and the protein(s) that receive were cultured in these medium containing FBS.
zinc from these zinc coordination compounds, have not been clarified yet.
Here, to examine whether zinc coordination compounds can serve 2.3. Isolation of RNA and quantification of specific mRNAs
as specific zinc protein functional modulators, we constructed a library
of zinc complexes. The transcription factor MTF-1 acts as cytosolic zinc Cells were cultured in 12-well culture plates and treated with
sensor to activate the expression of specific genes, including MT ZnSO4, CdCl2, or the indicated coordination compounds for 3 h before
and SLC30A1 [18]. MTF-1 has 6 Cys2His2-type zinc fingers. Canonical RNA extraction. Total RNAs were isolated with Isogen (Nippon Gene,
Cys2His2-type zinc fingers typically bind one zinc ion with high affinity Tokyo, Japan) according to the manufacturer's protocol. Reverse tran-
(10−9 − 10 −12 M). Each zinc finger in MTF-1 also bind one zinc ion, but scription was carried out with random hexamers and a PrimeScript
have zinc binding affinities in the nanomolar to sub-micromoler range. RT reagent kit (TaKaRa Bio, Otsu, Japan). The cDNAs were subjected
Zinc fingers in MTF-1 would be saturated with zinc under conditions of to real-time PCR analysis using mouse MT-I-, human MT-IIA, and
zinc excess. Zinc-sensing transcription by MTF-1 involves zinc binding human/mouse GAPDH-specific TaqMan® primers (Life Technologies,
to its unique zinc finger domain. So, MT-I mRNA levels were determined Carlsbad, CA, USA), as well as human HO-1 specific primers (forward
to assess MTF-1 activation, and cell stress-mediated gene transcription primer: 5′-agtcaggcagagggtgatagaag-3′, reverse primer: 5′-cataaag
was monitored by measuring the induction of heme oxygenase 1 ccctacagcaactgtc-3 ′) and mouse HO-1 specific primers (forward
(HO-1) mRNA, whose transcription is mediated by the cell-stress- primer: 5′-acagccccaccaagttcaaacagct-3′, reverse primer: 5′-ctgtcag
related transcription factor nuclear factor-erythroid 2-related factor 2 catcacctgcagctcctc-3′). The amount of MT or HO-1 mRNA was normal-
(Nrf2) [19,20]. From the library, we found a zinc complex that activates ized against the amount of GAPDH mRNA.
MT-I gene expression without inducing HO-1 gene expression. We also
determined the mechanism of this MTF-1 activation. 2.4. Transfection and luciferase reporter assay

2. Materials and methods The reporter plasmid pGL4.12-MT − 264/+42 containing the
mouse MT-I promoter, which spans bases − 264 to + 42 (relative to
2.1. Materials the transcription start site), was used for the MT-I promoter reporter
assay. The promoter constructs were cloned into a Firefly reporter
Pyrithione (Py; 1-hydroxypyridine-2-thione) and zinc pyrithione plasmid, pGL4.12[luc2CP], as previously described [41]. For transfec-
(ZnPy; 1-hydroxypyridine-2-thione zinc salt) were purchased from tion, FuGENE 6 (Roche, Basel, Switzerland) was used for MEFs. MEFs
Sigma (St. Louis, MO, USA). The zinc coordination compounds were were cultured in 12-well culture plates and transfected at 70%–80%
synthesized using previously described protocols referenced in Table 1 confluence using FuGENE 6 according to the manufacturer's proto-
(Z01, Z02 and Z05) and Supplemental Table S1 (Z06–Z42) [21–40]. cols. Briefly, 3 μL of FuGENE 6 was diluted with 100 μL of serum-
Zinc acetylacetonate (Z27) was purchased from Sigma-Aldrich. and antibiotic-free Opti-MEM medium (Life Technologies). Then
(Toluene-3,4-dithiolato)zinc (Z28) and bis(8-quinolinolato)zinc 1.0 μg of Firefly reporter plasmid pGL4.12-MT–264/+42 and
(Z32) were purchased from Tokyo Chemical Industry (Tokyo, Japan). 0.25 ng of the control Renilla reporter plasmid pRL-SV40 were

Table 1
Zinc coordination compounds.

No Name Structure Formula, MW Reference

Z01 Na2[Zn(cys)2] C6H22Na2O10N2S2Zn, 457.74 [21]

Z02 Zn(his)2 C12H18N6O6Zn, 407.7 [21]

Z05 Zn(mal)2 C12H15O8.5Zn, 360.64 [22]


142 T. Kimura et al. / Journal of Inorganic Biochemistry 117 (2012) 140–146

added; the mixture was incubated for 15 min at room temperature. 2.10. Western blotting analysis
The FuGENE 6:DNA complex remained on the cells for 24 h. For the
luciferase assay, the cells were lysed using 1 × Passive Lysis Buffer MEFs were pre-cultured for 24 h in DMEM — 1.0 g/L glucose
(Promega, Madison, WI, USA), and luciferase activity was measured supplemented with 1% FBS. After the pre-culture, MEFs were washed
using the Dual-Luciferase Reporter Assay System (Promega) and a three times with ice-cold PBS, scraped into PBS, and collected by
luminometer (GloMax 20/20n Luminometer; Promega). Firefly lucif- centrifugation. Cell lysate was prepared by sonication in lysis buffer
erase activity was normalized to Renilla luciferase activity for normal- (20 mM Tris, pH 7.5; 150 mM NaCl; 1 mM EDTA; 1 mM EGTA; 1% Triton
ization of the transfection efficiency. X-100; 2.5 mM sodium pyrophosphate; 1 mM β-glycerol phosphate;
1 mM Na3VO4; 1 mg/mL leupeptin; 1 mM phenylmethylsulfonyl fluo-
2.5. Quantification of intracellular zinc ride). Loading buffer was added to the protein extracts, and the mixture
was incubated at 65 °C for 15 min. Extract volumes equivalent to 20 μg
Cells were cultured in 10-cm dishes and treated with ZnSO4, Z01, of protein were separated by using 10% SDS-PAGE gels, transferred
or ZnPy for 1 h before cell collection. The cell layer was washed twice to polyvinylidene difluouride membranes, and probed using anti-
with Ca 2+, Mg 2+-free phosphate-buffered saline (PBS) and solubi- phosphorylated ERK1/2 (Cell Signaling Technology, Inc., Beverly, MA,
lized in 0.5 mL SDS buffer solution (50 mM Tris, 2% SDS, 10% glycerol, USA) and ERK1/2 (BD Biosciences, Franklin Lakes, NJ, USA) antibodies
pH 6.8). The cell lysate was incubated for 10 min at 95 °C; the portion followed by an anti-mouse or rabbit IgG antibody conjugated with
of the lysate was degraded with nitric acid–H2O2 in an aluminum horseradish peroxidase. Complexes were detected with Western Light-
dry-block bath at 130 °C for 2 days and then diluted with 0.1 M nitric ning Chemiluminescence Reagent Plus (PerkinElmer Life Sciences,
acid and used for determination of the zinc concentration by means of Boston, MA, USA).
inductively coupled plasma mass spectrometry (ICP-MS, ELAN DRC II,
PerkinElmer, MA, USA). 2.11. Statistical analyses

65 Data were analyzed with Tukey's test using PASW Statistics 18


2.6. Zn incorporation assay
software (IBM, Armonk, NY, USA). Differences between groups were
considered significant if P b 0.05.
Radioactive 65Zn (270 MBq/nmol) was purchased from RIKEN (Wako,
Japan) as a 0.1 N HCl solution. For 65ZnSO4 treatment, the 65ZnCl2 solution
3. Results and discussion
was diluted with 100 vol of 100 mM ZnSO4 (5.4 MBq/mmol). For synthe-
sis of 65Zn-labeled Z01 (65Zn\Z01), 10 pmol of 65Zn and 0.5 mmol of
3.1. Coordination compounds alter MT-I and HO-1 mRNA concentrations
ZnCl2 were used as zinc sources. MEFs were cultured in 12-well culture
in MEFs
plates and treated with 100 μM 65Zn-labeled Z01 (about 50 cpm/nmol)
or 65ZnSO4 (about 50 cpm/nmol) for 1 h at 4 °C, 1 h at 37 °C, 3 h at
First, we measured MT-I and HO-1 mRNA concentrations to find
4 °C, and 3 h at 37 °C. After the indicated times, the cells were washed
MT-I mRNA-specific inducers. We treated MEFs for 3 h with the coor-
three times with ice-cold PBS. Cells were collected, and radioactivity
dination compounds (see Table 1 and Supplemental Table 1) at a con-
was measured by using a γ-counter (Auto Well Gamma System
centration of 100 μM; however, when compounds showed cytotoxicity
ARC-380CL; Aloka, Tokyo, Japan).
at 100 μM, we tested them at 20, 1, or 0.1 μM. We also examined ZnSO4,
the strong MT inducer CdCl2, and the Zn ionophore ZnPy to compare
2.7. Calculation of logP values
inducing capability. In our experimental condition, background zinc
concentration in culture medium was several μM. Weak MT-I mRNA
The logP values of Z01 and ZnPy were calculated by using the
induction by 100 μM ZnSO4 was observed without HO-I mRNA in-
ALOGPS 2.1 program (http://www.vcclab.org/lab/alogps/) [42].
duction. Strong MT-I mRNA induction by 200 μM ZnSO4, 10–20 μM
CdCl2, and 100 nM ZnPy was observed with strong HO-I mRNA in-
2.8. In vitro transcription/translation duction. Most of the compounds induced both MT-I and HO-1
mRNA, whereas a few induced neither MT-I nor HO-1 mRNA, because
Recombinant mouse MTF-1 proteins were synthesized in vitro of cytotoxicity (Fig. 1A). Z02 strongly induced MT-I mRNA but with
with the TnT T7 quick coupled transcription/translation system weak HO-1 mRNA induction; this effect was not specific for MT-I
(Promega) using 1.0 μg of plasmid (pcDNA3.1/hygromycin (+) plas- mRNA. In contrast, Z01 induced MT-I mRNA selectively at the con-
mid expressing flag-tagged mouse MTF-1) per reaction as previously centration of 100 μM. High MT-I mRNA induction by Z01 was ob-
described [43]. served after treatment at 100 μM and then plateaued (Fig. 1B). In
addition, Z01 at 200 μM and 500 μM showed induction of HO-1
2.9. Electrophoretic mobility shift assay mRNA and MT-I mRNA. Too much intracellular Z01 might cause
cell-stress. We also found some compounds that induced both MT
Binding reactions in the electrophoretic mobility shift assay was and HO-1. Z12–Z17, which are phenanthrene derivatives, and are
performed using binding buffer containing 12 mM 4-(2-hydroxyethyl)- MT plus HO-1 inducers. Almost all phenanthrene derivatives in our
1-piperazineethanesulfonic acid (HEPES) (pH 7.9), 5 mM MgCl2, library had high MT-inducing activity and high HO-1-inducing activ-
60 mM KCl, 0.5 mM dithiothreitol, 2 μg of poly (dI-dC) with 0.5 μL ity and/or cytotoxicity. Polycyclic aromatic hydrocarbon metabo-
of recombinant MTF-1 protein in a total volume of 20 μL. The MTF-1 lites, produced by endogenous aldo-keto reductases (AKR), are
protein was incubated with ZnSO4, or the indicated coordination known to generate reactive oxygen species (ROS) and enhance
compounds in the binding buffer for 15 min at 37 °C. Then 50 fmol Nrf2-mediated transcription [44]. Phenanthrene derivatives might
of 32P-labeled double-stranded MRE oligonucleotide (MREs-sense; be metabolized by AKR and generate ROS under our experimental
5′-GATCCAGGGAGCTCTGCACACGGCCCGAAAAGTA-3′) was added, and conditions.
the mixture was placed on ice for 30 min. After this incubation,
protein-DNA complexes were separated electrophoretically at 4 °C 3.2. The ligand of Z01 does not induce MT-I gene transcription
on 8% nondenaturing polyacrylamide gels. The gels were dried, and
the labeled complexes were detected by radioluminography using a To examine whether zinc is essential for induction of MT-I mRNA
Fuji BAS1500 phosphorimager (Fujifilm, Tokyo, Japan). by Z01, we treated MEFs for 3 h with the ligand of Z01. Z01 consists
T. Kimura et al. / Journal of Inorganic Biochemistry 117 (2012) 140–146 143

Fig. 1. Effect of zinc coordination compounds on MT-I and HO-1 mRNA levels. (A) MEFs were treated with the indicated concentrations of zinc coordination compounds for 3 h, and
then total RNA was isolated. MT-I, HO-1, and GAPDH mRNAs were reverse transcribed and quantified by real-time PCR. MT-I (closed bars) and HO-1 (open bars) mRNA levels were
normalized to GAPDH mRNA levels and are shown as relative values against the group treated with 20 μM CdCl2. Values are expressed as the mean± S.D. of 3–7 independent
experiments. (B) Dose response studies of MT-I and HO-1 mRNA levels treated with zinc coordination compounds for 3 h. Values are expressed as the mean ± S.D. of 3–5 indepen-
dent experiments. *, significantly different from the untreated control (P b 0.05). n.d., not determined. cytotoxic, cytotoxicity under phase-contrast microscopy was observed.

of one zinc and two L-cysteine molecules. A two-fold greater concen-


tration (200 μM) of L-cysteine relative to the concentration of Z01was
used to determine whether L-cysteine could act as an MT-I mRNA
inducer. After treatment with the ligands of Z01, Z02, and Z05, neither
MT-I nor HO-1 transcription was increased (Fig. 2). The ligand of
ZnPy, Py, markedly induced HO-1 mRNA expression with a slight
increase in MT-I mRNA expression. Zinc ions are present in the culture
medium, so Py might form ZnPy with the zinc in the culture medium.
The HO-1 mRNA level after Py treatment was higher than that after
ZnPy treatment.

3.3. Z01 increases MT-I promoter–driven luciferase reporter gene


expression and the gene expression is mediated the transcription MTF-1

We examined the effects of Z01 on MT-I promoter activation


(Fig. 3A). Treatment with 100 μM ZnSO4 increased MT-I promoter‐
driven luciferase reporter gene expression. Treatment with 100 μM
Z01 and 0.2 μM ZnPy also increased MT-I promoter‐driven luciferase
reporter gene expression. These result showed that Z01 induced
MT-I gene transcription.
Heavy metal-induced MT-I gene transcription is mediated by
MTF-1. To confirm that the Z01-induced MT-I transcription was also
mediated by MTF-1, we examined the effect of Z01 on MT-I gene tran-
scription in MEFs from MTF-1 knockout mice (MTF-1 KO MEFs). MT-I
induction was not observed in MTF-1 KO MEFs (Fig. 3B). In contrast, Fig. 2. Effect of ligands on MT-I and HO-1 mRNA levels. MEFs were treated with the
indicated concentrations of ligands for 3 h. After treatment, MT-I (closed bars) and
Z01 induced MT-I gene transcription was observed in other HO-1 (open bars) mRNA levels were measured as described for Fig. 1. Values are
MTF-1-expressing cells (BALB/3 T3 A31-1-1 cells and Hepa 1–6 expressed as the mean ± S.D. of 3–5 independent experiments. *, significantly different
cells; Fig. 3C). In human cells, MT-IIA gene transcription was well from the untreated control (P b 0.05).
144 T. Kimura et al. / Journal of Inorganic Biochemistry 117 (2012) 140–146

A) Luciferase assay B

Fig. 3. Effect of Z01 on MT-I promoter-driven gene transcription and MTF-1-dependent gene transcription. (A) MEFs were transfected with the Firefly luciferase reporter plasmid
pGL4.12-MT–264/+ 42 and the Renilla reporter plasmid pRL-SV40. Transfected cells were cultured for 24 h and then incubated with the indicated concentrations of Z01. Twelve
hours after treatment, reporter gene expression was measured as luciferase activity. Values are expressed as the mean ± S.D. of 3 independent experiments. *, significantly different
from the untreated control (P b 0.05). (B) MEFs and MTF-1 KO MEFs were treated with the indicated compounds for 3 h. After treatment, MT-I (closed bars) and HO-1 (open bars)
mRNA levels were measured as described for Fig. 1. Values are expressed as the mean ± S.D. of 3–6 independent experiments. *, significantly different from the untreated control
(P b 0.05). (C) BALB/3 T3, Hepa, HepG2, and HEK293 cells were treated with the indicated compounds for 3 h. After treatment, mouse MT-I mRNA (BALB/3 T3 and Hepa cells) and
human MT-IIA mRNA (HepG2 and HEK293 cells) levels were measured as described for Fig. 1. Values are expressed as the mean ± S.D. of 3–5 independent experiments.
*, significantly different from the untreated control (P b 0.05). cytotoxic, cytotoxicity under phase-contrast microscopy was observed.

studied, and the transcription also mediated by MTF-1. Z01-induced treatments (Fig. 4A). The intracellular total zinc concentration is
MT-IIA gene transcription was observed in HepG2 and HEK293 cells 100–500 μM [46]. Zinc ions, supplied as ZnSO4, are incorporated via
(Fig. 3C). zinc importers (ZIPs) [4]. Long-term (24 h) zinc treatment increases
By contrast, HO-1 induction caused by Z01 treatment was ob- the intracellular zinc concentration [47]; however, a 3-h treatment
served in MTF-1 KO MEFs, and the induction was much higher than with ZnSO4 may be too short to increase the intracellular total zinc
that in WT MEFs. The induction of HO-1 mRNA in MTF-1 KO MEFs concentration. The intracellular free zinc concentration is estimated
was also observed after CdCl2, ZnPy, and Z02 treatment. MTF-1 KO to be ~ 0.4 nM [46], which means that only a small portion of zinc
MEFs express extremely low levels of MT [45]; therefore, increasing ions exists as free zinc ions. An increase in the intracellular free zinc
the intracellular zinc concentration following treatment with zinc concentration could occur without an increase in the total intracellular
coordination compounds might be toxic to MT-deficient cells. zinc concentration. We measured the intracellular 65Zn concentration
after 1-h and 3-h 65ZnSO4 and 65Zn\Z01 treatments to evaluate the
3.4. Z01 does not increase intracellular zinc concentration rate of incorporation. 65Zn was incorporated 3 h after 65ZnSO4 treat-
ment at 37 °C (Fig. 4B gray bar). This increase was not observed at
One hundred micromolar Z01 increased MT-I mRNA to a level that 4 °C (Fig. 4B open bar). A portion of the incorporated zinc ions may
was much higher than that induced by 100 μM ZnSO4. To examine the bind to MTF-1 and increase MTF-1-dependent MT gene transcription. In
possibility that the high inducibility of Z01 depends on a high intra- contrast, no significant increase in intracellular 65Zn concentration was
cellular zinc concentration, we measured the total zinc concentration observed after 65Zn\Z01 treatment at both 4 °C and 37 °C (Fig. 4B
after ZnSO4 and Z01 treatment by using ICP-MS. However, we found hatched and closed bar). This difference suggests that 65Zn\Z01 is in-
that the total zinc concentration was the same following both corporated through ZIPs-independent routes. Kim et al. reported that
T. Kimura et al. / Journal of Inorganic Biochemistry 117 (2012) 140–146 145

A) Total zinc concentration B) 65Zn incorporation

Fig. 4. Zinc incorporation after ZnSO4 and Z01 treatment. (A) MEFs were treated with the indicated compounds for 3 h at 37 °C. Then, cell lysates were prepared after the cells were
washed with ice-cold PBS; the total zinc concentration of the lysates was measured by using ICP/MS. (B) MEFs were treated with 65Zn-labeled compounds for 1 h and 3 h at 4 °C
and 37 °C. Then the 65Zn concentration in MEFs was measured as radioactivity. Values are expressed as the mean ± S.D. *, significantly different from the 4 °C group (P b 0.05).

the zinc ionophore ZnPy induces an increase in the intracellular zinc The amount of zinc incorporated after 100 μM Z01 treatment might
concentration within a few minutes and that the maximum zinc con- be similar to that incorporated after 0.4 μM ZnPy treatment.
centration is achieved within 5 minutes [48]. Moreover, when logP
values were estimated by using the ALOGPS 2.1 program, we found 3.6. Possibility of Z01 on selective zinc transfer to MTF
that ZnPy gave a high value (0.58 ±2.32). In contrast, Z01 gave a low
value (−3.79 ± 2.32), suggesting that Z01 is too hydrophilic to be As mentioned previously, zinc ions interact with various sets of
absorbed via the cell surface membrane. The high inducibility of Z01 molecules and modify their functions [5,8–11,18]. Z01 might, there-
was not due to a higher intracellular zinc concentration as a result of a fore, modify the functions of not only MTF-1 but also other molecules.
higher rate of incorporation. Some researchers have studied the To assess whether Z01 selectively transfers zinc to MTF-1, we mea-
application of zinc-amino acid complexes for medical use [15] and ana- sured the phosphorylation of ERK after Z01 treatment. Zinc inhibits
lyzed zinc bioavailability after oral administration [17]. Several the phosphatase activity of PTP1B and increases the phosphorylation
zinc-amino acid complexes, including the zinc-L-cysteine complex
(Z01), showed similar bioavailability. These zinc-amino acid complexes
might be incorporated into cells via the L-type amino acid transporter
LAT [49] or the organic anion transporter OAT [50]. Further investiga-
tion is needed to clarify the role of Z01 in incorporation.

3.5. Z01 facilitates MTF-1–MRE complex formation in vitro

To address the question why Z01 showed high MT-I inducibility


without a higher intracellular zinc concentration, we compared the
abilities of the coordination compounds to promote MTF-1–MRE
complex formation. MTF-1 has 6 Cys2His2-type zinc fingers; the coor-
dination type of these zinc fingers is Zn(S2N2). Zn(S2N2) coordination
compounds could, therefore, transfer zinc to the zinc fingers of
MTF-1. To examine this possibility, we compared the abilities of
ZnSO4, Z01, and ZnPy to promote the formation of MTF-1–MRE com-
plexes. Two micromolar Z01 was needed for the formation of the
MTF-1–MRE complex (Fig. 5). This concentration was similar to the
concentration of ZnPy needed for MTF-1–MRE complex formation.
However, for ZnSO4, 10 μM was needed for the formation of the
MTF-1–MRE complex. The concentration of Z01 was thus lower
than that of ZnSO4. ZnPy has Zn(S2O2) coordination, not Zn(S2N2)
coordination. Zinc coordination compounds, regardless of coordina-
tion type, might transfer zinc ions to MTF-1 more efficiently than Fig. 5. Electrophoretic mobility shift assays of ZnSO4−, Z01-, and ZnPy-dependent
ZnSO4. In the ZnPy treatment experiment, zinc ions may have been MRE-binding activity of MTF-1. MTF-1 was synthesized in vitro using a transcription/
translation system. Synthesized MTF-1 was adjusted to the indicated concentrations
incorporated as the coordination compound, ZnPy, at low concentra-
of ZnSO4, Z01, and ZnPy, and MRE-binding activity was measured as detailed in the
tions (0.1–0.4 μM), and the concentration of incorporated ZnPy and/or Materials and methods section. The arrow demarcates the specific MTF-1–MRE
zinc ions was sufficient to activate MTF-1–MRE binding (Fig. 1A, B). complex, and the line indicates the free MRE.
146 T. Kimura et al. / Journal of Inorganic Biochemistry 117 (2012) 140–146

[8] C. Kitabayashi, T. Fukada, M. Kanamoto, W. Ohashi, S. Hojyo, T. Atsumi, N. Ueda, I.


Azuma, H. Hirota, M. Murakami, T. Hirano, Int. Immunol. 22 (2010) 375–386.
[9] H. Haase, W. Maret, Exp. Cell Res. 291 (2003) 289–298.
[10] C. Klein, R.K. Sunahara, T.Y. Hudson, T. Heyduk, A.C. Howlett, J. Biol. Chem. 277
(2002) 11859–11865.
[11] X. Gao, Z. Du, T.B. Patel, J. Biol. Chem. 280 (2005) 2579–2586.
[12] S.K. Das, K. Ray, Nat. Clin. Pract. Neurol. 2 (2006) 482–493.
[13] D. Magda, P. Lecane, Z. Wang, W. Hu, P. Thiemann, X. Ma, P.K. Dranchak, X. Wang,
V. Lynch, W. Wei, V. Csokai, J.G. Hacia, J.L. Sessler, Cancer Res. 68 (2008)
5318–5325.
[14] W.Q. Ding, S.E. Lind, IUBMB Life 61 (2009) 1013–1018.
[15] Y. Yoshikawa, Y. Adachi, H. Sakurai, Life Sci. 80 (2007) 759–766.
[16] E.Z. Dajani, M.J. Klamut, Expert Opin Investig Drugs 9 (2000) 1537–1544.
[17] T. Matsukura, H. Tanaka, Biochemistry (Mosc) 65 (2000) 817–823.
[18] T. Kimura, N. Itoh, G.K. Andrews, J. Health Sci. 55 (2009) 484–494.
Fig. 6. Time course of ZnSO4- and Z01-induced ERK1/2 phosphorylation in MEFs. MEFs [19] J.W. Kaspar, S.K. Niture, A.K. Jaiswal, Free Radic. Biol. Med. 47 (2009) 1304–1309.
were treated with ZnSO4 and Z01 for 5 min and 10 min. Three independent MEF [20] M. Kobayashi, L. Li, N. Iwamoto, Y. Nakajima-Takagi, H. Kaneko, Y. Nakayama, M.
lysates were subjected to SDS-PAGE and Western blotting with anti-phospho-ERK1/2 Eguchi, Y. Wada, Y. Kumagai, M. Yamamoto, Mol. Cell. Biol. 29 (2009) 493–502.
and ERK1/2 antibodies. Values shown under the top panel represent the intensity of [21] S. Viladkar, M. Kamaluddin, Nath, Bull. Chem. Soc. Jpn. 66 (1993) 2922–2926.
[22] Y. Yoshikawa, E. Ueda, K. Kawabe, H. Miyake, H. Sakurai, Y. Kojima, Chem. Lett. 29
the phosphor-ERK1/2 band normalized to that of the ERK band for each sample;
(2000) 874–875.
these values are expressed as the mean ± S.D. for three independent experiments.
[23] A. Katoh, T. Tsukahara, R. Saito, K. Ghosh, Y. Yoshikawa, Y. Kojima, A. Tamura, H.
*, significantly different from the untreated control (P b 0.05).
Sakurai, Chem. Lett. 31 (2002) 114–115.
[24] G. Anderegg, E. Hubmann, N.G. Podder, F. Wenk, Helv. Chim. Acta (1977)
123–140.
of ERK [9]. We did not observe a significant increase in pERK after [25] J. Wirbser, H. Vahrenkamp, Z. Naturforsch 47b (1992) 962–968.
[26] E.C. Constable, J. Lewis, M.C. Liptrot, P.R. Raithby, Inorg. Chim. Acta 178 (1990)
treatment of cells with 100 μM Z01 (Fig. 6). This result shows that 47–54.
the zinc of Z01 was selectively utilized for MTF-1 rather than for the [27] M. Senoue, T. Iwaki, K. Seki, M. Yagi, J. Photochem. Photobiol. A: Chem. 101
ERK cascade. We measured only one zinc-regulated signaling path- (1996) 257–263.
[28] K.S. Anjali, J.T. Sampanthar, J. Vittal, Inorg. Chim. Acta 295 (1999) 9–17.
way of the ERK cascade. To clarify the selectivity of Z01, further inves- [29] G.A. Crosby, R.G. Highland, K.A. Truesdell, Coord. Chem. Rev. 64 (1985) 41–52.
tigation is needed. However, if Z01 can modulate other zinc-regulated [30] A.W. Kleij, M. Kuil, D.M. Tooke, M. Lutz, A.L. Spek, J.N.H. Reek, Chem. Eur. J. 11
signaling pathways, we can modify its structure to remove such an (2005) 4743–4750.
[31] Y. Mikata, M. Wakamatsu, S. Yano, Dalton Trans. (2005) 545–550.
effect.
[32] M. Dakoviać, Z. Popović, G. Giester, M. Rajić-Linarić, Polyhedron 27 (2008)
An MTF-1-specific activator might be of use for cancer prevention, 210–222.
because mouse strains that show higher metal- and γ-ray radiation- [33] K. Krishnan, R.A. Plane, Inorg. Chem. 5 (1966) 852–857.
responsive transcription of MTF-1, caused by single nucleotide poly- [34] C. Postmus, J.R. Ferraro, W. Wozniak, Inorg. Chem. 6 (1967) 2030–2032.
[35] M.W. Venteer, A.M. Heyns, Inorg. Chem. 26 (1987) 1095–1098.
morphisms, are resistant to γ-ray-induced lymphoma development [36] M. Kato, T. Ito, Inorg. Chem. 24 (1985) 504–508.
[51]. MT is a key factor for the detoxification of heavy metals by [37] S.B. Sanni, H.J. Behm, P.T. Beurskens, G.A.V. Albada, J. Reedijk, A.T.H. Lenstra, A.W.
chelation [52]. Moreover, overexpression of MT protects the myocar- Addison, M. Planiandavar, J. Chem. Soc. Dalton Trans. (1988) 1429–1435.
[38] M. Schulz, M. Klopfleisch, H. Görls, M. Kahnes, M. Westerhausen, Inorg. Chim.
dium from ischemia/reperfusion injury [53]. Metallothionein is also Acta 362 (2009) 4706–4712.
an anti-inflammatory mediator [54]. Furthermore, our library of zinc [39] A.J.M. Al-Karawi, W. Clegg, R.W. Harrington, R.A. Henderson, Dalton Trans. (2009)
coordination compounds had only 30 coordination compounds, but 564–570.
[40] M.R. Bermejo, A.M. González-Noya, R.M. Pedrido, M.J. Romero, M. Vázquez,
it had structural variety. By using other screening methods, we Angew. Chem. Int. Ed. 44 (2005) 4182–4187.
might find that one of these zinc coordination compounds regulates [41] T. Kimura, Y. Li, F. Okumura, N. Itoh, T. Nakanishi, T. Sone, M. Isobe, G.K. Andrews,
zinc distribution and acts as a zinc donor for specific molecules. We Biochem. J. 415 (2008) 477–482.
[42] I.V. Tetko, J. Gasteiger, R. Todeschini, A. Mauri, D. Livingstone, P. Ertl, V.A. Palyulin,
believe that our results show the potential of coordination com- E.V. Radchenko, N.S. Zefirov, A.S. Makarenko, V.Y. Tanchuk, V.V. Prokopenko,
pounds as protein-specific activators/inhibitors. J. Comput. Aided Mol. Des. 19 (2005) 453–463.
Supplementary data to this article can be found online at http:// [43] T. Kimura, N. Itoh, T. Sone, K. Tanaka, M. Isobe, J. Cell. Biochem. 93 (2004)
609–618.
dx.doi.org/10.1016/j.jinorgbio.2012.07.021.
[44] J.H. Park, A.B. Troxel, R.G. Harvey, T.M. Penning, Chem. Res. Toxicol. 19 (2006)
719–728.
Acknowledgements [45] R. Heuchel, F. Radtke, O. Georgiev, G. Stark, M. Aguet, W. Schaffner, EMBO J. 13
(1994) 2870–2875.
[46] J.L. Vinkenborg, T.J. Nicolson, E.A. Bellomo, M.S. Koay, G.A. Rutter, M. Merkx, Nat.
We are indebted to Dr. Kazuyuki Sato (Laboratory of Pharmaceuti- Methods 6 (2009) 737–740.
cal Chemistry, Faculty of Pharmaceutical Sciences, Setsunan Universi- [47] P. Coyle, J.C. Philcox, A.M. Rofe, Biochem. J. 309 (Pt 1) (1995) 25–31.
ty) for excellent technical assistance. [48] C.H. Kim, J.H. Kim, S.J. Moon, K.C. Chung, C.Y. Hsu, J.T. Seo, Y.S. Ahn, Biochem.
Biophys. Res. Commun. 259 (1999) 505–509.
[49] M. Palacin, V. Nunes, M. Font-Llitjos, M. Jimenez-Vidal, J. Fort, E. Gasol, M. Pineda,
References L. Feliubadalo, J. Chillaron, A. Zorzano, Physiology (Bethesda) 20 (2005) 112–124.
[50] F. Zhou, G. You, Pharm. Res. 24 (2007) 28–36.
[1] A.S. Prasad, Nutrition 11 (1995) 93–99. [51] Y. Tamura, M. Maruyama, Y. Mishima, H. Fujisawa, M. Obata, Y. Kodama, Y.
[2] B.L. Vallee, D.S. Auld, EXS 73 (1995) 259–277. Yoshikai, Y. Aoyagi, O. Niwa, W. Schaffner, R. Kominami, Oncogene 24 (2005)
[3] M. Murakami, T. Hirano, Cancer Sci 99 (2008) 1515–1522. 399–406.
[4] T. Kambe, Y. Yamaguchi-Iwai, R. Sasaki, M. Nagao, Cell Mol. Life Sci. 61 (2004) [52] C.D. Klaassen, J. Liu, J. Toxicol. Sci. 23 (Suppl. 2) (1998) 97–102.
49–68. [53] Y. Oshima, Y. Fujio, T. Nakanishi, N. Itoh, Y. Yamamoto, S. Negoro, K. Tanaka, T.
[5] C. Andreini, L. Banci, I. Bertini, A. Rosato, J. Proteome Res. 5 (2006) 196–201. Kishimoto, I. Kawase, J. Azuma, Cardiovasc. Res. 65 (2005) 428–435.
[6] F. Radtke, O. Georgiev, H.P. Muller, E. Brugnera, W. Schaffner, Nucleic Acids Res. [54] K. Inoue, H. Takano, A. Shimada, M. Satoh, Mediat. Inflamm. 2009 (2009) 101659.
23 (1995) 2277–2286.
[7] B.M. Potter, L.S. Feng, P. Parasuram, V.A. Matskevich, J.A. Wilson, G.K. Andrews,
J.H. Laity, J. Biol. Chem. 280 (2005) 28529–28540.

Você também pode gostar