induction of dna double-strand breaks in the h4iie cell line exposed to environmentally relevant...

10
This article was downloaded by: [UZH Hauptbibliothek / Zentralbibliothek Zürich] On: 26 August 2013, At: 11:43 Publisher: Taylor & Francis Informa Ltd Registered in England and Wales Registered Number: 1072954 Registered office: Mortimer House, 37-41 Mortimer Street, London W1T 3JH, UK Journal of Toxicology and Environmental Health, Part A: Current Issues Publication details, including instructions for authors and subscription information: http://www.tandfonline.com/loi/uteh20 Induction of DNA Double-Strand Breaks in the H4IIE Cell Line Exposed to Environmentally Relevant Concentrations of Copper, Cadmium, and Zinc, Singly and in Combinations Renate Haldsrud a & Åse Krkje a a Department of Biology, Norwegian University of Science and Technology (NTNU), Trondheim, Norway Published online: 30 Jan 2009. To cite this article: Renate Haldsrud & se Krkje (2009) Induction of DNA Double-Strand Breaks in the H4IIE Cell Line Exposed to Environmentally Relevant Concentrations of Copper, Cadmium, and Zinc, Singly and in Combinations, Journal of Toxicology and Environmental Health, Part A: Current Issues, 72:3-4, 155-163, DOI: 10.1080/15287390802538964 To link to this article: http://dx.doi.org/10.1080/15287390802538964 PLEASE SCROLL DOWN FOR ARTICLE Taylor & Francis makes every effort to ensure the accuracy of all the information (the “Content”) contained in the publications on our platform. However, Taylor & Francis, our agents, and our licensors make no representations or warranties whatsoever as to the accuracy, completeness, or suitability for any purpose of the Content. Any opinions and views expressed in this publication are the opinions and views of the authors, and are not the views of or endorsed by Taylor & Francis. The accuracy of the Content should not be relied upon and should be independently verified with primary sources of information. Taylor and Francis shall not be liable for any losses, actions, claims, proceedings, demands, costs, expenses, damages, and other liabilities whatsoever or howsoever caused arising directly or indirectly in connection with, in relation to or arising out of the use of the Content. This article may be used for research, teaching, and private study purposes. Any substantial or systematic reproduction, redistribution, reselling, loan, sub-licensing, systematic supply, or distribution in any form to anyone is expressly forbidden. Terms & Conditions of access and use can be found at http:// www.tandfonline.com/page/terms-and-conditions

Upload: ase

Post on 12-Dec-2016

212 views

Category:

Documents


0 download

TRANSCRIPT

This article was downloaded by: [UZH Hauptbibliothek / Zentralbibliothek Zürich]On: 26 August 2013, At: 11:43Publisher: Taylor & FrancisInforma Ltd Registered in England and Wales Registered Number: 1072954 Registered office: Mortimer House,37-41 Mortimer Street, London W1T 3JH, UK

Journal of Toxicology and Environmental Health, PartA: Current IssuesPublication details, including instructions for authors and subscription information:http://www.tandfonline.com/loi/uteh20

Induction of DNA Double-Strand Breaks in the H4IIECell Line Exposed to Environmentally RelevantConcentrations of Copper, Cadmium, and Zinc, Singlyand in CombinationsRenate Haldsrud a & Åse Kr⊘kje a

a Department of Biology, Norwegian University of Science and Technology (NTNU),Trondheim, NorwayPublished online: 30 Jan 2009.

To cite this article: Renate Haldsrud & se Krkje (2009) Induction of DNA Double-Strand Breaks in the H4IIE Cell Line Exposedto Environmentally Relevant Concentrations of Copper, Cadmium, and Zinc, Singly and in Combinations, Journal of Toxicologyand Environmental Health, Part A: Current Issues, 72:3-4, 155-163, DOI: 10.1080/15287390802538964

To link to this article: http://dx.doi.org/10.1080/15287390802538964

PLEASE SCROLL DOWN FOR ARTICLE

Taylor & Francis makes every effort to ensure the accuracy of all the information (the “Content”) containedin the publications on our platform. However, Taylor & Francis, our agents, and our licensors make norepresentations or warranties whatsoever as to the accuracy, completeness, or suitability for any purpose of theContent. Any opinions and views expressed in this publication are the opinions and views of the authors, andare not the views of or endorsed by Taylor & Francis. The accuracy of the Content should not be relied upon andshould be independently verified with primary sources of information. Taylor and Francis shall not be liable forany losses, actions, claims, proceedings, demands, costs, expenses, damages, and other liabilities whatsoeveror howsoever caused arising directly or indirectly in connection with, in relation to or arising out of the use ofthe Content.

This article may be used for research, teaching, and private study purposes. Any substantial or systematicreproduction, redistribution, reselling, loan, sub-licensing, systematic supply, or distribution in anyform to anyone is expressly forbidden. Terms & Conditions of access and use can be found at http://www.tandfonline.com/page/terms-and-conditions

Journal of Toxicology and Environmental Health, Part A, 72: 155–163, 2009Copyright © Taylor & Francis Group, LLCISSN: 1528-7394 print / 1087-2620 online DOI: 10.1080/15287390802538964

155

UTEHInduction of DNA Double-Strand Breaks in the H4IIE Cell Line Exposed to Environmentally Relevant Concentrations of Copper, Cadmium, and Zinc, Singly and in Combinations

DNA DOUBLE-STRAND BREAKS IN THE H4IIE CELL LINERenate Haldsrud and Åse KrøkjeDepartment of Biology, Norwegian University of Science and Technology (NTNU), Trondheim, Norway

Xenobiotics, including heavy metals, exist in nature as complexmixtures of compounds with possible interactions. Induction ofDNA damage such as DNA strand breaks may exert detrimentalconsequences to both individuals and populations. In this study,the induction of DNA double-strand breaks was assessed using theH4IIE rat hepatoma cell line following exposure to high and envi-ronmentally relevant concentrations of chloride salts of the metalscadmium (Cd), copper (Cu), and zinc (Zn), both singly and incombination. DNA strand break analysis was performed usingagarose gel electrophoresis. Median molecular lengths were calcu-lated from fragment size distributions acquired from gel imagedata and were used as a quantitative measure of DNA double-strand break induction. Exposure to high concentrations of Cuand Cd in combination produced a significant increase in theoccurrence of DNA strand break. However, exposing cells to highconcentrations of Cu, Cd, and Zn in combination resulted insignificantly lower DNA double-strand break compared to controlcells. Addition of low Zn to the Cd/Cu mixture restored DNAdamage level back to that of the control. Environmentally rele-vant concentrations of Cd, Cu, and Zn did not appear to induceDNA strand breaks in the H4IIE cell line.

Heavy metals can exist either as pure metal mixtures, or inmixture with other types of contaminants. Metals occur naturallyin all ecosystems, but in variable concentrations. They are redis-tributed in nature through geochemical cycles, but anthropogenicactivity interferes with these natural cycles, leading to excessiveaccumulation of metals in certain compartments. This studyfocused on the metals cadmium (Cd), copper (Cu), and zinc(Zn). When metals coexist in a compartment, the speciationproperties and bioavailability of the individual metals are

affected. Chemicals also compete for uptake routes in organisms,as well as toxic target sites, transport mechanisms, and excretionroutes within organisms (Eaton & Gilbert, 2008). Copper and Znare, as opposed to Cd, essential metals (Uauy et al., 1998; Valkoet al., 2005). Copper is involved in the normal function of differentenzymes such as cytochrome c-oxidase, Cu/Zn superoxide dis-mutase, and tyrosinase, as well as in metabolic complexes (Sternet al., 2007). Zinc serves as a cofactor for a number of metal-loenzymes and is also an essential component in many proteinsinvolved in gene expression and regulation of genetic activities(Berg & Shi, 1996; Watanabe et al., 1997). Many proteinsinvolved in DNA repair mechanisms and cell signalling path-ways are Zn finger proteins (Ho, 2004). Zinc deficiency in cellsmay therefore pose a larger threat to DNA integrity than excessZn. However, essential metals have the potential to becometoxic when present in excessive amounts.

DNA double-strand break formation is considered to be aserious form of DNA damage, due to destruction of templatesessential for DNA replication and transcription. Induction ofDNA double-strand breaks may lead to induction of apoptosis,mutations, chromosomal rearrangements and development ofcancer (Pfeiffer, 1998). DNA double-strand breaks may beinduced directly by ionizing radiation or radiomimetic com-pounds, or are formed during replication when the replicationfork encounters an unrepaired single-strand break (Helledayet al., 2007). Heavy metals produce DNA damage in differentways. Being a Fenton metal, Cu has the ability to participate inredox cycling, ultimately leading to formation of reactiveoxygen species (ROS) such as hydroxyl radicals (•OH) (Valkoet al., 2005). Cu also has the ability to bind directly to DNA,although the presence of hydrogen peroxide (H2O2) apparentlyis necessary for a genotoxic effect to occur (Sagripanti et al.,1991; Moriwaki et al., 2008). Cadmium does not form ROSdirectly. However, Cd was shown to be involved in indirectformation of ROS by decreasing the levels of free radical scav-engers such as glutathione, or by displacing Fenton metalsfrom proteins (Szuster-Ciesielska et al., 2000; Galán et al.,2001; Waisberg et al., 2003). Cadmium also has the ability toinhibit antioxidant enzymes such as catalase, glutathione

Financial support was obtained by the Norwegian Ministry ofForeign Affairs, organized by the Centre for International UniversityCooperation (SIU) via the project “ENLINO master programnetwork” (project CCP 03/02). The authors are grateful to ChrisBingham for his valuable assistance.

Address correspondence to Åse Krøkje, Department of Biology,Norwegian University of Science and Technology (NTNU) 7491,Trondheim, Norway. E-mail: [email protected]

Dow

nloa

ded

by [

UZ

H H

aupt

bibl

ioth

ek /

Zen

tral

bibl

ioth

ek Z

üric

h] a

t 11:

43 2

6 A

ugus

t 201

3

156 R. HALDSRUD AND Å. KRØKJE

peroxidase (GPox), and superoxide dismutase (SOD) involvedin removal of ROS (Waisberg et al., 2003; Filipi6 & Hei, 2004;Valko et al., 2005). Mechanisms underlying Cd-induced ROSformation involve disturbance of mitochondrial electron trans-fer chains and damage to lysosomal membranes (Bradley et al.,1987; Wang et al., 2004; Fotakis et al., 2005). Cadmium hasthe ability to interfere with DNA repair mechanisms such asnucleotide excision repair, possibly by competing with Zn inZn-finger proteins (Hartwig 1998, Asmuss et al., 2000). Jinet al. (2003) found that Cd interfered with mismatch repairmechanisms, and Fatur et al. (2003) showed that the metalinhibited steps in the base excision repair pathway. Base exci-sion repair is one of the main pathways for removal of oxida-tive DNA lesions such as 8-hydroxyguanine (Evans et al.,2004). Interference with cell cycle regulation by exposure toCd was also reported by Mukherjee et al. (2004). Oliveira et al.(2008) found that the metal was capable of binding directly toDNA. However, no oxidative DNA damage associated with Cdbinding was noted in their experiment. Zinc is, as opposed toCd and Cu, associated with antioxidant effects. Bray andBettger (1990) showed that Zn protected sulfhydryl groups andcompeted with transition metals for binding to target sites fortoxicity. However, the main antioxidant property associatedwith Zn is its ability to induce synthesis of metal bindingproteins such as metallothionein (MT) (Chan & Cherian,1992). Metallothionein may protect against metal toxicity by(1) limiting uptake of metals into the cells, (2) sequesteringmetals within the cells, or (3) increasing export of metals out ofcells (Park et al., 2001). Zinc and Cd are known to be potentinducers of metallothionein, but Cu also induces the protein(Klaassen et al., 1999). Asmuss et al. (2000) found that coex-posure to Zn and Cd produced less inhibition of nucleotideexcission repair than exposure to Cd alone.

Genotoxic compounds represent a major environmentalchallenge because they may lead to impaired reproduction,accelerated aging processes, and tumor induction in exposedindividuals. Genetic damage is also transmitted to offspring(Würgler & Kraemers, 1992). Development of methods that

predict genotoxic impact of exposure to environmental con-taminants before serious effects on the ecosystem level occuris, therefore, required. Several methods such as the alkalineunwinding assay, comet assay, and agarose gel electrophoresiswere previously applied for detection of DNA strand breaks incells and tissues (Shugart, 2000).

One possible strategy of biomarker development involvesmeasurements of adverse and genotoxic effects of pollutantsin isolated cell cultures or cell lines. Hepatoma cells are rele-vant study objects, due to the important role the liver playsin detoxification, bioactivation, and excretion of xenobioticsas well as endogenous compounds. Metal concentrationsmeasured in liver of different organisms are given in Table 1.The H4IIE rat hepatoma cell line has primarily been used foranalysis of CYP1A1 induction, owing to its low basal levelof this enzyme (Whyte et al., 2004). However, the cell linewas chosen for the present study because of its previous use inexperiment involving genotoxic endpoints such as DNA strandbreaks (Grant et al., 2001, Wätjen et al., 2005) and cell deathmechanisms (Kim et al., 2003). The aim of the present studywas to evaluate induction of DNA double-strand breaks as abiomarker of exposure to heavy metals in the H4IIE cell line,following exposure to both high and lower, environmentallyrelevant, concentrations of Cd, Cu and Zn. These three metalsfrequently occur together in nature, and obtaining knowledgeabout how the metals behave in mixtures is of ecotoxicologi-cal relevance. DNA double-strand break induction wasanalyzed by using a technique involving cell lysis andrelease of DNA within agarose gel plugs prior to agarose gelelectrophoresis.

MATERIALS/METHODS

ChemicalsFetal bovine serum (FBS) (heat inactivated) (10108-157),

L-glutamine medium supplement (200 mM) (25030-032),penicillin–streptomycin (5000 U/ml) (15070-063), and trypsin

TABLE 1 Levels of Cd, Cu, and Zn Measured in Liver Samples From Organisms at Different Sampling Sites

Species Location Cd (μg/g) Cu (μg/g) Zn (μg/g) Reference

Yellow perch (Perca flavescens) Quebec, Canada 30.8a 101.68a 167.37a Kraemer et al., 2005Brown trout (Salmo trutta) Røros, Norway 5.48b 103.42b 47.99b Olsvik et al., 2001Ringed seal (Pusa hispida) Holman, Canada 6.65b 9.25b 43.99b Dehn et al., 2005Loggerhead turtle (Caretta caretta) Mediterranean Sea, Italy 3.36b 7.69b 29.3b Storelli et al., 2005Caribou (Rangifer tarandus groenlandicus) Akia, Greenland 0.695b 21.8b 29.5b Aastrup et al., 2000Moose (Alces alces) Yukon, Canada 4.94b 40.32b 34.87b Gamberg et al., 2005Glaucous gull (Larus hyperboreus) Bear Island, Norway 6.06a 19.7a 111a Savinov et al., 2003Black-footed albatross (Diomedea nigripes) North Pacific, Japan 22b 5.1b 69b Ikemoto et al., 2004

Note. Superscripts denote: a, dry weight; b, wet weight.

Dow

nloa

ded

by [

UZ

H H

aupt

bibl

ioth

ek /

Zen

tral

bibl

ioth

ek Z

üric

h] a

t 11:

43 2

6 A

ugus

t 201

3

DNA DOUBLE-STRAND BREAKS IN THE H4IIE CELL LINE 157

(2.5%) (15090-046) were obtained from Gibco. Culturemedium RPMI 1640 was obtained from EuroClone (ECB9006L) and Sigma (R0883). Dimethyl sulfoxide (DMSO)(99.9%, spectrophotometric grade) (K21387950) was pur-chased from Merck. Ethidium bromide (EB) (10 mg/ml)(161-0433), high-strength analytical-grade agarose (ultrapureDNA grade) (162-0126), low-melt preparative grade agarose(ultra pure DNA grade) (162-0019), and sodium dodecyl sulfate(SDS) (electrophoresis grade) (161-0301) were obtained fromBio-Rad. λ DNA/HindIII digest (500 μg/ml) (#SM0101) andloading dye solution (6×) (R0611) were purchased fromFermentas. Thiazolyl blue tetrazolium bromide (MTT) (98%TLC) (M2128-1G) and proteinase K (39 U/mg) (P-2308) wereobtained from Sigma. Cadmium chloride dihydrate(CdCl2·2H2O) was purchased from Fluka Chemika (20906).Copper chloride dihydrate (CuCl2·2H2O) was obtained fromSigma (C-6641). Zinc chloride (ZnCl2) was purchased fromMerck (8816). All other chemicals were of analytical grade,and were obtained from Sigma, Merck, and Fluka Chemica.

Cell CultivationThe H4IIE rat hepatoma cell line was developed from the

Reuber Hepatoma H-35 cell line (Whyte et al., 2004). TheH4IIE cell line used in this experiment was supplied by theCentre for Applied Microbiology and Research (CAMR,ECACC, Salisbury, Wiltshire). The cells were grown in RPMI1640 medium, supplemented with fetal bovine serum (FBS)(5%), L-glutamine (200 mM) (1%), and penicillin–streptomycin(5000 U/ml) (1%) in a humid atmosphere of 5% CO2 in air at37°C. The cells were grown in 75-cm2 culture flasks(Sarstedt), and subcultured every 3–4 d after harvesting withtrypsin (0.0025%) and ethylenediamine tetraacetic acid(EDTA) (0.01%) in phosphate-buffered saline solution (PBS)and diluted 3–4 times. Cells were used at 75% monolayerconfluency.

MTT Cell Viability AssayThe 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium

bromide (MTT) assay (Mosmann, 1983) was performedaccording to Kim et al. (2003). The cells were plated in a 96-well plate (Costar) with 200 μl medium at a density of 105 cellsper well and incubated at 37°C in a humid atmosphere of 5%CO2 in air for 24 h prior to metal exposure. The stock solutionsof the metals (Cu, Cd, and Zn) were prepared in autoclaveddistilled water. The metal solutions were diluted in growthmedium to the desired concentrations, and 200 μl of the testexposure medium was applied to each well. When exposing thecells to individual metals, 6 CdCl2 concentrations in the range10-4–10 μM, 7 CuCl2 concentrations in the range 0.01–1000 μM,and 9 ZnCl2 concentrations in the range 0.01–500 μM wereused. When exposing the cells to combinations of metals, onehigh concentration and one low concentration were chosen foreach metal. These concentrations of metals are listed in Table 2.

For each exposure condition, a minimum of two parallels wereused, most conditions were tested with more than five paral-lels. At first, differences in cell viability were tested after 12,24, 36, or 48 h (data not shown). After these preliminary exper-iments, the cells were incubated with test solution for 36 hbefore determination of cell viability. After incubation, theexposure medium was removed, and the cells were incubatedfor another 4 h in medium containing MTT (0.5 mg/ml). Themedium was then removed, and to each well 200 μl dimethylsulfoxide (DMSO) was added to solubilize the produced for-mazan crystals. Relative concentration of formazan wasmeasured spectrophotometrically at 550 nm. Cell viability wasgiven as relative survival of exposed cells compared tountreated control cells, and was presented as percent of control.

Metal Exposure for DNA Double-Strand Break Assessment

The cells were seeded in 12-well plates (Costar), and incu-bated at 37ºC and in a humid atmosphere of 5% CO2 in air for24 h prior to metal exposure. After incubation, the growthmedium was replaced by 3 ml of fresh medium containing themetal for exposure. The metal concentrations used arepresented in Table 2. The cells were exposed to metals for 36 hat 37ºC and in a humid atmosphere of 5% CO2 in air. Afterexposure, the exposure medium was removed, and the cellswere washed with PBS (pH 7.4). The cells were harvested withtrypsin (0.0025%) and EDTA (0.01%) in PBS. The cell pelletobtained after centrifugation (5 min, 1200 × g) of the cellsuspension was dissolved in 500 μl TE-buffer (10 mM Tris, 1 mMEDTA, pH 8) prior to agarose plug preparation.

Agarose Plug PreparationThe agarose plugs for electrophoresis were prepared according

to the procedure described by Theodorakis et al. (1994) andKrøkje et al. (2006) with modifications. The cells obtainedafter harvesting cells were suspended in TE buffer, incubatedat 37°C, and mixed with an equal volume of premelted solutionof 1% low-melting-point-agarose at 37ºC. From this solution,agarose plugs were made by loading 20 μl of the low-melting-point agarose cell suspension into wells of a 1.5% standard

TABLE 2 High and Low Concentrations of the Metals Cadmium,

Copper, and Zinc Used for MTT Assay and DNA Strand Break Analyses in the H4IIE Cell Line After Exposure of the Metals

Singly and in Combination

Metal High concentration (μM) Low concentration (μM)

Cu 100 0.01Cd 1 0.0001Zn 10 0.01

Dow

nloa

ded

by [

UZ

H H

aupt

bibl

ioth

ek /

Zen

tral

bibl

ioth

ek Z

üric

h] a

t 11:

43 2

6 A

ugus

t 201

3

158 R. HALDSRUD AND Å. KRØKJE

agarose gel, and chilling the gel mold at 4°C for 1 h. The plugswere then cut from the mould, placed in 500 μl digestion buffer(10 mM Tris, 25 mM EDTA, 100 mM NaCl, 0.5% SDS, pH 8)with proteinase K (1 mg/ml), and incubated at 55°C overnight.After incubation, the plugs were chilled, loaded into the wellsof an agarose gel (0.6% agarose in TBE buffer (90 mM Trisbase, 90 mM boric acid, 2 mM EDTA, pH 8), and sealed with1% low-melting-point agarose prior to electrophoresis.

DNA Double-Strand Break AssayDNA from the exposed cells embedded in low-melting-point

agarose plugs was electrophoretically separated. Lambda-DNAHindIII digest was used as a molecular size marker. The sameTBE buffer used for gel preparation was used as running bufferfor electrophoresis. The electrophoresis was run at 1.5 V/cmfor 14 h, followed by staining of the gel with EB for 2 h.Gel image data was acquired using the BioRad Gel Doc 2000system for quantification of DNA double-strand breaks.Calculation of median molecular length (MML) of DNAfragments in the gel was performed, using densitometric dataobtained from gel image analysis. The DNA fragment sizedistribution determined from gel image data was comparedto a DNA size marker (Hind III digested λ-phage DNA) runon the same gel. Calculated MML values were used as thequantitative measure of the level of damage due to DNAdouble-strand breaks.

Statistical AnalysisStatistical differences between control cells and cells

exposed to different mixtures and concentrations of metalswere calculated using the nonparametric Mann–Witney test.The statistical tests were performed using MINITAB Release14. The criterion for significance was set at p < .05.

RESULTS

MTT Cell Viability AssayFor all metal concentrations analyzed, the cell viability was

expressed as percent formation of formazan crystals in exposedcells compared to controls. The preliminary experimentsperformed to assess differences in cell viability as a function ofexposure time indicated that no differences in survival as afunction of exposure time existed (data not shown). In furtherexperiments 36-h exposures were chosen.

The results from exposure of the cells to single metals inincreasing concentrations are presented in Figure 1. Whenexposing the H4IIE cells to CdCl2 concentrations in the rangeof 10-4–10 μM for 36 h, a decline in cell viability was detectedat 10 μM Cd (32.6% cell survival) (Figure 1A). After expos-ing the same cell line to CuCl2 concentrations in the range of0.01–1000 μM for 36 h, a fall in cell viability was detected at1000 μM Cu (39.4% survival) (Figure 1B). When exposing the

H4IIE cells to ZnCl2 concentrations in the range of 0.01–500 μMfor 36 h, the cell viability showed a marked decrease at 100 μMZn (34.8% survival) (Figure 1C).

Data obtained when exposing the H4IIE cells to differentcombinations of metals (concentrations given in Table 2) arepresented in Figure 2. When the cells were exposed to highconcentrations of Cu and Cd in combination for 36 h, a signifi-cant decrease in cell viability was observed. None of the othermetal combinations induced any significant change in cellviability.

DNA Double-Strand Break AssayMedian molecular length (MML) values for the DNA frag-

ments in the different exposure categories were calculatedbased on the size separation of DNA fragments in the agarosegel and presented in Figure 3. Cells exposed to high concentra-tions of all three metals (Cu, Cd, and Zn) in combination hadMML values that were significantly higher than control values,indicating a lower frequency of DNA double-strand breaks inthese cells. The cells exposed to high concentrations of Cu andCd in combination had significantly lower MML values thanboth control and cells exposed to high concentrations of allmetals in combination, indicating a higher frequency of DNAdouble-strand breaks in these cells. All the other exposureconditions tested did not yield MML values that differedsignificantly from the control. In a separate experiment, thefollowing mixtures were tested: high concentrations of Cu andCd with low concentration of Zn, low concentrations of Cu andCd, low concentration of Cu with high concentration of Cd,and high concentration of Cu with low concentration of Cd.None of these mixtures yielded MML values different fromcontrol values (data not shown).

DISCUSSIONThe aim of the present study was to evaluate induction of

DNA double-strand breaks in the H4IIE cell line as a biomarkerof exposure to environmentally relevant concentrations ofmetals Cu, Cd, and Zn. These metals are common environmentalpollutants that often are found together in nature, related tomining runoffs or emissions from smelting industries.

H4IIE Cell Viability After Exposure to Cd, Cu, and ZnFor assessment of cell viability, the H4IIE cells were

exposed to the metals Cd, Cu, and Zn both singly and in com-binations. When exposing the cells to a range of increasing Cdconcentrations, a marked decline in cell viability was observedbetween 1 and 10 μM Cd. In the same cell line, Kim et al.(2003) also reported a decrease in cell viability after exposureto 0.3 μM Cd for 12 h, and after exposure to concentrationsbetween 1 and 2 μM Cd (Son et al., 2001), which are in agree-ment with our findings. However, in the experiments by Kimet al. (2003), the Cd concentration producing a decline in cell

Dow

nloa

ded

by [

UZ

H H

aupt

bibl

ioth

ek /

Zen

tral

bibl

ioth

ek Z

üric

h] a

t 11:

43 2

6 A

ugus

t 201

3

DNA DOUBLE-STRAND BREAKS IN THE H4IIE CELL LINE 159

viability was lower than the findings in our study. This maypossibly be explained by the fact that the H4IIE cells in theexperiments by Kim et al. (2003) had been starved for 12 hprior to exposure. Cadmium cytotoxicity has been associated withthe metal interfering with mitochondrial membrane potentials,leading to ROS formation and lipid peroxidation (Pourahmad &O’Brien, 2000).

When exposing the H4IIE cells to a range of Cu concen-trations, a fall in cell viability was found to occur between500 and 1000 μM Cu. Rau et al. (2004) found cell survival of75% after exposing H4IIE cells to 500 μM of Cu for 12 h.This indicates lower cell viability than the findings in ourexperiment. In a study with another cell line, Hep G2, Sethet al. (2004) showed a decline in cell viability after exposureto 50 μM Cu after both 24 and 48 h of exposure, indicatingthat the H4IIE cell line is relatively tolerant to Cu exposure

compared to the Hep G2 cell line. Copper bound to metal-lothionein enters cellular lysosomes. Copper cytotoxicity wasshown to arise from production of ROS in lysosomes, withfollowing lysosomal membrane damage and release of lysosomalproteases and phospholipases into the intracellular matrix(Pourahmad et al., 2001).

After exposing the H4IIE cells to a range of increasing Znconcentrations, a decrease in cell viability was observedbetween 10 and 100 μM. No comparable experiments assess-ing cell viability after Zn exposure in the H4IIE cell line wasfound in the literature. Devergnas et al. (2004) showed adecline in cell viability in the HeLa cell line at concentrationsof 150 μM and higher, which may indicate that the H4IIE cellline is more sensitive to Zn exposure than HeLa cells.

Exposing H4IIE cells to combinations of the three metalsgave variable results. The cells exposed to a combination of

FIG. 1. Cell viability in the H4IIE cells given as percent of control values after 36 h of exposure to (A) CdCl2, where each point represents the mean ± SD of 5parallels for each concentration; (B) CuCl2, where the circular points represents the mean ± SD for 7 parallels, the triangular point the mean ± SD for 2 parallels,and the square point the mean ± SD for 5 parallels; and (C) ZnCl2 where the circular points represent the mean ± SD for 2 parallels, the triangular points the mean± SD for 3 parallels, and the square points the mean ± SD for 5 parallels.

µM CdCl2

10–5 10–4 10–3 10–2

(a) (b)

(c)

10–1 100 101 102

µM CuCl2

10–3 10–2 10–1 100 101102 103 104

µM ZnCl2

10–3 10–2 10–1 100 101 102 103

% C

ell v

iabi

lity

% C

ell v

iabi

lity

0

20

40

60

80

100

120

140

160

% C

ell v

iabi

lity

0

20

40

60

80

100

120

140

0

20

40

60

80

100

120

140

160

Dow

nloa

ded

by [

UZ

H H

aupt

bibl

ioth

ek /

Zen

tral

bibl

ioth

ek Z

üric

h] a

t 11:

43 2

6 A

ugus

t 201

3

160 R. HALDSRUD AND Å. KRØKJE

1 μM Cd and 100 μM Cu showed a significant reduction in cellviability compared to control. This is in agreement with Fotakisand Timbrell (2006), who found a decrease in cell viability whencoexposing the HTC cell line and the HepG2 cell line to Cu

and Cd, as measured by the neutral red (NR) assay. However,in the same study, coexposure to Cu and Cd showed no effectson lactate dehydrogenase (LDH) leakage, and the authors sug-gested that Cu affected the lysosomal stability in these cell

FIG. 2. Cell viability in the H4IIE cells given as percent of control after 36 h of exposure to combinations of the metals Cd, Cu, and Zn (given in Table 2). Eachbar represents the mean ± SD for each exposure category. The control bar is based on 29 parallels, with the bars representing high concentrations of Cu and Cdcombined, and low concentrations of Cu and Cd combined are based on 13 parallels. The rest of the bars are based on 6 parallels.

FIG. 3. DNA fragment size distributions given as MML values in H4IIE cells exposed to the metals Cd and Cu singly, and to Cd, Cu, and Zn in differentcombinations (concentrations are given in Table 2). In each box, the top line represents the 75% fractile and the bottom line represents the 25% fractile. The solid-drawn line inside the box represents the median value, and the dotted line represents the mean. The bars above the box represent the 90% fractile, and the barsbelow the box represent the 10% fractile.

Dow

nloa

ded

by [

UZ

H H

aupt

bibl

ioth

ek /

Zen

tral

bibl

ioth

ek Z

üric

h] a

t 11:

43 2

6 A

ugus

t 201

3

DNA DOUBLE-STRAND BREAKS IN THE H4IIE CELL LINE 161

lines. Urani et al. (2005) found that preexposure of the HepG2cell line to non-cytotoxic concentrations of Zn prior to Cdexposure decreased the cytotoxicity of Cd, possibly due tometallothionein induction by Zn. None of the other metal com-binations tested were significantly different from control. Withthe exception of cells exposed to high concentrations of Cu andCd in combination, the findings in our study indicated no addi-tive effects or synergistic interactions between the metalsoccurred.

DNA Double-Strand Break InductionAssessment of DNA double-strand break induction in the

H4IIE cells after metal exposure was performed by comparingMML values calculated from the agarose gel image dataobtained from electrophoretic analysis of the differentiallyexposed cell cultures. A significant reduction in MML valueswas found in cells exposed to 1 μM Cd and 100 μM Cu in com-bination. Both Cu and Cd are known ROS inducers (Waisberget al., 2003; Valko et al., 2005). Cadmium has, as previouslymentioned, the ability to participate in ROS formation throughdisturbances of mitochondrial electron transfer chains (Wanget al., 2004) and processes involving lysosomal membranedamage (Fotakis et al., 2005), while Cu produces ROS throughthe Fenton mechanism (Valko et al., 2005). Cadmium has beenassociated with formation of hydrogen peroxide, and superox-ide radicals, which are converted to hydrogen peroxide bySOD. Hydrogen peroxide is catalyzed to reactive hydroxyl rad-icals via the Fenton reaction (Szuster-Ciesielska et al., 2000;Shih et al., 2004; Bertin & Averbeck, 2006). An increased pro-duction of hydroxyl radicals when coexposing the cells to Cuand Cd might explain the decrease in cell viability and increasein DNA double-strand break formation that was observed inour study. ROS induce single-strand breaks directly by attack-ing deoxyribose, or indirectly as intermediate steps in the baseexcision repair pathway for repair of oxidized DNA bases. Thisfurther leads to induction of DNA double-strand breaks duringreplication (Evans et al., 2004; Helleday et al., 2007; Hedgeet al., 2008). The ability of Cd to inhibit DNA repair mecha-nisms (Dally & Hartwig, 1997; Asmuss et al., 2000; Jin et al.,2003) might also contribute to the observed effect.

When cells were exposed to 1 μM Cd, 100 μM Cu, and 10 μMZn in combination, the DNA fragment sizes had significantlyhigher values than both control cells and cells exposed to highconcentrations of Cu and Cd in combination without Znpresent. Zinc is an essential element that plays a critical role inantioxidant enzymes such as Cu/Zn-SOD and in Zn fingerproteins involved in cell signal pathways, DNA repair mecha-nisms, and transcription factors (Ho et al., 2004). The SigmaRPMI 1640 medium formulation does not include Zn salts,which may explain why the DNA damage level in our study islower than in control cells when high concentrations of thiselement are present. Hormesis is a favorable biologicalresponse to low levels of toxins, and is characterized by induction

of stress proteins such as heat-shock protein 70 (Hsp70) andmetallothionein (Damelin et al., 2000). All the metals in ourstudy are known inducers of Hsp70, and both Cd and Zninduce metallothionein (Murata et al., 1999). As previouslymentioned, both Cu and Cd are known to induce ROS. Zincwas found to protect sulfhydryl groups against oxidativedamage (Bray & Bettger, 1990), compete with toxic metals forcellular binding sites, and induce metallothionein (Kägi &Schäffer, 1988). Metallothionein is present in the H4IIE cellline at detectable levels (Son et al., 2001). The binding affinityto metallothionein for the metals in our study is as follows:Cu > Cd > Zn (Kägi & Schäffer, 1988). The induction poten-cies of the metals and the binding affinity taken togethermight offer an explanation for our results, with Zn inducingmetallothionein, which in turn binds Cu and Cd. Fotakis andTimbrell (2006) found a decrease in Cd uptake in the HepG2and HTC hepatoma cell lines when Zn was present and pro-posed competition for uptake routes as a possible mecha-nism. The same mechanism was suggested to be involved inan in vivo study concerning Cd-induced hepatotoxicity andZn protection in male LE rats (Takamure et al., 2006).Fotakis and Timbrell (2006) also reported an increase in Cduptake in the HTC cells when Cu was present, while theuptake of Cd decreased when Zn was present. Increased Cduptake in the hepatoma cell line with the presence of Cu mayperhaps help explain both the decrease in cell viability andincrease in DNA double-strand break levels found in ourexperiment, whereas the decreased uptake shown by coexpo-sure to zinc might explain some of the protective effect ofthis metal. When coexposing HeLa cells to Cd and Zn,Szuster-Ciesielska et al. (2000) showed that the metalsmutually influenced and decreased the penetration of themetals into the cells.

The other exposure conditions tested did not differ signifi-cantly from controls. Environmentally relevant concentrationsof the metals Cd, Cu, and Zn did not appear to produce anelevated level of DNA double-strand breaks in exposed H4IIEcell line. However, extrapolation of data obtained in vitro to invivo conditions is not a straightforward matter. Samplesobtained from the environment also, in addition to the metalsin question, contain a wide range of other components with theability to affect the response of the cultured cells.

REFERENCESAastrup, P., Riget, F., Dietz, R., and Asmund, G. 2000. Lead, zinc, cadmium,

mercury, selenium and copper in Greenland caribou and reindeer (Rangifertarandus). Sci. Total Environ. 245:149–159.

Asmuss, M., Mullenders, L. H. F., Eker, A., and Hartwig, A. 2000. Differen-tial effects of toxic metal compounds on the activities of Fpg and XPA,two zinc finger proteins involved in DNA repair. Carcinogenesis21:2097–2104.

Berg, J. M., and Shi, Y. 1996. The galvanization of biology: A growing appre-ciation for the roles of zinc. Science 271:1081–1085.

Bertin, G., and Averbeck, D. 2006. Cadmium: cellular effects, modifications ofbiomolecules, modulation of DNA repair and genotoxic consequences (areview). Biochimie 88:1549–1559.

Dow

nloa

ded

by [

UZ

H H

aupt

bibl

ioth

ek /

Zen

tral

bibl

ioth

ek Z

üric

h] a

t 11:

43 2

6 A

ugus

t 201

3

162 R. HALDSRUD AND Å. KRØKJE

Bradley, M. O., Taylor, V. I., Armstrong, M. J., and Galloway, S. M. 1987.Relationships among cytotoxicity, lysosomal breakdown, chromosomeaberrations, and DNA double-strand breaks. Mutat. Res. 189:69–79.

Bray, T. M., and Bettger, W. J. 1990. The physiological role of zinc as an anti-oxidant. Free Radical Biol. Med. 8:281–291.

Chan, H. M., and Cherian, M. G. 1992. Protective roles of metallothionein andglutathione in hepatotoxicity of cadmium. Toxicology 72:281–290.

Dally, H., and Hartwig, A. 1997. Induction and repair inhibition of oxidativeDNA damage by nickel(II) and cadmium(II) in mammalian cells. Carcino-genesis 18:1021–1026.

Damelin L. H., Vokes, S., Whitcutt, J. M., Damelin, S. B., and Alexander, J. J.2000. Hormesis: A stress response in cells exposed to low levels of heavymetals. Hum. Exp. Toxicol. 19:420–430.

Dehn, L.-A., Sheffield, G. G., Follmann, E. H., Duffy, L. K., Thomas, D. L.,Bratton, G. R., Taylor, R. J., and O’Hara, T. M. 2005. Trace elements intissues of phocid seals harvested in the Alaskan and Canadian Arctic:Influence of age and feeding ecology. Can. J. Zool. 83:726–746.

Devergnas, S., Chimienti, F., Naud, N., Pennequin, A., Coquerel, Y., Chantegrel, J.,Favier, A., and Seve M., 2004. Differential regulation of zinc efflux trans-porters ZnT-1, ZnT-5 and ZnT-7 gene expression by zinc levels: A real-time RT–PCR study. Biochem. Pharmacol. 68:699–709.

Eaton, D. L., and Gilbert, S. S. 2008. Principles of toxicology. In Casarett &Doull’s toxicology: The basic science of poisons, 7th ed., ed. C. D.Klaasen, pp. 11–44. New York: McGraw-Hill.

Evans, M. D., Dizdaroglu, M., and Cooke, M. S. 2004. Oxidative DNAdamage and disease: induction, repair and significance. Mutat. Res.568:1–61.

Fatur, T., Lah, T. T., and Filipi6, M. 2003. Cadmium inhibits repair of UV-,methyl methanesulfonate- and N-methyl-N-nitrosourea-induced DNAdamage in Chinese hamster ovary cells. Mutat. Res. 529:109–116.

Filipi6, M., and Hei, T. K., 2004. Mutagenicity of cadmium in mammaliancells: implication of oxidative DNA damage. Mutat. Res. 546:81–91.

Fotakis, G., and Timbrell, J. A., 2006. Role of trace elements in cadmium chlo-ride uptake in hepatoma cell lines. Toxicol. Lett. 164:97–103.

Fotakis, G., Cemeli, E., Anderson, D., and Timbrell, J. A., 2005. Cadmiumchloride-induced DNA and lysosomal damage in a hepatoma cell line.Toxicol. In Vitro 19:481–489.

Galán, A., García-Bermejo, L., Troyano, A., Vilaboa, N. E., Fernández, C.,de Blas, E., and Aller, P. 2001. The role of intracellular oxidation indeath induction (apoptosis and necrosis) in human promonocytic cellstreated with stress inducers (cadmium, heat, X-rays). Eur. J. Cell Biol.80:312–320.

Gamberg, M., Palmer, M., and Roach, P. 2005. Temporal and geographictrends in trace element concentrations in moose from Yukon, Canada. Sci.Total Environ. 351–352:530–538.

Grant, G. M., Jackman, S. M., Kolanko, C. J., and Stenger, D. A. 2001. JP-8 jetfuel-induced DNA damage in H4IIE rat hepatoma cells. Mutat. Res.490:67–75.

Hartwig, A. 1998. Carcinogenicity of metal compounds: possible role of DNArepair inhibition. Toxicol. Lett. 102/103:235–239.

Hedge, M. L., Hazra, T. K., and Mitra, S. 2008. Early steps in the DNA baseexcision/single strand interruption repair pathway in mammalian cells. CellRes. 18:27–47.

Helleday, T., Lo, J., van Gent, D. C., Engelward, and B. P. 2007. DNA doublestrand break repair: From mechanistic understanding to cancer treatment.DNA Repair 6:923–936.

Ho, E., Courtemanche C., and Ames, B. N. 2003. Zinc deficiency inducesoxidative DNA damage and increases p53 expression in human lung fibro-blasts. J. Nutr. 133:2543–2548.

Ho, E. 2004. Zinc deficiency, DNA damage and cancer risk. J. Nutr. Biochem.15:572–578.

Ikemoto, T., Kunito, T., Tanaka, H., Baba, N., Miyazaki, N., and Tanabe, S.2004. Detoxification mechanism of heavy metals in marine mammals andseabirds: Interaction of selenium with mercury, silver, copper, zinc, andcadmium in liver. Arch. Environ. Contam. Toxicol. 47:402–413.

Jin, Y. H., Clark, A. B., Slebos, R. J. C., Al-Refai, H., Taylor, J. A., Kunkel, T.A., Resnick, M. A., and Gordenin, D. A. 2003. Cadmium is a mutagen thatacts by inhibiting mismatch repair. Nat. Genet. 34:326–329.

Kägi, J. H. R., and Schäffer, A. 1988. Biochemistry of metallothionein. Bio-chemistry 27:8509–8515.

Kim, S. C., Cho, M. K., and Kim, S. G. 2003. Cadmium-induced non-apoptoticcell death mediated by oxidative stress under the condition of sulfhydryldeficiency. Toxicol. Lett. 144:325–336.

Klaassen, J. D., Liu, J., and Choudhuri, S. 1999. Metallothionein: An intracel-lular protein to protect against cadmium toxicity. Annu. Rev. Pharmacol.Toxicol. 39:267–294.

Kraemer, L. D., Campbell, P. G. C., and Hare, L. 2005. A field study examin-ing metal elimination kinetics in juvenile yellow perch (Perca flavescens).Aquat. Toxicol. 75:108–126.

Krøkje, Å., Bingham, C., Tuven, R. H., and Gabrielsen, G. W. 2006. Chromo-some aberrations and DNA strand breaks in glaucous gull (Larus hyper-boreus) chicks fed environmentally contaminated gull eggs. J. Toxicol.Environ. Health A 69:159–174.

Liu, J., Goyer, R. A., and Waalkes, M. P. 2008. Toxic effects of metals. InCasarett & Doull’s toxicology: The basic science of poisons, eds.C. D. Klaasen, M. O. Amdur, and J. Doull, pp. 931–979. New York:McGraw-Hill.

Moriwaki, H., Osborne, M. R., and Phillips, D. H. 2008. Effects of mixingmetal ions on oxidative DNA damage mediated by a Fenton-type reduc-tion. Toxicol. In Vitro 22:36–44.

Mosmann, T. 1983. Rapid colorimetric assay for cellular growth and survival:Application to proliferation and cytotoxicity assays. J. Immunol. Methods65:55–63.

Mukherjee, J. J., Gupta, S.K., Kumar, S., and Sikka, H. C. 2004. Effects of cad-mium (II) on (±)-anti-benzo[a]pyrene-7,8-diol-9,10-epoxide-induced DNAdamage response in human fibroblasts and DNA repair: A possible mecha-nism of cadmium’s cogenotoxicity. Chem. Res. Toxicol. 17:287–293.

Murata, M., Gong, P., Suzuki, K., and Koizumi, S. 1999. Differential metalresponse and regulation of human heavy metal-inducible genes. J. Cell.Physiol. 180:105–113.

Oliveira, S. C. B., Corduneanu, O., and Oliveira-Brett, A. M. 2008. In situevaluation of heavy metal–DNA interactions using an electrochemicalDNA biosensor. Bioelectrochemistry 72:53–58.

Olsvik, P. A., Gundersen, P., Andersen, R. A., and Zachariassen, K. E. 2001.Metal accumulation and metallothionein in brown trout, Salmo trutta, fromtwo Norwegian rivers differently contaminated with Cd, Cu and Zn. Comp.Biochem. Physiol. C 128:189–201.

Park, J. D., Liu, Y., and Klaassen, C. D. 2001. Protective effect of metallothioneinagainst the toxicity of cadmium and other metals. Toxicology 163:93–100.

Pfeiffer, P. 1998. The mutagenic potential of DNA double-strand break repair.Toxicol. Lett. 96–97:119–129.

Pourahmad, J., and O’Brien, P. J. 2000. A comparison of hepatocyte cytotoxicmechanisms for Cu2+ and Cd2+. Toxicology 143:263–273.

Pourahmad, J., Ross, S., and O’Brien, P. J. 2001. Lysosomal involvement inhepatocyte cytotoxicity induced by Cu2+ but not Cd2+. Free Radical Biol.Med. 30:89–97.

Rau, M. A., Whitaker, J., Freedman, J. H., and Di Giulio, R. T. 2004. Differen-tial susceptibility of fish and rat liver cells to oxidative stress and cytotoxic-ity upon exposure to prooxidants. Comp. Biochem. Physiol. 137:335–342.

Sagripanti, J.-L., Goering, P. L., and Lamanna, A. 1991. Interactions of copperwith DNA and antagonism by other metals. Toxicol. Appl. Pharmacol.110:477–485.

Savinov, V., Gabrielsen, G. W., and Savinova, T. N. 2003. Cadmium, zinc, cop-per, arsenic, selenium and mercury in seabirds from the Barents Sea: Levels,inter-specific and geographical differences. Sci. Total Environ. 306:133–158.

Seth, R., Yang, S., Choi, S., Sabean, M., and Roberts, E. A. 2004. In vitroassessment of copper-induced toxicity in the human hepatoma line, HepG2. Toxicol. In Vitro 18:501–509.

Shih, C.-M., Ko, W.-C., Wu, J.-S., Wei, Y.-H., Wang, L.-F., Chang, E.-E.,Lo, T.-Y-, Cheng, H.-H., and Chen, C.-T. 2004. Mediating of caspase-independent apoptosis by cadmium through the mitochondria-ROS path-way in MRC-5 fibroblasts. J. Cell. Biochem. 91:384–397.

Shugart, L. R. 2000. DNA damage as a biomarker of exposure. Ecotoxicology9:329–340.

Son, M. H., Kang, K. W., Lee, C. H., and Kim, S. G. 2001. Potentiation ofcadmium-induced cytotoxicity by sulfur amino acid deprivation through

Dow

nloa

ded

by [

UZ

H H

aupt

bibl

ioth

ek /

Zen

tral

bibl

ioth

ek Z

üric

h] a

t 11:

43 2

6 A

ugus

t 201

3

DNA DOUBLE-STRAND BREAKS IN THE H4IIE CELL LINE 163

activation of extracellular signal-regulated kinase1/2 (ERK1/2) in conjunc-tion with p38 kinase or c-jun N-terminal kinase (JNK) Complete inhibitionof the potentiated toxicity by U0126 an ERK1/2 and p38 kinase inhibitor.Biochem. Pharmacol. 62:1379–1390.

Stern, B. R., Solioz, M., Krewski, D., Aggett, P., Aw, T.-C., Baker, S., Crump, K.,Dourson, M., Haber, L., Hertzberg, R., Keen, C., Meek, B., Rudenko, L.,Schoeny, R., Slob., W., and Starr, T. 2007. Copper and human health: Bio-chemistry, genetics and strategies for modelling dose-response relationships.J. Toxicol. Environ. Health B 10:157–222.

Storelli, M. M., Storelli, A., D’Addabbo, R., Marano, C., Bruno, R., andMarcotrigiano, G. O. 2005. Trace elements in loggerhead turtles (Carettacaretta) from the eastern Mediterranean Sea: overview and evaluation.Environ. Pollut. 135:163–170.

Szuster-Ciesielska, A., Stachura, A., Slotwinska, M., Kaminska, T., Sniezko,R., Paduch, R., Abramczyk, D., Filar, J., and Kandefer-Szerszen, M. 2000.The inhibitory effect of zinc on cadmium-induced cell apoptosis and reac-tive oxygen species (ROS) production in cell cultures. Toxicology145:159–171.

Takamure, Y., Shimada, H., Kiyozumi, M., Yasutake, A., and Imamurac, Y.2006. A possible mechanism of resistance to cadmium toxicity in maleLong–Evans rats. Environ. Toxicol. Pharmacol. 21:231–234.

Theodorakis, C. W., D’Surney, S. J., and Shugart, L. R. 1994. Detection ofgenotoxic insult as DNA strand breaks in fish blood cells by agarose gelelectrophoresis. Environ. Toxicol. Chem. 13:1023–1031.

Uauy, R., Olivares, M., and Gonzalez, M. 1998. Essentiality of copper inhumans. Am. J. Clin. Nutr. 67:952S–959S.

Urani, C., Melchioretto, P., Canevali, C., and Crosta, G. F. 2005. Cytotoxicityand induction of protective mechanisms in HepG2 cells exposed tocadmium. Toxicol. In Vitro 19:887–892.

Valko, M., Morris, H., and Cronin, M. T. D. 2005. Metals, toxicity and oxida-tive stress. Curr. Med. Chem. 12:1161–1208.

Waisberg, M., Joseph, P., Hale, B., and Beyersmann, D. 2003. Molecularand cellular mechanisms of cadmium carcinogenesis. Toxicology192:95–117.

Wang, Y., Fang, J., Leonard, S. S., and Rao, K. M. K. 2004. Cadmium inhibitsthe electron transfer chain and induces reactive oxygen species. Free Radi-cal Biol. Med. 36:1434–1443.

Watanabe, T., Kiron, V., and Satoh, S. 1997. Trace minerals in fish nutrition.Aquaculture 151:185–207.

Wätjen, W., Michels, G., Steffan, B., Niering, P., Chovolou, Y., Kampkötter,A., Tran-Thi, Q.-H., Proksch, P., and Kahl, R. 2005. Low concentrations offlavonoids are protective in rat H4IIE cells whereas high concentrationscause DNA damage and apoptosis. J. Nutr. 135:525–531.

Whyte, J. J., Schmitt, C. J., and Tillitt, D. E. 2004. The H4IIE cell bioassay asan indicator of dioxin-like chemicals in wildlife and the environment. Crit.Rev. Toxicol. 34:1–83.

Würgler, F. E., and Kramers, P. G. N. 1992. Environmental effects of genotoxins(eco-genotoxicology). Mutagenesis 7:321–327.

Dow

nloa

ded

by [

UZ

H H

aupt

bibl

ioth

ek /

Zen

tral

bibl

ioth

ek Z

üric

h] a

t 11:

43 2

6 A

ugus

t 201

3