Effects of in Utero PFOS Exposure on Transcriptome, Lipidome, and

Jun 27, 2017 - A Reduced Transcriptome Approach to Assess Environmental Toxicants Using Zebrafish Embryo Test. Environmental Science & Technology...
4 downloads 3 Views 381KB Size
Subscriber access provided by Arizona State University Libraries

Article

Effects of In Utero PFOS Exposure on Transcriptome, Lipidome and Function of Mouse Testis Keng Po Lai, Jetty Chung-Yung Lee, Hin Ting Wan, Jing Woei Li, Aman YiMan Wong, Ting-Fung Chan, Camille Oger, Jean-Marie Galano, Thierry Durand, Kin Sum Leung, Cherry C. Leung, Rong Li, and Chris Kong-Chu Wong Environ. Sci. Technol., Just Accepted Manuscript • DOI: 10.1021/acs.est.7b02102 • Publication Date (Web): 27 Jun 2017 Downloaded from http://pubs.acs.org on June 29, 2017

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

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

Page 1 of 31

Environmental Science & Technology

1 2 3 4

6

Effects of In Utero PFOS Exposure on Transcriptome, Lipidome and Function of Mouse Testis.

7

Short title: Omics Analysis of PFOS Effects on Testis.

5

8 Keng Po Lai 2#, Jetty Chung-Yung Lee3#, Hin Ting Wan1, Jing Woei Li 4, Aman Yi-Man Wong1, Ting Fung Chan4, Camille Oger5, Jean-Marie Galano5, Thierry Durand5, Kin Sum Leung3, Cherry C Leung1, Rong Li1, Chris Kong-Chu Wong1*

9 10 11 12 13 14 15 16

1

Partner State Key Laboratory of Environmental and Biological Analysis, Croucher Institute for Environmental Sciences, Department of Biology, Hong Kong Baptist University, 2Department of Biology and Chemistry, City University of Hong Kong, 3School of Biological Sciences, The University of Hong Kong, 4School of Life Sciences, The Chinese University of Hong Kong, 5Institut des Biomolécules Max Mousseron, UMR 5247 CNRS, ENSCM, Université de Montpellier, France

17 18 19 20 21 22 23 24 25 26 27

#

28 29 30 31 32

The authors declare they have no actual or potential competing financial interests.

Equal contribution

*Corresponding author, Dr Chris KC Wong Address: Partner State Key Laboratory of Environmental and Biological Analysis, Croucher Institute for Environmental Sciences, Department of Biology, Hong Kong Baptist University. Phone: (852) 34117053 Email address: [email protected].

Keywords: Lipid mediators, steroidogenesis; epididymal sperm.

1 ACS Paragon Plus Environment

Environmental Science & Technology

33

Abstract

F0

(1) Chemical & physiological analyses

P1 Prenatal PFOS exposure

34

Page 2 of 31

(2) Transcriptomic & lipidomic analyses

F1

P63 (3) Assess the reproductive parameters

1. Alteration of lipid metabolisms, oxidative stress and cell junction signaling; 2. Perturbations of lipid mediators; 3. Reductions in serum testosterone & epididymal sperm count.

35

Transcriptomic and LC-MS/MS-based targeted lipidomic analyses were conducted to identify

36

the effects of in utero PFOS exposure on neonatal testes and its relation to testicular

37

dysfunction in adult offspring. Pregnant mice were orally administered 0.3 and 3 µg PFOS/g

38

body weight until term. Neonatal testes (P1) were collected for the detection of PFOS, and

39

were subjected to omics study. Integrated pathway analyses using DAVID, KEGG and IPA

40

underlined the effects of PFOS exposure on lipid metabolism, oxidative stress and cell

41

junction signaling in testes. LC-MS/MS analysis showed that the levels of adrenic acid and

42

docosahexaenoic acid (DHA) in testes were significantly reduced in the PFOS treatment

43

groups. A significant linear decreasing trend in eicosapentaenoic acid and DHA with PFOS

44

concentrations was observed. Moreover, LOX-mediated 5-hydroxyeicosatetraenoic acids

45

(HETE) and 15-HETE from arachidonic acid in the testes were significantly elevated and a

46

linear increasing trend of 15-HETE concentrations was detected with doses of PFOS. The

47

perturbations of lipid mediators suggested that PFOS has potential negative impacts on

48

testicular functions. Postnatal analysis of male offspring at P63 showed significant reductions

49

in serum testosterone and epididymal sperm count. This study sheds light into the as yet

50

unrevealed action of PFOS on lipid mediators in affecting testicular functions.

51

2 ACS Paragon Plus Environment

Page 3 of 31

Environmental Science & Technology

52

Introduction

53

The incidence of reproductive problems is a globally prevalent issue. Recent census data

54

from the World Bank show a continuous decline of global average fertility rate from >5.0 in

55

1960 to 0.3

206

and FDR < 0.05 were considered as differentially expressed genes (DEGs). The DEGs were

207

subjected to GO functional enrichment analysis and Kyoto Encyclopedia of Genes and

208

Genomes (KEGG) analysis using The Database for Annotation, Visualization and Integrated

209

Discovery (DAVID) v6.8.53,54 Furthermore, Ingenuity Pathway Analysis (IPA®, QIAGEN

210

Redwood City, www.qiagen.com/ingenuity) was used to decipher the molecular interaction

211

networks that are deregulated by in utero PFOS treatment. Canonical pathways with P < 0.05

212

were considered as statistically significant.

213

214

Quantitative PCR analysis

215

Fetal testes (four dam per group) were used for verification. Reverse transcription was

216

conducted using the SuperScript™ VILO™ cDNA Synthesis Kit (ThermoFisher). qPCR was

217

performed using SYBR FAST qPCR Master Mix (Kapa Biosystems, Woburn, MA) on a

9 ACS Paragon Plus Environment

Environmental Science & Technology

Page 10 of 31

218

StepOnePlus Real-Time PCR System (Life Technologies). It was performed with a 3 min

219

initial denaturation step at 95°C followed by 40 cycles of 95°C for 5 s and 60°C for 20 s. The

220

GAPDH housekeeping gene was used for internal normalization for each gene. To calculate

221

gene expression, the comparative Ct method (∆∆Ct) was used. The following gene-specific

222

primers were used: GAPDH-forward, 5'-gggttcctataaatacggactgc-3; GAPDH-reverse, 5'-

223

ccattttgtctacgggacga-3'; SPTBN1-forward, 5'-atctgccagcacccagagta-3'; SPTBN1-reverse, 5'-

224

acgctggtgggtaaggtct-3';

225

catccgtggaaaacaccag-3'; MAPK9-forward, 5'-acacctgtctatggcttcagg-3'; and MAPK9-reverse,

226

5'-ctgatgcactgtgggacttc-3';

227

reverse, 5'-ggcaaaattacaaccgacca-3; SCD2-forward, 5'-agctggtgatgttccagagg-3'; SCD2-reverse,

228

5'-caagaaggtgctaacgcaca-3'; HDS17B12–forward, 5'-ctttccgtttgcaaggtgac-3'; HSD17B12-

229

reverse,

230

CTNNB1-reverse, 5'-aagaacggtagctgggatca-3'; APPL2-forward, 5'-cccacggacagatcaacttt-3';

231

APPL2-reverse, 5'-tccagttccacctgaatgc-3'; OXTR-forward, 5'-gttctcaaccatcctcggca-3'; OXTR-

232

reverse, 5'-gttctcaaccatcctcggca-3'.

ITGB1-forward,

5'-atgcaggttgcggtttgt-3';

PIK3R1-forward,

5'-tgttgagaatcacccctttagat-3';

ITGB1-reverse,

5'-ataaactaaagttggtcttttgacgag-3';

CTNNB1-forward,

5'-

PIK3R1-

5'-tgcagatcttggactggacat-3';

233

234

Lipidomic Analysis of Neonatal testes

235

At P1, the neonate testes from 4 dams (n = 4) per treatment group were collected. The lipid

236

portion was extracted from the testes samples which were stored in -80ºC following the Lee et

237

al. method with modifications.55 In brief, the tissues (~0.1 g) were homogenized in 10 mL of

238

ice-cold Folch solution (chloroform/methanol 2:1 v/v + 0.05% BHT) using a blade

239

homogenizer (T25, ULTRA-TURRAX, IKA) at 24,000 rpm in 2×20 s bursts. Afterwards, 2

240

mL of 0.9% NaCl solution were added into samples to generate phase separation. After

241

centrifugation at 3000 x g for 10 min at 4ºC, the lower chloroform phase was collected into a

242

glass vial and dried under stream of nitrogen gas. The dried samples were re-suspended in 1 10 ACS Paragon Plus Environment

Page 11 of 31

Environmental Science & Technology

243

mL of 1 M KOH in methanol and 1 mL of PBS (pH 7.4) and then hydrolyzed overnight in the

244

dark, at room temperature on an orbital shaker. Hydrolysis was terminated by the addition of

245

hydrochloric acid and formic acid. Finally, the samples were purified by solid phase

246

extraction (SPE) using 60 mg mixed anionic SPE cartridges (MAX Oasis, Waters, USA). The

247

SPE was cleaned and conditioned with methanol and formic acid respectively. The sample

248

prepared were then loaded on the SPE, cleaned with 2% ammonium hydroxide and hexane,

249

and the final elute (hexane/ethanol/acetic acid solution) was dried under a stream of nitrogen

250

gas and the extracts were reconstituted in 100 µL of acetonitrile for the analysis of

251

polyunsaturated fatty acids (PUFAs) and their oxidized lipid products. The extracted lipid

252

products were quantified by liquid chromatography tandem mass spectrometry (LC-

253

MS/MS)56 using Kinetex C18 column (150 × 21 mm, 2.6 µm, Phenomenex, USA)

254

maintained at 30ºC in the 1290 Infinity LC system (Agilent Technologies, USA).

255

A QTrap 3200 triple quadrupole mass spectrometer (Sciex Applied Biosystems, MA, USA)

256

was operated in a negative atmospheric pressure chemical ionization (APCI) mode with the

257

source temperature set at 750ºC. The mobile phase included Milli-Q water with 0.1% formic

258

acid (A) and acetonitrile with 0.1% formic acid (B). The flow rate was set to 300 µL/min. A

259

elution gradient was set with 80% of elute A for 2 min, then a gradual decrease in elute A to

260

2% in 8 min and then maintained for 5 min. Finally, elute A was returned to 80% for 5 min

261

prior to the next run. The injection volume of the sample to LC-MS/MS was 10 µL. The

262

PUFAs including arachidonic (AA), adrenic (AdA), α-linolenic (ALA), n-3 docosapentaenoic

263

(DPA), eicosapentaenoic (EPA) and docosahexaenoic (DHA) acids and its oxidized lipid

264

products, namely prostaglandins, hydroeicosatetraenoic acids (5-, 8-, 9-, 11-, 12-, 15-, 20-

265

HETEs), hydroxyl-DHA (4-, 7-, 8-, 10-, 11-, 13-, 14-, 16-, 17-, 20-HDoHE), isoprostanoids

266

(isoprostanes, dihomo-isoprostanes, neuroprostanes, phtytoprostanes) and isofuranoids

267

(isofurans, dihomo-isofurans, neurofurans), were analyzed using multiple reactions 11 ACS Paragon Plus Environment

Environmental Science & Technology

Page 12 of 31

268

monitoring (MRM) mode summarized in Supplementary Table S2. A total of 43 oxidized

269

lipid products were determined and a typical mass spectra is shown in Supplementary Figure

270

S1. Quantification of PUFAs and their oxidized lipid products were determined by relating

271

the peak area with its corresponding heavy-labeled isotopic internal standard peak. For

272

compounds without a heavy-labeled isotope, their quantification were determined by using a

273

heavy-labeled isotope with the closest chemical structure (Supplementary Table S2). All

274

PUFAs and their oxidized lipid product standards were purchased from Cayman Chemical Co.

275

(Ann Arbor, MI, USA) or in-house synthesized by the Institut des Biomolecules Max

276

Mousseron (IBMM, Montpellier, France).57-60

277

278

Epididymal Sperm Count and Serum Testosterone Analysis

279

At P63, blood serum and epididymides of adult offspring from 5 dams (n=5) per group were

280

collected. Epididymis was cut into small pieces. They were then resuspeded in PBS (3%

281

BSA) and incubated at 37 °C for an hour. Epididymal spermatozoa were stained with eosin-

282

nigrosin (0.67% eosin Y, 10% nigrosin, 0.9% sodium chloride) and at least 200 epididymal

283

spermatozoa (magnification x400) were examined under a light microscope.

284

Serum testosterone levels were determined by use of ELISA kits (MP Biomedicals, Ohio,

285

USA). The reagents including the working hormone-HRP conjugate (100µl), rabbit anti-

286

hormone solution (50µl) and samples or standards (25 µl), were added and mixed in wells

287

and incubated at 37 °C for 90 min. The wells were then rinsed five times with distilled water,

288

followed by an addition of 100 µl TMB and incubation at room temperature. The reaction

289

was then stopped by 1N HCl solution. Absorbance was measured at 450 nm.

290

12 ACS Paragon Plus Environment

Page 13 of 31

Environmental Science & Technology

291

Statistical Analysis

292

Statistical evaluation was conducted using SPSS16. Data normality was checked by Shapiro-

293

Wilk and Q-Q Plot. The Spearman’s correlation analysis was used to examine the possible

294

correlation between BPs and PFAAs in the samples. Data are presented as mean ± SD.

295

Groups are considered significantly different if P < 0.05.

296

297

Results and Discussion

298

LCMS/MS Analysis of PFOS Concentrations in Neonatal Testes

299

Throughout the course of the exposure, there were no significant differences in the body

300

weights of the maternal mice between the control and the treatment groups [low (0.3 µg/g)

301

and high (3 µg/g)]. The average numbers of litters in the control and the treatment groups

302

were similar (Fig. 1A). No significant changes in the weights of neonatal testes were

303

measured (Fig. 1B). The data suggested that the exposure did not cause any observable

304

maternal and developmental toxicity. Nevertheless, placental transfer of PFOS was obvious

305

through the detection of PFOS in the neonatal testes. Using LC-MS/MS analysis of the PFOS

306

concentrations in neonatal testes, considerable levels of PFOS were detected in the testes of

307

the treatment groups. The levels of the testicular uptake of PFOS were in a dose-dependent

308

manner (Fig. 1C) and were proportionate with the maternal exposure doses.

309

310

Transcriptomic Analysis of Neonatal Testes upon in Utero PFOS Exposure

311

In an attempt to understand the molecular mechanisms underlying the effect of in utero PFOS

312

exposure on neonatal testes, comparative transcriptomic analysis was conducted (n = 4 per group).

313

A total of 56 and 319 DEGs were identified in the low and high dose of PFOS treatments, as

13 ACS Paragon Plus Environment

Environmental Science & Technology

Page 14 of 31

314

compared to control group, respectively (Supplementary Table S3 and S4). The DEGs were then

315

subjected to the Database for Annotation, Visualization and Integrated Discovery (DAVID) v6.7

316

and Ingenuity Pathway Analysis (IPA) to determine the biological functions, KEGG pathways

317

and canonical pathways altered by PFOS exposures. The results of the DAVID analysis showed

318

that PFOS treatment caused the alteration of biological processes related to fatty acid synthesis

319

and metabolism, such as oxidation-reduction process, fatty acid metabolic process, fatty acid

320

beta-oxidation, lipid metabolic process and coenzyme A biosynthetic process (Fig. 2A &

321

Supplementary Table S5). In the KEGG pathway analysis, the highest ranked pathway were the

322

metabolic pathways, suggesting that the prenatal PFOS exposure could alter different

323

metabolisms in testes. In addition, a large number of pathways related to fatty acid metabolism

324

were underlined. These included fatty acid metabolism, fatty acid degradation, fatty acid

325

elongation and biosynthesis of unsaturated fatty acids (Fig. 2B & Supplementary Table S6).

326

These findings are concordant to the previous reports that PFOS exposure could alter fatty

327

metabolism, but most of the studies worked on liver tissue. For instance, PFOS could induce the

328

expression of the lipid catabolism genes involved in fatty acid and triglyceride synthesis, leading

329

to liver steatosis.61 Another mouse fetal liver study showed that prenatal PFOS exposure could

330

activate the synthesis and metabolism of fatty acids and lipids, leading to liver damage in the

331

fetus.45 When we compared our result with the transcriptomic analysis of human primary

332

hepatocytes exposed to perfluorooctanoic acid (PFOA), we found that the response of liver and

333

testis tissues is different. In the liver, PFOA could stimulate gene expression of the proto-

334

oncogenes c-Jun and c-Fos and alter the cell cycle of hepatocytes.62 Using Ingenuity Pathway

335

Analysis (IPA), 13 canonical pathways (P < 0.05) were identified at the low dose of PFOS

336

exposure. Of these, pathways directly related to testicular development and functions include (i)

337

epithelial adherens junction signaling, (ii) Wnt/β-catenin signaling, (iii) androgen biosynthesis

338

and (iv) embryonic stem cell pluripotency (Fig. 2C & Supplementary Table S7). It has been

14 ACS Paragon Plus Environment

Page 15 of 31

Environmental Science & Technology

339

reported that Wnt/β-catenin signaling is important to testicular functions. For example,

340

constitutive Wnt/β-catenin signaling would disrupt Sertoli cells differentiation and

341

spermatogenesis.63 In addition, Wnt/β-catenin signaling is essential for epididymal coiling and

342

proliferation of undifferentiated spermatogonia.64,65 At the high dose of in utero PFOS exposure,

343

a greater number of canonical pathways (59 pathways, P < 0.05) were highlighted

344

(Supplementary Table S8). The altered pathways that were associated with PFOS toxicity and

345

testicular function included (i) nuclear receptor activation (LXR/RXR and PXR/RXR activation),

346

(ii) hormonal function (IGF-1 signaling), (iii) lipid metabolism (biosynthesis of cholesterol,

347

zymosterol, stearate, oleate or degradation of ceramide, fatty acid), (iv) redox response

348

(superoxide radicals degradation and oxidized GTP/dGTP detoxification) and (v) xenobiotic

349

metabolism signaling. Epithelial adherens junction-related pathways like (vi) germ cell-Sertoli

350

cell and (vii) Sertoli-Sertoli junctional signaling were also affected (Fig. 2C). Consistently, the

351

results of the pathways analysis from the transcriptomic data suggested that the in utero PFOS

352

exposure perturbed lipid metabolism, redox and cell adhesion functions of the testes. Our

353

previous studies had already demonstrated the negative effects of PFOS on the disruption of

354

epithelial homeostasis at the Sertoli-Sertoli and Sertoli-germ cell interface in the seminiferous

355

epithelium,66 leading to the loss of germ cell adhesion, spermatid orientation, and

356

dysregulation of spermatogenesis. However, little attention has been drawn to the effects of

357

PFOS on the disruption of lipid signaling mediators, which are known to play critical roles in

358

steroidogenesis, early male sexual development and masculinization.32,33,36,37 In order to

359

validate the findings from the transcriptomic analysis, qPCR analysis was performed. 4 gene

360

clusters involved in (i) germ cell-sertoli cell junction and sertoli-sertoli cell junction signaling

361

including spectrin beta, non-erythrocytic 1 (SPTBN1), integrin beta 1 (ITGB1), mitogen-activated

362

protein kinase 9 (MAPK9), catenin, beta 1 (CTNNB1) and phosphatidylinositol 3-kinase,

363

regulatory subunit, polypeptide 1 (PIK3R1); (ii) fatty acid metabolism including stearoyl-

15 ACS Paragon Plus Environment

Environmental Science & Technology

Page 16 of 31

364

Coenzyme A desaturase 2 (SCD2) and hydroxysteroid (17-beta) dehydrogenase 12 (HSD17B12);

365

(iii) steroid hormone response such as oxytocin receptor (OXTR); and (iv) Wnt/β-catenin

366

signaling pathway including catenin, beta 1 (CTNNB1) and adaptor protein, phosphotyrosine

367

interaction, PH domain and leucine zipper containing 2 (APPL2) were selected. We found that

368

the result of qPCR analysis well matched with transcriptomic data (Fig. 2D).

369 370

Lipidomic Analysis of Neonatal Testes upon in Utero PFOS Exposure

371

The mechanistic action of PFOS has been suggested to be mediated via nuclear receptors.

372

While biochemical phenotypes associated with PFOS toxicities are closely linked to the

373

perturbation of lipid metabolism and redox status.55 With the benefit of hindsight and the

374

bioinformatics analysis obtained in this study, the PFOS-perturbed fatty acid metabolism,

375

redox and other physiological outcomes may be related to the modulation of lipid

376

metabolism and signaling. To gain a further understanding about this biological phenomenon,

377

targeted lipidomic analysis was performed to determine the levels of polyunsaturated fatty

378

acids and its oxidized lipid products, and to endeavor their relationship with the

379

transcriptome evaluation.

380

We evidently showed that in utero PFOS exposure to the dams influenced DHA metabolism

381

in the pup testes (P1). We analyzed the impact of PFOS exposure on the metabolism of

382

polyunsaturated fatty acids (PUFAs) in relation to n-6 (arachidonic and adrenic acids) and n-

383

3 (α-linolenic, n-3 docosapentaenoic, eicosapentaenoic and docosahexaenoic acids) PUFAs

384

pathways, which are currently known to take part in male reproduction.67 Among them, the

385

levels of adrenic acid (AdA) and docosahexaenoic acid (DHA) in testes were significantly

386

reduced by low and high PFOS concentrations respectively (Fig. 3A), showing disruption of

387

PUFA metabolism. The reduction of AdA at low PFOS concentration corresponds to the

16 ACS Paragon Plus Environment

Page 17 of 31

Environmental Science & Technology

388

upregulation of PTGIS (prostaglandin I2) gene indicating an increase in inflammation (Fig.

389

3B). However, this effect is transient since AdA was not suppressed at higher PFOS

390

concentration. Although no reports are available on AdA in testes, in endothelial cells, a

391

small portion of AdA can retro-convert to AA.68 In agreement, the upregulation of ELOVL5

392

gene which is shared by n-6 and n-3 PUFA metabolism displayed possible competition for

393

fatty acid elongation of AA to AdA and EPA to n-3 DPA69 when exposed by PFOS (Fig. 3B).

394

We also observed a significant linear decreasing trend in EPA and DHA with PFOS

395

concentration (Fig. 3A), indicating the possible role of ELOVL5 to favor n-6 PUFA

396

metabolism (Fig. 3B). Suppression of DHA is detrimental to the pup testicular development

397

and consequently reproduction. Deficiency of essential n-3 PUFA, noticeably DHA, is

398

recognized to cause testicular malfunction in adults, where sperm mobility was found to be

399

reduced in boars.70 Lower levels of DHA in human seminal plasma were found in the male

400

partners of infertile couples compared to fertile males.71 Recent studies found that

401

supplementation of DHA to healthy men prevented sperm DNA fragmentation72 and rescued

402

spermatogenesis in null-mice lacking delta-6 desaturase, an essential rate-limiting enzyme in

403

the synthesis of PUFAs.73 However, it is not known if the suppression of DHA in the testis by

404

PFOS exposure is permanent, as our previous study showed that pups exposed to PFOS for

405

longer days had elevated DHA in the liver, the central tissue for lipid metabolism.55 PUFAs

406

are prone to oxidation via enzymatic or non-enzymatic pathway due to the numerous skipped

407

diene bonds. DHA, having the most number of double bonds, is the most vulnerable PUFA

408

for oxidation. Oxidation products from n-6 PUFA are often toxic and related to inflammation,

409

whereas those of n-3 PUFA are lipid mediators that assist in resolving inflammation and take

410

part in normalizing microvascular functions.74,75

411

Bioinformatics analysis of the transcriptome data underlined that PFOS exposure perturbed

412

the redox responses and oxidation-reduction processes in neonatal testes. We anticipated 17 ACS Paragon Plus Environment

Environmental Science & Technology

Page 18 of 31

413

PFOS to induce oxidative stress in the testes, since comparable amounts of PUFAs were

414

reported in the neonatal liver.55 However, the data showed a different oxidized lipid profile in

415

the testes as compared to the liver. In the analysis of oxidized lipid products, only 15 of the

416

43 enzymatic (e.g. COX, LOX and cytochrome P450 mediated (Table 1)) and non-enzymatic

417

(e.g. isoprostanoids and isofuranoids) oxidized lipid products were detected in the testes. The

418

generation of isoprostanes predominates under normal oxidative stress, whereas isofuranoids

419

predominates under extreme oxidative stress and high oxygen tension. The non-detectable

420

products do not signify that it is absent in the tissue but probably in trace concentrations

421

below the level of detection (0.05 ng/mg tissue) by the LC-MS/MS. It is proposed PFOS

422

initiates non-enzymatic lipid oxidation through the presence of reactive oxygen species such

423

as hydrogen peroxide and hydroxyl radicals55,76 and it appears the young testis lacks the

424

presence of these species. Further, the difference might also be attributed by the age of the

425

pups where lipid metabolism is not fully developed. Exposure of PFOS significantly elevated

426

LOX-mediated 5-HETE and 15-HETE from arachidonic acid (AA) in the testes. A significant

427

linear trend with an increase of PFOS concentration was observed for 15-HETE. LOX-

428

mediated hydroxy-DHA (HDoHE) products were not detected in the testes. HDoHE are

429

precursors of pro-resolving factors of inflammation such as resolvins (RvD, RvE),75 and our

430

data indicated that the presence of PFOS might inhibit the regulation of HDoHE. No

431

significant changes were observed for oxidized lipid products released via cytochrome P450

432

after PFOS exposure (Table 2). Surprisingly, we found no noticeable changes in the non-

433

enzymatic oxidized lipid products in the neonatal testes. This included the reputable 15-F2t-

434

IsoPs, which is known to be the best biomarker for oxidative stress through free radical/ROS

435

pathway in vivo. No linear trend with an increment of PFOS concentration was noticed for all

436

non-enzymatic oxidized lipid products detected. To date, the upregulation of 15-HETE is

437

associated to inflammation, proliferation, apoptosis, membrane blebbing and chromatin

18 ACS Paragon Plus Environment

Page 19 of 31

Environmental Science & Technology

438

condensation in cancer cells77,78 (Fig. 3B), but its role in testis is poorly understood.

439

Nevertheless, lipoxygenase-catalyzed products of the AA cascade play important roles in

440

early male sexual development, steroidogenesis and masculinization.31-41 It is proposed that

441

PFOS initiates non-enzymatic lipid oxidation through the presence of reactive oxygen species

442

such as hydrogen peroxide and hydroxyl radicals,55,76 and it appears that the young testes lack

443

the presence of these species. Nonetheless, the observations warrant the determination of

444

whether or not PFOS exposure in utero causes a predisposition to reproductive dysfunction in

445

male fetus, and therefore contributing to the disturbance in testosterone production and

446

spermatogenesis in adult offspring.

447

448

Characterization of Testicular Functions at Postnatal Growth of the Male Offspring

449

We then determined the effects of in utero PFOS exposure on the testicular function of pups. At

450

P63, significant reductions in the serum testosterone levels (Fig. 4A) and epididymal sperm count

451

(Fig. 4B) were observed in pups from the high-dose exposed group. This observation supports

452

the pathway analysis data of neonatal testes in which in utero PFOS exposure perturbed the

453

expression of genes associated with androgen synthesis. The identified canonical pathways

454

provide a mechanistic insight to revealing the effect of in utero PFOS exposure on neonatal

455

testes, as androgen is important for the development of male reproductive system and sexual

456

maturation.79 Our omics and physiological data are consistent with the findings of other

457

studies. For example, in utero exposure of female rats to PFOS (5 or 20 mg·kg-1·day-1

458

maternal dose) reduced the number of fetal Leydig cells and impaired cell functions in

459

comparison to the untreated control group through the decrease in steroidogenic activity and

460

testosterone levels.80 Furthermore, PFOA exposure (1, 3, 5, 10, 20 or 40 mg·kg-1·day-1) in

461

timed-pregnant CD-1 mice (GD 1-17) affected sexual maturation in male offspring.81 Adult

19 ACS Paragon Plus Environment

Environmental Science & Technology

Page 20 of 31

462

rats treated with 0.05–0.5% PFOA exhibited reduced activities of testicular-metabolic

463

enzymes.82,83 Using microsomal preparations of human and rat testes, PFCs were found to

464

target steroidogenic enzymes, resulting in a reduction of testosterone production.84 Moreover,

465

the perturbation of androgen synthesis affects the process of masculinization while the

466

disruption of epithelial adherens affects Sertoli-germ cells interaction. Our IPA analysis of

467

differentially expressed transcripts from low- and high-dose of in utero PFOS exposure

468

underlined the epithelial adherens junction-signaling pathway, which is known to regulate the

469

blood-testis barrier for spermatogenesis. Our previous study has demonstrated that the cell

470

adherens proteins are the molecular targets of PFOS.85 The prenatal perturbations of this

471

important pathway were found to be associated with a significant reduction of epididymal sperm

472

counts in adult offspring.

473

Collectively, the present study analyzed the dynamic changes of global mRNA expression to

474

provide compiled experimental data of the integrated pathways using different

475

computational tools. To advance the significances of these findings, targeted lipidomics

476

analysis was implemented to track the changes in the levels of PUFAs and their oxidized

477

lipid products, serving the purposes of revealing further molecular targets/pathways to

478

unravel the effects of PFOS on neonatal testes.

479 480

Acknowledgements

481

This work was supported by the Partner State Key Laboratory of Environmental and

482

Biological Analysis (SKLP-16-17-P01) to Dr Chris KC Wong (Hong Kong Baptist

483

University).

484 485

Availability of supporting data 20 ACS Paragon Plus Environment

Page 21 of 31

Environmental Science & Technology

486

The sequence data from this study have been submitted to the NCBI Sequence Read Archive

487

(SRA) (http://www.ncbi.nlm.nih.gov/sra) under the accession number SRP108484.

488

489

21 ACS Paragon Plus Environment

Environmental Science & Technology

Page 22 of 31

490

FIGURE LEGENDS

491

Figure 1. The pregnant mice were orally administered 0, 0.3 or 3 µg PFOS/g body weight by

492

gavage with corn oil throughout gestation. On postnatal day 1, the average number of litters

493

per dam was counted, neonatal testes from each dam were collected for weight measurement

494

and PFOS analysis. The figures shows the effects of in utero PFOS exposure on the (A)

495

number of litters per dam, (B) relative testicular weights and (C) concentrations of PFOS in

496

the testes of neonates.

497

Figure 2. The Database for Annotation, Visualization and Integrated Discovery (DAVID)

498

analysis highlighted the alteration of (A) biological processes and (B) pathways caused by

499

high dose (3 µg PFOS/g body weight) PFOS treatment. (C) Ingenuity Pathway Analysis (IPA)

500

highlighted the changes in canonical pathways related to testicular development, testicular

501

functions and toxicity caused by low dose (0.3 µg PFOS/g body weight) and high dose (3 µg

502

PFOS/g body weight) PFOS exposure. Expression of selected genes for each significantly

503

dysregulated pathways are presented in heat-map format. Color of each gene corresponds to

504

the log2 fold change of PFOS treatment versus control. (D) Quantitative PCR analysis to

505

validate the findings of RNA-sequencing. n = 4, *P < 0.05.

506

Figure 3. Lipidomic analysis reveals the effect of prenatal PFOS exposure on level of

507

polyunsaturated fatty acids. (A) Concentrations of polyunsaturated fatty acids in post-natal

508

mice (P1) exposed to low or high dose of PFOS during dam pregnancy. Values are mean ±

509

SD, n=4. *P < 0.05 versus control. P-trend indicates linear trend with PFOS concentration.

510

AA: arachidonic acid; AdA: adrenic acid; ALA: α-linolenic acid; EPA: eicosapentaenoic acid;

511

DPA: docosapentaenoic acid; DHA: docosahexanoic acid. (B) integrated analysis of

512

transcriptomic and lipidomic data.

22 ACS Paragon Plus Environment

Page 23 of 31

Environmental Science & Technology

513

Figure 4. At P63, significant reductions in (A) serum testosterone levels and (B) epididymal

514

sperm counts were recorded.

23 ACS Paragon Plus Environment

Environmental Science & Technology

515

Page 24 of 31

Literature Cited

516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562

(1) Sharlip, I. D.; Jarow, J. P.; Belker, A. M.; Lipshultz, L. I.; Sigman, M.; Thomas, A. J.; Schlegel, P. N.; Howards, S. S.; Nehra, A.; Damewood, M. D.; Overstreet, J. W.; Sadovsky, R. Best practice policies for male infertility. Fertil. Steril. 2002, 77 (5), 873-882. (2) Matzuk, M. M.; Lamb, D. J. The biology of infertility: research advances and clinical challenges. Nat. Med. 2008, 14 (11), 1197-1213. (3) CHEMTrust Effects of pollutants on the reproductive health of male vertebrate wildlife - males under threat; 2008. (4) Joensen, U. N.; Bossi, R.; Leffers, H.; Jensen, A. A.; Skakkebaek, N. E.; Jorgensen, N. Do perfluoroalkyl compounds impair human semen quality? Environ. Health Perspect. 2009, 117 (6), 923-927. (5) Toft, G.; Jonsson, B. A.; Lindh, C. H.; Giwercman, A.; Spano, M.; Heederik, D.; Lenters, V.; Vermeulen, R.; Rylander, L.; Pedersen, H. S.; Ludwicki, J. K.; Zviezdai, V.; Bonde, J. P. Exposure to perfluorinated compounds and human semen quality in Arctic and European populations. Hum. Reprod. 2012, 27 (8), 2532-2540. (6) Joensen, U. N.; Veyrand, B.; Antignac, J. P.; Blomberg, J. M.; Petersen, J. H.; Marchand, P.; Skakkebaek, N. E.; Andersson, A. M.; Le Bizec, B.; Jorgensen, N. PFOS (perfluorooctanesulfonate) in serum is negatively associated with testosterone levels, but not with semen quality, in healthy men. Hum. Reprod. 2013, 28 (3), 599608. (7) Specht, I. O.; Hougaard, K. S.; Spano, M.; Bizzaro, D.; Manicardi, G. C.; Lindh, C. H.; Toft, G.; Jonsson, B. A.; Giwercman, A.; Bonde, J. P. Sperm DNA integrity in relation to exposure to environmental perfluoroalkyl substances - a study of spouses of pregnant women in three geographical regions. Reprod. Toxicol. 2012, 33 (4), 577583. (8) Vested, A.; Ramlau-Hansen, C. H.; Olsen, S. F.; Bonde, J. P.; Kristensen, S. L.; Halldorsson, T. I.; Becher, G.; Haug, L. S.; Ernst, E. H.; Toft, G. Associations of in utero exposure to perfluorinated alkyl acids with human semen quality and reproductive hormones in adult men. Environ. Health Perspect. 2013, 121 (4), 453455. (9) Bateson, P.; Barker, D.; Clutton-Brock, T.; Deb, D.; D'Udine, B.; Foley, R. A.; Gluckman, P.; Godfrey, K.; Kirkwood, T.; Lahr, M. M.; McNamara, J.; Metcalfe, N. B.; Monaghan, P.; Spencer, H. G.; Sultan, S. E. Developmental plasticity and human health. Nature. 2004, 430 (6998), 419-421. (10) Gluckman, P. D.; Hanson, M. A.; Bateson, P.; Beedle, A. S.; Law, C. M.; Bhutta, Z. A.; Anokhin, K. V.; Bougneres, P.; Chandak, G. R.; Dasgupta, P.; Smith, G. D.; Ellison, P. T.; Forrester, T. E.; Gilbert, S. F.; Jablonka, E.; Kaplan, H.; Prentice, A. M.; Simpson, S. J.; Uauy, R.; West-Eberhard, M. J. Towards a new developmental synthesis: adaptive developmental plasticity and human disease. Lancet. 2009, 373 (9675), 1654-1657. (11) Gluckman, P. D.; Hanson, M. A.; Beedle, A. S.; Spencer, H. G. Predictive adaptive responses in perspective. Trends Endocrinol. Metab. 2008, 19 (4), 109-110. (12) Hanson, M. A.; Gluckman, P. D. Developmental origins of health and disease: new insights. Basic Clin. Pharmacol. Toxicol. 2008, 102 (2), 90-93. (13) Gluckman, P. D.; Hanson, M. A. Developmental plasticity and human disease: research directions. J. Intern. Med. 2007, 261 (5), 461-471. 24 ACS Paragon Plus Environment

Page 25 of 31

563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612

Environmental Science & Technology

(14) Gluckman, P. D.; Hanson, M. A.; Beedle, A. S. Early life events and their consequences for later disease: a life history and evolutionary perspective. Am. J. Hum. Biol. 2007, 19 (1), 1-19. (15) Gluckman, P. D.; Hanson, M. A.; Pinal, C. The developmental origins of adult disease. Matern. Child Nutr. 2005, 1 (3), 130-141. (16) Gluckman, P. D.; Cutfield, W.; Hofman, P.; Hanson, M. A. The fetal, neonatal, and infant environments-the long-term consequences for disease risk. Early Hum. Dev. 2005, 81 (1), 51-59. (17) Andersson, A. M.; Jorgensen, N.; Main, K. M.; Toppari, J.; Rajpert-De, M. E.; Leffers, H.; Juul, A.; Jensen, T. K.; Skakkebaek, N. E. Adverse trends in male reproductive health: we may have reached a crucial 'tipping point'. Int. J. Androl. 2008, 31 (2), 7480. (18) Skakkebaek, N. E.; Jorgensen, N.; Main, K. M.; Rajpert-De Meyts, E.; Leffers, H.; Andersson, A. M.; Juul, A.; Carlsen, E.; Mortensen, G. K.; Jensen, T. K.; Toppari, J. Is human fecundity declining? Int. J. Androl. 2006, 29 (1), 2-11. (19) Yiee, J. H.; Baskin, L. S. Environmental factors in genitourinary development. J. Urol. 2010, 184 (1), 34-41. (20) Ma, L. Endocrine disruptors in female reproductive tract development and carcinogenesis. Trends Endocrinol. Metab. 2009, 20 (7), 357-363. (21) Crinnion, W. J. Maternal levels of xenobiotics that affect fetal development and childhood health. Altern. Med. Rev. 2009, 14 (3), 212-222. (22) Lyche, J. L.; Gutleb, A. C.; Bergman, A.; Eriksen, G. S.; Murk, A. J.; Ropstad, E.; Saunders, M.; Skaare, J. U. Reproductive and developmental toxicity of phthalates. J. Toxicol. Environ. Health B Crit Rev. 2009, 12 (4), 225-249. (23) Grellier, J.; Bennett, J.; Patelarou, E.; Smith, R. B.; Toledano, M. B.; Rushton, L.; Briggs, D. J.; Nieuwenhuijsen, M. J. Exposure to disinfection by-products, fetal growth, and prematurity: a systematic review and meta-analysis. Epidemiology. 2010, 21 (3), 300-313. (24) Wong, E. W.; Cheng, C. Y. Impacts of environmental toxicants on male reproductive dysfunction. Trends Pharmacol. Sci. 2011, 32 (5), 290-299. (25) Llopis, J.; Westin, S.; Ricote, M.; Wang, Z.; Cho, C. Y.; Kurokawa, R.; Mullen, T. M.; Rose, D. W.; Rosenfeld, M. G.; Tsien, R. Y.; Glass, C. K. Ligand-dependent interactions of coactivators steroid receptor coactivator-1 and peroxisome proliferatoractivated receptor binding protein with nuclear hormone receptors can be imaged in live cells and are required for transcription. Proc. Natl. Acad. Sci. U. S. A. 2000, 97 (8), 4363-4368. (26) Schultz, R.; Yan, W.; Toppari, J.; Volkl, A.; Gustafsson, J. A.; Pelto-Huikko, M. Expression of peroxisome proliferator-activated receptor alpha messenger ribonucleic acid and protein in human and rat testis. Endocrinology. 1999, 140 (7), 2968-2975. (27) Abbott, B. D.; Wolf, C. J.; Das, K. P.; Zehr, R. D.; Schmid, J. E.; Lindstrom, A. B.; Strynar, M. J.; Lau, C. Developmental toxicity of perfluorooctane sulfonate (PFOS) is not dependent on expression of peroxisome proliferator activated receptor-alpha (PPAR alpha) in the mouse. Reprod. Toxicol. 2009, 27 (3-4), 258-265. (28) Rosen, M. B.; Schmid, J. R.; Corton, J. C.; Zehr, R. D.; Das, K. P.; Abbott, B. D.; Lau, C. Gene Expression Profiling in Wild-Type and PPARalpha-Null Mice Exposed to Perfluorooctane Sulfonate Reveals PPARalpha-Independent Effects. PPAR. Res. 2010, 2010 . (29) Kim, H. S.; Jun, K. S.; Sik, H. E.; Seok, K. T.; Hee, K. S.; Young, H. S. Induction of apoptosis and CYP4A1 expression in Sprague-Dawley rats exposed to low doses of perfluorooctane sulfonate. J. Toxicol. Sci. 2011, 36 (2), 201-210. 25 ACS Paragon Plus Environment

Environmental Science & Technology

613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661

Page 26 of 31

(30) Rosen, M. B.; Schmid, J. E.; Das, K. P.; Wood, C. R.; Zehr, R. D.; Lau, C. Gene expression profiling in the liver and lung of perfluorooctane sulfonate-exposed mouse fetuses: comparison to changes induced by exposure to perfluorooctanoic acid. Reprod. Toxicol. 2009, 27 (3-4), 278-288. (31) Abayasekara, D. R.; Band, A. M.; Cooke, B. A. Evidence for the involvement of phospholipase A2 in the regulation of luteinizing hormone-stimulated steroidogenesis in rat testis Leydig cells. Mol. Cell Endocrinol. 1990, 70 (2), 147-153. (32) Adams, I. R.; McLaren, A. Sexually dimorphic development of mouse primordial germ cells: switching from oogenesis to spermatogenesis. Development. 2002, 129 (5), 1155-1164. (33) Amateau, S. K.; McCarthy, M. M. Induction of PGE2 by estradiol mediates developmental masculinization of sex behavior. Nat. Neurosci. 2004, 7 (6), 643-650. (34) Didolkar, A. K.; Sundaram, K. Arachidonic acid is involved in the regulation of hCG induced steroidogenesis in rat Leydig cells. Life Sci. 1987, 41 (4), 471-477. (35) Dix, C. J.; Habberfield, A. D.; Sullivan, M. H.; Cooke, B. A. Inhibition of steroid production in Leydig cells by non-steroidal anti-inflammatory and related compounds: evidence for the involvement of lipoxygenase products in steroidogenesis. Biochem. J. 1984, 219 (2), 529-537. (36) Gupta, C.; Goldman, A. S. The arachidonic acid cascade is involved in the masculinizing action of testosterone on embryonic external genitalia in mice. Proc. Natl. Acad. Sci. U. S. A. 1986, 83 (12), 4346-4349. (37) Gupta, C. The role of prostaglandins in masculine differentiation: modulation of prostaglandin levels in the differentiating genital tract of the fetal mouse. Endocrinology. 1989, 124 (1), 129-133. (38) Kojima, I.; Kojima, K.; Rasmussen, H. Possible role of phospholipase A2 action and arachidonic acid metabolism in angiotensin II-mediated aldosterone secretion. Endocrinology. 1985, 117 (3), 1057-1066. (39) Lopez-Ruiz, M. P.; Choi, M. S.; Rose, M. P.; West, A. P.; Cooke, B. A. Direct effect of arachidonic acid on protein kinase C and LH-stimulated steroidogenesis in rat Leydig cells; evidence for tonic inhibitory control of steroidogenesis by protein kinase C. Endocrinology. 1992, 130 (3), 1122-1130. (40) Maloberti, P.; Cornejo, M. F.; Castillo, A. F.; Castilla, R.; Duarte, A.; Toledo, M. F.; Meuli, F.; Mele, P.; Paz, C.; Podesta, E. J. Enzymes involved in arachidonic acid release in adrenal and Leydig cells. Mol. Cell Endocrinol. 2007, 265-266 113-120. (41) Solano, A. R.; Dada, L.; Podesta, E. J. Lipoxygenase products as common intermediates in cyclic AMP-dependent and -independent adrenal steroidogenesis in rats. J. Mol. Endocrinol. 1988, 1 (2), 147-154. (42) Fang, X.; Dillon, J. S.; Hu, S.; Harmon, S. D.; Yao, J.; Anjaiah, S.; Falck, J. R.; Spector, A. A. 20-carboxy-arachidonic acid is a dual activator of peroxisome proliferator-activated receptors alpha and gamma. Prostaglandins Other Lipid Mediat. 2007, 82 (1-4), 175-184. (43) Huang, J. T.; Welch, J. S.; Ricote, M.; Binder, C. J.; Willson, T. M.; Kelly, C.; Witztum, J. L.; Funk, C. D.; Conrad, D.; Glass, C. K. Interleukin-4-dependent production of PPAR-gamma ligands in macrophages by 12/15-lipoxygenase. Nature. 1999, 400 (6742), 378-382. (44) Wang, S. Y.; Lau, K.; Lai, K. P.; Zhang, J. W.; Tse, A. C.; Li, J. W.; Tong, Y.; Chan, T. F.; Wong, C. K.; Chiu, J. M.; Au, D. W.; Wong, A. S.; Kong, R. Y.; Wu, R. S. Hypoxia causes transgenerational impairments in reproduction of fish. Nat. Commun. 2016, 7 12114.

26 ACS Paragon Plus Environment

Page 27 of 31

662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711

Environmental Science & Technology

(45) Lai, K. P.; Li, J. W.; Cheung, A.; Li, R.; Billah, M. B.; Chan, T. F.; Wong, C. K. Transcriptome sequencing reveals prenatal PFOS exposure on liver disorders. Environ. Pollut. 2017, 223 416-425. (46) Fida, T. T.; Moreno-Forero, S. K.; Breugelmans, P.; Heipieper, H. J.; Roling, W. F.; Springael, D. Physiological and Transcriptome Response of the Polycyclic Aromatic Hydrocarbon Degrading Novosphingobium sp. LH128 after Inoculation in Soil. Environ. Sci. Technol. 2017, 51 (3), 1570-1579. (47) European Food Safety Authority Perfluorooctane sulfonate (PFOS), perfluorooctanic acid (PFOA) and their salts - Scientific Opinion of the Panel on Contaminants in the Food Chain. The EFSA Journal. 2008, 653 1-131. (48) Wan, H. T.; Leung, P. Y.; Zhao, Y. G.; Wei, X.; Wong, M. H.; Wong, C. K. Blood plasma concentrations of endocrine disrupting chemicals in Hong Kong populations. J. Hazard. Mater. 2013, 261 763-769. (49) Li, J. W.; Lai, K. P.; Ching, A. K.; Chan, T. F. Transcriptome sequencing of Chinese and Caucasian population identifies ethnic-associated differential transcript abundance of heterogeneous nuclear ribonucleoprotein K (hnRNPK). Genomics. 2014, 103 (1), 56-64. (50) Lai, K. P.; Li, J. W.; Wang, S. Y.; Chiu, J. M.; Tse, A.; Lau, K.; Lok, S.; Au, D. W.; Tse, W. K.; Wong, C. K.; Chan, T. F.; Kong, R. Y.; Wu, R. S. Tissue-specific transcriptome assemblies of the marine medaka Oryzias melastigma and comparative analysis with the freshwater medaka Oryzias latipes. BMC. Genomics. 2015, 16 135. (51) Yates, A.; Akanni, W.; Amode, M. R.; Barrell, D.; Billis, K.; Carvalho-Silva, D.; Cummins, C.; Clapham, P.; Fitzgerald, S.; Gil, L.; Giron, C. G.; Gordon, L.; Hourlier, T.; Hunt, S. E.; Janacek, S. H.; Johnson, N.; Juettemann, T.; Keenan, S.; Lavidas, I.; Martin, F. J.; Maurel, T.; McLaren, W.; Murphy, D. N.; Nag, R.; Nuhn, M.; Parker, A.; Patricio, M.; Pignatelli, M.; Rahtz, M.; Riat, H. S.; Sheppard, D.; Taylor, K.; Thormann, A.; Vullo, A.; Wilder, S. P.; Zadissa, A.; Birney, E.; Harrow, J.; Muffato, M.; Perry, E.; Ruffier, M.; Spudich, G.; Trevanion, S. J.; Cunningham, F.; Aken, B. L.; Zerbino, D. R.; Flicek, P. Ensembl 2016. Nucleic Acids Res. 2016, 44 (D1), D710D716. (52) Robinson, M. D.; McCarthy, D. J.; Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010, 26 (1), 139-140. (53) Huang, d. W.; Sherman, B. T.; Lempicki, R. A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 2009, 4 (1), 4457. (54) Huang, d. W.; Sherman, B. T.; Lempicki, R. A. Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 2009, 37 (1), 1-13. (55) Lee, Y. Y.; Wong, C. K.; Oger, C.; Durand, T.; Galano, J. M.; Lee, J. C. Prenatal exposure to the contaminant perfluorooctane sulfonate elevates lipid peroxidation during mouse fetal development but not in the pregnant dam. Free Radic. Res. 2015, 49 (8), 1015-1025. (56) Lee, Y. Y.; Crauste, C.; Wang, H.; Leung, H. H.; Vercauteren, J.; Galano, J. M.; Oger, C.; Durand, T.; Wan, J. M.; Lee, J. C. Extra Virgin Olive Oil Reduced Polyunsaturated Fatty Acid and Cholesterol Oxidation in Rodent Liver: Is This Accounted for Hydroxytyrosol-Fatty Acid Conjugation? Chem. Res. Toxicol. 2016, 29 (10), 1689-1698. (57) de la Torre, A.; Lee, Y. Y.; Mazzoni, A.; Guy, A.; Bultel-Ponce, V.; Durand, T.; Oger, C.; Lee, J. C.; Galano, J. M. Total syntheses and in vivo quantitation of novel 27 ACS Paragon Plus Environment

Environmental Science & Technology

712 713 714 715 716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744 745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760

(58)

(59)

(60)

(61)

(62)

(63)

(64) (65)

(66)

(67)

(68)

(69)

(70)

(71)

(72)

Page 28 of 31

neurofuran and dihomo-isofuran derived from docosahexaenoic acid and adrenic acid. Chemistry. 2015, 21 (6), 2442-2446. de La, T. A.; Lee, Y. Y.; Oger, C.; Sangild, P. T.; Durand, T.; Lee, J. C.; Galano, J. M. Synthesis, discovery, and quantitation of dihomo-isofurans: biomarkers for in vivo adrenic acid peroxidation. Angew. Chem. Int. Ed Engl. 2014, 53 (24), 6249-6252. Guy, A.; Oger, C.; Heppekausen, J.; Signorini, C.; De, F. C.; Furstner, A.; Durand, T.; Galano, J. M. Oxygenated metabolites of n-3 polyunsaturated fatty acids as potential oxidative stress biomarkers: total synthesis of 8-F3t-IsoP, 10-F4t-NeuroP and [D4]10-F4t-NeuroP. Chemistry. 2014, 20 (21), 6374-6380. Oger, C.; Bultel-Ponce, V.; Guy, A.; Balas, L.; Rossi, J. C.; Durand, T.; Galano, J. M. The handy use of Brown's P2-Ni catalyst for a skipped diyne deuteration: application to the synthesis of a [D4]-labeled F4t-neuroprostane. Chemistry. 2010, 16 (47), 13976-13980. Das, K. P.; Wood, C. R.; Lin, M. T.; Starkov, A. A.; Lau, C.; Wallace, K. B.; Corton, J. C.; Abbott, B. D. Perfluoroalkyl acids-induced liver steatosis: Effects on genes controlling lipid homeostasis. Toxicology. 2017, 378 37-52. Buhrke, T.; Kruger, E.; Pevny, S.; Rossler, M.; Bitter, K.; Lampen, A. Perfluorooctanoic acid (PFOA) affects distinct molecular signalling pathways in human primary hepatocytes. Toxicology. 2015, 333 53-62. Tanwar, P. S.; Kaneko-Tarui, T.; Zhang, L.; Rani, P.; Taketo, M. M.; Teixeira, J. Constitutive WNT/beta-catenin signaling in murine Sertoli cells disrupts their differentiation and ability to support spermatogenesis. Biol. Reprod. 2010, 82 (2), 422-432. Kumar, M.; Syed, S. M.; Taketo, M. M.; Tanwar, P. S. Epithelial Wnt/betacatenin signalling is essential for epididymal coiling. Dev. Biol. 2016, 412 (2), 234-249. Takase, H. M.; Nusse, R. Paracrine Wnt/beta-catenin signaling mediates proliferation of undifferentiated spermatogonia in the adult mouse testis. Proc. Natl. Acad. Sci. U. S. A. 2016, 113 (11), E1489-E1497. Wan, H. T.; Mruk, D. D.; Wong, C. K.; Cheng, C. Y. The apical ES-BTB-BM functional axis is an emerging target for toxicant-induced infertility. Trends Mol. Med. 2013, 19 (7), 396-405. Yan, L.; Bai, X. L.; Fang, Z. F.; Che, L. Q.; Xu, S. Y.; Wu, D. Effect of different dietary omega-3/omega-6 fatty acid ratios on reproduction in male rats. Lipids Health Dis. 2013, 12 33. Mann, C. J.; Kaduce, T. L.; Figard, P. H.; Spector, A. A. Docosatetraenoic acid in endothelial cells: formation, retroconversion to arachidonic acid, and effect on prostacyclin production. Arch. Biochem. Biophys. 1986, 244 (2), 813-823. Zhang, J. Y.; Kothapalli, K. S.; Brenna, J. T. Desaturase and elongase-limiting endogenous long-chain polyunsaturated fatty acid biosynthesis. Curr. Opin. Clin. Nutr. Metab Care. 2016, 19 (2), 103-110. Am-in, N.; Kirkwood, R. N.; Techakumphu, M.; Tantasuparuk, W. Lipid profiles of sperm and seminal plasma from boars having normal or low sperm motility. Theriogenology. 2011, 75 (5), 897-903. Oborna, I.; Wojewodka, G.; De Sanctis, J. B.; Fingerova, H.; Svobodova, M.; Brezinova, J.; Hajduch, M.; Novotny, J.; Radova, L.; Radzioch, D. Increased lipid peroxidation and abnormal fatty acid profiles in seminal and blood plasma of normozoospermic males from infertile couples. Hum. Reprod. 2010, 25 (2), 308-316. Martinez-Soto, J. C.; Domingo, J. C.; Cordobilla, B.; Nicolas, M.; Fernandez, L.; Albero, P.; Gadea, J.; Landeras, J. Dietary supplementation with docosahexaenoic

28 ACS Paragon Plus Environment

Page 29 of 31

761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 804 805 806 807

Environmental Science & Technology

(73)

(74)

(75)

(76)

(77) (78)

(79) (80)

(81)

(82)

(83)

(84)

(85)

acid (DHA) improves seminal antioxidant status and decreases sperm DNA fragmentation. Syst. Biol. Reprod. Med. 2016, 62 (6), 387-395. Roqueta-Rivera, M.; Stroud, C. K.; Haschek, W. M.; Akare, S. J.; Segre, M.; Brush, R. S.; Agbaga, M. P.; Anderson, R. E.; Hess, R. A.; Nakamura, M. T. Docosahexaenoic acid supplementation fully restores fertility and spermatogenesis in male delta-6 desaturase-null mice. J. Lipid Res. 2010, 51 (2), 360-367. Galano, J. M.; Lee, J. C.; Gladine, C.; Comte, B.; Le Guennec, J. Y.; Oger, C.; Durand, T. Non-enzymatic cyclic oxygenated metabolites of adrenic, docosahexaenoic, eicosapentaenoic and alpha-linolenic acids; bioactivities and potential use as biomarkers. Biochim. Biophys. Acta. 2015, 1851 (4), 446-455. Gabbs, M.; Leng, S.; Devassy, J. G.; Monirujjaman, M.; Aukema, H. M. Advances in Our Understanding of Oxylipins Derived from Dietary PUFAs. Adv. Nutr. 2015, 6 (5), 513-540. Jin, L.; Zhang, P.; Shao, T.; Zhao, S. Ferric ion mediated photodecomposition of aqueous perfluorooctane sulfonate (PFOS) under UV irradiation and its mechanism. J. Hazard. Mater. 2014, 271 9-15. Cho, K. J.; Seo, J. M.; Kim, J. H. Bioactive lipoxygenase metabolites stimulation of NADPH oxidases and reactive oxygen species. Mol. Cells. 2011, 32 (1), 1-5. Ma, J.; Zhang, L.; Zhang, J.; Liu, M.; Wei, L.; Shen, T.; Ma, C.; Wang, Y.; Chen, Y.; Zhu, D. 15-lipoxygenase-1/15-hydroxyeicosatetraenoic acid promotes hepatocellular cancer cells growth through protein kinase B and heat shock protein 90 complex activation. Int. J. Biochem. Cell Biol. 2013, 45 (6), 1031-1041. Holterhus, P. M. Molecular androgen memory in sex development. Pediatr. Endocrinol. Rev. 2011, 9 Suppl 1 515-518. Zhao, B.; Li, L.; Liu, J.; Li, H.; Zhang, C.; Han, P.; Zhang, Y.; Yuan, X.; Ge, R. S.; Chu, Y. Exposure to perfluorooctane sulfonate in utero reduces testosterone production in rat fetal Leydig cells. PLoS. One. 2014, 9 (1), e78888. Lau, C.; Thibodeaux, J. R.; Hanson, R. G.; Narotsky, M. G.; Rogers, J. M.; Lindstrom, A. B.; Strynar, M. J. Effects of perfluorooctanoic acid exposure during pregnancy in the mouse. Toxicol. Sci. 2006, 90 (2), 510-518. Mehrotra, K.; Morgenstern, R.; Lundqvist, G.; Becedas, L.; Bengtsson, A. M.; Georgellis, A. Effects of peroxisome proliferators and/or hypothyroidism on xenobiotic-metabolizing enzymes in rat testis. Chem. Biol. Interact. 1997, 104 (2-3), 131-145. Mehrotra, K.; Morgenstern, R.; Ahlberg, M. B.; Georgellis, A. Hypophysectomy and/or peroxisome proliferators strongly influence the levels of phase II xenobiotic metabolizing enzymes in rat testis. Chem. Biol. Interact. 1999, 122 (2), 73-87. Zhao, B.; Hu, G. X.; Chu, Y.; Jin, X.; Gong, S.; Akingbemi, B. T.; Zhang, Z.; Zirkin, B. R.; Ge, R. S. Inhibition of human and rat 3beta-hydroxysteroid dehydrogenase and 17beta-hydroxysteroid dehydrogenase 3 activities by perfluoroalkylated substances. Chem. Biol. Interact. 2010, 188 (1), 38-43. Wan, H. T.; Mruk, D. D.; Wong, C. K.; Cheng, C. Y. Perfluorooctanesulfonate (PFOS) perturbs male rat Sertoli cell blood-testis barrier function by affecting F-actin organization via p-FAK-Tyr(407): an in vitro study. Endocrinology. 2014, 155 (1), 249-262.

29 ACS Paragon Plus Environment

Environmental Science & Technology

1 2 3

4 5 6 7

Page 30 of 31

Table 1. Non-enzymatic lipid products in testis tissues in post-natal mice (P1) exposed to 0.3 (low) or 3 µg (high) PFOS/g body weight during dam pregnancy. Control

Low

High

p-trend

AA-derived 5-F2t-IsoP 15-F2t-IsoP Total F2t-IsoPs IsoF

34.98±14.12 1.53±0.92 36.51±14.94 23.08±9.25

14.91±4.13 1.11±0.78 16.02±4.74 22.08±2.26

54.04±52.26 2.43±0.78 56.47±52.87 43.42±24.60

0.412 0.151 0.398 0.092

AdA-derived 7-Dihomo-IsoP 17-Dihomo-IsoP Total Dihomo-IsoP

6.79±2.75 15.22±9.24 22.01±11.73

4.99±2.79 12.58±3.09 17.57±5.58

6.01±3.78 14.78±2.21 20.79±2.61

0.731 0.917 0.826

DHA-derived 16-HDoHE 56.68±5.21 39.50±9.12 43.55±24.04 0.252 4-NeuroF 215.57±75.24 194.52±63.22 192.73±34.14 0.604 Values are mean ± SD, n=4. P-trend indicates linear trend with PFOS concentration. AA: arachidonic acid; AdA: adrenic acid; DHA: docosahexaenoic acid; IsoP: isoprostane; IsoF: isofurans; HDoHE: hydroxy-DHA; NeuroF: neurofuran.

ACS Paragon Plus Environment

Page 31 of 31

1 2

3 4 5 6

Environmental Science & Technology

Table 2. Enzymatic lipid products in testis tissues in post-natal mice (P1) exposed to 0.3 or 3 µg PFOS/g body weight during dam pregnancy. Control Low High p-trend COX-mediated 0.68±0.16 0.29±0.13 0.86±0.50 0.449 PGF2α LOX-mediated 5-HETE 8-HETE 12-HETE 15-HETE Total

215.21±57.86 325.66±80.02 93.06±38.11 21.26±12.25 655.19±166.50

614.51±231.50** 382.76±223.57 24.08±15.03 47.85±35.92 1069.21±324.20*

99.85±40.25 284.49±105.64 148.89±111.63 127.34±37.84** 660.57±126.34

0.273 0.707 0.280 0.001 0.974

CYP-mediated 9-HETE 11-HETE 20-HETE Total

137.52±65.90 450.73±158.29 209.01±100.15 797.27±185.31

319.21±250.81 721.94±501.31 314.97±126.83 1356.12±617.82

127.63±59.14 913.92±381.37 237.23±80.23 1278.78±399.94

0.929 0.115 0.711 0.155

Enzymatic lipid products presented in testis tissues. Values are mean ± SD, n=4. *p