Danio rerio

Dydrogesterone causes male bias and accelerates sperm maturation in zebrafish .... extraction, the gonads and brain of three female and four male fish...
0 downloads 0 Views 1MB Size
Subscriber access provided by Kaohsiung Medical University

Ecotoxicology and Human Environmental Health

Dydrogesterone causes male bias and accelerates sperm maturation in zebrafish (Danio rerio) Wen-Jun Shi, Yu-Xia Jiang, Guo-Yong Huang, Jian-Liang Zhao, JinNa Zhang, You-Sheng Liu, Lingtian Xie, and Guang-Guo Ying Environ. Sci. Technol., Just Accepted Manuscript • DOI: 10.1021/acs.est.8b02556 • Publication Date (Web): 13 Jul 2018 Downloaded from http://pubs.acs.org on July 13, 2018

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 29

Environmental Science & Technology

graphic abstract 63x43mm (300 x 300 DPI)

ACS Paragon Plus Environment

Environmental Science & Technology

1

Page 2 of 29

Dydrogesterone causes male bias and accelerates sperm maturation in zebrafish (Danio rerio)

2 3

Wen-Jun Shi†,§, Yu-Xia Jiang†,§, Guo-Yong Huang†, Jian-Liang Zhao†, Jin-Na Zhang†,§, You-Sheng

4

Liu†, Ling-Tian Xie†, Guang-Guo Ying†, *

5 6



7

Chemistry, South China Normal University, Guangzhou 510006, China

8

§

9

Pollution and Control, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences,

10

The Environmental Research Institute, MOE Key Laboratory of Environmental Theoretical

State Key Laboratory of Organic Geochemistry, CAS Research Centre of PRD Environmental

Guangzhou 510640, China

11 12

*Corresponding author (GG Ying)

13

Tel.: +86 20 39310796; Fax: +86 20 85290200.

14

E-mail address: [email protected]; [email protected]

15

1

ACS Paragon Plus Environment

Page 3 of 29

Environmental Science & Technology

16

ABSTRACT: Synthetic progestins are widely used in human and veterinary medicine. They can

17

enter aquatic environments mainly via wastewater discharge and agricultural runoff, thus affecting

18

fish populations in receiving waters. Here we investigated the chronic effects of dydrogesterone

19

(DDG) on zebrafish from 21 days post fertilization (dpf) to 140 dpf at 3.39, 33.1 and 329 ng L-1.

20

The results showed that the male ratio increased with exposure concentration, and after 120 d of

21

exposure to 329 ng L-1 98% of the fish were males. The DDG exposure during sex differentiation

22

significantly increased the transcription of dmrt1 (1.83 fold) and apoptosis related genes, but

23

suppressed the transcription of cyp19a1a (3.16 fold). Histological analysis showed that the exposure

24

to DDG at 329 ng L-1caused 61.5% of mature spermatocytes in males, while the exposure to DDG

25

at 33.1 ng L-1 resulted in 14.7% of atretic follicles in females. Microarray analysis identified

26

spermatogenesis related gene ontology (endothelial barrier and immune response) in the testes at all

27

concentrations. Genes from phagosome, lysosome and sphingolipid metabolism pathways were

28

enriched and could be responsible for sperm maturation. The findings from this study demonstrate

29

that DDG in the aquatic environment can cause male bias and accelerate sperm maturation in

30

zebrafish, resulting in potential high ecological risks to fish populations.

31

2

ACS Paragon Plus Environment

Environmental Science & Technology

32 33

Page 4 of 29

INTRODUCTION Dydrogesterone (DDG) is one of the commonly used progestins in contraception and therapy

34

of menstrual disorders and endometriosis in human and animals. 1 The consumption of DDG was

35

approximately 34 kg in Switzerland in 2010 and 209 kg in UK in 2006. 2,3 In France, the

36

consumption of DDG reached 744.70 kg in 2004. 4 Though the consumption of DDG is not clear in

37

China, DDG has been detected in the effluents of wastewater treatment plants (WWTPs) and

38

receiving waters. 5 The measured concentrations of DDG were up to 35.1 ng L-1 in WWTP effluents,

39

and 9.6 ng L-1 in river water in China. 5 Furthermore, some extreme values (up to 2790 ng L-1) for

40

DDG have been reported in the flush water of swine farms. 5 The lowest observed effect

41

concentration (LOEC) for DDG on gene expressions in zebrafish embryos has been reported to be

42

as low as 4.8 ng L-1, 4 which is below the reported levels in surface water (9.6 ng L-1) and in

43

effluents of WWTP (35 ng L-1). 5 This clearly suggests that DDG may pose a risk for fish at

44

environmentally realistic concentrations. DDG is relatively easy to be degraded in the exposure

45

medium. 4 In addition, the half-life of DDG is estimated to be approximately 5 - 7 hr in humans. 6

46

In general, DDG is purposed to resemble progesterone mainly in its progestogenic activity, but

47

less in the androgenic, anti-androgenic and glucocorticoid activities. 4 An in vivo study showed that

48

DDG increased the frequency of mature spermatocytes in zebrafish, implying DDG might have

49

potential androgenic activity. 4 An in vitro study using the GripTiteTM 293 cell line derived from

50

human embryonic kidney cell (HEK 293) demonstrated a negligible androgenic activity of DDG by

51

GeneBLAzer® Nuclear Receptor Cell-based assays. 1 Previous studies showed some androgenic

52

progestins such as norgestrel (NGT), and levonorgestrel (LNG) can cause skewed sex ratio in

53

zebrafish. 7,8 LNG and NGT caused male bias in zebrafish by affecting transcription of cytochrome

54

P450, family 19, subfamily A, polypeptide 1a (cyp19a1a) and double sex/mab-3 related 3

ACS Paragon Plus Environment

Page 5 of 29

Environmental Science & Technology

55

transcription factor 1 (dmrt1). 7,8 Genes such as dmrt1, forkhead transcription factor gene L2 (foxl2)

56

and cyp19a1a have been demonstrated to be associated with the process of sex differentiation in

57

zebrafish. 9 In addition, the oocyte apoptosis is the mechanism of gonadal differentiation in

58

zebrafish. 10 For instance, the synthetic estrogen 17α-ethinylestradiol (EE2) disturbs the gonadal

59

differentiation via affecting apoptosis pathway. 11 However, the effects of DDG on the sex

60

differentiation and sex determination in fish have yet to be determined.

61

Previous studies showed that progestins can affect the gonad development in fish. 4,7-8,12 These

62

studies have addressed the acute and chronic effects of progestins on the frequency of gonadal

63

maturity stages. 8,12 However, the mechanism of histological alterations in gonads after exposure to

64

progestins is still not clear. Compared with the acute exposure, chronic exposure at environmentally

65

realistic concentrations from fish larval stages to sexual maturity will provide better understanding

66

of the effects of progestins like DDG in fish.

67

The aim of this study was to investigate the chronic effects of DDG on gonad and brain in

68

zebrafish. To achieve our goal, the transcriptional expression of genes related to gonad

69

differentiation and apoptosis was examined in the zebrafish larvae at 35 dpf. In addition, microarray

70

analysis was conducted and histology of the gonads was evaluated in the sexually mature zebrafish

71

(140 dpf). The study can help us better understand the mechanisms for the effects of DDG on sex

72

differentiation and gonadal development.

73 74 75

MATERIALS AND METHODS Chemical. Dydrogesterone (DDG, CAS number: 152–62–5; purity: 98%) was obtained from

76

US Pharmacopeia (Rockville, MD). The stock solution of DDG was prepared in dimethyl sulfoxide

77

(DMSO), and then stored at -20 °C in the dark. 4

ACS Paragon Plus Environment

Environmental Science & Technology

78

Page 6 of 29

Test Organisms and Exposure Experiment. Zebrafish were maintained in the Aquatic

79

Toxicology Laboratory of South China Normal University. Zebrafish embryos were obtained from

80

spawning adults placed in groups of 6 males and 3 females. Normally developed embryos were

81

selected for the exposure. The embryos were collected and transferred into petri dishes with 50 mL

82

charcoal-filtered dechlorinated tap water, and kept at 26 ± 1 °C with a photoperiod of 14 h: 10 h

83

(light: dark). The zebrafish larvae were fed homogenate of brine shrimp twice daily from 5 dpf until

84

18 dpf. Larvae were fed with newly hatched brine shrimp after 18 dpf (Shandong, China).

85

The experimental set-up consisted of one solvent control (SC, 0.001% (v/v) of DMSO) and

86

three DDG groups: 5 (L treatment), 50 (M treatment), and 500 ng L−1 (H treatment) with the same

87

DMSO concentration. Each treatment had four replicates. At 21 dpf, 25 larvae per replicate were

88

randomly selected and exposed to DDG in 5 L glass aquaria with 3 L of exposure media. The

89

exposure media were renewed daily. Dead and malformed fish in all aquaria were removed during

90

the exposure. At 35 dpf, 9 larvae from each replicate (n = 4) were randomly taken and anesthetized

91

with 0.01% tricaine methane sulfonate (MS-222, Sigma–Aldrich) and were pooled as a composite

92

sample and transferred to RNAlater for later qPCR analysis. At 60 dpf, the larvae from each

93

aquarium were transferred to the 10 L glass tanks each containing 6 L of exposure media to

94

continue exposure until 140 dpf. At 140 dpf, all fish were anesthetized with 0.01% MS-222. The

95

body length (cm) and weight (g) were measured. The plasma samples from three individual fish of

96

the same sex were pooled as a composite sample using glass capillary and pooled as a composite

97

sample. The blood samples were transferred into heparin sodium-rinsed centrifuge tubes. After

98

centrifugation (7,000 g for 5 min at 4 °C), the plasma samples were stored at -80 °C for hormone

99

analysis.

100

All remaining fish were dissected. The sampling time continued for approximately four hours. 5

ACS Paragon Plus Environment

Page 7 of 29

Environmental Science & Technology

101

Two male and one female fish per replicate were randomly taken and dissected out for the gonads.

102

The gonads were fixed in Bouin’s solution for 24 h prior to histological analysis. For RNA

103

extraction, the gonads and brain of three female and four male fish per replicate were pooled and

104

preserved in RNAlater at −80 °C. All experiments were conducted with the approval of the Ethics

105

Committee of South China Normal University for the Care and Use of Laboratory Animals.

106

Chemical Analysis. The exposure concentration of DDG in each aquarium was determined

107

according to our previous method. 13 The concentrations of DDG at two different sampling dates

108

(i.e., 60 dpf for juvenile stage; 140 dpf for adult stage) were measured. The limits of detection and

109

quantification for DDG were 0.05 ng L-1 and 0.18 ng L-1, respectively. The recovery was 101% ±

110

3%. For detailed information about chemical analysis, please refer to the Supporting Information

111

(SI Text S1) and our previous work. 13

112

Microarray Analysis. Microarray analysis was performed to investigate the global

113

transcription profile following the exposure to DDG. Single-color zebrafish oligo microarray in a 4

114

× 44 K slide format (Agilent, design ID 026437) was used in the microarray analysis as described

115

previously. 14 The testis and brain samples stored at RNAlater were sent to Capital-Bio Inc. (Beijing,

116

China) for microarray analysis. In total, 16 microarrays were analyzed for testis or brain (SC, L, M,

117

and H treatments, 4 biological replicates per treatment). Capital-Bio cRNA Amplification and

118

Labeling Kit (CapitalBio, Beijing, China) was used to label the samples with a fluorescent dye (Cy5

119

and Cy3-dCTP) to produce yields of labeled cDNA. Hybridization was performed at 42 °C for

120

overnight in an Agilent hybridization oven with a rotation speed of 20 rpm. The arrays were washed

121

and scanned by Agilent G2565CA Microarray Scanner (Agilent, Santa Clara, USA). 15

122

Quantitative Real-time PCR assay. Details on total RNA extraction, reverse-transcription of

123

RNA into cDNA and qPCR reaction were described in a previous study. 16 Synthesis of cDNA was 6

ACS Paragon Plus Environment

Environmental Science & Technology

Page 8 of 29

124

carried out from 500 ng of total RNA using ReverTra Ace® qPCR RT Master Mix with gDNA

125

Remover (Toyobo, Japan) in a total volume of 50 µL. The qPCR analysis was conducted on the

126

Applied Biosystems ViiATM 7 Dx (ABI) using the THUNDERBIRD SYBR® qPCR Mix (Toyobo,

127

Japan). The intron/exon boundary-spanning primers were preferred to minimize DNA

128

contamination. Primer-BLAST was employed to check the specificity for the primers (Table S1). In

129

order to ensure specificity of the primers, a melting curve analysis (60 - 95 °C) was completed on

130

each sample at the end of qPCR reactions. The ribosomal protein L (RpL13α) and elongation factor

131

1 alpha (Ef1−α) showed high gene expression stabilities after chronic exposure to DDG (SI, Figure

132

S1). The average Ct value (threshold cycle) of the reference genes was employed to normalize the

133

expression of mRNA for the target genes. Relative mRNA expression was determined by the

134

delta-delta CT method. 17

135

Sex Hormone Analysis. The determination of sex hormone concentration was performed as

136

previously described. 18 The detailed information was provided in the Supporting Information (SI

137

Text S1). The concentrations of estradiol (E2) and 11-ketotestosterone (11-KT) were measured

138

using ELISA kits (Cayman Chemical Company, Ann Arbor, MI, USA).

139

Histological Examination. Histological preparation was performed as described in previous

140

studies. 14,19 Briefly, the fixed gonad samples were dehydrated through a graded series of ethanol

141

solutions, and the dehydrated samples were embedded in paraffin. Three longitudinal sections (4

142

µm) from different regions of the ovary and testis were cut through the gonadal region. The sections

143

were stained by hematoxylin-eosin, and examined under a Nikon Eclipse 50 i light microscope

144

(Nikon, Japan). Testis and ovary staging and histopathological alterations were evaluated according

145

to OECD guidelines and previous studies. 14,19,20

146

Data Statistics and Analysis. The microarray data had been submitted to the Gene Expression 7

ACS Paragon Plus Environment

Page 9 of 29

Environmental Science & Technology

147

Omnibus (GEO) database in accordance with the minimum information about a microarray

148

experiment (MIAME) standards with an accession No.GSE116641. GeneSpring software V13

149

(Agilent) was used to analyze the microarray data. Due to the relatively low number of genes from

150

the L and M treatments selected based on the false discovery rate (FDR) analysis (< 0.05, see also

151

the statistical analysis section in Text S2 in SI), differentially expressed genes (DEGs) were

152

selected according to the threshold values (≥ 2) as compared with solvent control and the p value

153

calculated following t-test. 15,21-24 Database for Annotation, Visualization, and Integrated Discovery

154

(DAVID) was used instead of Kyoto Encyclopedia of Genes and Genomes (KEGG) as DAVID can

155

combine the functionally descriptive data with intuitive graphical displays, which can facilitate the

156

interpretation of the DEGs and the determination of significantly gene ontology (GO) categories

157

and biological pathways. 25 The significantly altered biological pathways and GO categories were

158

identified based on the p values (≤ 0.05).

159

Hierarchical clustering (HAC) maps were constructed using the cluster 3.0 Software. The

160

chi-square test was used to examine the differences in the sex ratio between the solvent control and

161

other treatments. One way analysis of variance (ANOVA) was used to analyze the data followed by

162

Tukey multiple comparison tests. Prior to ANOVA analysis, Kolmogorov–Simirnov and Levene’s

163

tests were used to test the assumptions of normality and homogeneity of variances, respectively.

164

The statistical analyses were conducted in SPSS (version 13.0). Data were considered significantly

165

different at p < 0.05.

166 167

RESULTS and DISCUSSION

168

Measured DDG Exposure Concentrations. The concentrations of DDG were measured at the

169

initiation of exposure (T0) and prior to water renewal (T24) during the juvenile and adult stages (See 8

ACS Paragon Plus Environment

Environmental Science & Technology

Page 10 of 29

170

Chemical Analysis section). The measured concentrations of DDG were close to the nominal values

171

at T0, but remained approximately 25% - 30% of their nominal concentrations at T24 (SI Table S2).

172

The average measured concentrations of DDG for 5, 50, and 500 ng L-1 treatments were 3.79, 37.9

173

and 374 ng L-1 during juvenile stage, and 2.97, 28.3, and 285 ng L-1 during adult stage, respectively.

174

For simplicity, the average of the measured concentrations of DDG (i.e., 3.39, 33.1, 329 ng L-1 for

175

the L, M and H treatments, respectively) during juvenile and adult stages was used for the

176

presentation of the data throughout the study. In the present study, the decrease trend and magnitude

177

of DDG concentrations during the exposure for juvenile and adult fish were similar to those

178

observed in other studies on the effects of progestins in zebrafish. 4,12

179

Sex Differentiation and Transcriptional Alterations. Results on the sex ratio of

180

DDG-exposed populations are presented in Figure. 1A. DDG exposure severely disturbed sex

181

differentiation in the H treatment with approximately 98% males. Previous studies have

182

demonstrated that NGT and LNG caused 100% males due to their strong androgenic activity. 7,26 In

183

the present study, DDG affected the sex ratio of the zebrafish populations, leading to a shift towards

184

males. To the best of our knowledge, it is the first time to show that the exposure to DDG resulted

185

in male-biased zebrafish populations.

186

To further understand the possible mechanisms underlying the effects of DDG on sex

187

differentiation, we examined the transcription of genes related to sex differentiation and apoptosis

188

pathway at 35 dpf. The results showed that DDG suppressed the transcription of cyp19a1a

189

(3.16-fold decrease) but stimulated the expression of dmrt1 mRNA (1.83-fold increase) in the H

190

treatment (Figure. 1B). It is generally believed that dmrt1 is predominantly expressed in testis

191

during sex differentiation in fish. 9 The over expression of dmrt1 mRNA would probably lead to

192

differentiation of juvenile ovary into testis, which is consistent with the observed male-biased 9

ACS Paragon Plus Environment

Page 11 of 29

Environmental Science & Technology

193

zebrafish populations in the present study. Moreover, dmrt1 promotes the early formation of testes

194

by suppressing the ovarian developmental pathway via the repression of aromatase transcription and

195

estrogen production in the gonads, 9 which was also evidenced by the low transcription of cyp19a1a

196

in the DDG-exposed zebrafish. Previous studies also demonstrated that the male-biased zebrafish

197

populations caused by exposure to NGT and LNG had low expression of cyp19a1a mRNA. 16,26,27

198

Therefore, the up-regulation of dmrt1 and down-regulation of cyp19a1a at least partially explained

199

the male bias in zebrafish populations chronically exposed to DGG.

200

The suppression of cyp19a1a is most likely to decrease the production of aromatase and

201

estrogen. The depletion of aromatase and estrogen could induce oocyte apoptosis during sex

202

differentiation, therefore resulting in male bias. 10 Caspases (including casp1b, casp9, and casp3a)

203

are a family of enzymes playing major roles in apoptosis. 28,29 In the present study, DDG

204

significantly increased the transcription of casp1b, casp3a and casp9 in the H treatment. It was

205

possible that the up-regulation of casp1b, casp9, and casp3a was closely related to sex

206

determination in zebrafish (Figure 1B). Therefore, it is implied that over expression of dmrt1

207

mRNA suppressed the transcription of cyp19a1a which activated the apoptosis pathway, thereby

208

resulting in enhanced oocyte apoptosis and accelerating the differentiation of the juvenile ovary into

209

testis in zebrafish. 10

210

Blood Plasma Hormone Levels. Chronic exposure to DDG had no significant effect on the

211

plasma concentrations of 17β-estradiol (E2) in females and 11-ketotestosterone (11-KT) in males

212

(SI Figure S5), which were consistent with a previous study. 4 However, several studies have

213

showed that progestins (LNG, norethindrone (NET) and megestrol acetate (MTA)) decreased the

214

plasma hormone levels in fish, which might be due to the differences in disrupting potency of the

215

chemicals and fish species 4,8,18. 10

ACS Paragon Plus Environment

Environmental Science & Technology

216

Page 12 of 29

Global Gene Expression Analysis. Based on the sex ratio results, the brain and testis in male

217

fish were selected for microarray analysis. Volcano plots showed that more DEGs were identified in

218

the H treatment in both the testis and brain (Figure 2A, B). The number of up-regulated DEGs by

219

DDG showed a concentration dependent pattern in the testis. In contrast, the number of

220

up-regulated DEGs by DDG in the brain decreased with increasing DDG concentrations (Figure 2A,

221

B, SI Figure S6).

222

The Venn diagrams depicted the overlap of DEGs and Gene Ontology (GO) at all

223

concentrations (Figure 2C, D). In the testis, there were 6 overlapped DEGs mainly involved in

224

immune responses (mhc1uea and mhc1ufa), endothelial barrier and regulation of cell migration

225

(tnfrsf19) (SI Table S3). Similarly, in the brain, there were 17 overlapped DEGs, which were

226

mainly involved in the rhythmic processes (clocka, antl2, antl2b, cry5, cyr1ab, per1b, nr1d2b,

227

si:ch211-132b12.7, nfil3-5, nfil3-6 and mapk8b) and in responses to chemical stimuli (cyp2ad6,

228

nr1d4b and ddb2) (SI Table S3). Overlapping DEGs were identical to those overlapping GOs both

229

in the brain and testis of male fish.

230

Confirmation by qPCR. The DEGs predicted by microarray analysis were validated by qPCR

231

in both the testis and the brain of male fish (Table 1; SI Table S11). In the testis, the qPCR results

232

of p2rx4b, il1rapl1a and tnfrsf19 were consistent with microarray results, while the fold change of

233

mhc1uea, mhc1ufa and trim66 from the qPCR data was higher than that of the microarray data. The

234

transcriptional alterations of asah1b, casp1b, casp3a, and casp9 were also consistent with the

235

microarray results (Table 1). In the brain, the qPCR results showed the transcriptional levels of

236

clocka, nr1d4b, arntl2, cyp2ad6, cry1ab, cry5, ddb2, nr1d2b, gabrr1 and zgc:112266 were

237

significantly enhanced, which was consistent with the microarray results. The fold change of nfil3-5

238

and si:ch211-132b12.7 by qPCR was slightly higher than that by microarray analysis, while the 11

ACS Paragon Plus Environment

Page 13 of 29

Environmental Science & Technology

239

fold change of nfil3-6, si:ch211-168n16.1 and nfil3-6 by qPCR analysis was relatively lower than

240

that by microarray analysis (Table 1).

241

We further evaluated the transcription of the overlapping DEGs in the ovary and brain of

242

female fish. In the ovary, the suppression of mhc1uea and mhc1ufa was observed in the L treatment,

243

while up-regulation of mhc1uea, mhc1ufa, asah1b and casp3a was observed in the H treatment

244

(Figure 3 A). In the brain, the transcriptional alteration of the overlapping DEGs in females was

245

very similar to that of the males (Figure 3 B). DDG exposure increased transcription of clocka and

246

arntl2, but decreased the transcription of per1b, cry5, nr1d2b and cyp2ad6. Heatmap analysis

247

revealed a significant distinction between brain and gonad (Figure 3 C). The target genes from the

248

brain were clustered on the top, except for tnfrsf19. The genes in females in the M treatment were

249

clustered near the genes of male in the H treatment, indicating that DDG was androgenic to

250

zebrafish and therefore indirectly corroborated the observed male-biased zebrafish populations.

251

Histological Alterations and Functional Genomics Analysis in the Gonads. The

252

histological alterations in gonads are presented in Figure 4. The typical histological sections of

253

gonads of zebrafish in the SC and the H treatment in the testes, and in the SC and M treatment in

254

the ovaries treatment are shown in Figure 4 A-D. Other DDG treatment groups are presented in

255

Figure S3 and S4. Compared with the control gonads, DDG exposure significantly increased the

256

percentage of mature spermatids in the testes in the H treatment, and atretic follicles (AF) in the L

257

and M treatments in the ovaries (Figure 4 E, F).

258

In the present study, GOs related to immune response, endothelial barrier and regulation of cell

259

migration were enriched at all concentrations. A previous study showed 21 days of exposure to

260

progesterone (P4) and drospirenone (DRSP) mainly affected GOs related to the ion channel in the

261

ovary of adult zebrafish, 12 which was different from our results. Endothelial barrier, regulation of 12

ACS Paragon Plus Environment

Environmental Science & Technology

Page 14 of 29

262

cell migration, and phagosome are essential for maintaining the spermatogenesis in the testis. 30,31

263

Phagosome plays important roles in tissue remodeling by engulfing the regressing tissues (e.g., the

264

residual body) during spermatogenesis. 32,33 Defective elimination of residual bodies in mammals is

265

associated with impairment of sperm quantity. 34 Moreover, endothelial barrier is established at the

266

end of meiotic phase, which controls the movement of spermatocytes from the basal to the

267

adluminal compartment via disassembles and reassembles. 31 Therefore, the enriched GOs from the

268

above mentioned three processes in the testes from DDG-exposed zebrafish implied that DDG

269

might influence spermatogenesis.

270

In the testis, interestingly, exposure to DDG increased the percentage of mature spermatids in

271

the H treatment. Similar results were observed in zebrafish exposed to 1263 ng L-1 DDG for 21

272

days.4 In the present study, lysosome, phagosome and sphingolipid metabolism pathways were

273

mostly affected in the H treatment by KEGG analysis (Table 2). Since the mechanisms of the

274

effects of progestins on spermatogenesis in fish are largely unknown, it is assumed that these three

275

pathways are responsible for the alterations of spermatogenesis caused by exposure to DDG based

276

on the following evidence. First, these three pathways are involved in regulation of spermatogenesis,

277

especially for the acrosome biogenesis. 30,35 The acrosome is regarded as a modified lysosome or a

278

novel lysosome-related organelle (LRO). 35 In the present study, microarray analysis showed DDG

279

exposure up-regulated the transcription of neu1, naga and asah1b in the H treatment. Neu1 and

280

naga encode glycosidase which is located on the surface of acrosome and participates in the

281

regulation of spermatogenesis, epididymis maturation and acrosome reaction. 36,37 Meanwhile, the

282

activity of glycosidase encoded by naga in the seminal plasma is considered as an indicator of

283

sperm cell membrane integrity during semen maturation in the reproductive tract of the common

284

carp (Cyprinus carpio). 37 Therefore, the over expression of neu1 and naga mRNA might accelerate 13

ACS Paragon Plus Environment

Page 15 of 29

Environmental Science & Technology

285

the formation of acrosome, resulting in more mature spermatids, as evidenced from our histological

286

observations. Secondly, during acrosome biogenesis, the lysosome fuses with an autophagosome to

287

form LRO. 37 Previous studies showed that interference with autophagosome-lysosome fusion, a

288

key step in acrosome biogenesis, could affect acrosome biogenesis and sperm quality in zebrafish.

289

35,38

In the present study, asah1b and cers2a, two genes involved in the regulation of autophagy,

290

39,40

were identified in the sphingolipid metabolism pathway. In addition, stx17, a gene encoding the

291

protein required for autophagosome-lysosome fusion, 41 was enriched in Golgi transport GOs

292

functioning in regulating the acrosome biogenesis. 35 Its expression was significantly enhanced in

293

the H treatment. Therefore, the elevated expression of stx17 suggested that DDG promoted the

294

autophagosome-lysosome fusion, and accelerated the formation of acrosome biogenesis. Finally,

295

once spermatids are differentiated into spermatozoa, the bulk cytoplasmic contents are segregated

296

into the residual body, which is phagocytosed via the phagosome pathway. 32 In the present study,

297

DDG exposure increased transcription of target genes (e.g., mhc1ufa and fc50e01) involved in the

298

phagosome pathway by microarray analysis. The activation of the phagosome pathway implied a

299

more efficient removal of the residual bodies and a more accelerated production of spermatozoa, as

300

evidenced from our histological observations. Taking together, it is reasonable to conclude that

301

DDG increased the level of sperm maturation via sphingolipid metabolism and lysosome pathways,

302

and enhanced the efficient removal of residual bodies via the phagosome pathway.

303

In the ovary, DDG increased the frequency of AF. The ovarian follicle atresia is an apoptotic

304

process leading to reabsorption of maturing oocytes. 42 The elevated transcription of casp3a in the

305

ovary in all treatments implied that DDG was likely to cause apoptosis, which was supported by the

306

histological data. Moreover, it is known that mhc1uea and mhc1ufa are expressed on the surface of

307

the regression tissues (e.g., the ovarian follicle) and then induce the T cells and macrophages to 14

ACS Paragon Plus Environment

Environmental Science & Technology

Page 16 of 29

308

clear and engulf the regression tissues. 32,33,43 The over expression of mhc1uea and mhc1ufa mRNA

309

implied more removal of regression tissues, indicating more AF occurred, which was supported by

310

the increased frequency of AF. On the contrary, the lower expression of mhc1uea and mhc1ufa in

311

the L treatment implied the less removal of residues, which increases the space stress and induces

312

the apoptosis as well. 43,44 Therefore, the increased frequency of AF was also noticed in the L

313

treatment.

314

Functional Genomics Analysis in Brain. The majority of the overlapping DEGs were

315

involved in circadian rhythm network. Up-regulation of clocka and arntl2 and down-regulation of

316

per1b, cry1ab and nr1d2b were fitted well to the circadian rhythm network in both female and male

317

brains. 45 This was consistent with those from previous studies. 4,12,46 Interestingly,

318

si:ch211-132b12.7 was ascribed to be a negative regulator of circadian rhythm by microarray

319

analysis. The suppressed transcription of si:ch211-132b12.7 was most likely to stimulate the

320

expression of clocka, arntl2 and arntl2b, which was observed in the present study. It is worth noting

321

that the transcriptional expressions of genes in the circadian rhythm were altered even at –3.39 ng

322

L-1, suggesting that circadian rhythm pathways are vulnerable to the DDG exposure, and these

323

genes are sensitive to the exposure to DDG and other progestins in general as well. 4,12,46

324

Circadian rhythm network is vital to mediate crucial cellular and physiological processes, such

325

as cell cycle, metabolism, hormone secretion, and reproduction. 47 For instance, endogenous

326

circadian clocks in hypothalamic−pituitary axis participate in regulating the release of leuthine

327

hormone (LH) from pituitary gonadotropin cells in rodents. 48 In the present study, it seemed that

328

circadian rhythm might affect other physiological processes in zebrafish. The microarray analysis

329

showed two genes (nr1d4a and nr1d4b) involved in both the rhythmic process and in response to

330

steroid hormone were identified after chronic exposure to DDG. 45 Therefore, exposure to DDG 15

ACS Paragon Plus Environment

Page 17 of 29

Environmental Science & Technology

331

might affect the metabolism of steroid hormones via the alterations in the transcriptional genes in

332

the circadian rhythm network in the brain. It is known that steroid hormones play an important role

333

in sex differentiation and gonad development. However, whether the circadian rhythm network

334

affects the reproductive system of teleost requires further investigation. Previous studies suggested

335

disorder of the circadian rhythm affected fish reproduction in zebrafish and Japanese medaka,

336

probably via the HPG-Liver axis. 46,49 In the present study, the HPG axis related pathways were not

337

significantly enriched, and hormone levels in blood plasma were not changed (SI Figure S5).

338

Furthermore, the transcriptional changes of the genes of the circadian rhythm in the brain were not

339

well coordinated with the histological changes in the gonads. Slight transcriptional alteration of

340

genes related to circadian rhythm network was observed at the period of sex differentiation (35 dpf)

341

(SI Figure S10). Future research on the correlation between the circadian rhythm network and fish

342

reproduction is still warranted.

343

Intriguingly, the transcription of cyp2ad6 was stimulated at all concentrations in the brain. Up

344

to now, the catalytic or biological functions of cyp2ad6 have yet to be determined in fish. 50 In the

345

present study, the cyp2ad6 was enriched in the arachidonic acid metabolic process by GO analysis.

346

It is showed that cyp2ad6 could have similar catalytic functions to cyp2j2 which is important in the

347

oxidation of arachidonic acid in human, 51 and this was manifested by our GO analysis. The

348

transcriptional alteration of cyp2ad6 implied that DDG might influence arachidonic acid

349

metabolism in zebrafish.

350

Our results showed that exposure to DDG induced multiple transcriptional responses, caused

351

male bias and affected the gonadal development in fish. Progestins such as DDG, NGT and LNG

352

cause male bias and have similar effects on the transcription of dmrt1 and cyp19a1a during sex

353

differentiation in zebrafish but with different potentials. These progestins may simultaneously exist 16

ACS Paragon Plus Environment

Environmental Science & Technology

354

in contaminated aquatic environments. The effects (for example, on the endocrine system) of the

355

mixture of progestins in natural fish populations deserve further research.

Page 18 of 29

356 357

ASSOCIATED CONTENT

358

Supporting Information. The detail materials and methods (Text S1), statistical analyses

359

(Text S2), primer sequences (Table S1), measured concentrations (Table S2), overlapping GOs and

360

DEGs (Table S3), the period of exposure, endpoints and number of replicates (Table S4),,

361

overlapping GOs (Table S5-S10), confirmation of the DEGs (Table S11), top 10 pathways at three

362

concentrations (Table S12-17), list of DEGs (p < 0.05; FC ≥2) at three concentrations (Table

363

S18-23), stability analysis of RpL13a and EF1-α (Figure S1), condition factors and gonadosomatic

364

index at 140 dpf (Figure S2), typical histological sections of gonad (Figure S3, 4), plasma

365

concentrations (Figure S5), number of DEGs in testis and brain (Figure S6), transcriptional

366

alteration of target genes in brain and gonad (Figure S7-8), schematic representations of the

367

spermatid mature (Figure S9), transcriptional alteration of genes in circadian rhythm network at 35

368

dpf (Figure S10).

369 370

ACKNOWLEDGEMENTS

371

The authors would like to acknowledge the financial support from the National Natural Science

372

Foundation of China (U1401235 and 41273119) and National Water Pollution Control Program

373

(2014ZX07206-005), as well as Guangdong Provincial Key Research Program (2015B020235012)

374

and Guangdong Natural Science Foundation (2015A030313738).

375 376

Notes 17

ACS Paragon Plus Environment

Page 19 of 29

377

Environmental Science & Technology

The authors declare no competing financial interest.

378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418

REFERENCES (1) Rižner, T. L.; Brožič, P.; Doucette, C.; Tirek-Etienne, T.; Mueller-Vieira, U.; Sonneveld, E.; van der Burg, B.; Böcker, C.; Husen, B. Selectivity and potency of the retro progesterone dydrogesterone in vitro. Steroids 2011, 76 (6), 607−15. (2) Fent, K. Progestins as endocrine disrupters in aquatic ecosystems: Concentrations, effects and risk assessment. Environ. Int. 2015, 84, 115–130. (3) Runnalls T. J.; Margiotta-Casaluci, L.; Kugathas, S.; Sumpter, J. P. Pharmaceuticals in the aquatic environment: steroids and anti-steroids as high priorities for research. Hum. Ecol. Risk. Assess. 2010, 16, 1318–1338. (4) Zhao, Y.; Castiglioni, S.; Fent, K. Synthetic progestins medroxyprogesterone acetate and dydrogesterone and their binary mixtures adversely affect reproduction and lead to histological and transcriptional alterations in zebrafish (Danio rerio). Environ. Sci. Technol. 2015, 49 (7), 4636–4645. (5) Liu, S. S.; Ying G. G.; Liu, Y. S.; Yang, Y. Y.; He, L. Y.; Chen, J.; Liu, W. R.; Zhao, J. L. Occurrence and removal of progestagens in two representative swine farms: effectiveness of lagoon and digester treatment. Water. Res. 2015, 77, 146–154. (6) Available from: https://www.medicines.org.uk/EMC/medicine/2493/SPC/Femoston+1+10mg/. Electronic Medicines Compendium, Prescription only medicine, dydrogesterone estradiol hemihydrates, 1995, Mylan Products Ltd. United Kingdom. (7) Brockmeier, E. K.; Scott, P. D.; Denslow, N. D.; Leusch, F. D. Transcriptomic and physiological changes in Eastern Mosquitofish (Gambusia holbrooki) after exposure to progestins and anti-progestagens. Aquat Toxicol. 2016,179, 8-17. (8) Runnalls, T. J.; Beresford, N.; Losty, E.; Scott, A. P.; Sumpter, J. P. Several synthetic progestins with different potencies adversely affect reproduction of fish. Environ. Sci. Technol. 2013, 47 (4), 2077−2084. (9) Von Hofsten, J.; Olsson, P. E. Zebrafish sex determination and differentiation: involvement of FTZ-F1 genes. Reprod. Biol. Endocrinol. 2005, 3, 63. (10) Uchida, D.; Yamashita, M.; Kitano, T.; Iguchi, T. Oocyte apoptosis during the transition from ovary-like tissue to testes during sex differentiation of juvenile zebrafish. J. Exp. Biol. 2002, 205, 711–718. (11) Luzio, A.; Matos, M.; Santos, D.; Fontaínhas-Fernandes, A. A.; Monteiro, S. M.; Coimbra, A. M. Disruption of apoptosis pathways involved in zebrafish gonad differentiation by 17α-ethinylestradiol and fadrozole exposures. Aquat. Toxicol. 2016, 177, 269–284. (12) Zucchi, S.; Mirbahai, L.; Castiglioni, S.; Fent, K. Transcriptional and physiological responses induced by binary mixtures of drospirenone and progesterone in zebrafish (Danio rerio) at environmental concentrations. Environ. Sci. Technol. 2014, 48 (6), 3523−3531. (13) Liu, S. S.; Ying, G. G.; Liu, S.; Lai, H. J.; Chen, Z. F.; Pan, C. G.; Zhao, J. L.; Chen, J. Analysis of 21 progestagens in various matrices by ultra–high–performance liquid chromatography tandem mass spectrometry (UHPLC-MS/MS) with diverse sample pretreatment. Anal. Bioanal. Chem. 2014, 406, 7299–7311. (14) Shi, W. J.; Zhao, J. L.; Jiang, Y. X.; Huang, G. Y.; Liu, Y. S.; Zhang, J. N.; Ying, G. G. Transcriptional and histological alterations in gonad of adult zebrafish after exposure to the synthetic progestin norgestrel. Environ. Toxicol. Chem. 2017, 36 (12), 3267–3276. (15) Ørom, U. A.; Derrien, T.; Beringer, M.; Gumireddy, K.; Gardini, A.; Bussotti, G. Long noncoding RNAs 18

ACS Paragon Plus Environment

Environmental Science & Technology

Page 20 of 29

419 420

with enhancer−like function in human cells. Cell 2010, 143, 46–58. (16) Liang, Y.Q.; Huang, G.Y.; Liu, S.S.; Zhao, J.L.; Yang, Y.Y.; Chen, X.W.; Tian, F.; Jiang, Y.X.; Ying, G.G.

421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463

Long−term exposure to environmentally relevant concentrations of progesterone and norgestrel affects sex differentiation in zebrafish (Danio rerio). Aquat Toxicol. 2015, 160, 172–179. (17) Livak, K. J.; Schmittgen, T. D. Analysis of relative gene expression data using real−time quantitative PCR and the 2−∆∆ Ct method. Methods 2001, 25, 402–408. (18) Hua, J.; Han, J.; Wang, X.; Guo, Y.; Zhou, B. S. The binary mixtures of megestrol acetate and 17α-ethinylestradiol adversely affect zebrafish reproduction. Environ. Pollut. 2016, 213, 776-784. (19) Silva, P.; Rocha, M. J.; Cruzeiro, C.; Malhão, F.; Reis, B.; Urbatzka, R.; Monteiro, R. A. F.; Rocha, E. Testing the effects of ethinylestradiol and of an environmentally relevant mixture of xenoestrogens as found in the Douro River (Portugal) on the maturation of fish gonads-a stereological study using the zebrafish (Danio rerio) as model. Aquat. Toxicol. 2012, 124, 1–10. (20) Johnson, R.; Wolf, J.; Braunbeck, T. Guidance Document on the diagnosis of endocrine-related histopathology in fish gonads. OECD, 2009, Paris, France: 5–95. (21) Patterson, T. A.; Lobenhofer, E. K.; Fulmer-Smentek, S. B.; Collins, P. J.; Chu, T. M.; Bao, W. J.; Fang, H.; Kawasaki, E. S.; Hager, J.; Tikhonova, I. R.; Walker, S. T.; Zhang, L.; Hurban, P.; De Longueville, M.; Fuscoe, J. C.; Tong, W.; Shi, L.; Wolfinger, R. D. Performance comparison of one-color and two-color platforms within the microarray quality control (MAQC) project. Nat. Biotechnol. 2006, 24, 1140–1150. (22) Yan, S. H.; Wang, M.; Zha0, J. M.; Zhu, L. F.; Li, W.; Luo, Q.; Sun, J.; Wang, Z. J. Environmentally relevant concentrations of carbamazepine caused endocrine-disrupting effects on nontarget organisms, Chinese Rare Minnows (Gobiocypris rarus). Environ. Sci. Technol. 2018, 52(2), 886–894. (23) Ray, A.; Bhaduri, A.; Srivastava, N.; Mazumder, S. Identification of novel signature genes attesting arsenic-induced immune alterations in adult zebrafish (Danio rerio). J. Hazard. Mater. 2017, 321, 121–131. (24) Zhang, S.; Cao, J. Close examination of double filtering with fold change and t test in microarray analysis. BMC bioinformatics 2009, 10(1): 402. (25) Dennis, J. G.; Sherman, B. T.; Hosack, D. A.; Yang, J.; Gao, W.; Lane, H. C. DAVID: database for annotation, visualization, and integrated discovery. Genome. Biol. 2003, 4, R60.1–R60.11. (26) Svensson, J.; Mustafa, A.; Fick, J.; Schmitz, M.; Brunström, B. Developmental exposure to progestins causes male bias and precocious puberty in zebrafish (Danio rerio). Aquat. Toxicol. 2016, 177, 316–323. (27) Han, J.; Wang, Q. W.; Wang, X. F.; Li, Y. G.; Wen, S.; Liu, S.; Ying, G. G.; Guo, Y. Y.; Zhou, B. S. The synthetic progestin megestrol acetate adversely affects zebrafish reproduction. Aquat Toxicol. 2014, 150, 66–72. (28) Karami, A.; Groman, D. B.; Wilson, S. P.; Ismail, P.; Neela, V. K. Biomarker responses in zebrafish (Danio rerio) larvae exposed to pristine low-density polyethylene fragments. Environ. Pollut. 2017, 223, 466–475. (29) Kuida, K.; Haydar, T. F.; Kuan, C. Y.; Gu, Y.; Taya, C.; Karasuyama, H.; Su, M. S. S.; Rakic, P.; Flavell, R. A. Reduced apoptosis and cytochrome c–mediated caspase activation in mice lacking caspase 9. Cell 1998, 94(3), 325–337. (30) Yoshida, S. From cyst to tubule: innovations in vertebrate spermatogenesis. WIREs. Dev. Biol. 2016, 5, 119–131. (31) Leal, M. C.; Cardoso, E. R.; Nóbrega, R. H.; Batlouni, S. R.; Bogerd, J.; França, L. R.; Schulz, R. W. Histological and stereological evaluation of zebrafish (Danio rerio) spermatogenesis with an emphasis on spermatogonia generations. Biol. Reprod. 2009, 81(1), 177–187. (32) Tang, E. I.; Lee, W. M.; Cheng, C. Y. Coordination of actin-and microtubule-based cytoskeletons supports transport of spermatids and residual bodies/phagosomes during spermatogenesis in the rat testis. Endocrinology 2016, 1, 47–62. (33) Stuart, L. M.; Ezekowitz, R. A. B. Phagocytosis: elegant complexity. Immunity 2005, 22(5), 539–550. 19

ACS Paragon Plus Environment

Page 21 of 29

464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508

Environmental Science & Technology

(34) Huang, J.; Wang, H.; Chen, Y.; Wang, X.; Zhang, H. Residual body removal during spermatogenesis in C. elegans requires genes that mediate cell corpse clearance. Development 2012, 139(24), 4613–4622. (35) Berruti, G.; Paiardi, C. Acrosome biogenesis: revisiting old questions to yield new insights. Spermatogenesis 2011, 1, 95–98. (36) Ma, F.; Wu, D.; Deng, L. W.; Secrest, P.; Zhao, June.; Varki, Nissi.; Lindheim, Steven.; Gagneux, Pascal. Sialidases on mammalian sperm mediate deciduous sialylation during capacitation. J. Biol. Chem. 2012, 287 (45), 38073–38079. (37) Cejko, B. I.; Sarosiek, B.; Krejszef, S.; Kowalski, R. K. Multiple collections of common carp Cyprinus carpio L. semen during the reproductive period and its effects on sperm quality. Anim. Reprod. Sci. 2018, 188: 178–188. (38) Zhang, J.; Zhang, X M.; Liu, Y. J.; Su, Z. H.; Dawar, F. U.; Dan, Hong.; He, Y.; Gui, J. F.; Mei, J. Leucine mediates autophagosome-lysosome fusion and improves sperm motility by activating the PI3K/Akt pathway. Oncotarget 2017, 8 (67), 111807–111818. (39) Martens, S.; Nakamura, S.; Yoshimori, T. Phospholipids in autophagosome formation and fusion. J. Mol. Boil. 2016, 428(24), 4819–4827. (40) Young, M. M.; Kester, M.; Wang, H. G. Sphingolipids: regulators of crosstalk between apoptosis and autophagy. J. Lipid. Res. 2013, 54(1), 5–19. (41) Itakura, E.; Kishi-Itakura, C.; Mizushima, N. The hairpin-type tail-anchored SNARE syntaxin 17 targets to autophagosome for fusion with endosomes/lysosomes. Cell 2012, 151(6), 1256–1269. (42) Blüthgena, N.; Castiglioni, S.; Sumpter, J. P.; Fent, K. Effects of low concentrations of the antiprogestin mifepristone (RU486) in adults and embryos of zebrafish (Danio rerio): 1. Reproductive and early developmental effects. Aquat. Toxicol. 2013, 144, 83–95. (43) Xu, X.; Zhao, X.; Lu, L.; Duan, X.; Qin, H.; Du, X.; Li G. Q.; Tao, Z. G.; Zhong S. L.; Wang, G. Transcriptomic analysis of different stages of pigeon ovaries by RNA-sequencing. Mol. Reprod. Dev. 2016, 83(7), 640–648. (44) Huang, J.; Wang, H.; Chen, Y.; Wang, X.; Zhang, H. Residual body removal during spermatogenesis in C. elegans requires genes that mediate cell corpse clearance. Development 2012, 139(24), 4613–4622. (45) Ko, C. H.; Takahashi, J. S. Molecular components of the mammalian circadian clock. Hum. Mol. Genet. 2006, 15 (2), 271−277. (46) Zhao, Y. B.; Castiglioni, S.; Fent, K. Environmental progestins progesterone and drospirenone alter the circadian rhythm network in zebrafish (Danio rerio). Environ. Sci. Technol. 2015, 49 (16), 10155–10164. (47) Gachon, F.; Nagoshi, E.; Brown, S. A.; Ripperger, J.; Schibler, U. The mammalian circadian timing system: From gene expression to physiology. Chromosoma 2004, 113 (3), 103−112. (48) Cano, P.; Jiménez-Ortega, V.; Larrad, Á.; Toso, C. F. R.; Cardinali, D. P.; Esquifino, A. I. Effect of a high-fat diet on 24-h pattern of circulating levels of prolactin, luteinizing hormone, testosterone, corticosterone, thyroid-stimulating hormone and glucose, and pineal melatonin content, in rats. Endocrine 2008, 33(2), 118−125. (49) Zhang, X. W.; Hecker, M.; Jones, P. D.; John, N.; Au, D.; Kong, R.; Wu, R. S. S.; Giesy, J. P. Responses of the medaka HPG axis PCR array and reproduction to prochloraz and ketoconazole. Environ. Sci. Technol. 2008, 42 (17), 6762–6769. (50) Goldstone, J. V.; McArthur, A. G.; Kubota, A.; Zanette, J.; Parente, T.; Jönsson, M. E.; . Nelson, D. R.; Stegeman, J. J. Identification and developmental expression of the full complement of Cytochrome P450 genes in zebrafish. BMC Genomics 2010, 11(1), 643. (51) Arnold, W. R.; Baylon, J. L.; Tajkhorshid, E.; Das, A. Asymmetric binding and metabolism of polyunsaturated fatty acids (PUFAs) by CYP2J2 epoxygenase. Biochemistry 2016, 55(50), 6969−6980.

20

ACS Paragon Plus Environment

Environmental Science & Technology

Page 22 of 29

509

Table 1. Confirmation of the differentially expressed genes in brain and testis of male

510

zebrafish by qPCR. Fold change a Organ

Gene

3.39 ng L-1 vs SC b

33.1 ng L-1 vs SC

329 ng L-1 vs SC

qPCR

Micro array

qPCR

Micro array

qPCR

Micro array

Testis

mhc1uea mhc1ufa p2rx4b il1rapl1a tnfrsf19 asah1b casp1b casp3a casp9

-10.71*** -9.75*** -2.20*** -1.24* 1.51** 0.50 -0.66 -0.38 0.30

-7.07* -7.76* -1.96* -1.13** 1.89* 0.40 -0.48 -0.56 0.14

-9.00*** -8.50*** -1.22* -1.20* 1.47** 0.21 -0.73 0.49 -0.36

-6.48* -7.06* -1.51** -1.05** 1.05* 0.63 -0.19 0.19 -0.35

1.82* 1.60* -0.73 -0.67 1.72** 1.10* -0.62 0.65 0.53

1.50** 1.16* -1.10* -1.28*** 1.11* 1.04** -0.22 0.25 0.07

Brain

clocka nr1d4b arntl2 cry5 cyp2ad6 per1b si:ch211-13 2b12.7 zgc:112266 ddb2

1.71*** c 3.56*** 2.16*** -1.56* -1.76** -2.07***

1.42** 3.88** 1.78*** -1.50** -1.82** -1.73**

1.90*** 2.92*** 1.76** -1.87*** -0.85 -2.11***

1.39* 3.23** 1.91* -1.62** -1.17*** -2.39*

2.23*** 4.85*** 2.31*** -1.59* -1.25* -2.13***

1.74** 4.53*** 2.07*** -1.65*** -1.76*** -2.06***

-2.23**

-1.58**

-1.92**

-1.39*

-2.70***

-1.86**

-1.98** -1.38*

-1.84* -1.40*

-1.42* -1.85***

-2.00* -1.54***

-2.31** -1.18*

-2.74** -1.37***

511

a

512

2−∆∆CT method and ratio of array spot intensity, respectively;

513

b

SC: solvent control;

514

c

Data are shown as mean of four replicates;

515

* p < 0.05, ** p < 0.01, and *** p < 0.001.

Fold changes (log2) of qPCR and microarray compared to solvent controls was determined by

516

21

ACS Paragon Plus Environment

Page 23 of 29

Environmental Science & Technology

517

Table 2. The top 10 affected signaling pathways in testis of zebrafish from the H treatment as

518

compared to the solvent control. Signaling Pathway

FDR a

p value

DEGs b

0.62989

actb2, hbl4, sec61al2, fc50e01, mhc1uea, mhc1uxa2.c tubb1.d

0.62989

neu1, cers2a, asah1b. smpd3.

0.62989

neu1, naga, ctsc, fc50e01, asah1b, cd164.

0.62989

aldh7a1.

0.06196

0.70939

adrb3a. ltc4s, cyp2p9, ggt1l2.2.

Calcium signaling pathway

0.07089

0.70939

mylk3, tnnc1b. p2rx4b, cacna1i, adrb2b, pdgfra, mylk4b.

Biosynthesis of amino acids

0.07273

0.70939

cth, gapdh, aldh7a1. gpt2l.

Cell adhesion molecules (CAMs)

0.07821

0.70939

mhc1uea, lrrc4.1, mpz, mhc1uxa2. cntn1a.

Neuroactive ligand-receptor interaction

0.08800

0.70939

adrb3a, c5ar1. p2rx4b, gria3a, vipr1b, nmur1, chrm4a, adrb2b.

Nicotinate and nicotinamide metabolism

0.10005

0.70939

enpp1, nt5c2b.

Phagosome

0.00649

Sphingolipid metabolism

0.02354

Lysosome

0.03485

Lysine biosynthesis

0.03669

Arachidonic acid metabolism

**

*

* *

519

* p < 0.05 and ** p < 0.01;

520

a

FDR: False discovery rate;

521

b

DEGs: Differentially expressed genes;

522

c

Red color: up-regulation;

523

d

Green color: down-regulation.

524 525

22

ACS Paragon Plus Environment

Environmental Science & Technology

Page 24 of 29

526

Figure Captions

527

Figure 1. Sex differentiation and transcriptional alterations. (A) The sex ratio of zebrafish after

528

exposure to dydrogesterone (DDG) from 21 dpf to 140 dpf. The sex of adult fish was determined by

529

the morphology of its gonad. N represents the total number of fish from four replicate aquariums in

530

each treatment. Abbreviations: SC: solvent control; L: 3.39 ng L-1; M: 33.1 ng L-1; H: 329 ng L-1. (B)

531

Transcriptional alterations of dmrt1, cyp19a1a, foxl2, casp1b, casp3a, casp9, esr and ar in juvenile

532

zebrafish after 14 days (from 21 to 35 dpf) of exposure to DDG compared to solvent control. Data

533

are shown as mean ± SD of four replicates. * p < 0.05, ** p < 0.01, and *** p < 0.001. L: 3.39 ng

534

L-1; M: 33.1 ng L-1; H: 329 ng L-1.

535 536

Figure 2. Multivariate analysis of the transcriptomics data. (A-B) Volcano plots depicting fold

537

change (log2, x-axis) and statistical significance (–log10 p value, y-axis) in the testis (A) and brain

538

(B) of male zebrafish. The fold change cut-off is +/− 1 and −log10 p value cut-off is 1. The upper

539

corners of the plot represent the genes that show both statistical significance and large fold changes.

540

Red dots: significant up-regulation; Green dots: significant down-regulation; Black dots: no

541

significant alteration. (C) Venn diagram of the overlapping DEGs (C1) and GOs (C2) at all

542

concentrations in the testis compared to solvent control. (D) Venn diagram of the overlapping

543

DEGs (D1) and GOs (D2) at all concentrations in the brain of male zebrafish compared to solvent

544

control. One hundred percent (100%) stands for the sum of all the numbers in the pie charts.

545

Abbreviations: Key: SC: solvent control; L: 3.39 ng L-1; M: 33.1 ng L-1; H: 329 ng L-1.

546 547

Figure 3. The qPCR analysis of DEGs in females. (A, B) Transcriptional alteration of target

548

genes in the ovary (A) and brain of females (B). Data are shown as mean ± SD of four replicates. * 23

ACS Paragon Plus Environment

Page 25 of 29

Environmental Science & Technology

549

p < 0.05, ** p < 0.01, and *** p < 0.001. Abbreviations: Key: SC: solvent control; L: 3.39 ng L-1;

550

M: 33.1 ng L-1; H: 329 ng L-1. (C) Hierarchical clustering analysis of qPCR data in the brain and

551

gonads of females (F-) and males (M-). L: 3.39 ng L-1; M: 33.1 ng L-1; H: 329 ng L-1. Due to

552

insufficient number of females in the H treatment (male-biased populations), no data on qPCR are

553

available for females.

554 555

Figure 4. Histological alterations in the gonads at 140 dpf. (A, B) Typical histological sections

556

of testis of adult male zebrafish in SC (A) and H treatment (329 ng L-1 of DDG) (B). Examples of

557

spermatozoa maturation stages include Sg (spermatogonia), Sc (spermatocytes), St (spermatids) and

558

Sz (spermatozoa). (C, D) Typical histological sections of ovary of adult female zebrafish in SC (C)

559

and M treatment (33.1 ng L-1) (D). Examples of oocyte developmental stages include PO

560

(perinucleolar oocyte), CO (cortical alveolar oocyte), EV (early vitellogenic oocyte), LV (Mid-late

561

and Late- vitellogenic oocyte), POF (postovulatory follicles) and AF (atretic follicles). (E)

562

Percentage of mature (MA) and immature (IMA) spermatocytes in the testis of zebrafish at 140 dpf.

563

MA includes St and Sz, and IMA includes Sg and Sc. (F) Percentage of oocytes at different

564

developmental stages in the ovary of zebrafish at 140 dpf. Bar from top to bottom refers to AF, POF,

565

LV, EV, CO and PO. Data are shown as mean values of four replicates. * p < 0.05, ** p < 0.01, and

566

*** p < 0.001. Abbreviations: SC: solvent control; L: 3.39 ng L-1; M: 33.1 ng L-1; H: 329 ng L-1.

567

24

ACS Paragon Plus Environment

Environmental Science & Technology

Figure 1

%

A 1.0

Male Female

0.5

N=49 N=54 N=51 *** 0.0 N=53 SC H M L 570 571

B4 Fold Change

568 569

Page 26 of 29

2

SC L M ** H

**

** **

0 -2 ***

-4 a 1 2 b a 9 r r 1 rt xl 1 3 sp es a 19a dm fo casp casp ca p cy

25

ACS Paragon Plus Environment

Page 27 of 29

572 573

Environmental Science & Technology

Figure 2

574 575

26

ACS Paragon Plus Environment

Environmental Science & Technology

576 577

Page 28 of 29

Figure 3

578 579

27

ACS Paragon Plus Environment

Page 29 of 29

580 581

Environmental Science & Technology

Figure 4

582 583

28

ACS Paragon Plus Environment