Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Environmental Pollution 241 (2018) 674e683

Contents lists available at ScienceDirect

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

New insights into the mechanism of phthalate-induced


developmental effects*
Xiyan Mu a, *, Ying Huang a, Jia Li a, Ke Yang a, Wenbo Yang a, Gongming Shen a,
Xuxing Li a, Yunlei Lei a, Sen Pang b, Chengju Wang b, Xuefeng Li b, Yingren Li a, **
a
Fishery Resource and Environment Research Center, Chinese Academy of Fishery Sciences, Beijing, PR China
b
College of Sciences, China Agricultural University, Beijing 100193, PR China

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

Article history: To investigate the biological pathways involved in phthalate-induced developmental effects, zebrafish
Received 6 February 2018 embryos were exposed to different concentrations of di-(2-ethylhexyl) (DEHP) and di-butyl phthalate
Received in revised form (DBP) for 96 h. Embryonic exposure to DEHP and DBP induced body length decrease, yolk sac
5 May 2018
abnormities, and immune responses (up-regulation of immune proteins and genes). The lipidomic re-
Accepted 31 May 2018
sults showed that at a concentration of 50 mg/L, DEHP and DBP significantly reduced the levels of fatty
acids, triglycerides, diacylglycerol, and cholesterol. These effects are partly explained by biological
pathway enrichment based on data from the transcriptional and proteomic profiles. Co-exposure to DBP
Keywords:
Di-(2-ethylhexyl) phthalate
and ER antagonist did not significantly relieve the toxic symptoms compared with exposure to DBP
Di-butyl phthalate alone. This indicates that phthalate-induced developmental abnormities in zebrafish might not be
Developmental effects mediated by the ER pathway. In conclusion, we identified the possible biological pathways that mediate
Immune response phthalate-induced developmental effects and found that these effects may not be driven by estrogenic
Lipid homeostasis activation.
Mechanism © 2018 Elsevier Ltd. All rights reserved.

1. Introduction contamination occur (more than 100 mg/L) in heavily industrialised


areas (Fromme et al., 2002; Yuwatini et al., 2006; Khan and Jung,
Phthalates are commonly used in a variety of industrial and 2008). As a result of widespread contamination, these chemicals
consumer products, including food packaging, children's toys, and can enter the human body through daily ingestion and inhalation.
building materials. Di-(2-ethylhexyl) phthalate (DEHP) is the most Phthalates are ubiquitously detected in urine samples of all age
commonly used phthalate plasticizer in polyvinyl chloride (PVC) groups from all around the world (Guo et al., 2011; Gao et al., 2017;
formulations; DEHP accounts for 80% of phthalate production in Merida-Ortega et al., 2016; Lee et al., 2017).
China and approximately 18% of all plasticizers used in Western Given the wide distribution of phthalates, their negative effects
Europe (Zolfaghari et al., 2014; European Union, 2008). Di-butyl on humans and animals have drawn considerable attention. Most
phthalate (DBP) is typically used in personal care products (Lien research efforts have focused on endocrine-disrupting effects, pri-
et al., 2015). Because phthalates are not chemically bound to the marily the alteration of hypothalamus-pituitary-thyroid axis and
plastics, they can leach into food, medicine, water, indoor dust, and hypothalamus-pituitary-gonad axis. Previous studies found that
air, resulting in human and animal exposure (Zota et al., 2016; Jia the urinary phthalate concentrations in humans are negatively
et al., 2017; Net et al., 2015; Kang et al., 2012; Gaspar et al., 2014). associated with the levels of urinary thyroid hormones in children
The detected concentration of DEHP and DBP in drinking water and and serum T4 in adults (Boas et al., 2010; Huang et al., 2017). In
surface water is generally below 1 and 10 mg/L respectively (Liu addition, a variety of studies have shown that phthalates cause
et al., 2014; Zeng et al., 2008). However, hotspots of reproductive toxicity in both male and female individuals. Serum
phthalates are negatively associated with serum testosterone, sex
hormone-binding globulin, semen volume, semen quality, and total
*
This paper has been recommended for acceptance by Dr. Chen Da. sperm count, whereas they are positively associated with DNA
* Corresponding author. damage in the sperm of adult men (Joensen et al., 2012; Pant et al.,
** Corresponding author.
2008; Specht et al., 2014). In male fish, decreased plasma
E-mail addresses: muxiyan@cafs.ac.cn (X. Mu), liyr@cafs.ac.cn (Y. Li).

https://doi.org/10.1016/j.envpol.2018.05.095
0269-7491/© 2018 Elsevier Ltd. All rights reserved.
X. Mu et al. / Environmental Pollution 241 (2018) 674e683 675

testosterone (T) concentrations, reduced ability to fertilise oocytes 2.2. Chemicals and reagents
spawned by untreated females, decreased sperm motility and ve-
locity, and altered spermatogenesis were observed after DEHP Standard water was prepared in the lab according to the formula
exposure (Uren-Webster et al., 2010; Crago and Klaper, 2012; of iso-7346-3: 2 mmol L1 Ca2þ, 0.5 mmol L1 Mg2þ, 0.77 mmol L1
Golshan et al., 2015). In females, urinary concentrations of DEHP Naþ, and 0.08 mmol L1 Kþ (ISO, 1996). Di-(2-ethylhexyl phthalate
metabolites are negatively associated with oocyte yield, clinical (DEHP, 99.5%, CAS: 117-81-7) and dibutyl phthalate (DBP, 99%, CAS:
pregnancy, and live birth (Hauser et al., 2016). Ye et al. (2014) re- 84-74-2) were purchased from Sigma-Aldrich (Darmstadt, Ger-
ported that female marine medaka (Oryzias melastigma) showed many). Stock solutions of DEHP and DBP for drug-exposure ex-
decreased egg production, increased concentrations of T and E2 and periments were prepared using acetone AR. ICI-182,780 (98%, CAS
up-regulation of ldlr. 129453-61-8) was purchased from Sigma-Aldrich (Darmstadt,
In addition to the effects on reproduction and hormone levels, Germany).
the developmental effects of phthalates have been increasingly
studied in recent years. Tsai et al. (2016) demonstrated that daily 2.3. Exposure and sample collection
DEHP intake is negatively associated with height percentile,
weight percentile, bone age/chronological age, and insulin-like Experiments were performed in accordance with current Chi-
growth factor 1 (IGF-1) levels in children. The levels of urinary nese legislation and were approved by the independent animal
phthalate metabolites are associated with delayed pubic hair ethics committee at Chinese Academy of Fishery Sciences.
development in boys and earlier breast development in girls
(Zhang et al., 2015). Yamasaki et al. (2009) reported that maternal
2.3.1. Exposure for morphological endpoints
exposure to dicyclohexyl phthalate can induce a series of devel-
Test solutions of DEHP and DBP with concentrations of 0 (con-
opmental effects in male offspring, including prolonged preputial
trol), 50, and 250 mg/L (based on pre-experiment data) were made
separation, reduced ano-genital distance, increased areola/nipple
using standard water. Embryos at approximately 2 h post-
retention, and decreased ventral prostate and levator ani/bulbo-
fertilization (hpf) were randomly transferred into the test solu-
cavernosus muscle weights. In addition, neurodevelopment has
tions in 24-well plates. Exposure solutions for solvent control and
been identified as another target of phthalates. Research on
all treatment groups contained the same concentration of acetone
humans and other mammals has shown that phthalate exposure
(0.05 mL/L). Each treatment was replicated three times. The num-
can result in attention problems along with aggressive or rule-
ber of dead individuals was determined, and the dead embryos
breaking behavior in children and induce cell apoptosis in hip-
were removed daily. Embryo death was judged using the lethal
pocampal neurons as well as impair spatial learning and memory
toxicological endpoints proposed by Nagel (2002). The hatching
in rat pups (Kobrosly et al., 2014; Engel et al., 2010; Li et al., 2013;
and development status of embryos were checked daily. Terato-
Li et al., 2009). However, the mechanism of action and pathways
genic effects were identified and recorded using a ZEISS Vert.A1
involved in phthalate-induced developmental effects remain
microscope (Jena, Germany). Body length along with yolk sac width
uncharacterized, although these symptoms have been considered
and height were measured linearly using the microscope software.
as the downstream health outcomes that resulted from the dis-
See the Supplemental Information for more details.
order of hormone, for instance lower production of estradiol or
progesterone (Kay et al., 2013).
2.3.2. Exposure for transcriptomic and proteomic analysis
Currently, zebrafish (Danio rerio) embryos are an important
Embryos were randomly transferred into test solutions (50 mg/L)
model for toxicological investigations of hazardous chemicals
in 1-L beakers at 2 hpf. Each beaker contained 500 mL of exposure
(Zhang et al., 2017; Brox et al., 2016; Huang et al., 2016). Zebrafish
solution and approximately 100 embryos, and there were three
embryos possess many advantages, including in vitro fertilization,
beakers in each treatment group. At 96 hpf, 75 hatched (or decor-
high fecundity, rapid embryonic development, and optical trans-
ticated) larvae from each replicate (beaker) were collected and
parency, which make it easy to detect morphological endpoints or
washed twice with standard water (25 for RNA extraction and 50
observe the development process in early life stages (Mu et al.,
for protein extraction). The embryo samples were stored at 80  C
2016). Previous studies have shown that phthalates have several
until analysis.
negative effects on zebrafish, including steroidogenic alteration,
cardiac defects, reproductive toxicity and oxidative stress (Sohn
et al., 2016; Sun and Liu, 2017; Zhang et al., 2014; Corradetti 2.3.3. Exposure for lipidomic analysis
et al., 2013). In this study, we applied several biological ap- Embryos were randomly transferred into test solutions (50 mg/L)
proaches with an experimental morphological investigation using in 200-mL beakers at 2 hpf. Each beaker contained 100 mL of
zebrafish embryo as a model to identify the biological pathways exposure solution and 40 embryos, and there were eight beakers in
that mediate phthalate-induced developmental effects. each treatment group. At 96 hpf, 20 embryos were collected from
each beaker and washed with standard water. The embryo samples
were stored at 80  C until analysis.

2. Materials and methods 2.3.4. Exposure for immune response and estrogenic activity test
Embryos were randomly transferred into test solutions (50 and
2.1. Zebrafish maintenance and embryo collection 250 mg/L) in 1-L beakers at 2 hpf. 50 and 250 mg/L 17b-estradiol was
used as positive control in the estrogenic activity test. Each beaker
Wild-type zebrafish (AB strain) were purchased from a local contained 500 mL of exposure solution and approximately 200
provider. All adult zebrafish were maintained in flow-through embryos, and there were three beakers in each treatment group. At
feeding equipment (made by Esen Corp.) at 26  C with a photope- 96 hpf, 120 hatched (or decorticated) larvae from each replicate
riod of 14/10 (light/dark). The zebrafish were fed daily with live were collected and washed twice with standard water (30 for
brine shrimp (Artemia salina). The preparation and collection of mRNA extraction and 90 for the ELISA assay of hormones, ERa and
zebrafish embryos followed the procedure described in our previ- immune protein levels). The embryo samples were stored at 80  C
ous work (Mu et al., 2013). until analysis.
676 X. Mu et al. / Environmental Pollution 241 (2018) 674e683

2.4. Illumina sequencing and transcriptomic analysis 2.8. Gene expression analysis

Total RNA was extracted from zebrafish embryos using a spin RNA extraction, cDNA synthesis and real-time PCR reactions
column adsorption method following the manufacturer's protocol were performed as previously described (Mu et al., 2018).
(Tiangen Biotech). RNA quality was assessed using an Agilent 2100 Zebrafish-specific primers were designed for the genes of interest
BioAnalyzer (Agilent Technologies, Santa Clara, CA, USA) and a using Primer 5.0 software (Table S6). The housekeeping gene b-
Qubit Fluorometer (Invitrogen). Total RNA samples that met the actin was used as an internal control. Transcript quantification was
following requirements were used in subsequent experiments: performed using the 2DdCt method. Please see the Supplemental
RNA integrity number >7.0 and 28S:18S > 1.8. Libraries were con- Information for more details (Tables S7eS8).
structed from three samples of embryos exposed to the solvent
control and three samples of embryos exposed to 50 mg/L phtha- 2.9. ER inhibition test
lates using a Next Ultra RNA Library Prep Kit for Illumina (NEB).
mRNA molecules with poly(A) tails were enriched from 1 mg total Exposure was conducted in the same way as for morphological
RNA using a Next Poly(A) mRNA Magnetic Isolation Module (NEB) endpoints. Embryos were exposed to 250 mg/L DBP or co-exposed
kit. After RT-qPCR validation, the libraries were subjected to paired- to 250 mg/L DBP and 5 mM (z3.04 mg/L) ICI 182,780 in a 24-well
end sequencing with pair end 125-base pair reading length using plate from 2 to 96 hpf. The body lengths and yolk sac sizes of the
an Illumina HiSeq 2500 sequencer (Illumina). Please see the embryos were assayed.
Supplemental Information for more details on RNA-Seq
(Tables S1eS3 and Figures S5eS6). 2.10. Chemical confirmation of phthalate concentration

2.5. Proteomics The actual concentrations of DEHP and DBP were measured at
0 hpf (at the beginning of exposure), 24 hpf (before the first
For each sample, 50 embryos were ground in liquid nitrogen and replacement) and 96 hpf (at the end of exposure). Based on the
lysed using protein-extraction buffer on ice for 30 min and then monitoring results, the deviation between the nominal and actual
centrifuged at 16,000  g for 15 min at 4  C. The supernatant was phthalate concentrations was generally less than 20%
collected, and the total protein content was determined using a (Tables S9eS11). Therefore, the nominal concentrations of DEHP
bicinchoninic acid protein assay (Pierce, USA). The lysis was stored and DBP could be used to represent the actual concentrations in
at 80  C before further processing. Tandem mass tags (TMT6/10, exposure solution in this work.
Pierce, USA) with different reporter ions (126e131 Da) were
applied as isobaric tags for relative quantification. TMT labeling was 2.11. Statistical analysis
performed according to the manufacturer's instructions. LC-MS/MS
analysis was carried out at Capitbalbio Technology using a Q All statistical analyses were performed using SPSS 16.0 software.
Exactive mass spectrometer (Thermo Scientific, CA). Proteome Differences were determined by one-way ANOVA complemented
Discoverer software (Version 1.4, Thermo Scientific, USA) was used by Dunnett post hoc comparison. P < 0.05 was considered signifi-
to perform database searching against the Oryctolagus cuniculus cant. Semantic similarity scatter plots were constructed using
database (46601 proteins) using the Sequest algorithms. More de- REVIGO online software according to the instructions (Supek et al.,
tails regarding sample digestion, TMT labeling, LC-MS/MS param- 2011).
eters and data analysis are provided in the Supplemental
Information (Tables S4eS5). 3. Results

2.6. Lipidomics 3.1. Solvent effect

Lipid analysis was performed using a Q Exactive Orbitrap mass No significant difference was found between the solvent and
spectrometer (Thermo, CA, USA). Mobile phase A was prepared by blank control for all tested indicators (data not shown). The control
dissolving ammonium acetate (0.77 g) in 400 mL of water followed data shown in all figures and tables are the experimental data for
by adding 600 mL of acetonitrile. Mobile phase B was prepared by the solvent control.
mixing 100 mL of acetonitrile with 900 mL of isopropanol. An
XSelect CSH C18 column was used for lipid analysis at a column 3.2. Developmental effects caused by phthalates
temperature of 45  C. Lipids were identified and quantified using
Lipid Search 4.1.30 (Thermo, CA, USA). Mass tolerances of 5 and Under the tested concentrations, DEHP and DBP exposure had
10 ppm were applied for the precursor and product ions. A reten- no significant effects on the survival and hatching of zebrafish
tion time shift of 0.25 min was performed in “alignment.” The M- embryos (Figures S1b and 1c). However, the body length of the
score and chromatographic areas were used to reduce false posi- embryos in the DBP-250 group was significantly reduced compared
tives. Please see the Supplemental Information for more details. with that of the control (Figure S1a). Furthermore, the yolk sac was
significantly affected by DEHP and DBP, which is reflected in two
2.7. ELISA aspects: (1) the proportion of the fish body made up by the yolk sac
was larger in phthalate-treated embryos than in control embryos
Protein and hormone levels were measured via ELISA. 90 em- (Fig. 1a); and (2) the yolk sac size was larger in phthalate-treated
bryos were homogenized with saline (1:10, w/v) on ice in centri- embryos than in control embryos (Fig. 1b and c).
fuge tube using an electric homogenizer (Tiangen Biotech, Beijing,
China). After centrifugation for 10 min at 3000 rpm and 4  C, the 3.3. Global changes in protein abundance
supernatant was collected for ELISA. ELISA was conducted accord-
ing to the ELISA Kit instructions (Dongge Biotechnology Co., Ltd, Global proteome profiling indicated the significant alteration of
Beijing, China). Please see the Supplemental Information for more the abundances of 257 proteins (217 up-regulated and 40 down-
details. regulated) and 235 proteins (217 up-regulated and 18 down-
X. Mu et al. / Environmental Pollution 241 (2018) 674e683 677

Fig. 1. a Yolk sac area (red dotted line) of embryos in the control and DEHP-250 groups at 48 hpf. The phthalate-treated yolk sac accounted for a higher proportion of the larvae body
than the control yolk sac. b Widths and heights of embryo yolk sacs in the control and phthalate-treated groups at 48 hpf. c Yolk sac size (including width and height) of embryos in
the control, DBP-50 and DBP-250 groups at 48 hpf. (For interpretation of the references to colour in this figure legend, the reader is referred to the Web version of this article.)

regulated) in embryos exposed to 50 mg/L DBP and DEHP, respec- 3.4. Changes in transcriptional profile
tively (Fig. 2a). The enrichment analysis of proteome data revealed
that several processes, including response to xenobiotic stimulus, The transcript profiles also indicated that the biological pro-
lipoprotein metabolism, regulation of lipid catabolism, regulation cesses involved in lipid metabolism, skeletal development, and the
of fatty-acid metabolism, and regulation of triglyceride catabolism, immune system (e.g., steroid binding, lipid binding, fatty-acid
were significantly induced in DBP-treated embryos. In DEHP- metabolism, lipid transport, skeletal muscle tissue development,
exposed embryos, lipase activity regulation, lipid absorption, lipid stress response, and chemokine activity) were significantly altered
catabolism and lipid metabolism were significantly up-regulated by DEHP and DBP (Figure S3 a-c). However, the transcription levels
(Figure S2). of genes involved in the synthesis of sex hormones, including es-
To identify the main pathway involved in phthalate exposure, trogen receptor isomers, androgen receptors, and estradiol syn-
we further conducted a REVIGO similarity analysis. According to the thetase, remained unchanged (Figure S3d).
similarity scatterplot of significantly enriched GO terms, lipid
metabolism, lipid transport, cell locomotion, digestive system, 3.5. Phthalates altered lipid profiles
lipase activity regulation, and cell adhesion are the main molecular
pathways altered by DBP exposure (Fig. 2b). Similar to the DBP Since both the proteomic and transcriptomic results showed
results, the main active pathways affected by DEHP exposure are that the pathways involved in lipid metabolism were significantly
lipid digestion, lipid metabolism, lipid transport and lipase activity altered by DEHP and DBP, we further assessed the influence of
regulation (Fig. 2c). Please see the Supplemental Information for phthalates on the zebrafish lipid profile. After the analysis of pos-
more details on the REVIGO analysis (Table S12 and S13). itive and negative adducts, 207 lipids [4 acyl carnitines (Acca), 26
678 X. Mu et al. / Environmental Pollution 241 (2018) 674e683

Fig. 2. a Hierarchical clustering of differentially expressed proteins (fold change compared to control > 1.5) in zebrafish embryos exposed to 50 mg/L DEHP and DBP. b-c REViGO
semantic similarity scatter plot of significantly enriched (p < 0.01) biological processes. Ontology terms after DBP (b) and DEHP (c) exposure.

ceramides (Cer), 11 neutral glycosphingolipids (Cerg1 and Cerg2), 3.6. Phthalates induced immune responses
two cholesterol esters (Che), 21 diglycerides (DG), 98 triglycerides
(TG), six lysophosphatidylcholines (LPC), four lysophosphatidyle- To further investigate the immune stress induced by DEHP and
thanolamines (LPE), 34 sphingomyelins (SM), and one sphingosine DBP, the embryonic levels of immune proteins and genes were
(So)] and 254 lipids [21 cardiolipins (CL), five ceramides (Cer), 30 assayed. Both the protein and transcription levels of TNFa, il-1b,
fatty acids (FA), 17 phosphatidylserines (PS), 13 phosphatidylino- and il-8 apparently increased after DEHP and DBP exposure (Fig. 3a,
sitols (PI), two phosphatidylglycerols (PG), 70 phosphatidyletha- b, d, and e). Phthalate concentrations of 50 and 250 mg/L signifi-
nolamines (PE), 85 phosphatidylcholines (PC), two LPEs, and two cantly induced the level of protein tp63 (Fig. 3c), while 250 mg/L
LPCs], respectively, were identified. Most of the identified lipid DEHP and DBP inhibited the transcription level of tp63. In addition,
species remained unchanged after phthalate exposure. Compared the expression level of NF-kb was elevated after phthalate
with the control, exposure to 50 mg/L DBP significantly reduced the exposure.
levels of Cer, Cerg2, Che, PC, DG, TG, and FA, whereas exposure to
50 mg/L DEHP significantly reduced the levels of Cerg1, Che, DG, TG, 3.7. Phthalate-induced endocrine disruption
and FA (Figure S4a). The significantly altered lipids are detailed in
Figures S4b and S4c. Among all the lipids, TG (16:0/16:0/18:1) and We further assessed the estrogenic activation and hormone
TG (16:0/18:1/18:1) were the most significantly affected by disruption in zebrafish embryos exposed to phthalates. ELISA test
phthalate exposure. In addition, CerG2 (d18:1/22:0), ChE (22:6), PC showed that 250 mg/L E2 performed significant activation towards
(17:1/16:0), DG (18:0/18:0), DG (18:0/16:0), and FA (18:1) levels both 17-estradiol and ERa protein level, however, had no obvious
were significantly reduced by DBP exposure; the relative levels impact on transcription level of ESR1 and AR (Fig. 4c, d and 4e).
compared with the control were 0.44, 0.58, 0.56, 0.49, 0.5, and 0.49, DEHP and DBP could significantly induce the level of 17b-estradiol
respectively. DG (16:0/16:1), DG (16:0/16:0), ChE (22:6), and FA and ERa protein, and the induction is lower than that of E2. Further,
(18:1) were significantly reduced by DEHP, with relative levels of DEHP and DBP induced sharp up-regulation of ESR1 expression.
0.59, 0.64, 0.57, and 0.59, respectively. Although exposure to 50 mg/L phthalates had no apparent effect
X. Mu et al. / Environmental Pollution 241 (2018) 674e683 679

Fig. 3. a Relative protein levels of TNFa, il-1b (b), tp63 (c) and il-8 (d). eRelative embryonic transcription levels of TNFa, NF-kB, il-8, il-1b, tp63 in the control and phthalate-treated
groups. Asterisks denote significant differences between treatments and the control (determined by Dunnett post-hoc comparison; p < 0.05*, p < 0.01**).

on thyroid hormone levels, at the concentration of 250 mg/L, the yolk sac stores and provides the nutrients needed for embryonic
phthalates significantly induced triiodothyronine (T3) and tetraio- development (Wiegand, 1996). Thus, the reduction in body length
dothyronine (T4) levels (Fig. 4a and b). observed in this study might be a down-stream symptom of
nutritional deficiencies originating from the yolk sac abnormities.
3.8. ER inhibition test Sant et al. (2016) demonstrated that monoethylhexyl phthalic acid
(MEHP) exposure led to nutritional deficiencies in developing
To further investigate the receptor mechanism of the DBP- mouse embryos, including changes in uptake and availability of
induced developmental effects, embryos were simultaneously nutrients and cofactor uptake; this suggests that embryonic expo-
exposed to DBP and an ER antagonist (ICI-182,780). The morpho- sure to phthalates may reduce essential nutrient supplies, resulting
logical results showed that the yolk sac size at 48 hpf and the body in stunted growth in mammals, consistent with the present study.
length at 96 hpf were significantly affected in both the DBP group Many previous studies have suggested that phthalates can alter
and DBP-ICI co-exposure group compared with the control (Fig. 5a, human development, including changes in body size, body weight
b, and 5c). Co-exposure to ICI did not provide any apparent relief for and brain development (Boas et al., 2010; Engel et al., 2010;
the developmental effects of DBP. Maresca et al., 2016; Cho et al., 2010). The majority of these
symptoms have been recognized as downstream health outcomes
resulting from phthalate-induced endocrine disruption (e.g., es-
4. Discussion trogenic effects or thyroid disruption) (Kay et al., 2013; Meeker and
Ferguson, 2011). However, in the present study, developmental
Phthalates, which are commonly used as plasticizers in flexible indicators seemed to be more sensitive to DEHP and DBP exposure
PVC formulations, are a class of ubiquitous environmental toxi- than endocrine indicators. The concentration of DEHP or DBP
cants. Thus, humans and animals can be exposed to phthalates required to affect body length, yolk sac morphology, and skeletal
through oral, dermal, and inhalation routes. In addition, fetal and development transcripts was 50 mg/L, lower than the concentration
newborn individuals can be maternally exposed to phthalates required to induce symptoms related to endocrine disruption
during early life stages. The present study showed that embryonic (250 mg/L for the alteration of ER protein, ER transcription, estrogen,
exposure of zebrafish to DEHP and DBP during the period from 2 to and thyroxine levels). Therefore, we infer that the developmental
96 hpf induced negative effects, including yolk sac abnormalities abnormities caused by phthalates in this work may not have been
and reductions in body length. The change in the transcript profile driven by estrogen effects. To further confirm whether the devel-
also indicated significant alteration in the expression levels of opmental effects caused by DEHP and DBP arose from the estrogen
genes participating in skeletal development, which might be receptor-mediated pathway, zebrafish embryos were co-exposed to
associated with the reduced body length. In zebrafish embryos, the
680 X. Mu et al. / Environmental Pollution 241 (2018) 674e683

Fig. 4. a Relative T3 level of embryos in control and phthalate-treated groups. b Relative T4 level of embryos in control and phthalate-treated groups. c Relative 17b-estradiol level of
embryos in control, positive control (E2) and phthalate-treated groups. d Relative embryonic transcription level of ESR1 and AR in control, positive control (E2) and phthalate-
treated groups. e Relative ERa protein level of embryos in control, positive control (E2) and phthalate-treated groups. Asterisks denote significant differences between treat-
ments and control (determined by Dunnett post-hoc comparison, p < 0.05*; p < 0.01**).

Fig. 5. a Body lengths of zebrafish embryos in the control, ICI (3.04 mg/L), DBP (250 mg/L) and DBP-ICI (250 mg/Lþ3.04 mg/L) groups at 96 hpf. b Yolk sac heights of zebrafish
embryos in the control, ICI, DBP and DBP-ICI groups at 48 hpf. c Yolk sac widths of zebrafish embryos in the control, ICI, DBP and DBP-ICI groups at 48 hpf. Asterisks denote
significant differences between treatments and the control (determined by Dunnett post-hoc comparison; p < 0.05*, p < 0.01**).

phthalate and an ER antagonist. The results showed that co- have shown that exposure to phthalates can cause abnormal lipid
exposure to the ER antagonist did not relieve the developmental metabolism in humans and other mammals (Lind et al., 2012;
effects induced by DBP. This failure to antagonize DBP effects Schmidt et al., 2012). Schmidt et al. (2012) reported that exposure
indicated that DBP might not act through common nuclear or to environmental concentrations of DEHP enhanced the expression
membrane-bound ERs since the same concentration (5 mM) of ICI levels of genes related to adipogenesis (PPAR isoforms and FABP4)
182,780 was sufficient to antagonize ER activity in zebrafish em- and led to increases in body weight and visceral fat deposits in C3H/
bryos (Figure S7) (Kinch et al., 2015). Considering the above dis- N mice. Chiang et al. (2016) showed that the MEHP-treated adi-
cussion, the phthalate-induced growth defects observed in this pocytes exhibited significant increases in lipolysis and the tran-
work may not be mediated by ER. scription of genes involved in adipogenesis and lipid metabolism.
Lipid metabolism disorder is a global public health problem However, less is known about the phthalate-induced disruption of
attributable to complex interactions between genetic, behavioral, lipid metabolism in aquatic organisms. In this work, proteomic data
and environmental factors. In recent years, a number of studies showed that DEHP and DBP may alter lipid homeostasis in zebrafish
X. Mu et al. / Environmental Pollution 241 (2018) 674e683 681

embryos by inducing the expression of proteins involved in certain level of plasma 17b-estradiol was significantly increased in both
biological processes. The transcription profile also showed that male and female zebrafish (Zhu et al., 2016). In the present study,
genes involved in fatty-acid metabolism, steroid binding, and lipid the trends in hormone levels after phthalate exposure were
transport were up-regulated by DEHP and DBP exposure, which is consistent with the above studies; however, the phthalate con-
consistent with previous studies. However, the lipidomic results centration (250 mg/L) required for hormone alteration was much
showed that most lipid species remained unchanged after phtha- higher than reported in previous studies. This may be attributed to
late exposure, while several species were reduced, including di- the shorter duration of exposure in this study or differences in
glycerides, triglycerides, ceramides, phosphatidylcholines, phthalate sensitivity between different species/different develop-
cholesterol esters, and fatty acids; this contradicts previous results mental stages. In addition, DEHP and DBP treatment resulted in up-
for mammals (Schmidt et al., 2012; Chiang et al., 2016). In fish eggs, regulation of ESR1, which is different with the results of E2 groups
the endogenous lipid reserves, mainly phospholipids and tri- which showed no regulation towards ESR1. This indicated that the
acylglycerols, are in the form of yolk globules, and alterations in action mode of DEHP and DBP in zebrafish embryos is not exactly
lipid metabolism may affect the yolk sac (Wiegand, 1996). Previous the same with E2. Further, T3 and T4 levels were significantly
studies have reported yolk sac abnormities occurred along with elevated after exposure to 250 mg/L phthalates, which is in consis-
disruptions in lipid metabolism in zebrafish embryos after expo- tent with a previous study in which 400 mg/L DEHP enhanced the
sure to triazole fungicides (Mu et al., 2013; Hermsen et al., levels of T3 and T4 in zebrafish larvae (Jia et al., 2016).
2011).39,52 Thus, the disruption of lipid metabolism is a possible Our research indicated that phthalates can induce a variety of
pathway that could explain the yolk sac abnormities observed in effects during zebrafish embryonic development, including
this study. decreased body length, yolk sac abnormities, immune response,
Both in vivo and in vitro assays have demonstrated that estrogenic effects, and reduced lipid levels. The effective concen-
phthalates can induce immune response (Kimber and Dearman, tration required to produce immune response, lipid homeostasis
2010). In human tests, in utero exposure to DEHP induced the and yolk sac development was lower than that required for estro-
expression of immune response markers and increased the serum genic effects and hormone disruption, indicating that these pro-
TNF level in postnatal day 60 male offspring (Campioli et al., 2014). cesses might be more sensitive to phthalate exposure than
Xu et al. (2013) reported that the transcription of innate immune- endocrine system. Furthermore, an estrogen receptor inhibition
related genes including IL-1b, TNFa, CC-chemokine, CXCL-clc, and test revealed that DBP-induced morphological symptoms are not
lysozyme were up-regulated in zebrafish upon exposure to DBP and mediated by ERs. Taken together, the results identified a series of
DEP. In the present study, an apparent immune response was developmental abnormities caused by phthalates that may not
observed after phthalate exposure, including the up-regulation of originate from estrogenic effects.
the expressions of immune proteins and their encoding genes.
Previous studies also reported that DEHP and DBP can significantly Acknowledgements
affect fish immune systems through the inhibition of macrophage
formation and neutrophil bodies as well as the induction of il-1b, This work was supported by the Central Public-interest Scien-
IFNg, CCL20, and rag1 transcription (Huang et al., 2015; Xu et al., tific Institution Basal Research Fund, CAFS (No. 2016C008) and the
2015). It has been demonstrated that estrogens are involved in Central Public-interest Scientific Institution Basal Research Fund,
immune processes, and estrogen-receptor proteins in fish immune CAFS (NO. 2017HYZD0201).
cells participate in the regulation of immune response (Milla et al.,
2011). Thus, exogenous estrogen may act on the fish immune sys- Appendix A. Supplementary data
tem by altering the level of ER protein (Casanova-Nakayama et al.,
2011). However, in this work, 50 mg/L DEHP and DBP induced sig- Supplementary data related to this article can be found at
nificant immune responses but had no effect on estrogen level, ERa https://doi.org/10.1016/j.envpol.2018.05.095.
protein level, or ESR1 transcription. Based on the above results, the
immune system disruption caused by DEHP and DBP might not
References
originate from the alteration of estrogen receptors. Previous
research have found that immune response is related to the alter- Blanc, M., Hsieh, W.Y., Robertson, K.A., Watterson, S., Shui, G., Lacaze, P.,
ation of lipid homeostasis (Im et al., 2011; Blanc et al., 2011). Khondoker, M., Dickinson, P., Sing, G., Rodríguez-Martín, S., Phelan, P.,
Further, levels of multiple lipid species were altered by DEHP and Forster, T., Strobl, B., Müller, M., Riemersma, R., Osborne, T., Wenk, M.R.,
Angulo, A., Ghazal, P., 2011. Host defense against viral infection involves inter-
DBP in the present work. Thus we inferred that the disruption of feron mediated down-regulation of sterol biosynthesis. PLoS Biol. 9 (3)
lipid levels is a possible pathway mediating the phthalates-induced e1000598.
immune response. Boas, M., Frederiksen, H., Feldt-Rasmussen, U., Skakkebæk, N.E., Hegedüs, L.,
Hilsted, L., Juul, A., Main, K.M., 2010. Childhood exposure to phthalates: asso-
The endocrine-disrupting effect has been the focus of consid- ciations with thyroid function, insulin-like growth factor I, and growth. Environ.
erable ecotoxicity research on phthalate contaminants. Over the Health Perspect. 118 (10), 1458e1464.
past decade, a large number of studies have focused on the effects Brox, S., Seiwert, B., Küster, E., Reemtsma, T., 2016. Toxicokinetics of polar chemicals
in zebrafish embryo (Danio rerio): influence of physicochemical properties and
of phthalates on fish hormone levels. Wang et al. (2013) examined of biological processes. Environ. Sci. Technol. 50 (18), 10264e10272.
the effects of long-term DEHP exposure at environmentally rele- Campioli, E., Martinez-Arguelles, D.B., Papadopoulos, V., 2014. In utero exposure to
vant concentrations on the endocrine systems of adult rare min- the endocrine disruptor di-(2-ethylhexyl) phthalate promotes local adipose and
systemic inflammation in adult male offspring. Nutr. Diabetes 4, e115.
nows (Gobiocypris rarusrare). They found that the plasma 17b- Casanova-Nakayama, A., Wenger, M., Burki, R., Eppler, E., Krasnov, A., Segner, H.,
estradiol level was significantly increased in male fish and signifi- 2011. Endocrine disrupting compounds: can they target the immune system of
cantly decreased in female fish after exposure to DEHP at concen- fish? Mar. Pollut. Bull. 63, 412e416.
Chiang, H., Kuo, Y., Shen, C., Lin, Y., Wang, S., Tsou, T., 2016. Mono (2-ethylhexyl)
trations of 39.4 mg/L or higher. Guo et al. (2015) reported that after
phthalate accumulation disturbs energy metabolism of fat cells. Arch. Toxicol.
six months of DEHP exposure (from larvae to adult), the fecundity, 90 (3), 589e601.
plasma 17-b estradiol level, and plasma testosterone level of female Cho, S.C., Bhang, S.Y., Hong, Y.C., Shin, M.S., Kim, B.N., Kim, J.W., et al., 2010. Rela-
rare minnows were significantly decreased, and Cyp17 and cyp19b tionship between environmental phthalate exposure and the intelligence of
school-age children. Environ. Health Perspect. 118, 1027e1032.
transcription were down-regulated in the ovaries. After 81 days of Corradetti, B., Stronati, A., Tosti, L., Manicardi, G., Carnevali, O., Bizzaro, D., 2013. Bis-
exposure to low MEHP concentrations (0.46, 4.0, and 37.5 mg/L), the (2-ethylexhyl) phthalate impairs spermatogenesis in zebrafish (Danio rerio).
682 X. Mu et al. / Environmental Pollution 241 (2018) 674e683

Reprod. Biol. 13 (3), 195e202, 2013. Lee, K.M., Kho, Y., Kim, P.G., Park, S.H., Lee, J.H., 2017. Urinary levels of phthalate
Crago, J., Klaper, R., 2012. A mixture of an environmentally realistic concentration of metabolites and associations with demographic characteristics in Korean
a phthalate and herbicide reduces testosterone in male fathead minnow adults. Environ. Sci. Pollut. Res. Int. 24 (17), 14669e14681.
(Pimephales promelas) through a novel mechanism of action. Aquat. Toxicol Lien, Y., Ku, H., Su, P., Chen, S., Chen, H., Liao, P., Chen, W., Wang, S., 2015. Prenatal
110e111, 74e83. exposure to phthalate esters and behavioral syndromes in children at 8 Years of
Engel, S.M., Miodovnik, A., Canfield, R.L., Zhu, C., Silva, M.J., Calafat, A.M., et al., 2010. age: taiwan maternal and infant cohort study. Environ. Health Perspect. 123 (1),
Prenatal phthalate exposure is associated with childhood behavior and exec- 95e100.
utive functioning. Environ. Health Perspect. 118, 565e571. Lind, P.M., Roos, V., Ro €nn, M., Johansson, L., Ahlstro €m, H., Kullberg, J., Lind, L., 2012.
European Union, 2008. European Union Risk Assessment Report. Bis (2-ethylhexyl) Serum concentrations of phthalate metabolites are related to abdominal fat
Phthalate (DEHP). Available: http://www.dehp-facts.com/upload/documents/ distribution two years later in elderly women. Environ. Health 11, 21.
webpage/DEHP%20RA%20report%20full.pdf. (Accessed 24 June 2012). Li, X.J., Jiang, L., Chen, L., Chen, H.S., Li, X., 2013. Neurotoxicity of dibutyl phthalate in
Fromme, H., Küchler, T., Otto, T., Pilz, K., Müller, J., Wenzel, A., 2002. Occurrence of brain development following perinatal exposure: a study in rats. Environ.
phthalates and bisphenol A and F in the environment. Water Res. 36, Toxicol. Pharmacol. 36, 392e402.
1429e1438. Li, Y., Zhuang, M.Z., Li, T., Shi, N., 2009. Neurobehavioral toxicity study of dibutyl
Gao, H., Zhu, Y.D., Xu, Y.Y., Zhang, Y.W., Yao, H.Y., Sheng, J., Jin, Z.X., Ren, L.L., phthalate on rats following in utero and lactational exposure. J. Appl. Toxicol.
Huang, K., Hao, J.H., Tao, F.B., 2017. Season-dependent concentrations of urinary 29, 603e611.
phthalate metabolites among Chinese pregnant women: repeated measures Liu, X., Shi, J., Bo, T., Zhang, H., Wu, W., Chen, Q., Zhan, X., 2014. Occurrence of
analysis. Environ. Int. 104, 110e117. phthalic acid esters in source waters: a nationwide survey in China during the
Gaspar, F.W., Castorina, R., Maddalena, R.L., Nishioka, M.G., McKone, T.E., period of 2009-2012. Environ. Pollut. 184, 262e270.
Bradman, A., 2014. Phthalate exposure and risk assessment in California child Maresca, M.M., Hoepner, L.A., Hassoun, A., Oberfield, S.E., Mooney, S.J., Calafat, A.M.,
care facilities. Environ. Sci. Technol. 48 (13), 7593e7601. Ramirez, J., Freyer, G., Perera, F.P., Whyatt, R.M., Rundle, A.G., 2016. Prenatal
Golshan, M., Hatef, A., Socha, M., Milla, S., Butts, I.A.E., Carnevali, O., Rodina, M., exposure to phthalates and childhood body size in an urban cohort. Environ.
Sokołowska-Mikołajczyk, M., Fontaine, P., Linhart, O., Alavi, S.M.H., 2015. Di-(2- Health Perspect. 124 (4), 514e520.
ethylhexyl)-phthalate disrupts pituitary and testicular hormonal functions to Meeker, J.D., Ferguson, K.K., 2011. Relationship between urinary phthalate and
reduce sperm quality in mature goldfish. Aquat. Toxicol 163, 16e26. bisphenol A concentrations and serum thyroid measures in U.S. adults and
Guo, Y., Alomirah, H., Cho, H., Minh, T.B., Mohd, M.A., Nakata, H., Kannan, K., 2011. adolescents from the National Health and Nutrition Examination Survey
Occurrence of phthalate metabolites in human urine from several asian coun- (NHANES) 2007-2008. Environ. Health Perspect. 119 (10), 1396e1402.
tries. Environ. Sci. Technol. 45 (7), 3138e3144. Me rida-Ortega, A.1,  Hernandez-Alcaraz, C.1, Herna ndez-Ramírez, R.U.2, García-
Guo, Y., Yang, Y., Gao, Y., Wang, X., Zhou, B., 2015. The impact of long term exposure Martínez, A.1, Trejo-Valdivia, B.1, Salinas-Rodríguez, A.1, Svensson, K.3,
to phthalic acid esters on reproduction in Chinese rare minnow (Gobiocypris Cebria n, M.E.4, Franco-Marina, F.5, Lo pez-Carrillo, L.6, 2016. Phthalate exposure,
rarus). Environ. Pollut. 203, 130e136. flavonoid consumption and breast cancer risk among Mexican women. Environ.
Hauser, R., Gaskins, A.J., Souter, I., Smith, K.W., Dodge, L.E., Ehrlich, S., Meeker, J.D., Int. 96, 167e172.
Calafat, A.M., Williams, P.L., EARTH Study Team, 2016. Urinary phthalate Milla, S., Depiereux, S., Kestemont, P., 2011. The effects of estrogenic and androgenic
metabolite concentrations and reproductive outcomes among women under- endocrine diruptors on the immune system of fish: a reveiw. Ecotoxicology 20,
going in vitro fertilization: results from the EARTH study. Environ. Health 305e319.
Perspect. 124 (6), 831e839. Mu, X., Chai, T., Wang, K., Zhu, L., Huang, Y., Shen, G., Li, Y., Li, X., Wang, C., 2016. The
Hermsen, S.A.B., Pronk, T.E., Brandhof, E., Ven, L.T.M., Piersma, A.H., 2011. Chemical developmental effect of difenoconazole on zebrafish embryos: a mechanism
class-specific gene expression changes in the zebrafish embryo after exposure research. Environ. Pollut. 212, 18e26.
to glycol ether alkoxy acids and 1,2,4-triazole antifungals. Reprod. Toxicol. 32, Mu, X., Pang, S., Sun, X., Gao, J., Chen, J., Chen, X., Li, X., Wang, C., 2013. Evaluation of
245e252. acute and developmental effects of difenoconazole via multiple stage zebrafish
Huang, H.B., Pan, W.H., Chang, J.W., Chiang, H.C., Guo, Y.L., Jaakkola, J.J., Huang, P.C., assays. Environ. Pollut. 175, 147e157.
2017. Does exposure to phthalates influence thyroid function and growth hor- Mu, X., Huang, Y., Li, X., Lei, Y., Teng, M., Li, X., Wang, C., Li, Y., 2018. Developmental
mone homeostasis? The Taiwan Environmental Survey for Toxicants (TEST) effects and estrogenicity of bisphenol a alternatives in a zebrafish embryo
2013. Environ. Res. 153, 63e72. model. Environ. Sci. Technol. 52 (5), 3222e3231.
Huang, S.S.Y., Benskin, J.P., Chandramouli, B., Butler, H., Helbing, C.C., Cosgrove, J.R., Nagel, R., 2002. DarT: the embryo test with the zebrafish Danio rerio e a general
2016. Xenobiotics produce distinct metabolomic responses in zebrafish larvae model in ecotoxicology and toxicology. ALTEX 19, 38e48.
(Danio rerio). Environ. Sci. Technol. 50 (12), 6526e6535. Net, S., Sempe re , R., Delmont, A., Paluselli, A., Ouddane, B., 2015. Occurrence, fate,
Huang, Q., Chen, Y.1, Chi, Y., Lin, Y., Zhang, H., Fang, C., Dong, S., 2015. Immunotoxic behavior and ecotoxicological state of phthalates in different environmental
effects of perfluorooctane sulfonate and di(2-ethylhexyl) phthalate on the matrices. Environ. Sci. Technol. 49 (7), 4019e4035.
marine fish Oryzias melastigma. Fish Shellfish Immunol. 44, 302e306. Pant, N., Shukla, M., Kumar Patel, D., Shukla, Y., Mathur, N., Kumar Gupta, Y., et al.,
Im, S., Yousef, L., Blaschitz, C., Liu, J.Z., Edwards, R.A., Young, S.G., Raffatellu, M., 2008. Correlation of phthalate exposures with semen quality. Toxicol. Appl.
Osborne, T.F., 2011. Linking lipid metabolism to the innate immune response in Pharmacol. 231 (1), 112e116.
macrophages through sterol regulatory element binding protein -1a. Cell Sant, K.E., Dolinoy, D.C., Jilek, J.L., Shay, B.J., Harris, C., 2016. Mono-2-ethylhexyl
Metabol. 13 (5), 540e549. phthalate (MEHP) alters histiotrophic nutrition pathways and epigenetic pro-
ISO, 1996. Water Quality e Determination of the Acute Lethal Toxicity of Substances cesses in the developing conceptus. J. Nutr. Biochem. 27, 211e218.
to a Freshwater Fish [Brachydanio rerio Hamiltone Buchanan (Teleostei, Cyp- Schmidt, J., Schaedlich, K., Fiandanese, N., Pocar, P., Fischer, B., 2012. Effects of di(2-
rinidae)]. In: Part 3: Flow-through Method. ethylhexyl) phthalate (DEHP) on female fertility and adipogenesis in C3H/N
Jia, L.L., Lou, X.Y., Guo, Y., Leung, K.S., Zeng, E.Y., 2017. Occurrence of phthalate esters mice. Environ. Health Perspect. 120 (8), 1123e1129.
in over-the-counter medicines from China and its implications for human Sohn, J., Kim, S., Koschorreck, J., Kho, Y., Choi, K., 2016. Alteration of sex hormone
exposure. Environ. Int. 98, 137e142. levels and steroidogenic pathway by several low molecular weight phthalates
Jia, P., Ma, Y., Lu, C., et al., 2016. The effects of disturbance on hypothalamus- and their metabolites in male zebrafish (Danio rerio) and/or human adrenal cell
pituitary-thyroid (HPT) Axis in zebrafish larvae after exposure to DEHP. PLoS (H295R) line. J. Hazard Mater. 320, 45e54.
One 11 (5) e0155762. Specht, I.O., Toft, G., Hougaard, K.S., Lindh, C.H., Lenters, V., Jo €nsson, B.A.,
Joensen, U.N., Frederiksen, H., Blomberg Jensen, M., Lauritsen, M.P., Olesen, I.A., Heederik, D., Giwercman, A., Bonde, J.P., 2014. Associations between serum
Lassen, T.H., Andersson, A.M., Jørgensen, N., 2012. Phthalate excretion pattern phthalates and biomarkers of reproductive function in 589 adult men. Environ.
and testicular function: a study of 881 healthy Danish men. Environ. Health Int. 66, 146e156.
Perspect. 120 (10), 1397e1403. Sun, G., Liu, K., 2017. Developmental toxicity and cardiac effects of butyl benzyl
Kang, Y., Man, Y.B., Cheung, K.C., Wong, M.H., 2012. Risk assessment of human phthalate in zebrafish embryos. Aquat. Toxicol. 192, 165e170.
exposure to bioaccessible phthalate esters via indoor dust around the pearl 
Supek, F., Bosnjak, M., Skunca, 
N., Smuc, T., 2011. REVIGO summarizes and visualizes
river delta. Environ. Sci. Technol. 46 (15), 8422e8430. long lists of Gene Ontology terms. PLoS One 6 (7), e21800.
Kay, V.R., Chambers, C., Foster, W.G., 2013. Reproductive and developmental effects Tsai, Y.A., Lin, C.L., Hou, J.W., Huang, P.C., Lee, M.C., Chen, B.H., Wu, M.T., Chen, C.C.,
of phthalate diesters in females. Crit. Rev. Toxicol. 43, 200e219. Wang, S.L., Lee, C.C., Hsiung, C.A., Chen, M.L., RAPIT Group, 2016. Effects of high
Khan, H.H., Jung, J.Y., 2008. Ozonation catalyzed by homogeneous and heteroge- di(2-ethylhexyl) phthalate (DEHP) exposure due to tainted food intake on pre-
neous catalysts for degradation of DEHP in aqueous phase. Chemosphere 72, pubertal growth characteristics in a Taiwanese population. Environ. Res. 149,
690e696. 197e205.
Kimber, I., Dearman, R.J., 2010. An assessment of the ability of phthalates to influ- Uren-Webster, T.M., Lewis, C., Filby, A.L., Paull, G.C., Santos, E.M., 2010. Mechanisms
ence immune and allergic responses. Toxicology 271, 73e82. of toxicity of di(2-ethylhexyl) phthalate on the reproductive health of male
Kinch, C.D., Ibhazehiebo, K., Jeong, J., Habibi, H.R., Kurrasch, D.M., 2015. Low-dose zebrafish. Aquat. Toxicol 99, 360e369.
exposure to bisphenol A and replacement bisphenol S induces precocious hy- Wang, X., Yang, Y., Zhang, L., Ma, Y., Han, J., Yang, L., Zhou, B., 2013. Endocrine
pothalamic neurogenesis in embryonic zebrafish. Proc. Natl. Acad. Sci. Unit. disruption by di-(2-ethylhexyl)-phthalate (DEHP) in Chinese rare minnow
States Am. 112 (5), 1475e1480. (Gobiocypris rarus). Environ. Toxicol. Chem. 32 (8), 1846e1854.
Kobrosly, R.W., Evans, S., Miodovnik, A., Barrett, E.S., Thurston, S.W., Calafat, A.M., Wiegand, M.D., 1996. Composition, accumulation and utilization of yolk lipids in
Swan, S.H., 2014. Prenatal phthalate exposures and neurobehavioral develop- teleost fish. Rev. Fish Biol. Fish. 6, 259e286.
ment scores in boys and girls at 6-10 years of age. Environ. Health Perspect. 122 Xu, H., Dong, X., Zhang, Z., Yang, M., Wu, X., Liu, H., Lao, Q., Li, C., 2015. Assessment
(5), 521e528. of immunotoxicity of dibutyl phthalate using live zebrafish embryos. Fish
X. Mu et al. / Environmental Pollution 241 (2018) 674e683 683

Shellfish Immunol. 45, 286e292. fate and effects of di (2-ethylhexyl) phthalate in wastewater treatment plants: a
Xu, H., Shao, X., Zhang, Z., Zou, Y., Wu, X., Yang, L., 2013. Oxidative stress and im- review. Environ. Pollut. 194, 281e293.
mune related gene expression following exposure to di-n-butyl phthalate and Zota, A.R., Phillips, C.A., Mitro, S.D., 2016. Recent fast food consumption and
diethyl phthalate in zebrafish embryos. Ecotoxicol. Environ. Saf. 93, 39e44. bisphenol a and phthalates exposures among the U.S. Population in NHANES,
Yamasaki, K., Okuda, H., Takeuchi, T., Minobe, Y., 2009. Effects of in utero through 2003-2010. Environ. Health Perspect. 124 (10), 1521e1528.
lactational exposure to dicyclohexyl phthalate and p,p’-DDE in Sprague-Dawley Zhang, C., Yang, X., He, Z., Zhong, Q., Guo, J., Hu, X.J., Xiong, L., Liu, D., 2014. Influence
rats. Toxicol. Lett. 189, 14e20. of BBP exposure on nervous system and antioxidant system in zebrafish. Eco-
Ye, T., Kang, M., Huang, Q., Fang, C., Chen, Y., Shen, H., Dong, S., 2014. Exposure to toxicology 23 (10), 1854e1857, 2014.
DEHP and MEHP from hatching to adulthood causesreproductive dysfunction Zhang, X., Zhou, Q., Zou, W., Hu, X., 2017. Molecular mechanisms of developmental
and endocrine disruption in marine medaka (Oryzias melastigma). Aquat. Tox- toxicity induced by graphene oxide at predicted environmental concentrations.
icol 146, 115e126. Environ. Sci. Technol. 51 (14), 7861e7871.
Yuwatini, E., Hata, N., Taguchi, S., 2006. Behavior of di(2-ethylhexyl) phthalate Zhang, Y., Cao, Y., Shi, H., Jiang, X., Zhao, Y., Fang, X., Xie, C., 2015. Could exposure to
discharged from domestic waste water into aquatic environment. J. Environ. phthalates speed up or delay pubertal onset and development? A 1.5-year
Monit. 8, 191e196. follow-up of a school-based population. Environ. Int. 83, 41e49.
Zeng, F., Cui, K., Xie, Z., Liu, M., Li, Y., Lin, Y., Zeng, Z., Li, F., 2008. Occurrence of Zhu, Y., Hua, R., Zhou, Y., Li, H., Quan, S., Yu, Y., 2016. Chronic exposure to mono-(2-
phthalate esters in water and sediment of urban lakes in a subtropical city, ethylhexyl)-phthalate causes endocrine disruption and reproductive dysfunc-
Guangzhou, South China. Environ. Int. 34, 372e380. tion in zebrafish. Environ. Toxicol. Chem. 35 (8), 2117e2124.
Zolfaghari, M., Drogu, P., Seyhi, B., Brar, S.K., Buelna, G., Dube , R., 2014. Occurrence,

You might also like