Plasticizer bis(2-ethylhexyl) phthalate causes meiosis defects and

3 days ago - Bis(2-ethylhexyl) phthalate (DEHP) is a widely used plasticizer in polyvinyl chloride (PVC) plastics. Humans and animals are widely and ...
0 downloads 0 Views 5MB Size
Subscriber access provided by WEBSTER UNIV

Food Safety and Toxicology

Plasticizer bis(2-ethylhexyl) phthalate causes meiosis defects and decreases fertilization ability of mouse oocytes in vivo Zhenzhen Lu, Chengtu Zhang, Chengquan Han, Quanli An, Yuyao Cheng, Yongzhong Chen, Ru Meng, Yong Zhang, and Jianmin Su J. Agric. Food Chem., Just Accepted Manuscript • DOI: 10.1021/acs.jafc.9b00121 • Publication Date (Web): 28 Feb 2019 Downloaded from http://pubs.acs.org on March 1, 2019

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 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 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.

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 44

Journal of Agricultural and Food Chemistry

1

Plasticizer bis(2-ethylhexyl) phthalate causes meiosis defects and

2

decreases fertilization ability of mouse oocytes in vivo

3 4

Running title: DEHP impairs mouse oocyte quality

5 6

Zhenzhen Lu † , § , Chengtu Zhang ‡ , § , Chengquan Han † , Quanli An † , Yuyao

7

Cheng†, Yongzhong Chen‡, Ru Meng‡, Yong Zhang* †, and Jianmin Su*



8

9

† College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi

10

Province, 712100, PR China.

11

‡ Xining animal husbandry and veterinary station, Xining, Qinghai Province, 810003,

12

PR China.

13

§The first two authors contributed equally to this manuscript.

14

* Corresponding author:

15

[email protected]; [email protected]

16 17 18

1

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

19

Abstract

20

Bis(2-ethylhexyl) phthalate (DEHP) is a widely used plasticizer in polyvinyl

21

chloride (PVC) plastics. Humans and animals are widely and continuously exposed to

22

DEHP, especially in dietary respect, which is associated with reproductive diseases.

23

Nevertheless, the effects and underlying mechanisms of DEHP exposure on oocytes

24

in vivo remain ambiguous. In this study, we found that oral administration of DEHP

25

(40 µg/kg body weight per day for 14 days) markedly reduced the maturation and

26

fertilization of oocytes in vivo. In addition, DEHP caused oxidative stress, increased

27

reactive oxygen species generation, promoted early apoptosis, and resulted in DNA

28

damage in mouse oocytes. Moreover, DEHP exposure caused mitochondrial damage,

29

reduced ATP content, down-regulated actin expression, and disturbed the spindle

30

assembly and chromosome alignment in mouse oocytes. Furthermore, DEHP

31

exposure remarkably impaired the localization and protein level of Juno, the sperm

32

receptor on the membrane of oocytes. The levels of DNA methylation, H3K9me3,

33

and H3K9ac were aslo altered in the DEHP-exposed mouse oocytes. Thus, our results

34

indicated that DEHP exposure reduced the maturation and fertilization capabilities of

35

mouse oocytes by affecting cytoskeletal dynamics, oxidative stress, early apoptosis,

36

meiotic spindle morphology, mitochondria, ATP content, Juno expression, DNA

37

damage, and epigenetic modifications in mouse oocytes.

38 39

Keywords: DEHP, Oocyte, Oxidative stress, Apoptosis, Epigenetic modification

2

ACS Paragon Plus Environment

Page 2 of 44

Page 3 of 44

Journal of Agricultural and Food Chemistry

41

Introduction

42

Bis(2-ethylhexyl) phthalate (DEHP) is a widely used plasticizer in polyvinyl

43

chloride (PVC) plastics for daily necessities and medical devices. More than 8.3

44

billion metric tons of virgin plastics have been produced, and about 6.3 billion metric

45

tons of them have turned into plastic waste over the world 1. In China, the estimated

46

daily dietary intake of DEHP for adults was 1.60 µg/kg/day 2. Given its weak

47

molecular linkages with the PVC polymers, DEHP can easily be released from

48

industrial settings and plastic wastes to enter and pollute the air, soil, and water

49

Therefore, humans and animals are widely and continuously exposed to high levels of

50

DEHP through ingestion, inhalation, or skin absorption 6. In general, the Agency for

51

Toxic Substances and Disease Registry reported that humans are only allowed 3-30

52

µg/kg/day exposure to DEHP, but this level can reach 700 mg/kg in household dust 7-8.

53

Consequently, more than 95% of human blood samples and 100% of urine samples

54

contain DEHP or DHEP metabolites. Moreover, more than 90% of amniotic fluid,

55

newborn blood, human breast milk, and follicular fluid samples contain DEHP residue

56

6-7, 9-11.

3-5.

57

DEHP exposure of humans and animals is associated with reproductive diseases,

58

including reproductive hormone alteration, anovulation, suppressed or delayed

59

ovulation, irregular estrogen secretion, small preovulatory follicles, sterility, and

60

damaged sperm DNA

61

DEHP could impair estrous cycle and inhibit ovulation in female rats 16. Hannon et al.

62

demonstrated that low doses of DEHP disrupt the estrous cycle and accelerate

6, 12-15.

An earlier report showed that oral administration of

3

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 4 of 44

63

primordial follicle recruitment in adult mice; they hypothesized that DEHP interferes

64

with normal reproductive function and disrupts reproductive senescence

65

research has shown that DEHP exposure could impair the meiotic progression of

66

oocytes in vitro in mares, bovines, and mice

67

DEHP exposure on the meiotic progression of mouse oocytes in vitro and suggested

68

that DEHP could perturb the meiosis of oocytes and disturb the expression of genes

69

associated with ovarian development

70

DEHP negatively affects bovine oocyte maturation and influences oocyte

71

developmental competence in vitro

72

mechanisms of DEHP in vivo exposure on oocytes remain unclear.

22.

18-22.

17.

Recent

Liu et al. evaluated the effects of

In addition, some studies suggested that

19-21.

Nevertheless, the effects and underlying

73

In the present study, we established a mouse model to explore the toxic effects and

74

underlie mechanisms of DEHP on mammalian oocytes in vivo. We found that

75

maturation of oocytes and in vitro fertilization decreased in mice orally administered

76

with DEHP. Our results indicated that DEHP can affect the cytoskeletal dynamics,

77

reactive oxygen species (ROS) levels, apoptosis, energy, and epigenetic modifications

78

of mouse oocytes.

79 80

Materials and methods

81

Animals

82

All animal experiments were carried out in accordance with the Animal Care

83

Commission of the College of Veterinary Medicine, Northwest A&F University.

84

(certificate no.: SCXK [SHAAN] 2017-003). Six to eight-week-old ICR female mice

4

ACS Paragon Plus Environment

Page 5 of 44

Journal of Agricultural and Food Chemistry

85

were kept under stable temperature (24°C–26°C) and illumination (12 h light–dark

86

cycle) with free food and water available.

87 88 89

Chemicals Anti-DNA/RNA damage, anti-H3K9ac, and anti-H3K9me3 were purchased from

90

Abcam

(Cambridge,

UK).

Annexin

V-FITC

Apoptosis

Detection

Kit,

91

Mito-Tracker@Green, 4,6-diamidino-2-phenylindole (DAPI), anti-α-tubulin were

92

purchased from Beyotime (Haimen, China). Folr4-FITC was purchased from

93

BioLegend (CA, USA), CellROX® Green Reagent was purchased from life

94

technologies (CA, USA), BODIPY FL ATP was obtained from Thermo Fisher

95

Scientific (MA, USA). G-IVF medium was from Vitrolife (Goteborg, Sweden). All

96

other reagents and chemicals used in this study were obtained by Sigma Chemical

97

Company unless otherwise indicated.

98 99

Feeding treatment

100

Oocytes were harvested from 6–8-week-old female ICR mice treated with oral

101

doses of DEHP. In brief, the mice orally received 0, 10, 40, or 80 µg/kg body weight

102

daily dose of DEHP with a gavage needle (Engineering 360, NY, USA) in a corn oil

103

carrier at 6:00 pm every day for 14 days before oocyte collection. In the control and

104

the 40 µg/kg DEHP-treated group, there were 30 mice were used respectively. In the

105

10 µg/kg and 80µg/kg DEHP-treated group, there were 7 and 6 mice were analyzed,

106

respectively.

5

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

107 108

Hematoxylin and eosin staining

109

Ovaries were collected from mice and immediately fixed in 4% paraformaldehyde

110

for 24 h, then the ovaries were embedded by paraffin. Serially ovaries were

111

frozen-sectioned at 5 μm thick along the longitudinal direction. Ovary sections were

112

stained with hematoxylin for a minute and washed with dripping water. The sections

113

were then stained with eosin for 5–10 s and washed with dripping water. Finally, the

114

stained sections were analyzed for the pathological changes of the ovaries under a

115

microscope (Carl Zeiss LSM 700 META, Jena, Germany).

116 117

In vitro maturation of oocytes

118

Germinal vesicle oocytes were collected, washed with M2 medium, and then

119

incubated in M2 medium with mineral oil covering the surface at 37 °C in 5% CO2

120

for 16 h until maturation. The rate of the first polar body extrusion was calculated as

121

the oocyte maturation rate.

122 123

Metaphase II (MII) oocyte collection

124

Except calculating maturation rates, all oocytes used in other experiments were MII

125

oocytes in vivo. MII-stage oocytes were collected from the oviductal ampullae. In

126

brief, female mice were intraperitoneally injected with 10 IU pregnant mare serum

127

gonadotrophin for the stimulation of follicle growth, followed by 10 IU human

128

chorionic gonadotrophin (hCG) 48 h later. At 13-15 h after hCG injection, oocytes

6

ACS Paragon Plus Environment

Page 6 of 44

Page 7 of 44

129

Journal of Agricultural and Food Chemistry

were collected in M2 medium.

130 131

In vitro fertilization

132

Sperms were collected from 12-week-old male mice. In brief, caudal epididymides

133

and spermaduct were obtained by surgery, washed with G-IVF (Vitrolife, Goteborg,

134

Sweden) medium, and then placed in a new dish of G-IVF to release the sperm with

135

shears and tweezers. Sperms floated on the surface of 3 mL G-IVF in a 15 mL

136

centrifuge tube for 30 min at 37 °C in 5% CO2. The sperm suspension was

137

centrifuged at 735 × g for 5 min, transferred to 200 µL of G-IVF, and then incubated

138

with MII oocytes for 6 h at 37 °C in 5% CO2. Zygotes were counted for fertilization

139

rate statistics.

140 141

Immunofluorescence staining of oocytes

142

Oocytes were fixed with in Immunol Staining Fix Solution (Beyotime, Haimen,

143

China) for 2 h, permeabilized with 0.5% Triton X-100 for 20 min at room

144

temperature, and then blocked with Immunol Staining Blocking Solution (Beyotime,

145

Haimen, China) at 4°C overnight. The samples were stained with anti-DNA/RNA

146

damage (1:500, Abcam, Cambridge, UK ), anti-α-tubulin (1:100, Beyotime),

147

anti-vimentin (1:500, Sigma, St. Louis, MO, USA), Phalloidin-TRITC (1:200, sigma),

148

Folr4-FITC (1:100, BioLegend, CA, USA), anti-H3K9ac (1:500, Abcam, Cambridge,

149

UK), and anti-H3K9me3 (1:500, Abcam) antibodies for 12 h at 4 °C. After extensive

150

washing, oocytes were incubated with Alexa Fluor 488/555-labeled IgG (Beyotime)

7

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

151

at room temperature for 3 h. After washing three times, the oocytes were

152

counterstained with 4,6-diamidino-2-phenylindole (DAPI, Beyotime) for 4-5 min at

153

room temperature. Finally, the oocytes were mounted on glass slides and analyzed

154

using a confocal laser scanning microscope (Ador REVOLUTION WD, Northern

155

Ireland, England), Those oocytes with Anti-DNA/RNA Damage antibody were

156

analyzed under a fluorescence microscope (Carl Zeiss LSM 700 META, Jena,

157

Germany).

158

For 5mC and 5hmC staining, permeabilized samples were treated with 4 N HCl for

159

10 min and neutralized with Tris-HCl (pH 8.5) for 10 min at room temperature. Other

160

procedures were in accordance with our previous work 23, except DAPI staining was

161

performed for 10 min.

162

The fluorescence intensity was measured and analyzed using Image-Pro Plus 6.0

163

(Media Cybernetics, Silver Spring, MD, USA) based on previous reports of our

164

laboratory

165

were measured. The average normalized fluorescence intensity for oocytes was

166

represented by ‘sum IOD / sum area’. To quantify fluorescence intensity, the intensity

167

levels of DEHP-treated oocytes were presented relative to the mean intensity level of

168

control oocytes.

. Using Image-Pro plus, the integrated optical density (IOD) and area

24

169 170

ROS detection and Annexin-V staining

171

CellROX® Green Reagent (1:500, life technologies, CA, USA) was used to

172

examined the ROS level in living oocytes. In brief, MII oocytes were incubated in M2

8

ACS Paragon Plus Environment

Page 8 of 44

Page 9 of 44

Journal of Agricultural and Food Chemistry

173

medium with 50 μL of CellROX® Green Reagent for 30 min at 37 °C in 5% CO2.

174

After washing three times in PBS-PVA, the oocytes were placed on glass slides and

175

examined immediately under the fluorescence microscope.

176

For Annexin-V staining, Annexin V-FITC Apoptosis Detection Kit (Beyotime) was

177

used to identify phosphatidylserine exteriorization in early apoptotic cells. After

178

washing three times, living oocytes were stained for 20 min in darkness with 195 µL

179

of Annexin V-FITC binding buffer containing 5 µL of Annexin-V-FITC and 10 µL of

180

propidium iodide at room temperature. After washing three times in PBS-PVA, the

181

oocytes were placed on glass slides and examined immediately under the confocal

182

laser scanning microscope.

183 184

Staining of mitochondria in oocytes

185

Mito-Tracker@Green (Beyotime) was used to evaluate mitochondrial activity in the

186

oocytes. After washing three times, MII oocytes were stained with 200 nM of

187

Mito-Tracker@Green in M2 medium for 30 min at 37 °C in 5% CO2. The oocytes

188

were washed three times in PBS-PVA, mounted onto glass slides, and examined

189

immediately under the fluorescence microscope.

190 191

ATP content assay

192

After washing three times in PBS-PVA, denuded oocytes were incubated in M2

193

medium supplemented with BODIPY FL ATP (1:10000, Thermo Fisher Scientific,

194

MA, USA) for 30 min at room temperature in darkness. The oocytes were washed

9

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

195

three times in PBS-PVA, mounted onto glass slides, and examined immediately under

196

the fluorescence microscope.

197 198

Statistical analysis

199

Statistical analysis was performed with the SPSS 19.0 software (SPSS Inc.,

200

Chicago, IL, USA). Each experiment was repeated three times. When multiple

201

comparisons were made, data were analyzed by one-way ANOVA and LSD tests.

202

When only two comparisons were made, Student’s t test was used. Data were

203

presented as means ± SEM. Differences were considered statistically significant when

204

p < 0.05.

205 206

Results

207

DEHP causes primary follicle damage

208

In the control group (Fig. 1A), at the center of the primary follicle is the oocyte

209

which has clear nucleus and zona pellucida, and the oocyte is surrounded by

210

granulosa cells of normal shape. This is the morphology of a normal primary follicle.

211

However, in the DEHP-treated group, in the primary follicle, the oocyte nucleus

212

disappeared (Fig. 1B), the granular cells around the oocyte were extremely loose and

213

detached, and exhibited disorganized and disrupted morphologies (Fig. 1C). In

214

addition, degraded oocyte was found in the DEHP-exposed follicle (Fig. 1D).

215

10

ACS Paragon Plus Environment

Page 10 of 44

Page 11 of 44

216

Journal of Agricultural and Food Chemistry

DEHP interferes with mouse oocyte maturation and fertilization

217

Considering that good oocyte quality is essential for female fertility, we analyzed

218

the maturation and fertilization of mouse oocytes exposed to different doses of DEHP.

219

After 16 h of culture, more than 76% oocytes reached the MII stage in the control

220

group (76.75% ± 1.33%, n = 224; Fig. 2A, 2B). However, the maturation rate

221

significantly decreased with DEHP treatment (10 µg/kg/day: 71.77% ± 0.87%, n =

222

223, p < 0.05; 40 µg/kg/day: 50.86% ± 2.14%, n = 235, p < 0.01; 80 µg/kg/day:

223

34.62% ± 0.55%, n = 228, p < 0.01; Fig. 2A, 2B). These phenomena indicate that

224

DEHP can inhibit mouse oocyte maturation in a dose-dependent manner. Then, we

225

detected the fertilization rates in DEHP-exposed oocytes. As shown in figure 2, more

226

than 83% oocytes successfully fertilized and initiated cleavage into 2-cell embryos

227

(83.61% ± 2.47%, n = 183; Fig. 2C, 2D). However, exposure to 40 and 80 µg/kg

228

DEHP significantly decreased fertilization rates (40 µg/kg: 64.47% ± 2.78%, n = 104,

229

p < 0.01; 80 µg/kg, 5.36% ± 1.06%, n = 97, p < 0.01; Fig. 2C, 2D). We demonstrated

230

that 40 µg/kg and 80 µg/kg DEHP treatment could cause both oocyte maturation and

231

fertilization defects. However, the toxicity of 80 µg/kg dose, which causes extremely

232

low fertilization, is too high to explore the toxicity mechanism of DEHP in living

233

oocytes. Therefore, we used 40 µg/kg DEHP treatment (DEHP group) for the

234

following experiments.

235 236 237

DEHP exposure increases ROS level and early apoptosis in mouse oocytes To determine the specific mechanism by which DEHP affects oocyte maturation 11

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

238

and fertilization, we measured the ROS levels in mouse oocytes exposed to DEHP.

239

Compared to control group (40.24 ± 6.95, n = 52), the fluorescence intensity of ROS

240

in oocytes dramatically increased after DEHP exposure (92.11 ± 8.5, n=58, p < 0.01;

241

Fig. 3A, 3B). Considering that high ROS levels can induce early apoptosis through

242

cellular oxidative stress, we examined early apoptosis in DEHP-exposed oocytes. As

243

refer to previously study 25, if the external cellular membrane of oocyte with a clearly

244

green circle fluorescent signal, it was defined as Annexin V positive, otherwise it

245

would be defined as negative. The immunofluorescence signals of early apoptosis

246

increased in the DEHP-treated oocytes (Fig. 3C). The results indicated that the early

247

apoptosis rate of mouse oocytes significantly increased after DEHP exposure (DEHP:

248

72.14% ± 1.49%, n =65 vs. control: 34.91% ± 1.11%, n =43, p < 0.01; Fig. 3D). The

249

following data was also shown as DEHP-treated group vs. control group.

250 251

DEHP exposure causes DNA damage in mouse oocytes

252

8-Oxo-7,8-dihydroguanine is one of the most common DNA damages resulting

253

from ROS. Therefore, anti-DNA/RNA Damage antibody was used to investigate

254

whether DEHP exposure could cause DNA damage in mouse oocytes. As shown in

255

figure 4, DNA damage level significantly increased with DEHP exposure (1.93 ±

256

0.11, n=26 vs. 1.00 n=29; p < 0.01; Fig. 4A, 4B).

257

258

DEHP exposure causes mitochondrial damage and reduces ATP content in

259

mouse oocytes 12

ACS Paragon Plus Environment

Page 12 of 44

Page 13 of 44

Journal of Agricultural and Food Chemistry

260

Mitochondrion is a determinant factor in oocyte maturation and fertilization26-27.

261

Thus, we detected the mitochondrial activation in DEHP-exposed oocytes. The

262

quantification of fluorescence intensity showed that the signals of the mitochondria

263

was lower in the DEHP-exposed groups than in the controls (0.68 ± 0.01, n=83 vs.

264

1.00, n=102, p < 0.01; Fig. 5A, 5B) As produced in mitochondria, ATP content is

265

closely associated with mitochondrial activity, and ATP content could affect oocyte

266

biological process by serving as an energy source. Further examination showed that

267

the ATP content was significantly lower in the DEHP-exposed oocytes than in the

268

control group (0.38 ± 0.03, n=67 vs. 1.00, n=66, p < 0.01; Fig. 6A, 6B).

269 270

DEHP disturbs the spindle assembly and chromosome alignment during mouse

271

oocyte meiotic maturation

272

Aberrant spindle organization often activates the spindle assembly checkpoint,

273

resulting in meiotic arrest. Therefore, we analyzed the morphologies of meiotic

274

spindles in DEHP-treated and control oocytes through α-tubulin-FITC staining to

275

observe the spindle apparatus and DAPI staining to show the chromosome alignment.

276

As shown in figure 7, most oocytes in the control group showed typical barrel-shaped

277

spindle morphologies and well-aligned chromosome nearby the equatorial plate

278

(normal spindles). However, most of DEHP-exposed oocytes exhibited disorganized

279

and disrupted spindle morphologies (abnormal spindles). (Fig. 7A). Statistically, the

280

proportion of abnormal spindles significantly increased in the DEHP-exposed oocytes

281

compared with the control oocytes (72.24% ± 0.83%, n=108 vs. 11.71% ± 1.07%, 13

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

282

n=109; p < 0.01; Fig. 7B). Normal chromosome was a well-aligned chromosome on

283

the equatorial plate, otherwise it would be misaligned chromosomes. The rate of

284

misaligned chromosomes also significantly increased in the DEHP-exposed oocytes

285

(66.81% ± 2.18%, n = 108 vs. 12.09% ± 0.62%, n =109; p < 0.01; Fig. 7C). Taken

286

together, these phenomena indicate that DEHP impairs the meiotic maturation of

287

mouse oocyte by disturbing spindle assembly and chromosome alignment.

288 289

DEHP exposure reduces actin expression in mouse oocytes

290

Actin and vimentin, together with microtubule, constitute the cytoskeleton and

291

function in cell morphology maintenance and cell movement. In this study, exposure

292

to DEHP significantly reduced actin level (0.53 ± 0.08, n=36 vs. 1.00, n=27, p < 0.01;

293

Fig. 8A, 8B). However, exposure to DEHP did not significantly affect vimentin

294

expression (1.07 ± 0.02, n=22 vs. 1.00, n = 30, p > 0.05; Fig. 8C, 8D).

295 296

DEHP exposure reduces the protein level of Juno in the membrane of mouse

297

oocytes

298

The lack of Juno protein could disrupt sperm–egg fusion. Therefore, we analyzed

299

the localization and quantification of Juno on the oocyte membrane using

300

immunofluorescence. The Juno protein was equably distributed on the oocyte

301

membrane in the control group, but it was partly or entirely lost in the membrane of

302

the DEHP-exposed oocytes (Fig. 9A). The proportion of mislocalized Juno was

303

considerably higher in the DEHP-exposed oocytes than in the controls (48.10% ± 14

ACS Paragon Plus Environment

Page 14 of 44

Page 15 of 44

Journal of Agricultural and Food Chemistry

304

0.99%, n=86 vs. 17.33% ± 1.01%, n=82, p < 0.01; Fig. 9B). Interestingly, the

305

fluorescence signals of Juno were remarkably lower in the DEHP-treated oocytes than

306

in the control group ( 28.34 ± 0.70, n=86 vs.78.99 ±3.29, n=82, p < 0.05; Fig. 9C).

307

Collectively, we speculated that the reduction of Juno protein on DEHP-exposed

308

oocyte membrane is partly responsible for the low fertilization rate.

309 310

DEHP exposure results in epigenetic alterations in mouse oocytes

311

Correct and precise epigenetic modifications in oocytes are essential for

312

fertilization and zygote cleavage. We examined DNA methylation, histone

313

methylation, and acetylation levels in DEHP-exposed oocytes. As shown in figure 10,

314

5mC level significantly increased in the DEHP-exposed oocytes (1.36 ± 0.08, n = 54

315

vs. 1.00, n = 33, p < 0.05; Fig. 10A, 10B), whereas 5hmC level significantly

316

decreased with DEHP exposure (0.58 ± 0.01, n = 54 vs. 1.00, n = 33, p < 0.05; Fig.

317

10A, 10C). Moreover, DEHP treatment could significantly reduce H3K9me3 level

318

(0.62 ± 0.04, n = 52 vs. 1.00, n = 28, p < 0.05; Fig. 11A, 11B). By contrast, the

319

signals of H3K9ac were significantly higher in the DEHP-exposed oocytes than in the

320

control group (1.49 ± 0.03, n = 24 vs. 1.00, n = 33, p < 0.01; Fig. 11C, 11 D). Our

321

findings suggest that DEHP exposure could alter DNA methylation, histone

322

methylation, and acetylation modifications in mouse oocytes.

323 324

Discussion

15

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

325

Increasing evidence suggests that environmental agents cause reproductive

326

disorders in human populations and animals. DEHP is the most bounteous phthalate

327

in the environment

328

from the U.S. National Toxicology Program’s Center, thought that DEHP has the

329

potential to cause adverse effects in human reproduction. Phthalates exist in our daily

330

life as plasticizers. These substances are present in many personal-care necessities,

331

such as nail polish, hair spray, shampoo, soap, deodorants, lotions, and fragrances.

332

They are also used in some medical devices and pharmaceuticals, plastic products

333

(e.g., plastic raincoats and automotive plastics), adhesives, detergents, solvents, vinyl

334

tiles, and flooring. Humans are exposed to DEHP daily

335

the specific contribution of DEHP to oocyte development failure is important.

28.

Panel, an expert of evaluation of risks to human reproduction

28.

Therefore, understanding

336

In the present study, we performed a series of experiments on mouse models in

337

oocytes to investigate the negative effects of DEHP exposure on oocyte development.

338

We investigated cytoskeleton, apoptosis, ROS levels, epigenetic modifications, and

339

receptor protein Juno in mouse oocytes exposed to DEHP. We found that DEHP

340

could decline the maturation and fertilization abilities of oocytes by disturbing the

341

structures or biological processes in mouse oocytes.

342

ROS participate not only in signal transduction, initiating mitosis, and pathogen

343

defense but also in follicular development, ovulation, corpus luteum function,

344

follicular atresia, and oocyte maturation

345

could increase oxidative stress levels in different cells. Consistent with their results,

346

our findings indicated that ROS accumulated in mouse oocytes exposed to DEHP.

29.

Previous studies have shown that DEHP

16

ACS Paragon Plus Environment

Page 16 of 44

Page 17 of 44

Journal of Agricultural and Food Chemistry

347

Many previous reports have claimed that ROS could lead to apoptosis 30-32 in different

348

cells. ROS, such as H2O2, could induce apoptosis in neutrophils

349

increasing evidence showed that ROS could cause apoptosis in other cell types 33. In

350

addition, excess ROS can cause DNA damage and can induce damage to cell

351

structure, such as lipids, cell membranes, and proteins

352

showed that DEHP cause ROS accumulation and DNA damage in mouse oocytes.

353

29, 33.

32.

Moreover,

The results of this study

Mitochondria are the source and target of ROS, and ROS could induce oxidative 34.

354

damage to mitochondria

355

associated with ATP production because mitochondrial clustering may promote ATP

356

production

357

disrupted

358

and electron transport chain

359

ROS, apoptosis, and DNA damage based on our results. In brief, high ROS levels

360

may cause early apoptosis and DNA damage in mouse oocytes. After DEHP

361

treatment, the mitochondrial ROS levels increased, which may cause mitochondrial

362

oxidative damage in turn. In addition, mitochondria are the source of ATP. Therefore,

363

ATP levels decreased in the mouse oocytes treated with 40 µg/kg DEHP. As the

364

energy source for cellular biological processes, ATP provides energy for centrosome,

365

cytoskeleton, and chromosomal activity during meiosis and mitosis. ATP content

366

decrease can interfere with the polymerization and depolymerization of tubulin on the

367

spindle, thereby affecting chromosome segregation and leading to aneuploidy

368

Thus, we further explored the changes in cytoskeleton on DEHP-exposed mouse

35-37,

37.

Earlier studies showed that mitochondrial state is

and ATP production decreases when mitochondrial clusters are

ROS could reduce ATP levels by affecting the tricarboxylic acid cycle 38.

We summarize the underlying connections among

17

ACS Paragon Plus Environment

39.

Journal of Agricultural and Food Chemistry

369

Page 18 of 44

oocytes.

370

In general, the cytoskeleton mainly contains microtubules, microfilaments, and

371

intermediate fibers present in the cytoplasm. In addition, the actin filament regulates

372

the spindle movement, activates cell meiosis, causes polar body extrusion, and

373

regulates the microtubules forming meiotic spindles, which promote the aggregation

374

and separation of chromosomes in oocytes. Mycotoxins and zearalenone reduce the

375

maturation rate of mouse oocytes by altering the expression and localization of actin

376

filaments and α-tubulin, the basic components of microtubules

377

showed that DEHP exposure reduced cytoskeletal actin expression and increased

378

spindle abnormalities and chromosome misalignment. This result suggests that DEHP

379

exposure causes the arrest of oocyte meiosis possibly by damaging spindle

380

organization and chromosome alignment. Among the three cytoskeletal types, the

381

intermediate filaments are the most complex. When treated with a high-salt solution

382

or nonionic detergent, most of the cytoskeleton components in the cells are destroyed,

383

leaving only the intermediate fibers. Vimentin is the composition protein of the

384

intermediate filaments. In the present study, vimentin expression showed no

385

significant difference between the DEHP-exposed and control groups.

386

40-41.

Our experiments

Previous reports demonstrated that epigenetic modifications could be changed 42.

387

after exposure to environmental chemicals in various cell types

388

cytoskeleton could be affected by epigenetic modification states, especially DNA

389

methylation and histone methylation and acetylation. Aberrant epigenetic

390

modifications usually indicate slim survivability in cells

18

ACS Paragon Plus Environment

43.

Cell cycle and

Epigenetic modification

Page 19 of 44

Journal of Agricultural and Food Chemistry

44-45.

391

states are also involved in meiosis and development during oocyte maturation

392

The epigenetic modification of cells is constantly adjusted through a range of

393

time-precise biological events, such as gamete development, fertilization, and fetal

394

development, which can be particularly vulnerably interfered by endocrine-disrupting

395

chemicals (EDCs) 46. These epigenetic modification changes could pass on to the next

396

generation

397

plastics and other EDC members could cause mammal oocytes meiosis defects and

398

mouse reproductive diseases in the F3 generation

399

DEHP exposure could alter DNA methylation, histone methylation, and acetylation

400

modifications in mouse oocytes, indicating that DEHP could affect epigenetic

401

modifications in mouse oocytes.

47-49.

Plasticizers are EDCs, and earlier reports showed that exposure to

50-53.

On the basis of our results,

402

Sperm-egg fusion is influenced by many factors, the most important of which is

403

Juno, a receptor protein on the oocyte, which binds to the Izumo1 protein on the

404

sperm. The lack of Juno protein causes a disorder in sperm-egg fusion. In the present

405

study, Juno was significantly reduced in the oocytes exposed to DEHP. This

406

phenomenon suggests that DEHP could impair oocyte fertilization.

407

According to the Food and Drug Administration, the oral tolerable intake range for

408

DEHP is 40 µg/kg/day 54, and the Environmental Protection Agency reported that 20

409

and 50 µg/kg/day of DEHP may cause hepatomegaly and testicular toxicity,

410

respectively 55. In addition, some previous studies found that 40 µg/kg/day of DEHP

411

could impair oocyte maturation through oral administration or injection

412

Therefore, we chose 0-80 µg/kg/day of DEHP as the oral dose to evaluate the effect of

19

ACS Paragon Plus Environment

56-57.

Journal of Agricultural and Food Chemistry

413

DEHP exposure on oocyte quality.

414

In conclusion, our results indicated that DEHP exposure affected actin and Juno

415

expression, meiotic spindle morphology, apoptosis, oxidative stress, ATP content,

416

mitochondria, DNA damage, and epigenetic modifications in mouse oocytes. These

417

alterations may be responsible for the decreased maturation and fertilization rates in

418

DEHP-exposed oocytes. The doses we used in this study are within the range of

419

environmental exposure levels in humans. Therefore, our findings provide some

420

supporting insights into the reproductive toxicity of DEHP in humans.

421 422

Acknowledgments

423

The authors wish to thank Dr. Bo Xiong and Dr. Shaochen Sun from Nanjing

424

Agricultural University for his generous technical assistance. This work was

425

supported by the National Natural Science Foundation of China (31873001),

426

National Key R&D Program of China (2018YFD0502304), and Qinghai Science and

427

Technology Project (2017-NK-111, 2018-NK-132).

428 429

Author contributions

430

JMS designed and conceived experiments. ZZL performed the experiments with

431

assistance from CTZ, CQH, QLA, YYC, YZC, and YM. ZZL analyzed data and

432

wrote the manuscript. JMS and YZ provided research guidance, critical revision of

433

the manuscript, and approval of the article.

434

20

ACS Paragon Plus Environment

Page 20 of 44

Page 21 of 44

Journal of Agricultural and Food Chemistry

435

Declaration of conflicting interests

436

The authors report that no conflict of interest exists to prejudice the impartiality of the

437

research reported.

438 439 440 441

References (1) Geyer, R.; Jambeck, J. R.; Law, K. L., Production, use, and fate of all plastics ever made. Sci Adv 2017, 3 (7), e1700782.

442

(2) Guo, Y.; Zhang, Z.; Liu, L.; Li, Y.; Ren, N.; Kannan, K., Occurrence and profiles of phthalates in

443

foodstuffs from China and their implications for human exposure. Journal of agricultural and food

444

chemistry 2012, 60 (27), 6913-6919.

445 446 447 448 449 450 451 452

(3) Bosnir, J.; Puntaric, D.; Skes, I.; Klaric, M.; Simic, S.; Zoric, I., Migration of phthalates from plastic products to model solutions. Coll Antropol 2003, 27 Suppl 1, 23-30. (4) Petersen, J. H.; Breindahl, T., Plasticizers in total diet samples, baby food and infant formulae. Food Addit Contam 2000, 17 (2), 133-141. (5) Liang, D. W.; Zhang, T.; Fang, H. H.; He, J., Phthalates biodegradation in the environment. Appl Microbiol Biotechnol 2008, 80 (2), 183-198. (6) Heudorf, U.; Mersch-Sundermann, V.; Angerer, J., Phthalates: toxicology and exposure. Int J Hyg Environ Health 2007, 210 (5), 623-634.

453

(7) Becker, K.; Seiwert, M.; Angerer, J.; Heger, W.; Koch, H. M.; Nagorka, R.; Rosskamp, E.;

454

Schluter, C.; Seifert, B.; Ullrich, D., DEHP metabolites in urine of children and DEHP in house dust.

455

Int J Hyg Environ Health 2004, 207 (5), 409-417.

456

(8) Agency for Toxic Substances and Disease Registry (ATSDR), . In: Production, Import, Use, and Disposal Potential for Human

458

Exposure. Atlanta, GA: U.S. Department of Health and Human Services, Public Health Service 2002,

459

176–213.

460

(9) Marsee, K.; Woodruff, T. J.; Axelrad, D. A.; Calafat, A. M.; Swan, S. H., Estimated daily

461

phthalate exposures in a population of mothers of male infants exhibiting reduced anogenital distance.

462

Environ Health Perspect 2006, 114 (6), 805-809.

463

(10) Silva, M. J.; Barr, D. B.; Reidy, J. A.; Malek, N. A.; Hodge, C. C.; Caudill, S. P.; Brock, J. W.;

464

Needham, L. L.; Calafat, A. M., Urinary levels of seven phthalate metabolites in the U.S. population

465

from the National Health and Nutrition Examination Survey (NHANES) 1999-2000. Environ Health

466

Perspect 2004, 112 (3), 331-338.

467

(11) Hogberg, J.; Hanberg, A.; Berglund, M.; Skerfving, S.; Remberger, M.; Calafat, A. M.; Filipsson,

468

A. F.; Jansson, B.; Johansson, N.; Appelgren, M.; Hakansson, H., Phthalate diesters and their

469

metabolites in human breast milk, blood or serum, and urine as biomarkers of exposure in vulnerable

470

populations. Environ Health Persp 2008, 116 (3), 334-339.

471

(12) Hauser, R.; Meeker, J. D.; Singh, N. P.; Silva, M. J.; Ryan, L.; Duty, S.; Calafat, A. M., DNA

472

damage in human sperm is related to urinary levels of phthalate monoester and oxidative metabolites.

473

Hum Reprod 2007, 22 (3), 688-695.

474

(13) Li, L.; Liu, J. C.; Lai, F. N.; Liu, H. Q.; Zhang, X. F.; Dyce, P. W.; Shen, W.; Chen, H., Di

475

(2-ethylhexyl) Phthalate Exposure Impairs Growth of Antral Follicle in Mice. PLoS One 2016, 11 (2),

476

e0148350.

477 478

(14) Hauser, R.; Calafat, A. M., Phthalates and human health. Occup Environ Med 2005, 62 (11), 806-818.

22

ACS Paragon Plus Environment

Page 22 of 44

Page 23 of 44

479 480

Journal of Agricultural and Food Chemistry

(15) Krotz, S. P.; Carson, S. A.; Tomey, C.; Buster, J., Phthalates and bisphenol do not accumulate in human follicular fluid. J Assist Reprod Genet. 2012, 29 (8), 773-777.

481

(16) Hirosawa, N.; Yano, K.; Suzuki, Y.; Sakamoto, Y., Endocrine disrupting effect of di ‐ (2 ‐

482

ethylhexyl) phthalate on female rats and proteome analyses of their pituitaries. J Proteomics 2006, 6

483

(3), 958-971.

484

(17) Hannon, P. R.; Peretz, J.; Flaws, J. A., Daily exposure to Di(2-ethylhexyl) phthalate alters estrous

485

cyclicity and accelerates primordial follicle recruitment potentially via dysregulation of the

486

phosphatidylinositol 3-kinase signaling pathway in adult mice. Biol Reprod 2014, 90 (6), 1-11.

487

(18) Ambruosi, B.; Uranio, M. F.; Sardanelli, A. M.; Pocar, P.; Martino, N. A.; Paternoster, M. S.;

488

Amati, F.; Dell'Aquila, M. E., In vitro acute exposure to DEHP affects oocyte meiotic maturation,

489

energy and oxidative stress parameters in a large animal model. PLoS One 2011, 6 (11), e27452.

490 491

(19) Anas, I. M. K.; Suzuki, C.; Iwamura, S.; Yoshioka, K., Effect of mono-(2-ethylhexyl) phthalate on bovine oocyte maturation in vitro. Biology of Reproduction 2001, 64 (3), 139-139.

492

(20) Beker van Woudenberg, A.; Grollers-Mulderij, M.; Snel, C.; Jeurissen, N.; Stierum, R.;

493

Wolterbeek, A., The bovine oocyte in vitro maturation model: a potential tool for reproductive

494

toxicology screening. Reprod Toxicol 2012, 34 (2), 251-260.

495

(21) Grossman, D.; Kalo, D.; Gendelman, M.; Roth, Z., Effect of di-(2-ethylhexyl) phthalate and

496

mono-(2-ethylhexyl) phthalate on in vitro developmental competence of bovine oocytes. Cell Biol

497

Toxicol 2012, 28 (6), 383-396.

498

(22) Liu, J. C.; Lai, F. N.; Li, L.; Sun, X. F.; Cheng, S. F.; Ge, W.; Wang, Y. F.; Li, L.; Zhang, X. F.;

499

De Felici, M.; Dyce, P. W.; Shen, W., Di (2-ethylhexyl) phthalate exposure impairs meiotic

500

progression and DNA damage repair in fetal mouse oocytes in vitro. Cell Death Dis 2017, 8 (8), e2966.

23

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

501

(23) Su, J. M.; Wang, Y. S.; Xing, X. P.; Zhang, L.; Sun, H. Z.; Zhang, Y., Melatonin significantly

502

improves the developmental competence of bovine somatic cell nuclear transfer embryos. J Pineal Res

503

2015, 59 (4), 455-468.

504

(24) Xu, W.; Wang, Y.; Li, Y.; Wang, L.; Xiong, X.; Su, J.; Zhang, Y., Valproic acid improves the in

505

vitro development competence of bovine somatic cell nuclear transfer embryos. Cellular

506

reprogramming 2012, 14 (2), 138-145.

507

(25) An Q, Peng W, Cheng Y, Lu Z, Zhou C, Zhang Y, Su J. Melatonin supplementation during in

508

vitro maturation of oocyte enhances subsequent development of bovine cloned embryos. J Cell Physiol.

509

2019,1-12, doi: 10.1002/jcp.28357..

510 511

(26) Roth, Z., Reduction in oocyte developmental competence by stress is associated with alterations in mitochondrial function. Journal of Dairy Science 2018, 101 (4), 3642-3654.

512

(27) Jonathan, V. B.; Patrick, D.; Vicky, M.; Samuel, A., Domains of high-polarized and

513

low-polarized mitochondria may occur in mouse and human oocytes and early embryos. Human

514

Reproduction 2002, 17 (2), 393-406.

515

(28) Akingbemi, B. T.; Sottas, C. M.; Koulova, A. I.; Klinefelter, G. R.; Hardy, M. P., Inhibition of

516

testicular steroidogenesis by the xenoestrogen bisphenol A is associated with reduced pituitary

517

luteinizing hormone secretion and decreased steroidogenic enzyme gene expression in rat Leydig cells.

518

Endocrinology 2004, 145 (2), 592-603.

519

(29) Das, S.; Chattopadhyay, R.; Ghosh, S.; Ghosh, S.; Goswami, S.; Chakravarty, B.; Chaudhury.,

520

Reactive oxygen species level in follicular fluid—embryo quality marker in IVF? J Human

521

Reproduction 2006, 21 (9), 2403-2407.

522

(30) Tatemoto, H.; Sakurai, N.; Muto, N., Protection of porcine oocytes against apoptotic cell death

24

ACS Paragon Plus Environment

Page 24 of 44

Page 25 of 44

Journal of Agricultural and Food Chemistry

523

caused by oxidative stress during In vitro maturation: role of cumulus cells. Biol Reprod 2000, 63 (3),

524

805-810.

525 526

(31) Pierce, G. B.; Parchment, R. E.; Lewellyn, A. L., Hydrogen peroxide as a mediator of programmed cell death in the blastocyst. Differentiation 2010, 46 (3), 181-186.

527

(32) Kasahara, Y.; Iwai, K.; Yachie, A.; Ohta, K.; Konno, A.; Seki, H.; Miyawaki, T.; Taniguchi, N.,

528

Involvement of reactive oxygen intermediates in spontaneous and CD95 (Fas/APO-1)-mediated

529

apoptosis of neutrophils. Blood 1997, 89 (5), 1748-1753.

530 531 532 533

(33) Simon, H. U.; Haj-Yehia, A.; Levi-Schaffer, F., Role of reactive oxygen species (ROS) in apoptosis induction. Apoptosis 2000, 5 (5), 415-418. (34) Kawamura, K.; Qi, F.; Kobayashi, J., Potential relationship between the biological effects of low-dose irradiation and mitochondrial ROS production. J Radiat Res 2018, 59 (suppl_2). ii91–ii97

534

(35) Van Blerkom, J.; Davis, P. W.; Lee, J., ATP content of human oocytes and developmental

535

potential and outcome after in-vitro fertilization and embryo transfer. Hum Reprod 1995, 10 (2),

536

415-424.

537

(36) Wilding, M.; Dale, B.; Marino, M.; di Matteo, L.; Alviggi, C.; Pisaturo, M. L.; Lombardi, L.; De

538

Placido, G., Mitochondrial aggregation patterns and activity in human oocytes and preimplantation

539

embryos. Hum Reprod 2001, 16 (5), 909-917.

540

(37) Yu, Y.; Dumollard, R.; Rossbach, A.; Lai, F. A.; Swann, K., Redistribution of mitochondria leads

541

to bursts of ATP production during spontaneous mouse oocyte maturation. J Cell Physiol 2010, 224

542

(3), 672-680.

543 544

(38) Brookes, P. S.; Yoon, Y.; Robotham, J. L.; Anders, M. W.; Sheu, S. S., Calcium, ATP, and ROS: a mitochondrial love-hate triangle. Am J Physiol Cell Physiol 2004, 287 (4), C817-C833.

25

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

545

(39) Cohen, J.; Scott, R.; Alikani, M.; Schimmel, T.; Munne, S.; Levron, J.; Wu, L.; Brenner, C.;

546

Warner, C.; Willadsen, S., Ooplasmic transfer in mature human oocytes. Mol Hum Reprod 1998, 4 (3),

547

269-280.

548

(40) Song, Y.; Hai, T.; Wang, Y.; Guo, R.; Li, W.; Wang, L.; Zhou, Q., Epigenetic reprogramming,

549

gene expression and in vitro development of porcine SCNT embryos are significantly improved by a

550

histone deacetylase inhibitor--m-carboxycinnamic acid bishydroxamide (CBHA). Protein & cell 2014,

551

5 (5), 382-393.

552

(41) Hou, Y. J.; Zhu, C. C.; Xu, Y. X.; Cui, X. S.; Kim, N. H.; Sun, S. C., Zearalenone exposure

553

affects mouse oocyte meiotic maturation and granulosa cell proliferation. Environ Toxicol 2015, 30

554

(10), 1226-1233.

555 556 557 558

(42) Bolt, A. M.; Klimecki, W. T., Autophagy in toxicology: self-consumption in times of stress and plenty. J Appl Toxicol 2012, 32 (7), 465-479. (43) Sui, X. B.; Zhu, J.; Zhou, J. C.; Wang, X.; Li, D.; Han, W. D.; Fang, Y.; Pan, H. M., Epigenetic modifications as regulatory elements of autophagy in cancer. Cancer Lett 2015, 360 (2), 106-113.

559

(44) Eppig, J. J.; Schroeder, A. C., Capacity of mouse oocytes from preantral follicles to undergo

560

embryogenesis and development to live young after growth, maturation, and fertilization in vitro. Biol

561

Reprod 1989, 41 (2), 268-276.

562 563 564 565 566

(45) Schultz, R. M., The molecular foundations of the maternal to zygotic transition in the preimplantation embryo. Hum Reprod Update 2002, 8 (4), 323-331. (46) Murphy, S. K.; Jirtle, R. L., Imprinting evolution and the price of silence. Bioessays 2003, 25 (6), 577-588. (47) Skinner, M. K.; Manikkam, M.; Guerrero-Bosagna, C., Epigenetic transgenerational actions of

26

ACS Paragon Plus Environment

Page 26 of 44

Page 27 of 44

567 568 569 570 571

Journal of Agricultural and Food Chemistry

endocrine disruptors. Reprod Toxicol 2011, 31 (3), 337-343. (48) Anway, M. D.; Cupp, A. S.; Uzumcu, M.; Skinner, M. K., Epigenetic transgenerational actions of endocrine disruptors and male fertility. Science 2005, 308 (5727), 1466-1469. (49) Janesick, A. S.; Shioda, T.; Blumberg, B., Transgenerational inheritance of prenatal obesogen exposure. Molecular and cellular endocrinology 2014, 398 (1-2), 31-35.

572

(50) Nilsson, E.; Larsen, G.; Manikkam, M.; Guerrero-Bosagna, C.; Savenkova, M. I.; Skinner, M. K.,

573

Environmentally induced epigenetic transgenerational inheritance of ovarian disease. PLoS One 2012,

574

7 (5), e36129.

575

(51) Zhang, M.; Miao, Y.; Chen, Q.; Cai, M.; Dong, W.; Dai, X.; Lu, Y.; Zhou, C.; Cui, Z.; Xiong, B.,

576

BaP exposure causes oocyte meiotic arrest and fertilization failure to weaken female fertility. FASEB J

577

2018, 32 (1), 342-352.

578

(52) Miao, Y.; Zhou, C.; Bai, Q.; Cui, Z.; Shiyang, X.; Lu, Y.; Zhang, M.; Dai, X.; Xiong, B., The

579

protective role of melatonin in porcine oocyte meiotic failure caused by the exposure to

580

benzo(a)pyrene. Human Reproduction 2018, 33 (1), 116-127.

581 582

(53) Z, Y.; T, W.; M, L.; XW, Z.; YL, L.; XS, C.; NH, K.; SC, S., Melatonin protects oocytes from MEHP exposure-induced meiosis defects in porcine. Biology of Reproduction 2018, 98 (3), 286-298.

583

(54) Zhang T, Li L, Qin XS, Zhou Y, Zhang XF, Wang LQ, De FeliciM, Chen H, Qin GQ, Shen W,

584

Di-(2-ethylhexyl) phthalate and bisphenol A exposure impairs mouse primordial follicle assembly in

585

vitro. Environ Mol Mutagen.2013, 55(4):343-353

586 587 588

(55) Serrano, S. E.; Braun, J.; Trasande, L.; Dills, R.; Sathyanarayana, S., Phthalates and diet: a review of the food monitoring and epidemiology data. Environ Health 2014, 13 (1), 43. (56) Zhang, X. F.; Zhang, L. J.; Li, L.; Feng, Y. N.; Chen, B.; Ma, J. M.; Huynh, E.; Shi, Q. H.; De

27

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

589

Felici, M.; Shen, W., Diethylhexyl phthalate exposure impairs follicular development and affects

590

oocyte maturation in the mouse. Environ Mol Mutagen 2013, 54 (5), 354-361.

591

(57) Li, L.; Zhang, T.; Qin, X. S.; Ge, W.; Ma, H. G.; Sun, L. L.; Hou, Z. M.; Chen, H.; Chen, P.; Qin,

592

G. Q.; Shen, W.; Zhang, X. F., Exposure to diethylhexyl phthalate (DEHP) results in a heritable

593

modification of imprint genes DNA methylation in mouse oocytes. Mol Biol Rep 2014, 41 (3),

594

1227-1235.

28

ACS Paragon Plus Environment

Page 28 of 44

Page 29 of 44

Journal of Agricultural and Food Chemistry

595

Figure legends

596

Figure 1. DEHP causes primary follicle damage

597

(A) Representative pictures of excellent primary follicles from the control group. The

598

black arrow indicates primary follicle. Scale bar=10 μm. (B, C, D) Representative

599

pictures of primary follicles from the DEHP-exposed group.

600 601

Figure 2. Effects of DEHP on mouse oocyte maturation and in vitro fertilization

602

(A) Representative images of the first polar body extrusion (PBE 1) in the control and

603

DEHP-exposed (10, 40, and 80 µg/kg) mouse oocytes. Scale bar=100 μm. (B) PBE

604

rates were recorded in the control and DEHP-exposed (10, 40, and 80 µg/kg) oocytes.

605

Data are presented as mean ± SEM (*p < 0.05, **p < 0.01). (C) Representative

606

images of fertilized oocytes in the control and DEHP-exposed (10, 40, and 80 µg/kg)

607

mouse oocytes. Scale bar=100 μm. (D) In vitro fertilization rates were analyzed in the

608

control and DEHP-exposed mouse oocytes. Data are presented as mean ± SEM (**p