You are on page 1of 15

Environmental Pollution 230 (2017) 163e177

Contents lists available at ScienceDirect

Environmental Pollution
journal homepage: www.elsevier.com/locate/envpol

Genetic and epigenetic alterations in normal and sensitive COPD-


diseased human bronchial epithelial cells repeatedly exposed to air
pollution-derived PM2.5*
B. Leclercq a, b, A. Platel a, S. Antherieu a, L.Y. Alleman b, E.M. Hardy c, E. Perdrix b,
N. Grova c, V. Riffault b, B.M. Appenzeller c, M. Happillon a, F. Nesslany a, P. Coddeville b,
J-M. Lo-Guidice a, G. Garçon a, *
a
Univ. Lille, CHU Lille, Institut Pasteur de Lille, EA4483 IMPECS-IMPact de l’Environnement Chimique sur la Sant
e humaine, France
b
IMT Lille Douai, Univ. Lille, SAGE-D epartement Sciences de l’Atmosph
ere et G
enie de l’Environnement, F-59000 Lille, France
c
Human Biomonitoring Research Unit, Department of Population Health, Luxembourg Institute of Health, L-4354 Esch-sur-Alzette, Luxembourg

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

Article history: Even though clinical, epidemiological and toxicological studies have progressively provided a better
Received 28 February 2017 knowledge of the underlying mechanisms by which air pollution-derived particulate matter (PM) exerts
Received in revised form its harmful health effects, further in vitro studies on relevant cell systems are still needed. Hence, aiming
15 May 2017
of getting closer to the human in vivo conditions, primary human bronchial epithelial cells derived from
Accepted 12 June 2017
normal subjects (NHBE) or sensitive chronic obstructive pulmonary disease (COPD)-diseased patients
(DHBE) were differentiated at the air-liquid interface. Thereafter, they were repeatedly exposed to air
pollution-derived PM2.5 to study the occurrence of some relevant genetic and/or epigenetic endpoints.
Keywords:
Air pollution-derived PM2.5
Concentration-, exposure- and season-dependent increases of OH-B[a]P metabolites in NHBE, and to a
Human bronchial epithelial cells lesser extent, COPD-DHBE cells were reported; however, there were more tetra-OH-B[a]P and 8-OHdG
Healthy and COPD phenotypes DNA adducts in COPD-DHBE cells. No increase in primary DNA strand break nor chromosomal aberra-
Sensitivity tion was observed in repeatedly exposed cells. Telomere length and telomerase activity were modified in
Genetic and epigenetic hallmarks a concentration- and exposure-dependent manner in NHBE and particularly COPD-DHBE cells. There
were a global DNA hypomethylation, a P16 gene promoter hypermethylation, and a decreasing DNA
methyltransferase activity in NHBE and notably COPD-DHBE cells repeatedly exposed. Changes in site-
specific methylation, acetylation, and phosphorylation of histone H3 (i.e., H3K4me3, H3K9ac, H3K27ac,
and H3S10ph) and related enzyme activities occurred in a concentration- and exposure-dependent
manner in all the repeatedly exposed cells. Collectively, these results highlighted the key role played
by genetic and even epigenetic events in NHBE and particularly sensitive COPD-DHBE cells repeatedly
exposed to air pollution-derived PM2.5 and their different responsiveness. While these specific epigenetic
changes have been already described in COPD and even lung cancer phenotypes, our findings supported
that, together with genetic events, these epigenetic events could dramatically contribute to the shift from
healthy to diseased phenotypes following repeated exposure to relatively low doses of air pollution-
derived PM2.5.
© 2017 Elsevier Ltd. All rights reserved.

1. Introduction

In an effort to improve ambient air quality, awareness is now


*
paid to the need to re-evaluate the environmental quality stan-
This paper has been recommended for acceptance by David Carpenter.
dards, aiming not only at lower particulate matter (PM) concen-
* Corresponding author. EA4483 IMPECS-IMPact de l’Environnement Chimique
 humaine, De
sur la Sante partement de Toxicologie, Sante publique et Environne- tration thresholds, but also focusing on specific reduction of PM
ment, Faculte des Sciences Pharmaceutiques et Biologiques de Lille, 3 rue du Pro- toxicity (Cassee et al., 2013). Even though clinical, epidemiological
fesseur Laguesse, BP83 59006 Lille Cedex, France. and toxicological studies have progressively provided a better
E-mail address: guillaume.garcon@univ-lille2.fr (G. Garçon).

http://dx.doi.org/10.1016/j.envpol.2017.06.028
0269-7491/© 2017 Elsevier Ltd. All rights reserved.
164 B. Leclercq et al. / Environmental Pollution 230 (2017) 163e177

knowledge of the underlying mechanisms by which air pollution- imprinting, chromosome stability, and gene transcription (Liu et al.,
derived PM exerts its harmful health effects worldwide, new 2010). Global hypermethylation, as well as hypomethylation of
in vitro studies on relevant cell systems are still needed. With the transposable repetitive elements, has been associated with reduced
aim of getting closer to the human in vivo conditions, we developed chromosomal stability and altered genome function (Herceg and
a primary culture model of normal human bronchial epithelial Vaissiere, 2011). The methylation state is actively maintained by
(NHBE) and sensitive chronic obstructive pulmonary disease the activity of DNA methyltransferases (DNMTs). Histones are
(COPD)-diseased human bronchial epithelial (DHBE) cells, with the essential to the DNA compaction and covalent histone modifica-
cells being grown and, thereafter, differentiated at the air-liquid tions, such as methylation or acetylation, and have shown to play an
interface (ALI). These innovative in vitro models integrate a num- essential role in the regulation of gene expression by modulating
ber of sensitive factors related to pre-existing chronic pulmonary chromatin structure (Liu et al., 2010). These post-translational
inflammation and also allow to obtain a differentiation state as modifications are regulating by enzymes such as histone acetyl-
close as possible to the cell exposure conditions reported in vivo transferases (HATs), deacetylases (HDACs), methyltransferases, and
(Leclercq et al., 2016). Moreover, the use of ALI cultures also offers a demethylases (HDMs) (Ho et al., 2012). H3 and H4 histones have
unique opportunity for the direct deposition of air pollution- long tails which can be covalently modified to allow DNA and nu-
derived PM onto the semi-dry apical cell surfaces, a situation that clear proteins binding changes that lead to chromatin structure and
mimics the deposition of particles onto the lung surface in vivo. gene expression modulations.
Using a more relevant in vitro model such as these primary cells Hence, carrying on from the results recently published by
differentiated at ALI is also critical to the better understanding of Leclercq et al. (2016), the aim of the present study was to better
the toxicological mechanisms related to the repeat exposure to air determine whether genetic and/or epigenetic endpoints will occur
pollution-derived PM (Be rube et al., 2009, 2010; Boublil et al., 2013; in primary culture models of NHBE and sensitive COPD-DHBE cells
Klein et al., 2013; Leclercq et al., 2016; Pezzulo et al., 2011). repeatedly exposed to relatively low doses of air pollution-derived
As argued by our previous works, oxidative stress, inflamma- PM2.5, collected during two different seasons (i.e., autumn-winter,
tion, genotoxicity and cell cycle deregulation were among the main AW; and spring-summer, SS). After checking cytotoxicity, genetic
adverse cell outcomes after single short-term exposure of lung cell and/or epigenetic endpoints were also investigated through the
lines to air pollution-derived PM (Abbas et al., 2009, 2010, 2013, determination of oxidative and benzo[a]pyrene (B[a]P)-related
2016; Billet et al., 2007, 2008; Dagher et al., 2005a,b, 2006, 2007; DNA adducts (i.e., 8-OHdG, and tetra-OH-B[a]P DNA adducts), pri-
Dergham et al., 2012, 2015; Garçon et al., 2006; Saint-Georges et al., mary DNA and chromosomal damages (i.e., in vitro comet assay, and
2008, 2009). All the above-mentioned processes could also play an in vitro micronucleus test, respectively), telomere integrity (i.e., TL
important role in lung diseases, such as asthma, allergy, COPD, and and telomerase activity), DNA methylation (i.e., LINE-1 methyl-
even cancer. However, to date, only few studies have been per- ation, P16 and RB gene promotor methylation, and DNMT activity),
formed using innovative in vitro exposure systems such as NHBE and histone H3 post-translational modifications (i.e., H3K4me3,
and even COPD-DHBE cell models repeatedly exposed to air- H3K9me2, H3K9me3, H3K9ac, H3K27me2, H3K27me3, H3K27ac,
pollution-derived PM (Boublil et al., 2013; Leclercq et al., 2016; H3S10ph; and HAT, HDAC, and HDM activities).
Loxham et al., 2015).
Air pollution-derived PM exposure has been linked to genetic 2. Materials and methods
alterations (for review, see also IARC Publications, 2015). Among
them, two main types of DNA lesions have been described. Firstly, 2.1. Chemicals
the oxidative stress related to xenobiotic-metabolism enzyme ac-
tivity or to metabolites or transition metal redox properties can All the culture reagents we used for the proliferation and dif-
induce DNA strand breaks and oxidized bases (i.e., 8-hydroxy-2’- ferentiation at ALI of NHBE and COPD-DHBE cells were provided by
deoxyguanosine, 8-OHdG). Secondly, highly biologically reactive LONZA (Verviers, Belgium). Chemicals were from Sigma-Aldrich
metabolites resulting from phase I enzyme activity can also link (Saint-Quentin Fallavier, France). CellTiter-Glo® Luminescent Cell
DNA bases yielding to DNA adducts. Besides, telomeres, a complex Viability was from Promega (Charbonnie re-les-Bains, France).
of DNA tandem repeats, protect chromosomes from nucleolytic Vybrant Cytotoxicity Assay Kit and all the molecular biology re-
degradation, end-to-end fusion and breakage-fusion-bridge cycles agents were from Thermo-Scientific (Illkirch, France). Normal and
(Zhang et al., 2013). Telomere shortening can be accelerated by the low-melting point agaroses were from Bio-Rad (Marnes-la-
exposure to several environmental factors known to cause oxida- Coquette, France). QIAamp DNA Mini Kits were from Qiagen
tive stress and chronic inflammation. Recent studies supported that (Courtaboeuf, France). Merck-Millipore (St Quentin-en-Yvelines,
telomere length (TL) modifications are associated with diseases, France) provided CpGenome Direct Prep Bisulfite Modification
such as cancers (Zhang et al., 2013). Telomere integrity is mostly Kit, CpG WIZ® P16 and RB1 Amplification Kits, and TRAPeze® RT
maintained by an enzyme named telomerase which plays a key role Telomerase Detection Kit. Highly Sensitive 8-OHdG Check was from
against chromosome erosion by adding hexameric repeats to the Gentaur France SARL (Paris, France). Global DNA Methylation LINE-
telomeric ends of the chromosome. Therefore, telomeric DNA is 1 Assay, DNMT Assay kit, Histone Purification Mini Kit, Histone H3
dynamic, and TL is considered to be the result of a balance between PTM Multiplex Assay, and HAT, HDAC, and HDM Assay kits were
telomere shortening and lengthening processes (Kordinas et al., from Active Motif (La Hulpe, Belgium).
2016). Evidence for a close link between the genetic and epige-
netic events has recently emerged; in other words, genes that 2.2. Field campaign and PM2.5 sampling description
directly control the epigenome are also frequent targets of acti-
vating or inactivating mutations (Li et al., 2015). Air pollution-derived PM2.5 samples were collected during two
The current field of epigenetics includes a number of mecha- distinct seasons from April 2013 to July 2013 (i.e., AW) and from
nisms, and notably DNA methylation and histone modifications (Liu November 2013 to April 2014 (i.e., SS), respectively, in the center of
et al., 2010). DNA methylation is a covalent modification, heritable Lille (France). The Lille city has a population of 230,000 inhabitants
by somatic cells after cell division, primarily found on CpG di- (density: 6600 inhab/km2) while the metropolitan area represents
nucleotides, and closely involved in regulating many cellular pro- more than 1.1 million inhabitants. The sampling site (50 370 53.200 N;
cesses, including chromatin structure and remodeling, genomic 3 040 31.500 E) is surrounded by medium-sized buildings and located
B. Leclercq et al. / Environmental Pollution 230 (2017) 163e177 165

less than 500 m from a high traffic road (N265) and a large train (Table 2). Overall, the concentrations of chemicals were higher in
P
station. During these two campaigns, the average PM2.5 mass the AW PM2.5 as compared to the SS PM2.5 (i.e., inorganic ele-
P
concentrations, measured at a monitoring station from the regional ments: 207,004 versus 160,246 mg/g, PAHs: 69.4 versus 25.4 mg/g,
P P
air quality monitoring network, were 16.8 ± 10.1 mg/m3 and PCDDs: 13.9 versus 11.5 fg/g, PCDFs: 6.37 versus 4.45 fg/g,
P P
16.6 ± 8.4 mg/m3 for the AW and SS seasons, respectively. The high DL-PCBs: 44.5 versus 24.3 ng/g, and M-PCBs: 233 versus
volume cascade impactor sampler (Tish Environmental Inc., Village 158 ng/g).
of Cleves, OH, USA) we used was localized on the rooftop of a
building (i.e., 15 m above ground) and operated at 35 m3/h on 6
stages from 0.39 mm to 10.2 mm. Only the last 3 stages collecting 2.4. NHBE and COPD-DHBE cell culture and exposure
particles with aerodynamic equivalent diameter between 2.1 mm
and 0.39 mm (i.e., also referred as PM2.5 in this paper) were NHBE and COPD-DHBE cells (LONZA, Verviers, Belgium) were
retrieved. Collected particles were directly weighed, homogenized, established from biopsies isolated from five healthy and five COPD-
and kept at 20  C until sub-sampling for chemical and toxico- diseased donors, respectively. The characteristics of the healthy and
logical studies. COPD-diseased donors and cells are shown as supplemental data
(Table S2). Except for the smoking habits, no significant difference
was reported between the characteristics of the healthy and COPD-
2.3. PM2.5 physical and chemical characterization diseased status. Cells were cultured and differentiated at ALI in
0.4 mm-pore polyester permeable membrane inserts in 6-well
The size distributions of the PM2.5 samples (i.e., AW and SS Transwell® microplate supports (Corning; Sigma-Aldrich), accord-
PM2.5) were studied using a Zetasizer Nano ZSP™ (Malvern In- ing to Leclercq et al. (2016). Permeable membrane inserts we used
struments SARL, Orsay, France). The size distributions, as evaluated were as follow: membrane type: tissue culture treated-polyester;
by PM volume and number, were 1214 nm (95.9%), and 172.3 nm pore diameter: 0.4 mm; nominal pore density: 4  106 pores/cm2;
(73.9%) for the AW season (i.e., AW PM2.5), and 1215 nm (91.5%), and insert diameter: 24 mm; insert membrane growth area: 4.67 cm2;
184.5 nm (92.7%) for the SS season (i.e., SS PM2.5), respectively nominal membrane thickness: 10 mm; membrane-coating: 0.03%
(supplemental data: Table S1). Metal analyses were performed (v/v) type-I collagen from rat tail in PBS). After 21 days of differ-
using inductively coupled plasma-atomic emission spectroscopy entiation, NHBE and COPD-DHBE cells were exposed at ALI for 4 h,
(IRIS Intrepid, Thermo-Scientific) and -mass spectrometry (NeXion one or three times at 24 h intervals, to vehicle solution (i.e., sterile
300x, Perkin Elmer, Villebon-sur-Yvette, France) according to HBSS, 500 mL, used as negative control) or AW and SS PM2.5 (i.e.,
Alleman et al. (2010) and Mbengue et al. (2014). Elemental com- concentrations ranging from 1 to 20 mg PM2.5/cm2, suspended in
positions showed not only some elements usually associated with sterile HBSS, 500 mL), directly deposited by pipetting on the
natural environment (e.g., Al, Ca, Na, Mg, Ti, Sr), but also so-called permeable membrane inserts. Just before cell exposure, air
anthropogenic elements (e.g., Ba, Cr, Cd, Cu, Fe, Ni, Mn, Pb, Zn) pollution-derived AW and SS PM2.5 were suspended at the ex-
(Table 1). Polycyclic aromatic hydrocarbon (PAH) analyses were pected concentrations in sterile HBSS supplemented with ampho-
performed using a high-pressure liquid chromotography Waters tericin B (20 mg/mL) at 2% (v/v), and sonicated for 2  1 min.
2695 Alliance system (Waters SA, Saint-Quentin-en-Yvelines, Theoretical final doses of particles applied to NHBE or COPD-DHBE
France) coupled to an on-line 996-photodiode array and a 2475- cells were respectively 2 or 10 mg/cm2 after a single exposure and 6
fluorimetric detector according to Crenn et al. (2017). High reso- and 30 mg/cm2 after repeated exposures. Twenty-four hours after
lution gas chromatography/high resolution mass spectrometry af- exposure, 1 mL-aliquots of cell-free culture media were collected
ter accelerated solvent extraction were used to quantify and quickly frozen at 80  C. Adherent cells were washed once
polychlorinated dibenzo-p-dioxin and furans (PCDD/Fs) and with 1 mL-aliquots of cold sterile PBS, and quickly frozen at 80  C.
dioxin-like and marker polychlorinated biphenyls (DL/M-PCBs) For the study of primary DNA damage by the alkaline comet assay,
(Billet et al., 2007, 2008). Several congeners of PAHs, PCDD/Fs, and NHBE and COPD-DHBE cells were cultured at ALI and exposed to air
DL- and M-PCBs were detected within the two PM2.5 samples pollution-derived AW and SS PM2.5 for 4 h without any recovery

Table 1
Elemental concentrations of bulk AW and SS PM2.5.

[Element] (mg/g) AW PM2.5 SS PM2.5 [Element] (mg/g) AW PM2.5 SS PM2.5

As 15.8 15.2 Sc 3.0 2.6


Ba 471 290 V 50.5 42.5
Cd 5.7 9.4 Al 15,929 11,633
Ce 23.5 17.3 Ca 45,001 30,417
Co 7.8 6.8 Fe 24,897 18,126
Cs 1.8 2.7 K 8013 6515
Cu 6727 694 Mg 8606 7925
Hg 12.6 18.4 Na 39,148 41,329
La 13.7 13.0 Si 53,681 39,461
Li 14.5 10.6 Sn 160 91
Mn 430 445 Sr 158 119
Mo 212 27.3 Th 2.4 1.7
Ni 61.7 53.9 Ti 1266 1169
Pb 275 300 Tl 1.1 1.8
Pd 0.7 0.5 U 0.9 0.7
Pt 0.2 0.3 Zn 1485 1199
Rb 26.8 23.6 Bi 6.8 6.2
Sb 122 134 Cr 153 119
P
Se 20.9 25.7 elementsa 207,004 160,246
a
S metals was calculated as the sums of elements.
166 B. Leclercq et al. / Environmental Pollution 230 (2017) 163e177

Table 2
Organic compound concentrations of bulk AW and SS PM2.5.

[PAHs]a (mg/g) AW PM2.5 SS PM2.5 [PCDD/Fs]b (fg/g) AW PM2.5 SS PM2.5

FLA 4.71 2.10 2,3,7,8 TCDD <0.03 <0.04


PYR 4.49 1.55 1,2,3,7,8 PeCDD <0.07 <0.13
BcPHE 0.77 0.25 1,2,3,4,7,8 HxCDD 0.11 <0.11
BaA 4.03 1.00 1,2,3,6,7,8 HxCDD 0.38 0.40
CHR 5.94 1.68 1,2,3,7,8,9 HxCDD 0.24 0.33
5MCHR 0.50 0.14 1,2,3,4,6,7,8 HpCDD 4.11 3.25
BeP 6.25 2.34 OCDD 9.12 7.53
P
BbF 10.0 4.62 PCDDs 13.9 11.5
BkF 3.94 1.58
BaP 6.86 1.86 2,3,7,8 TCDF 0.19 0.12
DalP 0.60 0.21 1,2,3,7,8 PeCDF 0.34 0.19
DahA 0.96 0.48 2,3,4,7,8 PeCDF 0.63 0.45
BghiP 7.15 2.99 1,2,3,4,7,8 HxCDF 0.53 0.50
IP 7.64 2.78 1,2,3,6,7,8 HxCDF 0.53 0.41
DaeP 1.81 0.74 2,3,4,6,7,8 HxCDF 0.75 0.64
ANT 1.17 0.05 1,2,3,7,8,9 HxCDF 0.35 0.28
COR 2.60 1.04 1,2,3,4,6,7,8 HpCDF 1.57 1.50
P
PAHs 69.4 25.4 1,2,3,4,7,8,9 HpCDF 0.38 0.37
OCDF 1.06 0.97
P
[DL-PCBs]c (ng/g) AW PM2.5 SS PM2.5 PCDFs 6.37 5.45

PCB 81 <0.53 <0.80


P
PCB 77 2.17 <0.80 PCDD/Fs 20.3 16.9
PCB 123 0.43 <0.59
PCB 118 26.4 16.3
PCB 114 <0. <0.5731

PCB 105 11.17 7.96 [M-PCBs]c (ng/g) AW PM2.5 SS PM2.5

PCB 126 <0.44 <0.51 PCB 28 32 28


PCB 167 <0.32 <0.65 PCB 52 71 54
PCB 156 4.35 <0.65 PCB 101 28 20
PCB 157 <0.35 <0.65 PCB 138 40 21
PCB 169 <0.42 <0.74 PCB 153 40 19
PCB 189 <0.11 <0.25 PCB 180 21 15
P P
DL-PCBs 44.5 24.3 M-PCBs 233 158
a
PAHs; Polyccylic Aromatic hydrocarbons; FLA: Fluoranthene; PYR: Pyrene; BcPHE: Benzo[c]phenanthrene; BaA: Benzo[a]anthracene; CHR: Chrysene; 5MCHR: 5-
Methylchrysene; BeP: Benzo[e]pyrene; BbF: Benzo[b]fluoranthene; BkF: Benzo[k]fluoranthene; BaP: Benzo[a]pyrene; DalP: Dibenzo[a,l]pyrene; DahA: Dibenzo[a,h]anthra-
cene; BghipP Benzo[g,h,i]perylene; IP: Indeno[1,2,3-c,d]pyrene; DaeP: Dibenzo[a,e]pyrene; ANT: Anthanthrene; COR: Coronene. SPAHs was calculated as the sums of PAHs.
b
Tera- (T), Penta- (Pe), Hexa- (Hx), Hepta- (Hp), and Octa (O)-chlorinated Dibenzo-p-Dioxin (PCDDs) and Dibenzo-Furans (PCDFs). SPCDD, SPCDF, and SPCDD/Fs were
calculated as the sums of PCDDs, PCDFs, or PCDDs and PCDFs, respectively.
c
DL-PCB: Dioxin-Like PolyChlorinated Biphenyls; M-PCB: marker PolyChlorinated Biphenyls. SDL-PCB and SM-PCB were calculated as the sums of DL-PCB and M-PCB,
respectively.

time. Cells-exposed to methyl methanesulfonate (MMS; 15 mg/mL) HAT, HDAC, and HDM activities were studied after cell lysis with
served as positive controls. Because of the absolute need of prolif- CHAPS buffer (Sigma-Aldrich). Oxidative DNA adduct 8-OHdG
erative cells for the study of chromosomal aberrations by the concentration was studied using a commercially available enzyme
in vitro micronucleus test, NHBE and COPD-DHBE cells were kept immunoassay (Highly Sensitive 8-OHdG Check, Gentaur France
undifferentiated, grown submerged, and exposed to air pollution- SARL), as published elsewhere (Blasco et al., 2017). The methods
derived AW and SS PM2.5 for 24 h with an additional 5 day recov- developed for the analysis of PAHs and their OH-metabolites in cell
ery period. Cells-exposed to mitomycin C (MMC; 0.25 mg/mL) lysates, on the one hand, and for the analysis of tetra-OH-B[a]P
served as positive controls. isomers released from cell DNA hydrolysis, on the other hand, were
based upon gas chromatography tandem mass spectrometry (GC-
2.5. Toxicological endpoints MS/MS) and have been adapted from Grova et al. (2011, 2014, 2016).
The comet assay was performed under alkaline conditions
Twenty-four hours after the last exposure, the cytotoxicity in (pH > 13) in compliance with previously described protocol (Singh
NHBE and COPD-DHBE cells was evaluated through the determi- et al., 1988; Tice et al., 2000). For the alkaline comet assay, cyto-
nation of intracellular ATP concentration (CellTiter-Glo® Lumines- toxicity was assessed at harvest using the Trypan Blue dye exclu-
cent Cell Viability, Promega, Charbonnie re-les-Bains, France) and sion assay. Results were expressed as percent of cell survival (i.e.,
extracellular glucose-6-phosphate dehydrogenase (G6PD) activity the percent ratio of viable unstained cells to non-viable stained
(Vybrant Cytotoxicity Assay Kit, Thermo Fisher Scientific, Illkirch, cells in exposed versus control groups). Concentrations which led
France). The further study of the toxicological endpoints needed the to more than 70% of viability were then submitted to a genotoxicity
preparation of different cell lysates: (i) B[a]P and its metabolites, study. For the in vitro micronucleus test, cells were submitted to a
OH-B[a]P DNA adducts, and histone H3 post-translational modifi- hypotonic shock and a pre-fixation step, before being twice fixed,
cations were studied after cell lysis with RIPA buffer (Sigma- stained for 10 min with 2% (v/v) Giemsa water solution, and inde-
Aldrich), (ii) 8-OHdG, global DNA methylation, P16 and RB pro- pendently coded and analyzed blind. MN, identified according to
moter methylation, and TL were studied after DNA extraction with recommended criteria, were scored (Fenech et al., 2007). For the
the QIAamp DNA Mini Kit (Qiagen), and (iii) telomerase, DNMT, in vitro micronucleus test, cytotoxicity was evaluated by calculating
B. Leclercq et al. / Environmental Pollution 230 (2017) 163e177 167

the percentage of relative population doubling where population together, these results supported the use of 2 mg PM2.5/cm2 as a
doubling ¼ [log (post-treatment cell number/initial cell number)]/ non-cytotoxic dose and 10 mg PM2.5/cm2 as a low cytotoxic dose of
log(2), as recommended by the OECD test Guideline 487. Concen- the AW and SS PM2.5 to apply for the further study of genetic and
trations which led to a relative population doubling greater than epigenetic endpoints.
45 ± 5% were considered as having low or no cytotoxicity and were
then submitted to genotoxicity study. TL was studied using 3.2. PAH metabolism and DNA adducts in PM2.5-exposed NHBE and
monochrome multiplex quantitative PCR method, as published COPD-DHBE cells
elsewhere (Cawthon, 2009). Telomerase activity was studied using
the TRAPeze® RT Telomerase Detection Kit, (Merck-Millipore, St In order to better determine whether genetic and/or epigenetic
Quentin-en-Yvelines, France). The Global DNA Methylation LINE-1 endpoints will occur in NHBE and sensitive COPD-DHBE cells
Assay and DNMT Assay provided a measure of global DNA repeatedly exposed to relatively low doses of air pollution-derived
methylation shifts and DNMT activity in NHBE and COPD-DHBE PM2.5, we firstly reported concentration- and exposure-dependent
cells, respectively (Active motif, La Hulpe, Belgium). The CpGe- increases of PAHs, B[a]P, PAH metabolites, and tetra-OH-B[a]P
nome Direct Prep Bisulfite Modification Kit provided bisulfite metabolites in NHBE and COPD-DHBE cells (p < 0.01; Fig. 2-A, B, C,
conversion and the CpG WIZ® P16 and RB1 Amplification Kits were and D, respectively). Negligible concentrations of PAHs (i.e.,
used for determining the methylation status of the P16 and RB1 11.7 ± 5.8 and 12.7 ± 3.9 pg/mg protein), B[a]P (i.e., 0.2 ± 0.1 and
gene promoters by methylation-specific PCR, respectively (Merck- 0.1 ± 0.07 pg/mg protein), PAH metabolites (i.e., 0.25 ± 0.10 and
Millipore). Active Motif's Histone Purification Mini Kit was used to 0.10 ± 0.04 pg/mg protein), and tetra-OH-B[a]P (i.e., non-
purify core histones and specific H3 modifications (i.e., H3K4me3, detectable) were observed in control NHBE and COPD-DHBE cells.
H3K9me2, H3K9me3, H3K9ac, H3K27me2, H3K27me3, H3K27ac, Moreover, PAH and B[a]P concentrations did not significantly differ
H3S10ph) were studied using Histone H3 PTM Multiplex Assay between NHBE and COPD-DHBE cells; they were also rather related
(Active Motif). HAT, HDAC, and HDM assays were from Active motif. to the total dose of the PM2.5 we applied onto the cells (Fig. 2-A and
B). In contrast, although they were concentration- and exposure-
2.6. Statistical analysis dependent, the levels of PAH metabolites and tetra-OH-B[a]P me-
tabolites were less important in COPD-DHBE vs NHBE cells. With
Cytotoxicity data were analyzed with the GraphPad Prism6 regards to PAH metabolites, concentrations measured in NHBE cells
Software (v6.01) (GraphPadSoftware Inc., USA). For the other 24 h after one or three exposures to 2 and 10 mg/cm2 AW or SS PM2.5
toxicological endpoints, except in vitro micronucleus test, NHBE were raised by 9.8e76.1-fold vs control NHBE cells, whereas those
and COPD-DHBE cells exposed to air pollution-derived PM2.5 were in COPD-DHBE cells 24 h after one or three exposures to 2 and
compared with those from their respective controls (i.e., sterile 10 mg/cm2 AW or SS PM2.5 were raised by 6.3e35.8-fold vs control
HBSS) using the Mann-Whitney U test, with p value correction for COPD-DHBE cells (Fig. 2-C). In addition, except after one exposure
multiple comparisons. For the in vitro micronucleus test, the sta- to 2 mg/cm2 of AW PM2.5, all the exposure conditions to AW or SS
tistical significance of difference between groups (i.e., each expo- PM2.5 induced lower concentrations of PAH metabolites in COPD-
sure versus controls) was determined using the CHI-squared test. DHBE vs NHBE cells (i.e., from 1.21 to 2.05-fold increases,
Moreover, for all the toxicological endpoints under study and all the p < 0.05). Very similar results, indicating lower concentrations of
exposure conditions we tested, we looked for statistically signifi- tetra-OH-B[a]P metabolite concentrations in COPD-DHBE cells 24 h
cant differences between NHBE and COPD-DHBE cells using the after unique or repeated exposures to 2 and 10 mg/cm2 of AW or SS
Mann-Whitney U test, with p value correction for multiple com- PM2.5 vs NHBE cells, were reported (Fig. 2-D). More accurately,
parisons. The concentration-related increase was also evaluated except after one exposure to 2 mg/cm2 of SS PM2.5, all the exposure
with an ANOVA trend test. Statistical analyses were carried out with conditions to AW or SS PM2.5 produced lower concentrations of
the IBM SPSS Software (v20) (IBM SPSS Software, Inc., USA). Sta- tetra-OH-B[a]P metabolites in COPD-DHBE vs NHBE cells (i.e. from
tistical analyses for comet assay and in vitro micronucleus test were 1.64 to 5.82-fold increases, p < 0.05). Taken together, these results
performed using the statistical StatView® Software (v5.0) (SAS supported higher metabolic capacities of NHBE vs COPD-DHBE cells
Institute Inc., USA). Statistically significant differences were re- towards PAHs. In contrast, there were concentration- and
ported with p values < 0.05. exposure-dependent increases of OH-B[a]P DNA adduct concen-
trations in NHBE and particularly in COPD-DHBE cells (p < 0.01)
3. Results (Fig. 2-E). Indeed, OH-B[a]P DNA adduct concentrations measured
in NHBE cells 24 h after one or three exposures to 2 and 10 mg/cm2
3.1. Cytotoxicity in PM2.5-exposed NHBE and COPD-DHBE cells of AW or SS PM2.5 were raised by 1.61e5.51-fold vs control NHBE
cells, whereas those in COPD-DHBE cells 24 h after one or three
Before studying whether genetic and/or epigenetic endpoints exposures to 2 and 10 mg/cm2 of AW or SS PM2.5 were raised by
will occur in NHBE and sensitive COPD-DHBE cells repeatedly 4.73e11.35-fold vs control COPD-DHBE cells. Moreover, except after
exposed to relatively low doses of air pollution-derived PM2.5, we one exposure to 2 mg/cm2 SS PM2.5, all the exposure conditions to
firstly checked the low cytotoxicity of both the doses we tested. AW or SS PM2.5 generated higher concentrations of OH-B[a]P DNA
Indeed, AW and SS PM2.5 exposure induced dose- and time- adducts in COPD-DHBE vs NHBE cells (i.e. from 1.64 to 5.82-fold
dependent decreases of ATP concentrations in NHBE and COPD- increases, p < 0.05). Hence, PAH- and notably B[a]P-coated onto
DHBE cells (Fig. 1). Significant decreases of ATP concentrations air pollution-derived AW and SS PM2.5 were metabolized by NHBE
were observed after one exposure to AW and SS PM2.5 only at their and to a lesser extent by COPD-DHBE cells, thereby producing
highest concentrations (i.e., 20 mg/cm2, p < 0.05) (Fig. 1-A, C, E, and reactive metabolites at different concentrations. Consequently,
G). After three exposures to AW and SS PM2.5, significant decreases some of them (i.e., tetra-OH-B[a]P) were able to interact with
of ATP concentrations, higher than those above-reported, were genomic DNA to form DNA adducts, which can favor the genetic
observed for the two highest concentrations (i.e., 10 and 20 mg/cm2, instability, in NHBE and particularly COPD-DHBE cells. According to
p < 0.01). Statistically significant G6PD activity was reported only Fig. 2-F, the production of 8-OHdG oxidative DNA adducts after
after three exposures to AW and SS PM2.5 at their highest concen- exposure to AW and SS PM2.5 were concentration- and exposure-
trations (i.e., 20 mg/cm2; p < 0.01) ((Fig. 1-B, D, F, and H). Taken dependent in NHBE, and, more markedly, in COPD-DHBE cells,
168 B. Leclercq et al. / Environmental Pollution 230 (2017) 163e177

Fig. 1. Normal human bronchial epithelial (NHBE; Figure 1-A, B, E, and F) and chronic obstructive pulmonary disease (COPD) diseased human bronchial epithelial (DHBE; Figure 1-C,
D, G, and H) cells were grown and thereafter differentiated for 21 days at air-liquid interface. ATP concentration (Figure 1-A, C, E, G) and glucose-6-phoshate dehydrogenase (G6PD;
Figure 1- B, D, F, H) release in cell-free culture media, were measured 24 h after one or three 4 h exposures with 24 h intervals to increasing concentrations (i.e., ranging from 1 to
20 mg/cm2) of air-pollution-derived PM2.5 collected during autumn-winter (AW) and spring-summer (SS) seasons. Values are depicted as means and standard deviations (i.e., n ¼ 5).
(Mann-Whitney U test, with p value correction for multiple comparisons; versus NHBE or COPD-DHBE controls: *: p < 0.05; **: p < 0.01).
B. Leclercq et al. / Environmental Pollution 230 (2017) 163e177 169

Fig. 2. Polycyclic aromatic hydrocarbons (PAHs, Figure 2-A) benzo[a]pyrene (B[a]P; Figure 2-B), PAH metabolites (Figure 2-C), tetra-OH-B[a]P (Figure 2-D), and the DNA adduct
formation of its OH metabolites (Figure 2-E), and the 8-hydroxy-2’-deoxyguanosine (8-OHdG; Figure 2-F) formation were determined in normal human bronchial epithelial (NHBE)
and chronic obstructive pulmonary disease (COPD) diseased human bronchial epithelial (DHBE) cells 24 h after one or three 4 h exposures with 24 h intervals to 2 and 10 mg/cm2 of
air-pollution-derived PM2.5 collected during autumn-winter (AW) and spring-summer (SS) seasons. Values are depicted as means and standard deviations (i.e., n ¼ 5). (Mann-
Whitney U test, with p value correction for multiple comparisons; versus NHBE controls: **: p < 0.01; versus COPD-DHBE controls: b: p < 0.01).

and showed similar changes as those above-described for the DNA, respectively; 1.36-fold increase, p < 0.01). Moreover, 8-OHdG
production of OH-B[a]P DNA adducts. Statistically significant dif- concentrations measured in NHBE cells 24 h after one or three
ference of 8-OHdG concentrations were reported between control exposures to 2 and 10 mg/cm2 of AW or SS PM2.5 were raised by
NHBE and COPD-DHBE cells (i.e., 5.88 ± 0.70 and 7.97 ± 0.95 pg/mg 1.07e4.60-fold vs control NHBE cells and those measured in COPD-
170 B. Leclercq et al. / Environmental Pollution 230 (2017) 163e177

DHBE cells 24 h after one or three exposures to 2 and 10 mg/cm2 of NHBE (i.e., from 0.96 to 1.38-fold decreases and from 1.05 to 1.50-
AW or SS PM2.5 were raised by 1.17e4.77-fold vs control COPD- fold increases, respectively; p < 0.01) and in COPD-DHBE cells
DHBE cells. All the exposure conditions to AW or SS PM2.5 we (i.e., from 0.98 to 1.48-fold decreases and 1.07e1.57-fold increases,
tested generated higher concentrations of 8-OHdG in COPD-DHBE respectively, p < 0.01). Moreover, only repeated exposure to AW or
vs NHBE cells (i.e. from 1.21 to 1.49-fold increases, p < 0.05). SS PM2.5 induced lower TL and higher telomerase activity in COPD-
Interestingly, NHBE and notably COPD-DHBE cells repeatedly DHBE vs NHBE cells (i.e. from 1.10 to 1.20-fold decreases, p < 0.05,
exposed to air-pollution-derived AW PM2.5 induced high oxidative and from 1.07 to 1.17-fold increases, p < 0.05, respectively). Taken
DNA adduct formation. Collectively, these findings supported that together, these data suggest that despite they did not significantly
PAH-coated onto air pollution-derived AW and SS PM2.5 were induce any DNA stand break nor chromosomal damage, air
differently metabolized by NHBE and COPD-DHBE cells and their pollution-derived AW and SS PM2.5 samples were able to signifi-
reactive metabolites, together with reactive oxygen species (ROS), cantly alter TL, also considered as a genetic alterations, and telo-
were able to dramatically interact with DNA to form DNA adducts, merase activity, in NHBE and particularly sensitive COPD-DHBE
notably in COPD-DHBE cells, which can favor the genetic instability. cells.

3.3. Genetic damages in PM2.5-exposed NHBE and COPD-DHBE cells 3.4. Epigenetic alterations in PM2.5-exposed NHBE and COPD-DHBE
cells
Having demonstrated that DNA adducts occurred in NHBE and
particularly in sensitive COPD-DHBE cells repeatedly exposed to Accordingly, we next compared the sensitivity of NHBE and
relatively low doses of air pollution-derived PM2.5, we next sought COPD-DHBE cells repeatedly exposed to relatively low doses of air
to elucidate whether AW and SS PM2.5 contribute to other genetic pollution-derived PM2.5 to the occurrence of epigenetic alterations.
alterations. We observed that the exposure to AW and SS PM2.5 did We also reported concentration- and exposure-dependent changes
not induce any biologically nor statistically significant increases in of global DNA methylation, P16 gene promoter methylation, and
DNA strand breaks. Primary DNA damage was evaluated using the DNMT activity between control NHBE and COPD-DHBE cells
in vitro comet assay under alkaline conditions. To exclude cyto- (p < 0.05; Fig. 5-A, B, and C). Indeed, global DNA methylation and
toxicity as a confounding factor, the cell viability should be more DNMT activity were lower in control COPD-DHBE vs control NHBE
than 70% for the comet assay. Results of the mean %DNA in the cells (i.e., 0.94 ± 0.03 vs 1 ± 0.04 fold change, 1.07-fold decrease,
comet tail and the corresponding relative cell viability obtained in p < 0.05; 0.94 ± 0.04 vs 1 ± 0.04 fold change, 1.06-fold decrease,
NHBE and COPD-DHBE cells are shown in Fig. 3-A and C. No sig- p < 0.05, respectively), whereas P16 gene promoter methylation
nificant difference was observe between the mean %DNA in the was higher in control COPD-DHBE vs control NHBE cells (i.e.,
comet tail of NHBE and COPD-DHBE cells vs their respective con- 1.17 ± 0.10 vs 1 ± 0.06 fold change, 1.17-fold decrease, p < 0.05).
trols. Moreover, no statistically significant cytotoxic effect up to the Moreover, there were concentration- and exposure-dependent
highest concentration we tested (i.e., 10 mg/cm2) was reported in decreases in the global DNA methylation in NHBE and, more
NHBE nor COPD-DHBE cells exposed to AW or SS PM2.5. Under the significantly, in COPD-DHBE cells (p < 0.01). The concentrations of
experimental conditions above-described, the exposure to AW and 5-mC associated with detectable CpG residues detected in NHBE
SS PM2.5 did not induce any biologically nor statistically significant cells 24 h after one or three exposures to 2 and 10 mg/cm2 of AW or
increases in DNA strand breaks, whatever the concentration (i.e., 2 SS PM2.5 were decreased by 1.06e1.79-fold vs control NHBE cells,
and 10 mg/cm2) or the period of sampling (i.e., SS and AW). Both the whereas those in COPD-DHBE cells 24 h after one or three expo-
PM2.5 samples we tested were, therefore, considered as non- sures to 2 and 10 mg/cm2 of AW or SS PM2.5 were decreased by
genotoxic in NHBE and sensitive COPD-DHBE cells. In addition, 1.17e2.23-fold vs control COPD-DHBE cells (Fig. 5-A). Overall, very
the in vitro micronucleus test was used to assess the induction of similar results, indicating 1.07e1.52-fold decreases of DNMT ac-
chromosomal damages (i.e., clastogenicity and/or aneuploidy) 24 h tivity in NHBE cells 24 h after unique or repeated exposures to 2 and
after NHBE and COPD-DHBE cells exposure to the two PM2.5 sam- 10 mg/cm2 AW or SS PM2.5 vs control NHBE cells, and 1.10e1.62-fold
ples (Fig. 3-B and D). According to the last draft of OECD guideline decreases of DNMT activity in COPD-DHBE cells 24 h after unique or
487 (2014), the genotoxic effects must be associated with low or no repeated exposures to 2 and 10 mg/cm2 AW or SS PM2.5 vs control
cytotoxicity in order to avoid any false positive response. Only COPD-DHBE cells, were reported (Fig. 5-C). In contrast, there were
concentrations leading to more than 45 ± 5% of cell viability were concentration- and exposure-dependent methylation increases of
analyzed. Note that contrary to mean %DNA in the comet tail, mean P16 gene promotor in NHBE and, to a higher extend, in COPD-DHBE
MN were higher in control COPD-DHBE vs control NHBE cells (i.e., cells (i.e., from 0.96 to 2.33-fold increases and from 1.30 to 2.53-fold
7.00 ± 2.83 vs 1.75 ± 0.35, 4.0-fold increase, p < 0.05). Moreover, increases, respectively; p < 0.01) (Fig. 5-B). However, whatever the
NHBE and sensitive COPD-DHBE cell exposure to the relatively low exposure conditions we tested, no significant change was reported
concentrations of AW and SS PM2.5 we tested did not produce any in the methylation status of the RB1 gene promotor in NHBE and
observable chromosomal damage since no biologically nor statis- COPD-DHBE cells (data not shown). Moreover, all the exposure
tically significant effects were observed. In addition, 24 h after their conditions to AW or SS PM2.5 samples generated lower DNA
exposure to the lowest concentration (i.e., 2 mg/cm2) of AW and SS methylation (i.e., from 1.18 to 1.34-fold decreases, p < 0.05) but
PM2.5, mean %DNA in the comet tail and mean MN in COPD-DHBE higher methylation of P16 gene promotor (i.e., from 1.21 to 1.54-fold
cells were respectively lower and higher than in NHBE cells (i.e., increases, p < 0.05) in COPD-DHBE vs NHBE cells. In addition, only
from 5.87 to 8.65-fold decreases and from 3.1 to 3.2-fold increases, the exposure to the highest doses of AW or SS PM2.5 produced lower
p < 0.05). As shown in Fig. 4-A and B, it is also worth noting that TL DNMT activity in COPD-DHBE vs NHBE cells (i.e. from 1.14 to 1.20-
was slightly lower in control COPD-DHBE cells vs control NHBE cells fold decreases, p < 0.05). Statistically significant differences were
(i.e., 0.91 ± 0.04 vs 1 ± 0.06 fold change, 1.10-fold decrease, p < 0.05) reported for site-specific methylation, acetylation, and phosphor-
whereas telomerase activity was somewhat higher in control ylation of histone H3 (i.e., H3K4me3, H3K9ac, H3K27ac, and
COPD-DHBE cells vs control NHBE cells (i.e., 1.07 ± 0.04 vs 1 ± 0.03 H3S10ph), and for HAT and HDAC activities between control NHBE
fold change, 1.05-fold increase, p < 0.05). Moreover, TL and telo- and COPD-DHBE cells (p < 0.05; Table 3 and Fig. 6-A, B, and C).
merase activity were significantly reduced and increased, respec- Indeed, post-translational histone modifications were higher in
tively, in a concentration- and exposure-dependent manner in control COPD-DHBE vs control NHBE cells (i.e., H3K4me3:
B. Leclercq et al. / Environmental Pollution 230 (2017) 163e177 171

Fig. 3. Genotoxicity was determined in normal human bronchial epithelial (NHBE; Figure 3-A and B) and chronic obstructive pulmonary disease (COPD; Figure 3-C and D) after
exposure to 2 and 10 mg/cm2 of air-pollution-derived PM2.5 collected during autumn-winter (AW) and spring-summer (SS) seasons. Primary DNA damage were determined with the
alkaline comet assay immediately after 4 h exposure at the ALI (Figure 3-A and 3-C). For the alkaline comet assay, cytotoxicity was assessed at harvest using the Trypan Blue dye
exclusion assay. Results were expressed as percent of cell survival (i.e., the percent ratio of viable unstained cells to non-viable stained cells in exposed versus control groups).
Concentrations which led to more than 70% of viability were then submitted to a genotoxicity study. Chromosomal aberrations were determined with the micronucleus tests after
24 h exposure in submerged conditions followed by a 5 days recovery period (Figure 3-B and 3-D). For the in vitro micronucleus test, cytotoxicity was evaluated by calculating the
percentage of relative population doubling where population doubling ¼ [log (post-treatment cell number/initial cell number)]/log(2), as recommended by the OECD test Guideline
487. Concentrations which led to a relative population doubling greater than 45 ± 5% were considered as having low or no cytotoxicity and were then submitted to genotoxicity
study. Values are depicted as means and standard deviations (i.e., n ¼ 5). (Mann-Whitney U test, CHI-squared test, and ANOVA trend test with p value correction for multiple
comparisons; versus NHBE controls or COPD-DHBE controls: *: p < 0.05).

1.30 ± 0.14 vs 1 ± 0.10 fold change, 1.30-fold increase, p < 0.05; NHBE and more importantly in COPD-DHBE cells (p < 0.01). Indeed,
H3K9ac: 1.31 ± 0.14 vs 1 ± 0.14 fold change, 1.31-fold increase, H3K4 trimethylation was significantly increased in NHBE (i.e., from
p < 0.05; H3K27ac: 1.40 ± 0.20 vs 1 ± 0.06 fold change, 1.40-fold 1.40 to 1.57-fold increases) and COPD-DHBE (i.e., from 1.60 to 1.84-
increase, p < 0.05; H3S10ph: 1.20 ± 0.10 vs 1 ± 0.06 fold change, fold increases) cells repeatedly exposed to the highest concentra-
1.20-fold increase, p < 0.05). Moreover, significant changes in HDM, tion of AW or SS PM2.5 (p < 0.01; Table 3). As shown in Fig. 6-A,
HAT, and HDAC activities were reported in control COPD-DHBE vs HDM activity was slightly reduced in NHBE and COPD-DHBE cells
control NHBE cells (i.e., HDM: 0.90 ± 0.07 vs 1.03 ± 0.07 fold change, only after their repeated exposure to the highest concentration of
1.14-fold decrease, p < 0.05; HAT: 1.10 ± 0.03 vs 1.00 ± 0.06 fold the two PM2.5 samples (p < 0.05). Besides, as shown in Table 3,
change, 1.10-fold increase, p < 0.05; HDAC: 0.93 ± 0.03 vs there was a significant acetylation of H3K9 and H3K27 in NHBE (i.e.,
1.00 ± 0.04 fold change, 1.08-fold decrease, p < 0.05). There were from 1.34 to 1.91-fold increases, and from 1.30 to 1.85-fold in-
concentration- and exposure-dependent changes in histone post- creases, respectively) and COPD-DHBE (i.e., from 1.44 to 2.24-fold
translational modifications and related enzymes activities in increases, and from 1.50 to 1.96-fold increases, respectively) cells
172 B. Leclercq et al. / Environmental Pollution 230 (2017) 163e177

H3K9ac: 1.27e1.48-fold increase, p < 0.05; H3K27ac: 1.43e1.83-


fold increase, p < 0.05; H3S10ph: 1.11e1.43-fold increase,
p < 0.05), lower HDM and HDAC activities (i.e., HDM: 1.09e1.14-fold
decrease, p < 0.05; HDAC: 1.08e1.12-fold decrease, p < 0.05), and
higher HAT activity (i.e., 1.07e1.23-fold increase, p < 0.05) in COPD-
DHBE vs NHBE cells. Collectively, these results also closely sup-
ported the higher sensitivity of the diseased cell model to the
epigenetic alterations-induced by AW and SS PM2.5, thereby
underlining the real need to use sensitive diseased models to better
consider the sensitive individuals in terms of air pollution-derived
PM2.5-induced harmful health effects.

4. Discussion

Despite the high number of in vitro approaches reported in


respiratory toxicology literature, researchers are still far from
having a complete detailed mechanistic explanation of the causal
relation between PM and health effects (Longhin et al., 2016).
However, the use of more relevant in vitro models, such as primary
HBE cells, would be appropriate to better address this issue. Using
cells isolated from diseased-subjects such as COPD is important to
include sensitive diseased phenotypes. Hence, aiming to get closer
to in vivo exposure conditions, NHBE and sensitive COPD-DHBE cell
models repeatedly exposed to relatively low doses of air pollution-
derived PM2.5-0.39 (i.e., also referred as PM2.5 in this paper) were
used to better evaluate the harmful occurrence of genetic and
epigenetic alterations.
According to their chemical characterization (i.e., see also sup-
plemental data), AW and SS PM2.5 samples we collected contained
some PAHs, which could, directly or indirectly, through their
metabolic activation, participate to the toxicity in NHBE cells
(Boublil et al., 2013; Dergham et al., 2012, 2015; Longhin et al., 2013,
2016; Leclercq et al., 2016; Zhou et al., 2014. Concentration- and
exposure-dependent increases of the concentrations of PAHs, PAH
metabolites, B[a]P, and, in particular, tetra-OH-B[a]P metabolites in
NHBE cells, and to a lesser extent in COPD-DHBE cells, were re-
ported. Interestingly, in repeatedly exposed COPD-DHBE cells,
despite the lower gene expression of PAH-related xenobiotic
metabolizing enzymes and the subsequent lower formation of PAH
metabolites, there was a more pronounced formation of OH-B[a]P
DNA adducts (Leclercq et al., 2016). Indeed, as a result of the
Fig. 4. Telomere length (TL; Figure 4-A) and telomerase activity (Figure 4-B) were
commercial origin of the COPD-DHBE cells, leading to less choice
evaluated in normal human bronchial epithelial (NHBE) and chronic obstructive pul-
monary disease (COPD) diseased human bronchial epithelial (DHBE) cells 24 h after
among the diseased cell donors, each of them had smoking habits
one or three 4 h exposures with 24 h intervals to 2 and 10 mg/cm2 of air-pollution- and their cells seemed also to have a moderate capacity to convert
derived PM2.5 collected during autumn-winter (AW) and spring-summer (SS) sea- PAHs into metabolites, such as OH-B[a]P, probably related to their
sons. Values are depicted as means and standard deviations (i.e., n ¼ 5). (Mann- inflammatory state. However, they appeared to be more sensitive to
Whitney U test, with p value correction for multiple comparisons; versus NHBE con-
the formation and/or the sustainability of OH-B[a]P adducts,
trols: *: p < 0.05 and **: p < 0.01; versus COPD-DHBE controls: a: p < 0.05 and b:
p < 0.01). probably because both their smoking habits and diseased state
could affect their defense capacities and, in particular, their DNA
damage repair capacities (Pauk et al., 2013; Rode et al., 2013). In a
repeatedly exposed to the highest concentration of AW and SS similar manner, 8-OHdG levels increased in a concentration- and
PM2.5 (p < 0.01). Accordingly, HAT and HDAC activities were exposure-dependent manner in NHBE cells, and notably sensitive
increased and decreased, respectively, in a concentration- and COPD-DHBE cells, thereby supporting the highest sensitivity of the
exposure-dependent manner in NHBE (i.e., from 1.04 to 1.36-fold COPD-diseased model. Indeed, in addition to the metal-catalyzed
increases and from 1.03 to 1.43-fold increases, respectively; ROS generation, the metabolic activation of PAHs in AW and SS
p < 0.01) and in particular in COPD-DHBE cells (i.e., from 1.13 to PM2.5-exposed NHBE and COPD-DHBE cells would closely
1.54-fold increases and from 1.09 to 1.57-fold increases, respec- contribute to ROS generation (Boublil et al., 2013; Gualtieri et al.,
tively; p < 0.01) (Fig. 6-B and C). Statistically significant decreases in 2010, 2011). Accordingly, the AW PM2.5 sample, and, to a lesser
HAT and HDAC activity were only reported in NHBE and COPD- extent, the SS PM2.5 sample, contained some metals (e.g., Al, As, Cd,
DHBE cells after one or three exposures to the highest concentra- Cr, Cu, Fe, Hg, Mn, Ni, Pb, and Zn) and organic chemicals (e.g. PAHs,
tion we tested (i.e., 10 mg/cm2). Changes in H3S10 phosphorylation PCDD/Fs) generally associated with redox reactions, which could
were almost similar as those above-described for H3K4 trimethy- therefore damage macromolecules such as DNA (i.e., 8-OHdG)
lation (Table 3). Importantly, all the exposure conditions to AW or (Dergham et al., 2012, 2015; Garçon et al., 2006; Gualtieri et al.,
SS PM2.5 we tested produced higher post-translational histone 2010, 2011; Leclercq et al., 2016, 2017). Moreover, the highest
modifications (i.e., H3K4me3: 1.21e1.52-fold increase, p < 0.05; sensitivity of COPD-diseased cells towards the PM2.5-induced
B. Leclercq et al. / Environmental Pollution 230 (2017) 163e177 173

formation of oxidative DNA adducts was factually consistent with


the following statement that COPD disease is generally associated
with a rising oxidative stress, rendering these cells more sensitive
to the harmful effects of inhaled pollutants. Taken together, our
results supported that both OH-B[a]P and 8-OHdG DNA adducts
caused by the repeated exposure to AW and SS PM2.5 were more
marked in COPD-DHBE versus NHBE cells, thereby clearly con-
firming their higher sensitivity to external injuries and their higher
genetic instability (Caramori et al., 2014).
Despite the significant occurrence of alkyl and oxidative DNA
adducts in NHBE and particularly COPD-DHBE cells repeatedly
exposed, no biologically nor significant increases in primary DNA
strand breaks nor chromosomal aberrations were reported. How-
ever, there is now overwhelming evidence that ambient air con-
tains hundreds of genotoxic compounds (IARC Publications, 2015).
In almost all of the available studies, mutagenic and/or genotoxic
potential was determined from extracted organic matter (EOM).
However, as reported by Andre  et al. (2011), extrapolations to po-
tential effects on human health are limited for many reasons,
including the too important chemical concentrations in EOM, the
bioavailability differences between EOM and the whole complex
mixture-coated onto PM, and the interactions between surface and
core chemicals. To date, only a few works test directly the geno-
toxicity of air pollution-derived PM. Among them, Lepers et al.
(2014), using BEAS-2B cell line once exposed to several native
PM2.5 samples, supported, despite the lack of a clear concentration-
dependent effect that some PM2.5 samples would be genotoxic. This
apparent discrepancy might probably rely on the use of various
exposure strategies (e.g., ALI vs submerged exposed cells, acute vs
repeated exposure) on the one hand, and of primary cultures vs
transformed cells, on the other hand. It reinforced the importance
of using innovative in vitro exposure systems and repeated expo-
sure protocols, which could also help to better reflect the real
exposure conditions. Nevertheless, in the present work, TL and
telomerase activity were respectively reduced and increased in a
concentration- and exposure-dependent manner in NHBE and
particularly COPD-DHBE cells. Telomere shortening has been
shown to contribute to a persistent DNA damage response and the
irreversible loss of division potential of somatic cells, and thereby to
an increasing risk of chronic diseases (Hewitt et al., 2012).
Accordingly, telomere shortening is frequently associated with
decreased lung functions and a rising risk of developing COPD
pathology or cancers, and telomerase activity is regularly raised in
COPD and in lung cancer patients (Ferna ndez-Marcelo, 2015;
Bozkus et al., 2016; Rode et al., 2013). Moreover, oxidative stress
and chronic inflammation, which can also be mediated by envi-
ronmental exposures including traffic-related air pollution (e.g.,
PM, black carbon, PAHs), are considered to be major mechanisms
that influence TL and telomerase activity, two possible biological
mechanisms leading to diseases such as COPD or cancer (Zhang
et al., 2013). Taken together, these results clearly supported that
despite no significant DNA stand break nor chromosomal damage
were observed, other genetic alterations than those above-reported
(i.e., OH-B[a]P and 8-OHdG DNA adducts) will occur (i.e., TL
shortening and telomerase activity increase) in NHBE and partic-
ularly sensitive COPD-DHBE cells repeatedly exposed to air
pollution-derived AW and SS PM2.5.
Fig. 5. Global DNA methylation (Figure 5-A), P16 promoter methylation (Figure 5-B),
In addition to DNA damage, which could induce genetic ab-
and DNA methyltransferase (DNMT; Figure 5-C) activity were evaluated in normal
human bronchial epithelial (NHBE) and chronic obstructive pulmonary disease (COPD)
normalities, recent evidence supported that air pollution-derived
diseased human bronchial epithelial (DHBE) cells 24 h after one or three 4 h exposures PM could alter the epigenome, leading to dramatically dysregula-
with 24 h intervals to 2 and 10 mg/cm2 of air-pollution-derived PM2.5 collected during tion of gene expression (Li et al., 2015). Epigenetic alterations affect
autumn-winter (AW) and spring-summer (SS) seasons. Values are depicted as means the gene expression pattern, DNA accessibility and chromatin
and standard deviations (i.e., n ¼ 5). (Mann-Whitney U test, with p value correction for
compaction, the integrity and function of the genome, and the
multiple comparisons; versus NHBE controls: *: p < 0.05 and **: p < 0.01; versus
COPD-DHBE controls: b: p < 0.01). maintenance of the nuclear organization in a manner that
174 B. Leclercq et al. / Environmental Pollution 230 (2017) 163e177

Table 3
Histone modifications in NHBE and COPD-DHBE cells exposed to air pollution-derived PM2.5.

Season Exposure [PM] Cell Histone modification (Fold change vs control)


(n) (mg/cm2)
H3K4me3 H3K9ac H3K27ac H3S10ph

NHBE 1.00 ± 0.10 1.00 ± 0.14 1.00 ± 0.06 1.00 ± 0.06


COPD-DHBE 1.30 ± 0.14 ** 1.31 ± 0.14 ** 1.40 ± 0.20 ** 1.20 ± 0.10 **
AW 1 2 NHBE 1.07 ± 0.13 1.10 ± 0.17 1.08 ± 0.08 1.06 ± 0.08
COPD-DHBE 1.34 ± 0.14 ** 1.50 ± 0.25 ** 1.68 ± 0.20 ** 1.23 ± 0.09 **
10 NHBE 1.18 ± 0.16 1.34 ± 0.20 * 1.30 ± 0.18 * 1.11 ± 0.11
COPD-DHBE 1.44 ± 0.20 ** 1.98 ± 0.18 **, a 2.38 ± 0.18 *, a 1.34 ± 0.18 *
3 2 NHBE 1.18 ± 0.16 1.24 ± 0.18 1.25 ± 0.25 1.14 ± 0.08
COPD-DHBE 1.49 ± 0.20 * 1.57 ± 0.27 * 1.88 ± 0.18 * 1.37 ± 0.13 *
10 NHBE 1.57 ± 0.24 ** 1.91 ± 0.30 ** 1.85 ± 0.25 ** 1.36 ± 0.16 **
COPD-DHBE 2.39 ± 0.37 **, b 2.94 ± 0.47 **, b 2.74 ± 0.40 **, b 1.94 ± 0.21 **, b
SS 1 2 NHBE 1.05 ± 0.09 1.08 ± 0.12 1.10 ± 0.09 1.04 ± 0.05
COPD-DHBE 1.27 ± 0.10 ** 1.37 ± 0.23 ** 1.66 ± 0.20 ** 1.20 ± 0.10 **
10 NHBE 1.04 ± 0.07 1.28 ± 0.14 * 1.26 ± 0.16 * 1.08 ± 0.09
COPD-DHBE 1.30 ± 0.12 ** 1.88 ± 0.24 **, a 2.10 ± 0.30 **, a 1.28 ± 0.12 **
3 2 NHBE 1.20 ± 0.14 1.20 ± 0.18 1.22 ± 0.16 1.14 ± 0.10
COPD-DHBE 1.46 ± 0.18 ** 1.58 ± 0.28 ** 1.75 ± 0.25 ** 1.26 ± 0.13 **
10 NHBE 1.40 ± 0.20 ** 1.84 ± 0.24 ** 1.68 ± 0.20 ** 1.30 ± 0.16 **
COPD-DHBE 2.06 ± 0.21 **, b 2.60 ± 0.34 **, b 2.48 ± 0.24 **, b 1.80 ± 0.15 **, b

Histone post-translational modification in normal human bronchial epithelial (NHBE) and chronic obstructive pulmonary disease (COPD) diseased human bronchial epithelial
(DHBE) cells 24 h after one or three 4 h exposures with 24 h intervals to 2 and 10 mg/cm2 of air-pollution-derived PM2.5 collected during autumn-winter (AW) and spring-
summer (SS) seasons. Values are depicted as means and standard deviations (i.e., n ¼ 6). (Mann-Whitney U test, with p value correction for multiple comparisons; versus NHBE
controls: *: p < 0.05 and **: p < 0.01; versus COPD-DHBE controls: a: p < 0.05 and b: p < 0.01).

determines the healthy or diseased cell state. Therefore, a better mechanisms involved in COPD biology, and generally related to CS
knowledge of the epigenetic changes induced by PM exposure may exposure. Indeed, a number of very rewarding studies have looked
provide an important tool for analyzing the association between at the effects of CS exposure on histone post-translational modifi-
PM exposure and the induction and/or exacerbation of chronic cations. Accordingly, Sundar et al. (2010) found that CS-induced
inflammatory lung diseases. A global DNA hypomethylation, a P16 post-translational modification of histone H3 (e.g., H3K4me3 and
gene promoter hypermethylation, and a DNMT activity decrease H3K27ac) in human lung epithelial H292 cells. Acetylation of lysine
occurred in control COPD-DHBE vs control NHBE cells, which is also residues on H3 has also been suggested to have a direct relationship
consistent with the COPD diseased state (Liu et al., 2010; Zong et al., with the transcription of genes that promote inflammation (Chen
2015). Accordingly, Wang et al. (2012) demonstrated that changes et al., 2015; Vucic et al., 2014).Moreover, in agreement with our
in genome-wide methylation can be found in COPD, and dynamic, above-described results, total HDAC activity is generally decreased
site-specific methylation changes appear in response to smoking in peripheral lung tissue samples, in alveolar macrophages, and in
and may contribute to the extended risks associated with cigarette bronchial-biopsy from patients with COPD; this decrease is corre-
smoking that persist after cessation. The progressive genomic lated with both the disease severity and the intensity of the in-
hypomethylation and locoregional DNA hypermethylation induced flammatory response (To et al., 2016). Lastly, in compliance to our
by cigarette smoke (CS) coincided with an important clonogenicity, results reporting concentration- and exposure-dependent changes
supporting thereby that CS induces ‘COPD and/or cancer-associ- of site-specific phosphorylation of histone H3 (i.e., H3S10ph) in
ated’ epigenomic alterations. Moreover, acute exposure to air NHBE and particularly sensitive COPD-DHBE cells repeatedly
pollution-derived PM resulted in P16 gene promoter hyper- exposed to AW and SS PM2.5, Ibuki et al. (2014) reported that CS
methylation, which can favor the genomic instability and the cell exposure phosphorylated H3S10. The results of some other inter-
cycle progression (Baccarelli and Vollati, 2009; Soberanes et al., esting studies, focusing their attention on the ability of air
2012; Tarantini et al., 2009). Collectively, our results supported pollution-derived PM and some of its chemical constituents to
global and specific methylation changes in NHBE and particularly post-translationally modify histones, also closely supported the
sensitive COPD-DHBE cells repeatedly exposed to AW and SS PM2.5, original findings we above-reported. Cantone et al. (2014) indicated
which could contribute to the induction and/or exacerbation of that metal rich air pollution-derived PM2.5 modify the levels of
COPD and/or cancer phenotypes. Moreover, although the exact role specific extracellular histone modifications, notably H3K4 trime-
of histone modifications in chromatin remodeling of chronic lung thylation and H3K9 acetylation. Furthermore, Wang et al. (2012)
diseases still remains unclear, histone acetylation/deacetylation or reported that air pollution-derived PM contributed to dysregu-
methylation/demethylation could contribute to air pollutant- lated H3 acetylation (i.e., reduced by Ni and Cr but increased by
induced lung inflammation (Li et al., 2015). Indeed, concentra- PAHs). Recent evidences, clearly strengthening that altered activ-
tion- and exposure-dependent changes of site-specific methyl- ities of HDM, HAT, and HDAC may contribute to the epigenetic
ation, acetylation, and phosphorylation of histone H3 (i.e., changes induced by PM or PM-related chemicals, also confirmed
H3K4me3, H3K9ac, H3K27ac, and H3S10ph), and of HDM, HAT and our present findings (Li et al., 2015). Lastly, in keeping with the
HDAC activities were reported in NHBE and particularly sensitive present results, Ding et al. (2017) studying the early effects, cu-
COPD-DHBE cells repeatedly exposed to AW and SS PM2.5. However, mulative effects, dose-effect responses, and interactions of the
to our current knowledge, only very few data are already available traffic-related air pollution in affecting DNA methylation and H3K9
about the post-translational modifications of histones induced by acetylation, showed that the early effect of traffic-related air
air pollutants in general, and, in particular, air pollution-derived pollution exposure mainly included global DNA hypomethylation
PM2.5, in cell or animal models. It is nonetheless important to and changes in the inflammation-related gene, while prolonged
note that our results were in accordance with those indicating that exposure could effectively affect the methylation of specific genes,
H3K4 methylation and H3K27 hyperacetylation are important and that high level of traffic-related air pollution exposure had
B. Leclercq et al. / Environmental Pollution 230 (2017) 163e177 175

significant H3K9 acetylation. Overall, our results highlighted the


crucial occurrence of specific epigenetic changes in DNA methyl-
ation and histone post-translational modification in NHBE cells and
notably sensitive COPD-DHBE cells repeatedly exposed to air
pollution-derived AW and SS PM2.5. While these specific epigenetic
changes have been already described in COPD and even lung cancer
phenotypes, our findings supported that, together with genetic
events, these epigenetic events could dramatically contribute to the
shift from healthy to diseased phenotypes following repeated
exposure to relatively low doses of air pollution-derived PM2.5. In
other words, taken together, our results supported that exposure of
NHBE cells to air pollution-derived PM2.5 significantly induced
some genetic and epigenetic modifications generally observed
within sensitive COPD-DHBE cells, apart from any exposure,
thereby suggesting the possible PM2.5-induced shift of the healthy
phenotype (i.e., NHBE cells) to the COPD phenotype (i.e., COPD-
DHBE cells). The use of innovative in vitro exposure systems such
as NHBE and sensitive COPD-DHBE cells could therefore be
consider as a very useful and powerful promising tool in the field of
the respiratory toxicology. However, further studies based on NHBE
or sensitive COPD-DHBE cell models will be needed to better
evaluate the consequences of longer-term effects produced by air
pollution-derived PM with respect to relevant genetic and mainly
epigenetic alterations, and their possible interplay.

5. Conclusions

To conclude, with the aim of being closer to environmental


human exposure conditions, we used an original experimental
strategy allowing to repeatedly expose NHBE and sensible COPD-
DHBE cells to relatively low doses of air pollution-derived PM2.5,
also directly collected from an urban surrounding. By studying
some relevant genetic and epigenetic endpoints, we highlighted
both the key role played by all these underlying mechanisms and
the difference between the two cell models in their responsiveness
to two different air pollution-derived PM2.5. Indeed, COPD-DHBE
cells were more sensitive to pollution-derived PM2.5 and, there-
fore, showed an exacerbation of some of the underlying mecha-
nisms normally induced, thereby underlining the real need to use
sensitive diseased models to better consider the sensitive in-
dividuals. Moreover, our work allowed us to identify some epige-
netic modifications involved in lung diseases arguably linked to
environmental exposures (i.e., COPD, cancer); however, their
possible use as biomarkers for the early detection of PM-induced
harmful effects needs further investigations to confirm their rele-
vance and to identify new ones, which can reflect the whole past
exposures and predict the future diseases risks.

Formatting and funding sources

This work benefited from grants from the ITMO Cancer (i.e., Plan
Cancer, 2009e2013, Contract n ENV201210), the University of Lille,
gion Nord-Pas
and IMT Lille Douai. BL's PhD is supported by the Re
de Calais and IMT Lille Douai (Contract n 13005131).

Conflict of interest disclosure

Fig. 6. Histone demethylase (HDM; Figure 6-A), acetyl transferase (HAT; Figure 6-B) All authors of this manuscript disclosed any conflict of interest:
and histone deacetylase (HDAC; Figure 6- C) activities were determined in normal they did not have any financial and personal relationships with
human bronchial epithelial (NHBE) and chronic obstructive pulmonary disease (COPD) other people or organizations that could inappropriately influence
diseased human bronchial epithelial (DHBE) cells 24 h after one or three 4 h exposures
with 24 h intervals to 2 and 10 mg/cm2 of air-pollution-derived PM2.5 collected during
their work.
autumn-winter (AW) and spring-summer (SS) seasons. Values are depicted as means
and standard deviations (i.e., n ¼ 5). (Mann-Whitney U test, with p value correction for Acknowledgments
multiple comparisons; versus NHBE controls: *: p < 0.05 and **: p < 0.01; versus
COPD-DHBE controls: a: p < 0.05 and b: p < 0.01).
The authors would also like to convey their sincere thanks to the
176 B. Leclercq et al. / Environmental Pollution 230 (2017) 163e177

Lille University of Health and Law which gave us the opportunity to 513e526.
Crenn, V., Fronval, I., Petitprez, D., Riffault, V., 2017. Fine particles sampled at an
easily access to the sampling site and skillfully helped us in the
urban background site and an industrialized coastal site in Northern France -
setting up of the sampling materials needed to collect air pollution- Part 1: seasonal variations and chemical characterization. Sci. Total Environ.
derived fine particles. They gratefully acknowledge B. MALET for his 578, 203e218.
helpful technical support in the sampling of air pollution-derived Dagher, Z., Garçon, G., Billet, S., Ledoux, F., Courcot, D., Aboukais, A., Puskaric, E.,
Shirali, P., 2005a. Role of Nuclear Factor-kappa B Activation in the adverse ef-
particles and in the analyses of metals. They also sincerely thank fects induced by air pollution particulate matter (PM2.5) in human epithelial
Dr A. HACHIMI and Dr P-E. LAFARGUE, from MicroPolluants Tech- lung cells (L132) in culture. J. Appl. Toxicol. 27, 284e290.
nologie S.A. (Saint Julien Les Metz, France), for their highly skillful Dagher, Z., Garçon, G., Gosset, P., Ledoux, F., Surpateanu, G., Courcot, D., Aboukais, A.,
Puskaric, E., Shirali, P., 2005b. Pro-inflammatory effects of Dunkerque city air
assistance in the analyses of polychlorinated dibenzo-p-dioxin and pollution particulate matter 2.5 in human epithelial lung cells (L132) in culture.
furans, and dioxin-like and marker polychlorinated biphenyls. J. Appl. Toxicol. 25, 166e175.
Dagher, Z., Garçon, G., Billet, S., Gosset, P., Ledoux, F., Courcot, D., Aboukais, A.,
Puskaric, E., Shirali, P., 2006. Activation of different pathways of apoptosis by
Appendix A. Supplementary data Dunkerque city air pollution particulate matter (PM2;5) in human epithelial
lung cells (L132) in culture. Toxicology 225, 12e24.
Dagher, Z., Garçon, G., Billet, S., Ledoux, F., Courcot, D., Aboukais, A., Puskaric, E.,
Supplementary data related to this article can be found at http:// Shirali, P., 2007. Role of Nuclear Factor-kappa B Activation in the adverse effects
dx.doi.org/10.1016/j.envpol.2017.06.028. induced by air pollution particulate matter (PM2.5) in human epithelial lung
cells (L132) in culture. J. Appl. Toxicol. 27, 284e290.
Dergham, M., Lepers, C., Verdin, A., Billet, S., Cazier, F., Courcot, D., Shirali, P.,
References Garçon, G., 2012. Prooxidant and proinflammatory potency of air pollution
particulate matter (PM0.3-2.5) produced in rural- urban- or industrial sur-
Abbas, I., Saint-Georges, F., Billet, S., Verdin, A., Mulliez, P., Shirali, P., Garçon, G., roundings in human bronchial epithelial cells (BEAS-2B). Chem. Res. Toxicol. 25,
2009. Air pollution Particulate Matter (PM2.5)-induced gene expression of 904e919.
volatile organic compound and/or polycyclic aromatic hydrocarbon- Dergham, M., Lepers, C., Verdin, A., Billet, S., Cazier, F., Courcot, D., Shirali, P.,
metabolizing enzymes in an in vitro coculture lung model. Toxicol vitro 2, Garçon, G., 2015. Temporal-spatial variations of the physicochemical charac-
37e46. teristics of air pollution particulate matter (PM0.3-2.5) and toxicological effects in
Abbas, I., Garçon, G., Saint-Georges, F., Billet, S., Verdin, A., Gosset, P., Mulliez, P., human bronchial epithelial cells BEAS-2B. Env. Res. 137, 256e267.
Shirali, P., 2010. Occurrence of molecular abnormalities of cell cycle in L132 cells Ding, R., Jin, Y., Liu, X., Ye, H., Zhu, Z., Zhang, Y., Wang, T., Xu, Y., 2017. Dose- and
after in vitro short-term exposure to air pollution PM2.5. Chem. Biol. Interact. time- effect responses of DNA methylation and histone H3K9 acetylation
188, 558e565. changes induced by traffic-related air pollution. Sci. Rep. 7, 43737e43745.
Abbas, I., Garçon, G., Saint-Georges, F., Andre , V., Gosset, P., Billet, S., Le Goff, J., Fenech, M., Bolognesi, C., Kirsch-Volders, M., Bonassi, S., Zeiger, E., Knasmüller, S.,
Verdin, A., Mulliez, P., Sichel, F., Shirali, P., 2013. Polycyclic aromatic hydrocar- Holland, N., 2007. Harmonisation of the micronucleus assay in human buccal
bons within airborne particulate matter (PM2.5) produced DNA bulky stable cells-a Human Micronucleus (HUMN) project (www,humn,org) initiative
adducts in a human lung cell coculture model. J. Appl. Toxicol. 33, 109e119. commencing in 2007. Mutagenesis 22, 3e4.
Abbas, I., Verdin, A., Escande, F., Saint-Georges, F., Cazier, F., Mulliez, P., Courcot, D., Fernandez-Marcelo, T., 2015. Telomere length and telomerase activity in non-small
Shirali, P., Gosset, P., Garçon, G., 2016. In vitro short-term exposure to air cell lung cancer prognosis: clinical usefulness of a specific telomere status.
pollution PM2.5-0.3 induced cell cycle alterations and genetic instability in a J. Exp. Clin. Cancer Res. 34 (1), 78.
human lung cell coculture model. Env. Res. 147, 146e158. Garçon, G., Dagher, Z., Zerimech, F., Ledoux, F., Courcot, D., Aboukais, A., Puskaric, E.,
Alleman, L.Y., Lamaison, L., Perdrix, E., Robache, A., Galloo, J.C., 2010. PM10 metal Shirali, P., 2006. Dunkerque city air pollution particulate matter-induced cyto-
concentrations and source identification using positive matrix factorization and toxicity oxidative stress and inflammation in human epithelial lung cells (L132)
wind sectoring in a French industrial zone. Atmos. Res. 96, 612e625. in culture. Toxicol vitro 20, 519e528.
Andre , V., Billet, S., Pottier, D., Le Goff, J., Pottier, I., Garçon, G., Shirali, P., Sichel, F., Grova, N., Salque bre, G., Schroeder, H., Appenzeller, B.M., 2011. Determination of
2011. Mutagenicity and genotoxicity of PM2,5 issued from an urbano- PAHs and OH-PAHs in rat brain by gas chromatography tandem triple quad-
industrialized area of Dunkerque (France). J. Appl. Toxicol. 31 (2), 131e138. rupole mass spectrometry. Chem. Res. Toxicol. 24 (10), 1653e1667.
Baccarelli, A., Vollati, B., 2009. Epigenetics and environmental chemicals. Curr. Opin. Grova, N., Salque bre, G., Hardy, E.M., Schroeder, H., Appenzeller, B.M., 2014. Tetra-
Pediatr. 21, 243e251. hydroxylated-benzo[a]pyrene isomer analysis after hydrolysis of DNA-adducts
rube
Be , K.A., Prytherch, Z., Job, C., Hughes, T., 2009. Human primary bronchial lung isolated from rat and human white blood cells. J. Chromatogr. A 1364, 183e191.
cell constructs: the new respiratory models. Toxicology 278, 311e318. Grova, N., Hardy, E.M., Meyer, P., Appenzeller, B.M., 2016. Analysis of tetrahy-
rube
Be , K.A., Prytherch, Z., Job, C., Hughes, T., 2010. Human primary bronchial lung droxylated benzo[a]pyrene isomers in hair as biomarkers of exposure to benzo
cell constructs: the new respiratory models. Toxicology 278, 311e318. [a]pyrene. Anal. Bioanal. Chem. 408 (8), 1997e2008.
Billet, S., Garçon, G., Dagher, Z., Verdin, A., Ledoux, F., Courcot, D., Aboukais, A., Gualtieri, M., Øvrevik, J., Holme, J.A., Perrone, M.G., Bolzacchini, E., Schwarze, P.E.,
Shirali, P., 2007. Ambient particulate matter (PM2.5): physicochemical charac- Camatini, M., 2010. Differences in cytotoxicity versus pro-inflammatory potency
terization and metabolic activation of the organic fraction in human lung of different PM fractions in human epithelial lung cells. Toxicol vitro 24, 29e39.
epithelial cells A549. Environ. Res. 105, 212e223. Gualtieri, M., Ovrevik, J., Mollerup, S., Asare, N., Longhin, E., Dahlman, H.J.,
Billet, S., Abbas, I., Le Goff, J., Verdin, A., Andre, V., Lafargue, P.,E., Hachimi, A., Camatini, M., Holme, J.A., 2011. Airborne urban particles (Milan winter-PM2.5)
Cazier, F., Sichel, F., Shirali, P., Garçon, G., 2008. Genotoxic potential of polycyclic cause mitotic arrest and cell death: effects on DNA mitochondria AhR binding
aromatic hydrocarbons-coated onto airborne particulate matter (PM2.5) in hu- and spindle organization. Mutat. Res. 713, 18e31.
man lung epithelial A549 cells. Cancer Lett. 270, 144e155. Herceg, Z., Vaissie re, T., 2011. Epigenetic mechanisms and cancer. An interface be-
Blasco, H., Garçon, G., Patin, F., Veyrat-Durebex, C., Boyer, J., Devos, D., Vourc'h, P., tween the environment and the genome. Epigenetics 6 (7), 804e819.
Andres, C.R., Corcia, P., 2017. Panel of oxidative stress and inflammatory bio- Hewitt, G., Jurk, D., Marques, F.D.M., Correia-melo, C., Hardy, T., Gackowska, A.,
markers in ALS: a pilot study. Can. J. Neurol. Sci. 44 (1), 90e95. Anderson, R., Taschuk, M., Mann, J., Passos, J.F., 2012. Telomeres are favoured
Boublil, L., Asse mat, E., Borot, M.C., Boland, S., Martinon, L., Sciare, J., Baeza- targets of a persistent DNA damage response in ageing and stress-induced
Squiban, A., 2013. Development of a repeated exposure protocol of human senescence. Nat. Commun. 3, 708e717.
bronchial epithelium in vitro to study the long-term effects of atmospheric IARC Publications, 2015. In: Straif, Kurt, Cohen, Aaron, Samet, Jonathan (Eds.), Air
particles. Toxicol vitro 27, 533e542. Pollution and Cancer. IARC Scientific Publication No. 161 download at http://
Bozkus, F., Guler, S., Simsek, S., 2016. Serum telomerase levels and COPD exacer- www.iarc.fr/en/publications/books/sp161/.
bations. Respir. Care 61 (3), 359e365. Ho, S.M., Johnson, A., Tarapore, P., Janakiram, V., Zhang, X., Leung, Y.K., 2012.
Cantone, L., Angelici, L., Bollati, V., Bonzini, M., Apostoli, P., Tripodi, A., Bertazzi, P.A., Environmental epigenetics and its implication on disease risk and health out-
Baccarelli, A.A., 2014. Extracellular histones mediate the effects of metal-rich air comes. ILAR J. 53 (3/4), 289e305.
particles on blood coagulation. Environ. Res. 132, 76e82. Ibuki, Y., Toyooka, T., Zhao, X., Yoshida, I., 2014. Cigarette sidestream smoke induces
Caramori, G., Adcock, I.,M., Casolari, P., Ito, K., Jazrawi, E., Tsaprouni, L., Villetti, G., histone H3 phosphorylation via JNK and PI3K/Akt pathways leading to the
Civelli, M., Carnini, C., Chung, K.,F., Barnes, P.,J., Papi, A., 2014. Unbalanced expression of proto-oncogenes. Carcinogenesis 35 (6), 1228e1237.
oxidant-induced DNA damage and repair in COPD: a link towards lung cancer. Klein, S.G., Serchi, T., Hoffmann, L., Blo€ meke, B., Gutleb, A.C., 2013. An improved 3D
Thorax 66, 521e527. tetraculture system mimicking the cellular organisation at the alveolar barrier
Cassee, F.R., Heroux, M.E., Gerlofs-Nijland, M.E., Kelly, F.J., 2013. Particulate matter to study the potential toxic effects of particles on the lung. Part Fibre Toxicol. 26
beyond mass: recent health evidence on the role of fractions chemical con- (10), 31.
stituents and sources of emission. Inhal. Toxicol. 25, 802e812. Kordinas, V., Ioannidis, A., Chatzipanagiotou, S., 2016. The telomere/telomerase
Cawthon, R.M., 2009. Telomere length measurement by a novel monochrome system in chronic inflammatory diseases, cause or effect? Genes (Basel) 3 (7), 9.
multiplex quantitative PCR method. Nucleic Acids Res. 37 (3), e21. Leclercq, B., Happillon, M., Antherieu, S., Hardy, E.M., Alleman, L.Y., Grova, N.,
Chen, X., Guan, X.J., Luan, C.Y., Guo, X.J., 2015. Acetylation of lysine 9 on histone H3 Perdrix, E., Appenzeller, B.M., Lo Guidice, J.M., Coddeville, P., Garçon, G., 2016.
is associated with increased pro-inflammatory cytokine release in a cigarette Differential responses of healthy and chronic obstructive pulmonary diseased
smoke-induced rat model through HDAC1 depression. Inflamm. Res. 64, human bronchial epithelial cells repeatedly exposed to air pollution-derived
B. Leclercq et al. / Environmental Pollution 230 (2017) 163e177 177

PM4. Env. Poll. 218, 1074e1088. Saint-Georges, F., Garçon, G., Escande, F., Abbas, I., Verdin, A., Gosset, P., Mulliez, P.,
Leclercq, B., Alleman, L.Y., Perdrix, E., Riffault, V., Happillon, M., Strecker, A., Lo- Shirali, P., 2009. Role of air pollution particulate matter (PM2.5) in the occur-
Guidice, J.M., Garçon, G., Coddeville, P., 2017. Particulate metal bioaccessibility rence of loss of heterozygosity in multiple critical regions of 3p chromosome in
in physiological fluids and cell culture media: toxicological perspectives. Envi- human epithelial lung cells L132. Toxicol. Lett. 187, 172e179.
ron. Res. 156, 148e157. Singh, N.P., McCoy, M.T., Tice, R.R., Schneider, E.L., 1988. A simple technique for
Lepers, C., DerghamM, Armand L., Billet, S., Verdin, A., Andre , V., Pottier, D., quantitation of low levels of DNA damage in individual cells. Exp. Cell Res. 175,
Courcot, D., Shirali, P., Sichel, F., 2014. Mutagenicity and clastogenicity of native 184e191.
airborne particulate matter samples collected under industrial urban or rural Soberanes, S., Gonzalez, A., Urich, D., Chiarella, S.E., Radigan, K.A., Osornio-
influence. Toxicol Vitro 28, 866e874. Vargas, A., Joseph, J., Kalyanaraman, B., Ridge, K.M., Chandel, N.S., Mutlu, G.M.,
Li, J., Li, W.X., Bai, C., Song, Y., 2015. Particulate matter-induced epigenetic changes De Vizcaya-Ruiz, A., Budinger, G.R., 2012. Particulate matter air pollution in-
and lung cancer. Clin. Respir. J. http://dx.doi.org/10.1111/crj.12389. duces hypermethylation of the P16 promoter via a mitochondrial ROS-JNK-
Liu, F., Killian, J.K., Yang, M., Walker, R.L., Hong, J.A., Zhang, M., Davis, S., Zhang, Y., DNMT1 pathway. Sci. Rep. 2, 275e285.
Hussain, M., Xi, S., Rao, M., Meltzer, P.A., Schrump, D.S., 2010. Epigenomic al- Sundar, I.K., Nevid, M.Z., Friedman, A.E., Rahman, I., 2010. Cigarette smoke induces
terations and gene expression profiles in respiratory epithelia exposed to distinct histone modifications in lung cells: implications for the pathogenesis of
cigarette smoke condensate. Oncogene 29, 3650e3664. COPD and lung cancer. J. Proteome Res. 13, 982e996.
Longhin, E., Pezzolato, E., Mantecca, P., Holme, J.A., Franzetti, A., Camatini, M., Tarantini, L., Bonzini, M., Apostoli, P., Pegoraro, V., Bollati, V., Marinelli, B.,
Gualtieri, M., 2013. Season linked responses to fine and quasi-ultrafine Milan Cantone, L., Rizzo, G., Hou, L., Schwartz, J., Bertazzi, P.A., Baccarelli, A., 2009.
PM in cultured cells. Toxicol vitro 27, 551e559. Effects of particulate matter on genomic DNA methylation content and iNOS
Longhin, E., Capasso, L., Battaglia, C., Proverbio, M.,C., Consentino, C., Cifola, I., promoter methylation. Environ. Health Perspect. 117 (2), 217e222.
Mangano, E., Camatini, M., Gualtieri, M., 2016. Integrative transcriptomic and Tice, R.R., Agurell, E., Anderson, D., Burlinson, B., Hartmann, A., Kobayashi, H.,
protein analysis of human bronchial BEAS-2B exposed to seasonal urban par- Miyamae, Y., Rojas, E., Ryu, J.C., Sasaki, Y.F., 2000. Single cell gel/comet assay:
ticulate matter. Environ. Poll. 209, 87e98. guidelines for in vitro and in vivo genetic toxicology testing. Environ. Mol.
Loxham, M., Morgan-Walsh, R.J., Cooper, M.J., Blume, C., Swindle, E.J., Mutagen 35, 206e221.
Dennison, P.W., Howarth, P.H., Cassee, F.R., Teagle, D.A.H., Palmer, M.R., To, T., Zhu, J., Larsen, K., Simatovic, J., Feldman, L., Ryckman, K., Gershon, A.,
Davies, D.E., 2015. The effects on bronchial epithelial mucociliary cultures of Lougheed, M.D., Licskai, C., Chen, H., Villeneuve, P.J., Crighton, E., Su, Y.,
coarse fine and ultrafine particulate matter from an underground railway sta- Sadatsafavi, M., Williams, D., Carlsten, C., 2016. Canadian Respiratory Research
tion. Toxicol. Sci. 145 (1), 98e107. Network, 2016. Progression from asthma to chronic obstructive pulmonary
Mbengue, S., Alleman, L.Y., Flament, P., 2014. Size-distributed metallic elements in disease, COPD; is air pollution a risk factor? Am. J. Respir. Crit. Care Med. 194 (4),
submicronic and ultrafine atmospheric particles from urban and industrial 429e438.
areas in northern France. Atmos. Res. 135e136, 35e47. Vucic, E.A., Chari, R., Thu, K.L., Wilson, I.M., Cotton, A.M., Kennett, J.Y., Zhang, M.,
OECD, 2014. Test N 487: In Vitro Mammalian Cell Micronucleus Test OECD Lonergan, K.M., Steiling, K., Brown, C.J., McWilliams, A., Ohtani, K.,
Guidelines for Testing of Chemicals. Section 4. OECD Publishing, Paris. Lenburg, M.E., Sin, D.D., Spira, A., MacAulay, C.E., Lam, S., Lam, W.L., 2014. DNA
Pauk, N., Klimesova, S., Kara, J., Topinka, J., Labaj, J., 2013. The relevance of moni- methylation is globally disrupted and associated with expression changes in
toring of antibodies against the polycyclic aromatic hydrocarbon (PAH) and chronic obstructive pulmonary disease small airways. Am. J. Respir. Cell Mol.
PAH-DNA adducts in serum in relation to lung cancer and chronic obstructive Biol. 50 (5), 912e922.
pulmonary disease (COPD). Neoplasma 60 (2), 182e187. Wang, T., Garcia, J.G.N., Zhang, W., 2012. Epigenetic regulation in particulate matter-
Pezzulo, A.A., Starner, T.D., Scheetz, T.E., Traver, G.L., Tilley, A.E., Harvey, B.G., mediated cardiopulmonary toxicities: a systems biology perspective. Curr.
Crystal, R.G., McCray Jr., P.B., Zabner, J., 2011. The aireliquid interface and use of Pharmacogenomics Person. Med. 10 (4), 314e321.
primary cell cultures are important to recapitulate the transcriptional profile of Zhang, X., Lin, S., Funk, W.E., Hou, L., 2013. Environmental and occupationalexpo-
in vivo airway epithelia. Am. J. Physiol. Lung Cell Mol. Physiol. 300, L25eL31. sure to chemicals and telomere length in human studies. Occup. Environ. Med.
Rode, L., Bojesen, S.E., Weischer, M., Vestbo, J., Nordestgaard, B.G., 2013. Short 70 (10), 743e749.
telomere length lung function and chronic obstructive pulmonary disease in 46 Zhou, B., Liang, G., Qin, H., Peng, X., Huang, J., Li, Q., Qing, L., Zhang, L., Chen, L., Ye, L.,
396 individuals. Thorax 68, 429e435. Niu, P., Zou, Y., 2014. p53-dependent apoptosis induced in human bronchial
Saint-Georges, F., Abbas, I., Billet, S., Verdin, A., Gosset, P., Mulliez, P., Shirali, P., epithelial (16-HBE cells) by PM2,5 sampled from air in Guangzhou, China.
Garçon, G., 2008. Gene expression induction of volatile organic compound and/ Toxicol. Mech. Methods 24, 552e559.
or polycyclic aromatic hydrocarbon-metabolizing enzymes in isolated human Zong, D.D., Ouyang, R.J., Chen, P., 2015. Epigenetic mechanisms in chronic
alveolar macrophages in response to airborne particulate matter PM2.5. Toxi- obstructive pulmonary disease. Eur. Rev. Med. Pharmacol. Sci. 19 (5), 844e856.
cology 244, 220e230.

You might also like