You are on page 1of 16

Environmental Pollution 213 (2016) 809e824

Contents lists available at ScienceDirect

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

Review

Biological impact of environmental polycyclic aromatic hydrocarbons


(ePAHs) as endocrine disruptors*
Yanyan Zhang a, Sijun Dong b, Hongou Wang b, Shu Tao a, Ryoiti Kiyama c, *
a
College of Urban and Environmental Sciences, Peking University, Beijing 100871, PR China
b
Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, PR China
c
Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki 305-8566, Japan

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

Article history: Polycyclic aromatic hydrocarbons (PAHs) are often detected in the environment and are regarded as
Received 23 October 2015 endocrine disruptors. We here designated mixtures of PAHs in the environment as environmental PAHs
Received in revised form (ePAHs) to discuss their effects collectively, which could be different from the sum of the constituent
3 March 2016
PAHs. We first summarized the biological impact of environmental PAHs (ePAHs) found in the atmo-
Accepted 20 March 2016
sphere, sediments, soils, and water as a result of human activities, accidents, or natural phenomena.
ePAHs are characterized by their sources and forms, followed by their biological effects and social impact,
and bioassays that are used to investigate their biological effects. The findings of the bioassays have
Keywords:
Endocrine disruptor
demonstrated that ePAHs have the ability to affect the endocrine systems of humans and animals. The
Social impact pathways that mediate cell signaling for the endocrine disruptions induced by ePAHs and PAHs have also
Evaluation assay been summarized in order to obtain a clearer understanding of the mechanisms responsible for these
Cell signaling pathway effects without animal tests; they include specific signaling pathways (MAPK and other signaling
Estrogenic activity pathways), regulatory mechanisms (chromatin/epigenetic regulation, cell cycle/DNA damage control, and
cytoskeletal/adhesion regulation), and cell functions (apoptosis, autophagy, immune responses/inflam-
mation, neurological responses, and development/differentiation) induced by specific PAHs, such as benz
[a]anthracene, benzo[a]pyrene, benz[l]aceanthrylene, cyclopenta[c,d]pyrene, 7,12-dimethylbenz[a]
anthracene, fluoranthene, fluorene, 3-methylcholanthrene, perylene, phenanthrene, and pyrene as well
as their derivatives. Estrogen signaling is one of the most studied pathways associated with the
endocrine-disrupting activities of PAHs, and involves estrogen receptors and aryl hydrocarbon receptors.
However, some of the actions of PAHs are contradictory, complex, and unexplainable. Although several
possibilities have been suggested, such as direct interactions between PAHs and receptors and the
suppression of their activities through other pathways, the mechanisms underlying the activities of PAHs
remain unclear. Thus, standardized assay protocols for pathway-based assessments are considered to be
important to overcome these issues.
© 2016 Elsevier Ltd. All rights reserved.

1. Introduction exchange processes (Baek et al., 1991; Buehler and Hites, 2002). We
herein referred to PAHs associated with environmental media such
Polycyclic aromatic hydrocarbons (PAHs) are a large group of as particulate matter, soil, water, and sediment as environmental
organic compounds comprised of two or more fused benzene rings PAHs (ePAHs). ePAHs can be classified or characterized by their
arranged in various configurations (Mumtaz et al., 1996). They forms/sources, biological effects, social impacts and bioassays used
generally originate from combustion processes and are widely to detect and evaluate them. The importance of ePAHs is empha-
distributed in the natural environment as a result of atmospheric sized here because their effects could be different from the sum of
transportation, wet and dry deposition, and surface-to-air the effects by their constituent PAHs due to their additive or sub-
tractive contributions and the contribution of the matrix, which
normally reduces the bioavailability of PAHs to target organisms
*
This paper has been recommended for acceptance by Frank Arthur von Hippel. (Ramesh et al., 2004). More than one hundred PAHs exist in the
* Corresponding author. environment and often occur as complex mixtures (Mumtaz et al.,
E-mail address: kiyama.r@aist.go.jp (R. Kiyama).

http://dx.doi.org/10.1016/j.envpol.2016.03.050
0269-7491/© 2016 Elsevier Ltd. All rights reserved.
810 Y. Zhang et al. / Environmental Pollution 213 (2016) 809e824

1996). Sixteen PAHs are included among the 129 priority pollutants DNA (Bostro €m et al., 2002; Koganti et al., 2001; Ramesh et al., 2004;
announced by the U.S. Environmental Protection Agency (Keith and Stegeman and Lech, 1991). In non-occupational settings, PAHs have
Telliard, 1979), some of which are carcinogenic (Denissenko et al., been identified as causative agents of cardiopulmonary and car-
1996; Bostro € m et al., 2002). ePAHs and the constitutent PAHs diovascular diseases. Previous studies using cellular and animal
may be taken up by living organisms and accumulate via the food models have suggested that PAHs associated with ambient partic-
chain, and are exposed to humans via inhalation and the ingestion ulate matter generate reactive oxygen species (ROS), resulting in
of food; therefore, they pose potential threats to the ecosystem and oxidative stress and inflammatory responses, which account for the
human health (Ramesh et al., 2004; Zhang et al., 2014). For prevalence and exacerbation of asthma and allergic diseases. In
example, Zhang et al. (2009) estimated that the overall population addition, chronic exposure to low-dose PAHs may be responsible
attributable fraction for lung cancer caused by the inhalation of for immune-mediated pregnancy loss, and maternal exposure to
PAHs was 1.6% in China. Due to the extent of environmental PAH PAHs has been suggested to affect fetal development such as birth
contamination, especially in developing countries, identifying the length, weight, and head circumference.
biological effects of ePAHs is considered essential for pollution in-
dications and the prevention of adverse effects on population
health. In this section, we summarized the source and forms of 1.3. Social impact of ePAHs
ePAHs in Table 1, and presented their direct biological effects in
living organisms and humans as well as the implications of their A deeper understanding of the biological effects of ePAHs is
social impact on the ecosystem and population health. essential for understanding their social impacts, such as bio-
monitoring, pollution control, pollution indication, remediation
1.1. Source and forms of ePAHs and risk prevention (Table 1). For example, the abundance of
nematode communities in addition to nah and pdo1 genes (PAH-
ePAHs can be found in various sources, such as air, dust, smoke, degrading genes) were previously reported to be positively influ-
sediment, water, soil and oil in the environment and/or pollutants, enced by PAH levels in soil, and may be used as bioindicators for
as forms such as particulate matter and materials in oven, fuel and PAH contamination and in the isolation of bacterial strains that
tar. PAHs in the air mainly originate from the incomplete com- degrade PAHs (Han et al., 2014). The biological effects of ePAHs in
bustion of carbonaceous materials such as fossil fuel and biomass fish and the invertebrates described above (Section 1.2) may assist
including emissions from coke and coal burning in occupational in the development of early warning signals for PAH contamination
settings and forest fire, the exhaust fumes of motor vehicles, flues of and remediation and mitigation strategies, and also in the assess-
biomass burning for cooking and heating in rural areas, and tobacco ment of restoration performance. Regarding population health, the
smoke (Baek et al., 1991; Rubin, 2001; Shen et al., 2013). Due to genotoxic effects of PAHs such as DNA adducts, chromosome ab-
their semivolatile nature, PAHs in the atmosphere may exist either errations, and ras oncogene overexpression, may be used as bio-
in a gaseous or particulate phase, depending on their molecular markers for PAH exposure and cancer risk assessments. By
weights (Li et al., 2014). PAHs in soil and water are mainly derived monitoring maternal PAH exposure and fetal development, pre-
from atmospheric deposition, especially in remote places such as dictive biomarkers may be established for fetal health and even
high-altitude lake areas (McVeety and Hites, 1988; Nam et al., 2008, childhood intelligence and cognitive development (Perera et al.,
2009), or from industrial process such as produced water. Oil spills 1998, 1999). The identification of early biological effects (chromo-
are a prominent source of PAHs in marine water, which is then somal aberrations, sister chromatid exchanges, and micronuclei)
introduced to marine sediments. may also be applied to heavy PAH-exposed workers for the purpose
of cancer prevention; however, contradictory findings have been
1.2. Biological effects of ePAHs reported (Porru et al., 1997). Furthermore, identifying the impact of
tobacco smoke on health as well as the underlying mechanisms will
PAHs in the environment may be exposed to living organisms be beneficial for regulating tobacco control policies.
via different routes and result in various biological effects (Table 1). As summarized here, quite a variety of biological activities of
Many studies have reported correlations between the induction of ePAHs have been reported, which include acute/chronic toxicity,
cytochrome P450 (CYP), elevated 7-ethoxyresorufin-O-deethylase reproductive effects, embryotoxicity, teratogenicity, mutagenicity,
(EROD) activities, lysosomal membrane destabilization, DNA dam- caricinogenicity and endocrine disruptions (Lisouza et al., 2012;
age, and endocrine and reproductive effects in fish and in- Kameda et al., 2012). While the genotoxic effect of ePAHs is
vertebrates with PAH contamination in the water and sediments important, we focused here on the endocrine disruptor action by
they inhabit. Plants also uptake atmospheric and soil PAHs, and this ePAHs.
contamination induces leaf injuries, decreased biomass production,
and other adverse biological and physiological effects. Furthermore,
the abundance and composition of the soil microbial community 2. Biological activities of ePAHs
are strongly influenced by PAH contamination levels in soil.
Humans may be exposed to environmental PAHs via the intake A variety of bioassays have been developed and applied to un-
of PAH-contaminated food as well as the unintentional ingestion of derstand the role of ePAHs as endocrine disruptors. The bioassays
soil and dust via hand-to-mouth behaviors, inhalation, and dermal used to investigate the biological effects of ePAHs are summarized
contact (Buratti et al., 2007; Harris et al., 2013; McClean et al., 2007; according to the types or sources of ePAHs (Table 2), which have
Pampanin et al., 2014; Zhang et al., 2014). Epidemic studies have been roughly categorized into two types: environmental samples
demonstrated that heavy exposure to PAHs from occupational en- containing various types of PAHs, or pollutants themselves with
vironments increases the risk of developing various cancers known chemicals or their sources (see Table 1). The assays used
including lung, skin, bladder, and larynx cancers (Bostro € m et al., have been categorized into animal/plant tests, cell assays, microbial
2002; Boffetta et al., 1997; Tolbert, 1997; Wagner et al., 2015). The tests, reporter-gene assays, protein assays, and other assays. The
carcinogenicity of PAHs is associated with their potential to form effects of ePAHs as endocrine disruptors was initially demonstrated
reactive diol epoxide intermediates catalyzed by CYP, which may be in animal tests, and then by mechanism-based analyses using cell
induced by PAH exposure, and their subsequent covalent binding to and protein assays.
Y. Zhang et al. / Environmental Pollution 213 (2016) 809e824 811

Table 1
Biological effects and social impact of ePAHs.

Form/source Biological effect Social impact Reference

Environment
Air/diesel/dust/particulate/smoke/volatile
Ambient air Plant growth/symbiosis/photosynthesis Risk assessment Desalme et al., 2013
Ambient air Lung, bladder, and skin cancers Prevention of cancer Porru et al., 1997
Ambient air PAH-DNA adducts PAH exposure and lung cancer Kriek et al., 1993
Particulate matter ROS/oxidative stress/inflammation Prevalence of allergic diseases Huang et al., 2015
Particulate matter Generation of ROS Prevalence of childhood asthma Karimi et al., 2015
Particulate matter Cardiovascular diseases Toxicology/risk control Schnelle-Kreis et al., 2009
Particulate matter Genotoxic effects Biomarkers for PAH exposure Farmer et al., 2003
Particulate matter Fetal development Childhood intelligence/development Perera et al., 1999
Smoke (tobacco) Immune-mediated pregnancy loss Pollution control Detmar and Jurisicova, 2010
Smoke (tobacco) Atherosclerosis Prevention of cardiovascular diseases Ramos and Moorthy, 2005
Smoke (tobacco) Cardiovascular diseases Assessment of cardiotoxicity Korashy and El-Kadi, 2006
Smoke (tobacco) Carcinogenicity Synergistic mechanisms by PAHs Rubin, 2001
Settled house dust Mutagenic potency Carcinogenic risk Maertens et al., 2004
Diesel exhaust particles B-cell differentiation/IgE production Allergic airway diseases Pampanin et al., 2014
Coal/coke/combustion/fossil fuel/tar
Coke oven Increased oxidative stress in workers Biomarkers for toxic responses Gao et al., 2014
Coke oven Oxidative stress and lipid peroxidation Immunological effects in workers Jeng et al., 2011
Coke oven DNA methylation states in workers Epigenetic markers Pavanello et al., 2009
Coke oven CYP1B1 mRNA in peripheral blood Biomarker validation Hanaoka et al., 2002
Coal tar mixtures Carcinogenicity Effects of small PAHs Benford et al., 2010
Coal burning Fetal development Establishing predictive biomarkers Perera et al., 1998
Coal-tar/creosote/bitumen/juniper tar/engine oils Formation of DNA adducts Hazards by epidermal contact Phillips et al., 1990
Fumes bitumens Carcinogenicity Biomarkers of non-priority PAHs Binet et al., 2002
Biomass fuel emissions Respiratory/cardiovascular diseases Prevention of health risks Kim et al., 2011a
Biomass fuel emissions Effects on human blood Pollution control Kamal et al., 2015
Pollutant
Oil
Marine oil Phase-I enzymes/DNA adducts/neoplasm Impact on marine ecosystems Hylland, 2006
Crude oil spill Increase in fish bile FACs/CYP response Toxicity/lesions/reproductive failure Lee and Anderson, 2005
Bunker fuel spill Lysosomal membrane destabilization Sublethal effects in bivalves Hwang et al., 2014
Oil spill Elevated EROD activities in eelpouts Multi-biomarkers for PAH exposure Sturve et al., 2014
Oil spill Adverse effects on eelpouts Adverse biological effects Kreitsberg et al., 2012
Oil spill Elevated metabolites/CYP1A induction Identifying extra exposure pathways Jung et al., 2011
Oil spill Detoxification/defense systems in mussel Biomonitoring of acute exposure Sureda et al., 2011
Oil spill DNA strand break in mussels and clams Assessment of exposure/damage Thomas et al., 2007
Oil spill DNA damage via the food chain Identifying genotoxic contaminants Lemiere et al., 2004
Sediment
Harbor sediment Bacterial population Remediation/mitigation strategies Slater et al., 2008
Marine sediment Accumulation in marine organisms Impact on marine populations Meador et al., 1995
Lake sediment Toxic to photoluminescent bacteria Potential risk upon disturbances Hyo€ tyla
€inen and Olkari, 1999
Lake sediment Forming DNA adducts in the eel Indicators for PAH exposure van Schooten et al., 1995
River sediment CYP1A activity/porphyrin accumulation Biomonitoring Huuskonen et al., 2000
Sediment (rivers/lakes/marines) Salmonella mutagenic potency Health and viability of aquatic biota Chen and White, 2004
Sewage/sludge/waste/wastewater
Contaminated water Induction of cytochrome P-450 Monitoring exposure to carcinogens Stegeman and Lech, 1991
Wastewater (oil company) Mutagenicity in bacterial strains Field monitoring Re
ategui-Zirena et al., 2014
Marine water EROD activity in leaping mullet Risk elimination Sen et al., 2010
Produced water Gene expression in zebrafish Monitoring early warning signals Holth et al., 2008
Produced water DNA damage in mosquito fish Toxicology of produced water Caliani et al., 2009
Produced water Reproductive effects to cod Application of multiple biomarkers Sundt and Bjo €rkblom, 2011
Produced water Effects to cod and blue mussel Population and ecosystem effects Bakke et al., 2013
Soil
Surface soil Ecological risk Remedial measures Lang et al., 2012
Surface soil Soil nematode community Indicators for PAH soil pollution Chen et al., 2009
Contaminated soil Abundance of nah and pdoI genes Indicators for PAH soil pollution Han et al., 2014
Coal tar contaminated soil DNA adducts in the mouse lung Main components inducing responses Koganti et al., 2001
Creosote-contaminated soil Microbial community composition Isolation of PAH degraders €rneman et al., 2008
To

Abbreviations: EROD: ethoxyresorufin-O-deethylase; FAC: fluorescent aromatic compound; PLHC-1: a fish hepatoma cell line; PM: particulate matter; ROS: reactive oxygen
species.

2.1. Assays for biological activities of ePAHs exposure, whereas no significant increase was noted in lung tumors
(Soffritti et al., 2013). Serum and tissues obtained from Wistar Han
2.1.1. Animal/plant tests rats were used to evaluate genotoxicity and the induction of
Various tests have been conducted using animals and plants in oxidative stress after long-term exposure to diesel exhaust fumes
order to evaluate the biological activities of ePAHs. For example, containing PAHs (Hallberg et al., 2015). The toxicity and mutage-
Sprague-Dawley rats were used to examine the carcinogenic effects nicity of fractions of crude coal tar containing various PAHs were
of coarse particles containing PAHs as dominant components, and also investigated using a chick embryotoxicity screening test
the increase observed in the incidence of terminal bronchiolar (CHEST) in addition to a Salmonella/microsome bioassay, and a
hyperplasia and squamous metaplasia was associated with PAH significant relationship was observed between PAH types and their
812 Y. Zhang et al. / Environmental Pollution 213 (2016) 809e824

Table 2
Bioassays for the study of ePAHs.

Form/Source Bioassay Reference

Environmental
Air/diesel/dust/particulate/smoke/volatile
Air (coke oven) Ames test Dobias et al., 1999
DEPs (compressor) Ames test Mutlu et al., 2013
Dust Life-span carcinogenicity bioassay (rats) Soffritti et al., 2013
Extractable organic matter (urban air) DNA microarray Líbalova et al., 2014
Particulate matter (biomass burning) Micronucleus bioassay de Oliveira Alves et al., 2011
Particulate matter (cashew nut roasting) Micronucleus bioassay Galv~ao et al., 2014
Particulate matter (smoked meat) CALUX assay Kuhn et al., 2008
Particulate matter (urban air) Ames test Villalobos-Pietrini et al., 2007; Traversi et al., 2011
Semi-volatiles (vehicle exhaust) Ames test/AhR receptor affinity test Westerholm and Egeba €ck, 1994
Smoke (tobacco) Ames test DeMarini et al., 1995
Soot particles (biomass fuel) Yeast estrogen screen Wang et al., 2005
Urban air CALUX assay/Salmonella/microsome assay Ciganek et al., 2004
Urban air/DEPs Comet assay/ELISA/rat bioassay Hallberg et al., 2015
Coal/coke/combustion/fossil fuel/tar
Coal tar Chick test/Salmonella/microsome assay Mayura et al., 1999
Coal tar/coke/oil/soot/smoke Cancer bioassays Collins et al., 1998 (review)
Combustion emissions Bioassays Lewtas, 2007 (review)
Combustion products Amphipod/polychaete/clam bioassays Van Dolah et al., 2005
Combustion products (meat/wood/charcoal) Yeast estrogen screen Muthumbi et al., 2003
Pollutant
Oil
Oil (lampblack) Dermal bioassay (human skin section) Stroo et al., 2005
Oil production discharges EROD assay/ELISA Harman et al., 2010
Oil shale EROD assay Huuskonen et al., 2000
Oil spills/bay sediments AhR reporter-gene assay Puy-Azurmendi et al., 2010
Petrogenic extracts Comet assay/fish micronucleus assay Le Bihanic et al., 2014
Sediment
Bay sediments P450 reporter-gene assay Koh et al., 2001
Bay sediments Sea urchin/oyster bioassays Geffard et al., 2001
Bay sediments EROD assay/yeast estrogen screen Annavarapu et al., 2004
Bay sediments EROD assay/yeast estrogen screen Weston et al., 2010
Creek sediments (hazardous waste) Flow cytometry Matson et al., 2009
Lagoon sediments Sea urchin bioassays D'Adamo et al., 2014
Lagoon sediments Comet assay/yeast estrogen screen Amaeze et al., 2015
Lake sediments Ames test Marvin et al., 2000
Lake sediments AhR/ER reporter-gene assay/snail bioassay Mazurova  et al., 2008
Lake sediments (coal tar) Ames test Marvin et al., 1999
Marine sediments English sole feeding bioassay Rice et al., 2000
River sediments CALUX assay/ER reporter-gene assay/Salmonella umu-test Vondra cek et al., 2001
River sediments Mutatox bioassay (luminescent bacteria test) Thomas et al., 2002
River sediments Animal bioassays/Microtox bioassay den Besten & van den Brink, 2005
River sediments LUMIStox bioassay/duckweed bioassay Olajire et al., 2005
Sewage/sludge/waste/water
Groundwater Adult hydra bioassay Ake et al., 2003
Groundwater EROD assay/CFDA-AM assay Bopp et al., 2007
Lake water Ames test/EROD assay/yeast estrogen screen Rastall et al., 2004
Sewage sludge ToxAlert bioassay (luminescent bacteria test) rez et al., 2001
Pe
Sewage sludge composts Phytotoxkit (seed germination) test Oleszczuk, 2008
Soil
Bioremediated soils Salmonella/microsome assay Brooks et al., 1998
Bioremediated soils Microtox bioassay (luminescent bacteria test) Lau et al., 2003
Contaminated soils Earthworm survival bioassay Charrois et al., 2001
Contaminated soils LUMIStox bioassay (luminescent bacteria test) Hirmann et al., 2007
Contaminated soils (MGP) Ames test Park et al., 2008
Contaminated soils Earthworm/lettuce/nematode bioassays Cofield et al., 2008
Contaminated soils CALUX assay/comet assay Andersson et al., 2009
Contaminated soils AhR reporter-gene assay Larsson et al., 2013
Contaminated soils lacZ-transgene mutation assay Lemieux et al., 2015

Abbreviations: AhR: aryl hydrocarbon receptor; CALUX chemical-activated luciferase gene expression; CFDA-AM: 5-carboxyfluorescein diacetate acetoxymethyl ester; DEP:
diesel exhaust particle; ELISA: enzyme-linked immunosorbent assay; ER: estrogen receptor; EROD: ethoxyresorufin-O-deethylase; Luc: luciferase; MGP: manufactured gas
plant.

concentrations and embryonic mortality (Mayura et al., 1999). A activities of ePAHs. For example, the phytoremediation of soil from
predator-prey interaction was also used as a bioassay to represent a former manufacturing gas plant site was assessed using a com-
realistic ecological processes; English soles fed sediment/PAH- bination of bioassays based on earthworm survivability, nematode
exposed polychaetes in the juvenile period exhibited growth mortality, lettuce germination and emergence, and microbial
retardation, the increased expression of CYP1A, and/or accumula- respiration (Cofield et al., 2008). The toxicity of PAHs in the sedi-
tion of hepatic PAH-DNA adducts (Rice et al., 2000). ments of estuarine wetlands near highways was examined by
Invertebrates have been employed to study the biological bioassays using amphipods, polychaetes, and clams, among which
Y. Zhang et al. / Environmental Pollution 213 (2016) 809e824 813

only clams showed toxicity, even though the sensitivity of the other used in this test, and bacterial colonies form on agar plates only
assays was enhanced by ultraviolet light (Van Dolah et al., 2005). after this bacterium is exposed to chemicals, which may revert its
The fresh water snail Potamopyrgus antipodarum has been used to genotype to prototrophy. Furthermore, since some chemicals are
assess the procreation toxicity of sediments highly contaminated only mutagenic after metabolic activation, mammalian microsomal
with powdered waste coal, and the findings obtained revealed that, enzymes have been complemented to overcome the shortage of
after an 8-week exposure, the fecundity of snails was stimulated at metabolic activation. An improved protocol with two new tester
low concentrations, but inhibited at high concentrations (Mazurova  strains was developed to increase the sensitivity and applicability
et al., 2008). Sea urchins and oysters were previously used to of the Ames test (Maron and Ames, 1983). Alternatively, the umu-
evaluate the embryotoxicity of marine sediments containing PAHs, test has been used to detect environmental carcinogens (Oda et al.,
and the findings obtained indicated that PAHs enhanced embryonic 1985). Salmonella typhimurium TA1535 carrying a plasmid
death, and oysters were more sensitive to PAHs than sea urchins (pSK1002) containing the fusion gene umuC'-'lacZ, in which umuC
(Geffard et al., 2001). may be activated by the presence of mutagens in SOS/DNA damage
Phytotoxicity has gained significant attention among bioassays responses, was constructed, and, after carcinogens induced SOS/
to assess ePAHs. The emergence of lettuce seedlings has been used DNA damage responses and activated the umuC'-'lacZ gene, b-
to assess phytoremediation (Cofield et al., 2008). Similarly, the galactosidase (the lacZ gene product) was produced and the level of
toxicity of contaminated sediments was assessed by analyzing the DNA damage was quantified based on b-galactosidase activity in a
number of fronds of the duckweed, the findings of which sug- colorimetric assay.
gesting that, although ePAHs inhibited growth, no significant dose- Luminescent bacteria, which emit light generated by an
dependent relationship was observed in this particular case (Olajire enzyme-catalyzed chemiluminescence reaction, have also been
et al., 2005). Moreover, sewage sludge collected at different widely used to evaluate ePAHs. For example, the genotoxicity of
wastewater treatment plants in Poland was assessed by a toxicity PAHs in sediments from estuaries was evaluated using the Mutatox
test using a commercial Phytotoxkit, in which the seed germination bioassay, a commercially available assay involving the dark variant
and growth of young cress roots were examined, and some sludge (strain M169) marine bioluminescent bacterium, Photobacterium
samples were found to inhibit seed germination by 100% phosphoreum, which emits light when it is exposed to mutagenic
(Oleszczuk, 2008). compounds and reverts to the wild-type (Thomas et al., 2002). The
LUMIStox bioassay, in which the inhibition of bioluminescence
2.1.2. Cell assay from the marine bacterium Vibrio fischeri, is quantified, has been
Assays using various types of cells have been widely used to used to assess the acute toxicities of ePAH-contaminated sediments
evaluate the biological activities of ePAHs. Due to its rapidity and in southern Nigeria (Olajire et al., 2005). ToxAlert and Microtox
sensitivity, the chemical-activated luciferase gene expression bioassays are based on a similar principle to the LUMIStox assay; a
(CALUX) assay has commonly been used to detect aryl hydrocarbon decrease in the rate of luminescence by the inhibition of respiration
receptor (AhR)-active compounds including PAHs in various envi- in V. fischeri by toxicants is quantified. The ToxAlert bioassay was
ronmental samples, such as soil, water, and sediments. A rat hep- previously used in combination with a composition analysis by gas
atoma (H4IIE) cell line, stably transfected with a construct chromatography-mass spectrometry (GC-MS) in order to evaluate
containing the dioxin-responsive element (DRE) and luciferase the toxicity of sewage sludge (Perez et al., 2001). On the other hand,
reporter gene, was used in this assay to detect the activities of AhR- the effects of spent mushroom compost on reductions in the
active compounds, such as 2,3,7,8-tetrachlorodibenzo-p-dioxin toxicity of PAHs were verified by combining the Microtox bioassay
(TCDD), in sediments and pore waters (Murk et al., 1996). On the and GC-MS (Lau et al., 2003). The protocol of the LUMIStox assay
other hand, a comet assay, which measures the migration, under was improved by miniaturization with 96-microplates, for which
alkaline conditions, of DNA from cells encapsulated into low- dark microplates are recommended because they may prevent
melting-point agarose gels, represents a simple method, but has overestimations of the level of PAHs when soil is colored (Hirmann
been widely used to quantify DNA damage and repair with high et al., 2007). Assay results are highly variable when compared
specificity and sensitivity (Singh et al., 1988). In a collaborative among the microbial tests used or with other bioassays because of
study with the International Programme on Chemical Safety, a differences in the sensitivity, specificity, and durability of chem-
Tradescantia pallida (spiderwort) micronucleus (Trad-MCN) icals; nevertheless, these assays are useful when cost, time, and
bioassay was employed to evaluate clastogens and was found to be portability are considered.
efficient and reliable for monitoring contamination in water or the
ambient atmosphere (Ma et al., 1994). Small teleostomi are easily 2.1.4. Reporter-gene assay
fed and exposed to chemical toxicants in laboratories, and exhibit Various marker genes and proteins have become available in
similar toxic responses to those observed in mammals. Conse- bioassays to evaluate ePAHs by molecular biology-based studies on
quently, gill cells have been used to examine micronucleus fre- the mechanisms of their actions. Reporter-gene assays have also
quencies in order to evaluate environmental genotoxins (Hayashi been developed to more efficiently detect these markers through
et al., 1998). The genotoxic effects of PAHs in the ambient envi- the construction of plasmid vectors equipped with appropriate sets
ronment were previously assayed by flow cytometry, and the of reporter genes, such as those encoding b-galactosidase (the lacZ
findings obtained verified a significant increase in chromosomal gene product), luciferase, chloramphenicol acetyltransferase (CAT),
damage following exposure to sediments from a hazardous waste and green fluorescent protein (GFP), and regulatory sequences for
site (Matson et al., 2009). marker genes, such as the DRE and estrogen-responsive element
(ERE), which bind to the corresponding receptors. Since PAHs
2.1.3. Microbial test exhibit AhR-mediated activity, estrogenicity, and/or anti-
Microbes, such as Salmonella and luminescent bacteria, have androgenicity depending on the chemicals examined and cell
also been commonly used in in vitro assays to analyze the biological types used, various assay systems have been employed. For
activities of ePAHs. The Ames test is widely used to screen potential example, the growth inhibition of recombinant yeast containing
carcinogens and mutagens based on their mutational activities AhR was previously used to examine the dioxin-like activity of
(Ames et al., 1975). Histidine requirement strains of Salmonella ePAHs in estuary sediments, in which the lacZ gene was used as a
typhimurium, which cannot grow in histidine-free medium, are reporter (Puy-Azurmendi et al., 2010). Alternatively, a recombinant
814 Y. Zhang et al. / Environmental Pollution 213 (2016) 809e824

yeast strain, containing the human estrogen receptor (ER) gene and 2.1.7. Other assays
ERE-driven lacZ as a reporter gene, has been widely used to eval- More assays are used to assess the biological activities of ePAHs.
uate the estrogenic effects of ePAHs (Routledge and Sumpter, 1996). For example, the TCDD receptor affinity test was performed to
Exposure to estrogen induces lacZ gene expression and changes the evaluate biological effects of PAHs in exhaust emissions, which is
color of the medium from yellow to red. The estrogenicity and based on TCDD and PAHs having affinity to AhR (Westerholm and
dioxin-like activity of contaminated sediments have also been Egeba €ck, 1994). Furthermore, dermal contact with PAHs has been
examined using assays with mammalian cell lines containing assessed by quantifying their amounts with high-performance
luciferase as a reporter, the expression of which is regulated by ERE liquid chromatography (HPLC) after their direct absorption using
or DRE (Mazurova  et al., 2008). A previous study assessed the car- mounted adult human skin sections (Stroo et al., 2005). Other as-
cinogenicity of PAHs in bay sediments using a CYP reporter-gene says, such as ligand-binding assays, signaling pathway analyses,
system in order to determine whether ePAHs promoted tumori- and yeast two-hybrid assays have also been employed (see Section
genesis through the AhR pathway by inducing the expression of 3.2).
CYP1A1 (Koh et al., 2001). On the other hand, the mutagenic activity
of PAH-contaminated soils has been assessed by quantifying the 2.2. ePAHs as endocrine disruptors
rates of mutations in the lacZ gene in mouse FE1 cells (Lemieux
et al., 2015). There are significant differences between yeast and PAHs in the environment are regarded as endocrine disruptors
mammalian assays in the response to chemicals, such as the that interfere with the homeostasis of organisms by mimicking
discrimination between agonists and antagonists, and the differ- endogenous hormones. The best example is benzo[a]pyrene (B[a]
ences caused by membrane permeability, transport proteins and P), which has been identified as a contaminant in food, water,
signal transduction pathways (Kiyama and Wada-Kiyama, 2015). cigarette smoke, and vehicle exhaust fumes or in the body
following occupational exposure. For example, hepatocytes isolated
from male Wistar rats exposed to B[a]P for 72 h were examined for
2.1.5. Protein assay
thyroid hormone activity and related RNA expression profiles, and
A variety of qualitative and quantitative assays were developed
the findings obtained revealed that B[a]P reduced the metabolic
to detect proteins to evaluate endocrine disruptor action, such as
rate by breaking down thyroid hormone (Schraplau et al., 2015).
mass spectrometry, Western blotting, the enzyme-linked immu-
B[a]P is also toxic to the female reproductive system. When fe-
nosorbent assay (ELISA) and immunohistochemistry. Western
male scallops (Chlamys farreri) were exposed to B[a]P, the levels of
blotting has been used widely to quantify marker proteins. For
the sex steroids, 17b-estradiol, testosterone, and progesterone, in
example, AhR and ERa were used to evaluate the response to
the ovaries were significantly reduced in a dose-dependent
endocrine disruptor action (Safe and Wormke, 2003).
manner, and the expression of the estrogen-metabolizing en-
ELISA is also commonly used for protein analyses. For example,
zymes, hydroxysteroid dehydrogenases and CYP17, was also
ELISA has been employed to quantify the amounts of proteins, such
decreased, implying that steroidogenesis is restrained following
as vitellogenin, and examine the estrogenicity of ePAHs. For
exposure to B[a]P (Tian et al., 2013). A previous study reported that
example, ER-mediated vitellogenin production was previously
exposure to B[a]P increased plasma estrogen and progesterone
quantified by ELISA in order to monitor the toxicity of ePAHs in oil
levels as well as the expression of ERa in early pregnancy, whereas
production discharges, and the level of vitellogenin was found to
the expression of the progesterone receptor was reduced, and these
decrease with increases in the concentrations of ePAHs due to the
changes ultimately impaired endometrium receptivity and
presence of ER antagonists (Harman et al., 2010). ELISA has also
enhanced embryonic mortality (Zhao et al., 2014). Significant his-
been used to detect complex materials other than proteins, such as
topathological changes have also been observed in the ovary after
the 8-hydroxy-deoxyguanosine (8-OHdG) adduct, which is an in-
exposure to B[a]P in the postnatal period, such as an increase in the
dicator of oxidative stress and DNA damage (Hallberg et al., 2015).
total number of nonatretic antral follicles due to the thickening of
The quantification of enzymatic activities is another example of
theca cell layers and deficient apoptosis in granulosa cells (Kummer
how protein assays are used in the study of ePAHs. The CYP-
et al., 2013). Exposure to B[a]P may also induce endocrine disrup-
associated monooxygenase, EROD, belongs to the CYP family of
tions in the male reproductive system. For example, testosterone
enzymes, and its activity is used as a marker for the AhR-mediated
levels in the serum and intratesticular fluid as well as testosterone
toxicity of dioxin-like chemicals including PAHs (Pohl and Fouts,
production and the expression of related genes in Leydig cells were
1980; Kennedy and Jones, 1994). Thus, the activity of EROD is
examined in adult male Sprague-Dawley rats fed B[a]P for 90 days
often employed to evaluate ePAHs (see Table 2).
(Chung et al., 2011). The findings obtained suggested that the ste-
roidogenic acute regulatory protein (StAR) targeted by B[a]P
2.1.6. Transcription assay disturbed testosterone levels and degraded the quality of epidid-
Various transcription assays have been developed due to the ymal sperms.
recent progress in gene- or genome-based biotechnology, such as The impact of dietary exposure to B[a]P on the multigenera-
DNA microarray assay, next-generation sequencing-based RNA tional growth and development of animals has been examined.
sequencing (RNA-seq) and reverse transcription polymerase chain Adult zebrafish (F0) fed B[a]P were subjected to hatching in order to
reaction (RT-PCR). Several groups have developed DNA microarray obtain further generations and conduct multigenerational pheno-
assays for gene expression profiling of cancer cells treated with typic examinations, and mortality and premature hatching were
ePAHs/PAHs (Hockley et al., 2006, 2009; Líbalova  et al., 2014), observed more in the first filial generation (F1) of the treatment
because of the advantage of the assay for profiling more than group than in the control group, whereas no significant alterations
10,000 genes simultaneously. Statistically significant numbers of were detected in their descendants (F2, F3 and F4) and deformities
genes were up- or down-regulated by the treatment with B[a]P, were only noted in F1 (Corrales et al., 2014).
including those involved in xenobiotic metabolism, cell cycle Vertebrates have been used to examine the endocrine and
regulation and oxidative stress response (Hockley et al., 2006). The developmental effects of PAHs. Fish (polar or Atlantic cod) were
genes involved in signaling pathways, such as TGF-b and Wnt exposed to PAHs or ePAHs (dispersed oil) and their effects on the
pathways, responded to the treatment with the extractable organic endocrine and reproductive systems were analyzed by examining
 et al., 2014).
matters from respirable airborne particles (Líbalova phenotypes and endocrine markers such as plasma vitellogenin
Y. Zhang et al. / Environmental Pollution 213 (2016) 809e824 815

levels; limited, but distinct effects were observed in these studies, epigenetic regulation, cell cycle/DNA damage control, and cyto-
such as increases in vitellogenin levels, decreases in the number of skeletal/adhesion regulation), and cell functions (apoptosis, auto-
oocytes in females, and the inhibition of spermatogenesis in males phagy, immune responses/inflammation, neurological responses,
(Tollefsen et al., 2011; Geraudie et al., 2014). Zebrafish have also and development/differentiation), where PAHs, such as 2-
been used to assess the endocrine effects of PAHs. Zebrafish em- aminoanthracene, benz[a]anthracene (B[a]A), benzo[a]pyrene (B
bryos exposed to PAHs for 120 h showed distinct developmental [a]P), B[a]P-4,5-dihydroepoxide (BPE), B[a]P diol epoxide (BPDE),
effects, such as a dioxin-like phenotype, which may have been benzo[e]pyrene (B[e]P), benz[j]aceanthrylene (B[j]A), benzo[k]flu-
mediated through activation of the AhR pathway (Hawliczek et al., oranthene (B[k]F), benz[l]aceanthrylene (B[l]A), cyclopenta[c,d]
2012). Furthermore, juvenile mullets exposed to heavy fuel oil pyrene (CPP), dibenzo[a,l]pyrene (DB[al]P), 7,12-dimethylbenz[a]
containing ePAHs exhibited effects including transient neuroen- anthracene (DMBA), DMBA-3,4-diol, fluoranthene, fluorene, indeno
docrine effects, which may have negative impacts on reproduction, [1,2,3-c,d]pyrene (IND), 3-methylcholanthrene, 1-methylpyrene, 1-
behavior, and sex differentiation (Bilbao et al., 2010). The endocrine nitropyrene, perylene, phenanthrene, and pyrene are found in
effects of ePAHs have also been examined in the migratory bird, the sources, such as aerosol, air pollutants, asphalt fume, coal tar/coke/
Northern gannet by analyzing two hormones, prolactin and corti- tobacco/wood smoke particles, coastal sediments, diesel exhaust
costerone, which influence reproduction (Franci et al., 2014). particles (DEPs), electronic waste, and urban dust (Table 3).
Meanwhile, there were preliminary studies of the effects of ePAHs Various signaling pathways are involved in the biological ac-
on invertebrates by evaluating endocrine markers because they tivities induced or modulated by ePAHs/PAHs, which include
represent more than 95% of all animal species (Zapata-Perez et al., receptor-mediated pathways, growth factor/cytokine-signaling
2005), although there were few cases of mechanism-based pathways, and key-regulator/modulator-dependent pathways
comparisons. (Table 3). For example, B[a]A is involved in the ROS/MAPK pathway
It is important to note that difficulties are associated with in cell migration (Song et al., 2011), the PPARa/PPARb/d pathway in
obtaining clear endocrine effects in animal tests due to genetic carcinogenesis (Kim et al., 2005) and the AhR/NF-kB/IL-6 pathway
variations and detoxification activities in addition to the ethical in cytokine response (Kolasa et al., 2013). B[a]P is involved in the
issues linked to the use of animals for bioassays; therefore, in vitro MAPK/JNK pathway in stress response (Jarvis et al., 2014), p53-
assays need to be developed, are expected to replace animal assays dependent apoptosis via FOXO3a (Al-Anati et al., 2014), cell cycle/
with equivalent quality, sensitivity, and reliability, and include DNA damage control in cell proliferation (Volkov et al., 2012; Guo
pathway-based assays as described below. et al., 2000), cell adhesion regulation in macrophage differentia-
tion or hemocyte immunomodulation (van Grevenynghe et al.,
3. Mechanisms underlying biological effects induced by 2003; Croxton et al., 2012), microtubule dynamics (Deng et al.,
ePAHs and constituent PAHs 2014), cytokine response associated with asthma (Ple  et al., 2015),
TGF-b-induced pro-inflammatory response (Curfs et al., 2005),
Recent technological advances have provided a clearer insight TLRs-induced immune response (Do et al., 2012), Parkinson's dis-
into the mechanisms responsible for the biological effects induced ease (Konstandi et al., 2007), and development/differentiation,
by ePAHs and constituent PAHs. PAHs are known to stimulate AhR- such as neurodevelopment (Wormley et al., 2004), atherogenesis
mediated cell signaling, and their mechanisms of actions have been (Knaapen et al., 2007) and hematopoiesis (N'jai et al., 2011). BPDE is
examined extensively (Hankinson, 1995; Tian, 2009). AhR is a involved in cell functions, such as chromatin structure in carcino-
ligand-activated transcription factor that binds to planar aromatic genesis (Feng et al., 2003), G1/S cell cycle regulation (Mukherjee
hydrocarbons, such as flavonoids, polyphenols, indoles, and the and Kumar, 2013), and T-dependent antibody response (Li et al.,
polyaromatic and halogenated aromatic hydrocarbons of natural 2010). DMBA is involved in PPARb/d-AhR crosstalk in carcinogen-
and industrial origins. Previous studies described the association or esis (Borland et al., 2014), and the Wnt/b-catenin and other path-
crosstalk between AhR and various cell signaling pathways, such as ways in carcinogenesis (Currier et al., 2005), and cell functions such
mitogen-activated protein kinase (MAPK), NF-kB, and cell cycle/ as pre-B cell apoptosis (Mann et al., 2001), tumor necrosis factor a
apoptosis-related pathways (Schmidt and Bradfield, 1996; Puga (TNF-a)-dependent bone marrow toxicity (Page et al., 2004), T-
et al., 2002, 2009; Henklova  et al., 2008; Tian, 2009). Since AhR is dependent antibody response (Li et al., 2010), and development/
not a direct regulator of hormonal actions, the mechanisms un- differentiation, such as B lymphopoiesis (Mann et al., 2001) and
derlying the endocrine-disrupting effects of AhR and its ligands hematopoiesis (N'jai et al., 2011). 3-Methylcholanthrene is involved
require the involvement of hormone receptors and/or regulators in signaling pathways such as TGF-b signaling in hepatocyte func-
(Murphy et al., 2007). Since estrogen and estrogenic chemicals are tions (Abdel-Razzak et al., 1994); cell functions, such as chromatin
an important category of endocrine modulators and sometimes act condensation in apoptosis (Reynaud et al., 2004) and cell adhesion
as endocrine disruptors, the involvement of estrogen signaling regulation during epithelial-mesenchymal transition (Oh et al.,
associated with AhR-mediated cell signaling stimulated with PAHs 2014); and in diseases, such as rheumatoid arthritis (Tamaki
has been investigated in an attempt to elucidate the mechanisms et al., 2004).
responsible for their biological effects (Safe et al., 2000; On the other hand, ePAHs from various sources are involved in
Swedenborg and Pongratz, 2010; Feng et al., 2013). signaling pathways, such as the MAPK/AKT (Sumanasekera et al.,
In this section, we summarized the cell signaling pathways 2007), NF-kB/MAPK (Bonvallot et al., 2001), IGF-1 (Xu et al.,
stimulated by ePAHs and PAHs. We then focused on the estrogen 2013), insulin (Choi et al., 2015) pathways; cell functions, such as
activities of ePAHs and PAHs in order to elucidate the mechanisms DNA methylation (Liu et al., 2013; Yan et al., 2014; Hew et al., 2015),
underlying the biological effects of ePAHs as endocrine disruptors. histone modification (Klingbeil et al., 2014), FASL-dependent
apoptosis (Crew et al., 2007), infectious response (Ma and Ma,
3.1. Pathways mediating cell signaling induced by ePAHs/PAHs 2002), DNA damage response (Mahadevan et al., 2005; Duan
et al., 2009; Foresta et al., 2014), actin function (Tuominen et al.,
The biological activities of and signaling pathways stimulated 2003; Hooven and Baird, 2008), cell adhesion (Wu et al., 2011),
and/or mediated by ePAHs/PAHs are summarized in Table 3. These cytokine response (Ple  et al., 2015) and inflammatory response
activities are categorized by specific signaling pathways (MAPK and (Gate et al., 2006; Totlandsdal et al., 2014; Wittkopp et al., 2013;
other signaling pathways), regulatory mechanisms (chromatin/ Yang et al., 2016; Danielsen et al., 2011; Totlandsdal et al., 2015;
Table 3
Biological activities of and cell signaling by ePAHs/PAHs.

Pathway/cascade/ ePAH/PAH Reference


function

MAPK signaling
MAPK/AKT/NO ePAHs (DEPs) Sumanasekera et al., 2007
MAPK/JNK B[a]P Jarvis et al., 2014
NF-kB/MAPK ePAHs (DEPs) Bonvallot et al., 2001
ROS/MAPK B[a]A/B[k]F/IND Song et al., 2011
Other signaling (see Table 4 for ER signaling)
PPARa/PPARb/d B[a]A Kim et al., 2005
PPARb/d/AhR DMBA Borland et al., 2014
crosstalk
Chromatin/epigenetic regulation
Chromatin 3-Methylcholanthrene Reynaud et al., 2004
condensation
Chromatin BPDE Feng et al., 2003
structure
DNA methylation ePAHs (aerosol, air pollutant), Ph Liu et al., 2013; Yan et al., 2014; Hew et al., 2015
Histone ePAHs (aerosol) Klingbeil et al., 2014 (review)
modification
Apoptosis
FASL-dependent ePAHs (tobacco/DNA adduct) Crew et al., 2007
apoptosis
Infectious response ePAHs (DEPs) Ma and Ma, 2002
p53-dependent B[a]P/B[j]A/B[l]A/CPP/DB[a,l]P/Fla/1-Nitropyrene Solhaug et al., 2004; Su et al., 2008; Pru et al., 2009; Huang et al., 2012; Al-Anati et al., 2014
apoptosis
Autophagy
Starvation stress Ph McVeigh et al., 2006
Cellular metabolism
AMPK signaling 1-Nitropyrene Podechard et al., 2011
IGF-1 signaling ePAHs (electronic waste) Xu et al., 2013
Insulin resistance ePAHs (air pollutant) Choi et al., 2015
Insulin/AMPK 2-Aminoanthracene Mattis et al., 2014
signaling
Cell cycle/DNA damage
G1/S cell cycle B[a]P/BPDE Volkov et al., 2012; Mukherjee & Kumar, 2013
regulation
G2/M DNA damage B[a]P/B[a]P-7,8-diol/DMBA Guo et al., 2000; Caino et al., 2007; Nogueira et al., 2009
checkpoint
DNA damage ePAHs (coke/tobacco/urban dust) Mahadevan et al., 2005; Duan et al., 2009; Foresta et al., 2014
response
Cytoskeletal regulation and adhesion
Actin function B[a]P/BPDE/BPE, ePAHs (coal tar/DEPs/tobacco) Tuominen et al., 2003; Hooven and Baird, 2008
Cell adhesion B[a]P/Fla/Fluorene/3-Methylcholanthrene/1- van Grevenynghe et al., 2003; Pillai et al., 2003; Bahri et al., 2010; Wu et al., 2011; Croxton
Methylpyrene/Perylene/Ph, ePAHs (coke) et al., 2012; Oh et al., 2014
Microtubule B[a]P Deng et al., 2014
dynamics
Immunology and inflammation
B-cell development DMBA Allan et al., 2003; Ryu et al., 2003; Ryu et al., 2005
Cytokine response B[a]A/B[a]P/DMBA/Fla/3-Nitrobenzanthrone/3- Page et al., 2004; Ovrevik et al., 2010; Kolasa et al., 2013; Hao and Whitelaw, 2013; Kaba tkov a
Nitro-Fla/1-Nitropyrene/Ph/Pyrene, ePAHs (DEPs)  et al., 2015
et al., 2014; Ple
Inflammatory B[a]P/B[e]P, ePAHs (asphalt fume/DEPs/wood smoke Curfs et al., 2005; Gate et al., 2006; den Hartigh et al., 2010; Danielsen et al., 2011; Tsuji et al.,
response particles) 2012; Wittkopp et al., 2013; Totlandsdal et al., 2014; Yang et al., 2016; Totlandsdal et al., 2015
Rheumatoid 3-Methylcholanthrene Tamaki et al., 2004
arthritis
T-dependent B[a]P-7,8-diol/BPDE/DB[a,l]P/Dibenzo[def,p] Li et al., 2010; Lauer et al., 2013
antibody response chrysene/DMBA/DMBA-3,4-diol
TLRs-induced B[a]P Do et al., 2012
immune response
Neuroscience
Behavior/brain 16 PAH mixture peaux et al., 2012
Cre
metabolism
Neurodevelopment B[a]P/Pyrene Wormley et al., 2004; He et al., 2012
Parkinson's disease B[a]P Konstandi et al., 2007
Development and differentiation
Atherogenesis B[a]P Knaapen et al., 2007
B lymphopoiesis DMBA Mann et al., 2001
Development B[a]P/Fla, ePAHs (coastal sediments) Kerambrun et al., 2012; Jayasundara et al., 2015
Differentiation B[a]P/DMBA/DMBA-3,4-diol Ge et al., 2012; Le Vee et al., 2014
Hematopoiesis B[a]P/DMBA N'jai et al., 2011
TGF-b signaling 3-Methylcholanthrene, ePAHs (DEPs) Abdel-Razzak et al., 1994; Pacheco et al., 2001
Wnt/b-catenin DMBA Currier et al., 2005
signaling

The classification of pathways/cascades/functions was described in Kiyama and Zhu (2014). Only representative chemicals are listed for each category. Abbreviations: AhR:
aryl hydrocarbon receptor; AMPK: AMP-dependent protein kinase; B[a]A: benz[a]anthracene; B[a]P: benzo[a]pyrene; B[e]P: benzo[e]pyrene; B[j]A: benz[j]aceanthrylene; B
[k]F: benzo[k]fluoranthene; B[l]A: benz[l]aceanthrylene; BPDE: benzo[a]pyrene diol epoxide; BPE: benzo[a]pyrene-4,5-dihydroepoxide; CPP: cyclopenta[c,d]pyrene; DB[a,l]P:
dibenzo[a,l]pyrene; DEP: diesel exhaust particle; DMBA: 7,12-dimethylbenz[a]anthracene; ER: estrogen receptor; FASL: Fas ligand; Fla: fluoranthene; IGF-1: insulin-like
growth factor 1; IND: indeno[1,2,3-c,d]pyrene; JNK: c-Jun N-terminal kinase; MAPK: mitogen-activated protein kinase; NO: nitric oxide; ROS: reactive oxygen species; Ph:
phenanthrene; PI3K: phosphoinositide 3-kinase; PPAR: peroxisome proliferator-activated receptor.
Y. Zhang et al. / Environmental Pollution 213 (2016) 809e824 817

den Hartigh et al., 2010); and development and differentiation, analysis, and cell viability assay), ligand-binding assays, protein
such as TGF-b signaling (Kerambrun et al., 2012). assays (e.g., Western blotting and immunoassay), reporter-gene
assays (e.g., CALUX assay, MVLN-cell assay, and YES assay),
3.2. Estrogenic activity of ePAHs/PAHs signaling pathway analyses, transcription assays (e.g., RT-PCR and
DNA microarrays), and yeast two-hybrid assays, the time, cost,
In an attempt to more clearly understand the endocrine dis- sensitivity, accuracy/reliability, safety/ethics, and purpose of which
ruptor activities of ePAHs/PAHs, we searched for estrogenic PAHs vary (Kiyama and Zhu, 2014; Kiyama and Wada-Kiyama, 2015). For
and their signaling pathways (Table 4). Various PAHs, such as example, Arcaro et al. reported that PAH fractions extracted from
anthraquinone, B[a]A, B[a]P, chrysene, naphthalene, and pyrene, environmental samples showed anti-estrogenic activity by ER-
and their hydroxyl products, including alizarin, 9,10-dihydroxy-B[a] binding and estrogen-metabolism assays, and suggested that the
P, 2-hydroxyanthraquinone, 3-hydroxy-B[a]P, 7-hydroxy-B[a]P, 9- competition of PAHs against the binding of estrogen to the ER and
hydroxy-B[a]P, 2-hydroxychrysene, 2-hydroxyphenanthrene, 9- the metabolic degradation of estrogen are the mechanisms for the
hydroxyphenanthrene, 1-hydroxypyrene, 1-naphthol, and 2- activity (Arcaro et al., 1999). Meanwhile, a reporter-gene assay us-
naphthol, were previously reported to be estrogenic (agonistic) or ing human breast cancer MCF-7 cells showed that some PAHs, such
anti-estrogenic (antagonistic), and signaling pathways, such as as B[a]P and B[a]A, were weak inducers of estrogenic activity
those involving AhR, AKT, BRCA1, MAPK, and phosphoinositide 3- (Vondra cek et al., 2002; Plískov a et al., 2005).
kinase (PI3K), are known to be involved in estrogen signaling. The Several groups reported differences in estrogenicity between
assays used to examine estrogenicity include animal tests, cell as- PAHs (non-estrogenic) and hydroxyl PAHs (estrogenic), and/or in
says (e.g., cell migration assay, cell proliferation assay, cell-shape the estrogenicity of the same PAHs among the different assays or

Table 4
Estrogenic PAHs and associated signaling pathways.

Chemical Signaling pathway Reference (assaya)

PAH
Alizarin, 2-hydroxyanthraquinone ER Kurihara et al., 2005 (Y)
Anthraquinone, benz[a]acridine, B[a]A -7,12-dione, benzanthrone (estrogenic) ER Machala et al., 2001 (R)
B[a]A (estrogenic) AhR/PKCs/ERK/ERa Kolasa et al., 2012 (S)
B[a]A, B[a]P, B[b]F, B[c]Ph, monohydroxy PAHs (agonistic) ERa Fertuck et al., 2001 (L, R)
B[a]A, B[a]P, Fla (estrogenic); B[k]F (anti-estrogenic) ER Kummer et al., 2008 (A)
B[a]A, B[a]P, DMBA, 3-methylcholanthrene (anti-estrogenic) AhR/ER Chaloupka et al., 1992 (C, P)
B[a]A, dibenz[a,h]anthracene (estrogenic) ER Villeneuve et al., 2002 (R)
B[a]F (estrogenic) ERa Kim et al., 2011b (A, P)
B[a]P, 3-hydroxy-B[a]P, 9-hydroxy-B[a]P, 9,10-dihydroxy-B[a]P (estrogenic) ERa Charles et al., 2000 (R)
B[a]P (anti-estrogenic) AhR/CYP19/ER Patel et al., 2006 (A, P, T)
B[a]P (anti-estrogenic) ER Booc et al., 2014 (A)
B[a]P (estrogenic) ER/MAPK, PI3K/AKT/COX-2 Tsai et al., 2004 (S)
B[a]P (anti-estrogenic) AhR/ER/BRCA1 Jeffy et al., 2002 (C, R, T)
B[a]P, B[b]F, B[k]F, chrysene, dibenz[a,h]anthracene, IND (anti-estrogenic) ER Arcaro et al., 1999 (C, L)
B[a]P, 2,3-benzofluorene, dibenz[a,h]anthracene, 6-hydroxychrysene (anti-estrogenic) ER Tran et al., 1996 (L, R)
B[a]A, B[a]P, dibenz[a,h]anthracene (antagonistic) ER Ohta et al., 2012 (A)
DMBA þ E2 (estrogenic) AhR/CYP1A1/ER Singhal et al., 2008 (A, P, T)
DMBA þ naringenin (anti-estrogenic) AhR/CYP1B1/ER Poon et al., 2013 (P, R, T)
7-Hydroxy-B[a]P, 1-hydroxypyrene (estrogenic) ERa Van de Wiele et al., 2005 (R)
2-Hydroxychrysene, 2-hydroxy-Ph, 1-hydroxypyrene, 1-naphthol, 2-naphthol (estrogenic) ER Wenger et al., 2009 (R)
2-Hydroxyfluorene, 2-hydroxy- Ph, 3-hydroxy-Ph, 1-hydroxypyrene (estrogenic) ER Kamiya et al., 2005 (Y)
9-Hydroxy-Ph, 1-hyroxypyrene, 1-naphthol (estrogenic) ERb Sievers et al., 2013 (L, P, R, T)
3-Methylcholanthrene þ E2 (estrogenic) AhR/CYP1A/ER Navas and Segner, 2000 (P)
3-Methylcholanthrene (estrogenic) ERa Abdelrahim et al., 2006 (L, R, T)
Naphthalene (anti-estrogenic) ER Tintos et al., 2006 (A)
6-Nitrochrysene (anti-estrogenic) AhR/ERa Sun et al., 2013 (A, P)
2-Nitrofluorene (estrogenic) ER Fujimoto et al., 2003 (R)
ePAH
Ambient particulate matter (estrogenic) ER Wenger et al., 2009 (R) (see above)
Bay sediments (estrogenic) ER Koh et al., 2002 (R)
Charcoal flue gas (estrogenic) ER Muthumbi et al., 2003 (R)
Cigarette smoke (estrogenic) ER Kamiya et al., 2005 (Y) (see above)
Coastal sediments (estrogenic/anti-estrogenic) ER Koh et al., 2005 (R)
Coastal sediments/water (estrogenic) ER Weston et al., 2010 (R)
Coastal/river sediments (estrogenic) ER David et al., 2010 (R)
Contaminated oil (anti-estrogenic) ER Aarab et al., 2004 (A, P)
Dispersed oil (anti-estrogenic) ER Tollefsen et al., 2011 (L, P)
Lagoon sediments (estrogenic) ER Amaeze et al., 2015 (R)
Lake sediments (estrogenic) ER Mazurova  et al., 2008 (R)
Lake sediments (estrogenic) ER Garcia-Reyero et al., 2005 (R)
Marine sediments (antagonistic) ER Grung et al., 2011 (R)
Produced water (anti-estrogenic) ER Harman et al., 2010 (A, P)
River sediments (anti-estrogenic) ER Arcaro et al., 1999 (C, L) (see above)
River sediments (estrogenic) ER Hilscherova et al., 2002 (R)
a
Abbreviations for assays are: animal test (A), cell assay (C), ligand-binding assay (L), protein assay (P), reporter-gene assay (R), signaling pathway analysis (S), transcription
assay (T), and yeast two-hybrid assay (Y) (see Kiyama and Wada-Kiyama, 2015). B[a]A: benz[a]anthracene; B[a]P: benzo[a]pyrene; B[b]F: benzo[b]fluoranthene; B[c]Ph: benzo
[c]phenanthrene; B[k]F: benzo[k]fluoranthene; DMBA: 7,12-dimethylbenz[a]anthracene; ERK: extracellular signal-regulated kinase; Fla: fluoranthene; IND: indeno[1,2,3-c,d]
pyrene; Ph: phenanthrene; PKC: protein kinase C.
818 Y. Zhang et al. / Environmental Pollution 213 (2016) 809e824

tissues used (Tran et al., 1996; Arcaro et al., 1999; Fertuck et al., produced water, and contaminated/dispersed oil (Table 4). PAHs are
2001; Machala et al., 2001; Van de Wiele et al., 2005; Ohta et al., estrogenic or anti-estrogenic, depending on the types and locations
2012). These differences may be explained by non-hydroxylated of the samples. Although the mechanisms responsible for their
PAHs being metabolized to estrogenic hydroxyl PAHs by PAH- activities currently remain unclear, several possibilities have been
inducible CYP enzymes such as aromatase (CYP19), AhR agonists suggested, such as a direct interaction between PAHs and ERs (es-
indirectly interfering with ER-responsive transactivation functions trogenic), the AhR-mediated suppression of estrogenic activity
or directly competing with estrogen, and differences in metabolism (anti-estrogenic), and the modulation of estrogen or other signaling
or bioaccumulation in tissues or cells, thereby changing the status pathways (estrogenic or anti-estrogenic), which are based on an-
of the estrogenicity of PAHs in cells. The contribution of hydroxyl alyses of specific PAHs (see Section 3.2). For example, Koh et al.
PAHs to the estrogenic activity of environmental samples is sup- found that bay sediments were estrogenic and attributed this to the
ported by the yeast two-hybrid estrogen assay, in which hydroxyl estrogenicity of B[a]A and dibenz[a,h]anthracene (Koh et al., 2002).
PAHs, such as 4-hydroxy-B[a]A, 3-hydroxy-B[a]A, and 2- This relationship between estrogenicity and PAHs has been
hydroxychrysense, showed estrogenic activity, whereas their cor- observed in other sediments (Garcia-Reyero et al., 2005; Mazurova 
responding PAHs did not (Hayakawa et al., 2008). However, the et al., 2008). However, only a limited number of environmental
metabolic activation of PAHs is not regulated by ERs in estrogen- samples have exhibited clear estrogenicity (Muthumbi et al., 2003;
irresponsive, but ER-expressing tissues such as the lung (Berge Amaeze et al., 2015) or anti-estrogenicity (Harman et al., 2010).
et al., 2004). Furthermore, metabolites of PAHs, such as B[b]F, B[k] Such inconsistent findings on estrogenicity have been obtained for
F, chrysene, and IND, were previously shown to be anti-estrogenic other estrogenic chemicals, such as selective ER modulators
in human breast cancer MCF-7 cells (Arcaro et al., 1999), and the (Kiyama and Wada-Kiyama, 2015). Wenger et al. found that only
same PAHs examined by the same types of assays (B[a]A and B[a]P 0.01e0.2% of the overall estrogenic activity of atmospheric partic-
by animal tests) (Ohta et al., 2012) or the same PAHs (for example ulate matter was attributed to the major PAHs, 2-
DMBA) mixed with different types of estrogens (E2 and naringenin, hydroxyphenanthrene, 2-naphthol and 1-naphthol, and suggested
Table 4; Singhal et al., 2008; Poon et al., 2013), gave opposite ac- the presence of other estrogenic compounds not yet identified
tivities, suggesting the presence of other mechanisms and/or the (Wenger et al., 2009). However, Hilscherova et al. indicated that the
contribution of metabolites other than hydroxyl PAHs to contribution of alkylphenols to estrogenicity in river sediments was
estrogenicity. relatively small (Hilscherova et al., 2002). Thus, standardized assay
Cases of complicated and unexplainable estrogen actions have protocols based on pathway-based assessments are needed in or-
been reported as follows. While B[a]A exposure reduced estradiol der to overcome these inconsistencies (Kiyama and Wada-Kiyama,
concentrations in fish, no effect was observed in the endocrine 2015).
system (Booc et al., 2014). Hydroxy PAHs, such as 9- Since a number of cells do not have hormone receptors, there
hydroxyphenanthrene, 1-hydroxypyrene, and 1-naphthol, showed should be a mechanism of communication of signaling, or signal
even differential actions between ERb (estrogenic) and ERa (non- crosstalk, between hormone and other signaling pathways. Among
estrogenic) (Sievers et al., 2013). So, the balance of the concentra- the cases of signal crosstalk, the one between the AhR and ER has
tions of estrogenic and anti-estrogenic PAHs (Hilscherova et al., been extensively studied (Safe and Wormke, 2003); inhibitory
2002) or the contribution of other compounds not yet identified crosstalk between AhR and ER is explained at the gene expression
by chemical analysis (Wenger et al., 2009) is important. Further- level as well as by the direct interaction of receptors/signal medi-
more, the activation of ERa was found to occur through a direct ators and metabolic degradation of estrogen. In addition, since the
interaction between 3-methylcholanthrene and its receptor in AhR has been reported to have association with other hormone-
MCF-7 cells and was independent of AhR (Abdelrahim et al., 2006). signaling pathways, such as androgen and thyroid hormone
There are also cases in which the contribution of estrogen to the signaling pathways (Ohtake et al., 2011; Murphy et al., 2007), the
overall responses of environmental samples containing PAHs was involvement of ePAHs/PAHs in the mechanisms of endocrine dis-
low (Vondra cek et al., 2001) and other estrogenic chemicals ruptor action will be studied with significant attention.
appeared to contribute to this activity (David et al., 2010). The
contradictory and unexplainable effects observed for ePAHs were 4. Conclusions
observed not only because of unreproducible and unreliable results
due to technical limitation/immaturity, but also because of the lack We here summarized the biological activities of ePAHs as
of knowledge about the mechanisms and methods to solve such follows.
problems. We emphasized here the importance of signaling path-
ways to understand the endocrine disruptor action of ePAHs/PAHs (1) ePAHs are found in the atmosphere, sediments, soils, and
because the outcome-based assessment of the action, such as cell water as a result of human activities, accidents, or natural
proliferation, could be influenced by various conditions, such as the phenomena, and are categorized by their sources and forms,
directions of signaling pathways controlled by the amounts of re- followed by their biological effects and social impact.
ceptors and signal mediators within the cell and the balance among (2) ePAHs and their constitutent PAHs have been analyzed by
cell functions attributable to various environmental factors, which bioassays, such as animal/plant tests, cell assays, microbial
could result in quite different outcomes. At the level of signaling tests, reporter-gene assays, and other assays, which have
pathways, we could monitor the outcome of signaling qualitatively demonstrated that ePAHs/PAHs affect the endocrine systems
and quantitatively, and thus, we could obtain more reproducible of humans and animals.
and reliable results, which can be used in risk assessment. (3) The mechanisms underlying endocrine disruption induced
by ePAHs/PAHs have been studied based on cell signaling
3.3. Estrogenic cell signaling induced by ePAHs/PAHs pathways, such as the MAPK pathway, regulatory mecha-
nisms, including chromatin/epigenetic regulation, cell cycle/
Estrogenic activity has been detected in ePAHs/PAHs, such as DNA damage control, and cytoskeletal/adhesion regulation,
those extracted from the sediments of various sources (bays, coasts, and cell functions, such as apoptosis, autophagy, immune
lagoons, lakes, rivers, and other marine sites), and from ambient responses/inflammation, neurological responses, and devel-
particulate matter, charcoal flue gas, cigarette smoke, coastal/ opment/differentiation.
Y. Zhang et al. / Environmental Pollution 213 (2016) 809e824 819

(4) Estrogen signaling has been examined in association with Binet, S., Pfohl-Leszkowicz, A., Brandt, H., Lafontaine, M., Castegnaro, M., 2002.
Bitumen fumes: review of work on the potential risk to workers and the pre-
the endocrine-disrupting activities induced by ePAHs/PAHs,
sent knowledge on its origin. Sci. Total Environ. 300 (1e3), 37e49.
which involve receptors, such as ERs and AhR. Boffetta, P., Jourenkova, N., Gustavsson, P., 1997. Cancer risk from occupational and
(5) Due to sensitivity, reliability, accessibility (such as time and environmental exposure to polycyclic aromatic hydrocarbons. Cancer Causes
cost), and ethical reasons, pathway-based in vitro assays are Control 8 (3), 444e472.
Bonvallot, V., Baeza-Squiban, A., Baulig, A., Brulant, S., Boland, S., Muzeau, F.,
becoming more important, and standardized protocols are Barouki, R., Marano, F., 2001. Organic compounds from diesel exhaust particles
needed in order to minimize inconsistencies. elicit a proinflammatory response in human airway epithelial cells and induce
cytochrome p450 1A1 expression. Am. J. Respir. Cell. Mol. Biol. 25 (4), 515e521.
Booc, F., Thornton, C., Lister, A., MacLatchy, D., Willett, K.L., 2014. Benzo[a]pyrene
Acknowledgments effects on reproductive endpoints in Fundulus heteroclitus. Toxicol. Sci. 140 (1),
73e82.
Bopp, S.K., McLachlan, M.S., Schirmer, K., 2007. Passive sampler for combined
This research was supported partly by a Knowledge Cluster chemical and toxicological long-term monitoring of groundwater: the ceramic
Initiative program and a Grant-in-Aid for Basic Areas from the toximeter. Environ. Sci. Technol. 41 (19), 6868e6876.
Ministry of Education, Culture, Sports, Science and Technology of Borland, M.G., Krishnan, P., Lee, C., Albrecht, P.P., Shan, W., Bility, M.T., Marcus, C.B.,
Lin, J.M., Amin, S., Gonzalez, F.J., Perdew, G.H., Peters, J.M., 2014. Modulation of
Japan, and by the Guideline Program for Medical Device Develop- aryl hydrocarbon receptor (AHR)-dependent signaling by peroxisome
ment from the Ministry of Economy, Trade and Industry of Japan. proliferator-activated receptor b/d (PPARb/d) in keratinocytes. Carcinogenesis
35 (7), 1602e1612.
Bostro€m, C.E., Gerde, P., Hanberg, A., Jernstro € m, B., Johansson, C., Kyrklund, T.,
References Rannug, A., To €rnqvist, M., Victorin, K., Westerholm, R., 2002. Cancer risk
assessment, indicators, and guidelines for polycyclic aromatic hydrocarbons in
Aarab, N., Minier, C., Lemaire, S., Unruh, E., Hansen, P.D., Larsen, B.K., Andersen, O.K., the ambient air. Environ. Health Perspect. 110 (Suppl. 3), 451e488.
Narbonne, J.F., 2004. Biochemical and histological responses in mussel (Mytilus Brooks, L.R., Hughes, T.J., Claxton, L.D., Austern, B., Brenner, R., Kremer, F., 1998.
edulis) exposed to North Sea oil and to a mixture of North Sea oil and alkyl- Bioassay-directed fractionation and chemical identification of mutagens in
phenols. Mar. Environ. Res. 58 (2e5), 437e441. bioremediated soils. Environ. Health Perspect. 106 (Suppl. 6), 1435e1440.
Abdelrahim, M., Ariazi, E., Kim, K., Khan, S., Barhoumi, R., Burghardt, R., Liu, S., Buehler, S.S., Hites, R.A., 2002. The Great Lakes' integrated atmospheric deposition
Hill, D., Finnell, R., Wlodarczyk, B., Jordan, V.C., Safe, S., 2006. 3- network. Environ. Sci. Technol. 36 (17), 354Ae359A.
Methylcholanthrene and other aryl hydrocarbon receptor agonists directly Buratti, M., Campo, L., Fustinoni, S., Cirla, P.E., Martinotti, I., Cavallo, D., Foa, V., 2007.
activate estrogen receptor alpha. Cancer Res. 66 (4), 2459e2467. Urinary hydroxylated metabolites of polycyclic aromatic hydrocarbons as bio-
Abdel-Razzak, Z., Corcos, L., Fautrel, A., Campion, J.P., Guillouzo, A., 1994. Trans- markers of exposure in asphalt workers. Biomarkers 12 (3), 221e239.
forming growth factor-beta 1 down-regulates basal and polycyclic aromatic Caino, M.C., Oliva, J.L., Jiang, H., Penning, T.M., Kazanietz, M.G., 2007. Benzo[a]pyr-
hydrocarbon-induced cytochromes P-450 1A1 and 1A2 in adult human hepa- ene-7,8-dihydrodiol promotes checkpoint activation and G2/M arrest in human
tocytes in primary culture. Mol. Pharmacol. 46 (6), 1100e1110. bronchoalveolar carcinoma H358 cells. Mol. Pharmacol. 71 (3), 744e750.
Ake, C.L., Wiles, M.C., Huebner, H.J., McDonald, T.J., Cosgriff, D., Richardson, M.B., Caliani, I., Porcelloni, S., Mori, G., Frenzilli, G., Ferraro, M., Marsili, L., Casini, S.,
Donnelly, K.C., Phillips, T.D., 2003. Porous organoclay composite for the sorption Fossi, M.C., 2009. Genotoxic effects of produced waters in mosquito fish
of polycyclic aromatic hydrocarbons and pentachlorophenol from groundwater. (Gambusia affinis). Ecotoxicology 18 (1), 75e80.
Chemosphere 51 (9), 835e844. Chaloupka, K., Krishnan, V., Safe, S., 1992. Polynuclear aromatic hydrocarbon car-
Al-Anati, L., Kadekar, S., Ho€gberg, J., Stenius, U., 2014. PCB153, TCDD and estradiol cinogens as antiestrogens in MCF-7 human breast cancer cells: role of the Ah
compromise the benzo[a]pyrene-induced p53-response via FoxO3a. Chem. Biol. receptor. Carcinogenesis 13 (12), 2233e2239.
Interact. 219, 159e167. Charles, G.D., Bartels, M.J., Zacharewski, T.R., Gollapudi, B.B., Freshour, N.L.,
Allan, L.L., Mann, K.K., Matulka, R.A., Ryu, H.Y., Schlezinger, J.J., Sherr, D.H., 2003. Carney, E.W., 2000. Activity of benzo[a]pyrene and its hydroxylated metabolites
Bone marrow stromal-B cell interactions in polycyclic aromatic hydrocarbon- in an estrogen receptor-alpha reporter gene assay. Toxicol. Sci. 55 (2), 320e326.
induced pro/pre-B cell apoptosis. Toxicol. Sci. 76 (2), 357e365. Charrois, J.W., McGill, W.B., Froese, K.L., 2001. Acute ecotoxicity of creosote-
Amaeze, N.H., Schnell, S., Sozeri, O., Otitoloju, A.A., Egonmwan, R.I., Arlt, V.M., contaminated soils to Eisenia fetida: a survival-based approach. Environ. Tox-
Bury, N.R., 2015. Cytotoxic and genotoxic responses of the RTgill-W1 fish cells in icol. Chem. 20 (11), 2594e2603.
combination with the yeast oestrogen screen to determine the sediment quality Chen, G., Qin, J., Shi, D., Zhang, Y., Ji, W., 2009. Diversity of soil nematodes in areas
of Lagos lagoon, Nigeria. Mutagenesis 30 (1), 117e127. polluted with heavy metals and polycyclic aromatic hydrocarbons (PAHs) in
Ames, B.N., Mccann, J., Yamasaki, E., 1975. Methods for detecting carcinogens and Lanzhou, China. Environ. Manag. 44 (1), 163e172.
mutagens with the Salmonella/mammalian-microsome mutagenicity test. Chen, G., White, P.A., 2004. The mutagenic hazards of aquatic sediments: a review.
Mutat. Res. 31 (6), 347e364. Mutat. Res. 567 (2e3), 151e225.
Andersson, E., Rotander, A., von Kronhelm, T., Berggren, A., Ivarsson, P., Hollert, H., Choi, Y.H., Kim, J.H., Hong, Y.C., 2015. Sex-dependent and body weight-dependent
Engwall, M., 2009. AhR agonist and genotoxicant bioavailability in a PAH- associations between environmental PAHs exposure and insulin resistance:
contaminated soil undergoing biological treatment. Environ. Sci. Pollut. Res. Korean urban elderly panel. J. Epidemiol. Community Health 69 (7), 625e631.
Int. 16 (5), 521e530. Chung, J.Y., Kim, Y.J., Kim, J.Y., Lee, S.G., Park, J.E., Kim, W.R., Yoon, Y.D., Yoo, K.S.,
Annavarapu, S., Foran, C.M., Gardinali, P., Metzger, C., Willett, K.L., 2004. Comparison Yoo, Y.H., Kim, J.M., 2011. Benzo[a]pyrene reduces testosterone production in rat
of two sites in Mobile Bay using in vivo biomarkers in largemouth bass, sedi- Leydig cells via a direct disturbance of testicular steroidogenic machinery. En-
ment bioassays, and sediment contaminant analysis. Arch. Environ. Contam. viron. Health Perspect. 119 (11), 1569e1574.
Toxicol. 46 (4), 502e510. Ciganek, M., Neca, J., Adamec, V., Janosek, J., Machala, M., 2004. A combined
Arcaro, K.F., O'Keefe, P.W., Yang, Y., Clayton, W., Gierthy, J.F., 1999. Antiestrogenicity chemical and bioassay analysis of traffic-emitted polycyclic aromatic hydro-
of environmental polycyclic aromatic hydrocarbons in human breast cancer carbons. Sci. Total Environ. 334e335, 141e148.
cells. Toxicology 133 (2e3), 115e127. Cofield, N., Banks, M.K., Schwab, A.P., 2008. Lability of polycyclic aromatic hydro-
Baek, S.O., Field, R.A., Goldstone, M.E., Kirk, P.W., Lester, J.N., Perry, R., 1991. A review carbons in the rhizosphere. Chemosphere 70 (9), 1644e1652.
of atmospheric polycyclic aromatic hydrocarbons: sources, fate and behavior. Collins, J.F., Brown, J.P., Alexeeff, G.V., Salmon, A.G., 1998. Potency equivalency fac-
Water Air Soil Pollut. 60 (3e4), 279e300. tors for some polycyclic aromatic hydrocarbons and polycyclic aromatic hy-
Bahri, R., Saidane-Mosbahi, D., Rouabhia, M., 2010. Cytokine release and cytotoxicity drocarbon derivatives. Regul. Toxicol. Pharmacol. 28 (1), 45e54.
in human keratinocytes induced by polycyclic aromatic hydrocarbons (1- Corrales, J., Thornton, C., White, M., Willett, K.L., 2014. Multigenerational effects of
methylpyrene and perylene). J. Toxicol. Environ. Health A 73 (8), 552e564. benzo[a]pyrene exposure on survival and developmental deformities in
Bakke, T., Klungsøyr, J., Sanni, S., 2013. Environmental impacts of produced water zebrafish larvae. Aquat. Toxicol. 148, 16e26.
and drilling waste discharges from the Norwegian offshore petroleum industry. peaux, G., Bouillaud-Kremarik, P., Sikhayeva, N., Rychen, G., Soulimani, R.,
Cre
Mar. Environ. Res. 92, 154e169. Schroeder, H., 2012. Late effects of a perinatal exposure to a 16 PAH mixture:
Benford, D., Dinovi, M., Setzer, R.W., 2010. Application of the margin-of-exposure increase of anxiety-related behaviours and decrease of regional brain meta-
(MoE) approach to substances in food that are genotoxic and carcinogenic bolism in adult male rats. Toxicol. Lett. 211 (2), 105e113.
e.g.: benzo[a]pyrene and polycyclic aromatic hydrocarbons. Food Chem. Toxicol. Crew, K.D., Gammon, M.D., Terry, M.B., Zhang, F.F., Agrawal, M., Eng, S.M., Sagiv, S.K.,
48 (Suppl. 1), S42eS48. Teitelbaum, S.L., Neugut, A.I., Santella, R.M., 2007. Genetic polymorphisms in the
Berge, G., Mollerup, S., ØVrebø, S., Hewer, A., Phillips, D.H., Eilertsen, E., Haugen, A., apoptosis-associated genes FAS and FASL and breast cancer risk. Carcinogenesis
2004. Role of estrogen receptor in regulation of polycyclic aromatic hydrocar- 28 (12), 2548e2551.
bon metabolic activation in lung. Lung Cancer 45 (3), 289e297. Croxton, A.N., Wikfors, G.H., Schulterbrandt-Gragg, R.D., 2012. Immunomodulation
Bilbao, E., Raingeard, D., de Cerio, O.D., Ortiz-Zarragoitia, M., Ruiz, P., Izagirre, U., in eastern oysters, Crassostrea virginica, exposed to a PAH-contaminated,
Orbea, A., Marigo mez, I., Cajaraville, M.P., Cancio, I., 2010. Effects of exposure to microphytobenthic diatom. Aquat. Toxicol. 118e119, 27e36.
Prestige-like heavy fuel oil and to perfluorooctane sulfonate on conventional Curfs, D.M., Knaapen, A.M., Pachen, D.M., Gijbels, M.J., Lutgens, E., Smook, M.L.,
biomarkers and target gene transcription in the thicklip grey mullet Chelon Kockx, M.M., Daemen, M.J., van Schooten, F.J., 2005. Polycyclic aromatic hy-
labrosus. Aquat. Toxicol. 98 (3), 282e296. drocarbons induce an inflammatory atherosclerotic plaque phenotype
820 Y. Zhang et al. / Environmental Pollution 213 (2016) 809e824

irrespective of their DNA binding properties. FASEB J. 19 (10), 1290e1292. Garcia-Reyero, N., Pin ~ a, B., Grimalt, J.O., Fern andez, P., Fonts, R., Polvillo, O.,
Currier, N., Solomon, S.E., Demicco, E.G., Chang, D.L., Farago, M., Ying, H., Martrat, B., 2005. Estrogenic activity in sediments from European mountain
Dominguez, I., Sonenshein, G.E., Cardiff, R.D., Xiao, Z.X., Sherr, D.H., Seldin, D.C., lakes. Environ. Sci. Technol. 39 (6), 1427e1435.
2005. Oncogenic signaling pathways activated in DMBA-induced mouse Gate, L., Langlais, C., Micillino, J.C., Nunge, H., Bottin, M.C., Wrobel, R., Binet, S., 2006.
mammary tumors. Toxicol. Pathol. 33 (6), 726e737. Bitumen fume-induced gene expression profile in rat lung. Toxicol. Appl.
D'Adamo, R., Specchiulli, A., Cassin, D., Botter, M., Zonta, R., Fabbrocini, A., 2014. The Pharmacol. 215 (1), 83e92.
effect of floods on sediment contamination in a microtidal coastal lagoon: the Ge, C., Ye, J., Wang, Q., Zhang, C., Yang, J.M., Qian, G., 2012. Polycyclic aromatic
lagoon of Lesina, Italy. Arch. Environ. Contam. Toxicol. 67 (3), 297e309. hydrocarbons suppress meiosis in primordial germ cells via the AHR signaling
Danielsen, P.H., Møller, P., Jensen, K.A., Sharma, A.K., Wallin, H., Bossi, R., Autrup, H., pathway. Toxicol. Lett. 210 (3), 285e292.
Mølhave, L., Ravanat, J.L., Briede , J.J., de Kok, T.M., Loft, S., 2011. Oxidative stress, Geffard, O., Budzinski, H., Augagneur, S., Seaman, M.N., His, E., 2001. Assessment of
DNA damage, and inflammation induced by ambient air and wood smoke sediment contamination by spermiotoxicity and embryotoxicity bioassays with
particulate matter in human A549 and THP-1 cell lines. Chem. Res. Toxicol. 24 sea urchins (Paracentrotus lividus) and oysters (Crassostrea gigas). Environ.
(2), 168e184. Toxicol. Chem. 20 (7), 1605e1611.
David, A., Gomez, E., Aït-Aïssa, S., Rosain, D., Casellas, C., Fenet, H., 2010. Impact of Geraudie, P., Nahrgang, J., Forget-Leray, J., Minier, C., Camus, L., 2014. In vivo effects
urban wastewater discharges on the sediments of a small Mediterranean river of environmental concentrations of produced water on the reproductive func-
and associated coastal environment: assessment of estrogenic and dioxin-like tion of polar cod (Boreogadus saida). J. Toxicol. Environ. Health A 77 (9e11),
activities. Arch. Environ. Contam. Toxicol. 58 (3), 562e575. 557e573.
DeMarini, D.M., Shelton, M.L., Levine, J.G., 1995. Mutation spectrum of cigarette Grung, M., Næs, K., Fogelberg, O., Nilsen, A.J., Brack, W., Lübcke-von Varel, U.,
smoke condensate in Salmonella: comparison to mutations in smoking- Thomas, K.V., 2011. Effects-directed analysis of sediments from polluted marine
associated tumors. Carcinogenesis 16 (10), 2535e2542. sites in Norway. J. Toxicol. Environ. Health A 74 (7e9), 439e454.
den Besten, P.J., van den Brink, P.J., 2005. Bioassay responses and effects on benthos Guo, N., Faller, D.V., Vaziri, C., 2000. A novel DNA damage checkpoint involving
after pilot remediations in the delta of the rivers Rhine and Meuse. Environ. post-transcriptional regulation of cyclin A expression. J. Biol. Chem. 275 (3),
Pollut. 136 (2), 197e208. 1715e1722.
Deng, X., Pan, L., Miao, J., Cai, Y., Hu, F., 2014. Digital gene expression analysis of Hallberg, L.M., Ward, J.B., Hernandez, C., Ameredes, B.T., Wickliffe, J.K., HEI Health
reproductive toxicity of benzo[a]pyrene in male scallop chlamys farreri. Eco- Review Committee, 2015. Part 3. Assessment of genotoxicity and oxidative
toxicol. Environ. Saf. 110, 190e196. damage in rats after chronic exposure to new-technology diesel exhaust in the
den Hartigh, L.J., Lame , M.W., Ham, W., Kleeman, M.J., Tablin, F., Wilson, D.W., 2010. ACES bioassay. Res. Rep. Health Eff. Inst. 184, 87e105.
Endotoxin and polycyclic aromatic hydrocarbons in ambient fine particulate Han, X.M., Liu, Y.R., Zheng, Y.M., Zhang, X.X., He, J.Z., 2014. Response of bacterial
matter from Fresno, California initiate human monocyte inflammatory re- pdo1, nah, and C12O genes to aged soil PAH pollution in a coke factory area.
sponses mediated by reactive oxygen species. Toxicol. Vitro 24 (7), 1993e2002. Environ. Sci. Pollut. Res. Int. 21 (16), 9754e9763.
Denissenko, M.F., Pao, A., Tang, M., Pfeifer, G.P., 1996. Preferential formation of Hanaoka, T., Yamano, Y., Pan, G., Hara, K., Ichiba, M., Zhang, J., Zhang, S., Liu, T., Li, L.,
benzo[a]pyrene adducts at lung cancer mutational hotspots in P53. Science 274 Takahashi, K., Kagawa, J., Tsugane, S., 2002. Cytochrome P450 1B1 mRNA levels
(5286), 430e432. in peripheral blood cells and exposure to polycyclic aromatic hydrocarbons in
de Oliveira Alves, N., Matos Loureiro, A.L., Dos Santos, F.C., Nascimento, K.H., Chinese coke oven workers. Sci. Total Environ. 296 (1e3), 27e33.
Dallacort, R., de Castro Vasconcellos, P., de Souza Hacon, S., Artaxo, P., de Hankinson, O., 1995. The aryl hydrocarbon receptor complex. Annu. Rev. Pharmacol.
Medeiros, S.R., 2011. Genotoxicity and composition of particulate matter from Toxicol. 35, 307e340.
biomass burning in the eastern Brazilian Amazon region. Ecotoxicol. Environ. Hao, N., Whitelaw, M.L., 2013. The emerging roles of AhR in physiology and im-
Saf. 74 (5), 1427e1433. munity. Biochem. Pharmacol. 86 (5), 561e570.
Desalme, D., Binet, P., Chiapusio, G., 2013. Challenges in tracing the fate and effects Harman, C., Farmen, E., Tollefsen, K.E., 2010. Monitoring North Sea oil production
of atmospheric polycyclic aromatic hydrocarbon deposition in vascular plants. discharges using passive sampling devices coupled with in vitro bioassay
Environ. Sci. Technol. 47 (9), 3967e3981. techniques. J. Environ. Monit. 12 (9), 1699e1708.
Detmar, J., Jurisicova, A., 2010. Embryonic resorption and polycyclic aromatic hy- Harris, K.L., Banks, L.D., Mantey, J.A., Huderson, A.C., Ramesh, A., 2013. Bio-
drocarbons: putative immune-mediated mechanisms. Syst. Biol. Reprod. Med. accessibility of polycyclic aromatic hydrocarbons: relevance to toxicity and
56 (1), 3e17. carcinogenesis. Expert Opin. Drug Metab. Toxicol. 9 (11), 1465e1480.
Do, K.N., Fink, L.N., Jensen, T.E., Gautier, L., Parlesak, A., 2012. TLR2 controls intes- Hawliczek, A., Nota, B., Cenijn, P., Kamstra, J., Pieterse, B., Winter, R., Winkens, K.,
tinal carcinogen detoxication by CYP1A1. PLoS One 7 (3), e32309. Hollert, H., Segner, H., Legler, J., 2012. Developmental toxicity and endocrine
Dobias, L., Kusova , J., Gajdos, O., Vidov a, P., Gajdosova, D., Havra nkova, J., Fried, M., disrupting potency of 4-azapyrene, benzo[b]fluorene and retene in the zebra-
Binkova , B., Topinka, J., 1999. Bioassay-directed chemical analysis and detection fish Danio rerio. Reprod. Toxicol. 33 (2), 213e223.
of mutagenicity in ambient air of the coke oven. Mutat. Res. 445 (2), 285e293. Hayakawa, K., Onoda, Y., Tachikawa, C., Yoshita, M., Toriba, A., Kameda, A., Tang, N.,
Duan, H., Leng, S., Pan, Z., Dai, Y., Niu, Y., Huang, C., Bin, P., Wang, Y., Liu, Q., Chen, W., 2008. Interaction of hydroxylated polycyclic aromatic hydrocarbons to estrogen
Zheng, Y., 2009. Biomarkers measured by cytokinesis-block micronucleus receptor. Polycycl. Aromat. Compd. 28, 382e391.
cytome assay for evaluating genetic damages induced by polycyclic aromatic Hayashi, M., Ueda, T., Uyeno, K., Wada, K., Kinae, N., Saotome, K., Tanaka, N.,
hydrocarbons. Mutat. Res. 677 (1e2), 93e99. Takai, A., Sasaki, Y.F., Asano, N., Sofuni, T., Ojima, Y., 1998. Development of
Farmer, P.B., Singh, R., Kaur, B., Sram, R.J., Binkova, B., Kalina, I., Popov, T.A., Garte, S., genotoxicity assay systems that use aquatic organisms. Mutat. Res. 399 (2),
Taioli, E., Gabelova, A., Cebulska-Wasilewska, A., 2003. Molecular epidemiology 125e133.
studies of carcinogenic environmental pollutants. Effects of polycyclic aromatic He, C., Wang, C., Li, B., Wu, M., Geng, H., Chen, Y., Zuo, Z., 2012. Exposure of
hydrocarbons (PAHs) in environmental pollution on exogenous and oxidative Sebastiscus marmoratus embryos to pyrene results in neurodevelopmental
DNA damage. Mutat. Res. 544 (2e3), 397e402. defects and disturbs related mechanisms. Aquat. Toxicol. 116e117, 109e115.
Feng, S., Cao, Z., Wang, X., 2013. Role of aryl hydrocarbon receptor in cancer. Bio- Henklova , P., Vrzal, R., Ulrichova , J., Dvora
k, Z., 2008. Role of mitogen-activated
chim. Biophys. Acta 1836 (2), 197e210. protein kinases in aryl hydrocarbon receptor signaling. Chem. Biol. Interact.
Feng, Z., Hu, W., Rom, W.N., Costa, M., Tang, M.S., 2003. Chromium(VI) exposure 172 (2), 93e104.
enhances polycyclic aromatic hydrocarbon-DNA binding at the p53 gene in Hew, K.M., Walker, A.I., Kohli, A., Garcia, M., Syed, A., McDonald-Hyman, C.,
human lung cells. Carcinogenesis 24 (4), 771e778. Noth, E.M., Mann, J.K., Pratt, B., Balmes, J., Hammond, S.K., Eisen, E.A.,
Fertuck, K.C., Kumar, S., Sikka, H.C., Matthews, J.B., Zacharewski, T.R., 2001. Inter- Nadeau, K.C., 2015. Childhood exposure to ambient polycyclic aromatic hy-
action of PAH-related compounds with the alpha and beta isoforms of the es- drocarbons is linked to epigenetic modifications and impaired systemic im-
trogen receptor. Toxicol. Lett. 121 (3), 167e177. munity in T cells. Clin. Exp. Allergy 45 (1), 238e248.
Foresta, M., Izzotti, A., La Maestra, S., Micale, R., Poggi, A., Vecchio, D., Frosina, G., Hilscherova, K., Kannan, K., Holoubek, I., Giesy, J.P., 2002. Characterization of es-
2014. Accelerated repair and reduced mutagenicity of DNA damage induced by trogenic activity of riverine sediments from the Czech Republic. Arch. Environ.
cigarette smoke in human bronchial cells transfected with E.coli for- Contam. Toxicol. 43 (2), 175e185.
mamidopyrimidine DNA glycosylase. PLoS One 9 (1), e87984. Hirmann, D., Loibner, A.P., Braun, R., Szolar, O.H., 2007. Applicability of the biolu-
Franci, C.D., Guillemette, M., Pelletier, E., Chastel, O., Bonnefoi, S., Verreault, J., 2014. minescence inhibition test in the 96-well microplate format for PAH-solutions
Endocrine status of a migratory bird potentially exposed to the Deepwater and elutriates of PAH-contaminated soils. Chemosphere 67 (6), 1236e1242.
Horizon oil spill: a case study of northern gannets breeding on Bonaventure Hockley, S.L., Arlt, V.M., Brewer, D., Giddings, I., Phillips, D.H., 2006. Time- and
Island. East. Can. Sci. Total Environ. 473e474, 110e116. concentration-dependent changes in gene expression induced by benzo(a)
Fujimoto, T., Kitamura, S., Sanoh, S., Sugihara, K., Yoshihara, S., Fujimoto, N., Ohta, S., pyrene in two human cell lines, MCF-7 and HepG2. BMC Genomics 7, 260.
2003. Estrogenic activity of an environmental pollutant, 2-nitrofluorene, after Hockley, S.L., Mathijs, K., Staal, Y.C., Brewer, D., Giddings, I., van Delft, J.H.,
metabolic activation by rat liver microsomes. Biochem. Biophys. Res. Commun. Phillips, D.H., 2009. Interlaboratory and interplatform comparison of micro-
303 (2), 419e426. array gene expression analysis of HepG2 cells exposed to benzo(a)pyrene.
Galva~o, M.F., de M.Cabral, T., de Andre , P.A., Andrade Mde, F., de Miranda, R.M., OMICS 13 (2), 115e125.
Saldiva, P.H., de C.Vasconcellos, P., de Medeiros, S.R., 2014. Cashew nut roasting: Holth, T.F., Nourizadeh-Lillabadi, R., Blaesbjerg, M., Grung, M., Holbech, H.,
chemical characterization of particulate matter and genotocixity analysis. En- Petersen, G.I., Alestro €m, P., Hylland, K., 2008. Differential gene expression and
viron. Res. 131, 145e152. biomarkers in zebrafish (Danio rerio) following exposure to produced water
Gao, M.L., Chen, L., Li, Y.F., Xue, X.C., Chen, L., Wang, L.N., Shah, W., Kong, Y., 2014. components. Aquat. Toxicol. 90 (4), 277e291.
Synergistic increase of oxidative stress and tumor markers in PAH-exposed Hooven, L.A., Baird, W.M., 2008. Proteomic analysis of MCF-7 cells treated with
workers. Asian Pac. J. Cancer Prev. 15 (17), 7105e7112. benzo[a]pyrene, dibenzo[a,l]pyrene, coal tar extract, and diesel exhaust extract.
Y. Zhang et al. / Environmental Pollution 213 (2016) 809e824 821

Toxicology 249 (1), 1e10. Koh, C.H., Khim, J.S., Villeneuve, D.L., Kannan, K., Giesy, J.P., 2002. Analysis of trace
Huang, M.C., Chen, F.Y., Chou, M.T., Su, J.G., 2012. Fluoranthene enhances p53 organic contaminants in sediment, pore water, and water samples from Onsan
expression and decreases mutagenesis induced by benzo[a]pyrene. Toxicol. Bay, Korea: instrumental analysis and in vitro gene expression assay. Environ.
Lett. 208 (3), 214e224. Toxicol. Chem. 21 (9), 1796e1803.
Huang, S.K., Zhang, Q., Qiu, Z., Chung, K.F., 2015. Mechanistic impact of outdoor air Koh, C.H., Khim, J.S., Villeneuve, D.L., Kannan, K., Johnson, B.G., Giesy, J.P., 2005.
pollution on asthma and allergic diseases. J. Thorac. Dis. 7 (1), 23e33. Instrumental and bioanalytical measures of dioxin-like and estrogenic com-
Huuskonen, S.E., Tuvikene, A., Trapido, M., Fent, K., Hahn, M.E., 2000. Cytochrome pounds and activities associated with sediment from the Korean coast. Eco-
P4501A induction and porphyrin accumulation in PLHC-1 fish cells exposed to toxicol. Environ. Saf. 61 (3), 366e379.
sediment and oil shale extracts. Arch. Environ. Contam. Toxicol. 38 (1), 59e69. Koh, C.H., Kim, G.B., Maruya, K.A., Anderson, J.W., Jones, J.M., Kang, S.G., 2001. In-
Hwang, H.M., Stanton, B., McBride, T., Anderson, M.J., 2014. Polycyclic aromatic duction of the P450 reporter gene system bioassay by polycyclic aromatic hy-
hydrocarbon body residues and lysosomal membrane destabilization in mus- drocarbons in Ulsan Bay (South Korea) sediments. Environ. Pollut. 111 (3),
sels exposed to the Dubai Star bunker fuel oil (intermediate fuel oil 380) spill in 437e445.
San Francisco Bay. Environ. Toxicol. Chem. 33 (5), 1117e1121. Kolasa, E., Balaguer, P., Houlbert, N., Fardel, O., 2012. Phorbol ester-modulation of
Hylland, K., 2006. Polycyclic aromatic hydrocarbon (PAH) ecotoxicology in marine estrogenic genomic effects triggered by the environmental contaminant
ecosystems. J. Toxicol. Environ. Health A 69 (1e2), 109e123. benzanthracene. Toxicol. Vitro 26 (6), 807e816.
Hyo € tyla
€inen, T., Oikari, A., 1999. The toxicity and concentrations of PAHs in Kolasa, E., Houlbert, N., Balaguer, P., Fardel, O., 2013. AhR- and NF-kB-dependent
creosote-contaminated lake sediment. Chemosphere 38 (5), 1135e1144. induction of interleukin-6 by co-exposure to the environmental contaminant
Jarvis, I.W., Bergvall, C., Morales, D.A., Kummrow, F., Umbuzeiro, G.A., benzanthracene and the cytokine tumor necrosis factor-a in human mammary
Westerholm, R., Stenius, U., Dreij, K., 2014. Nanomolar levels of PAHs in extracts MCF-7 cells. Chem. Biol. Interact. 203 (2), 391e400.
from urban air induce MAPK signaling in HepG2 cells. Toxicol. Lett. 229 (1), Konstandi, M., Harkitis, P., Thermos, K., Ogren, S.O., Johnson, E.O., Tzimas, P.,
25e32. Marselos, M., 2007. Modification of inherent and drug-induced dopaminergic
Jayasundara, N., Van Tiem Garner, L., Meyer, J.N., Erwin, K.N., Di Giulio, R.T., 2015. activity after exposure to benzo(alpha)pyrene. Neurotoxicology 28 (4),
AHR2-Mediated transcriptomic responses underlying the synergistic cardiac 860e867.
developmental toxicity of PAHs. Toxicol. Sci. 143 (2), 469e481. Korashy, H.M., El-Kadi, A.O., 2006. The role of aryl hydrocarbon receptor in the
Jeffy, B.D., Chirnomas, R.B., Romagnolo, D.F., 2002. Epigenetics of breast cancer: pathogenesis of cardiovascular diseases. Drug Metab. Rev. 38 (3), 411e450.
polycyclic aromatic hydrocarbons as risk factors. Environ. Mol. Mutagen 39 Kreitsberg, R., Tuvikene, A., Barsiene, _ J., Fricke, N.F., Rybakovas, A., Andreikenait
_ _ L.,
e,
(2e3), 235e244. Rumvolt, K., Vilbaste, S., 2012. Biomarkers of environmental contaminants in
Jeng, H.A., Pan, C.H., Diawara, N., Chang-Chien, G.P., Lin, W.Y., Huang, C.T., Ho, C.K., the coastal waters of Estonia (Baltic Sea): effects on eelpouts (Zoarces vivipa-
Wu, M.T., 2011. Polycyclic aromatic hydrocarbon-induced oxidative stress and rus). J. Environ. Monit. 14 (9), 2298e2308.
lipid peroxidation in relation to immunological alteration. Occup. Environ. Med. Kriek, E., Van Schooten, F.J., Hillebrand, M.J., Van Leeuwen, F.E., Den Engelse, L., De
68 (9), 653e658. Looff, A.J., Dijkmans, A.P., 1993. DNA adducts as a measure of lung cancer risk in
Jung, J.H., Kim, M., Yim, U.H., Ha, S.Y., An, J.G., Won, J.H., Han, G.M., Kim, N.S., humans exposed to polycyclic aromatic hydrocarbons. Environ. Health Perspect.
Addison, R.F., Shim, W.J., 2011. Biomarker responses in pelagic and benthic fish 99, 71e75.
over 1 year following the Hebei Spirit oil spill (Taean, Korea). Mar. Pollut. Bull. Kuhn, K., Nowak, B., Klein, G., Behnke, A., Seidel, A., Lampen, A., 2008. Determi-
62 (8), 1859e1866. nation of polycyclic aromatic hydrocarbons in smoked pork by effect-directed
Kaba tkov a, M., Svobodova, J., Pe
n , K., Mohatad, D.S., Smerdova
cíkova , L., Kozubík, A., bioassay with confirmation by chemical analysis. J. Food Prot. 71 (5), 993e999.
Machala, M., Vondr a
cek, J., 2014. Interactive effects of inflammatory cytokine Kummer, V., Maskova , J., Zralý, Z., Faldyna, M., 2013. Ovarian disorders in immature
and abundant low-molecular-weight PAHs on inhibition of gap junctional rats after postnatal exposure to environmental polycyclic aromatic hydrocar-
intercellular communication, disruption of cell proliferation control, and the bons. J. Appl. Toxicol. 33 (2), 90e99.
AhR-dependent transcription. Toxicol. Lett. 232 (1), 113e121. Kummer, V., Maskova , J., Zralý, Z., Neca, J., Simeckova , P., Vondr
acek, J., Machala, M.,
Kamal, A., Cincinelli, A., Martellini, T., Malik, R.N., 2015. A review of PAH exposure 2008. Estrogenic activity of environmental polycyclic aromatic hydrocarbons in
from the combustion of biomass fuel and their less surveyed effect on the blood uterus of immature Wistar rats. Toxicol. Lett. 180 (3), 212e221.
parameters. Environ. Sci. Pollut. Res. Int. 22 (6), 4076e4098. Kurihara, R., Shiraishi, F., Tanaka, N., Hashimoto, S., 2005. Presence and estrogenicity
Kameda, T., Akiyama, A., Toriba, A., Tang, N., Hayakawa, K., 2012. Hydroxylated nitro of anthracene derivatives in coastal Japanese waters. Environ. Toxicol. Chem. 24
polycyclic aromatic compounds: atomospheric occurrence and health impact. (8), 1984e1993.
In: Bandeira, G.C., Meneses, H.E. (Eds.), Handbook of Polycyclic Aromatic Lang, Y., Wang, N., Gao, H., Bai, J., 2012. Distribution and risk assessment of poly-
Hydrocarbons-chemistry, Occurrence and Health Issues. Nova Publishers, New cyclic aromatic hydrocarbons (PAHs) from Liaohe estuarine wetland soils. En-
York, pp. 235e264. viron. Monit. Assess. 184 (9), 5545e5552.
Kamiya, M., Toriba, A., Onoda, Y., Kizu, R., Hayakawa, K., 2005. Evaluation of es- Larsson, M., Hagberg, J., Rotander, A., van Bavel, B., Engwall, M., 2013. Chemical and
trogenic activities of hydroxylated polycyclic aromatic hydrocarbons in ciga- bioanalytical characterisation of PAHs in risk assessment of remediated PAH-
rette smoke condensate. Food Chem. Toxicol. 43 (7), 1017e1027. contaminated soils. Environ. Sci. Pollut. Res. Int. 20 (12), 8511e8520.
Karimi, P., Peters, K.O., Bidad, K., Strickland, P.T., 2015. Polycyclic aromatic hydro- Lau, K.L., Tsang, Y.Y., Chiu, S.W., 2003. Use of spent mushroom compost to bio-
carbons and childhood asthma. Eur. J. Epidemiol. 30 (2), 91e101. remediate PAH-contaminated samples. Chemosphere 52 (9), 1539e1546.
Keith, L., Telliard, W., 1979. ES&T special report: priority pollutants: I-a perspective Lauer, F.T., Walker, M.K., Burchiel, S.W., 2013. Dibenzo[def,p]chrysene (DBC) sup-
view. Environ. Sci. Technol. 13 (4), 416e423. presses antibody formation in spleen cells following oral exposures of mice.
Kennedy, S.W., Jones, S.P., 1994. Simultaneous measurement of cytochrome P4501A J. Toxicol. Environ. Health A 76 (1), 16e24.
catalytic activity and total protein concentration with a fluorescence plate Le Bihanic, F., Morin, B., Cousin, X., Le Menach, K., Budzinski, H., Cachot, J., 2014.
reader. Anal. Biochem. 222 (1), 217e223. Developmental toxicity of PAH mixtures in fish early life stages. Part I: adverse
Kerambrun, E., Henry, F., Marechal, A., Sanchez, W., Minier, C., Filipuci, I., Amara, R., effects in rainbow trout. Environ. Sci. Pollut. Res. Int. 21 (24), 13720e13731.
2012. A multibiomarker approach in juvenile turbot, Scophthalmus maximus, Lee, R.F., Anderson, J.W., 2005. Significance of cytochrome P450 system responses
exposed to contaminated sediments. Ecotoxicol. Environ. Saf. 80, 45e53. and levels of bile fluorescent aromatic compounds in marine wildlife following
Kim, K.H., Jahan, S.A., Kabir, E., 2011a. A review of diseases associated with oil spills. Mar. Pollut. Bull. 50 (7), 705e723.
household air pollution due to the use of biomass fuels. J. Hazard. Mater. 192 Lemiere, S., Cossu-Leguille, C., Bispo, A., Jourdain, M.J., Lanhers, M.C., Burnel, D.,
(2), 425e431. Vasseur, P., 2004. Genotoxicity related to transfer of oil spill pollutants from
Kim, A., Park, M., Yoon, T.K., Lee, W.S., Ko, J.J., Lee, K., Bae, J., 2011b. Maternal mussels to mammals via food. Environ. Toxicol. 19 (4), 387e395.
exposure to benzo[b]fluoranthene disturbs reproductive performance in male Lemieux, C.L., Long, A.S., Lambert, I.B., Lundstedt, S., Tysklind, M., White, P.A., 2015.
offspring mice. Toxicol. Lett. 203 (1), 54e61. Cancer risk assessment of polycyclic aromatic hydrocarbon contaminated soils
Kim, J.H., Yamaguchi, K., Lee, S.H., Tithof, P.K., Sayler, G.S., Yoon, J.H., Baek, S.J., 2005. determined using bioassay-derived levels of benzo[a]pyrene equivalents. En-
Evaluation of polycyclic aromatic hydrocarbons in the activation of early growth viron. Sci. Technol. 49 (3), 1797e1805.
response-1 and peroxisome proliferator activated receptors. Toxicol. Sci. 85 (1), Le Vee, M., Kolasa, E., Jouan, E., Collet, N., Fardel, O., 2014. Differentiation of human
585e593. placental BeWo cells by the environmental contaminant benzo(a)pyrene. Chem.
Kiyama, R., Wada-Kiyama, Y., 2015. Estrogenic endocrine disruptors: molecular Biol. Interact. 210, 1e11.
mechanisms of action. Environ. Int. 83, 11e40. Lewtas, J., 2007. Air pollution combustion emissions: characterization of causative
Kiyama, R., Zhu, Y., 2014. DNA microarray-based gene expression profiling of es- agents and mechanisms associated with cancer, reproductive, and cardiovas-
trogenic chemicals. Cell. Mol. Life Sci. 71 (11), 2065e2082. cular effects. Mutat. Res. 636 (1e3), 95e133.
Klingbeil, E.C., Hew, K.M., Nygaard, U.C., Nadeau, K.C., 2014. Polycyclic aromatic Li, Q., Lauer, F.T., Liu, K.J., Hudson, L.G., Burchiel, S.W., 2010. Low-dose synergistic
hydrocarbons, tobacco smoke, and epigenetic remodeling in asthma. Immunol. immunosuppression of T-dependent antibody responses by polycyclic aromatic
Res. 58 (2e3), 369e373. hydrocarbons and arsenic in C57BL/6J murine spleen cells. Toxicol. Appl.
Knaapen, A.M., Curfs, D.M., Pachen, D.M., Gottschalk, R.W., de Winther, M.P., Pharmacol. 245 (3), 344e351.
Daemen, M.J., Van Schooten, F.J., 2007. The environmental carcinogen benzo[a] Li, W., Wang, C., Wang, H., Chen, J., Shen, H., Shen, G., Huang, Y., Wang, R., Wang, B.,
pyrene induces expression of monocyte-chemoattractant protein-1 in vascular Zhang, Y., Chen, H., Chen, Y., Su, S., Lin, N., Tang, J., Li, Q., Wang, X., Liu, J., Tao, S.,
tissue: a possible role in atherogenesis. Mutat. Res. 621 (1e2), 31e41. 2014. Atmospheric polycyclic aromatic hydrocarbons in rural and urban areas of
Koganti, A., Singh, R., Ma, B.L., Weyand, E.H., 2001. Comparative analysis of PAH: northern China. Environ. Pollut. 192, 83e90.
DNA adducts formed in lung of mice exposed to neat coal tar and soils Lisouza, F.A., Owuor, P.O., Lalah, J.O., 2012. Environmental fate of polycyclic aromatic
contaminated with coal tar. Environ. Sci. Technol. 35 (13), 2704e2709. hydrocarbons emitted from indoor burning of fuel biomass in poorly ventilated
822 Y. Zhang et al. / Environmental Pollution 213 (2016) 809e824

households: a case of study in the traditional rural households in Western particles. Environ. Mol. Mutagen 54 (9), 719e736.
Kenya. In: Bandeira, G.C., Meneses, H.E. (Eds.), Handbook of Polycyclic Aromatic Nam, J.J., Sweetman, A.J., Jones, K.C., 2009. Polynuclear aromatic hydrocarbons
Hydrocarbons-chemistry, Occurrence and Health Issues. Nova Publishers, New (PAHs) in global background soils. J. Environ. Monit. 11 (1), 45e48.
York, pp. 1e43. Nam, J.J., Thomas, G.O., Jaward, F.M., Steinnes, E., Gustafsson, O., Jones, K.C., 2008.
Liu, J., Zhang, L., Winterroth, L.C., Garcia, M., Weiman, S., Wong, J.W., Sunwoo, J.B., PAHs in background soils from Western Europe: influence of atmospheric
Nadeau, K.C., 2013. Epigenetically mediated pathogenic effects of phenanthrene deposition and soil organic matter. Chemosphere 70 (9), 1596e1602.
on regulatory T cells. J. Toxicol. 2013, 967029. Navas, J.M., Segner, H., 2000. Antiestrogenicity of beta-naphthoflavone and PAHs in
Líbalov a, H., Kr
ckova, S., Uhlírov
a, K., Klema, J., Ciganek, M., Ro  a
€ssner Jr., P., Sr m, R.J., cultured rainbow trout hepatocytes: evidence for a role of the arylhydrocarbon
Vondr a
cek, J., Machala, M., Topinka, J., 2014. Analysis of gene expression receptor. Aquat. Toxicol. 51 (1), 79e92.
changes in A549 cells induced by organic compounds from respirable air par- N'jai, A.U., Larsen, M.C., Bushkofsky, J.R., Czuprynski, C.J., Jefcoate, C.R., 2011. Acute
ticles. Mutat. Res. 770, 94e105. disruption of bone marrow hematopoiesis by benzo(a)pyrene is selectively
Ma, J.Y., Ma, J.K., 2002. The dual effect of the particulate and organic components of reversed by aryl hydrocarbon receptor-mediated processes. Mol. Pharmacol. 79
diesel exhaust particles on the alteration of pulmonary immune/inflammatory (4), 724e734.
responses and metabolic enzymes. J. Environ. Sci. Health C 20 (2), 117e147. Nogueira, P., Lourenço, J., Rodriguez, E., Pacheco, M., Santos, C., Rotchell, J.M.,
Ma, T.H., Cabrera, G.L., Chen, R., Gill, B.S., Sandhu, S.S., Vandenberg, A.L., Mendo, S., 2009. Transcript profiling and DNA damage in the European eel
Salamone, M.F., 1994. Tradescantia micronucleus bioassay. Mutat. Res. 310 (2), (Anguilla anguilla L.) exposed to 7,12-dimethylbenz[a]anthracene. Aquat. Tox-
221e230. icol. 94 (2), 123e130.
Machala, M., Ciganek, M., Bla ha, L., Minksov ck, J., 2001. Aryl hydro-
a, K., Vondra Oda, Y., Nakamura, S., Oki, I., Kato, T., Shinagawa, H., 1985. Evaluation of the new
carbon receptor-mediated and estrogenic activities of oxygenated polycyclic system (umu-test) for the detection of environmental mutagens and carcino-
aromatic hydrocarbons and azaarenes originally identified in extracts of river gens. Mutat. Res. 147 (5), 219e229.
sediments. Environ. Toxicol. Chem. 20 (12), 2736e2743. Oh, J., Kwak, J.H., Kwon, D.Y., Kim, A.Y., Oh, D.S., Je, N.K., Lee, J., Jung, Y.S., 2014.
Maertens, R.M., Bailey, J., White, P.A., 2004. The mutagenic hazards of settled house Transformation of mouse liver cells by methylcholanthrene leads to phenotypic
dust: a review. Mutat. Res. 567 (2e3), 401e425. changes associated with epithelial-mesenchymal transition. Toxicol. Res. 30 (4),
Mahadevan, B., Keshava, C., Musafia-Jeknic, T., Pecaj, A., Weston, A., Baird, W.M., 261e266.
2005. Altered gene expression patterns in MCF-7 cells induced by the urban Ohta, R., Takagi, A., Ohmukai, H., Marumo, H., Ono, A., Matsushima, Y., Inoue, T.,
dust particulate complex mixture standard reference material 1649a. Cancer Ono, H., Kanno, J., 2012. Ovariectomized mouse uterotrophic assay of 36
Res. 65 (4), 1251e1258. chemicals. J. Toxicol. Sci. 37 (5), 879e889.
Mann, K.K., Doerre, S., Schlezinger, J.J., Sherr, D.H., Quadri, S., 2001. The role of NF- Ohtake, F., Fujii-Kuriyama, Y., Kawajiri, K., Kato, S., 2011. Cross-talk of dioxin and
kappaB as a survival factor in environmental chemical-induced pre-B cell estrogen receptor signals through the ubiquitin system. J. Steroid Biochem. Mol.
apoptosis. Mol. Pharmacol. 59 (2), 302e309. Biol. 127 (1e2), 102e107.
Maron, D.M., Ames, B.N., 1983. Revised methods for the Salmonella mutagenicity Olajire, A.A., Altenburger, R., Küster, E., Brack, W., 2005. Chemical and ecotoxico-
test. Mutat. Res. 113 (3e4), 173e215. logical assessment of polycyclic aromatic hydrocarbonecontaminated sedi-
Marvin, C.H., McCarry, B.E., Lundrigan, J.A., Roberts, K., Bryant, D.W., 1999. Bioassay- ments of the Niger Delta. South. Niger. Sci. Total Environ. 340 (1e3), 123e136.
directed fractionation of PAH of molecular mass 302 in coal tar-contaminated Oleszczuk, P., 2008. Phytotoxicity of municipal sewage sludge composts related to
sediment. Sci. Total Environ. 231 (2e3), 135e144. physico-chemical properties, PAHs and heavy metals. Ecotoxicol. Environ. Saf.
Marvin, C.H., McCarry, B.E., Villella, J., Allan, L.M., Bryant, D.W., 2000. Chemical and 69 (3), 496e505.
biological profiles of sediments as indicators of sources of contamination in Ovrevik, J., Arlt, V.M., Oya, E., Nagy, E., Mollerup, S., Phillips, D.H., Låg, M.,
Hamilton Harbour. Part II: bioassay-directed fractionation using the Ames Holme, J.A., 2010. Differential effects of nitro-PAHs and amino-PAHs on cytokine
Salmonella/microsome assay. Chemosphere 41 (7), 989e999. and chemokine responses in human bronchial epithelial BEAS-2B cells. Toxicol.
Matson, C.W., Gillespie, A.M., McCarthy, C., McDonald, T.J., Bickham, J.W., Appl. Pharmacol. 242 (3), 270e280.
Sullivan, R., Donnelly, K.C., 2009. Wildlife toxicology: biomarkers of genotoxic Pacheco, K.A., Tarkowski, M., Sterritt, C., Negri, J., Rosenwasser, L.J., Borish, L., 2001.
exposures at a hazardous waste site. Ecotoxicology 18 (7), 886e898. The influence of diesel exhaust particles on mononuclear phagocytic cell-
Mattis, N.D., Jay, J.W., Barnett, G.W., Rosaldo, J.J., Howerth, E.W., Means, J.C., derived cytokines: IL-10, TGF-beta and IL-1 beta. Clin. Exp. Immunol. 126 (3),
Gato, W.E., 2014. Profile of select hepatic insulin signaling pathway genes in 374e383.
response to 2-aminoanthracene dietary ingestion. J. Biol. Regul. Homeost. Page, T.J., MacWilliams, P.S., Suresh, M., Jefcoate, C.R., Czuprynski, C.J., 2004. 7-12
Agents 28 (4), 693e704. Dimethylbenz[a]anthracene-induced bone marrow hypocellularity is depen-
Mayura, K., Huebner, H.J., Dwyer, M.R., McKenzie, K.S., Donnelly, K.C., Kubena, L.F., dent on signaling through both the TNFR and PKR. Toxicol. Appl. Pharmacol. 198
Phillips, T.D., 1999. Multi-bioassay approach for assessing the potency of com- (1), 21e28.
plex mixtures of polycyclic aromatic hydrocarbons. Chemosphere 38 (8), Pampanin, D.M., Larssen, E., Oysæd, K.B., Sundt, R.C., Sydnes, M.O., 2014. Study of
1721e1732. the bile proteome of Atlantic cod (Gadus morhua): multi-biological markers of
Mazurov a, E., Hilscherova , K., Ja
lov €hler, H.R., Triebskorn, R., Giesy, J.P.,
a, V., Ko exposure to polycyclic aromatic hydrocarbons. Mar. Environ. Res. 101, 161e168.
Blaha, L., 2008. Endocrine effects of contaminated sediments on the freshwater Park, J., Ball, L.M., Richardson, S.D., Zhu, H.B., Aitken, M.D., 2008. Oxidative muta-
snail Potamopyrgus antipodarum in vivo and in the cell bioassays in vitro. genicity of polar fractions from polycyclic aromatic hydrocarbon-contaminated
Aquat. Toxicol. 89 (3), 172e179. soils. Environ. Toxicol. Chem. 27 (11), 2207e2215.
McClean, M.D., Rinehart, R.D., Sapkota, A., Cavallari, J.M., Herrick, R.F., 2007. Dermal Patel, M.R., Scheffler, B.E., Wang, L., Willett, K.L., 2006. Effects of benzo(a)pyrene
exposure and urinary 1-hydroxypyrene among asphalt roofing workers. exposure on killifish (Fundulus heteroclitus) aromatase activities and mRNA.
J. Occup. Environ. Hyg. 4 (Suppl. 1), 118e126. Aquat. Toxicol. 77 (3), 267e278.
McVeety, B.D., Hites, R.A., 1988. Atmospheric deposition of polycyclic aromatic Pavanello, S., Bollati, V., Pesatori, A.C., Kapka, L., Bolognesi, C., Bertazzi, P.A.,
hydrocarbons to water surfaces: a mass balance approach. Atmos. Environ. 22 Baccarelli, A., 2009. Global and gene-specific promoter methylation changes are
(3), 511e536. related to anti-B[a]PDE-DNA adduct levels and influence micronuclei levels in
McVeigh, A., Moore, M., Allen, J.I., Dyke, P., 2006. Lysosomal responses to nutritional polycyclic aromatic hydrocarbon-exposed individuals. Int. J. Cancer 125 (7),
and contaminant stress in mussel hepatopancreatic digestive cells: a modelling 1692e1697.
study. Mar. Environ. Res. 62 (Suppl. l), S433eS438. Perera, F.P., Jedrychowski, W., Rauh, V., Whyatt, R.M., 1999. Molecular epidemiologic
Meador, J.P., Stein, J.E., Reichert, W.L., Varanasi, U., 1995. Bioaccumulation of poly- research on the effects of environmental pollutants on the fetus. Environ.
cyclic aromatic hydrocarbons by marine organisms. Rev. Environ. Contam. Health Perspect. 107 (Suppl. 3), 451e460.
Toxicol. 143, 79e165. Perera, F.P., Whyatt, R.M., Jedrychowski, W., Rauh, V., Manchester, D., Santella, R.M.,
Mukherjee, J.J., Kumar, S., 2013. DNA synthesis inhibition in response to benzo[a] Ottman, R., 1998. Recent developments in molecular epidemiology: a study of
pyrene dihydrodiol epoxide is associated with attenuation of p(34)cdc2: role of the effects of environmental polycyclic aromatic hydrocarbons on birth out-
p53. Mutat. Res. 755 (1), 61e67. comes in Poland. Am. J. Epidemiol. 147 (3), 309e314.
Mumtaz, M.M., George, J.D., Gold, K.W., Cibulas, W., DeRosa, C.T., 1996. ATSDR rez, S., Farre
Pe , M.L., Garcia, M.J., Barcelo
, D., 2001. Occurrence of polycyclic aromatic
evaluation of health effects of chemicals. IV. Polycyclic aromatic hydrocarbons hydrocarbons in sewage sludge and their contribution to its toxicity in the
(PAHs): understanding a complex problem. Toxicol. Ind. Health 12 (6), 742e971. toxalert 100 bioassay. Chemosphere 45 (6e7), 705e712.
Murk, A.J., Legler, J., Denison, M.S., Giesy, J.P., van de Guchte, C., Brouwer, A., 1996. Phillips, D.H., Schoket, B., Hewer, A., Grover, P.L., 1990. DNA adduct formation in
Chemical-activated luciferase gene expression (CALUX): a novel in vitro human and mouse skin by mixtures of polycyclic aromatic hydrocarbons. IARC
bioassay for Ah receptor active compounds in sediments and pore water. Sci. Publ. 104, 223e229.
Fundam. Appl. Toxicol. 33 (1), 149e160. Pillai, M.C., Vines, C.A., Wikramanayake, A.H., Cherr, G.N., 2003. Polycyclic aromatic
Murphy, K.A., Quadro, L., White, L.A., 2007. The intersection between the aryl hy- hydrocarbons disrupt axial development in sea urchin embryos through a beta-
drocarbon receptor (AhR)- and retinoic acid-signaling pathways. Vitam. Horm. catenin dependent pathway. Toxicology 186 (1e2), 93e108.
75, 33e67. , C., Fan, Y., Ait Yahia, S., Vorng, H., Everaere, L., Chenivesse, C., Balsamelli, J.,
Ple
Muthumbi, W., De Boever, P., Pieters, J.G., Siciliano, S., D'Hooge, W., Verstraete, W., Azzaoui, I., de Nadai, P., Wallaert, B., Lazennec, G., Tsicopoulos, A., 2015. Poly-
2003. Polycyclic aromatic hydrocarbons (PAHs) and estrogenic compounds in cyclic aromatic hydrocarbons reciprocally regulate IL-22 and IL-17 cytokines in
experimental flue gas streams. J. Environ. Qual. 32 (2), 417e422. peripheral blood mononuclear cells from both healthy and asthmatic subjects.
Mutlu, E., Warren, S.H., Matthews, P.P., King, C., Linak, W.P., Kooter, I.M., Schmid, J.E., PLoS One 10 (4), e0122372.
Ross, J.A., Gilmour, M.I., Demarini, D.M., 2013. Bioassay-directed fractionation Plískova , M., Vondracek, J., Vojtesek, B., Kozubík, A., Machala, M., 2005. Deregulation
and sub-fractionation for mutagenicity and chemical analysis of diesel exhaust of cell proliferation by polycyclic aromatic hydrocarbons in human breast
Y. Zhang et al. / Environmental Pollution 213 (2016) 809e824 823

carcinoma MCF-7 cells reflects both genotoxic and nongenotoxic events. Tox- saliens) from the highly contaminated Aliaga Bay. Turk. Environ. Monit. Assess.
icol. Sci. 83 (2), 246e256. 165 (1e4), 87e96.
Podechard, N., Tekpli, X., Catheline, D., Holme, J.A., Rioux, V., Legrand, P., Shen, H., Huang, Y., Wang, R., Zhu, D., Li, W., Shen, G., Wang, B., Zhang, Y., Chen, Y.,
Rialland, M., Fardel, O., Lagadic-Gossmann, D., Lecureur, V., 2011. Mechanisms Lu, Y., Chen, H., Li, T., Sun, K., Li, B., Liu, W., Liu, J., Tao, S., 2013. Global atmo-
involved in lipid accumulation and apoptosis induced by 1-nitropyrene in spheric emissions of polycyclic aromatic hydrocarbons from 1960 to 2008 and
Hepa1c1c7 cells. Toxicol. Lett. 206 (3), 289e299. future predictions. Environ. Sci. Technol. 47 (12), 6415e6424.
Pohl, R.J., Fouts, J.R., 1980. A rapid method for assaying the metabolism of 7- Sievers, C.K., Shanle, E.K., Bradfield, C.A., Xu, W., 2013. Differential action of mon-
ethoxyresorufin by microsomal subcellular fractions. Anal. Biochem. 107 (1), ohydroxylated polycyclic aromatic hydrocarbons with estrogen receptors a and
150e155. b. Toxicol. Sci. 132 (2), 359e367.
Poon, C.H., Wong, T.Y., Wang, Y., Tsuchiya, Y., Nakajima, M., Yokoi, T., Leung, L.K., Singh, N.P., McCoy, M.T., Tice, R.R., Schneider, E.L., 1988. A simple technique for
2013. The citrus flavanone naringenin suppresses CYP1B1 transactivation quantitation of low levels of DNA damage in individual cells. Exp. Cell Res. 175
through antagonising xenobiotic-responsive element binding. Br. J. Nutr. 109 (1), 184e191.
(9), 1598e1605. Singhal, R., Shankar, K., Badger, T.M., Ronis, M.J., 2008. Estrogenic status modulates
Porru, S., Assennato, G., Bergamaschi, E., Carta, P., Foa , V., Forni, A., Gabbani, G., aryl hydrocarbon receptoremediated hepatic gene expression and carcinoge-
Mastrangelo, G., Sartorelli, P., 1997. The toxicology and prevention of the risks of nicity. Carcinogenesis 29 (2), 227e236.
occupational exposure to aromatic polycyclic hydrocarbons. III. The effects: Slater, G.F., Cowie, B.R., Harper, N., Droppo, I.G., 2008. Variation in PAH inputs and
epidemiological evidence, early effects. Individual hypersusceptibility. Health microbial community in surface sediments of Hamilton Harbour: implications
Surveill. G Ital. Med. Lav. Ergon. 19 (4), 152e163. to remediation and monitoring. Environ. Pollut. 153 (1), 60e70.
Pru, J.K., Kaneko-Tarui, T., Jurisicova, A., Kashiwagi, A., Selesniemi, K., Tilly, J.L., 2009. Soffritti, M., Falcioni, L., Bua, L., Tibaldi, E., Manservigi, M., Belpoggi, F., 2013. Po-
Induction of proapoptotic gene expression and recruitment of p53 herald tential carcinogenic effects of world trade center dust after intratracheal
ovarian follicle loss caused by polycyclic aromatic hydrocarbons. Reprod. Sci. 16 instillation to Sprague-Dawley rats: first observation. Am. J. Ind. Med. 56 (2),
(4), 347e356. 155e162.
Puga, A., Ma, C., Marlowe, J.L., 2009. The aryl hydrocarbon receptor cross-talks with Solhaug, A., Refsnes, M., Låg, M., Schwarze, P.E., Husøy, T., Holme, J.A., 2004. Poly-
multiple signal transduction pathways. Biochem. Pharmacol. 77 (4), 713e722. cyclic aromatic hydrocarbons induce both apoptotic and anti-apoptotic signals
Puga, A., Marlowe, J., Barnes, S., Chang, C.Y., Maier, A., Tan, Z., Kerzee, J.K., Chang, X., in Hepa1c1c7 cells. Carcinogenesis 25 (5), 809e819.
Strobeck, M., Knudsen, E.S., 2002. Role of the aryl hydrocarbon receptor in cell Song, M.K., Kim, Y.J., Song, M., Choi, H.S., Park, Y.K., Ryu, J.C., 2011. Polycyclic aro-
cycle regulation. Toxicology 181e182, 171e177. matic hydrocarbons induce migration in human hepatocellular carcinoma cells
Puy-Azurmendi, E., Navarro, A., Olivares, A., Fernandes, D., Martínez, E., Lo pez de (HepG2) through reactive oxygen species-mediated p38 MAPK signal trans-
Alda, M., Porte, C., Cajaraville, M.P., Barcelo , D., Pin
~ a, B., 2010. Origin and dis- duction. Cancer Sci. 102 (9), 1636e1644.
tribution of polycyclic aromatic hydrocarbon pollution in sediment and fish Stegeman, J.J., Lech, J.J., 1991. Cytochrome P-450 monooxygenase systems in aquatic
from the biosphere reserve of Urdaibai (Bay of Biscay, Basque country, Spain). species: carcinogen metabolism and biomarkers for carcinogen and pollutant
Mar. Environ. Res. 70 (2), 142e149. exposure. Environ. Health Perspect. 90, 101e109.
Ramesh, A., Walker, S.A., Hood, D.B., Guille n, M.D., Schneider, K., Weyand, E.H., Stroo, H.F., Roy, T.A., Liban, C.B., Kreitinger, J.P., 2005. Dermal bioavailability of benzo
2004. Bioavailability and risk assessment of orally ingested polycyclic aromatic [a]pyrene on lampblack: implications for risk assessment. Environ. Toxicol.
hydrocarbons. Int. J. Toxicol. 23 (5), 301e333. Chem. 24 (6), 1568e1572.
Ramos, K.S., Moorthy, B., 2005. Bioactivation of polycyclic aromatic hydrocarbon Sturve, J., Balk, L., Liewenborg, B., Adolfsson-Erici, M., Fo €rlin, L., Carney Almroth, B.,
carcinogens within the vascular wall: implications for human atherogenesis. 2014. Effects of an oil spill in a harbor assessed using biomarkers of exposure in
Drug Metab. Rev. 37 (4), 595e610. eelpout. Environ. Sci. Pollut. Res. Int. 21 (24), 13758e13768.
Rastall, A.C., Neziri, A., Vukovic, Z., Jung, C., Mijovic, S., Hollert, H., Nikcevic, S., Su, J.G., Liao, P.J., Huang, M.C., Chu, W.C., Lin, S.C., Chang, Y.J., 2008. Aldo-keto
Erdinger, L., 2004. The identification of readily bioavailable pollutants in Lake reductase 1C2 is essential for 1-nitropyrene's but not for benzo[a]pyrene's in-
Shkodra/Skadar using semipermeable membrane devices (SPMDs), bioassays duction of p53 phosphorylation and apoptosis. Toxicology 244 (2e3), 257e270.
and chemical analysis. Environ. Sci. Pollut. Res. Int. 11 (4), 240e253. Sumanasekera, W.K., Ivanova, M.M., Johnston, B.J., Dougherty, S.M.,
tegui-Zirena, E.G., Stewart, P.M., Whatley, A., Chu-Koo, F., Sotero-Solis, V.E.,
Rea Sumanasekera, G.U., Myers, S.R., Ali, Y., Kizu, R., Klinge, C.M., 2007. Rapid effects
Merino-Zegarra, C., Vela-Paima, E., 2014. Polycyclic aromatic hydrocarbon of diesel exhaust particulate extracts on intracellular signaling in human
concentrations, mutagenicity, and Microtox® acute toxicity testing of Peruvian endothelial cells. Toxicol. Lett. 174 (1e3), 61e73.
crude oil and oil-contaminated water and sediment. Environ. Monit. Assess. 186 Sun, Y.W., Guttenplan, J.B., Cooper, T., Krzeminski, J., Aliaga, C., Boyiri, T.,
(4), 2171e2184. Kosinska, W., Zhao, Z.L., Chen, K.M., Berg, A., Amin, S., El-Bayoumy, K., 2013.
Reynaud, S., Duchiron, C., Deschaux, P., 2004. 3-Methylcholanthrene induces Mechanisms underlying the varied mammary carcinogenicity of the environ-
lymphocyte and phagocyte apoptosis in common carp (Cyprinus carpio L) mental pollutant 6-nitrochrysene and its metabolites (-)-[R,R]- and (þ)-[S,S]-
in vitro. Aquat. Toxicol. 66 (3), 307e318. 1,2-dihydroxy-1,2-dihydro-6-nitrochrysene in the rat. Chem. Res. Toxicol. 26
Rice, C.A., Myers, M.S., Willis, M.L., French, B.L., Casillas, E., 2000. From sediment (4), 547e554.
bioassay to fish biomarkereconnecting the dots using simple trophic re- Sundt, R.C., Bjo€ rkblom, C.J., 2011. Toxicol Effects of produced water on reproductive
lationships. Mar. Environ. Res. 50 (1e5), 527e533. parameters in prespawning Atlantic cod (Gadus morhua). J. Toxicol. Environ.
Routledge, E.J., Sumpter, J.P., 1996. Estrogenic activity of surfactants and some of Health A 74 (7e9), 543e554.
their degradation products assessed using a recombinant yeast screen. Environ. Sureda, A., Box, A., Tejada, S., Blanco, A., Caixach, J., Deudero, S., 2011. Biochemical
Toxicol. Chem. 15 (3), 241e248. responses of Mytilus galloprovincialis as biomarkers of acute environmental
Rubin, H., 2001. Synergistic mechanisms in carcinogenesis by polycyclic aromatic pollution caused by the Don Pedro oil spill (Eivissa Island, Spain). Aquat. Tox-
hydrocarbons and by tobacco smoke: a bio-historical perspective with updates. icol. 101 (3e4), 540e549.
Carcinogenesis 22 (12), 1903e1930. Swedenborg, E., Pongratz, I., 2010. AhR and ARNT modulate ER signaling. Toxicology
Ryu, H.Y., Emberley, J.K., Schlezinger, J.J., Allan, L.L., Na, S., Sherr, D.H., 2005. Envi- 268 (3), 132e138.
ronmental chemical-induced bone marrow B cell apoptosis: death receptor- Tamaki, A., Hayashi, H., Nakajima, H., Takii, T., Katagiri, D., Miyazawa, K., Hirose, K.,
independent activation of a caspase-3 to caspase-8 pathway. Mol. Pharmacol. Onozaki, K., 2004. Polycyclic aromatic hydrocarbon increases mRNA level for
68 (4), 1087e1096. interleukin 1 beta in human fibroblast-like synoviocyte line via aryl hydrocar-
Ryu, H.Y., Mann, K.K., Schlezinger, J.J., Jensen, B., Sherr, D.H., 2003. Environmental bon receptor. Biol. Pharm. Bull. 27 (3), 407e410.
chemical-induced pro/pre-B cell apoptosis: analysis of c-Myc, p27Kip1, and Thomas, K.V., Balaam, J., Barnard, N., Dyer, R., Jones, C., Lavender, J., McHugh, M.,
p21WAF1 reveals a death pathway distinct from clonal deletion. J. Immunol. 170 2002. Characterisation of potentially genotoxic compounds in sediments
(10), 4897e4904. collected from United Kingdom estuaries. Chemosphere 49 (3), 247e258.
Safe, S., Wormke, M., 2003. Inhibitory aryl hydrocarbon receptor-estrogen receptor Thomas, R.E., Lindeberg, M., Harris, P.M., Rice, S.D., 2007. Induction of DNA strand
alpha cross-talk and mechanisms of action. Chem. Res. Toxicol. 16 (7), 807e816. breaks in the mussel (Mytilus trossulus) and clam (Protothaca staminea)
Safe, S., Wormke, M., Samudio, I., 2000. Mechanisms of inhibitory aryl hydrocarbon following chronic field exposure to polycyclic aromatic hydrocarbons from the
receptor-estrogen receptor crosstalk in human breast cancer cells. J. Mammary Exxon Valdez spill. Mar. Pollut. Bull. 54 (6), 726e732.
Gland. Biol. Neoplasia 5 (3), 295e306. Tian, S., Pan, L., Sun, X., 2013. An investigation of endocrine disrupting effects and
Schmidt, J.V., Bradfield, C.A., 1996. Ah receptor signaling pathways. Annu. Rev. Cell toxic mechanisms modulated by benzo[a]pyrene in female scallop Chlamys
Dev. Biol. 12, 55e89. farreri. Aquat. Toxicol. 144e145, 162e171.
Schnelle-Kreis, J., Küpper, U., Sklorz, M., Cyrys, J., Briede , J.J., Peters, A., Tian, Y., 2009. Ah receptor and NF-kappaB interplay on the stage of epigenome.
Zimmermann, R., 2009. Daily measurement of organic compounds in ambient Biochem. Pharmacol. 77 (4), 670e680.
particulate matter in Augsburg, Germany: new aspects on aerosol sources and Tintos, A., Gesto, M., Alvarez, R., Míguez, J.M., Soengas, J.L., 2006. Interactive effects
aerosol related health effects. Biomarkers 14 (Suppl. 1), 39e44. of naphthalene treatment and the onset of vitellogenesis on energy metabolism
Schraplau, A., Schewe, B., Neuscha €fer-Rube, F., Ringel, S., Neuber, C., Kleuser, B., in liver and gonad, and plasma steroid hormones of rainbow trout Onco-
Püschel, G.P., 2015. Enhanced thyroid hormone breakdown in hepatocytes by rhynchus mykiss. Comp. Biochem. Physiol. C Toxicol. Pharmacol. 144 (2),
mutual induction of the constitutive androstane receptor (CAR, NR1I3) and 155e165.
arylhydrocarbon receptor by benzo[a]pyrene and phenobarbital. Toxicology Tolbert, P.E., 1997. Oils and cancer. Cancer Causes Control 8 (3), 386e405.
328, 21e28. Tollefsen, K.E., Sundt, R.C., Beyer, J., Meier, S., Hylland, K., 2011. Endocrine modu-
Sen, A., Ulutas, O.K., Tutuncu, B., Ertas, N., Cok, I., 2010. Determination of 7- lation in Atlantic cod (Gadus morhua L.) exposed to alkylphenols, poly-
ethoxyresorufin-o-deethylase (EROD) induction in leaping mullet (Liza aromatichydrocarbons, produced water, and dispersed oil. J. Toxicol. Environ.
824 Y. Zhang et al. / Environmental Pollution 213 (2016) 809e824

Health A 74 (7e9), 529e542. with polyaromatic hydrocarbons by chemical and in vitro bioassay techniques.
€rneman, N., Yang, X., Bååth, E., Bengtsson, G., 2008. Spatial covariation of mi-
To Environ. Toxicol. Chem. 20 (7), 1499e1506.
crobial community composition and polycyclic aromatic hydrocarbon concen- Wagner, M., Bolm-Audorff, U., Hegewald, J., Fishta, A., Schlattmann, P., Schmitt, J.,
tration in a creosote-polluted soil. Environ. Toxicol. Chem. 27 (5), 1039e1046. Seidler, A., 2015. Occupational polycyclic aromatic hydrocarbon exposure and
Totlandsdal, A.I., Låg, M., Lilleaas, E., Cassee, F., Schwarze, P., 2015. Differential risk of larynx cancer: a systematic review and meta-analysis. Occup. Environ.
proinflammatory responses induced by diesel exhaust particles with contrast- Med. 72 (3), 226e233.
ing PAH and metal content. Environ. Toxicol. 30 (2), 188e196. Wang, J., Xie, P., Kettrup, A., Schramm, K.W., 2005. Evaluation of soot particles of
Totlandsdal, A.I., Øvrevik, J., Cochran, R.E., Herseth, J.I., Bølling, A.K., Låg, M., biomass fuels with endocrine-modulating activity in yeast-based bioassay.
Schwarze, P., Lilleaas, E., Holme, J.A., Kuba tova, A., 2014. The occurrence of Anal. Bioanal. Chem. 381 (8), 1609e1618.
polycyclic aromatic hydrocarbons and their derivatives and the proin- Wenger, D., Gerecke, A.C., Heeb, N.V., Schmid, P., Hueglin, C., Naegeli, H., Zenobi, R.,
flammatory potential of fractionated extracts of diesel exhaust and wood 2009. In vitro estrogenicity of ambient particulate matter: contribution of hy-
smoke particles. J. Environ. Sci. Health A 49 (4), 383e396. droxylated polycyclic aromatic hydrocarbons. J. Appl. Toxicol. 29 (3), 223e232.
Tran, D.Q., Ide, C.F., McLachlan, J.A., Arnold, S.F., 1996. The anti-estrogenic activity of Westerholm, R., Egeba €ck, K.E., 1994. Exhaust emissions from light- and heavy-duty
selected polynuclear aromatic hydrocarbons in yeast expressing human estro- vehicles: chemical composition, impact of exhaust after treatment, and fuel
gen receptor. Biochem. Biophys. Res. Commun. 229 (1), 101e108. parameters. Environ. Health Perspect. 102 (Suppl. 4), 13e23.
Traversi, D., Schiliro , T., Degan, R., Pignata, C., Alessandria, L., Gilli, G., 2011. Weston, J., Warren, C., Chaudhary, A., Emerson, B., Argote, K., Khan, S., Willett, K.L.,
Involvement of nitro-compounds in the mutagenicity of urban Pm2.5 and Pm10 2010. Use of bioassays and sediment polycyclic aromatic hydrocarbon con-
in Turin. Mutat. Res. 726 (1), 54e59. centrations to assess toxicity at coastal sites impacted by Hurricane Katrina.
Tsai, K.S., Yang, R.S., Liu, S.H., 2004. Benzo[a]pyrene regulates osteoblast prolifera- Environ. Toxicol. Chem. 29 (7), 1409e1418.
tion through an estrogen receptor-related cyclooxygenase-2 pathway. Chem. Wittkopp, S., Staimer, N., Tjoa, T., Gillen, D., Daher, N., Shafer, M., Schauer, J.J.,
Res. Toxicol. 17 (5), 679e684. Sioutas, C., Delfino, R.J., 2013. Mitochondrial genetic background modifies the
Tsuji, G., Takahara, M., Uchi, H., Matsuda, T., Chiba, T., Takeuchi, S., Yasukawa, F., relationship between traffic-related air pollution exposure and systemic bio-
Moroi, Y., Furue, M., 2012. Identification of ketoconazole as an AhR-Nrf2 acti- markers of inflammation. PLoS One 8 (5), e64444.
vator in cultured human keratinocytes: the basis of its anti-inflammatory effect. Wormley, D.D., Ramesh, A., Hood, D.B., 2004. Environmental contaminant-mixture
J. Invest. Dermatol. 132 (1), 59e68. effects on CNS development, plasticity, and behavior. Toxicol. Appl. Pharmacol.
Tuominen, R., Warholm, M., Mo € ller, L., Rannug, A., 2003. Constitutive CYP1B1 mRNA 197 (1), 49e65.
expression in human blood mononuclear cells in relation to gender, genotype, Wu, M.T., Lee, T.C., Wu, I.C., Su, H.J., Huang, J.L., Peng, C.Y., Wang, W., Chou, T.Y.,
and environmental factors. Environ. Res. 93 (2), 138e148. Lin, M.Y., Lin, W.Y., Huang, C.T., Pan, C.H., Ho, C.K., 2011. Whole genome
Van de Wiele, T., Vanhaecke, L., Boeckaert, C., Peru, K., Headley, J., Verstraete, W., expression in peripheral-blood samples of workers professionally exposed to
Siciliano, S., 2005. Human colon microbiota transform polycyclic aromatic hy- polycyclic aromatic hydrocarbons. Chem. Res. Toxicol. 24 (10), 1636e1643.
drocarbons to estrogenic metabolites. Environ. Health Perspect. 113 (1), 6e10. Xu, X., Yekeen, T.A., Xiao, Q., Wang, Y., Lu, F., Huo, X., 2013. Placental IGF-1 and
Van Dolah, R.F., Riekerk, G.H., Levisen, M.V., Scott, G.I., Fulton, M.H., Bearden, D., IGFBP-3 expression correlate with umbilical cord blood PAH and PBDE levels
Sivertsen, S., Chung, K.W., Sanger, D.M., 2005. An evaluation of polycyclic aro- from prenatal exposure to electronic waste. Environ. Pollut. 182, 63e69.
matic hydrocarbon (PAH) runoff from highways into estuarine wetlands of Yan, Z., Zhang, H., Maher, C., Arteaga-Solis, E., Champagne, F.A., Wu, L.,
South Carolina. Arch. Environ. Contam. Toxicol. 49 (3), 362e370. McDonald, J.D., Yan, B., Schwartz, G.J., Miller, R.L., 2014. Prenatal polycyclic ar-
van Grevenynghe, J., Rion, S., Le Ferrec, E., Le Vee, M., Amiot, L., Fauchet, R., omatic hydrocarbon, adiposity, peroxisome proliferator-activated receptor
Fardel, O., 2003. Polycyclic aromatic hydrocarbons inhibit differentiation of (PPAR) g methylation in offspring, grand-offspring mice. PLoS One 9 (10),
human monocytes into macrophages. J. Immunol. 170 (5), 2374e2381. e110706.
van Schooten, F.J., Maas, L.M., Moonen, E.J., Kleinjans, J.C., van der Oost, R., 1995. Yang, L., Liu, G., Lin, Z., Wang, Y., He, H., Liu, T., Kamp, D.W., 2016. Pro-inflammatory
DNA dosimetry in biological indicator species living on PAH-contaminated soils response and oxidative stress induced by specific components in ambient
and sediments. Ecotoxicol. Environ. Saf. 30 (2), 171e179. particulate matter in human bronchial epithelial cells. Environ. Toxicol. in press.
Villalobos-Pietrini, R., Herna ndez-Mena, L., Amador-Mun ~ oz, O., Munive-Colín, Z., Zhang, Y., Ding, J., Shen, G., Zhong, J., Wang, C., Wei, S., Chen, C., Chen, Y., Lu, Y.,
Bravo-Cabrera, J.L., Go mez-Arroyo, S., Frías-Villegas, A., Waliszewski, S., Ram- Shen, H., Li, W., Huang, Y., Chen, H., Su, S., Lin, N., Wang, X., Liu, W., Tao, S., 2014.
írez-Pulido, J., Ortiz-Mun ~ iz, R., 2007. Biodirected mutagenic chemical assay of Dietary and inhalation exposure to polycyclic aromatic hydrocarbons and uri-
PM(10) extractable organic matter in Southwest Mexico City. Mutat. Res. 634 nary excretion of monohydroxy metabolitesea controlled case study in Beijing,
(1e2), 192e204. China. Environ. Pollut. 184, 515e522.
Villeneuve, D.L., Khim, J.S., Kannan, K., Giesy, J.P., 2002. Relative potencies of indi- Zhang, Y., Tao, S., Shen, H., Ma, J., 2009. Inhalation exposure to ambient polycyclic
vidual polycyclic aromatic hydrocarbons to induce dioxinlike and estrogenic aromatic hydrocarbons and lung cancer risk of Chinese population. Proc. Natl.
responses in three cell lines. Environ. Toxicol. 17 (2), 128e137. Acad. Sci. U. S. A. 106 (50), 21063e21067.
Volkov, M.S., Bolotina, N.A., Evteev, V.A., Koblyakov, V.A., 2012. Ah-receptor-inde- Zhao, Y., Chen, X., Liu, X., Ding, Y., Gao, R., Qiu, Y., Wang, Y., He, J., 2014. Exposure of
pendent stimulation of hepatoma 27 culture cell proliferation by polycyclic mice to benzo(a)pyrene impairs endometrial receptivity and reduces the
aromatic hydrocarbons. Biochem. (Mosc) 77 (2), 201e207. number of implantation sites during early pregnancy. Food Chem. Toxicol. 69,
Vondr acek, J., Kozubík, A., Machala, M., 2002. Modulation of estrogen receptor- 244e251.
dependent reporter construct activation and G0/G1-S-phase transition by Zapata-Perez, O., Del-Rio, M., Dominguez, J., Chan, R., Ceja, V., Gold-Bouchot, G.,
polycyclic aromatic hydrocarbons in human breast carcinoma MCF-7 cells. 2005. Preliminary studies of biochemical changes (ethoxycoumarin O-deethy-
Toxicol. Sci. 70 (2), 193e201. lase activities and vitellogenin induction) in two species of shrimp (Farfante-
Vondr acek, J., Machala, M., Minksova , K., Bla
ha, L., Murk, A.J., Kozubík, A., penaeus duorarum and Litopenaeus setiferus) from the Gulf of Mexico.
Hofmanova , J., Hilscherova, K., Ulrich, R., Ciganek, M., Neca, J., Svrckova , D., Ecotoxicol. Environ. Saf. 61 (1), 98e104.
Holoubek, I., 2001. Monitoring river sediments contaminated predominantly

You might also like