Top Banner
Please cite this article in press as: P. Rossner Jr., et al., Nonhomologous DNA end joining and chromosome aberrations in human embryonic lung fibroblasts treated with environmental pollutants, Mutat. Res.: Fundam. Mol. Mech. Mutagen. (2014), http://dx.doi.org/10.1016/j.mrfmmm.2014.03.006 ARTICLE IN PRESS G Model MUT 11334 1–11 Mutation Research xxx (2014) xxx–xxx Contents lists available at ScienceDirect Mutation Research/Fundamental and Molecular Mechanisms of Mutagenesis j ourna l h om epage: www.elsevier.com/l ocate/molmut Comm unit y ad dress: www.elsevier.com/locate/mutres Nonhomologous DNA end joining and chromosome aberrations in human embryonic lung fibroblasts treated with environmental pollutants Pavel Rossner Jr. , Andrea Rossnerova, Olena Beskid, Nana Tabashidze, Helena Libalova, Q1 Katerina Uhlirova, Jan Topinka, Radim J. Sram Department of Genetic Ecotoxicology, Institute of Experimental Medicine AS CR, Prague, Czech Republic a r t i c l e i n f o Article history: Received 18 January 2014 Received in revised form 25 February 2014 Accepted 7 March 2014 Available online xxx Keywords: Benzo[a]pyrene Chromosome aberrations Double-strand DNA breaks Extractable organic matter Nonhomologous DNA end joining repair a b s t r a c t In order to evaluate the ability of a representative polycyclic aromatic hydrocarbon (PAH) and PAH- containing complex mixtures to induce double strand DNA breaks (DSBs) and repair of damaged DNA in human embryonic lung fibroblasts (HEL12469 cells), we investigated the effect of benzo[a]pyrene (B[a]P) and extractable organic matter (EOM) from ambient air particles <2.5 m (PM2.5) on nonhomologous DNA end joining (NHEJ) and induction of stable chromosome aberrations (CAs). PM2.5 was collected in winter and summer 2011 in two Czech cities differing in levels and sources of air pollutants. The cells were treated for 24 h with the following concentrations of tested chemicals: B[a]P: 1 M, 10 M, 25 M; EOMs: 1 g/ml, 10 g/ml, 25 g/ml. We tested several endpoints representing key steps leading from DSBs to the formation of CAs including histone H2AX phosphorylation, levels of proteins Ku70, Ku80 and XRCC4 participating in NHEJ, in vitro ligation activity of nuclear extracts of the HEL12469 cells and the frequency of stable CAs assessed by whole chromosome painting of chromosomes 1, 2, 4, 5, 7 and 17 using fluorescence in situ hybridization. Our results show that 25 M of B[a]P and most of the tested doses of EOMs induced DSBs as indicated by H2AX phosphorylation. DNA damage was accompanied by induction of XRCC4 expression and an increased frequency of CAs. Translocations most frequently affected chromosome 7. We observed only a weak induction of Ku70/80 expression as well as ligation activity of nuclear extracts. In summary, our data suggest the induction of DSBs and NHEJ after treatment of human embryonic lung fibroblasts with B[a]P and complex mixtures containing PAHs. © 2014 Published by Elsevier B.V. 1. Introduction Combustion of organic material generates a complex mixture of chemicals and represents an important source of fine particle (PM2.5) air pollution [1]. PM2.5 is deposited in lung alveoli where it causes inflammation. Chemical compounds bound to PM2.5 may Abbreviations: B[a]P, benzo[a]pyrene; CA, chromosome aberration; c-PAH, carcinogenic polycyclic aromatic hydrocarbon; DSB, double strand DNA break; DMSO, dimethylsulfoxide; EOM, extractable organic matter; FG/100, genomic fre- quency of translocations per 100 cells; FISH, fluorescence in situ hybridization; -H2AX, phosphorylated H2AX; HEL12469, human embryonic lung fibroblasts; NEX, nuclear extract; NHEJ, nonhomologous end joining repair; PAH, polycyclic aromatic hydrocarbon; PM, particulate matter; PM2.5, particulate matter of aerodynamic diameter < 2.5 m; ROS, reactive oxygen species. Corresponding author at: Videnska 1083, 14220 Prague, Czech Republic. Tel.: +420 241062763; fax: +420 241062785. E-mail address: [email protected] (P. Rossner Jr.). then enter the bloodstream and cause damage to both lympho- cytes and cells of distant organs. Recently, the International Agency for Research on Cancer (IARC) classified outdoor air pollution and particulate matter from outdoor air pollution as carcinogenic to humans [2]. Mechanisms of carcinogenicity include direct DNA damage (chromosomal aberrations, bulky DNA adduct formation, oxidative stress), as well as changes in gene expression involved in DNA damage and repair pathways, inflammation, immune and oxidative stress response [2]. Carcinogenic polycyclic aromatic hydrocarbons (c-PAHs) are responsible for most of the genotoxic activity of organic extracts from particulate matter (EOM) resulting in the formation of bulky DNA adducts [3]. Benzo[a]pyrene (B[a]P), a model c-PAH, is classified as carcinogenic to humans [4]. There are three major pathways of detoxification/metabolic activation of PAHs: the pathway via dihydrodiol epoxides employ- ing CYP450 enzymes, the pathway via radical cations by one electron oxidation, and the pathway catalyzed by dihydrodiol dehydrogenases resulting in the formation of ortho-quinones [5]. http://dx.doi.org/10.1016/j.mrfmmm.2014.03.006 0027-5107/© 2014 Published by Elsevier B.V. 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45
11

Nonhomologous DNA end joining and chromosome aberrations in human embryonic lung fibroblasts treated with environmental pollutants

Apr 21, 2023

Download

Documents

Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: Nonhomologous DNA end joining and chromosome aberrations in human embryonic lung fibroblasts treated with environmental pollutants

M

Nhp

PQ1

KD

a

ARRAA

KBCDEN

1

o(i

cDq�nhd

T

h0

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

21

22

23

24

25

26

27

ARTICLE IN PRESSG ModelUT 11334 1–11

Mutation Research xxx (2014) xxx–xxx

Contents lists available at ScienceDirect

Mutation Research/Fundamental and MolecularMechanisms of Mutagenesis

j ourna l h om epage: www.elsev ier .com/ l ocate /molmutComm uni t y ad dress : www.elsev ier .com/ locate /mutres

onhomologous DNA end joining and chromosome aberrations inuman embryonic lung fibroblasts treated with environmentalollutants

avel Rossner Jr. ∗, Andrea Rossnerova, Olena Beskid, Nana Tabashidze, Helena Libalova,aterina Uhlirova, Jan Topinka, Radim J. Sram

epartment of Genetic Ecotoxicology, Institute of Experimental Medicine AS CR, Prague, Czech Republic

r t i c l e i n f o

rticle history:eceived 18 January 2014eceived in revised form 25 February 2014ccepted 7 March 2014vailable online xxx

eywords:enzo[a]pyrenehromosome aberrationsouble-strand DNA breaksxtractable organic matteronhomologous DNA end joining repair

a b s t r a c t

In order to evaluate the ability of a representative polycyclic aromatic hydrocarbon (PAH) and PAH-containing complex mixtures to induce double strand DNA breaks (DSBs) and repair of damaged DNA inhuman embryonic lung fibroblasts (HEL12469 cells), we investigated the effect of benzo[a]pyrene (B[a]P)and extractable organic matter (EOM) from ambient air particles <2.5 �m (PM2.5) on nonhomologousDNA end joining (NHEJ) and induction of stable chromosome aberrations (CAs). PM2.5 was collected inwinter and summer 2011 in two Czech cities differing in levels and sources of air pollutants. The cellswere treated for 24 h with the following concentrations of tested chemicals: B[a]P: 1 �M, 10 �M, 25 �M;EOMs: 1 �g/ml, 10 �g/ml, 25 �g/ml. We tested several endpoints representing key steps leading fromDSBs to the formation of CAs including histone H2AX phosphorylation, levels of proteins Ku70, Ku80and XRCC4 participating in NHEJ, in vitro ligation activity of nuclear extracts of the HEL12469 cells andthe frequency of stable CAs assessed by whole chromosome painting of chromosomes 1, 2, 4, 5, 7 and17 using fluorescence in situ hybridization. Our results show that 25 �M of B[a]P and most of the tested

doses of EOMs induced DSBs as indicated by H2AX phosphorylation. DNA damage was accompaniedby induction of XRCC4 expression and an increased frequency of CAs. Translocations most frequentlyaffected chromosome 7. We observed only a weak induction of Ku70/80 expression as well as ligationactivity of nuclear extracts. In summary, our data suggest the induction of DSBs and NHEJ after treatmentof human embryonic lung fibroblasts with B[a]P and complex mixtures containing PAHs.

© 2014 Published by Elsevier B.V.

28

29

30

. Introduction

Combustion of organic material generates a complex mixture

Please cite this article in press as: P. Rossner Jr., et al., Nonhohuman embryonic lung fibroblasts treated with environmental phttp://dx.doi.org/10.1016/j.mrfmmm.2014.03.006

f chemicals and represents an important source of fine particlePM2.5) air pollution [1]. PM2.5 is deposited in lung alveoli wheret causes inflammation. Chemical compounds bound to PM2.5 may

Abbreviations: B[a]P, benzo[a]pyrene; CA, chromosome aberration; c-PAH,arcinogenic polycyclic aromatic hydrocarbon; DSB, double strand DNA break;MSO, dimethylsulfoxide; EOM, extractable organic matter; FG/100, genomic fre-uency of translocations per 100 cells; FISH, fluorescence in situ hybridization;-H2AX, phosphorylated H2AX; HEL12469, human embryonic lung fibroblasts; NEX,uclear extract; NHEJ, nonhomologous end joining repair; PAH, polycyclic aromaticydrocarbon; PM, particulate matter; PM2.5, particulate matter of aerodynamiciameter < 2.5 �m; ROS, reactive oxygen species.∗ Corresponding author at: Videnska 1083, 14220 Prague, Czech Republic.el.: +420 241062763; fax: +420 241062785.

E-mail address: [email protected] (P. Rossner Jr.).

ttp://dx.doi.org/10.1016/j.mrfmmm.2014.03.006027-5107/© 2014 Published by Elsevier B.V.

31

32

33

34

35

36

37

38

39

40

41

then enter the bloodstream and cause damage to both lympho-cytes and cells of distant organs. Recently, the International Agencyfor Research on Cancer (IARC) classified outdoor air pollution andparticulate matter from outdoor air pollution as carcinogenic tohumans [2]. Mechanisms of carcinogenicity include direct DNAdamage (chromosomal aberrations, bulky DNA adduct formation,oxidative stress), as well as changes in gene expression involvedin DNA damage and repair pathways, inflammation, immune andoxidative stress response [2]. Carcinogenic polycyclic aromatichydrocarbons (c-PAHs) are responsible for most of the genotoxicactivity of organic extracts from particulate matter (EOM) resultingin the formation of bulky DNA adducts [3]. Benzo[a]pyrene (B[a]P),a model c-PAH, is classified as carcinogenic to humans [4].

There are three major pathways of detoxification/metabolic

mologous DNA end joining and chromosome aberrations inollutants, Mutat. Res.: Fundam. Mol. Mech. Mutagen. (2014),

activation of PAHs: the pathway via dihydrodiol epoxides employ-ing CYP450 enzymes, the pathway via radical cations by oneelectron oxidation, and the pathway catalyzed by dihydrodioldehydrogenases resulting in the formation of ortho-quinones [5].

42

43

44

45

Page 2: Nonhomologous DNA end joining and chromosome aberrations in human embryonic lung fibroblasts treated with environmental pollutants

ING ModelM

2 ion Re

Bandrsshstaosbrsbrgc

a(oOcp(aatset

2

2

tPHlssp2fEccyFndpfwC

2

b

46

47

48

49

50

51

52

53

54

55

56

57

58

59

60

61

62

63

64

65

66

67

68

69

70

71

72

73

74

75

76

77

78

79

80

81

82

83

84

85

86

87

88

89

90

91

92

93

94

95

96

97

98

99

100

101

102

103

104

105

106

107

108

109

110

111

112

113

114

115

116

117

118

119

120

121

122

123

124

125

126

127

128

129

130

131

132

133

134

135

136

137

138

139

140

141

142

143

144

145

146

147

148

149

150

151

152

153

154

155

156

157

158

159

160

161

162

163

ARTICLEUT 11334 1–11

P. Rossner Jr. et al. / Mutat

ulky DNA adducts represent a predominant form of DNA dam-ge following insult by c-PAHs. They are repaired by the action ofucleotide excision repair. However, PAH-o-quinones generated byihydrodiol dehydrogenases are redox active and may contribute toeactive oxygen species (ROS) formation and subsequent oxidativetress [6]. ROS may attack DNA and cause DNA single- or double-trand breaks. If repaired by nonhomologous end joining (NHEJ) oromologous repair, double strand DNA breaks (DSBs) may result intable chromosomal aberrations, as translocations and reciprocalranslocations. Apart from this mechanism, DSBs may be induceds an indirect result of bulky DNA adduct formation. The presencef adducts may block DNA synthesis and uncoupling of the othertrand thus resulting in DSB formation [7]. The induction of unsta-le chromosomal aberrations by c-PAHs or their metabolites wasepeatedly observed in various cell lines [8–14]. Although chromo-omal translocations are more serious than unstable aberrationsecause they may be fixed in the genome potentially leading toearrangements of regulatory elements of genes including onco-enes [15], very few studies analyzing the effect of c-PAHs on modelell lines in vitro have been published [16,17].

In the present study, we aimed to analyze processes associ-ted with the treatment of human embryonic lung fibroblastsHEL12469 cells) with EOMs and B[a]P. EOMs were prepared byrganic extraction from PM2.5 collected in two Czech cities (Prague,strava) differing in sources and concentrations of c-PAHs. Weoncentrated on the induction of DSBs (measured as levels of phos-horylated histone H2AX), expression of selected NHEJ proteinsKu70, Ku80, XRCC4), ligation activity of HEL12469 nuclear extractsnd the frequency of stable aberrations of chromosomes 1, 2, 4, 5, 7nd 17. We hypothesized that ROS generation after EOM and B[a]Preatment will result in the induction of DSBs, followed by expres-ion of NHEJ proteins and increased ligation activity of nuclearxtracts which in turn will increase the frequency of chromosomeranslocations.

. Materials and methods

.1. PM2.5 collection, preparation of EOM and chemical analysis

Sample collection, EOM preparation and chemical characteriza-ion were described in detail in our previous study [18]. Briefly,M2.5 was collected by a HiVol 3000 air sampler (model ECO-VS3000, Ecotech, Australia) on Pallflex filters T60A20 in two

ocations in the Czech Republic: Prague (a sampler was located out-ide the city center, with moderate traffic intensity) and Ostrava (aampler was located in a heavily polluted industrial area). The sam-les were collected in the winter and summer seasons of 2011, for4 h each day for 4–5 weeks. The total number of filters obtainedor individual location and sampling season ranged from 28 to 31.ach filter was extracted with a mixture of dichloromethane andyclohexane, the extracts (EOMs) were pooled and aliquots used forhemical analysis and cell treatment. Quantitative chemical anal-sis of PAHs was performed by HPLC with fluorimetric detection.or the in vitro experiments, EOM samples were evaporated to dry-ess under a stream of nitrogen and the residue redissolved inimethylsulfoxide (DMSO). The stock solution of each EOM sam-le contained 50 mg of EOM/ml DMSO. EOMs were kept in thereezer at −80 ◦C. The extraction of PM2.5 and chemical analysesere performed in the laboratories of the certified company ALSzech Republic, Prague (EN ISO CSN IEC 17025).

Please cite this article in press as: P. Rossner Jr., et al., Nonhohuman embryonic lung fibroblasts treated with environmental phttp://dx.doi.org/10.1016/j.mrfmmm.2014.03.006

.2. Cell cultivation and treatment

The growth characteristics of the HEL12469 cells (ECACC, Salis-ury, UK) used in the present study and cytotoxicity of the tested

PRESSsearch xxx (2014) xxx–xxx

chemicals were described in detail previously [18]. The cells weregrown at 37 ◦C in a humidified atmosphere containing 5% CO2 inminimal essential medium EMEM supplemented with 10% fetalbovine serum (FBS), 2 mM glutamine, 1% non-essential amino acids,0.2% sodium bicarbonate, 50 U/ml penicillin and 50 �g/ml strepto-mycin. After reaching confluency, the cells were treated with 0.25%trypsin/0.02% EDTA in PBS and used for individual applications.We tested the following concentrations of B[a]P (Sigma–Aldrich, St.Louis, MO, USA) and EOM: B[a]P: 1, 10 and 25 �M; EOMs: 1, 10 and25 �g/ml. We assessed the cytotoxicity of the selected concentra-tions of the tested compounds using the cell proliferation reagent,WST-1 (Roche, Mannheim, Germany), following the manufacturer’sinstructions (see [18] for details).

2.3. Expression of �-H2AX, Ku70, Ku80 and XRCC4 proteins

The cells (1 × 106) were seeded in 75 cm2 tissue culture flasks(TPP, Trasadingen, Switzerland) in 15 ml culture medium sup-plemented with 10% FBS and incubated for 2–3 days at 37 ◦C.Treatment involved replacing the culture medium with 10 ml freshmedium containing the test chemicals. The cells were treated for24 h, harvested by scraping and stored at −80 ◦C until furtherprocessing.

For Western blotting analysis, the cells were lysed in extractionbuffer (1% SDS, 10% glycerol, 100 mM Tris, pH 7.4) supplementedwith protease and phosphatase inhibitors, and the protein con-centration was estimated using the BCA reagent (Sigma, St. Louis,MO, USA). Proteins (12 �g/sample) were separated by SDS-PAGEin 4–12.5% gels (0.75 mm) and transferred onto a Hybond-P PVDFmembrane (GE Healthcare, Piscataway, NJ, USA). The membranewas blocked for 2 h at room temperature in TBS buffer (10 mMTris, 150 mM NaCl, pH 7.4) containing 2% ECL Prime BlockingAgent (GE Healthcare, Piscataway, NJ, USA). The proteins weredetected by incubation of the membrane with primary antibod-ies against �-H2AX (ab22551; 0.1 �g/ml), Ku70 + Ku80 (ab53126;dilution 1:2000) and XRCC4 (ab97351; dilution 1:500) from Abcam(Cambridge, UK) at 10 ◦C overnight and a secondary antibodyconjugated with horseradish peroxidase (for �-H2AX: NA931, GEHealthcare, Piscataway, NJ, USA, dilution 1:10,000; for Ku70 + Ku80and XRCC4: NA934, GE Healthcare, Piscataway, NJ, USA; dilution1:100,000 and 1:10,000 for Ku70 + Ku80 and XRCC4, respectively)at room temperature for 1 h. The antibodies were diluted in TBS-T buffer (TBS + 0.1% Tween 20) containing 2% ECL Prime BlockingAgent. To develop the chemiluminiscence signal the membranewas incubated with ECL Prime WB Detection Reagent (GE Health-care, Piscataway, NJ, USA) for 5 min at room temperature in the darkand exposed to Amersham Hyperfilm (GE Healthcare, Piscataway,NJ, USA). The intensity of the bands on the scanned films was ana-lyzed using ImageJ software (http://rsbweb.nih.gov/ij/index.html).The results were expressed as a percentage of the band intensityof the control sample normalized to a loading control [the mem-brane was stained with amido black solution (0.1% Amido Black10B, Sigma–Aldrich, St. Louis, MO, USA; 45% methanol; 10% aceticacid)].

2.4. NHEJ repair activity

The activity of NHEJ was measured in nuclear extracts ofHEL12469 cells essentially as described in [19,20], with some mod-ifications. The cells (4 × 105) were seeded in 25 cm2 tissue cultureflasks (TPP, Trasadingen, Switzerland) and cultivated in 10 ml cul-ture medium supplemented with 10% FBS for 2–3 days. Before

mologous DNA end joining and chromosome aberrations inollutants, Mutat. Res.: Fundam. Mol. Mech. Mutagen. (2014),

treatment, the medium was replaced with 5 ml fresh mediumcontaining the test chemicals. After 24 h treatment, the cellswere harvested and immediately used for nuclear extract (NEX)preparation. NEXs were prepared using the NE-PER Nuclear and

164

165

166

167

Page 3: Nonhomologous DNA end joining and chromosome aberrations in human embryonic lung fibroblasts treated with environmental pollutants

ING ModelM

on Res

CMMboU(

dwi8jM1ustsr(uiwecnesaa

2

(pTm2aibfio1ttpcsws

VC5fiClGas

cT

168

169

170

171

172

173

174

175

176

177

178

179

180

181

182

183

184

185

186

187

188

189

190

191

192

193

194

195

196

197

198

199

200

201

202

203

204

205

206

207

208

209

210

211

212

213

214

215

216

217

218

219

220

221

222

223

224

225

226

227

228

229

230

231

232

233

234

235

236

237

238

239

240

241

242

243

244

245

246

247

248

249

250

251

252

253

254

255

256

257

258

259

260

261

262

263

264

265

266

267

268

269

270

271

272

273

274

275

276

277

278

279

280

281

282

283

284

285

286

ARTICLEUT 11334 1–11

P. Rossner Jr. et al. / Mutati

ytoplasmic Extraction Reagents Kit (Thermo Scientific, Waltham,A, USA) with protease inhibitors (Thermo Scientific, Waltham,A, USA) added. The volume of the reagents depended on the num-

er of cells harvested. Five microliters of NEX were used for analysisf protein concentration using the BCA Kit (Sigma, St. Louis, MO,SA) and the remaining volume of NEX was mixed with glycerol

final concentration 20% v/v) and stored at −80 ◦C until analysis.Plasmid pUC18 (Invitrogen, Eugene, OR, USA) linearized by

igestion with EcoRI (Thermo Scientific, Waltham, MA, USA)as used as a substrate for the ligation reaction. NEX contain-

ng 4 �g of proteins in a total volume of 6 �l was mixed with0 ng (concentration 40 ng/�l) of linearized pUC18, 1 �l of end-

oining mix (containing 200 mM HEPES, 800 mM KCl, 100 mMgCl2), 1 mg/ml bovine serum albumin, 1 mM dithiothreitol and

mM adenosine triphosphate. The reaction mixture (total vol-me of 20 �l) was incubated for 2 h at 25 ◦C. The reaction wastopped by adding RNase (final concentration 1 mg/ml) and pro-einase K (final concentration 1 mg/ml). DNA fragments wereeparated by electrophoresis in 1% agarose gel in TBE bufferunning at 5 V/cm for 1 h. Gels were stained with SYBR® SafeInvitrogen, Eugene, OR, USA) diluted 1:10,000× and visualizedsing the GelDoc-ItTM Imaging System (UVP, Cambridge, UK). The

ntensity of the DNA fragments was analyzed using ImageJ soft-are (http://rsbweb.nih.gov/ij/index.html). The repair activity was

xpressed as a percentage of ligation of the linearized substratealculated as (intensity of ligation products/substrate) × 100. Aegative control (linearized pUC18 without NEX) was run in eachxperiment. To verify the method before running the test samples aeries of optimization experiments was conducted using differentmounts of NEX proteins and pUC18 in a reaction mixture as wells different incubation periods.

.5. Fluorescence in situ hybridization (FISH)

The cells (1 × 106) were seeded in 75 cm2 tissue culture flasksTPP, Trasadingen, Switzerland) in 15 ml culture medium sup-lemented with 10% FBS and incubated for 2–3 days at 37 ◦C.reatment involved replacing the culture medium with 10 ml freshedium containing the test chemicals. The cells were treated for

4 h. Colchicine (Sigma–Aldrich, St. Louis, MO, USA) was added at final concentration of 0.5 �g/ml three hours before the end of thencubation. After treatment, the cells were trypsinized, collectedy centrifugation, resuspended in a hypotonic 0.075 M KCl solution,xed with methanol/acetic acid and stored at −20 ◦C until droppingn slides. The FISH staining protocol of chromosomes 1, 2, 4, 5, 7 and7 was adapted from the manufacturer’s instructions (MetaSys-ems, Altlussheim, Germany). FISH analysis was performed usingwo sets of Customized XCP-Mix probes: the first set containingrobes for chromosome 1 (red), 4 (green) and 17 (red and green,onverted as yellow); the second set with the same color labels butpecific for chromosomes 2, 5, and 7. The slides were first stainedith the first set, analyzed, destained and re-hybridized with the

econd set.Analyses of FISH slides counterstained with DAPI mixed with

ectashield mounting medium (Vector Laboratories, Burlingame,A, USA) were performed using a Zeiss Axioscope and Olympus BX1 microscopes (final magnification: 1000×) equipped with a triplelter to visualize simultaneously the DAPI (blue), FITC (green) andy3 (red) signals. Color images with CAs were collected and ana-

yzed using ISIS software, version 5.0 (MetaSystems, Altlussheim,ermany). The number of scored metaphases per tested chemicalnd concentration was 1500–3000 for each set of painted chromo-

Please cite this article in press as: P. Rossner Jr., et al., Nonhohuman embryonic lung fibroblasts treated with environmental phttp://dx.doi.org/10.1016/j.mrfmmm.2014.03.006

omes.All aberrant cells were classified according to the Proto-

ol for Aberration Identification and Nomenclature (PAINT) [21].he genomic frequencies of translocations (FG) were calculated

PRESSearch xxx (2014) xxx–xxx 3

as suggested by Lucas et al. [22]. Exchange (translocations)frequencies obtained from each chromosome were calculated forthe whole genome by dividing the observed frequencies by thefactor of 2.05fp(1 − fp), where fp is the fraction of painted DNAconverted by individual chromosomes. The genomic frequencyof translocations obtained from the three chromosomes together(including the exchanges between painted chromosomes) wereestimated by dividing the observed frequencies by the factor2.05{(fr + fg + fy)[1 − (fr + fg + fy)] + frfg + fgfy + frfy} [23], where fr, fgand fy are the fractions of the genome [24] converted by chromo-somes 1, 4 and 17, and 2, 5, 7, for the first and second painted setof chromosomes, respectively. Genomic frequencies of transloca-tions (FG) were recalculated per 100 cells and expressed as genomicfrequencies of translocations per 100 cells (FG/100). Conversion ofpainted metaphase cells to whole-genome equivalents (defined ascell equivalents) was calculated using equations appropriate forthree-color paints [25]. Cell equivalents ranged from 463 to 927 forpainted chromosomes 1, 4 and 17 and from 509 to 1017 for paintedchromosomes 2, 5 and 7.

2.6. Statistical analysis

Differences between individual groups in the mean values ofNHEJ repair activity were tested using the two-tailed Student’st-test; a p-value of 0.05 or less indicated a statistically signifi-cant difference between the compared groups. The calculationswere performed using IBM SPSS (Chicago, IL, USA) v. 20 statisti-cal software. Observed frequencies of chromosomal translocationsin individual chromosomes were compared with expected valuesas described by Johnson et al. [26].

3. Results

3.1. Air sampling and chemical analysis of EOMs

Information on air sampling and chemical analysis of EOMswas reported in detail in our previous study [18]. Here we pro-vide a brief summary of the results. The PM2.5 samples werecollected in two cities (Prague, Ostrava) in two seasons (win-ter, summer) of 2011. The concentrations of PM2.5 (�g/m3)in the ambient air were as follows: 34.7, 49.2, 9.2 and 16.6,for Prague-winter, Ostrava-winter, Prague-summer and Ostrava-summer, respectively. The concentrations of B[a]P followed asimilar trend. Although for cell treatment we used equal con-centrations of EOMs (1, 10 and 25 �g/ml), the concentrations ofanalyzed PAHs in individual EOMs differed (Table 1). Total PAHconcentration was significantly higher in the Ostrava-winter thanin the Prague-winter EOM; the same was true for EOMs collectedin the summer season. It should be noted that the PAH concen-trations in EOMs used for cell treatment were several orders ofmagnitude lower than the concentrations of B[a]P tested in ourexperiments.

3.2. DSBs and expression of NHEJ proteins

We first examined the ability of B[a]P and EOMs to induceDSBs which are a prerequisite for the formation of chromosomeaberrations. B[a]P at the highest tested dose (25 �M) increased �-H2AX levels, and the doses of 1 �M and 10 �M seemed to haveno effect under the tested conditions (Fig. 1). Most EOMs werepotent inducers of DSBs. Although the effect of the EOM col-lected in the winter season in Prague was similar to B[a]P (2.5-fold

mologous DNA end joining and chromosome aberrations inollutants, Mutat. Res.: Fundam. Mol. Mech. Mutagen. (2014),

increase in �-H2AX levels after treatment with the highest dose),other EOMs induced H2AX phosphorylation ranging from 16- to84-fold above the control level (Fig. 2). Moreover, particularlyfor the Ostrava-winter EOM, we observed a clear dose–response

287

288

289

290

Page 4: Nonhomologous DNA end joining and chromosome aberrations in human embryonic lung fibroblasts treated with environmental pollutants

ARTICLE IN PRESSG ModelMUT 11334 1–11

4 P. Rossner Jr. et al. / Mutation Research xxx (2014) xxx–xxx

Table 1PAH content in pooled extractable organic matter (EOM) from individual samplings.

Analyzed PAHs Concentrations of PAHs in EOMs from individual sampling locations (ng/�g EOM)

Prague-winter* Ostrava-winter Prague-summer* Ostrava-summer

Benz[a]anthracene 0.22 0.41 0.01 0.11Benzo[a]pyrene 0.20 0.25 0.01 0.14Benzo[b]fluoranthene 0.30 0.41 0.03 0.34Benzo[g.h.i]perylene 0.16 0.16 0.02 0.17Benzo[k]fluoranthene 0.13 0.15 0.01 0.13Chrysene 0.34 0.38 0.02 0.16Dibenz[a.h]anthracene 0.02 0.03 0.01 0.02Indeno[1.2.3.cd]pyrene 0.15 0.14 0.02 0.13

Total PAHs 1.51 1.93 0.13 1.20

* gue-suS 8 to 3

ri

s

Fo

291

292

293

294

295

Significant differences (p < 0.001) in PAH concentrations between EOMs from Pratudent’s t-test was used to compare PAHs concentrations in EOMs obtained from 2

elationship between the tested concentrations of the EOM and

Please cite this article in press as: P. Rossner Jr., et al., Nonhohuman embryonic lung fibroblasts treated with environmental phttp://dx.doi.org/10.1016/j.mrfmmm.2014.03.006

nduction of DSBs.The effect of most tested compounds on Ku70 and Ku80 expres-

ion was weak (Figs. 1 and 2). A dose-dependent increase in Ku70

ig. 1. Induction of DSBs and expression of NHEJ proteins in HEL12469 cells treated with

f the respective bands. (B) Protein expression normalized per sample loading (assessed

mmer vs. Ostrava-summer and Prague-winter vs. Ostrava-winter. The two-tailed1 individual filters before pooling.

expression was observed after treatment with the Ostrava-summer

mologous DNA end joining and chromosome aberrations inollutants, Mutat. Res.: Fundam. Mol. Mech. Mutagen. (2014),

EOM (Fig. 2). We further observed inconsistent effects of otherEOMs on the expression of this protein. For the Ku80 protein, anincrease was noted only after treatment with the lowest dose of

B[a]P; a representative result of three independent western blot analyses. (A) Scansby Amido Black staining).

296

297

298

Page 5: Nonhomologous DNA end joining and chromosome aberrations in human embryonic lung fibroblasts treated with environmental pollutants

ARTICLE IN PRESSG ModelMUT 11334 1–11

P. Rossner Jr. et al. / Mutation Research xxx (2014) xxx–xxx 5

F ed wia ample

BBt4oXt

299

300

301

302

303

304

305

306

307

ig. 2. Induction of DSBs and expression of NHEJ proteins in HEL12469 cells treatnalyses. (A) Scans of the respective bands. (B) Protein expression normalized per s

[a]P (Fig. 1). In contrast, XRCC4 expression was induced by both[a]P (Fig. 1) and all tested EOMs (Fig. 2). The effect of B[a]P seemedo be more pronounced; the levels of expression ranged from a

Please cite this article in press as: P. Rossner Jr., et al., Nonhohuman embryonic lung fibroblasts treated with environmental phttp://dx.doi.org/10.1016/j.mrfmmm.2014.03.006

.60- to 11.1-fold increase when compared with the expressionf this protein in the control sample. All the tested EOMs affectedRCC4 expression to a similar extent (2- to 3-fold increase), with

he exception of the highest dose of the Ostrava-winter EOM which

th four different EOMs; a representative result of three independent western blot loading (assessed by Amido Black staining).

caused more than five-fold induction of the protein when comparedwith the control sample.

mologous DNA end joining and chromosome aberrations inollutants, Mutat. Res.: Fundam. Mol. Mech. Mutagen. (2014),

3.3. NHEJ repair activity

The activity of non-homologous DNA end-joining measured asthe ability of nuclear extracts to ligate a pUC18 plasmid substrate

308

309

310

Page 6: Nonhomologous DNA end joining and chromosome aberrations in human embryonic lung fibroblasts treated with environmental pollutants

ARTICLE IN PRESSG ModelMUT 11334 1–11

6 P. Rossner Jr. et al. / Mutation Research xxx (2014) xxx–xxx

F f HEL1t

dw5t

311

312

313

314

315

ig. 3. Activity of NHEJ repair measured as ligation efficiency of nuclear extracts ohree independent experiments.

igested with EcoRI was not significantly affected by treatment

Please cite this article in press as: P. Rossner Jr., et al., Nonhohuman embryonic lung fibroblasts treated with environmental phttp://dx.doi.org/10.1016/j.mrfmmm.2014.03.006

ith the tested samples (Fig. 3). The control NEX converted about0% of the substrate to end-joining products during 2 h incuba-ion. The NEX from the treated samples had a similar or about 10%

2469 cells treated with B[a]P and EOMs. The figure shows mean values ± SD from

lower ligation activity, but the differences were not statistically

mologous DNA end joining and chromosome aberrations inollutants, Mutat. Res.: Fundam. Mol. Mech. Mutagen. (2014),

significant. The NEX obtained from the Ostrava-summer samplesconsistently increased the repair activity to about 60%, but theresults were not significant.

316

317

318

Page 7: Nonhomologous DNA end joining and chromosome aberrations in human embryonic lung fibroblasts treated with environmental pollutants

ARTICLE IN PRESSG ModelMUT 11334 1–11

P. Rossner Jr. et al. / Mutation Research xxx (2014) xxx–xxx 7

00 ce

3

b

319

320

321

Fig. 4. Genomic frequency of translocations per 1

Please cite this article in press as: P. Rossner Jr., et al., Nonhohuman embryonic lung fibroblasts treated with environmental phttp://dx.doi.org/10.1016/j.mrfmmm.2014.03.006

.4. Stable chromosome aberrations

Genomic frequency of translocations per 100 cells calculatedased on the analysis of translocations of six chromosomes (1, 2,

lls. HEL12469 cells treated with B[a]P and EOMs.

mologous DNA end joining and chromosome aberrations inollutants, Mutat. Res.: Fundam. Mol. Mech. Mutagen. (2014),

4, 5, 7 and 17) was elevated above the control level (FG/100 = 0.19)after treatment with all tested samples (Fig. 4). However, we didnot find distinct differences between the compounds. To furthercharacterize the effect of B[a]P and EOMs on DNA damage we

322

323

324

325

Page 8: Nonhomologous DNA end joining and chromosome aberrations in human embryonic lung fibroblasts treated with environmental pollutants

ARTICLE IN PRESSG ModelMUT 11334 1–11

8 P. Rossner Jr. et al. / Mutation Research xxx (2014) xxx–xxx

F ressed( a]P on

cse1o

326

327

328

329

330

331

ig. 5. Frequency of chromosome translocations for individual chromosomes expcharts in the right column). Results shown for the effect of (A) B[a]P + EOMs; (B) B[

alculated the frequency of translocations for individual chromo-

Please cite this article in press as: P. Rossner Jr., et al., Nonhohuman embryonic lung fibroblasts treated with environmental phttp://dx.doi.org/10.1016/j.mrfmmm.2014.03.006

omes for all compounds and for B[a]P and EOMs separately. Wexpressed the results both as the number of translocations per000 cells and the number of translocations/1000 cells/1 giga basef a respective chromosome. The latter value takes into account

per 1000 cells (charts in the left column) and normalized per chromosome sizely; (C) EOMs only. Numbers below horizontal axes indicate chromosome number.

the chromosome size. The results reported in Fig. 5 indicate that

mologous DNA end joining and chromosome aberrations inollutants, Mutat. Res.: Fundam. Mol. Mech. Mutagen. (2014),

chromosome size does not significantly affect the frequency ofchromosome translocations. The pattern of frequencies of chromo-some translocations for B[a]P- and EOM-treated cells was different.EOMs preferentially induced DNA damage to chromosomes 7 and

332

333

334

335

Page 9: Nonhomologous DNA end joining and chromosome aberrations in human embryonic lung fibroblasts treated with environmental pollutants

ING ModelM

on Res

10Fcolrwcfa2o(

4

tomosit

opbnSoapDl[otpu

midowaailtibtcn�DwPaDc

336

337

338

339

340

341

342

343

344

345

346

347

348

349

350

351

352

353

354

355

356

357

358

359

360

361

362

363

364

365

366

367

368

369

370

371

372

373

374

375

376

377

378

379

380

381

382

383

384

385

386

387

388

389

390

391

392

393

394

395

396

397

398

399

400

401

402

403

404

405

406

407

408

409

410

411

412

413

414

415

416

417

418

419

420

421

422

423

424

425

426

427

428

429

430

431

432

433

434

435

436

437

438

439

440

441

442

443

444

445

446

447

448

449

450

451

452

453

454

455

456

457

458

459

ARTICLEUT 11334 1–11

P. Rossner Jr. et al. / Mutati

7, followed by chromosome 4. Frequencies of aberrations around.002/1000 cells/Gb were observed for chromosomes 5, 2 and 1.requencies of translocations caused by B[a]P were similar forhromosomes 7 and 4, followed by chromosome 2; frequenciesf translocations observed for chromosomes 5, 17 and 1 wereow (around 0.001/1000 cells/Gb). To further characterize theseesults we compared the observed frequencies of translocationsith expected values. For B[a]P treatment, the frequency of translo-

ations was 2.3-fold and 1.90-fold higher than the expected valueor chromosome 7 and 4, respectively. For EOM treatment, dam-ge to chromosomes 7 and 17 was similar and reached about-fold higher than expected frequency. For other chromosomes thebserved frequency was equal or lower than the expected valuedata not shown).

. Discussion

In the present study we conducted a series of experiments withhe aim of elucidating mechanisms associated with the treatmentf a human non-tumor lung cell line with carcinogenic PAHs, both aodel compound (B[a]P) and a complex mixture containing c-PAHs

btained from organic extracts from environments with differentources of air pollution. We analyzed a set of parameters related tonduction and repair of DSBs that should affect a final endpoint –he frequency of stable chromosome aberrations.

Double-strand DNA breaks are considered the most lethal formf DNA damage. It is estimated that in dividing cells about 10 DSBser cell per day are formed [27]. In general, DSBs can be repairedy two major mechanisms: homologous recombination (HR) andon-homologous end-joining. However, HR is limited only to the- and G2-phase of the cell cycle and requires sufficient homol-gy between repaired DNA strands. Therefore, NHEJ is regardeds the most important pathway of DSBs repair [28]. The repairrocess is started by binding of the Ku70/Ku80 heterodimer toNA ends. The Ku proteins are among the most abundant cel-

ular proteins and they exhibit a very high affinity to DNA ends28]. The DNA–Ku complex then attracts the catalytic subunitf the DNA-dependent protein kinase (DNA–PKcs) which causeshe phosphorylation of other proteins participating in the repairathway, including XRCC4. This protein stabilizes ligase IV whichltimately causes ligation of DNA strands (for details, see [28]).

Detection of phosphorylated histone H2AX is used as a reliablearker of DSBs [29]. It has been demonstrated that B[a]P treatment

nduces H2AX phosphorylation [30–32], although the intensityepends on the cell type used reflecting the metabolic activityf the tested model [33]. Similarly, extracts of soils contaminatedith PAH mixtures caused dsDNA damage in HepG2 cells [34]. As

DSB is a prerequisite for the formation of stable chromosomalberrations, we first determined whether the tested compoundsnduced phosphorylation of H2AX. We detected increased �-H2AXevels after treatment with all tested compounds, with the excep-ion of lower concentrations (1 and 10 �M) of B[a]P. As these dosesnduced chromosome translocations, this observation should note interpreted as the inability of lower concentrations of B[a]Po cause DSBs. The result rather suggests that at the time of cellollection DSBs had already been repaired. We also observed pro-ounced differences in the ability of individual EOMs to induce-H2AX. The Prague-summer EOM was the most potent inducer ofSBs, while the weakest effect (comparable with B[a]P treatment)as detected after the Prague-winter EOM treatment. Because the

Please cite this article in press as: P. Rossner Jr., et al., Nonhohuman embryonic lung fibroblasts treated with environmental phttp://dx.doi.org/10.1016/j.mrfmmm.2014.03.006

rague-summer EOM contained the lowest concentrations of B[a]Pnd other PAHs, we conclude that the ability of EOMs to induceSBs cannot by assigned exclusively to PAHs, but rather to otheronstituents that were not identified in the chemical analyses of

PRESSearch xxx (2014) xxx–xxx 9

the EOMs (e.g. other PAHs and their derivatives, n-alkanes, sterolsand various industrial contaminants [35]).

To the best of our knowledge, the effects of PAHs on the expres-sion of Ku70/80 and XRCC4 proteins in model cell lines has not yetbeen studied. The studies in cells exposed to X-rays [36] and etopo-side [37] suggested that expression of Ku70 and Ku80 proteins isnot induced by these factors. This may be explained by the fact thatKu proteins are one of the most abundant cellular proteins [28] andtheir levels are sufficient to perform their functions. In line withthis theory is our observation that the effect of B[a]P on Ku70/80expression is weak. However, EOMs, which in general were morepotent inducers of DSBs, exhibited some effect on the induction ofKu70 expression, which was mostly limited to the highest testeddoses of EOMs. Unlike Ku proteins, the expression of XRCC4 wasconsistently induced by treatment with the tested compounds and,particularly for EOMs, its expression levels were in agreement withlevels of phosphorylated H2AX.

Although protein expression analyses give some indication ofthe effect of tested compounds on the model organism, functionalassays should provide the ultimate information on repair and otherprocesses taking place in the cell. The in vitro NHEJ assays havebeen used in various set ups for more than 20 years (reviewedin [38]). The method we applied in our study is based on theprotocol of Perrault et al. [19] which employed pUC18 digestedwith EcoRI incubated with nuclear extracts of the treated cells.We hypothesized that treatment of the cells with the tested com-pounds would result in induction of NHEJ activity. In fact, althoughnumerous authors used the in vitro NHEJ assay for various appli-cations [38,39], none of them focused on inducibility of the NHEJreaction. Unexpectedly, we observed that the tested compoundshad a very small effect on NHEJ activity: the Ostrava summer EOMwas the only one to cause a consistent increase in the repair activ-ity, although the results were not statistically significant. Theremay be several reasons for the lack of a positive effect. First, lin-earized plasmid may not be an optimal substrate to mimic DNAdamage caused by the tested compounds, where ROS probablyplay a major role. It has been reported that naturally occurringoxidatively induced DSBs are far more complex than DNA breaksproduced by restriction enzymes and require further nucleolyticprocessing [39]. Thus, some enzymatic activities required for therepair of DSBs induced by PAHs and other compounds present inEOMs may not be detected by the in vitro NHEJ reaction. The effi-ciency of the NHEJ reaction is further dependent on the structure ofthe DNA break [40], thus we cannot exclude the possibility that theNHEJ reaction using the plasmid linearized by another restrictionenzyme would give different results. Finally, some authors advo-cate using whole-cell rather than nuclear extracts in the NHEJ assay[40] given the fact that some NHEJ proteins (e.g. Ku) are present inthe cytoplasm and transported to nuclei only after DNA damage[41]. However, this report was contradicted by another study [36]in which Ku proteins in human fibroblasts were found to be local-ized exclusively in the nucleus and only a minor fraction of XRCC4was detected in the cytoplasm. Moreover, the distribution of theproteins was not altered by irradiation of the cells [36]. The authorsof this study also measured DSB repair activity, but used a differentapproach (the pulldown assay) which allowed analysis of the activ-ity of the DNA–PK complex. They did not observe any induction ofthe repair activity upon X-ray irradiation of the fibroblasts.

To analyze the effects of the tested compounds on stable chro-mosome translocations we selected 6 chromosomes: 1, 2, 4, 5,7, and 17. The rationale for this selection was the fact that mostof these chromosomes represent the largest chromosomes thus

mologous DNA end joining and chromosome aberrations inollutants, Mutat. Res.: Fundam. Mol. Mech. Mutagen. (2014),

giving us an opportunity to analyze a significant proportion ofthe genome (35.8% of the human genome). Chromosome 17 wasincluded because the p53 gene, which is of particular interest withregard to human cancers, is located on its short arm. The number

460

461

462

463

Page 10: Nonhomologous DNA end joining and chromosome aberrations in human embryonic lung fibroblasts treated with environmental pollutants

ING ModelM

1 ion Re

oni[wsaAhi[lh

wEoloemi[cjwwstpnrmtithttrnoo

5

tcsiRaotbtiD

C

Q2

[

[

[

[

[

[

[

[

[

[

[

[

[

[

464

465

466

467

468

469

470

471

472

473

474

475

476

477

478

479

480

481

482

483

484

485

486

487

488

489

490

491

492

493

494

495

496

497

498

499

500

501

502

503

504

505

506

507

508

509

510

511

512

513

514

515

516

517

518

519

520

521

522

523

524

525

526

527

528

529

530

531

532

533

534

535

536

537

538

539

540

541

542

543

544

545

546

547

548

549

550

551

552

553

554

555

556

557

558

559

560

561

562

563

564

565

566

567

568

569

570

571

572

573

574

575

576

577

578

579

580

581

582

583

584

585

586

587

588

589

590

591

592

593

594

595

596

597

598

ARTICLEUT 11334 1–11

0 P. Rossner Jr. et al. / Mutat

f chromosomes analyzed for stable CAs and the fact that a model,on-tumor cell line was used make our study unique. Most stud-

es have been performed in human peripheral blood lymphocytes42]. The fraction of exchanges detected using our set of probesas 0.59 which is comparable with the results obtained for human

ubjects after painting six chromosomes [42]. Our data indicate thatll tested compounds induced stable chromosome translocations.lthough the ability of EOMs to induce chromosome aberrationsas not been reported so far, B[a]P was shown to cause abnormal-

ties of chromosome 8 and 5 in human mammary epithelial cells16]. Another study found that B[a]P diol epoxide, an active metabo-ite of B[a]P, caused instability of chromosomes 2, 3 and 16 in auman lung cancer cell line [17].

We further concentrated on the identification of chromosomeshich are preferentially damaged after treatment with B[a]P and

OMs. The sensitivity of the chromosomes does not seem to dependn their size: chromosome 7 was most commonly affected regard-ess of the tested compound. It has been reported that aberrationsf chromosomes 1–6 assessed in lymphocytes of human subjectsxposed to PAHs were nonrandomly distributed and that chro-osome 6 was most frequently damaged. The authors observed

ncreased aberration frequencies as chromosome size decreased43]. They argued that PAHs may be preferentially targeting specifichromosomes. Although this observation was made on human sub-ects exposed to air pollutants and thus is not directly comparable

ith our data, it suggests that there may be common principleshich affect damage to chromosomes. The position of chromo-

omes in the nucleus may be one of them. It has been shownhat in human diploid fibroblasts, smaller chromosomes, inde-endently of gene density, are located closer to the center of theucleus, while larger chromosomes are found closer to the nuclearim [44]. Thus, chromosomes located near the nucleus peripheryay be at increased risk of damage. This observation may explain

he higher frequency of chromosome 7 translocations. However,t is still unclear why chromosome 1 was the least damaged of allhe studied chromosomes. Interestingly, chromosome 1 codes theighest number of proteins. Some authors argue that in certain cellypes gene-rich chromosomes were preferentially located towardhe center of the nucleus (e.g. [45]), which again may decrease theisk of DNA damage. Thus, at present a definite conclusion can-ot be drawn. The fact that there are differences in the patternf translocation frequencies between B[a]P and EOMs suggest thatther unidentified mechanisms and EOM components play a role.

. Conclusions

In our study we demonstrated the ability of B[a]P and EOMso induce DSBs, activate the expression of NHEJ proteins and causehromosome translocations. Components of EOMs other than PAHseem to be more important in the induction of DNA damage whichs in agreement with the mechanisms of DSBs formation in whichOS play an important role. The tested compounds preferentiallyffected chromosome 7; however, different mechanisms of actionf B[a]P and EOMs are underscored by the high frequency of damageo chromosome 17 after EOMs treatment. Compounds responsi-le for these differences remain to be identified. Our data indicatehat HEL12469, the non-tumor human lung cell line is suitable forn vitro testing of compounds whose mechanism of action involvesSBs and induction of chromosome aberrations.

Please cite this article in press as: P. Rossner Jr., et al., Nonhohuman embryonic lung fibroblasts treated with environmental phttp://dx.doi.org/10.1016/j.mrfmmm.2014.03.006

onflict of interest statement

The authors declare that there are no conflicts of interest.[

PRESSsearch xxx (2014) xxx–xxx

Acknowledgement

This work was supported by the Grant Agency of the Czech

Republic (P503/11/0084).

References

[1] J. Lewtas, Air pollution combustion emissions: characterization of causativeagents and mechanisms associated with cancer, reproductive, and cardiovas-cular effects, Mutat. Res. 636 (2007) 95–133.

[2] D. Loomis, Y. Grosse, B. Lauby-Secretan, F. El Ghissassi, V. Bouvard, L.Benbrahim-Tallaa, N. Guha, R. Baan, H. Mattock, K. Straif, The carcinogenicityof outdoor air pollution, Lancet Oncol. 14 (2013) 1262–1263.

[3] J. Topinka, L.R. Schwarz, F.J. Wiebel, M. Cerna, T. Wolff, Genotoxicity of urban airpollutants in the Czech Republic. Part II. DNA adduct formation in mammaliancells by extractable organic matter, Mutat. Res. 469 (2000) 83–93.

[4] IARC, IARC Monographs on the Evaluation of the Carcinogenic Risk of Chemicalsto Humans, Chemical Agents and Related Occupations, IARC, Publications, Lyon,France, 2012.

[5] W. Xue, D. Warshawsky, Metabolic activation of polycyclic and heterocyclicaromatic hydrocarbons and DNA damage: a review, Toxicol. Appl. Pharmacol.206 (2005) 73–93.

[6] T.M. Penning, S.T. Ohnishi, T. Ohnishi, R.G. Harvey, Generation of reactive oxy-gen species during the enzymatic oxidation of polycyclic aromatic hydrocarbontrans-dihydrodiols catalyzed by dihydrodiol dehydrogenase, Chem. Res. Toxi-col. 9 (1996) 84–92.

[7] X. Bi, D.M. Slater, H. Ohmori, C. Vaziri, DNA polymerase kappa is specificallyrequired for recovery from the benzo[a]pyrene-dihydrodiol epoxide (BPDE)-induced S-phase checkpoint, J. Biol. Chem. 280 (2005) 22343–22355.

[8] B. van Agen, L.M. Maas, I.H. Zwingmann, F.J. Van Schooten, J.C. Kleinjans,B[a]P–DNA adduct formation and induction of human epithelial lung cell trans-formation, Environ. Mol. Mutagen. 30 (1997) 287–292.

[9] B.H. Lee, S.J. Lee, In vitro chromosome aberration assay using human bronchialepithelial cells, J. Toxicol. Environ. Health A 55 (1998) 325–329.

10] R. Meschini, E. Marotta, A. Berni, S. Filippi, M. Fiore, P. Mancinelli, A.T. Natarajan,F. Palitti, DNA repair deficiency and BPDE-induced chromosomal alterations inCHO cells, Mutat. Res. 637 (2008) 93–100.

11] W. Wei, C. Zhang, A.L. Liu, S.H. Xie, X.M. Chen, W.Q. Lu, PCB126 enhanced thegenotoxicity of BaP in HepG2 cells by modulating metabolic enzyme and DNArepair activities, Toxicol. Lett. 189 (2009) 91–95.

12] R. Meschini, A. Berni, E. Marotta, S. Filippi, M. Fiore, P. Mancinelli, A.T. Natarajan,F. Palitti, DNA repair mechanisms involved in the removal of DBPDE-inducedlesions leading to chromosomal alterations in CHO cells, Cytogenet. GenomeRes. 128 (2010) 124–130.

13] G. Sigounas, J.W. Hairr, C.D. Cooke, J.R. Owen, A.S. Asch, D.A. Weidner, J.E. Wiley,Role of benzo[alpha]pyrene in generation of clustered DNA damage in humanbreast tissue, Free Radic. Biol. Med. 49 (2010) 77–87.

14] J.P. Angeli, G.R. Barcelos, J.M. Serpeloni, F. Barbosa Jr., A. Nersesyan, M.S. Man-tovani, Evaluation of the genotoxic and anti-genotoxic activities of silybin inhuman hepatoma cells (HepG2), Mutagenesis 25 (2010) 223–229.

15] D.C. van Gent, J.H. Hoeijmakers, R. Kanaar, Chromosomal stability and the DNAdouble-stranded break connection, Nat. Rev. Genet. 2 (2001) 196–206.

16] J.A. Caruso, J.J. Reiners Jr., J. Emond, T. Shultz, M.A. Tainsky, M. Alaoui-Jamali,G. Batist, Genetic alteration of chromosome 8 is a common feature of humanmammary epithelial cell lines transformed in vitro with benzo[a]pyrene, Mutat.Res. 473 (2001) 85–99.

17] K. Shinmura, M. Iwaizumi, H. Igarashi, K. Nagura, H. Yamada, M. Suzuki, K.Fukasawa, H. Sugimura, Induction of centrosome amplification and chromo-some instability in p53-deficient lung cancer cells exposed to benzo[a]pyrenediol epoxide (B[a]PDE), J. Pathol. 216 (2008) 365–374.

18] P. Rossner Jr., A. Mrhalkova, K. Uhlirova, M. Spatova, A. Rossnerova, H. Libalova,J. Schmuczerova, A. Milcova, J. Topinka, R.J. Sram, Nucleotide excision repair isnot induced in human embryonic lung fibroblasts treated with environmentalpollutants, PLoS ONE 8 (2013) e69197.

19] R. Perrault, H. Wang, M. Wang, B. Rosidi, G. Iliakis, Backup pathways of NHEJare suppressed by DNA–PK, J. Cell Biochem. 92 (2004) 781–794.

20] N. Machella, M.B. Terry, J. Zipprich, I. Gurvich, Y. Liao, R.T. Senie, D.O. Kennedy,R.M. Santella, Double-strand breaks repair in lymphoblastoid cell lines fromsisters discordant for breast cancer from the New York site of the BCFR, Car-cinogenesis 29 (2008) 1367–1372.

21] J.D. Tucker, W.F. Morgan, A.A. Awa, M. Bauchinger, D. Blakey, M.N. Cornforth,L.G. Littlefield, A.T. Natarajan, C. Shasserre, A proposed system for scoring struc-tural aberrations detected by chromosome painting, Cytogenet. Cell Genet. 68(1995) 211–221.

22] J.N. Lucas, A. Awa, T. Straume, M. Poggensee, Y. Kodama, M. Nakano, K. Ohtaki,H.U. Weier, D. Pinkel, J. Gray, et al., Rapid translocation frequency analysis inhumans decades after exposure to ionizing radiation, Int. J. Radiat. Biol. 62(1992) 53–63.

23] S. Puerto, R. Marcos, M.J. Ramirez, P. Galofre, A. Creus, J. Surralles, Equal induc-

mologous DNA end joining and chromosome aberrations inollutants, Mutat. Res.: Fundam. Mol. Mech. Mutagen. (2014),

tion and persistence of chromosome aberrations involving chromosomes 1, 4and 10 in thyroid cancer patients treated with radioactive iodine, Mutat. Res.469 (2000) 147–158.

24] N.E. Morton, Parameters of the human genome, Proc. Natl. Acad. Sci. U.S.A. 88(1991) 7474–7476.

599

600

601

602

603

Page 11: Nonhomologous DNA end joining and chromosome aberrations in human embryonic lung fibroblasts treated with environmental pollutants

ING ModelM

on Res

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

604

605

606

607

608

609

610

611

612

613

614

615

616

617

618

619

620

621

622

623

624

625

626

627

628

629

630

631

632

633

634

635

636

637

638

639

640

641

642

643

644

645

646

647

648

649

650

651

652

653

654

655

656

657

658

659

660

661

662

663

664

665

666

667

668

669

ARTICLEUT 11334 1–11

P. Rossner Jr. et al. / Mutati

25] J.D. Tucker, J.W. Breneman, J.F. Briner, G.G. Eveleth, R.G. Langlois, D.H. Moore2nd, Persistence of radiation-induced translocations in rat peripheral blooddetermined by chromosome painting, Environ. Mol. Mutagen. 30 (1997)264–272.

26] K.L. Johnson, D.J. Brenner, J. Nath, J.D. Tucker, C.R. Geard, Radiation-inducedbreakpoint misrejoining in human chromosomes: random or non-random? Int.J. Radiat. Biol. 75 (1999) 131–141.

27] M.R. Lieber, J. Gu, H. Lu, N. Shimazaki, A.G. Tsai, Nonhomologous DNA end join-ing (NHEJ) and chromosomal translocations in humans, Subcell. Biochem. 50(2010) 279–296.

28] E. Mladenov, G. Iliakis, Induction and repair of DNA double strand breaks: theincreasing spectrum of non-homologous end joining pathways, Mutat. Res. 711(2011) 61–72.

29] L.J. Mah, A. El-Osta, T.C. Karagiannis, GammaH2AX: a sensitive molecularmarker of DNA damage and repair, Leukemia 24 (2010) 679–686.

30] T. Toyooka, Y. Ibuki, Co-exposure to benzo[a]pyrene and UVA induces phos-phorylation of histone H2AX, FEBS Lett. 579 (2005) 6338–6342.

31] T. Toyooka, G. Ohnuki, Y. Ibuki, Solar-simulated light-exposed benzo[a]pyreneinduces phosphorylation of histone H2AX, Mutat. Res. 650 (2008) 132–139.

32] A. Mattsson, B. Jernstrom, I.A. Cotgreave, E. Bajak, H2AX phosphorylation inA549 cells induced by the bulky and stable DNA adducts of benzo[a]pyreneand dibenzo[a,l]pyrene diol epoxides, Chem. Biol. Interact. 177 (2009) 40–47.

33] M. Audebert, A. Riu, C. Jacques, A. Hillenweck, E.L. Jamin, D. Zalko, J.P. Cravedi,Use of the gammaH2AX assay for assessing the genotoxicity of polycyclic aro-matic hydrocarbons in human cell lines, Toxicol. Lett. 199 (2010) 182–192.

34] M. Niziolek-Kierecka, K. Dreij, S. Lundstedt, U. Stenius, GammaH2AX, pChk1,and Wip1 as potential markers of persistent DNA damage derived fromdibenzo[a,l]pyrene and PAH-containing extracts from contaminated soils,Chem. Res. Toxicol. 25 (2012) 862–872.

35] H. Libalova, K. Uhlirova, J. Klema, M. Machala, R.J. Sram, M. Ciganek, J. Topinka,Global gene expression changes in human embryonic lung fibroblasts induced

Please cite this article in press as: P. Rossner Jr., et al., Nonhohuman embryonic lung fibroblasts treated with environmental phttp://dx.doi.org/10.1016/j.mrfmmm.2014.03.006

by organic extracts from respirable air particles, Part. Fibre Toxicol. 9 (2012) 1.36] U. Kasten-Pisula, S. Vronskaja, J. Overgaard, E. Dikomey, In normal human

fibroblasts variation in DSB repair capacity cannot be ascribed to radiation-induced changes in the localisation, expression or activity of major NHEJproteins, Radiother. Oncol. 86 (2008) 321–328.

[

PRESSearch xxx (2014) xxx–xxx 11

37] I. Schonn, J. Hennesen, D.C. Dartsch, Cellular responses to etoposide: celldeath despite cell cycle arrest and repair of DNA damage, Apoptosis 15 (2010)162–172.

38] P. Pfeiffer, E. Feldmann, A. Odersky, S. Kuhfittig-Kulle, W. Goedecke, Anal-ysis of DNA double-strand break repair by nonhomologous end joining incell-free extracts from mammalian cells, Methods Mol. Biol. 291 (2005)351–371.

39] E. Pastwa, R.I. Somiari, M. Malinowski, S.B. Somiari, T.A. Winters, In vitro non-homologous DNA end joining assays – the 20th anniversary, Int. J. Biochem.Cell Biol. 41 (2009) 1254–1260.

40] T. Poplawski, E. Pastwa, J. Blasiak, Non-homologous DNA end joining in normaland cancer cells and its dependence on break structures, Genet. Mol. Biol. 33(2010) 368–373.

41] M. Nagasawa, F. Watanabe, A. Suwa, K. Yamamoto, K. Tsukada, H. Teraoka,Nuclear translocation of the catalytic component of DNA-dependent proteinkinase upon growth stimulation in normal human T lymphocytes, Cell Struct.Funct. 22 (1997) 585–594.

42] A.J. Sigurdson, M. Ha, M. Hauptmann, P. Bhatti, R.J. Sram, O. Beskid, E.J.Tawn, C.A. Whitehouse, C. Lindholm, M. Nakano, Y. Kodama, N. Nakamura, I.Vorobtsova, U. Oestreicher, G. Stephan, L.C. Yong, M. Bauchinger, E. Schmid,H.W. Chung, F. Darroudi, L. Roy, P. Voisin, J.F. Barquinero, G. Livingston, D.Blakey, I. Hayata, W. Zhang, C. Wang, L.M. Bennett, L.G. Littlefield, A.A. Edwards,R.A. Kleinerman, J.D. Tucker, International study of factors affecting humanchromosome translocations, Mutat. Res. 652 (2008) 112–121.

43] K.A. Bocskay, M.A. Orjuela, D. Tang, X. Liu, D. Warburton, F.P. Perera, Fluo-rescence in situ hybridization is necessary to detect an association betweenchromosome aberrations and polycyclic aromatic hydrocarbon exposure inutero and reveals nonrandom chromosome involvement, Environ. Mol. Muta-gen. 48 (2007) 114–123.

44] A. Bolzer, G. Kreth, I. Solovei, D. Koehler, K. Saracoglu, C. Fauth, S. Muller, R. Eils,C. Cremer, M.R. Speicher, T. Cremer, Three-dimensional maps of all chromo-

mologous DNA end joining and chromosome aberrations inollutants, Mutat. Res.: Fundam. Mol. Mech. Mutagen. (2014),

somes in human male fibroblast nuclei and prometaphase rosettes, PLoS Biol.3 (2005) e157.

45] M. Cremer, J. von Hase, T. Volm, A. Brero, G. Kreth, J. Walter, C. Fischer, I. Solovei,C. Cremer, T. Cremer, Non-random radial higher-order chromatin arrange-ments in nuclei of diploid human cells, Chromosome Res. 9 (2001) 541–567.

670

671

672

673

674