You are on page 1of 10

Ecotoxicology and Environmental Safety 229 (2022) 113069

Contents lists available at ScienceDirect

Ecotoxicology and Environmental Safety


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

Comparison of the mechanisms of estrogen disrupting effects between


triphenyl phosphate (TPhP) and tris(1,3-dichloro-2-propyl)
phosphate (TDCIPP)
Xiaoya Ji a, b, Na Li a, *, Mei Ma a, c, *, Xinyan Li d, Kongrui Zhu a, c, Kaifeng Rao a, Zijian Wang c,
Jingfeng Wang e, Yanjun Fang e
a
Key Laboratory of Drinking Water Science and Technology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
b
School of Public Health, Qingdao University, Qingdao 266000, China
c
College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
d
Institute of Environmental and Ecological Engineering, Guangdong University of Technology, Guangzhou 510006, China
e
Tianjin Institute of Environmental and Operational Medicine, Key Laboratory of Risk Assessment and Control for Environment & Food Safety, Tianjin 300050, China

A R T I C L E I N F O A B S T R A C T

Edited by Dr. G. Liu As the typical aryl-organophosphate flame retardants (OPFRs), triphenyl phosphate (TPhP) and tris(1,3-dichloro-
2-propyl) phosphate (TDCIPP) were reported to be estrogen disruptors. However, estrogen receptor α (ERα)
Keywords: binding experiments could not explain their biological effects. In this study, their action on ERα, G protein-
Triphenyl phosphate (TPhP) coupled estrogen receptor (GPER) and the synthesis of 17β-estradiol (E2) were investigated using in vitro as­
Tris(1,3-dichloro-2-propyl) phosphate
says and molecular docking. The results showed that TPhP acted as an ERα agonist and recruited steroid receptor
(TDCIPP)
co-activator 1 (SRC1) and 3 (SRC3), which was found for the first time. Unlike TPhP, TDCIPP acted as an ERα
Estrogen disrupting effects
Estrogen receptor α (ERα) antagonist. However, both TPhP and TDCIPP activated the estrogen pathway by GPER in SKBR3 cells which were
G protein-coupled estrogen receptor (GPER) lack of ERα. Although molecular docking results revealed that both TPhP and TDCIPP could dock into ERα and
Estrogen biosynthesis GPER, their substituent groups and combination mode might affect the receptor activation. In addition, by using
estrogen biosynthesis assay in H295R cells, both of TPhP and TDCIPP were found to promote E2 synthesis and
E2/T ratio involving their different alteration on levels of progesterone, testosterone and estrone, and expression
of various key genes. Our data proposed estrogen-disrupting mechanism frameworks of TPhP and TDCIPP.
Moreover, our results will contribute to future construction of adverse outcome pathway (AOP) framework of
endocrine disruptors.

1. Introduction the environment, leading to their frequent detection in varied environ­


ment media (water, house dust, soil), aquatic organism and even in
In response to the gradual restriction of polybrominated diphenyl human samples (Brandsma et al., 2014; Kim et al., 2014; Reemtsma
ethers (PBDEs), there is growing need in the production and consump­ et al., 2008; Wan et al., 2016; Ya et al., 2019). In the 150 samples
tion of organophosphate flame retardants (OPFRs) as one of the poten­ consisting of river water, lake water, tap water, seawater and rainwater
tial alternatives (van der Veen and de Boer, 2012). Triphenyl phosphate collected from New York State, the average levels of TPhP and TDCIPP
(TPhP) and tris(1,3-dichloro-2-propyl) phosphate (TDCIPP) as shown in ranged from 0.53 to 11.0 and 4.75–21.1 ng/L, respectively (Kim and
Fig. 1 are typical aryl-OPFR and chlorinated-OPFR, respectively, and Kannan, 2018). The highest concentrations of TPhP and TDCIPP in 60
widely used in baby products, residential furniture, electronic equip­ floor dust samples located in Beijing, China reached 3514 and 3741
ment and building materials (Cooper et al., 2016; Stapleton et al., 2011; ng/g, respectively (Wu et al., 2016). TPhP and TDCIPP have also been
Wei et al., 2015). Because TPhP and TDCIPP as additive flame retardants found in human breast milk samples (Zheng et al., 2021), children’s
do not form chemical bonds with materials, they are easily released into urine (Y. Li et al., 2019; N. Li et al., 2019), human placenta (Ding et al.,

* Correspondence to: Key Laboratory of Drinking Water Science and Technology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences,
18 Shuangqing Road, Haidian District, Beijing 100085, China.
E-mail addresses: nali@rcees.ac.cn (N. Li), mamei@rcees.ac.cn (M. Ma).

https://doi.org/10.1016/j.ecoenv.2021.113069
Received 9 October 2021; Received in revised form 30 November 2021; Accepted 5 December 2021
Available online 8 December 2021
0147-6513/© 2021 The Authors. Published by Elsevier Inc. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
X. Ji et al. Ecotoxicology and Environmental Safety 229 (2022) 113069

effects of EDCs on GPER. The H295R cells are sensitive to the EDCs that
disrupt the steroidogenic processes including the E2 biosynthesis. In the
present study, the action on ERα of TPhP and TDCIPP was determined by
the MVLN cell assay, molecular docking and the ERα-SRCs recruitment
assay was conducted to characterize the SRCs recruitment of the ERα
agonist. Their interaction with GPER was confirmed by molecular
docking and their activation on GPER-mediated signaling was further
investigated in SKBR3 cells. Moreover, their effects on steroid hormone
levels (progesterone, estrone, testosterone and E2) and expression of
genes involved were measured in H295R cells.

2. Materials and methods

2.1. Chemicals and cell culture


Fig. 1. The structures of TPhP and TDCIPP.

Triphenyl phosphate (TPhP, 99%) and Tris(1,3-dichloro-2-propyl)


2016) and human blood (Zhao et al., 2016), which has aroused great
phosphate (TDCPP, 99%) were purchased from J&K Chemical (Bei­
concern about their potential adverse effects.
jing, China). The chemicals were dissolved in dimethyl sulfoxide
Several studies have demonstrated that exposure to TPhP and
(DMSO) and stored at − 20 ◦ C. To avoid cytotoxicity from DMSO, its
TDCIPP are associated with developmental toxicity (Fu et al., 2013; Y. Li
final concentration in the culture medium was below 0.1% (V/V).
et al., 2019; N. Li et al., 2019), endocrine-disrupting effects (Bajard
SKBR3 cells and H295R cells were purchased from the cell bank of
et al., 2021; Rosenmai et al., 2021; Zhang et al., 2016), immunotoxicity
the Chinese Academy of Sciences (Beijing, China). MVLN cells were
(Farhat et al., 2014), cardiotoxicity (Mitchell et al., 2018), obesity
MCF-7 cells that contained estrogen responsive element (ERE)-
(Hoffman et al., 2015) and neurotoxicity (Hong et al., 2018; J. Wang
controlled luciferase reporter gene, and were cultured in DMEM
et al., 2015; Q.W. Wang et al., 2015; Qiangwei Wang et al., 2015).
(HyClone, USA) with 10% fetal bovine serum and 1% penicillin/strep­
Currently, there is increasing evidence that TPhP and TDCIPP also have
tomycin (Gibco, USA) in an incubator at 37 ◦ C with 5% CO2. SKBR3 cell
potential to disrupt estrogen function which is thought to act vital role in
were cultured in RPMI 1640 medium (HyClone, USA) with 10% fetal
reproduction, growth and development (Kojima et al., 2013; Liu et al.,
bovine serum and 1% penicillin/streptomycin in an incubator at 37 ◦ C
2016; J. Wang et al., 2015; Q.W. Wang et al., 2015; Qiangwei Wang
with 5% CO2. H295R cells were cultured in DMEM/F12 medium
et al., 2015). In in vivo, exposure to TPhP and TDCIPP affect
(HyClone, USA) with 1% Ultroser G (Pall Corporation, USA), 1% ITS
hypothalamic-pituitary-gonadal (HPG) axis of zebrafish along with
(Gibco, USA) and 1% penicillin/streptomycin in an incubator at 37 ◦ C
alteration of plasma 17β-estradiol (E2) and testosterone (T), and sig­
with 5% CO2.
nificant increases of vitellogenin (VTG) (Liu et al., 2013), which suggests
that they can alter hormone metabolism and biosynthesis to induce es­
2.2. Cell viability
trogenic activities. However, the underlying mechanisms of TPhP and
TDCIPP to exert their estrogen disrupting effects were still unclear.
MVLN cells (1.5 × 104 cells/well), SKBR3 cells (3 × 103 cells) and
Although TPhP and TDCIPP have been reported to interact with estrogen
H295R cells (3 × 103 cells/well) were seeded in 96-well plates with
receptor α (ERα) to exert estrogen-disrupting activities. However, con­
100 μL medium for 24 h. The MVLN cells, SKBR3 cells and H295R cells
troversy remains regarding their effects on ERα in different in vitro
were treated with serial dilution of test compounds for 72 h, 24 h and
systems (Liu et al., 2012; Zhang et al., 2014). Therefore, the
48 h exposure, respectively. After treatment, cells were stained with
estrogen-disrupting activities of TPhP and TDCIPP might result from
Hoechst 33,342 for overnight. The images were acquired by an Oper­
various pathways, and understanding mechanisms involved is un­
etta™ High Content Screening instrument (PerkinElmer, Waltham, MA,
doubtedly critical to assessing their toxicity.
USA), and the cell number was analyzed by Harmony™ software. The
It is well known that the molecular mechanisms of estrogen-
relative cell viability was calculated as the ratio the cell number of the
disrupting effects induced by endocrine disrupting chemicals (EDCs)
DMSO control. The following experiments used non-cytotoxic concen­
are complicated. One of the classical genomic pathways is EDCs interact
trations of compounds.
with ERα to recruit co-activators to promote the transcription of target
genes (Zhu et al., 2019). Moreover, some EDCs such as bisphenol A
2.3. MVLN cell assay
(BPA) (Vinas and Watson, 2013), cadmium (Liu et al., 2019), hydrox­
ylated polybrominated diphenyl ethers (OH-PBDEs) (Cao et al., 2018b)
MVLN cells (1.5 × 104 cells/well) were seeded in 96-well plates and
and thiodiphenol (Lei et al., 2017) have potential to active G
cultured in phenol red-free DMEM with 10% charcoal-stripped FBS.
protein-coupled estrogen receptor (GPER)-mediated rapid non-genomic
After 24 h, the cells were treated with 20 μM, 10 μM, 2 μM, 1 μM,
pathways to exert effects including proliferation and migration of breast
0.2 μM and 0.1 μM of TPhP, and 10 μM, 2 μM, 1 μM, 0.2 μM and 0.1 μM
cancer cells. In addition, EDCs also can disrupt the E2 biosynthesis to
of TDCIPP, respectively. After 72 h exposure, the luciferase activity was
cause estrogen-disrupting effects, involving alteration in the levels of
detected by the Steady-Glo-Luciferase Assay System (Promega Corpo­
various steroid hormones and the expression of multiple key enzymes
ration, USA) and determined using a microplate reader (Tecan GENios
(Feng et al., 2016; He et al., 2008; J. Wang et al., 2015; Q.W. Wang et al.,
A-5002, Salzburg, Austria). The relative luciferase activity for each
2015; Qiangwei Wang et al., 2015). However, the toxicity mechanisms
sample well was normalized as the ratio the luciferase activity of the
involved in estrogen-disrupting activities of TPhP and TDCIPP had not
DMSO control. The anti-estrogenic activity of the test compounds was
been well understood and their action on various pathways remains to
determined by coincubation of MVLN cells with E2.
be elucidated.
Various in vitro cell models were developed to determine the modes
2.4. ERα-SRCs recruitment assay
of action of EDCs. The ERα positive human breast MVLN cells that
contained ERE-controlled reporter genes are used to investigate the ef­
The LanthaScreen ERα SRCs kit and 10 fluorescein-tagged SRCs
fects of EDCs on ERα-ERE pathway (Pons et al., 1990). The human breast
peptides including SRC1–1, SRC1–2, SRC1–3, SRC1–4, SRC2–1, SRC2–2,
SKBR3 cells (ERα-, GPER+) were routinely conducted to study the
SRC2–3, SRC3–1, SRC3–2 and SRC3–3, were purchased from Life

2
X. Ji et al. Ecotoxicology and Environmental Safety 229 (2022) 113069

Technology (CA, USA). According to the manufacturer’s instructions, 2.9. Data analysis
0.25 μM SRC peptide, 5 nM Tb anti-GST antibody, 3.5 nM ERα-LBD, and
serial concentrations including 0.01 μM, 0.1 μM, 1 μM, 10 μM, 100 μM All data was analyzed by OriginPro 2016 (Origin Lab, Northampton,
and 1000 μM of test compounds were gently mixed in a 384-well plate USA). All results were expressed as the mean ± SD (standard deviation)
(Corning, NY, USA). After 1 h incubation at room temperature from of at least three independent experiments. Levene’s test was used to
light, the fluorescence was read on Spark 10 M (TECAN) with TR-FRET check homogeneity of variances. Statistical significance was analyzed by
systems (EX = 340 nm, EM = 495 and 520 nm). one-way analysis of variance (ANOVA) followed by Tukey’s test and the
level of significant difference was p < 0.05.

2.5. Docking analysis 3. Results and discussion

The GPER homology protein model was kindly provided by Men­ 3.1. The effects of TPhP and TDCIPP on ERα
dez-Luna et al. (2016), and has been widely performed in reported
studies (Deng et al., 2017; Sarmiento et al., 2018). The Discovery Studio The ERα activation is a significant target of EDCs to induce estrogenic
4.0 software package was used to dock test compounds to ERα and GPER effects, and the molecular docking analysis and MVLN cells that contain
(Accelrys Software Inc., San Diego, CA). The protocol was performed estrogen respective element-controlled luciferase gene were used to
according to the CDOCKER protocol, in which protocol 20 random determine the action of TPhP and TDCIPP on ERα. The results showed
ligand conformations were generated and refined by grid-based simu­ that E2 formed hydrogen bonds with His524, Arg394 and Glu353
lated annealing in the binding site of GPER, and the top-ranking poses (Fig. 2A). TPhP formed a pi-sigma bond with Arg394 (Fig. 2B), and
from the docking were selected. TDCIPP formed a hydrogen bond with Arg394 (Fig. 2C), suggesting that
they had different interaction with ERα. In MVLN cells, the positive
compound E2 had concentration-dependent agonistic activities with
2.6. cAMP
EC50 (the concentration that had 50% of maximal activity) of 2.06 × 10-
11
mol/L (Fig. S1). According to the results of MVLN cells viability
The LANCE Ultra cAMP kit (PerkinElmer, Norwalk, CT, USA) was
(Fig. S2), the concentrations (≤ 2 ×10-5 M for TPhP and ≤ 1 ×10-5 M for
used to determine the cAMP levels. SKBR3 cells (3 × 103 cells) were
TDCIPP) that had no cytotoxicity were used. TPhP showed agonistic
seeded in a 96-well plate (PerkinElmer, USA) with 100 μL phenol red-
response in a dose-dependent manner with EC50 value and maximal
free RPMI 1640 medium for 24 h. The cells were rinsed with HBSS
relative luciferase of 1.02 × 10-5 mol/L and 28.31%, respectively
and treated with 20 μL mixture containing serial concentrations
(Fig. 2D). TDCIPP did not induce agonistic activity (Fig. S3), but it
including 0.1 μM, 1 μM and 10 μM of test compounds, 0.5 mM 3-isobu­
showed strong antagonistic activity inhibiting the luciferase activity
tyl-1-methylxanthine and Alexa-labeled antibodies for 30 min at room
induced by 1 × 10-8 mol/L E2 with a maximal inhibition of 24.82% and
temperature. Then, 20 μL detection solutions were added and incubated
IC50 (the concentration that had 50% of inhibitory activity) value of
for 60 min at room temperature. Finally, the signals at the module of
1.63 × 10-6 mol/L (Fig. 2E), suggesting that TPhP and TDCIPP had
time-resolved fluorescence resonance energy transfer were measured by
opposite effects on ERα. Specifically, TPhP acted as an ERα agonist to
Spark 10 M (TECAN, Switzerland), and the cAMP concentration was
activate ERE transcription, while TDCIPP acted as an ERα antagonist to
normalized according to the manufacturer’s instructions. For the G15 (a
compete with E2 for binding to ERα. Our results were also similar to
GPR30 inhibitor) inhibitory experiments, the cells were pretreated with
publicly available data from ToxCast that TPhP had agonistic activity on
1 μM G15 for 30 min
ERα with EC50 of 7.31 × 10-6 mol/L, while TDCIPP had antagonistic
activity on ERα with IC50 of 7.02 × 10-5 mol/L.
2.7. Hormone measurement Generally, in the ERα-mediated ERE pathway, the ERα agonist binds
to ERα to form an agonist-ERα complex, and then various co-activators
H295R cells (3 × 105 cells/well) were seeded in 24-well plates with are recruited to enhance target gene transcription. Moreover, the p160
1 mL complete medium for 24 h. The cells were treated with serial steroid receptor co-activators (SRCs) family containing SRC1, SRC2 and
concentrations including 0.1 μM, 1 μM, 2 μM and 10 μM of test com­ SRC3 are the first discovered co-activators, and a recent study has
pounds. After 48 h exposure, the medium was centrifuged, collected and demonstrated that differential recruitment of BPA and its analogs to
stored at − 80 ◦ C. According to the manufacturer’s instructions of ELISA SRCs were implicated with their effects on ERα-mediated ERE tran­
kits (Cayman Chemical, USA), the concentrations of progesterone, scription. In order to investigate the SRCs involved in estrogenicity of
testosterone and estradiol were determined. The concentrations of TPhP, the ERα-SRCs recruitment assay containing 10 SRCs peptides was
estrone were determined by an Estrone ELISA Kit (Abnova, Taiwan) used. E2-ERα recruited 9 SRCs peptides except for SRC3–3 (Fig. S4).
according to the manufacturer’s protocol. TPhP showed the concentration-dependent ERα agonistic interaction
with SRC1–1 and SRC3–3 with EC50 of 2.93 × 10-4 and 1.30 × 10-5 mol/
L, respectively (Fig. 2F), and had no effects on other 8 SRCs peptides
2.8. RNA isolation and qPCR (Fig. S5), suggesting that TPhP-ERα recruited SRC1–1 and SRC3–3. As
far as we know, this is the first study to investigate the interaction be­
H295R cells (3 × 105 cells/well) were seeded in 24-well plates with tween ERα and SRCs induced by TPhP, and it was content with our
1 mL complete medium for 24 h. The cells were treated with serial previous studies that TPhP did not induce the ERα interaction with SRC2
concentrations including 0.1 μM, 1 μM, 2 μM and 10 μM of test com­ in the two-hybrid yeast system (Ji et al., 2020b).
pounds. After 48 h exposure, the total RNA was extracted from the cells
using TRIzol reagent (Life Technologies, USA) and reverse transcribed in 3.2. The activation of TPhP and TDCIPP on GPER
a 20 μL reaction system by the FastQuant RT kit (TIANGEN, Beijing,
China). qPCR was performed using an ABI 7500 real-time quantitative The action of GPER has been demonstrated to be involved in
PCR system (Life Technologies, USA) according to the guidelines. The mammalian uterine proliferation, the progression of multiple
primer pairs are shown in the Supporting Information (Table S1), and reproduction-related cells and endocrine-related cells, and GPER is also
β-actin was used as the housekeeping gene for the mRNA expression implicated in the E2-activation of cancer-associated fibroblasts (Pross­
analysis. The expression of steroidogenesis gene was calculated with the nitz and Barton, 2011). There is increasing studies that some EDCs have
2− ΔΔCt method. potential to activate GPER and trigger various downstream pathways,

3
X. Ji et al. Ecotoxicology and Environmental Safety 229 (2022) 113069

Fig. 2. The effects of TPhP and TDCIPP on ERα action. (A-C) Molecular docking analysis of E2, TPhP and TDCIPP with ERα. (D) The agonistic activity of TPhP in the
MVLN cell assay. Data are presented as relative luciferase activity that was normalized to that of the DMSO control. (E) The antagonistic activity of TDCIPP in the
MVLN cell assay. Data are presented as the percent of activity that was inhibited relative to the maximum activity induced by 17β-estradiol (E2, 1 × 10-8 mol/L). (F)
TPhP induced ERα interaction with SRC1–1 and SRC3–3 peptides. Data are presented as the ratio of fluorescent units (520 nm/495 nm) emitted after excitation at
340 nm. Values represent the mean ± SD of three independent experiments.

4
X. Ji et al. Ecotoxicology and Environmental Safety 229 (2022) 113069

such as cyclic adenosine monophosphate (cAMP), phosphoinositide (≤1 ×10-5 M for TPhP and TDCIPP) that had no cytotoxicity were used.
3-kinase/protein kinase B (PI3K/AKT) and intracellular calcium mobi­ TPhP and TDCIPP showed significant agonistic effects on cAMP pro­
lization, to exert estrogen-disrupting effects, resulting in proliferation duction (Fig. 3C), and for the cells pretreated with 1 μM G15, their ef­
and migration of breast cancer cells including MCF-7 and SKBR3 (Cao fects were significantly inhibited (Fig. 3D), indicating that they
et al., 2017; Lei et al., 2017). In order to investigate the activation of activated the GPER-mediated cAMP pathway. Here, it was found for the
TPhP and TDCIPP on GPER, the molecular docking analysis was per­ first time that both TPhP and TDCIPP have potential to activate
formed to determine their interaction with GPER and their effects on GPER-cAMP, but their ways of interaction with GPER were different.
cAMP levels were measured in SKBR3 cells. In molecular docking
analysis, the results showed that E2 formed hydrogen bonds with
Tyr123 and Glu275, and Van der Waals forces with Cys205. G1, a GPER 3.3. The effects of TPhP and TDCIPP on E2 synthesis and mechanisms
agonist, had a hydrogen bond and a pi-pi bond with Gln138 and Phe208, involved
respectively (Fig. S6). TPhP formed a pi-sigma bond with Tyr142
(Fig. 3A), and the CDOCKER interaction energy was − 29.60 kcal/mol. In addition to estrogen receptors-mediated pathways, EDCs also have
TDCIPP formed a hydrogen bond with Gln138 that was similar to G1 been reported to disrupt non-receptor-mediated action concerning E2
(Fig. 3B), and the CDOCKER interaction energy was − 59.33 kcal/mol, synthesis to exert estrogen-disrupting activities including alteration of
suggesting that they had different interaction with GPER. The results VTG levels and promoting human breast cancer cell (MCF-7) prolifera­
from cAMP measurement showed E2 evoked a dose-dependent cAMP tion (Liu et al., 2007; Wrobel and Gregoraszczuk, 2013). In response, the
levels ranging from 1 × 10-10 to 1 × 10-7 mol/L (Fig. S7). According to USA Environmental Protection Agency (EPA) has recommended the
the results of SKBR3 cells viability (Fig. S8), the concentrations H295R cell line to evaluate the effects of EDCs on steroidogenesis, and
several EDCs, such as 2,4-dichlorophenol (Ma et al., 2012), naphthenic

Fig. 3. The effects of TPhP and TDCIPP on GPER action. (A)-(B) Molecular docking analysis of TPhP and TDCIPP with GPER. (C) cAMP production induced by
different concentrations of TPhP and TDCIPP in SKBR3 cells. (D) cAMP production induced by E2 (100 nM), 10 μM TPhP and TDCIPP in the absence or presence of
1 μM G15. The result of the cAMP production was expressed by setting the cAMP production of the DMSO control group as 100%. Values represent the mean ± SD of
three independent experiments. *p < 0.05, compared with the control. # p < 0.05, compared with the groups treated with the compounds in absence of G15.

5
X. Ji et al. Ecotoxicology and Environmental Safety 229 (2022) 113069

acids (J. Wang et al., 2015) and phthalates (Sohn et al., 2016), have been
demonstrated to promote E2 synthesis using H295R cell model, which
were in accordance with in vivo studies showing the increase of E2 levels
in zebrafish. Therefore, the effects of TPhP and TDCIPP on four hormone
concentrations in H295R cells were measured. The positive compound
forskolin increased concentrations of progesterone, estrone, testosterone
(T) and E2 (Fig. S9). According to the results of H295R cells viability
(Fig. S10), the concentrations (≤1 × 10-5 M for TPhP and TDCIPP) that
had no cytotoxicity were used. The E2 levels and E2/T ratio significantly
increased when H295R cells were exposed to TPhP and TDCIPP
(Fig. 4A–C), indicating that they could cause estrogenic response.
However, there was some difference in their effects on the levels of other
hormones involved in the process of E2 synthesis. TPhP increased the
levels of progesterone, but decreased the levels of estrone and testos­
terone (Fig. 4A), similarly, TPhP has been reported to promote proges­
terone synthesis in human placental choriocarcinoma JEC-3 cells (Hu
et al., 2017) and inhibit testosterone levels in Japanese medaka (Li et al.,
2018). TDCIPP increased the levels of estrone and testosterone, and did Fig. 5. The effects of TPhP and TDCIPP on the expression of genes related to
not cause any effects on the levels of progesterone (Fig. 4B). steroidogenesis in H295R cells. Values represent the mean ± SD of three in­
In order to investigate the mechanisms involved, the genes expres­ dependent experiments. Data are presented as the relative mRNA expression
sion of 9 key enzymes (HMGR, StAR, CYP11A1, 3β-HSD2, CYP17, 17β- levels in the exposed group to those in the DMSO control. β-actin was used for
HSD1, 17β-HSD4, CYP19 and SULT2A1) that are associated with the normalization.
synthesis of steroidogenesis were measured. As shown in Fig. 5, the
positive compound forskolin significantly increased expression of 8 and StAR mediates the transfer of cholesterol. The significant increase of
genes and inhibited SULT2A1 expression. Both TPhP and TDCIPP HMGR and StAR gene expression levels was induced by them, which
significantly promoted HMGR, StAR, CYP19 and 17β-HSD1 genes suggested that they promoted cholesterol synthesis and its transfer.
expression and significantly inhibited SULT2A1 gene expression CYP19 and 17β-HSD1 catalyze the conversion of androgen to estrogen
(Table S2). HMGR is a rate-limiting enzyme of cholesterol biosynthesis and the reduction of estrone into E2, respectively. It was found that

Fig. 4. The effects of TPhP and TDCIPP on steroidogenesis in H295R cells. (A) The effects of TPhP on the levels of progesterone, estrone, testosterone and 17β-
estradiol (E2). (B) The effects of TDCIPP on the levels of progesterone, estrone, testosterone and 17β-estradiol (E2). (C) The effects of TPhP and TDCIPP on the E2/T
ratio. Data are presented as relative levels that were normalized to that of the DMSO control. *p < 0.05, compared with the DMSO control.

6
X. Ji et al. Ecotoxicology and Environmental Safety 229 (2022) 113069

TPhP and TDCIPP up-regulated CYP19 and 17β-HSD1 expression to CYP11A1, 3β-HSD2 and CYP17 was not affected, and then the synthesis
promote the conversion of testosterone and estrone to E2, which resul­ and conversion of progesterone is normal, so that its levels remain un­
ted in an increase of E2 concentrations, similarly, up-regulation of changed. 17β-HSD4 catalyzes the reduction of androstenedione to
CYP19A gene and 17β-HSD1 gene have been reported to support an testosterone. No changes of 17β-HSD4 and increased CYP19 expression
increase of E2 induced by 5 phthalates (DEP, MEP, BBzP, MBzP and by TPhP caused that more testosterone is converted to E2 than synthetic
DiBP) (Sohn et al., 2016) and trichloroethylene (Tachachartvanich et al., testosterone, leading to decreased testosterone levels. This result is
2018), respectively. Moreover, the fold of 17β-HSD1 expression content with a previous study that perfluorooctanoic acid (PFOA) and
(10 μM = 3.6, 2 μM = 2.7) promoted by TPhP was significantly higher perfluorooctane sulfonate (PFOS) inhibited testosterone levels due to
than that of CYP19 (10 μM = 2.9, 2 μM = 2.0), implying that the reason their up-regulation of CYP19 expression and no effects on 17β-HSD4
for the lower estrone concentrations is that more estrone is converted to expression (Kang et al., 2016), While TDCIPP promoting 17β-HSD4
E2 than synthetic estrone. On the contrary, the fold of 17β-HSD1 expression resulted in increased testosterone concentrations.
expression (10 μM = 2.7, 2 μM = 2.1) promoted by TDCIPP was TPhP and TDCIPP have been demonstrated to act estrogenic activ­
significantly lower than that of CYP19 (10 μM = 3.7, 2 μM = 2.7), ities in in vivo, they up-regulated VTG levels in zebrafish (Liu et al.,
implying that the reason for the higher estrone concentrations is that 2013), which is thought to be one of biomarkers concerning
more estrone is synthesized than that for further conversion to E2. estrogen-like EDCs. According to the adverse outcome pathway (AOP)
SULT2A1 catalyzes the sulfonation of dehydroepiandrosterone, and framework for EDCs, an adverse outcome at the organism level might
their inhibition of SULT2A1 expression might lead to more dehydro­ result from various separate pathways, and understanding toxicity
epiandrosterone participating in the production of E2. However, they pathways of EDCs is better for their risk assessment on human health.
induced different effects on 4 genes (CYP11A1, 3β-HSD2, CYP17 and Thereby, in present study, the action on ERα, the activation on GPER and
17β-HSD4), which might cause their different action on other 3 hor­ E2 synthesis of TPhP and TDCIPP was investigated to form a systematic
mones levels. CYP11A1 catalyzes the conversion of cholesterol to and comprehensive toxicity mechanism framework of their
pregnenolone, 3β-HSD2 acts a crucial role in the synthesis of various estrogen-disrupting activities. As shown in Fig. 6, it was found that TPhP
steroids, and CYP17 catalyzes pregnenolone/progesterone to andro­ acted as a pure estrogen-like substance, it showed agonistic potential on
stenedione. TPhP increasing progesterone levels could result from the ERα to activate ERE transcription in MVLN cells, and induced ERα
up-regulation of CYP11A1 and 3β-HSD2 genes expression, and TPhP interaction with SRC1 and SCR3. It also formed a pi-sigma bond with
promoting CYP17 gene expression might cause more androstenedione GPER and activated GPER-mediated cAMP pathways. Additionally,
synthesis. In contrast, exposure to TDCIPP, since the expression of TPhP disrupted steroidogenesis which involved increased progesterone

Fig. 6. The proposed toxicity mechanisms framework of estrogen disrupting effects for TPhP and TDCIPP. Hormones including progesterone, estrone, testosterone
and 17β-estradiol which are marked with boxes and all genes in the figure were tested in this study. Red indicated a significant increase of levels, black indicated no
significant changes of levels and blue indicated a significant decrease of levels.

7
X. Ji et al. Ecotoxicology and Environmental Safety 229 (2022) 113069

levels, decreased testosterone and estrone levels, and alteration of androgen receptor (AR) with LOECs of 5 × 10-5 and 5 × 10-6 M for TPhP
various key genes, eventually resulting in up-regulation of E2 concen­ and TDCIPP, respectively, antagonism to thyroid hormone receptor (TR)
trations. In contrast, TDCIPP acted as a partial estrogen-like substance with LOECs of 1 × 10-5 M for TDCIPP, antagonism to glucocorticoid
leading to estrogen-disrupting activities. It showed antagonistic effects receptor (GR) with both LOECs of 1 × 10-6 M, antagonism to mineral­
on ERα in MVLN cells, but it formed a hydrogen bond with GPER to ocorticoid receptor (MR) with LOECs of 1 × 10-7 and 5 × 10-6 M for
activate GPER-cAMP pathways. Additionally, TDCIPP disrupted ste­ TPhP and TDCIPP, respectively, and antagonism to estrogen-related
roidogenesis which involved increased testosterone and estrone levels, receptor γ (ERRγ) with LOECs of 1 × 10-5 M for TPhP (Cao et al.,
and alteration of various key genes including 17β-HSD4, 17β-HSD1 and 2018a; Kojima et al., 2013; Zhang et al., 2016, 2017), indicating that the
CYP19, eventually resulting in up-regulation of E2 concentrations action of TPhP and TDCIPP toward various receptors might lead to
(Fig. 6). Obviously, TPhP induced the synergy of multiple pathways complex endocrine disrupting effects. Therefore, there is a need for
related to estrogenic effects including ERα and GPER agonistic response, further studies to assess their potential ecological and health risks
and promoting E2 synthesis, while TDCIPP showed more complicated effects.
mechanisms involving in its synergic effects between GPER activation In summary, we aimed to investigate the toxicity mechanisms con­
and promoting E2 synthesis, but antagonistic effects on ERα. The dif­ cerning estrogen disrupting effects of TPhP and TDCIPP. The results
ference in their toxicity mechanisms might be associated with their showed that TPhP acted as a pure estrogenic substance and TDCIPP
different substituent. In our previous studies, we found that in contrast acted as a partial estrogenic substance. Specifically, TPhP promoted
to TmCP and TpCP, ToCP with methyl group at the ortho position of the ERα-ERE transcription and recruited SRC1 and SRC3 which was found
aromatic ring showed higher activity in activating GPER (Ji et al., for the first time, while TDCIPP showed antagonistic effects on ERα.
2020a). Moreover, due to chlorination, TDCIPP induced more serious Both TPhP and TDCIPP activated GPER-mediated cAMP pathways.
toxicity than TnBP on DNA damage and apoptosis in C. fluminea (Yan Although TPhP and TDCIPP could dock into ERα and GPER, their sub­
et al., 2020). Of note, similar to BPA analogs, though they have similar stituent groups and combination mode might affect the receptor acti­
structure, some of them also exert different action in estrogen-disrupting vation. Additionally, TPhP and TDCIPP up-regulated E2 concentrations
activities, specifically, BPA, BPF, BFS and TCBPA showed agonistic ef­ and E2/T ratio, but they involved different alteration of progesterone,
fects on ERα in MVLN cells (Molina-Molina et al., 2013), while 9,9-bis testosterone and estrone levels, and various key genes expression. The
(4-hydroxyphenyl)-fluorene (BHPF) acted as an ERα antagonist due to difference in their mechanisms of their estrogen disrupting effects might
its inhibition on the coactivator recruitment (Cao et al., 2019). More­ be due to their different substituents. Our data proposed a framework of
over, BFAF and BFB displayed higher estrogenic response than BPA via toxicity mechanisms of estrogen disrupting effects for TPhP and TDCIPP,
GPER with different binding affinity (Cao et al., 2017). Furthermore, and provided valuable insight into future construction of AOP frame­
BPA and BPS inhibited hormone production in H295R cells by work of EDCs.
down-regulating steroidogenic genes, while BPF led to increased pro­
gesterone and E2 (Feng et al., 2016). In addition, our data provided for CRediT authorship contribution statement
the first time a more complete framework of toxicity mechanisms of
estrogen disrupting effects for TPhP and TDCIPP, and it still requires Xiaoya Ji: Conceptualization, Methodology, Validation, Formal
further study on their synergic effects involving various pathways. analysis, Investigation, Writing – original draft. Na Li: Methodology,
Currently, the molecular initiating event in the AOP framework for risk Formal analysis, Writing – review & editing, Funding acquisition. Mei
assessment of EDCs mainly focused on their activation on ERα, and their Ma: Visualization, Writing – review & editing, Project administration,
effects on GPER and E2 synthesis are needed to be considered in the Supervision, Funding acquisition. Xinyan Li: Investigation. Kongrui
future. Zhu: Validation. Kaifeng Rao: Visualization. Zijian Wang: Project
TPhP and TDCIPP have high frequency detection in aquatic envi­ administration, Supervision. Jingfeng Wang: Investigation. Yanjun
ronmental media, and their concentrations were in the levels of ng/L. Of Fang: Funding acquisition.
note, TPhP at the environmentally relevant concentrations of ng/L have
been demonstrated to cause increased plasma E2 and decreased testos­ Declaration of Competing Interest
terone, resulting in intersex in male medaka (Li et al., 2018), while
20 μg/L TDCIPP could increase plasma E2 in female zebrafish (J. Wang The authors declare that they have no known competing financial
et al., 2015; Q.W. Wang et al., 2015; Qiangwei Wang et al., 2015). interests or personal relationships that could have appeared to influence
Moreover, TPhP and TDCIPP have also been found in various biota the work reported in this paper.
samples, such as glaucous gull at levels of 1.2 and 22.52 ng/g wet weight
(ww), respectively, arctic fox at levels of 1.3 and 89 ng/g ww, respec­ Acknowledgments
tively, polar bear at levels of 6.5 and 54 ng/g ww, respectively, and
freshwater shrimp at levels of 5.4 and 2.0 ng/g ww, respectively (Hal­ We thank Dr. Méndez-Luna for kindly providing us the GPER ho­
langer et al., 2015; Strobel et al., 2018; Zhao et al., 2018), implying that mology modeling. This work was financially supported by the National
their potential to cause adverse effects on wildlife including aquatic and Key Research and Development Program of China (2019YFC1804604),
terrestrial organisms. Additionally, TPhP and TDCIPP were also detec­ the National Natural Science Foundation of China (41977208,
ted in human samples. In human milk samples, their maximal concen­ 52030003), Excellent Innovation Project of Research Center for Eco-
trations were 140 and 162 ng/g lipid wt, respectively (Kim et al., 2014), Environmental Sciences, CAS (RCEES-EEI-2019), and the special fund
which suggested that susceptible infants were at risk for exposure to from Key Laboratory of Drinking Water Science and Technology,
TPhP and TDCIPP, and might be easier to be affected by their Research Center for Eco-Environmental Sciences, Chinese Academy of
estrogen-disrupting potential. Note that maximal levels of 1.21 and Sciences (19204KLDWST), and Special Fund from key topics
3.41 ng/mL for TPhP and TDCIPP, respectively, were found in 257 (AWS18J004).
human blood samples from Shenzhen, China (Zhao et al., 2016), obvi­
ously, they are much lower than lowest-observed effect concentrations Appendix A. Supporting information
(LOECs) for TPhP and TDCIPP in present study of 326.3 (1 × 10-6 M)
and 430.9 ng/mL (1 × 10-6 M), respectively. However, our data sug­ Supplementary data associated with this article can be found in the
gested that TPhP and TDCIPP had potential to activate multiple path­ online version at doi:10.1016/j.ecoenv.2021.113069.
ways of estrogen-disrupting effects, and they also have been reported to
interfere with various steroid hormone receptors, such as antagonism to

8
X. Ji et al. Ecotoxicology and Environmental Safety 229 (2022) 113069

References Li, Y., Chen, R., He, J., Ma, H., Zhao, F., Tao, S., et al., 2019. Triphenyl phosphate at
environmental levels retarded ovary development and reduced egg production in
japanese medaka (Oryzias latipes). Environ. Sci. Technol. 53, 14709–14715.
Bajard, L., Negi, C.K., Mustieles, V., Melymuk, L., Jomini, S., Barthelemy-Berneron, J.,
Liu, C., Du, Y., Zhou, B., 2007. Evaluation of estrogenic activities and mechanism of
et al., 2021. Endocrine disrupting potential of replacement flame retardants-review
action of perfluorinated chemicals determined by vitellogenin induction in primary
of current knowledge for nuclear receptors associated with reproductive outcomes.
cultured tilapia hepatocytes. Aquat. Toxicol. 85, 267–277.
Environ. Int. 153, 106550.
Liu, J., Yu, L., Castro, L., Yan, Y., Sifre, M.I., Bortner, C.D., et al., 2019. A nongenomic
Brandsma, S.H., de Boer, J., van Velzen, M.J., Leonards, P.E., 2014. Organophosphorus
mechanism for “metalloestrogenic” effects of cadmium in human uterine leiomyoma
flame retardants (pfrs) and plasticizers in house and car dust and the influence of
cells through g protein-coupled estrogen receptor. Arch. Toxicol. 93 (10),
electronic equipment. Chemosphere 116, 3–9.
2773–2785.
Cao, H.M., Wang, L., Cao, M.X., Ye, T., Sun, Y.Z., 2019. Computational insights on
Liu, X., Ji, K., Choi, K., 2012. Endocrine disruption potentials of organophosphate flame
agonist and antagonist mechanisms of estrogen receptor alpha induced by bisphenol
retardants and related mechanisms in h295r and mvln cell lines and in zebrafish.
a analogues. Environ. Pollut. 248, 536–545.
Aquat. Toxicol. 114, 173–181.
Cao, L.Y., Ren, X.M., Li, C.H., Zhang, J., Qin, W.P., Yang, Y., et al., 2017. Bisphenol af
Liu, X., Ji, K., Jo, A., Moon, H.B., Choi, K., 2013. Effects of tdcpp or tpp on gene
and bisphenol b exert higher estrogenic effects than bisphenol a via g protein-
transcriptions and hormones of hpg axis, and their consequences on reproduction in
coupled estrogen receptor pathway. Environ. Sci. Technol. 51, 11423–11430.
adult zebrafish (Danio rerio). Aquat. Toxicol. 134, 104–111.
Cao, L.Y., Ren, X.M., Li, C.H., Guo, L.H., 2018a. Organophosphate esters bind to and
Liu, X.S., Jung, D., Jo, A., Ji, K., Moon, H.B., Choi, K., 2016. Long-term exposure to
inhibit estrogen-related receptor gamma in cells. Environ. Sci. Tech. Lett. 5, 68–73.
triphenylphosphate alters hormone balance and hpg, hpi, and hpt gene expression in
Cao, L.Y., Ren, X.M., Yang, Y., Wan, B., Guo, L.H., Chen, D., et al., 2018b. Hydroxylated
zebrafish (Danio rerio). Environ. Toxicol. Chem. 35, 2288–2296.
polybrominated biphenyl ethers exert estrogenic effects via non-genomic g protein-
Ma, Y., Han, J., Guo, Y., Lam, P.K., Wu, R.S., Giesy, J.P., et al., 2012. Disruption of
coupled estrogen receptor mediated pathways. Environ. Health Perspect. 126 (5),
endocrine function in in vitro h295r cell-based and in in vivo assay in zebrafish by
057005.
2,4-dichlorophenol. Aquat. Toxicol. 106–107, 173–181.
Cooper, E.M., Kroeger, G., Davis, K., Clark, C.R., Ferguson, P.L., Stapleton, H.M., 2016.
Mendez-Luna, D., Bello, M., Correa-Basurto, J., 2016. Understanding the molecular basis
Results from screening polyurethane foam based consumer products for flame
of agonist/antagonist mechanism of gper1/gpr30 through structural and energetic
retardant chemicals: assessing impacts on the change in the furniture flammability
analyses. J. Steroid Biochem. 158, 104–116.
standards. Environ. Sci. Technol. 50, 10653–10660.
Mitchell, C.A., Dasgupta, Subham, Zhang, Sharon, Stapleton, Heather M., Volz, D.C.,
Deng, L.J., Cheng, C., Wu, J., Wang, C.H., Zhou, H.B., Huang, J., 2017.
2018. Disruption of nuclear receptor signaling alters triphenyl phosphate-induced
Oxabicycloheptene sulfonate protects against beta-amyloid-induced toxicity by
cardiotoxicity in zebrafish embryos. Toxicol. Sci. 163, 307–318.
activation of pi3k/akt and erk signaling pathways via gper1 in c6 cells. Neurochem
Molina-Molina, J.M., Amaya, E., Grimaldi, M., Saenz, J.M., Real, M., Fernandez, M.F.,
Res. 42, 2246–2256.
et al., 2013. In vitro study on the agonistic and antagonistic activities of bisphenol-s
Ding, J., Xu, Z., Huang, W., Feng, L., Yang, F., 2016. Organophosphate ester flame
and other bisphenol-a congeners and derivatives via nuclear receptors. Toxicol.
retardants and plasticizers in human placenta in eastern china. Sci. Total Environ.
Appl. Pharm. 272, 127–136.
554–555, 211–217.
Pons, M., Gagne, D., Nicolas, J.C., Mehtali, M., 1990. A new cellular-model of response to
Farhat, Amani, Buick, K., Williams, Andrew, Yauk, C.L., O’Brien, M.J., Crump, Doug,
estrogens – a bioluminescent test to characterize (anti)estrogen molecules.
2014. Tris(1,3-dichloro-2-propyl) phosphate perturbs the expression of genes
Biotechniques 9, 450–459.
involved in immune response and lipid and steroid metabolism in chicken embryos.
Prossnitz, E.R., Barton, M., 2011. The g-protein-coupled estrogen receptor gper in health
Toxicol. Appl. Pharm. 275, 104–112.
and disease. Nat. Rev. Endocrinol. 7, 715–726.
Feng, Y.X., Jiao, Z.H., Shi, J.C., Li, M., Guo, Q.Z., Shao, B., 2016. Effects of bisphenol
Reemtsma, T., Quintana, J.B., Rodil, R., Garcı´a-López, M., Rodrı´guez, I., 2008.
analogues on steroidogenic gene expression and hormone synthesis in h295r cells.
Organophosphorus flame retardants and plasticizers in water and air i. Occurrence
Chemosphere 147, 9–19.
and fate. TrAC Trends Anal. Chem. 27, 727–737.
Fu, J., Han, J., Zhou, B., Gong, Z., Santos, E.M., Huo, X., et al., 2013. Toxicogenomic
Rosenmai, A.K., Winge, S.B., Moller, M., Lundqvist, J., Wedebye, E.B., Nikolov, N.G.,
responses of zebrafish embryos/larvae to tris(1,3-dichloro-2-propyl) phosphate
et al., 2021. Organophosphate ester flame retardants have antiandrogenic potential
(tdcpp) reveal possible molecular mechanisms of developmental toxicity. Environ.
and affect other endocrine related endpoints in vitro and in silico. Chemosphere 263,
Sci. Technol. 47, 10574–10582.
127703.
Hallanger, I.G., Sagerup, K., Evenset, A., Kovacs, K.M., Leonards, P., Fuglei, E., et al.,
Sarmiento, V., Ramirez-Sanchez, I., Moreno-Ulloa, A., Romero-Perez, D., Chavez, D.,
2015. Organophosphorous flame retardants in biota from svalbard, Norway. Mar.
Ortiz, M., et al., 2018. Synthesis of novel (-)-epicatechin derivatives as potential
Pollut. Bull. 101, 442–447.
endothelial gper agonists: evaluation of biological effects. Bioorg. Med. Chem. Lett.
He, Y., Murphy, M.B., Yu, R.M., Lam, M.H., Hecker, M., Giesy, J.P., et al., 2008. Effects of
28, 658–663.
20 pbde metabolites on steroidogenesis in the h295r cell line. Toxicol. Lett. 176,
Sohn, J., Kim, S., Koschorreck, J., Kho, Y., Choi, K., 2016. Alteration of sex hormone
230–238.
levels and steroidogenic pathway by several low molecular weight phthalates and
Hoffman, K., Butt, C.M., Chen, A., Limkakeng Jr., A.T., Stapleton, H.M., 2015. High
their metabolites in male zebrafish (Danio rerio) and/or human adrenal cell (h295r)
exposure to organophosphate flame retardants in infants: associations with baby
line. J. Hazard. Mater. 320, 45–54.
products. Environ. Sci. Technol. 49, 14554–14559.
Stapleton, H.M., Klosterhaus, S., Keller, A., Ferguson, P.L., van Bergen, S., Cooper, E.,
Hong, X., Chen, R., Hou, R., Yuan, L., Zha, J., 2018. Triphenyl phosphate (tphp)-induced
et al., 2011. Identification of flame retardants in polyurethane foam collected from
neurotoxicity in adult male chinese rare minnows (Gobiocypris rarus). Environ. Sci.
baby products. Environ. Sci. Technol. 45, 5323–5331.
Technol. 52, 11895–11903.
Strobel, A., Willmore, W.G., Sonne, C., Dietz, R., Letcher, R.J., 2018. Organophosphate
Hu, W., Gao, F., Zhang, H., Hiromori, Y., Arakawa, S., Nagase, H., et al., 2017. Activation
esters in east greenland polar bears and ringed seals: adipose tissue concentrations
of peroxisome proliferator-activated receptor gamma and disruption of progesterone
and in vitro depletion and metabolite formation. Chemosphere 196, 240–250.
synthesis of 2-ethylhexyl diphenyl phosphate in human placental choriocarcinoma
Tachachartvanich, P., Sangsuwan, R., Ruiz, H.S., Sanchez, S.S., Durkin, K.A., Zhang, L.,
cells: comparison with triphenyl phosphate. Environ. Sci. Technol. 51, 4061–4068.
et al., 2018. Assessment of the endocrine-disrupting effects of trichloroethylene and
Ji, X.Y., Li, N., Ma, M., Rao, K., Yang, R., Wang, Z., 2020a. Tricresyl phosphate isomers
its metabolites using in vitro and in silico approaches. Environ. Sci. Technol. 52,
exert estrogenic effects via g protein-coupled estrogen receptor-mediated pathways.
1542–1550.
Environ. Pollut. 264, 114747.
van der Veen, I., de Boer, J., 2012. Phosphorus flame retardants: properties, production,
Ji, X.Y., Li, N., Ma, M., Rao, K.F., Wang, Z.J., 2020b. In vitro estrogen-disrupting effects
environmental occurrence, toxicity and analysis. Chemosphere 88, 1119–1153.
of organophosphate flame retardants. Sci. Total Environ. 727, 138484.
Vinas, R., Watson, C.S., 2013. Bisphenol s disrupts estradiol-induced nongenomic
Kang, J.S., Choi, J.S., Park, J.W., 2016. Transcriptional changes in steroidogenesis by
signaling in a rat pituitary cell line: effects on cell functions. Environ. Health
perfluoroalkyl acids (pfoa and pfos) regulate the synthesis of sex hormones in h295r
Perspect. 121, 352–358.
cells. Chemosphere 155, 436–443.
Wan, W., Zhang, S., Huang, H., Wu, T., 2016. Occurrence and distribution of
Kim, J.W., Isobe, T., Muto, M., Tue, N.M., Katsura, K., Malarvannan, G., et al., 2014.
organophosphorus esters in soils and wheat plants in a plastic waste treatment area
Organophosphorus flame retardants (pfrs) in human breast milk from several asian
in china. Environ. Pollut. 214, 349–353.
countries. Chemosphere 116, 91–97.
Wang, J., Cao, X.F., Sun, J.H., Huang, Y., Tang, X.Y., 2015. Disruption of endocrine
Kim, U.J., Kannan, K., 2018. Occurrence and distribution of organophosphate flame
function in h295r cell in vitro and in zebrafish in vivo by naphthenic acids.
retardants/plasticizers in surface waters, tap water, and rainwater: implications for
J. Hazard. Mater. 299, 1–9.
human exposure. Environ. Sci. Technol. 52, 5625–5633.
Wang, Q.W., Lam, J.C.W., Han, J., Wang, X.F., Guo, Y.Y., Lam, P.K.S., et al., 2015.
Kojima, H., Takeuchi, S., Itoh, T., Iida, M., Kobayashi, S., Yoshida, T., 2013. In vitro
Developmental exposure to the organophosphorus flame retardant tris(1,3-dichloro-
endocrine disruption potential of organophosphate flame retardants via human
2-propyl) phosphate: estrogenic activity, endocrine disruption and reproductive
nuclear receptors. Toxicology 314, 76–83.
effects on zebrafish. Aquat. Toxicol. 160, 163–171.
Lei, B.L., Peng, W., Xu, G., Wu, M.H., Wen, Y., Xu, J., et al., 2017. Activation of g protein-
Wang, Qiangwei, Lam, James Chung-Wah, Man, Yin-Chung, Lai, NelsonLok-Shun,
coupled receptor 30 by thiodiphenol promotes proliferation of estrogen receptor
Kwok, Karen Ying, Guo, Yong yong, 2015. Bioconcentration, metabolism and
alpha-positive breast cancer cells. Chemosphere 169, 204–211.
neurotoxicity of the organophorous flame retardant 1,3-dichloro 2-propyl phosphate
Li, N., Ho, W., Sun Wu, R.S., Ying, G.G., Wang, Z., Jones, K., et al., 2019.
(tdcpp) to zebrafish. Aquat. Toxicol. 158, 108–115.
Organophosphate flame retardants and bisphenol a in children’s urine in hong kong:
Wei, G.L., Li, D.Q., Zhuo, M.N., Liao, Y.S., Xie, Z.Y., Guo, T.L., et al., 2015.
has the burden been underestimated? Ecotoxicol. Environ. Saf. 183, 109502.
Organophosphorus flame retardants and plasticizers: sources, occurrence, toxicity
Li, Y., Wang, C., Zhao, F., Zhang, S., Chen, R., Hu, J., 2018. Environmentally relevant
and human exposure. Environ. Pollut. 196, 29–46.
concentrations of the organophosphorus flame retardant triphenyl phosphate
impaired testicular development and reproductive behaviors in japanese medaka
(Oryzias latipes). Environ. Sci. Tech. Lett. 5, 649–654.

9
X. Ji et al. Ecotoxicology and Environmental Safety 229 (2022) 113069

Wrobel, A., Gregoraszczuk, E.L., 2013. Effects of single and repeated in vitro exposure of Zhang, Q., Ji, C.Y., Yin, X.H., Yan, L., Lu, M.Y., Zhao, M.R., 2016. Thyroid hormone-
three forms of parabens, methyl-, butyl- and propylparabens on the proliferation and disrupting activity and ecological risk assessment of phosphorus-containing flame
estradiol secretion in mcf-7 and mcf-10a cells. Pharmacol. Rep. 65, 484–493. retardants by in vitro, in vivo and in silico approaches. Environ. Pollut. 210, 27–33.
Wu, M., Yu, G., Cao, Z.G., Wu, D.K., Liu, K., Deng, S.B., et al., 2016. Characterization and Zhang, Quan, Wang, Jinghua, Zhu, Jianqiang, Liu, Jing, Zhao, M., 2017. Potential
human exposure assessment of organophosphate flame retardants in indoor dust glucocorticoid and mineralocorticoid effects of nine organophosphate flame
from several microenvironments of beijing, china. Chemosphere 150, 465–471. retardants. Environ. Sci. Technol. 51, 5803–5810.
Ya, M., Yu, N., Zhang, Y., Su, H., Tang, S., Su, G., 2019. Biomonitoring of Zhao, F., Wan, Y., Zhao, H., Hu, W., Mu, D., Webster, T.F., et al., 2016. Levels of blood
organophosphate triesters and diesters in human blood in jiangsu province, eastern organophosphorus flame retardants and association with changes in human
china: occurrences, associations, and suspect screening of novel metabolites. sphingolipid homeostasis. Environ. Sci. Technol. 50, 8896–8903.
Environ. Int. 131, 105056. Zhao, H., Zhao, F., Liu, J., Zhang, S., Mu, D., An, L., et al., 2018. Trophic transfer of
Yan, S.H., Wang, Q., Yang, L.H., Zha, J.M., 2020. Comparison of the toxicity effects of tris organophosphorus flame retardants in a lake food web. Environ. Pollut. 242,
(1,3-dichloro-2-propyl)phosphate (tdcipp) with tributyl phosphate (tnbp) reveals the 1887–1893.
mechanism of the apoptosis pathway in asian freshwater clams (Corbicula fluminea). Zheng, G.M., Schreder, E., Dempsey, J.C., Uding, N., Chu, V., Andres, G., et al., 2021.
Environ. Sci. Technol. 54, 6850–6858. Organophosphate esters and their metabolites in breast milk from the united states:
Zhang, Q., Lu, M.Y., Dong, X.W., Wang, C., Zhang, C.L., Liu, W.P., et al., 2014. Potential breastfeeding is an important exposure pathway for infants. Environ. Sci. Tech. Lett.
estrogenic effects of phosphorus-containing flame retardants. Environ. Sci. Technol. 8, 224–230.
48, 6995–7001. Zhu, Q., Liu, L., Zhou, X., Ma, M., 2019. In silico study of molecular mechanisms of
action: estrogenic disruptors among phthalate esters. Environ. Pollut. 255, 113193.

10

You might also like