You are on page 1of 8

Journal of Hazardous Materials 283 (2014) 897–904

Contents lists available at ScienceDirect

Journal of Hazardous Materials


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

Effect of titanium dioxide nanoparticles on the bioavailability,


metabolism, and toxicity of pentachlorophenol in zebrafish larvae
Qi Fang a,b , Xiongjie Shi c , Liping Zhang a , Qiangwei Wang a,b , Xianfeng Wang a,b ,
Yongyong Guo a , Bingsheng Zhou a,∗
a
State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
b
Graduate University of Chinese Academy of Sciences, Beijing 100039, China
c
College of Life Sciences, Wuhan University, Wuhan 430072, China

h i g h l i g h t s

• Effects of n-TiO2 on toxicity of PCP in zebrafish larvae were investigated.


• Co-exposure n-TiO2 enhanced metabolism of PCP to tetrachlorohydroquinone in larvae.
• Co-exposure n-TiO2 increased oxidative damage and developmental toxicity in larvae.
• NPs may influence toxicity of associated organic pollutants in the aquatic environment.

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

Article history: This study investigated the influence of titanium dioxide nanoparticles (n-TiO2 ) on the bioavailability,
Received 8 August 2014 metabolism, and toxicity of pentachlorophenol (PCP) in fish. Zebrafish (Danio rerio) embryos or larvae
Received in revised form 14 October 2014 (2-h post-fertilization) were exposed to PCP (0, 3, 10, and 30 ␮g/L) alone or in combination with n-TiO2
Accepted 27 October 2014
(0.1 mg/L) until 6 days post-fertilization. Results showed that n-TiO2 treatment alone did not induce
lipid peroxidation, DNA damage, as well as the generation of reactive oxygen species (ROS) in the larvae.
Keywords:
As compared with PCP treatment, the co-exposure of PCP and n-TiO2 enhanced the induction of ROS
Pentachlorophenol
generation, eventually leading to lipid peroxidation and DNA damage. The nuclear factor erythroid 2-
Nano-titanium dioxide
Oxidative damage
related factor 2 gene transcriptions were significantly upregulated in both PCP treatment alone and in
Bioavailability and metabolism combination with n-TiO2 . Chemical analysis and histological examination showed that n-TiO2 adsorb
Zebrafish larvae PCP, and n-TiO2 are taken up by developing zebrafish larvae; however, PCP content was not enhanced in
the presence of n-TiO2 , but the metabolism of PCP to tetrachlorohydroquinone was enhanced in larvae.
The results indicate that n-TiO2 enhanced the metabolism of PCP and caused oxidative damage and
developmental toxicity, suggesting that NPs can influence the fate and toxicity of associated organic
pollutants in the aquatic environment.
© 2014 Elsevier B.V. All rights reserved.

1. Introduction may influence the bioavailability, metabolism, fate, and toxicity of


a toxicant to aquatic biota. Thus, interactions between the toxicant
The increasing production and use of manufactured nanopar- and organisms are of particular concern for the environmental risk
ticles (NPs) have inevitably led to their release into the aquatic assessment of organic contaminants.
environment, thereby posing a threat to aquatic organisms [1,2]. Among NPs, titanium dioxide nanoparticles (n-TiO2 ) are one of
As a result, a mixture of NPs and other environmental toxicants is the most popular manufactured nanomaterials in various industrial
present in the environment. In this regard, NPs are highly insoluble and environmental applications, such as, catalysts for the decom-
and have a large surface area to volume ratio, suggesting that they position of organic contaminants [5]. As a consequence, n-TiO2
are likely to interact with other substances in water [2–4]. Thus, NPs are inevitably released into the environment and enter into the
aquatic environment [6]. Although it has been confirmed that n-
TiO2 exhibited low acute toxicity to fish species and other aquatic
invertebrates on the order of tens to hundreds of mg/L (e.g., Daph-
∗ Corresponding author. Tel.: +86 727768780042; fax: +86 727768780123. nia magna) [7–11], several studies have shown that n-TiO2 adsorb
E-mail address: bszhou@ihb.ac.cn (B. Zhou).

http://dx.doi.org/10.1016/j.jhazmat.2014.10.039
0304-3894/© 2014 Elsevier B.V. All rights reserved.
898 Q. Fang et al. / Journal of Hazardous Materials 283 (2014) 897–904

organic contaminants and enhances the uptake of organic pollu- sonication (50 W/L, 40 kHz) for 20 min. Test n-TiO2 solutions were
tants (e.g., tributyltin, polybrominated diphenyl ethers [PBDEs]) in prepared immediately prior to use by diluting the stock solutions
organisms [12,13]. Importantly, n-TiO2 are one of the common- with fresh charcoal-filtered water and sonicating (50 W/L, 40 kHz)
est materials used to degrade organic compounds, mainly on their for 20 min [34]. The test solution was renewed every 24 h with new
photocatalytic activity at the absorption of ultraviolet (UV) light n-TiO2 to maintain relatively consistent levels of n-TiO2 exposure.
at specific wavelengths (especially at 300–388 nm), via generation The properties of commercial n-TiO2 have been characterized
of reactive oxygen species (ROS) that can cause decomposition of in a previous study [13]. The crystal structure was confirmed
organic molecules [14–16]. Such interactions between n-TiO2 and by powder X-ray diffraction (XRD) using an X’Pert PRO XRD
other toxicants may modify the environmental behavior and/or instrument (PANalytical, Almelo, the Netherlands). The diameter
toxicity of toxicants to aquatic organisms, and therefore, of par- sizes were determined by transmission electron microscopy (TEM;
ticular concern for environmental risk assessments. JEOL2010F) at an acceleration voltage of 100 kV. The surface area of
Pentachlorophenol (PCP) has been extensively used as her- nano-TiO2 was measured by the Brunauer–Emmett–Teller method
bicide, algaecide, wood preservative, defoliant, and fungicide using an ASAP 2020 physisorption analyzer (Micromeritics, Atlanta,
[17–20]. Owing to the past widespread application and persistent US). The average diameter and ␨ potential in water (0.1 mg/L) were
properties, resistance to biodegradation, potential bioaccumu- determined by dynamic light scattering using a Zetasizer Nano ZS
lation, and biomagnification, PCP resulted in environmental (Malvern instruments, Worcestershire, UK).
contamination, and it is a common water contaminant [19–22].
The use of PCP in China was restricted in 1997. However, with 2.3. Zebrafish maintenance and experiment design
the re-emergence of schistosomiasis in the traditionally epidemic
areas, the production and use of PCP for snail elimination and The maintenance of wild-type zebrafish (Danio rerio, AB strain)
schistosomiasis control were warranted once again in some areas and exposure to their embryos were performed as described pre-
[23,24]. As a consequence, PCP has resulted in widespread envi- viously [13]. Briefly, embryos that had developed normally and
ronmental contamination in the local water environment where reached the blastula stage (2 h post-fertilization) were selected for
PCP has been detected in surface waters in China (up to 1.15 ␮g/L) the experiments. The zebrafish embryos were exposed to PCP alone
[20,25]. A high PCP concentration has been detected in Dongting or in combination with n-TiO2 in the exposure solution, containing
Lake (up to 103.7 ␮g/L), which is attributed to its application for the 0.2 mM of Ca(NO3 )2 , 0.13 mM of MgSO4 , 19.3 mM of NaCl, 0.23 mM
control of snailborne schistosomiasis [26–28]. Previous studies indi- of KCl, and 1.67 mM of HEPES [35]. For PCP exposure, the embryos
cate that principal mechanisms of action of PCP-induced toxicity were randomly distributed into glass beakers containing 500 mL
seem to be related to the uncoupling of oxidative phosphoryla- PCP solution at the environmental-related concentration (0, 3, 10,
tion in mitochondria and the generation of reactive oxygen species and 30 ␮g/L). In parallel, experiments were conducted with a com-
(ROS) [17,29]. PCP undergoes oxidative dechlorination to form bination of PCP and n-TiO2 (0, 3, 10, and 30 ␮g/L PCP; 0.1 mg/L
tetrachlorohydroquinone (TCHQ) by cytochrome P450 enzymes, n-TiO2 ). There were three replicates for each exposure concen-
resulting in oxidative stress on cells and tissues [30,31]. TCHQ tration, and each beaker contained ≈400 embryos. The n-TiO2
seems to be a more toxic metabolic product of PCP; it can induce exposure concentration was based on our previous findings: after
DNA strand breakage and deplete glutathione content [32,33]. exposure to this n-TiO2 concentration (0.1 mg/L), neither devel-
This study, quantifies the potential effect of n-TiO2 on PCP with opmental toxicity nor oxidative stress was observed in zebrafish
respect to the uptake, bioconcentration, and metabolism of PCP in larvae [13]. A previous study also indicated that exposure to n-
the presence of n-TiO2 in zebrafish larvae. As PCP was reported TiO2 (20 nm; up to 500 mg/L; during 96 h exposure) neither affected
to cause ROS generation, oxidative stress, and DNA damage, we hatching nor induced deformity in zebrafish embryos [12]. In addi-
investigated the potential effects on developmental toxicity as tion, with the increased production and application of n-TiO2 , levels
well as oxidative damage and genotoxicity using the 8-hydroxy- of n-TiO2 in the environment are expected to increase, and also
2-deoxyguanosine (8-OHdG) marker. Finally, responsive genes because of their persistent properties, 0.1 mg/L n-TiO2 is in the same
related to ROS stress (e.g., nuclear factor erythroid 2-related factor order of magnitude to the environmental concentration (predicted
2, Nrf2) were also examined. Our results suggest that the presence as 16 ␮g/L in Switzerland) [36].
of n-TiO2 enhanced the metabolism of PCP and caused oxidative DMSO 0.01% (v/v) was added to both control and expo-
damage, which lead to developmental toxicity in zebrafish larvae. sure groups. The embryos or larvae were exposed until 6 days
post-fertilization (dpf); at this stage, the yolk was absorbed com-
2. Materials and methods pletely. During the experimental period, the exposure solution was
renewed daily. The temperature was maintained at 28 ± 0.5 ◦ C, and
2.1. Reagents the photoperiod was set to 14-h light/10-h dark throughout the
entire experiment. At 6 dpf, the larvae were randomly sampled,
Pentachlorophenol (PCP) (>99%, CAS No. 87-86-5) and MS222 immediately frozen in liquid nitrogen, and stored at −80 ◦ C for
were purchased from Sigma (St. Louis, MO, USA). Stock solutions subsequent gene, ROS, maliondialdehyde (MDA), 8-OHdG, super-
were dissolved in dimethyl sulfoxide (DMSO) and were stored at oxide dismutase (SOD), glutathione (GSH), and chemical analysis.
4 ◦ C. n-TiO2 were purchased from Hangzhou Wan Jing New Material The developing toxicity (e.g., hatching, malformation, growth, and
Company (CAS: 13,463-67-7; purity > 99.9%, China). The diameter survival) was also recovered.
of n-TiO2 was 25 nm according to the manufacturer’s specifications.
All other chemicals used in this study were of analytical grade, 2.4. Chemical analysis of PCP and its metabolites in water and
while HPLC grade and trace metal analysis grade chemicals were larvae
used for the chemical analysis of PCP and for the measurement of
n-TiO2 , respectively. PCP could either undergo photolysis or be degraded by n-TiO2 ,
and the degradation products could be TCHQ and chlorophe-
2.2. Preparation and characterization of n-TiO2 nols (e.g. 2,3,4,6-tetrachlorophenol, 2,4,6-trichlorophenol, and
2,6-dichlorophenol) in water [37]. We measured PCP concen-
Stock solutions of 1 mg/mL n-TiO2 (25 nm) were prepared by trations and the possible photocatalytic degradation products in
dispersing the NPs in ultrapure water (Millipore, Billerica, MA) with exposure media. A water sample (100 mL) was taken at 6 dpf before
Q. Fang et al. / Journal of Hazardous Materials 283 (2014) 897–904 899

renewing the exposed water in all treated groups (n = 3 replicates). was performed with a calibration cure with a Ti standard. Ti
Briefly, the water sample was acidified to a pH of 4, and tribro- concentrations were converted to TiO2 concentrations by molec-
mophenol (TBP) was added as a standard to check the recovery of ular weight conversion. The recovery for this TiO2 quantification
the extraction procedure, and the TBP concentration was 10 ␮g/L. technique ranged from 94% to 107%. The measured n-TiO2 was
After being enriched by passing through an HLB column (Waters, expressed as TiO2 .
500 mg, 6 mL) [38], the solution was dried under nitrogen flow
and diluted in acetonitrile–ultrapure water (3:7) for analysis. The 2.7. Adsorption of PCP on n-TiO2
recovery of this method ranged from 83% to 105%.
For the bioavailability of PCP in the larvae, the extraction, clean We examined the adsorption of PCP on n-TiO2 . PCP (30 ␮g/L)
up, analysis, and quality assurance and quality control were per- was mixed with a suspension of n-TiO2 (0.1 mg/L). At 0, 2, 4, 8, 12,
formed according to China standard for determining PCP [39]. 20, and 24 h, 100 mL of the mixture was centrifuged for 5 min at
Briefly, 200 zebrafish larvae were collected, rinsed three times 12,000 × g using a high-speed centrifuge to separate n-TiO2 par-
with ultrapure water for 15 s to remove external n-TiO2 and PCP ticles. The supernatant was then collected and further analyzed
that was adsorbed on the surface. Previous studies have shown by HPLC–MS (Agilent 1100HPLC/MSD Trap, CA). The adsorption
that little n-TiO2 was adhered to the surface of fish larvae [40,41]. amount of PCP was evaluated as the decrease in the PCP concen-
After freeze-drying, the samples were weighed, added to 5 mL tration over time. This experiment was performed in a dark room
5% triethylamine in acetonitrile–water, and ultrasonicated for to avoid PCP degradation.
5 min. After centrifugation, the supernatant was collected. Through
enrichment by a MAX column (Oasis, 60 mg, 3 mL; Waters), the
2.8. ROS, lipid peroxidation assessment, and antioxidant enzyme
solution was dried under nitrogen flow and diluted by formic
assays
acid–methanol–water (v/v/v, 1:49:50) for analysis. Quantification
was conducted by a calibration curve created with a PCP standard.
The generation of ROS in the larvae at 6 dpf was measured using
Procedural blanks were analyzed simultaneously. The recovery of
dichlorofluorescein diacetate as previously described [43]. Briefly,
standard addition in the blank was 85.92% (n = 6).
20 larvae (n = 3) were washed two times with cold PBS (pH 7.4)
In this study, the TCHQ content was measured in the larvae
and then homogenized in cold buffer (0.32 mM sucrose, 20 mM
exposed to 30 ␮g/L PCP and co-exposure groups. The procedures
HEPES, 1 mM MgCl2 , and 0.5 mM) phenylmethylsulfonylfluoride
for extraction, clean up, and analysis were performed as previ-
(pH 7.4). The homogenate was centrifuged at 15,000 × g at 4 ◦ C for
ously described [42]. Briefly, 500 larvae were collected, cleaned,
20 min, and the supernatant was transferred to new tubes for fur-
freeze-dried, and weighed. After extraction with diethyl ether,
ther experiments. The fluorescence intensity was measured using
the solution was dried under nitrogen flow and diluted using
a microplate reader (Molecular Device, M2, Union City, CA) with
acetonitrile–ultrapure water (3:7). The recovery of standard addi-
excitation and emission at 485 and 530 nm, respectively. The ROS
tion in the blank was 80.33% (n = 3). Procedural blanks were
concentration was expressed in arbitrary units (DCF mg−1 protein).
analyzed simultaneously. Quantification was conducted by a cal-
Lipid peroxidation was assessed by measuring the amount of
ibration curve created with a TCHQ standard.
malondialdehyde (MDA) (trace MDA assay kit, Nanjing Jiancheng
The samples were analyzed for PCP, TCHQ, and several
Bioengineering Institute, Nanjing, China) according to the manu-
chlorophenols using HPLC/MS (Agilent 1100HPLC/MSD Trap, CA)
facturer’s introductions. Briefly, 100 larvae (n = 3) were collected
employed with a DAD detector. The limit of detection was 1 ␮g/L,
and homogenized with 200 ␮L normal saline. Then, the lysate was
which was calculated as three times the S/N from six runs for the
centrifuged at 5000 × g for 5 min, and the supernatant was collected
ongoing precision.
for MDA detection. The level of MDA is expressed as nanomoles per
milligram of protein.
2.5. TEM examination of n-TiO2 in larvae
The activity of total SOD and GSH content was determined using
a diagnostic reagent kit (Nanjing Jiancheng Bioengineering Insti-
Transmission electron microscopy was performed to confirm
tute, Nanjing, China) according to the manufacturer’s introductions.
the uptake of n-TiO2 by zebrafish larvae. Larvae were collected
The larvae (100, n = 3) were collected and homogenized with 400 ␮L
and euthanized in MS222, washed in phosphate buffer (PBS, 0.1 M,
normal saline. Then, the lysate was centrifuged at 5000 × g for
pH 7.4), and fixed with 2.5% glutaraldehyde in the same PBS
5 min, and the supernatant was collected for SOD and GSH detec-
overnight. They were then washed in the buffer, post-fixed with
tion. The absorbance was measured at 550 nm and 405 nm for SOD
1% osmium tetroxide, washed, dehydrated, and embedded in epoxy
and GSH, respectively. The level of GSH is expressed as micromoles
resin (Spurr’s). Ultrathin sections (60–80 nm) were obtained using
per gram of protein, and the activity of SOD is expressed as unit per
an ultramicrotome (Leica EMUC6, Germany) and observed using a
milligram of protein.
Hitachi (HT-7700) TEM at 80 kV. The samples were not stained to
better reveal n-TiO2 particles [40].
2.9. 8-OHdG detection
2.6. Quantification of n-TiO2 in larvae by HPLC–ICP-MS
8-OHdG was measured by the enzyme-linked immunosorbent
The quantification of n-TiO2 concentrations in larvae was per- assay (ELISA). The larvae (100, n = 3) were collected and homoge-
formed as previously described [13]. Briefly, 100 larvae from each nized with 200 ␮L normal saline. Then, the lysate was centrifuged
replicate were washed 5 times and weighed after at 5000 × g for 5 min, and the supernatant was collected for 8-OHdG
freeze-drying. Then, the samples were digested in 6 mL HNO3 detection (FISH 8-OHdG ELISA Kit Nanjing Jiancheng Bioengi-
using a microwave digestion system (Multiwave 3000, Anton neering Institute, Nanjing, China) according to the manufacturer’s
Paar, Austria) to decompose TiO2 into Ti4+ and evaporated introductions. The optical density was evaluated at 450 nm within
to dryness at 140 ◦ C. Nitric acid (2%) was added to the 15 min. The 8-OHdG concentration was calculated according to the
samples to a constant volume to 5 mL. There were three standard curve and expressed in arbitrary units (ng/mg protein).
replicates (i = 3) for the measurement for both the water sam- The protein content in our experiments was measured with a
ples and larvae. The Ti4+ concentrations were determined by Bradford protein assay kit (Beyotime Institute of Biotechnology,
HPLC–ICP-MS (NexION300X, PerkinElmer, MA). Quantification Jiangsu, China).
900 Q. Fang et al. / Journal of Hazardous Materials 283 (2014) 897–904

2.10. RNA isolation and quantitative real-time polymerase chain (A)


reaction (qRT-PCR)
12
PCP #
The total RNA was extracted from 30 homogenized zebrafish lar- PCP+n-TiO2

PCP content (µg/L)


vae (n = 3) using TRIzol Reagent (Invitrogen, Carlsbad, CA) according
to the manufacturer’s instructions. The extraction, purification, and 8
quantification of the total RNA and the synthesis of first-strand
cDNA were performed as described previously [43]. The primer
sequences of the selected genes were obtained using the online 4
program Primer3 (http://frodo.wi.mit.edu/; see Table S1). The ribo-
somal protein L8 (rpl8) was selected as the housekeeping gene,
which does not vary upon PCP exposure (data not shown). 0
0 3 10 30
2.11. Statistical analysis PCP conce ntration (µg/L)

All data were initially verified for normality and homogeneity


(B)
of variance using the Kolmogorov–Smirnov test and Levene’s test.
All data were expressed as the mean ± standard error (SEM). The 100
PCP
differences of the control and each exposure group were evaluated #

PCP content ( µg/g.dw)


PCP+n-TiO2
by one-way analysis of variance (ANOVA) followed by Dunett’s test 75
or two-way ANOVA followed by Tukey’s test using SPSS 13.0 (SPSS,
Chicago, IL). The differences between groups exposed to PCP alone
and co-exposure with n-TiO2 were also compared using the Stu- 50
dent’s t-test; a P value < 0.05 was considered to be a statistically
significant difference. 25

3. Results 0
0 3 10 30
3.1. Characterization of n-TiO2 PCP conce ntration (µg/L)

XRD revealed that peaks of n-TiO2 matched those expected Fig. 1. Content of PCP: (A) in exposure water and (B) in larvae measured after
for the crystal phase of anatase (JCPDF No: 89-4921). The mea- exposed to nominal PCP concentrations (0, 3, 10, and 30 ␮g/L) and/or in the presence
of 0.1 mg/L n-TiO2 for 6 days. Data are expressed as means ± SEM of three replicate
sured particle size was close to the manufacturer’s information samples. Significance differences (P < 0.05) are indicated by # vs. the corresponding
(mean ± SEM, n = 30, 27.73 ± 0.98 nm). The ␨ potential of n-TiO2 was PCP groups without n-TiO2 (two-way ANOVA followed by Tukey’s test).
−26.6 mV, and the particle size in suspension was 164.07 ± 6.99 nm,
which revealed that n-TiO2 could aggregate in water (0.1 mg/L) (Fig.
S1). The surface area of n-TiO2 was measured as 48.7 m2 /g. 3.5. Quantification of n-TiO2 in zebrafish larvae

In the larvae, the TiO2 concentration was measured as


3.2. Developmental toxicity 103.3 ± 4.8, 107.4 ± 5.4, 103.0 ± 22.3, and 111.7 ± 13.4 ␮g/g dry
weight (dw) in 0, 3, 10, and 30 ␮g/L PCP co-exposure with 0.1 mg/L
The overall hatching rates were over 90%, and survival rates n-TiO2 , respectively. There was no significant difference in the n-
were over 85% in all groups. As compared to the control, there TiO2 concentration between the n-TiO2 treatment alone and in
was no significant influence on hatching and mortality rates in combination of PCP, indicating that the presence of PCP did not
both PCP and PCP with n-TiO2- treated groups. A significant increase affect the uptake of n-TiO2 in larvae.
in the malformation was recorded in 30 ␮g/L PCP-treated group
(4.7 ± 0.29%) relative to the solvent control (2.5 ± 0.5%), while a
3.6. PCP concentrations in water
significantly higher malformation was further observed in the co-
exposure group (6.2 ± 0.6%) compared to the PCP-treated group
No degradation products of PCP were detected in all the
(4.7 ± 0.29%).
exposure water. The PCP concentrations in exposure water were
measured before the renewal of the exposure solutions. The
3.3. Adsorption of PCP on n-TiO2 measured PCP concentrations (n = 3 replicates) were 0.45 ± 0.01,
2.89 ± 0.50, and 8.91 ± 0.42 ␮g/L in the 3, 10, and 30 ␮g/L PCP expo-
The time-course adsorption kinetics is shown in Fig. S2. The sure groups, respectively (Fig. 1). In the groups exposed to PCP and
measured PCP concentration in water was stable over time. After n-TiO2 , the measured PCP concentrations (n = 3 replicates) were
mixing PCP (30 ␮g/L) with 0.1 mg/L n-TiO2 , the measured PCP con- 0.22 ± 0.01, 2.37 ± 0.32, and 7.11 ± 0.33 ␮g/L, respectively (Fig. 1).
centration in water decreased over time. The measured PCP concentration in the co-exposure group was sig-
nificant lower (25%) than that in the PCP-treated group (two-way
3.4. Transmission electron microscopy ANOVA followed by Tukey’s test, P < 0.001, Fig. 1).

TEM observation revealed that n-TiO2 aggregates were dis- 3.7. Bioconcentration and metabolism of PCP in larvae
tributed in larval tissues with no tissue specificity and uniform
distribution. In addition, n-TiO2 aggregates were also observed in The bioconcentration of PCP was measured (5.50 ± 1.64,
mitochondria; TEM observation thus confirmed that n-TiO2 was 18.30 ± 2.18, and 70.54 ± 6.89 ␮g/g, dry weight (dw)) in the larvae
taken up by zebrafish larvae (Fig. S3). exposed to 3, 10, and 30 ␮g/L PCP groups, respectively (Fig. 2A).
Q. Fang et al. / Journal of Hazardous Materials 283 (2014) 897–904 901

(A) Dunnett’s test, 3 ␮g/L, P < 0.05; 10 ␮g/L, P < 0.05; 30 ␮g/L, P < 0.05;
Fig. 3A); ROS generation was further increased in the co-exposure
8 groups compared with the respective PCP treatment (two-way

TCHQ content ( µ g/g.dw)


ANOVA followed by Tukey’s test, 3 ␮g/L, P < 0.05; 10 ␮g/L, P < 0.05;
# 30 ␮g/L, P < 0.05; Fig. 3A).
A significant increase in the MDA level was observed in the
30 ␮g/L PCP group (one-way ANOVA followed by Dunnett’s test,
P < 0.01; Fig. 3B). Compared with PCP exposure alone, the co-
4 exposure of PCP and n-TiO2 resulted in a significantly higher MDA
level (two-way ANOVA followed by Tukey’s test, 10 ␮g/L, P < 0.05;
30 ␮g/L, P < 0.05; Fig. 3B).
The 8-OHdG content was also significantly increased in 30 ␮g/L
PCP group (one-way ANOVA followed by Dunnett’s test, P < 0.05;
Fig. 3C). Likewise, the co-exposure of PCP and n-TiO2 resulted in a
significant higher 8-OHdG content, as compared to PCP treatment
0 alone (two-way ANOVA followed by Tukey’s test, P < 0.05; Fig. 3C).
PCP PCP+n-TiO2 The SOD activity was significantly decreased in 10 and 30 ␮g/L
PCP groups compared to the solvent control (one-way ANOVA fol-
lowed by Dunnett’s test; P < 0.05; Fig. 3D). The SOD activity was
(B) further decreased in the co-exposure groups relative to PCP-treated
groups (two-way ANOVA followed by Tukey’s test, 3 ␮g/L, P < 0.05;
0.18 10 ␮g/L, P < 0.05; 30 ␮g/L, P < 0.05; Fig. 3D).
The reduction of GSH content was observed in 30 ␮g/L PCP alone
compared to the solvent control (one-way ANOVA followed by
# Dunnett’s test; P < 0.05; Fig. 3E); a further reduction of GSH content
0.12 was measured in the co-exposure group compared with the PCP-
TCHQ/PCP

treated group (two-way ANOVA followed by Tukey’s test, P < 0.05;


Fig. 3E).

0.06 3.9. Gene transcription profiles

We observed the gene transcription of nrf2 and sod1 after


exposure to PCP with and without n-TiO2 (Fig. 4). A significant
0.00 upregulation of nrf2 gene transcription (2.2-fold) was observed
PCP PCP+n-TiO2 in 30 ␮g/L PCP-treated groups compared to the solvent control
(one-way ANOVA followed by Dunnett’s test; P < 0.01; Fig. 4A).
Fig. 2. Content of TCHQ in larvae exposed to 30 ␮g/L PCP and/or in the presence of n- In co-exposure groups, nrf2 gene transcription was significantly
TiO2 (0.1 mg/L) for 6 days: (A) TCHQ content and (B) TCHQ/PCP in larvae. Significance upregulated compared with the respective PCP-treated group (two-
differences (P < 0.05) are indicated by # vs. the corresponding PCP groups without way ANOVA followed by Tukey’s test; 10 ␮g/L, P < 0.05; 30 ␮g/L,
n-TiO2 (Student’s t-test).
P < 0.05; Fig. 4A).
A small but significant upregulation of sod1 gene transcription
In the groups co-exposed to PCP and n-TiO2 , PCP concentrations was observed in 10 and 30 ␮g/L PCP exposure groups (1.5-fold
showed a concentration dependence in the larvae (4.40 ± 1.02, and 1.8-fold, respectively) (one-way ANOVA followed by Dunnett’s
16.46 ± 3.39, and 46.74 ± 5.94 ␮g/g dw), respectively (Fig. 2A). We test; 10 ␮g/L, P < 0.05; 30 ␮g/L, P < 0.01; Fig. 4B). The sod1 gene
compared the PCP concentrations in the larvae for PCP exposure was significantly upregulated in the co-exposure group relative to
alone and in combination with n-TiO2 and revealed a significant the PCP-treated group (two-way ANOVA followed by Tukey’s test,
reduction of PCP concentrations in the 30 ␮g/L co-exposure groups P < 0.05; Fig. 4B).
(two-way ANOVA followed by Tukey’s test, P < 0.05, Fig. 2A).
TCHQ is the major metabolite of PCP [42]. The TCHQ con- 4. Discussion
tent was measured from the larvae exposed to 30 ␮g/L PCP alone
(4.56 ± 0.19 ␮g/g dw) and in combination with 0.1 mg/L n-TiO2 This study demonstrated a reduction of PCP content by zebrafish
(3.66 ± 0.16 ␮g/g dw) (Fig. 2B). TCHQ in larvae for 30 ␮g/L PCP alone larvae in the presence of n-TiO2 . Furthermore, in combination with
was significantly higher than that in combination of PCP with n- n-TiO2 , the metabolism of PCP could be enhanced, leading to PCP-
TiO2 (Student’s t-test, P < 0.05; Fig. 2B). However, the TCHQ/PCP induced oxidative damage and genotoxicity to zebrafish embryos
in the co-exposure group was significantly higher than that in the or larvae. PCP also caused the upregulation of genes related to
PCP-exposed group (Student’s t-test, P < 0.05; Fig. 2C). oxidative stress and glutathione metabolism (sod1).
An adsorption kinetics study showed that the PCP concentra-
3.8. Oxidative damage tions in the exposure media were stable in water, while there was a
marked decrease in the PCP concentration in the exposure media in
We observed oxidative damage effects including ROS content, the presence of n-TiO2 ; this result suggests that n-TiO2 can adsorb
MDA content, 8-OHdG level, SOD activity, and GSH content (Fig. 3). PCP. Previous studies have reported that n-TiO2 can adsorb heavy
Exposure of n-TiO2 alone to zebrafish larvae did not induce genera- metals [4,34,44] or organic toxicants, including tributyltin, PBDEs,
tion of ROS, and alter MDA, 8-OHdG, SOD, and GSH compared to the DDT and tetrachlorodibenzo-p-dioxin [TCDD] [12,13,45,46], while
solvent control. ROS was significantly increased in all PCP exposure study showed the aggregation and subsequent sedimentation of n-
groups relative to the solvent control (one-way ANOVA followed by TiO2 with tributyltin [12]. At the same time, we did not measure
902 Q. Fang et al. / Journal of Hazardous Materials 283 (2014) 897–904

(A)
9000
PCP

Fluorescence Intensity
PCP+ n-TiO2
# #

(DCF/µg prot)
#
** **
4500 ** ** **
*

0
0 3 10 30
PCP conce ntration (µg/L)

(B)
30
PCP
MDA (nmol/mg prot)

PCP+n-TiO2 #

# **
15 **
*

0
0 3 10 30
PCP conce ntration (µg/L)

Fig. 3. (A) ROS generation; (B) MDA content; (C) 8-OHdG level; (D) SOD activity; and (E) GSH content in larvae after exposure to PCP alone (0, 3, 10, and 30 ␮g/L) and in
the presence of n-TiO2 (0.1 mg/L) for 6 days. Significance differences (P < 0.05) are indicated by * vs. DMSO control (one-way ANOVA followed by Dunnett’s test); # vs. the
corresponding PCP groups without n-TiO2 ; P < 0.01 are indicated by ** vs. DMSO control (two-way ANOVA followed by Tukey’s test).

any metabolites of PCP in the exposure media. However, the pho- photocatalytic degradation of PCP observed in the exposure media
tocatalytic degradation of PCP could occur in the presence of high did not occur in our experiment because of negligible UV illu-
n-TiO2 concentrations (200 mg/L) under UV illumination condition mination. Hence, the decreased PCP in water was caused by
(450 W), via generation of ROS (e.g., hydroxyl radical) to attack para sedimentation and not by degradation.
position of the PCP ring to form a semiquinone radical, which in In our study, the presence of n-TiO2 reduced PCP content
turn disproportionates to yield detected metabolic products (e.g., in larvae. In contrast, several previous studies have reported
p-chloranil and tetrachlorohydroquinone) [37]. Likewise, there is enhanced bioavailability of organic toxicants to exposure organ-
ample evidence to indicate that photocatalytic degrade organic isms (e.g., tributyltin, BDE-209, DDT, TCDD) in the presence of
compounds are most likely driven by a UV-induced (especially at n-TiO2 [12,13,45,46], as n-TiO2 acted as a carrier that enables com-
300–388 nm) formation of ROS by n-TiO2 [14,47,48]. For toxicity pounds to adsorb on the organism surface. At the same time,
assay using aquatic organisms, typical laboratory lighting cycle is adsorption and sedimentation also normally occurred in organic
usually established by employing fluorescent lamps, which mainly toxicants with n-TiO2 in exposure media [12,46,51]. Thus, sedi-
emit visible light. The light does not provide wavelengths of UV mentation does not appear to be the major reason that affects the
radiation (below 400 nm) that trigger photoreactivity in most forms bioavailability of organic toxicants by organisms. To further inves-
of TiO2 [16], and no detectable UV light was measured from the fluo- tigate the potential mechanism to account for the reduced PCP
rescent lamps installed in the ceiling [49]. In our study, the intensity content in co-exposed zebrafish, we measured the metabolite in
of UV light is negligible from the fluorescent lamps (15 W/m2 : the larvae. We hypothesized that the observed PCP content in lar-
UVA, 6.5 × 10−3 W/m2 ; UVB, 1.1 × 10−3 W/m2 ) [50]. Thus, the vae could be caused by the increased metabolism of PCP to other
Q. Fang et al. / Journal of Hazardous Materials 283 (2014) 897–904 903

generation, increase lipid peroxidation production (MDA), and


decrease GSH [55]. A further significant change of these biomark-
ers as compared to PCP treatment alone could be accounted for
by enhanced ROS generation, and thus oxidative stress induced by
co-exposure with n-TiO2 . In our study, we observed increased mal-
formation rates in the PCP and co-exposure groups, as compared
to the respective control, which can be attribute to the increased
ROS generation and thus induced oxidative stress. ROS-induced
oxidative stress is thought to contribute to abnormal development
during embryogenesis [56].
The gene nrf2 is presumably one of the most important path-
ways for cells to deal with oxidative stress generated from exposure
to exogenous and endogenous chemicals [57,58]. The transcription
factor nrf2, which regulates the induction of antioxidant enzymes
with an antioxidant response element, played a crucial role in medi-
ating the oxidative damage induced by PCP [59]. In our study,
we also observed an upregulation of nrf2 transcription, and this
alteration should be considered as a regulatory response to the
enhanced ROS-induced oxidative stress. In addition, nrf2 could
induce the transcription of sod gene transcription [57]. The sod gene
transcription was upregulated, which would likely encode more
SOD protein, to protect cells from ROS-induced oxidative damage.
In conclusion, this study demonstrated an increased metabolism
of PCP in zebrafish larvae in the presence of n-TiO2 , which induced
enhanced oxidative damage in early developing zebrafish larvae.
However, the interaction between n-TiO2 and PCP, e.g., the poten-
tial influences of n-TiO2 on uptake and bioavailability, are not well
known. However, this study emphasizes the fact that n-TiO2 could
modify the bioavailability and metabolism of PCP, thus suggest-
ing the consideration of other widespread engineered NPs in the
environment and interaction between other toxicants during envi-
ronmental risks assessment.
Fig. 4. Gene transcription profiles in larvae after exposure to PCP (0, 3, 10, and
30 ␮g/L) alone or in the presence of 0.1 mg/L TiO2 for 6 days: (A) nrf2, (B) sod. The val-
ues are the means of three determinations on each of the three replicate exposures; Acknowledgements
they are presented as the mean ± SEM of three replicates. Significance differences
(P < 0.05) are indicated by * vs. DMSO control (one-way ANOVA followed by Dun-
nett’s test); # vs. the corresponding PCP groups without n-TiO2 (two-way ANOVA
This work was supported by grants from the National Nat-
followed by Tukey’s test); P < 0.01 are indicated by: ** vs. DMSO control; P < 0.001 ural Science Foundation of China (No. 21237005), the Strategic
are indicated by *** vs. DMSO control. Priority Research Program of the Chinese Academy of Sciences
(No. XDB14040103), the National Environmental Protection Public
Welfare Science and Technology Research Program of China (No.
products. TCHQ is a major metabolite of PCP in the mammalian sys- 201309047) and the State Key Laboratory of Freshwater Ecology
tem and also in fish liver cells [42,52]. In our study, TCHQ/PCP in and Biotechnology (No. 2011FBZ13).
larvae was significantly increased in the co-exposure group com-
pared with the PCP-treated group, supporting the evidence that Appendix A. Supplementary data
more PCP is transformed to TCHQ in the co-exposure group; how-
ever, the mechanism for the enhanced metabolism of PCP in the Supplementary data associated with this article can be found,
presence of n-TiO2 is not known. As a consequence, the increased in the online version, at http://dx.doi.org/10.1016/j.jhazmat.
production of TCHQ would generate a more potent source of ROS. 2014.10.039.
Previous studies have shown that TCHQ seems to be a more toxic
form of an organic toxicant, as it can autoxidize to its semiquinone
References
radicals under physiological conditions, which is the manner in
which PCP could generate a potent ROS source [32,33,53] and cause [1] S.J. Klaine, P.J. Alvarez, G.E. Batley, T.F. Fernandes, R.D. Handy, D.Y. Lyon, S.
oxidative damages to cells or tissues. Mahendra, M.J. McLaughlin, J.R. Lead, Nanomaterials in the environment:
behavior, fate, bioavailability, and effects, Environ. Toxicol. Chem. 27 (2008)
In our study, we observed a significant increase in ROS genera-
1825–1851.
tion in the PCP treatment relative to the solvent control; in addition, [2] R.D. Handy, R. Owen, E. Valsami-Jones, The ecotoxicology of nanoparticles and
a significantly increased generation of ROS was measured in the co- nanomaterials: current status, knowledge gaps, challenges, and future needs,
exposure groups, suggesting that an increased ROS amount could Ecotoxicology 17 (2008) 315–325.
[3] A. Baun, S.N. Sorensen, R.F. Rasmussen, N.B. Hartmann, C.B. Koch, Toxicity and
be due to the presence of n-TiO2 . The generation of ROS would affect bioaccumulation of xenobiotic organic compounds in the presence of aqueous
the antioxidant system (e.g., by decreasing SOD and GSH content), suspensions of aggregates of nano-C60, Aquat. Toxicol. 86 (2008) 379–387.
and if the oxidative stress overwhelmed the defending system, [4] W.W. Yang, A.J. Miao, L.Y. Yang, Cd2+ toxicity to green alga Chlamydomonas
reinhardtii as influenced by its adsorption on TiO2 engineered nanoparticles,
oxidative damage would occur, which could cause lipid peroxida- PLoS One 7 (2012) 1–8.
tion (measured as MDA) and genotoxicity (DNA damage, measured [5] X. Chen, S.S. Mao, Titanium dioxide nanomaterials: synthesis, properties,
as 8-OHdG). In agreement with this notion, we observed a decrease modifications, and applications, Chem. Rev. 107 (2007) 2891–2959.
[6] R. Kaegi, A. Ulrich, B. Sinnet, R. Vonbank, A. Wichser, S. Zuleeg, H. Simmler, S.
in GSH content, reduced SOD activity, and increased MDA and 8- Brunner, H. Vonmont, M. Burkhardt, M. Boller, Synthetic TiO2 nanoparticle
OHdG, a biomarker for DNA damage [54], in the PCP-treated groups. emission from exterior facades into the aquatic environment, Environ. Pollut.
PCP exposed to Carassius carassius hepatocytes could induce ROS 156 (2008) 233–239.
904 Q. Fang et al. / Journal of Hazardous Materials 283 (2014) 897–904

[7] G. Federici, B.J. Shaw, R.D. Handy, Toxicity of titanium dioxide nanoparticles tetrachlorohydroquinone, a toxic metabolite of pentachlorophenol, Chem.
to rainbow trout (Oncorhynchus mykiss): gill injury, oxidative stress, and other Biol. Interact. 105 (1997) 1–16.
physiological effects, Aquat. Toxicol. 84 (2007) 415–430. [34] C. Tan, W.H. Fan, W.X. Wang, Role of titanium dioxide nanoparticles in the
[8] X.S. Zhu, L. Zhu, Z.H. Duan, R. Qi, Y. Li, Y. Lang, Comparative toxicity of several elevated uptake and retention of cadmium and zinc in Daphnia magna,
metal oxide nanoparticle aqueous suspensions to zebrafish (Danio rerio) early Environ. Sci. Technol. 46 (2012) 469–476.
developmental stage, J. Environ. Sci. Health A 43 (2008) 278–284. [35] M. Westerfield, The Zebrafish Book, a Guide for the Laboratory Use of
[9] H. Ma, A. Brennan, S.A. Diamond, Phototoxicity of TiO2 nanoparticles under Zebrafish (Danio rerio), third ed., University of Oregon Press, Eugene, OR, USA,
solar radiation to two aquatic species: Daphnia magna and Japanese medaka, 1995.
Environ. Toxicol. Chem. 31 (2012) 1621–1629. [36] C.O. Robichaud, A.E. Uyar, M.R. Darby, L.G. Zucker, M.R. Wiesner, Estimates of
[10] V.K. Sharma, Aggregation and toxicity of titanium dioxide nanoparticles in upper bounds and trends in nano-TiO2 production as a basis for exposure
aquatic environment—a review, J. Environ. Sci. Health A 44 (2009) 1485–1495. assessment, Environ. Sci. Technol. 43 (2009) 4227–4233.
[11] C. Jacobasch, C. Völker, S. Giebner, J. Völker, H. Alsenz, T. Potouridis, H. [37] G. Mills, M.R. Hoffmann, Photocatalytic degradation of pentachlorophenol on
Heidenreich, G. Kayser, J. Oehlmann, M. Oetken, Long-term effects of TiO2 particles: identification of intermediates and mechanism of reaction,
nanoscaled titanium dioxide on the cladoceran Daphnia magna over six Environ. Sci. Technol. 27 (1993) 1681–1689.
generations, Environ. Pollut. 186 (2014) 180–186. [38] O. Jáuregui, E. Moyano, M.T. Galceran, Liquid chromatography-atmospheric
[12] X.S. Zhu, J. Zhou, Z.H. Cai, TiO2 nanoparticles in the marine environment: pressure chemical ionization mass spectrometry for chlorinated phenolic
impact on the toxicity of tributyltin to abalone (Haliotis diversicolor compounds: application to the analysis of polluted soils, J. Chromatogr. A 823
supertexta) embryos, Environ. Sci. Technol. 45 (2011) 3753–3758. (1998) 241–248.
[13] Q. Chen Q. Wang, W. P. Zhou, J. Wang Li, X. Wang C. Huang, B. Zhou K. Lin, [39] SN/T 2445-2010. Determination of Pentachlorophenol residues in food stuffs
Bioconcentration and metabolism of BDE-209 in the presence of titanium of animal origin for import and export-LC–MS/MS method.
dioxide nanoparticles and impact on the thyroid endocrine system and [40] O. Bar-Ilan, K.M. Louis, S.P. Yang, J.A. Pedersen, R.J. Hamers, R.E. Peterson, W.
neuronal development in zebrafish larvae, Nanotoxicology 8S1 (2014) Heideman, Titanium dioxide nanoparticles produce phototoxicity in the
196–207. developing zebrafish, Nanotoxicology 6 (2012) 670–679.
[14] U.I. Gaya, A.H. Abdullah, Heterogeneous photocatalytic degradation of organic [41] S. Li, X. Pan, L.K. Wallis, Z. Fan, Z. Chen, S.A. Diamond, Comparison of TiO2
contaminants over titanium dioxide: a review of fundamentals, progress and nanoparticle and graphene–TiO2 nanoparticle composite phototoxicity to
problems, J. Photochem. Photobiol. C 9 (2008) 1–12. Daphnia magna and Oryzias latipes, Chemosphere 112 (2014) 62–69.
[15] K. Nakata, T. Ochiai, T. Murakami, A. Fujishima, Photoenergy conversion with [42] G.R. Bruno, F.R. Jean, N.T. Thomas, Simultaneous assay of pentachlorophenol
TiO2 photocatalysis: new materials and recent applications, Electrochim. Acta and its metabolite tetrachlorohydroquinone, by gas chromatography without
84 (2012) 103–111. derivatization, J. Chromatogr. B 533 (1990) 111–124.
[16] H. Ma, A. Brennan, S.A. Diamond, Photocatalytic reactive oxygen species [43] J. Deng, L.Q. Yu, C.S. Liu, K. Yu, X.J. Shi, B.S. Zhou,
production and phototoxicity of titanium dioxide nanoparticles are Hexabromocyclododecane-induced developmental toxicity and apoptosis in
dependent on the solar ultraviolet radiation spectrum, Environ. Toxicol. zebrafish embryos, Aquat. Toxicol. 93 (2009) 29–36.
Chem. 31 (2012) 2099–2107. [44] X. Zhang, H. Sun, Z. Zhang, Q. Niu, Y. Chen, J.C. Crittenden, Enhanced
[17] D. Yin, Y. Gu, Y. Li, X. Wang, Q. Zhao, Pentachlorophenol treatment in vivo bioaccumulation of cadmium in carp in the presence of titanium dioxide
elevates point mutation rate in zebrafish p53 gene, Mutat. Res. 609 (2006) nanoparticles, Chemosphere 67 (2007) 160–166.
92–101. [45] Y. Shi, J.H. Zhang, M. Jiang, L.H. Zhu, H.Q. Tan, B. Lu, Synergistic genotoxic1ity
[18] K. Farhadi, M.A. Farajzadeh, A.A. Matin, P. Hashemi, Dispersive liquid–liquid caused by low concentration of titanium dioxide nanoparticles and p,p -DDT
microextraction and liquid chromatographic determination of in human hepatocytes, Environ. Mol. Mutagen. 51 (2010) 192–204.
pentachlorophenol in water, Cent. Eur. J. Chem. 7 (2009) 369–374. [46] C.L. Laura, G. Frenzilli, T. Balbi, M. Bernadeschi, C. Ciacci, S. Corsolini, C.D.
[19] W. Zheng, X. Wang, H. Yu, X. Tao, Y. Zhou, W. Qu, Global trends and diversity Torre, R. Fabbri, C. Faleri, S. Focardi, Interactive effects of n-TiO2 and
in pentachlorophenol levels in the environment and in humans: a 2,3,7,8-TCDD on the marine bivalve Mytilus galloprovincialis, Aquat. Toxicol.
meta-analysis, Environ. Sci. Technol. 45 (2011) 4668–4675. 153 (2014) 53–65, http://dx.doi.org/10.1016/j.aquatox.2013.11.002.
[20] W. Zheng, H. Yu, X. Wang, W. Qu, Systematic review of pentachlorophenol [47] D.S. Bhatkhande, V.G. Pangarkar, A. Beenackers, Photocatalytic degradation
occurrence in the environment and in humans in China: not a negligible for environmental applications—a review, J. Chem. Technol. Biotechnol. 77
health risk due to the reemergence of schistosomiasis, Environ. Int. 42 (2012) (2002) 102–116.
105–116. [48] F. Seitz, M. Bundschuh, A. Dabrunz, N. Bandow, G.E. Schaumann, R. Schulz,
[21] J. Muir, G. Eduljee, PCP in the freshwater and marine environment of the Titanium dioxide nanoparticles detoxify pirimicarb under UV irradiation at
European Union, Sci. Total Environ. 236 (1999) 41–56. ambient intensities, Environ. Toxicol. Chem. 31 (2012) 518–523.
[22] R.J. Letcher, W.A. Gebbink, C. Sonne, E.W. Born, M.A. Mckinney, R. Dietz, [49] Z. Clemente, V.L.S.S. Castro, M.A.M. Moura, C.M. Jonsson, L.F. Fraceto, Toxicity
Bioaccumulation and biotransformation of brominated and chlorinated assessment of TiO2 nanoparticles in zebrafish embryos under different
contaminants and their metabolites in ringed seals (Pusa hispida) and polar exposure conditions, Aquat. Toxicol. 147 (2014) 129–139.
bears (Ursus maritimus) from East Greenland, Environ. Int. 35 (2009) [50] Y. Tian, C.H. Dai, Y.X. Hao, C.P. Yao, J.L. Yu, H. Lu, W. Liu, Measurements of the
1118–1124. ultraviolet irradiation of artificial light sources in daily life, J. Clin. Dermatol.
[23] M.H. Zheng, B. Zhang, Z.C. Bao, Analysis of pentachlorophenol from water 35 (2006) 209–210 (in Chinese).
sediments, and fish bile of Dongting lake in China, Bull. Environ. Contam. [51] L. Wang, Y. Huang, A.T. Kan, M.B. Tomson, W. Chen, Enhanced transport of
Toxicol. 64 (2000) 16–19. 2,2 ,5,5 -polychlorinated biphenyl by natural organic matter (NOM) and
[24] D. Tan, J.B. Zhang, Estimates of PCP-Na consumption in districts and provinces surfactant-modified fullerene nanoparticles (nC60), Environ. Sci. Technol. 46
in China by the top-down calculation method, Environ. Pollut. Control 30 (2012) 5422–5429.
(2008) 17–20. [52] C. Pietsch, J. Hollender, F. Dorusch, P. Burkhardt-Holm, Cytotoxic effects of
[25] J.J. Gao, L.H. Liu, X.R. Liu, H.D. Zhou, S.B. Huang, Z.J. Wang, Levels and spatial pentachlorophenol (PCP) and its metabolite tetrachlorohydroquinone (TCHQ)
distribution of chlorophenols 2,4-dichlorophenol, 2,4,6-trichlorophenol, and on liver cells are modulated by antioxidants, Cell Biol. Toxicol. 30 (2014)
pentachlorophenol in surface water of China, Chemosphere 71 (2008) 233–252.
1181–1187. [53] S. Naito, Y. Ono, I. Somiya, S. Inoue, K. Ito, K. Yamamaoto, S. Kawanishi, Role of
[26] M. Pavlica, G. Klobucar, N. Vetma, R. Erben, D. Papes, Detection of micronuclei active oxygen species in DNA-damage by pentachlorophenol metabolites,
in haemocytes of zebra mussel and great ramshorn snail exposed to Mutat. Res. Fundam. Mol. Mech. 310 (1994) 79–88.
pentachlorophenol, Mutat. Res. Fundam. Mol. Mech. 465 (2000) 145–150. [54] A. Valavanidis, T. Vlahogianni, M. Dassenakis, M. Scoullos, Molecular
[27] Y. Guo, B. Zhou, Thyroid endocrine system disruption by pentachlorophenol: biomarkers of oxidative stress in aquatic organisms in relation to toxic
an in vitro and in vivo assay, Aquat. Toxicol. 142-143 (2013) 138–145. environmental pollutants, Ecotoxicol. Environ. Saf. 64 (2006) 178–189.
[28] T. Xu, J. Zhao, P. Hu, Z.J. Dong, J.Y. Li, H.C. Zhang, D.Q. Yin, Q.S. Zhao, [55] Y.L. Dong, P.J. Zhou, S.Y. Jiang, X.W. Pan, X.H. Zhao, Induction of oxidative
Pentachlorophenol exposure causes Warburg-like effects in zebrafish stress and apoptosis by pentachlorophenol in primary cultures, Comp.
embryos at gastrulation stage, Toxicol. Appl. Pharmacol. 277 (2014) 183–191. Biochem. Phys. C 150 (2009) 179–185.
[29] A.T. Proudfoot, Pentachlorophenol poisoning, Toxicol. Rev. 22 (2003) 3–11. [56] M. Yamashita, Apoptosis in zebrafish development, Comp. Biochem. Phys. B
[30] Z. Mehmood, M.P. Williamson, D.E. Kelly, S.L. Kelly, Metabolism of 136 (2003) 731–742.
organochlorine pesticides: the role of human cytochrome P4503A4, [57] X.J. Shi, B.S. Zhou, The role of nrf2 and mapk pathways in pfos-induced
Chemosphere 33 (1996) 759–769. oxidative stress in zebrafish embryos, Toxicol. Sci. 115 (2010) 391–400.
[31] Y.J. Wang, Y.S. Ho, J.H. Jeng, H.J. Su, C.C. Lee, Different cell death mechanisms [58] T.W. Kensler, N. Wakabayashi, S. Biswal, Cell survival responses to
and gene expression in human cells induced by pentachlorophenol and its environmental stresses via the Keap1-Nrf2-ARE pathway, Annu. Rev.
major metabolite, tetrachlorohydroquinone, Chem. Biol. Interact. 128 (2000) Pharmacol. 47 (2007) 89–116.
173–188. [59] T. Umemura, Y. Kuroiwa, Y. Kitamura, Y. Ishii, K. Kanki, Y. Kodama, K. Itoh, M.
[32] Y.J. Wang, J.K. Lin, Estimation of selected phenols in drinking-water with Yamamoto, A. Nishikawa, M. Hirose, A crucial role of nrf2 in vivo defense
in-situ acetylation and study on the DNA-damaging properties of against oxidative damage by an environmental pollutant, pentachlorophenol,
polychlorinated phenols, Arch. Environ. Contam. Toxicol. 28 (1995) 537–542. Toxicol. Sci. 90 (2006) 111–119.
[33] Y.J. Wang, Y.S. Ho, S.W. Chu, H.J. Lien, T.H. Liu, J.K. Lin, Induction of glutathione
depletion p53 protein accumulation and cellular transformation by

You might also like