Activation of Peroxisome Proliferator-Activated Receptor Gamma and

Mar 10, 2017 - ... Diphenyl Phosphate in Human Placental Choriocarcinoma Cells: Comparison with Triphenyl Phosphate ... *Phone/Fax: 86-10-62765520...
0 downloads 0 Views 2MB Size
Subscriber access provided by University of Newcastle, Australia

Article

Activation of Peroxisome Proliferator-activated Receptor Gamma and Disruption of Progesterone Synthesis of 2Ethylhexyl Diphenyl Phosphate in Human Placental Choriocarcinoma Cells: Comparison with Triphenyl Phosphate Wenxin Hu, Fumei Gao, Hong Zhang, Youhei Hiromori, Shuhei Arakawa, Hisamitsu Nagase, Tsuyoshi NAKANISHI, and Jianying Hu Environ. Sci. Technol., Just Accepted Manuscript • DOI: 10.1021/acs.est.7b00872 • Publication Date (Web): 10 Mar 2017 Downloaded from http://pubs.acs.org on March 12, 2017

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

Environmental Science & Technology is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 27

Environmental Science & Technology

1

Activation of Peroxisome Proliferator-activated Receptor Gamma and Disruption of

2

Progesterone Synthesis of 2-Ethylhexyl Diphenyl Phosphate in Human Placental

3

Choriocarcinoma Cells: Comparison with Triphenyl Phosphate

4

Wenxin Hu1, Fumei Gao1, Hong Zhang1, Youhei Hiromori2,3, Shuhei Arakawa2, Hisamitsu

5

Nagase2, Tsuyoshi Nakanishi2, and Jianying Hu1*

6 7

1

8

Peking University, Beijing 100871, China

9

2

MOE Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences,

Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical

10

University, 1-25-4 Daigaku-nishi, Gifu, Gifu, 501-1196, Japan

11

3

12

Minamitamagaki, Suzuka, Mie 513-8670, Japan

Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science 3500-3,

13 14 15 16

*Address for correspondence

17

Dr. Jianying Hu

18

College of Urban and Environmental Sciences

19

Peking University

20

Beijing 100871, China.

21

TEL & FAX: 86-10-62765520;

22 23

Email: [email protected]

1

ACS Paragon Plus Environment

Environmental Science & Technology

24 25

Abstract

26

2-Ethylhexyl diphenyl phosphate (EHDPP), an organophosphate flame retardants

27

(OPFRs), is frequently detected in human blood. In this study, sensitive dual-luciferase

28

reporter gene assay and molecular docking were used to investigate the activation of EHDPP

29

to human peroxisome proliferator-activated receptor gamma (PPARG). Results show that

30

EHDPP exhibited stronger PPARG activation (EC20: 2.04 µM) than triphenyl phosphate

31

(TPhP) (EC20: 2.78 µM). EHDPP upregulated the gene expression of 3β-hydroxysteroid

32

dehydrogenase type 1 (3β-HSD1) in human placental choriocarcinoma cells in a

33

dose-dependent manner, and the lowest observable effective concentration was 10 µM, lower

34

than that of TPhP (20 µM). EHDPP significantly altered progesterone secretion at a lower

35

concentration (10 µM) than that of TPhP (20 µM), and both EHDPP and TPhP significantly

36

promoted human chorionic gonadotropin (hCG) production at 20 µM. Furthermore,

37

inactivation of PPARG by either pharmacological inhibitor (GW9662) or small interfering

38

RNA (siRNA) abolished the change in progesterone secretion and gene expression in the cells

39

exposed to EHDPP, suggesting that the PPARG signaling pathway plays a role in the

40

upregulation of progesterone by the two OPFRs. This is the first report to show that OPFRs

41

can alter the biosynthesis of progesterone in the placenta, which could affect female

42

reproduction and fetal development.

43

Keywords: Organophosphate flame retardants; PPARG; Human placenta; Human chorionic

44

gonadotropin.

45

2

ACS Paragon Plus Environment

Page 2 of 27

Page 3 of 27

46

Environmental Science & Technology

Introduction

47

There is growing concern about the possible health threat posed by anthropogenic

48

compounds. Increasing evidence has shown that substances found in the environment, food,

49

and consumer products can interfere with hormone biosynthesis, metabolism, and activity,

50

resulting in deviation from normal homeostatic control or reproduction.1 The wide variety of

51

pollutants reported to disrupt endocrine function includes pesticides, polycyclic aromatic

52

hydrocarbons, phthalate plasticizers, polychlorinated biphenyls, dioxins, furans, alkylphenols,

53

and synthetic steroids.2

54

Of all environmental chemicals showing reproductive toxicity, organophosphate flame

55

retardants (OPFRs) are of particular concern. OPFRs are used as replacements for brominated

56

flame retardants (BFRs) such as polybrominated diphenyl ethers (PBDEs), thus their use has

57

increased significantly in recent years, with annual global production currently reaching

58

approximately 2,000,000 tons.3 OPFRs are widely used as plasticizers in various consumer

59

products, building materials, and baby products,4-6 and are therefore ubiquitous in various

60

environmental media such as house dust, sediment, sludge, river water, biota, and even in

61

human blood and placenta.7-13 There is increasing evidence that OPRFs exposure can disrupt

62

hormonal metabolism and biosynthesis in animals and humans. Tris(1,3-dichloro-2-propyl)

63

phosphate (TDCIPP) and triphenyl phosphate (TPhP) have been shown to reduce fecundity

64

by significantly increasing plasma estradiol levels and inhibiting androgen levels in

65

zebrafish.14 A similar phenomenon has also been observed in stably transfected human breast

66

cancer cell line MVLN and human adrenocortical carcinoma cell line (H295R).15 One recent

67

epidemiological study showed that OPFRs in house dust were associated with serum free 3

ACS Paragon Plus Environment

Environmental Science & Technology

68

thyroxin (T4), prolactin, and decreased semen quality in men.16 In addition to alteration of

69

hormonal metabolism and biosynthesis, OPFRs can also interact with various nuclear

70

receptors (NRs) such as constitutive androstane receptor (CAR), pregnane X receptor (PXR),

71

estrogen receptor (ER), androgen receptor (AR), glucocorticoid receptor (GR), and

72

peroxisome proliferator-activated receptor gamma (PPARG).17,18 Of these NRs, PPARG is

73

abundantly expressed in human placenta and serves as an essential regulator of endocrine

74

functions that play a vital role in maintaining pregnancy.19-22 Thus, it would be interesting to

75

determine whether OPFRs can disrupt endocrine functions in human placenta. Among

76

various OPFRs, only TPhP and tributyl phosphate (TBP) have been identified as agonists of

77

PPARG,17 for a broad range of OPFRs with diverse structures the activity to PPARG have not

78

been evaluated. Particularly, 2-ethylhexyl diphenyl phosphate (EHDPP), tris-(2-chloroethyl)

79

phosphate (TCEP), and tris-2-chloroisopropyl phosphate (TCPP) have been detected in the

80

blood of the Chinese population, with median concentrations of EHDPP (1.22 ng/ml) and

81

TCPP (0.71 ng/ml) found to be higher than that of TPhP (0.43 ng/ml).13 To date, however,

82

their activation to PPARG remain to be elucidated.

83

In this study, TCEP, TCPP and EHDPP PPARG activation were determined using

84

dual-luciferase reporter assay. The TBP and TPhP PPARG activation were also determined to

85

enable a direct comparison of the unknown PPARG activation by TCEP, TCPP and EHDPP

86

based on the same assay. We also investigated their effects on the synthesis of progesterone

87

and human chorionic gonadotropin (hCG) in human placental choriocarcinoma cell line

88

JEG-3, which retains characteristics of normal pregnancy trophoblast cells. Finally, the

89

potential role in promotion of progesterone production was also investigated by co-exposure 4

ACS Paragon Plus Environment

Page 4 of 27

Page 5 of 27

Environmental Science & Technology

90

with a pharmacological inhibitor of PPARG (GW9662) and small interfering RNA (siRNA)

91

targeting PPARG in JEG-3 cells.

92

Materials and Methods

93

Chemicals and Reagents. The standards of TCEP, TCPP, TBP, EHDPP, and TPhP were

94

purchased from Tokyo Chemical Industry Co (Tokyo, Japan). Rosiglitazone, GW9662,

95

3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT), and dimethyl sulfoxide

96

(DMSO) were purchased from Sigma-Aldrich (St. Louis, MO, USA). TRIzol was obtained

97

from Invitrogen (Carlsbad, CA, USA).

98

Cell Cultures and Treatments. The human placental choriocarcinoma cell line JEG-3

99

was cultured in DMEM-F12 medium (Hyclone, Logan, UT, USA) supplemented with 10%

100

fetal bovine serum (FBS, Gibco, Grand Island, NY, USA), and incubated at 37°C in a

101

humidified, 5% CO2 atmosphere. The JEG-3 cells were plated in 24-well plates in

102

DMEM-F12 medium and allowed to become confluent (2–3 d). To determine the effects of

103

EHDPP, TPhP, TCEP, TBP, and TCPP on synthesis-related gene expression and production of

104

progesterone and hCG, the JEG-3 cells were seeded, pre-cultured for 24 h, and then treated

105

with various concentrations of EHDPP, TPhP, TCEP, TBP, or TCPP in DMSO or with

106

vehicle-control (0.1% DMSO).

107

Reporter Assay. The 293T cells were seeded in 24-well plates in 500 µL of DMEM-F12

108

containing 10% FBS. After incubation for 24 h, the cells showed 70–80% confluence and

109

were then transiently transfected with pBIND-PPARG-LBD, GAL4-pGL4-luc (provided by

110

Shuyi Si, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and

111

Peking Union Medical College) and pGL4.74 (Promega, Madison, WI, USA) or GAL4-RXR, 5

ACS Paragon Plus Environment

Environmental Science & Technology

112

pG5-luc and pGL4.74 using Lipofectamine 2000 Transfection Reagent (Invitrogen, Carlsbad,

113

CA, USA) according to the manufacturer’s instructions.23 Following 24 h of incubation, the

114

medium was replaced and the cultures were treated with DMSO (0.1%), EHDPP, TPhP, TCEP,

115

TBP, or TCPP (0.05–25 µM) for 24 h. The extracts were prepared and assayed for firefly

116

luciferase (LUC) activity using the Dual-Luciferase Reporter Assay System (Promega,

117

Madison, WI, USA) and a LB 941 TriStar Multimode Microplate Reader (Berthold

118

Technologies, Bad Wildbad, Germany) according to the manufacturer’s instructions. In brief,

119

the cells were collected with phosphate buffered saline (PBS), and then passive lysis buffer

120

was added. The supernatant was collected to test the firefly LUC and Renilla LUC activities.

121

Results were expressed as the average relative firefly LUC activity of at least triplicated

122

samples.

123

Molecular Docking. Molecular simulation software Scigress (Ultra Version 3.0.0,

124

Fujitsu, USA) was used to dock flexible ligands into a rigid protein active site. In brief, the

125

PPARG ligand binding domain (PDB ID code 2PRG) was obtained from the Protein Data

126

Bank (PDB, http://www.rcsb.org/). Explicit hydrogen atoms were created and heteroatoms

127

such as water, ions, and cofactors were removed. Docking calculations were evaluated using

128

a genetic algorithm with a 15 × 15 × 15 Å grid box with 0.3 Å grid spacing, which contained

129

the active site for the original ligand. The procedure was set to run for 20000 generations with

130

an initial population size of 100, elitism of 8, crossover rate of 0.8, mutation rate of 0.5, and

131

convergence of 1.0. Other parameters were set at their default values. The Potential of Mean

132

Force (PMF) scores, a knowledge-based approach that extracts pairwise atomic potentials

133

from the structure information on known protein−ligand complexes contained in the Protein 6

ACS Paragon Plus Environment

Page 6 of 27

Page 7 of 27

Environmental Science & Technology

134

Data Bank, was determined and used to score the binding activity of a chemical to the active

135

site of human PPARG. The PMF of rosiglitazone, the native ligand in the complex, was

136

−91.15 kcal/mol when docked into the binding site. The root-mean-square error between the

137

predicted conformation and the actual conformation from the crystal structure was 1.78 Å,

138

smaller than the X-ray crystallography resolution (2.20 Å).24 Thus the parameter set used for

139

the docking simulation was considered to be reasonable.

140

Plasmid construction. pBIND-PPARG-LBD mutant construct, carrying a Ser342 to Ala

141

mutation, was generated by site-directed mutagenesis of pBIND-PPARG-LBD. The

142

sequences

143

5’-GGTTCTCATAGCAGAGGGCCAAGGCTTCATGAC-3’

144

CTTGGCCCTCTGCTATGAGAACCCCATCTTTAT-3’.

of

the

mutagenic

primers

are

and

5’-

145

Cell Viability Assays. Confluent JEG-3 cultures were treated with vehicle-control

146

(DMSO, 0.1%), EHDPP, TPhP, TCEP, TBP, or TCPP (5–40 µM) for 48 h. Cytotoxicity was

147

measured using MTT assay. Briefly, JEG-3 cells were seeded in 96-cell plates for 24 h, and

148

then treated with different concentrations of OPFRs for 48 h. At the end of treatment, 20 µL

149

of MTT was added and the solution was incubated for an additional 4 h. The formazan

150

crystals were then dissolved in 200 µL of DMSO. After shaking the plate for 10 min, cell

151

viability was assessed by measuring the absorbance at 490 nm using a spectrophotometer

152

(Bio-Rad Model 550; Bio-Rad Laboratories, Inc., Hercules, CA, USA). Cell viability was

153

also assessed by CellTiter-Glo Reagent (Promega, Madison, WI, USA) according to the

154

manufacturer’s instructions. In brief, cells were seeded, pre-cultured for 24 h, and then treated

155

with various concentrations of EHDPP, TPhP, TCEP, TBP, or TCPP in 0.1% DMSO or with 7

ACS Paragon Plus Environment

Environmental Science & Technology

156

vehicle-control (0.1% DMSO) for 48 h. After 100 µl of CellTiter-Glo Reagent was added to

157

each well, the plate and its contents were equilibrated at room temperature for approximately

158

30 min. The luminescence was determined using the LB 941 TriStar Multimode Microplate

159

Reader. Absorbance or luminescence in the experimental wells were normalized by dividing

160

the absorbance or luminescence in untreated cultures and reported as “Fold Change from

161

Medium”. Results were expressed as the average relative firefly LUC activity of at least

162

triplicated samples.

163

Determination of hCG and Progesterone Production. The JEG-3 cells were plated in

164

24-well plates. After 24 h of culture, the JEG-3 cells were treated with various concentrations

165

of EHDPP, TPhP, TCEP, TBP, or TCPP for a further 48 h. After incubation, the culture

166

supernatants were collected, and the concentrations of hCG and progesterone were

167

determined by radioimmunoassay (RIA) with a Progesterone Direct RIA Kit and hCG Direct

168

RIA Kit (ICN Biomedicals, Irvin, CA, USA) according to the manufacturer’s protocols.

169

Quantitative RT-PCR. Total RNA was extracted using TRIzol reagent (Invitrogen,

170

Carlsbad, CA, USA) according to the manufacturer’s protocols, and digested by DNaseI

171

(TaKaRa Biotechnology, Dalian, China) in case of genomic DNA contamination. The

172

concentration and quality of RNA was analyzed using a Nanovue Plus spectrophotometer

173

(GE Healthcare Life Science, Little Chalfont, Buckinghamshire, England). The purity of each

174

sample was between 1.8 and 2.0 (A260/A280 nm ratio). Total RNA was reverse transcribed

175

with MMLV reverse transcriptase in the presence of oligo (dT) and dNTP (TaKaRa

176

Biotechnology, Dalian, China). The complete reaction mixture was incubated at 37°C for 50

177

min, followed by 95°C for 5 min to stop the reaction. SYBR Green PCR Kits (TOYOBO, 8

ACS Paragon Plus Environment

Page 8 of 27

Page 9 of 27

Environmental Science & Technology

178

Osaka, Japan) were used for Q-PCR analysis. A real-time PCR profile was used: first,

179

enzyme activation at 95°C (10 min), 40 cycles at 95°C (30 s per cycle), and 60 s at 58–62°C,

180

depending on the target transcript, followed by post-run melt curve analysis (65°C).

181

Real-time fluorescence detection was carried out using the STEP ONE PLUS sequence

182

detection system (Applied Biosystems, Foster City, CA. USA). Relative gene expression was

183

evaluated by the 2-∆∆Ct method, as suggested by Applied Biosystems. The primers

184

sequences used are shown in Table S1.

185

RNA Interference Assay. Sequence-specific small interfering RNA (siRNA) targeting

186

PPARG (siPPARG) shown in Table S2 was purchased from Dharmacon/GE Healthcare

187

(Little Chalfont, Buckinghamshire, England). AllStars Negative Control siRNA purchased

188

from Qiagen (Valencia, CA) was used for the negative control siRNA. JEG-3 cells (3 × 105

189

cells/well) were seeded in 6-well plates and precultured at 37 °C for 24 h. The cells then were

190

transfected with siRNA duplexes (20 nM/well) by using Lipofectamine RNAiMAX

191

(Invitrogen/Thermo Fisher Scientific, Grand Island, NY) in accordance with the

192

manufacturer’s instructions.

193

Statistical Analysis. Results were presented as means ± standard errors of the mean

194

(SEM) and tested for statistical significance by analysis of variance (ANOVA) followed by

195

post hoc Dunnett’s test using SPSS 16.0 (SPSS Inc., Chicago, IL, USA). The number of

196

replicates is indicated in each figure legend. Dose response curves were performed with

197

Prism 5 (Graphpad Inc., La Jolla, CA). A p-value < 0.05 was considered statistically

198

significant.

199 9

ACS Paragon Plus Environment

Environmental Science & Technology

200

Results and Discussion

201

Human PPARG Activation. In this study, we determined the activation of TCEP, TCPP,

202

and EHDPP to PPARG using a sensitive dual-luciferase reporter assay, as shown in Figure

203

S1 (half effective concentration of rosiglitazone is 0.01µM). Although TPhP and TBP

204

PPARG activation have been reported previously,25 we show their PPARG activation in

205

Figure 1. The 20% effective concentrations (EC20) were 2.78 µM and 5.96 µM for TPhP and

206

TBP, respectively, slightly lower than those for TPhP (3.27 µM) and TBP (13.35 µM)

207

reported previously.25 Among the three OPFRs newly determined in this study, TCEP and

208

TCPP showed no significant PPARG activation under the testing concentrations, whereas

209

EHDPP showed significant PPARG activation compared with the vehicle-control, with a

210

EC20 value of 2.46 µM, showing stronger PPARG activation than that of TPhP. The EC20

211

value of EHDPP was similar to that of mono(2-ethylhexyl) phthalate (2.12 µM) and

212

3-hydroxide-2,2’,4,4’-tetrabromodiphendyl ether (3-OH-BDE47) (2.99 µM), but higher than

213

that of tetrabromobisphenol A (TBBPA) (0.41 µM) and 3,5,3′,5′-tetrachlorobisphenol A

214

(TCBPA) (0.47 µM).25 Although used as a replacement for PBDE, it should be noted that the

215

activation of EHDPP to PPARG was stronger than that of BDE47 (7.00 µM).25

216

Molecular docking was performed to understand the structural basis for the observed

217

activation of OPFRs to PPARG using Scigress. The PMF values of EHDPP, TPhP and TBP

218

were calculated to be −54.134, −44.288, and −32.534 kcal/mol, respectively, consistent with

219

their PPARG activation. As shown in Figure 2, hydrophobicity of TBP, EHDPP, and TPhP

220

plays an important role in human PPARG activation, as the nine important amino acid

221

residues (Leu333, Phe347, Ile341, Met348, Phe352, Leu270, Phe287, Ile281, and Phe363) 10

ACS Paragon Plus Environment

Page 10 of 27

Page 11 of 27

Environmental Science & Technology

222

present in the inner surface of the ligand-binding domain (LBD) of human PPARG form the

223

hydrophobic agonist pocket. EHDPP and TPhP both showed π-π interaction between their

224

benzene rings and Phe287 of PPARG-LBD (face to face). In contrast to TPhP, an important

225

feature of the EHDPP interaction was the formation of hydrogen bonds between EHDPP and

226

the residue Ser342 of PPARG-LBD (2.217 Å), which might contribute to the relatively high

227

PPARG activation by EHDPP. Such differences in binding mode might also explain the

228

differences in PPARG activation of the three chemicals. To verify the involvement of the

229

hydrogen bonds in the activation of EHDPP to PPARG, we substituted Ser342 of PPARG

230

with alanine and then carried out PPARG activation of EHDPP using the dual-luciferase

231

reporter assay. The activation of EHDPP to the PPARG mutant was found to be about 2.5 fold

232

lower than that of wild-type (Figure 3), demonstrating the importance of hydrogen bond in

233

PPARG activation by EHDPP.

234

To further demonstrate the PPARG activity of OPFRs, three main downstream genes of

235

PPARG, hCG, pregnancy-associated plasma protein-A (PAPP-A), and mucin gene Muc1

236

were also quantified in human placental choriocarcinoma JEG-3 cells.26,27 Confluent JEG-3

237

cells showed no change in cell viability or cell proliferation after treatment with EHDPP,

238

TPhP, TCEP, TBP, or TCPP at 5–40 µM for 48 h, and therefore the concentrations of 5, 10, 20,

239

and 40 µM were used in the following experiments (Figure S2). EHDPP and TPhP

240

significantly upregulated Muc1 and hCG expression but significantly down-regulated

241

PAPP-A gene expression compared with the control (Figure 4), further demonstrating

242

PPARG activation of TPhP and EHDPP in human placenta.

11

ACS Paragon Plus Environment

Environmental Science & Technology

243

Disruption of Progesterone Secretion. Since hCG governs the production of

244

progesterone in the corpus luteum during the first trimester,26,28 and significantly upregulated

245

hCG alterations may disrupt progesterone secretion. In addition to the gene expression of

246

hCG, its production alterations were also determined. Both TPhP and EHDPP significantly

247

increased hCG levels in the 20 and 40 µM groups compared with the control (Figure 5).

248

To investigate whether the five target OPFRs could disrupt progesterone biosynthesis,

249

JEG-3 cells were grown to confluence and treated with OPFRs at different concentrations,

250

and concentration of progesterone was determined. Among the OPFRs tested, EHDPP and

251

TPhP induced progesterone in a dose-dependent manner, while no changes in progesterone

252

level were observed in cells exposed to TBP, TCEP, or TCPP (Figure 5). The progesterone

253

concentration was significantly upregulated in the 20 and 40 µM TPhP exposure groups, with

254

1.29 ± 0.09 and 1.28 ± 0.07-fold increases, respectively, relative to that of the control (p