Iodoacetic acid disrupts the thyroid endocrine system - ACS Publications

Iodoacetic acid (IAA), an unregulated DBP, has been shown to. 24 be cytotoxic ... developmental toxicity of monohaloacids have shown that IAA might ha...
1 downloads 0 Views 1MB Size
Subscriber access provided by University of Winnipeg Library

Ecotoxicology and Human Environmental Health

Iodoacetic acid disrupts the thyroid endocrine system in vitro and in vivo Ying Xia, Yan Mo, Qiyuan Yang, Yang Yu, Meiyu Jiang, Shumao Wei, Du Lu, Huan Wu, Guodong Lu, Yunfeng Zou, Zhiyong Zhang, and Xiao Wei Environ. Sci. Technol., Just Accepted Manuscript • DOI: 10.1021/acs.est.8b01802 • Publication Date (Web): 29 May 2018 Downloaded from http://pubs.acs.org on May 29, 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 32

Environmental Science & Technology

Iodoacetic acid disrupts the thyroid endocrine system in vitro and in vivo

1 2 3

Ying Xia†, ||, Yan Mo†, ||, Qiyuan Yang†, ||, Yang Yu†, Meiyu Jiang†, Shumao Wei†, Du Lu†, Huan

4

Wu†, Guodong Lu∆, Yunfeng Zou∆, Zhiyong Zhang †,*, Xiao Wei †,*

5 6



Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning 530021, China

7 8 9 10



Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, China

11 12 13

||

These authors contributed equally to this work.

14 15

*Correspondence to: Xiao Wei, Shuang Yong Road 22, Nanning 530021, China. Tel:

16

86-771-5312371. Fax: 86-771-5312371. E-mail: [email protected]; [email protected]

17

Key words: Iodoacetic acid, thyroid endocrine disruption, disinfection byproducts, drinking

18

water

19 20

Competing financial interests declaration: The authors declare they have no actual or

21

potential competing financial interests.

1

ACS Paragon Plus Environment

Environmental Science & Technology

22

Abstract

23

Exposure to drinking water disinfection byproducts (DBPs) is potentially associated with

24

adverse developmental effects. Iodoacetic acid (IAA), an unregulated DBP, has been shown to

25

be cytotoxic, mutagenic, genotoxic, and tumorigenic. However, its endocrine-disrupting

26

effects remain unknown. This study evaluated the IAA-induced disruption of the thyroid

27

endocrine system using in vitro and in vivo assays. Rat pituitary tumor GH3 cells were treated

28

with IAA in the presence and absence of triiodothyronine (T3). IAA exposure significantly

29

reduced T3-activated GH3 cell proliferation, indicating the antagonistic activity of IAA in

30

vitro. Sprague-Dawley rats were also subjected to IAA treatment through oral gavage for 28

31

consecutive days. IAA exposure significantly down-regulated the mRNA expression levels of

32

the thyrotropin receptor (TSHR), the sodium/iodide symporter (NIS), and type I deiodinase

33

and simultaneously reduced the protein expression levels of TSHR and NIS. IAA exposure

34

decreased T3 levels but increased the weights of hypothalamus and the levels of thyrotropin

35

releasing hormone and thyrotropin. In addition, IAA induced the formation of smaller and

36

more depleted follicles or even vacuolization in the thyroid. These results suggested that IAA

37

potentially disrupts the thyroid endocrine system both in vitro and in vivo.

2

ACS Paragon Plus Environment

Page 2 of 32

Page 3 of 32

Environmental Science & Technology

38

Introduction

39

Iodoacetic acid (IAA) was first identified as a disinfection byproduct (DBP) in drinking

40

water in the United States Nationwide DBP Occurrence Study.1, 2 High iodide levels in source

41

water and chloramination are important factors in the formation of IAA.3, 4 IAA is generally

42

detected in drinking water at µg/L levels, but the cytotoxicity and genotoxicity of IAA are

43

higher than those of many regulated chloro/bromo-DBPs.1, 3, 5 A molecular mechanism of

44

IAA-induced cytotoxicity and genotoxicity is the generation of oxidative stress. IAA causes

45

toxicity in mammalian cells by inhibiting glyceraldehyde-3-phosphate dehydrogenase

46

(GAPDH) and reducing the levels of pyruvate and ATP.6-8 IAA even induces the malignant

47

transformation of NIH3T3 cells and the production of aggressive fibrosarcomas, showing that

48

IAA is also a potential carcinogen.9 However, IAA is still an unregulated DBP and does not

49

have a maximum contaminant level in regulations and guidelines.10

50

Previous studies mostly focused on the cytotoxicity, genotoxicity, and carcinogenicity of

51

IAA.1, 3, 9, 11 Furthermore, contaminants in drinking water have attracted much interest in

52

terms of their developmental toxicity. Because of their endocrine-disrupting effects, some

53

DBPs have become regarded as potential inducers in drinking water. Epidemiological studies

54

have revealed the association between longtime exposure to DBPs and increased risk of

55

adverse pregnancy outcomes such as miscarriage, intrauterine growth retardation, still birth,

56

low birth weight, and birth defects.12-17 A mouse whole embryo culture assay showing the

57

developmental toxicity of a series of haloacetic acids demonstrated their ability to affect

58

neural tube, eye, and heart development and to produce malpositioned and hypoplastic

59

pharyngeal arches.18 Quantitative structure-activity relationship (QSAR) analysis of the

60

developmental toxicity of monohaloacids have shown that IAA might have more

61

developmental toxicity than other monohaloacids.19

62

Thyroid hormone (TH), an essential endocrine hormone, is involved in the regulation of

63

metabolism and development, in mammals. TH abnormality may result in the hypogenesis of

64

bones, muscles, and the central nervous system in embryos.20, 21 Thyroid system function is

65

regulated by the hypothalamus-pituitary-thyroid (HPT) axis.22 Thus, its function can be 3

ACS Paragon Plus Environment

Environmental Science & Technology

66

disrupted by chemicals via multiple physiological steps within the HPT axis: (1) TH synthesis

67

[e.g., iodine uptake, the sodium/iodide symporter (NIS), thyroperoxidase (TPO),

68

thyroglobulin (TG), or the thyrotropin receptor (TSHR)], (2) TH transport and action [e.g.,

69

thyroxine-binding globulin, transthyretin, albumin, hepatic phase II enzyme, or TH receptors

70

(THR)], (3) TH metabolism [e.g., uridine diphosphoglucuronosyl transferases and deiodinases

71

(DIOs)], and (4) the HPT feedback mechanism.23 However, the effect of IAA on TH is not yet

72

fully understood.

73

The purposes of this study were to determine the ability of IAA to disturb thyroid system

74

functions in vitro and in vivo. A T-screen assay was initially performed to investigate whether

75

IAA disrupts TH-induced GH3 cell proliferation. A 28-day repeated-dose toxicity study in

76

Sprague-Dawley (SD) rats was subsequently conducted to confirm the effects of IAA on TH

77

levels and thyroid functions and to examine the possible pathways involved in the disruption

78

of the HPT axis.

79 80

Materials and methods

81

Reagents. IAA was procured from Sigma-Aldrich (MO, USA). Dulbecco’s modified Eagle’s

82

medium/nutrient mixture F-12 (DMEM/F-12), horse serum (HS), fetal bovine serum (FBS),

83

trypsin-EDTA, and TRIzol were purchased from Invitrogen (NY, USA). Antibodies for TSHR

84

(sc13936) and TPO (sc58432) were procured from Santa Cruz, Inc. (CA, USA). Antibodies

85

for NIS (ab17795) and TG (ab156008) were obtained from Abcam (MA, USA).

86 87

Cell culture. Rat pituitary tumor cell line GH3 was purchased from the Cell Center of the

88

Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (Beijing, China).

89

The cells were cultured in DMEM/F-12 medium containing 15% HS and 2.5% FBS at 37 °C

90

in a humidified 5% CO2 atmosphere.

91 92

Animals. SD rats were procured from the Laboratory Animal Center of Guangxi Medical

93

University and housed under the following conditions: 23 °C ± 2 °C, 60% ± 5% humidity, 4

ACS Paragon Plus Environment

Page 4 of 32

Page 5 of 32

Environmental Science & Technology

94

and a light/dark (12 h/12 h) cycle. Animal ethics approval was obtained from Guangxi

95

Medical University of Animal Ethics Committee.

96 97

T-screen assay. The T-screen assay was performed as published previously.24 GH3 cells were

98

transferred to serum-free medium (DMEM/F-12 medium supplemented with 10 ng/mL

99

sodium selenite, 10 µg/mL bovine insulin, 500 µg/mL bovine serum albumin, 10 µM

100

ethanolamine, and 10 µg/mL human apotransferrin) and maintained for 48 h. Then, GH3 cells

101

were detached and washed three times with serum-free medium. Approximately 2.5×103 cells

102

in 50 µL of serum-free medium per well were plated in 96-well microplates. After 24 h,

103

different concentrations of test compounds in 50 µL of serum-free medium were added to the

104

respective well for 96 h. Cell proliferation was analyzed using the crystal violet method.11, 25

105

Different concentrations of T3 were firstly tested to reach the optimal concentration. Then,

106

the desired concentrations of IAA (without cytotoxicity) were analyzed in the presence of the

107

optimal concentration of T3.

108 109

Animal treatment. The study was conducted as the Organization for Economic Cooperation

110

and Development (OECD) Test Guideline 407.26 Five-week-old SD rats with a weight of 100

111

± 10 g were randomly grouped (7 males and 7 females) at the beginning of the experiments.

112

They were kept in their cages for 1 week to allow them to acclimatize to the laboratory

113

conditions prior to the onset of the treatment study. The rats were then exposed to 0, 6, 12,

114

and 24 mg/kg bw of IAA in ultrapure water through oral gavage for 28 consecutive days. The

115

highest dose was determined based on an acute toxicity study of IAA in rats. Our preliminary

116

study indicated that 126 and 147 mg/kg bw were the acute oral LD50 values of IAA in male

117

and female rats, respectively. Clinical manifestations, food consumption, and body weight

118

were observed every day. All of the rats were sacrificed through exsanguination with 10%

119

chloral hydrate anesthesia after 28 days. Blood samples were collected for clinical

120

biochemical and hormonal analysis. Organs such as the hypothalamus, pituitary, thyroid,

121

kidney, liver, thymus, heart, testes, epididymides, prostate, ovaries, and uterus were weighed. 5

ACS Paragon Plus Environment

Environmental Science & Technology

Page 6 of 32

122

The relative weight of organs was calculated as follows: relative organ weight = organ weight

123

(g) / body weight (g).

124 125

Clinical biochemical and hormonal analysis. Clinical biochemical parameters included

126

alanine

127

aminotransferase-alanine aminotransferase ratio (AST/ALT), alkaline phosphatase (ALP),

128

total bilirubin (TB), total protein (TP), globulin (GLB), albumin (ALB), albumin-globulin

129

ratio (A/G), blood urea nitrogen (BUN), uric acid (UA), creatinine (CREA), sodium (Na),

130

potassium (K), and chlorine (Cl). Total triiodothyronine (TT3), free triiodothyronine (FT3),

131

total thyroxin (TT4), and free thyroxin (FT4) were determined using radioimmunoassay kits

132

from Cisbio Bioassays (Codolet, France). Thyrotropin (TSH), thyrotropin-releasing hormone

133

(TRH), TG, and TPO were measured using enzyme-linked immunosorbent assay (ELISA)

134

kits from EIAab (Hubei, China). These commercially available kits were widely employed to

135

measure TH, TSH, and TRH before and have been proved to be effective in identifying

136

hyperthyroidism or hypothyroidism.27-31

aminotransferase

(ALT),

aspartate

aminotransferase

(AST),

aspartate

137 138

Histopathology and immunohistochemistry. The obtained organs were fixed, dehydrated,

139

embedded, sectioned, and stained according to standard procedures for histopathological

140

examinations. Immunohistochemical analysis was developed in 4.0 µm sections of tissues

141

fixed with paraformaldehyde. After dewaxing and antigen retrieval, the sections were blocked

142

and incubated with anti-NIS, anti-TSHR, anti-TG, and anti-TPO antibody at 4 °C overnight,

143

combined with biotinylated secondary antibody, stained with diaminobenzidine (DAB), and

144

lightly counterstained with hematoxylin. Immunopositive reactions were randomly selected

145

from three different areas in each section and analyzed with Image-Pro Plus 6.0 (MD, USA).

146 147

RNA isolation and quantitative real-time reverse transcriptase-polymerase chain

148

reaction (RT-qPCR). The total RNA of the hepatic genes (THRα and THRβ1) and thyroid

149

genes (TSHR, NIS, DIO1, TPO, and TG) was isolated with TRIzol reagent and assessed 6

ACS Paragon Plus Environment

Page 7 of 32

Environmental Science & Technology

150

through A260/A280 ratio measurement and agarose-formaldehyde denaturing gel electrophoresis

151

as previously described.32, 33 RNA was reverse-transcribed to cDNA by using a PrimeScript

152

RT reagent kit from Takara (Shiga, Japan). qPCR analysis was conducted with a SYBR

153

Premix Ex Tap II kit from Takara (Shiga, Japan). PCR primers were designed using Oligo 6.0

154

(CO, USA) (Table S1) and synthesized by Takara Biotech Co., Ltd. (Liaoning, China). Each

155

sample was analyzed in triplicate, and the average cycle threshold (CT) was calculated. These

156

data were quantified by the 2

157

expression relative to the control.

-∆∆CT

method and are presented as the fold change of gene

158 159

Statistical analysis. Data were analyzed using SPSS 16.0 (IL, USA) and GraphPad Prism 5.0

160

(CA, USA). Significant differences among treatment groups were evaluated through a

161

one-way analysis of variance (ANOVA) test followed by Dunnett’s multiple comparison test

162

versus the control group. The incidence of histopathological changes was analyzed using

163

Fisher’s exact test. Two-tailed P< 0.05 indicated statistically significant differences. Data

164

were expressed as the mean ± standard deviation (SD).

165 166

Results

167

IAA reduced GH3 cell proliferation in the presence of T3.

168

The dose-dependent manner of GH3 cells stimulated by T3 is shown in Fig. 1A. The

169

lowest T3 concentration that induced significant cell proliferation was 0.1 nM. The highest

170

cell proliferation was found at 1 nM T3, and this observation was about 2-fold higher than

171

that of the control group. The GH3 cells were then exposed to 0.125 - 8 µM IAA without T3.

172

Fig. 1B shows that IAA was not significantly cytotoxic to GH3 cell lines within a range of

173

0.125–2 µM. After the GH3 cells were treated with IAA in the presence of 1 nM T3, 2 µM

174

IAA significantly reduced the GH3 cell growth compared with 0 µM IAA (Fig. 1C).

175 176

IAA altered the organ weights of SD rats but not their general observations, body weight

7

ACS Paragon Plus Environment

Environmental Science & Technology

177

or food consumption.

178

Male rats differed from female rats in their relative organ weights after IAA treatment

179

(Table 1). There was a significant dose-dependent decrease in the relative organ weight of the

180

heart and liver of males. However, in females, there was a decrease only in the ovaries. By

181

contrast, the relative weight of the hypothalamus increased significantly in female rats.

182

Additionally, there was an increasing trend in the thyroid relative weight of the female rats

183

but not of the male rats.

184

No abnormal clinical manifestations were observed in any group. As shown in Fig. S1

185

and S2, IAA did not induce significant changes in the body weight or food consumption of

186

SD rats.

187 188

IAA mediated the clinical biochemistry in SD rats.

189

In the blood biochemical findings after IAA treatment (Table S2), the male rats also

190

responded differently from the female rats in most clinical biochemical parameters, except a

191

common decrease in GLB and a common increase in A/G ratio in the 24 mg/kg bw IAA

192

groups and a common decrease in AST in the 6 mg/kg bw groups. The highest IAA dose (24

193

mg/kg bw) decreased the levels of ALP, TP, and BUN in the male rats but did not affect the

194

female rats. ALP tended to decrease in the female rats, but the differences failed to reach

195

statistical significance. Both AST and ALT decreased in the 6, 12, and 24 mg/kg bw groups

196

of female rats, leaving the AST/ALT ratio unchanged. Na and Cl increased only in the 12 and

197

24 mg/kg bw groups of the female rats.

198 199

IAA increased TRH, TSH, TG, and TPO but decreased TT3 in the serum of SD rats.

200

A common significant decrease of TT3 was observed at the dose of 24 mg/kg bw IAA in

201

both male and female rats (Table 2). This change was accompanied by increases in TG and

202

TPO in the 12 and 24 mg/kg bw IAA groups of both genders. In male rats, TSH was

203

increased in the 12 and 24 mg/kg bw groups, whereas in female rats, TRH was elevated in the

204

12 mg/kg bw group. 8

ACS Paragon Plus Environment

Page 8 of 32

Page 9 of 32

Environmental Science & Technology

205 206

IAA induced histopathological changes in the thyroid of SD rats.

207

The histopathological examination of the thyroid showed that the number of smaller and

208

depleted thyroid follicles in the IAA-treated male or female rats increased compared with that

209

of the control group but failed to reach statistical significance (Fig. 2B, 2C, and Table 3).

210

However, IAA induced a significant increase in total number of smaller thyroid follicles in the

211

male and female rats. The intrafollicular colloid was stained more faintly in these smaller or

212

depleted follicles. The cellular vacuolization of the thyroid was observed in one male rat in

213

the 24 mg/kg bw group (Fig. 2D). Slight edemas were observed in liver cells of most male

214

and female rats in the treatment groups (Fig. S3). No other changes appeared in periportal

215

zones after the rats were exposed to IAA.

216 217

IAA regulated the target gene expression in the thyroid and liver of SD rats.

218

Expression of the NIS gene was down-regulated in all IAA treatment groups of male and

219

female rats (Fig. 3). TSHR mRNA expression was also decreased at the doses of 12 mg/kg bw

220

in both genders and at 6 mg/kg bw in female rats. Meanwhile, expression of the DIO1 gene

221

was down-regulated in the 12 and 24 mg/kg bw groups of both genders. IAA enhanced the

222

expression of the TG and TPO genes and reduced the expression of the THRα gene at doses of

223

12 mg/kg bw in male rats. Both the THRβ1 gene in the 24 mg/kg bw group of male rats and

224

the THRα gene in the 6 mg/kg bw group of female rats were up-regulated.

225 226

IAA reduced NIS and TSHR protein expression in the thyroid of SD rats.

227

Consistent with the decrease in NIS and TSHR mRNA expression, the protein levels of

228

NIS and TSHR were down-regulated in all IAA treatment groups of male and female rats (Fig.

229

4, S4). However, there were no evident expression changes in the TG and TPO proteins

230

compared with the control.

231 232

Discussion 9

ACS Paragon Plus Environment

Environmental Science & Technology

233

Many epidemiological studies have shown the association between chronic DBPs

234

exposure and adverse developmental effects, but few DBPs have been tested for their direct

235

role in endocrine-disrupting effects. For all we know, this is the first study evaluating the

236

TH-disrupting effects of IAA in vitro and in vivo. The present study demonstrates that IAA is

237

a thyroid toxicant.

238

The T-screen assay is an in vitro model for chemicals that shows their potential

239

TH-disrupting effects. GH3 cell is suitable for T-screen assay because of its special growth

240

characteristics. In the ‘70 s and ‘80 s, Samuels34 and Hinkle35 et al. reported that T3 can

241

stimulate rat pituitary tumor cell proliferation and induce growth hormone secretion. Thus,

242

this assay was developed to evaluate the agonistic and antagonistic potency of chemicals in

243

the absence and presence of T3 by Hohenwarter et al. in 1996, the so-called T-screen assay.24

244

This assay has been successfully employed to detect the effects of many TH-disrupting

245

chemicals, such as pentachlorophenol, phosphorus-containing flame retardants, pesticides,

246

and hydroxylated polybrominated diphenyl ethers.36-40 In our study, IAA reduced GH3 cell

247

proliferation in the absence of T3, indicating potential non-agonistic activity. However, IAA

248

also inhibited cell proliferation when co-administered with T3. Thus, the assay revealed that

249

IAA exerted antagonistic effects on TH action. Previous studies have not yet displayed the

250

TH-disrupting effect of IAA in vitro.

251

For the in vivo assay, OECD Test Guideline 407, a repeated dose 28-day oral toxicity

252

study in rodents, is a screening assay conducted to detect endocrine disrupters acting through

253

various toxicity mechanisms. Several chemicals have been evaluated with this assay.41-43 In

254

our study, IAA treatment increased the weights of the hypothalamus and reduced the weights

255

of the ovaries of the female rats. The previous in vitro study, which reported that treatment

256

with IAA inhibits antral follicle growth and decreases estradiol levels in mouse ovarian

257

follicles.44 These results indicate that IAA may be an ovarian toxicant in vivo and in vitro. In

258

addition, as the dose of IAA increases, the relative thyroid weight has an upward trend in

259

female rats but not in male rats. Therefore, IAA might have thyroid-mediated effects.

260

Under normal physiological conditions, TRH secreted from the hypothalamus stimulates 10

ACS Paragon Plus Environment

Page 10 of 32

Page 11 of 32

Environmental Science & Technology

261

the pituitary to release TSH, which binds to TSHR on the surface of thyroid follicle cells.

262

Several thyroid genes and proteins, such as NIS, TPO, and TG, are triggered to participate in

263

the synthesis of TH.45 The uptake of iodide into thyroid follicular cells is the initial step in TH

264

synthesis. NIS can transport and concentrate iodide into the thyroid via Na+/K+-ATPase.46

265

NIS functioning is inhibited by many environmental toxicants, such as nitrate, iodide, and

266

bromide.47, 48 In the present study, the decrease in the mRNA and protein expression of TSHR

267

and NIS in the IAA-dosed group indicated that IAA is probably an inhibitor of TSHR and NIS.

268

In addition, Na+/K+-ATPase provides the driving force for the uptake of iodide by generating

269

a large concentration gradient in sodium. Na+/K+-ATPase pumps 3 Na+ out and takes 2 K+ in,

270

which would requires one ATP. However, IAA-induced GAPDH inhibition causes a severe

271

reduction of cellular ATP levels.6, 7 The reduction of ATP may inhibit Na+/K+-ATPase activity.

272

These studies indicate that IAA may affect metabolic processes at this level and inhibit TH

273

synthesis.

274

In thyroid follicular cells, iodide is activated by TPO. Then, the tyrosine residues of TG

275

are iodinated to produce monoiodinated and diiodinated residues, which coupled to form TH

276

(T4 and T3).22, 45 In this study, IAA treatment induced a significant change in the TPO and TG

277

transcript levels in the 12 mg/kg bw group of male rats, but no significant differences were

278

observed in the TPO and TG protein expression levels in the thyroid tissue. On the other hand,

279

the serum TPO and TG values increased in the 12 and 24 mg/kg bw groups of male and

280

female rats. Elevation of serum TG was proved to be an indicator of an abnormal or damaged

281

thyroid follicular structure.49, 50 Thus, the high TPO and TG levels in serum were possibly

282

attributable to the release of TPO and TG from the damaged thyroid follicular cells into the

283

serum under the dosing conditions of this study.

284

T4 is the main form of released TH. The biological activity of T3, however, is 4-fold

285

higher than that of serum T4. Serum T4 and T3 mostly bind to carrier proteins and are

286

transported to target tissues. The free (unbound) serum T4 and T3 levels were only 0.03% and

287

0.3%, respectively. Free THs, mainly T3, bind to THR in the nuclei of target cells to generate

288

their biological effects.45 Our study showed that THRα and THRβ1 mRNA expression levels 11

ACS Paragon Plus Environment

Environmental Science & Technology

289

were altered, but such changes were independent of dosage. Further studies on other target

290

tissues should be conducted to help elucidate the THR-disrupting effects of IAA. Another

291

major pathway of T3 production is the conversion of T4 to T3 by type I and type II

292

deiodinases.51 Thus, the reduction in DIO1 mRNA expression in the present study might be a

293

response to the decrease in T3 in the 24 mg/kg bw groups of both genders. These observations

294

were further supported by the inhibition of TSHR and NIS in T3 synthesis and the results of

295

the T-screen assay in vitro.

296

The reduction in TH (T4 and T3) will provoke an increase in TSH and TRH release

297

through the negative HPT feedback mechanism.22 In the present study, the weights of the

298

hypothalamus and TRH level of female rats increased in the IAA-dosed groups following the

299

reduction in T3. Similarly, the TSH level of male rats was stimulated by the reduction in T3

300

after IAA treatment. The release of TRH and TSH can stimulate the hyperplasia and

301

hypertrophy of the thyroid, which synthesizes and secretes more TH to satisfy physiological

302

requirements.22 The increase in TPO and TG mRNA expression levels in IAA-dosed groups

303

likely compensated for the reduction in T3. In the OECD Test Guideline 407, the

304

characteristic patterns of thyroid toxicants are to induce changes in the weight and/or

305

histopathology of the thyroid.26 Although IAA-treatment did not induce a statistically

306

significant difference in thyroid weights, the formation of small follicles was increasingly

307

observed in the exposed groups. These findings provided direct evidence supporting the

308

potential of IAA as a thyroid toxicant.

309

Decreased liver weight, abnormal AST, ALT, AST/ALT, ALP, TP, GLB, A/G, and BUN

310

values, and hepatocytic edema revealed the adverse effects of IAA on liver function. IAA has

311

been reported to significantly increase reactive oxygen species (ROS) levels5, 8 and to induce

312

cytotoxicity in primary rat hepatocytes in vivo.25 These results are in agreement with our

313

study. In general, decreased ALP in the serum, as observed in both genders, can be regarded

314

as a marker of reduced liver activity.52-54 ALP expression, which is TH dependent,55-60 may be

315

suppressed by IAA through the activation of TH-mediated effects.

316

In addition to in vitro assay, IAA had not been shown to exhibit T3-agonist activity in in 12

ACS Paragon Plus Environment

Page 12 of 32

Page 13 of 32

Environmental Science & Technology

317

vivo assay. In general, compounds that resemble the structure of TH can bind to nuclear THRs

318

in cells and then stimulate GH3 cell growth or regulate gene transcription and expression in

319

target organs. For example, Triac and Tetrac, which resemble the structures of T3 and T4,

320

respectively, induced GH3 cell growth.36 In contrast, this study indicates that IAA presents

321

antagonistic activity. In the T-screen assay, IAA inhibited GH3 cell growth in the presence of

322

T3. The possible mechanism of this antagonistic action was considered to be associated with

323

decreasing the rate of T3 entry into cells and inhibiting binding by nuclear receptors.61, 62

324

However, in vitro assay do not reflect the process of the absorption, distribution, metabolism,

325

or excretion of thyroid disruptors in vivo. Thyroid disruptors may interfere with TH functions

326

through multiple pathways and complicated feedback mechanisms in vivo. The present study

327

showed that IAA decreased T3 synthesis and metabolism by inhibiting the mRNA expression

328

of TSHR, NIS, and DIO1 in rats. Although IAA did not disrupt THR in the liver, consistent

329

with the findings of the T-screen assay, the expression of THR in other target organs still

330

needs to be evaluated in the future.

331

Overall, our results indicated that IAA reduced T3-mediated GH3 cell proliferation and

332

decreased T3 by down-regulating TSHR, NIS, and DIO1 mRNA expression in rats. Combined

333

with histopathological changes in the thyroid, this study suggests that IAA exposure disrupts

334

thyroid function and possibly induces potential health risks. Of particular note is that

335

exposure to IAA reduced the weights of the ovaries of the female rats. The finding revealed

336

that IAA may have reproductive toxicity. Hence, T-screen and OECD Test Guideline 407

337

assays are effective screening tests for evaluating the endocrine-mediated effects and

338

identifying thyroid toxicants. However, the effects of IAA on the physiological characteristics

339

of TH are more complex than previously assumed. Further mechanistic studies are therefore

340

required to elucidate IAA-induced anti-thyroid effects.

341 342

Associated Content

343

Supporting Information Available

344

Table S1. Primer sequences for RT-qPCR. 13

ACS Paragon Plus Environment

Environmental Science & Technology

Page 14 of 32

345

Table S2. Clinical biochemistry values in SD rats exposed to IAA for 28 days.

346

Fig. S1. Body weight changes in male and female SD rats treated with IAA for 28 days. Data

347

are shown as the mean ± SD, n=7.

348

Fig. S2. Food consumption of males and females SD rats treated with IAA for 28 days. Data

349

are shown as the mean of each group, n=7.

350

Fig. S3. Histological micrographs of the liver of SD rats treated with IAA for 28 days. (A)

351

Normal liver cells. (B) Slight edemas of liver cells were frequently observed in IAA-treated

352

rats.

353

Fig. S4. Immunohistochemical micrographs of NIS (A), TSHR (B), TG (C), and TPO (D)

354

protein expression in the thyroid of SD rats treated with IAA for 28 days.

355 356 357

Acknowledgements

358

This research was supported by grants from National Natural Science Foundation of

359

China (No. 81360421, 81560524), the China Postdoctoral Science Foundation (No.

360

2014T70839,

361

2012GXNSFBA053109), the Outstanding Young and Middle-aged Excellent Teachers'

362

Training in Higher Education Institutions of Guangxi (No. GXQG022014019), and the

363

Guangxi Medical University Training Program for Distinguished Young Scholars (No. 2017).

2013M540686),

the

Guangxi

Natural

Science

Foundation

(No.

364

We thank Professor Gang Chen and Dr. Jingjing Zeng (Department of Pathology,

365

Guangxi Medical University) for their technical support in the histological analyses. The

366

authors would like to thank the anonymous reviewers and the editor for their comments and

367

suggestions.

14

ACS Paragon Plus Environment

Page 15 of 32

Environmental Science & Technology

368

References

369

(1) Plewa, M. J.; Wagner, E. D.; Richardson, S. D.; Thruston, A. D., Jr.; Woo, Y. T.;

370

McKague, A. B., Chemical and biological characterization of newly discovered iodoacid

371

drinking water disinfection byproducts. Environ. Sci. Technol. 2004, 38 (18), 4713-4722.

372

(2) Krasner, S. W.; Weinberg, H. S.; Richardson, S. D.; Pastor, S. J.; Chinn, R.; Sclimenti, M.

373

J.; Onstad, G. D.; Thruston, A. D., Occurrence of a new generation of disinfection byproducts.

374

Environ. Sci. Technol. 2006, 40 (23), 7175-7185.

375

(3) Richardson, S. D.; Fasano, F.; Ellington, J. J.; Crumley, F. G.; Buettner, K. M.; Evans, J.

376

J.; Blount, B. C.; Silva, L. K.; Waite, T. J.; Luther, G. W.; McKague, A. B.; Miltner, R. J.;

377

Wagner, E. D.; Plewa, M. J., Occurrence and Mammalian Cell Toxicity of Iodinated

378

Disinfection Byproducts in Drinking Water. Environ. Sci. Technol. 2008, 42 (22), 8330-8338.

379

(4) Wei, X.; Chen, X.; Wang, X.; Zheng, W.; Zhang, D.; Tian, D.; Jiang, S.; Ong, C. N.; He,

380

G.; Qu, W., Occurrence of regulated and emerging iodinated DBPs in the Shanghai drinking

381

water. PLoS One 2013, 8 (3), e59677.

382

(5) Attene-Ramos, M. S.; Wagner, E. D.; Plewa, M. J., Comparative human cell

383

toxicogenomic analysis of monohaloacetic acid drinking water disinfection byproducts.

384

Environ. Sci. Technol. 2010, 44 (19), 7206-7212.

385

(6) Pals, J. A.; Ang, J. K.; Wagner, E. D.; Plewa, M. J., Biological mechanism for the toxicity

386

of haloacetic acid drinking water disinfection byproducts. Environ. Sci. Technol. 2011, 45 (13),

387

5791-5797.

388

(7) Dad, A.; Jeong, C. H.; Pals, J. A.; Wagner, E. D.; Plewa, M. J., Pyruvate remediation of

389

cell stress and genotoxicity induced by haloacetic acid drinking water disinfection

390

by-products. Environ. Mol. Mutagen. 2013, 54 (8), 629-637.

391

(8) Pals, J.; Attene-Ramos, M. S.; Xia, M.; Wagner, E. D.; Plewa, M. J., Human cell

392

toxicogenomic analysis linking reactive oxygen species to the toxicity of monohaloacetic acid

393

drinking water disinfection byproducts. Environ. Sci. Technol. 2013, 47 (21), 12514-12523.

394

(9) Wei, X.; Wang, S.; Zheng, W.; Wang, X.; Liu, X.; Jiang, S.; Pi, J.; Zheng, Y.; He, G.; Qu,

395

W.,

Drinking

water

disinfection

byproduct

iodoacetic

15

ACS Paragon Plus Environment

acid

induces

tumorigenic

Environmental Science & Technology

396

transformation of NIH3T3 cells. Environ. Sci. Technol. 2013, 47 (11), 5913-5920.

397

(10) Richardson, S. D.; Plewa, M. J.; Wagner, E. D.; Schoeny, R.; DeMarini, D. M.,

398

Occurrence, genotoxicity, and carcinogenicity of regulated and emerging disinfection

399

by-products in drinking water: A review and roadmap for research. Mutat. Res. 2007, 636

400

(1-3), 178-242.

401

(11) Plewa, M. J.; Simmons, J. E.; Richardson, S. D.; Wagner, E. D., Mammalian cell

402

cytotoxicity and genotoxicity of the haloacetic acids, a major class of drinking water

403

disinfection by-products. Environ. Mol. Mutagen. 2010, 51 (8-9), 871-878.

404

(12) Wright, J. M.; Schwartz, J.; Dockery, D. W., The effect of disinfection by-products and

405

mutagenic activity on birth weight and gestational duration. Environ. Health Perspect. 2004,

406

112 (8), 920-925.

407

(13) Jeong, C. H.; Wagner, E. D.; Siebert, V. R.; Anduri, S.; Richardson, S. D.; Daiber, E. J.;

408

McKague, A. B.; Kogevinas, M.; Villanueva, C. M.; Goslan, E. H.; Luo, W.; Isabelle, L. M.;

409

Pankow, J. F.; Grazuleviciene, R.; Cordier, S.; Edwards, S. C.; Righi, E.; Nieuwenhuijsen, M.

410

J.; Plewa, M. J., Occurrence and Toxicity of Disinfection Byproducts in European Drinking

411

Waters in Relation with the HIWATE Epidemiology Study. Environ. Sci. Technol. 2012, 46

412

(21), 12120-12128.

413

(14) Toledano, M. B.; Nieuwenhuijsen, M. J.; Best, N.; Whitaker, H.; Hambly, P.; de Hoogh,

414

C.; Fawell, J.; Jarup, L.; Elliott, P., Relation of trihalomethane concentrations in public water

415

supplies to stillbirth and birth weight in three water regions in England. Environ. Health

416

Perspect. 2005, 113 (2), 225-232.

417

(15) Nieuwenhuijsen, M. J.; Toledano, M. B.; Bennett, J.; Best, N.; Hambly, P.; de Hoogh, C.;

418

Wellesley, D.; Boyd, P. A.; Abramsky, L.; Dattani, N.; Fawell, J.; Briggs, D.; Jarup, L.; Elliott,

419

P., Chlorination disinfection by-products and risk of congenital anomalies in England and

420

Wales. Environ. Health Perspect. 2008, 116 (2), 216-222.

421

(16) Rivera-Nunez, Z.; Wright, J. M., Association of brominated trihalomethane and

422

haloacetic acid exposure with fetal growth and preterm delivery in Massachusetts. J. Occup.

423

Environ. Med. 2013, 55 (10), 1125-1134. 16

ACS Paragon Plus Environment

Page 16 of 32

Page 17 of 32

Environmental Science & Technology

424

(17) Wright, J. M.; Evans, A.; Kaufman, J. A.; Rivera-Nunez, Z.; Narotsky, M. G.,

425

Disinfection By-Product Exposures and the Risk of Specific Cardiac Birth Defects. Environ.

426

Health Perspect. 2017, 125 (2), 269-277.

427

(18) Hunter, E. S., 3rd; Rogers, E. H.; Schmid, J. E.; Richard, A., Comparative effects of

428

haloacetic acids in whole embryo culture. Teratology 1996, 54 (2), 57-64.

429

(19) Richard, A. M.; Hunter, E. S., 3rd, Quantitative structure-activity relationships for the

430

developmental toxicity of haloacetic acids in mammalian whole embryo culture. Teratology

431

1996, 53 (6), 352-360.

432

(20) Bernal, J., Action of thyroid hormone in brain. J Endocrinol. Invest. 2002, 25 (3),

433

268-288.

434

(21) Colicchia, M.; Campagnolo, L.; Baldini, E.; Ulisse, S.; Valensise, H.; Moretti, C.,

435

Molecular basis of thyrotropin and thyroid hormone action during implantation and early

436

development. Hum. Reprod. Update 2014, 20 (6), 884-904.

437

(22) Zoeller,

438

hypothalamic-pituitary-thyroid (HPT) axis. Crit. Rev. Toxicol. 2007, 37 (1-2), 11-53.

439

(23) Boas, M.; Feldt-Rasmussen, U.; Skakkebaek, N. E.; Main, K. M., Environmental

440

chemicals and thyroid function. Eur. J. Endocrinol. 2006, 154 (5), 599-611.

441

(24) Hohenwarter, O.; Waltenberger, A.; Katinger, H., An in vitro test system for thyroid

442

hormone action. Anal. Biochem. 1996, 234 (1), 56-59.

443

(25) Wang, S.; Zheng, W.; Liu, X.; Xue, P.; Jiang, S.; Lu, D.; Zhang, Q.; He, G.; Pi, J.;

444

Andersen, M. E.; Tan, H.; Qu, W., Iodoacetic acid activates Nrf2-mediated antioxidant

445

response in vitro and in vivo. Environ. Sci. Technol. 2014, 48 (22), 13478-13488.

446

(26) OECD guideline for the testing of chemicals: Repeated dose 28-day oral toxicity study in

447

rodents 407; Organisation for Economic Co-operation and Development (OECD): Paris,

448

France,

449

http://www.oecd-ilibrary.org/environment/test-no-407-repeated-dose-28-day-oral-toxicity-stu

450

dy-in-rodents_9789264070684-en;jsessionid=9012m5poq3np.x-oecd-live-03

451

(27) Walter, M. A.; Christ-Crain, M.; Schindler, C.; Muller-Brand, J.; Muller, B., Outcome of

R.

T.;

Tan,

S.

W.;

Tyl,

R.

W.,

General

background

on

the

2008;

17

ACS Paragon Plus Environment

Environmental Science & Technology

452

radioiodine therapy without, on or 3 days off carbimazole: a prospective interventional

453

three-group comparison. Eur. J. Nucl. Med. Mol. Imaging 2006, 33 (6), 730-737.

454

(28) Wang, Y.; Rogan, W. J.; Chen, P. C.; Lien, G. W.; Chen, H. Y.; Tseng, Y. C.; Longnecker,

455

M. P.; Wang, S. L., Association between maternal serum perfluoroalkyl substances during

456

pregnancy and maternal and cord thyroid hormones: Taiwan maternal and infant cohort study.

457

Environ. Health Perspect. 2014, 122 (5), 529-534.

458

(29) Liu, C.; Ha, M.; Cui, Y.; Wang, C.; Yan, M.; Fu, W.; Quan, C.; Zhou, J.; Yang, K., JNK

459

pathway decreases thyroid hormones via TRH receptor: a novel mechanism for disturbance of

460

thyroid hormone homeostasis by PCB153. Toxicology 2012, 302 (1), 68-76.

461

(30) Liu, C.; Li, L.; Ha, M.; Qi, S.; Duan, P.; Yang, K., The PI3K/Akt and ERK pathways

462

elevate thyroid hormone receptor beta1 and TRH receptor to decrease thyroid hormones after

463

exposure to PCB153 and p,p'-DDE. Chemosphere 2015, 118, 229-238.

464

(31)Han, C.; Xia, X.; Liu, A.; Zhang, X.; Zhou, M.; Xiong, C.; Liu, X.; Sun, J.; Shi, X.; Shan,

465

Z.; Teng, W., Circulating Betatrophin Is Increased in Patients with Overt and Subclinical

466

Hypothyroidism. BioMed research international 2016, 2016, 5090852.

467

(32) Yu, W. G.; Liu, W.; Jin, Y. H.; Liu, X. H.; Wang, F. Q.; Liu, L.; Nakayama, S. F., Prenatal

468

and postnatal impact of perfluorooctane sulfonate (PFOS) on rat development: a cross-foster

469

study on chemical burden and thyroid hormone system. Environ. Sci. Technol. 2009, 43 (21),

470

8416-8422.

471

(33) Pagano, L.; Servin, A. D.; De La Torre-Roche, R.; Mukherjee, A.; Majumdar, S.;

472

Hawthorne, J.; Marmiroli, M.; Maestri, E.; Marra, R. E.; Isch, S. M.; Dhankher, O. P.; White,

473

J. C.; Marmiroli, N., Molecular Response of Crop Plants to Engineered Nanomaterials.

474

Environ. Sci. Technol. 2016, 50 (13), 7198-7207.

475

(34) Samuels, H. H.; Tsai, J. S.; Cintron, R., Thyroid hormone action: a cell-culture system

476

responsive to physiological concentrations of thyroid hormones. Science 1973, 181 (4106),

477

1253-1256.

478

(35) Hinkle, P. M.; Kinsella, P. A., Thyroid hormone induction of an autocrine growth factor

479

secreted by pituitary tumor cells. Science 1986, 234 (4783), 1549-1552. 18

ACS Paragon Plus Environment

Page 18 of 32

Page 19 of 32

Environmental Science & Technology

480

(36) Gutleb, A. C.; Meerts, I. A.; Bergsma, J. H.; Schriks, M.; Murk, A. J., T-Screen as a tool

481

to identify thyroid hormone receptor active compounds. Environ. Toxicol. Pharmacol. 2005,

482

19 (2), 231-238.

483

(37) Zhang, Q.; Ji, C.; Yin, X.; Yan, L.; Lu, M.; Zhao, M., Thyroid hormone-disrupting

484

activity and ecological risk assessment of phosphorus-containing flame retardants by in vitro,

485

in vivo and in silico approaches. Environ. Pollut. 2016, 210, 27-33.

486

(38) Ghisari, M.; Long, M.; Tabbo, A.; Bonefeld-Jorgensen, E. C., Effects of currently used

487

pesticides and their mixtures on the function of thyroid hormone and aryl hydrocarbon

488

receptor in cell culture. Toxicol. Appl. Pharmacol. 2015, 284 (3), 292-303.

489

(39) Guo, Y.; Zhou, B., Thyroid endocrine system disruption by pentachlorophenol: an in

490

vitro and in vivo assay. Aquat. Toxicol. 2013, 142-143, 138-145.

491

(40) Chen, Q.; Wang, X.; Shi, W.; Yu, H.; Zhang, X.; Giesy, J. P., Identification of Thyroid

492

Hormone Disruptors among HO-PBDEs: In Vitro Investigations and Coregulator Involved

493

Simulations. Environ. Sci. Technol. 2016, 50 (22), 12429-12438.

494

(41) van der Ven, L. T.; Verhoef, A.; van de Kuil, T.; Slob, W.; Leonards, P. E.; Visser, T. J.;

495

Hamers, T.; Herlin, M.; Hakansson, H.; Olausson, H.; Piersma, A. H.; Vos, J. G., A 28-day

496

oral dose toxicity study enhanced to detect endocrine effects of hexabromocyclododecane in

497

Wistar rats. Toxicol. Sci. 2006, 94 (2), 281-292.

498

(42)Yamasaki, K.; Miyata, K.; Shiraishi, K.; Muroi, T.; Higashihara, N.; Oshima, H.; Minobe,

499

Y., Uterotrophic assay, Hershberger assay, and subacute oral toxicity study of

500

4,4'-butylidenebis(2-tert-butyl-5-methylphenol) and 3-(dibutylamino)phenol, based on the

501

OECD draft protocols. Arch. Toxicol. 2008, 82 (5), 301-311.

502

(43)Shin, J. H.; Moon, H. J.; Kang, I. H.; Kim, T. S.; Kim, I. Y.; Park, I. S.; Kim, H. S.; Jeung,

503

E. B.; Han, S. Y., Repeated 28-day oral toxicity study of ketoconazole in rats based on the

504

draft protocol for the "Enhanced OECD Test Guideline No. 407" to detect endocrine effects.

505

Arch. Toxicol. 2006, 80 (12), 797-803.

506

(44) Jeong, C. H.; Gao, L.; Dettro, T.; Wagner, E. D.; Ricke, W. A.; Plewa, M. J.; Flaws, J. A.,

507

Monohaloacetic acid drinking water disinfection by-products inhibit follicle growth and 19

ACS Paragon Plus Environment

Environmental Science & Technology

508

steroidogenesis in mouse ovarian antral follicles in vitro. Reprod. Toxicol. 2016, 62, 71-76.

509

(45) Yen, P. M., Physiological and molecular basis of thyroid hormone action. Physiol. Rev.

510

2001, 81 (3), 1097-1142.

511

(46) Dai, G.; Levy, O.; Carrasco, N., Cloning and characterization of the thyroid iodide

512

transporter. Nature 1996, 379 (6564), 458-460.

513

(47) Wolff, J., Perchlorate and the thyroid gland. Pharmacol. Rev. 1998, 50 (1), 89-105.

514

(48) Li, Q.; Mair, C.; Schedle, K.; Hellmayr, I.; Windisch, W., Effects of varying dietary

515

iodine supplementation levels as iodide or iodate on thyroid status as well as mRNA

516

expression and enzyme activity of antioxidative enzymes in tissues of grower/finisher pigs.

517

Eur J Nutr 2013, 52 (1), 161-168.

518

(49)Czarnywojtek, A.; Krysinska, I.; Lacka, K.; Stawny, B.; Rolski, M.; Jarzab, B.; Wloch, J.;

519

Gembicki, M., A study of thyroglobulin concentration in the thyroid and serum of patients

520

with different thyroid disorders. Arch. Immunol. Ther. Exp. (Warsz.) 2002, 50 (2), 143-148.

521

(50) Parkes, A. B.; Black, E. G.; Adams, H.; John, R.; Richards, C. J.; Hall, R.; Lazarus, J. H.,

522

Serum thyroglobulin: an early indicator of autoimmune post-partum thyroiditis. Clin.

523

Endocrinol. (Oxf) 1994, 41 (1), 9-14.

524

(51) St Germain, D. L.; Galton, V. A., The deiodinase family of selenoproteins. Thyroid 1997,

525

7 (4), 655-668.

526

(52) Fernandez, N. J.; Kidney, B. A., Alkaline phosphatase: beyond the liver. Vet Clin Pathol

527

2007, 36 (3), 223-233.

528

(53) Lammers, W. J.; van Buuren, H. R.; Hirschfield, G. M.; Janssen, H. L.; Invernizzi, P.;

529

Mason, A. L.; Ponsioen, C. Y.; Floreani, A.; Corpechot, C.; Mayo, M. J.; Battezzati, P. M.;

530

Pares, A.; Nevens, F.; Burroughs, A. K.; Kowdley, K. V.; Trivedi, P. J.; Kumagi, T.; Cheung,

531

A.; Lleo, A.; Imam, M. H.; Boonstra, K.; Cazzagon, N.; Franceschet, I.; Poupon, R.;

532

Caballeria, L.; Pieri, G.; Kanwar, P. S.; Lindor, K. D.; Hansen, B. E., Levels of alkaline

533

phosphatase and bilirubin are surrogate end points of outcomes of patients with primary

534

biliary cirrhosis: an international follow-up study. Gastroenterology 2014, 147 (6), 1338-1349.

535

e5. 20

ACS Paragon Plus Environment

Page 20 of 32

Page 21 of 32

Environmental Science & Technology

536

(54) Pratt, D. S.; Kaplan, M. M., Evaluation of abnormal liver-enzyme results in

537

asymptomatic patients. N. Engl. J. Med. 2000, 342 (17), 1266-1271.

538

(55) Milne, M.; Kang, M. I.; Quail, J. M.; Baran, D. T., Thyroid hormone excess increases

539

insulin-like growth factor I transcripts in bone marrow cell cultures: divergent effects on

540

vertebral and femoral cell cultures. Endocrinology 1998, 139 (5), 2527-2534.

541

(56) Hamers, T.; Kamstra, J. H.; Sonneveld, E.; Murk, A. J.; Kester, M. H.; Andersson, P. L.;

542

Legler, J.; Brouwer, A., In vitro profiling of the endocrine-disrupting potency of brominated

543

flame retardants. Toxicol. Sci. 2006, 92 (1), 157-173.

544

(57) Shetty, D.; Yung, G.; Russo, R.; Mansberg, R., Incidental 99mTc-HDP Uptake in a

545

Dominant Thyroid Nodule on Bone Scan. Clin Nucl Med 2017, 42 (5), 386-388.

546

(58) Ito, M.; Miyauchi, A.; Hisakado, M.; Yoshioka, W.; Ide, A.; Kudo, T.; Nishihara, E.;

547

Kihara, M.; Ito, Y.; Kobayashi, K.; Miya, A.; Fukata, S.; Nishikawa, M.; Nakamura, H.;

548

Amino, N., Biochemical Markers Reflecting Thyroid Function in Athyreotic Patients on

549

Levothyroxine Monotherapy. Thyroid 2017, 27 (4), 484-490.

550

(59)Kunt, H.; Senturk, I.; Gonul, Y.; Korkmaz, M.; Ahsen, A.; Hazman, O.; Bal, A.; Genc, A.;

551

Songur, A., Effects of electromagnetic radiation exposure on bone mineral density, thyroid,

552

and oxidative stress index in electrical workers. Onco Targets Ther 2016, 9, 745-754.

553

(60) Hoseini, S. M.; Hedayati, A.; Ghelichpour, M., Plasma metabolites, ions and thyroid

554

hormones levels, and hepatic enzymes activity in Caspian roach (Rutilus rutilus caspicus)

555

exposed to waterborne manganese. Ecotoxicol. Environ. Saf. 2014, 107, 84-89.

556

(61) Smith, P. J.; Surks, M. I., 5,5'-Diphenylhydantoin (dilantin) decreases cytosol and

557

specific nuclear 3,5,3'-triiodothyronine binding in rat anterior pituitary in vivo and in cultured

558

GC cells. Endocrinology 1984, 115 (1), 283-290.

559

(62) Zemel, L. R.; Biezunski, D. R.; Shapiro, L. E.; Surks, M. I., 5,5'-Diphenylhydantoin

560

decreases the entry of 3,5,3'-triiodo-L-thyronine but not L-thyroxine in cultured

561

GH-producing cells. Acta Endocrinol. 1988, 117 (3), 392-398.

21

ACS Paragon Plus Environment

Environmental Science & Technology

562

Table captions

563

Table 1. Relative organ weights in SD rats exposed to IAA for 28 days.

564

Table 2. Serum levels of hormones, TG and TPO in SD rats exposed to IAA for 28 days.

565

Table 3. Histopathological changes in the thyroid of SD rats exposed to IAA for 28 days at

566

different doses (mg/kg bw).

567 568

Figure captions

569

Fig. 1. (A) Dose-response relationship of the T3-activated GH3 cell response. (B)

570

Cytotoxicity of IAA to GH3 cell. (C) Antagonistic effects of different concentrations of IAA

571

in the T-screen assay. Data represent the mean ± SE from triplicate experiment. *P< 0.05,

572

Treatment groups compared with the control. #P< 0.05, 2 µM IAA treatment groups

573

compared with the 0 µM IAA treatment group in the presence of T3.

574 575

Fig. 2. Histological micrographs of the thyroid of SD rats treated with IAA for 28 days. (A)

576

Normal thyroid follicles. (B, C) Follicles are smaller (B) or even depleted (C). (D) Follicles

577

are more vacuolated.

578 579

Fig. 3. Gene expressions levels in the thyroid and liver of SD rats treated with IAA for 28

580

days. Data are the mean ± SD in each group, n=7. *P< 0.05, IAA treatment groups compared

581

with the negative control.

582 583

Fig. 4. Expression of NIS, TSHR, TG, and TPO protein in the thyroid of SD rats treated with

584

IAA for 28 days. Data are the mean ± SD in each group, n=7. *P< 0.05, IAA treatment groups

585

compared with the negative control.

22

ACS Paragon Plus Environment

Page 22 of 32

Page 23 of 32

Environmental Science & Technology

586

Table 1. Relative organ weights in SD rats exposed to IAA for 28 days. Relative organ weights

Control

IAA (mg/kg bw) 6

12

24

Male Hypothalamus (‰)

0.148±0.042

0.157±0.065

0.163±0.045

0.176±0.047

Pituitary (‰)

0.034±0.004

0.033±0.002

0.032±0.003

0.031±0.003

Thyroid (‰)

0.047±0.006

0.044±0.005

0.047±0.009

0.048±0.010

Kidney (%)

0.686±0.046

0.714±0.042

0.706±0.024

0.702±0.042

Liver (%)

4.220±0.212

4.022±0.214

3.851±0.294*

3.793±0.268*

Thymus (%)

0.183±0.022

0.192±0.021

0.176±0.022

0.174±0.039

Heart (%)

0.329±0.023

0.302±0.017*

0.307±0.017

0.296±0.016*

Testes (%)

1.019±0.091

0.997±0.084

1.081±0.081

1.119±0.168

Epididymides (%)

0.246±0.024

0.268±0.031

0.271±0.016

0.265±0.028

Prostate (%)

0.123±0.023

0.145±0.009

0.134±0.020

0.120±0.018

Hypothalamus (‰)

0.187±0.043

0.311±0.070*

0.344±0.071*

0.281±0.044*

Pituitary (‰)

0.062±0.006

0.060±0.008

0.057±0.011

0.059±0.007

Thyroid (‰)

0.049±0.009

0.056±0.006

0.058±0.007

0.055±0.010

Kidney (%)

0.719±0.054

0.784±0.041

0.724±0.058

0.756±0.063

Liver (%)

3.707±0.172

3.694±0.162

3.683±0.132

3.764±0.290

Thymus (%)

0.217±0.025

0.201±0.032

0.231±0.041

0.236±0.034

Female

23

ACS Paragon Plus Environment

Environmental Science & Technology

Page 24 of 32

Heart (%)

0.324±0.026

0.364±0.038

0.338±0.043

0.328±0.018

Ovaries (%)

0.091±0.020

0.063±0.005*

0.066±0.012*

0.062±0.012*

Uterus (%)

0.047±0.016

0.040±0.017

0.033±0.004

0.041±0.018

587

Data are expressed as the mean ± SD, n=7.

588

* Significantly different from the control at P < 0.05.

24

ACS Paragon Plus Environment

Page 25 of 32

Environmental Science & Technology

589

Table 2. Serum levels of hormones, TG, and TPO in SD rats exposed to IAA for 28 days. Parameters

Control

IAA (mg/kg bw) 6

12

24

Male TT3 (nM /L)

0.66±0.15

0.76±0.12

0.70±0.18

0.48±0.14*

TT4 (nM /L)

65.67±16.9

68.20±11.79

58.19±14.68

50.63±8.86

FT3 (pM /L)

4.51±0.40

4.79±1.03

4.55±0.37

4.50±0.56

FT4 (pM /L)

38.01±3.84

40.64±7.18

41.16±9.13

36.29±9.07

TSH (IU/L)

0.76±0.16

1.03±0.22

2.05±0.79*

2.50±0.50*

TRH (mIU/L)

6.72±2.02

6.90±2.50

5.11±0.28

4.66±1.97

TG (ng/L)

18.03±2.61

16.88±1.39

27.63±11.90*

36.43±3.39*

TPO (µg/L)

0.06±0.01

0.15±0.07

0.35±0.06*

0.32±0.06*

TT3 (nM /L)

0.90±0.25

0.77±0.17

0.75±0.10

0.61±0.06*

TT4 (nM /L)

36.64±7.21

38.23±7.81

45.78±4.54

45.53±8.79

FT3 (pM /L)

4.02±0.78

4.27±0.82

4.64±1.04

3.90±0.70

FT4 (pM /L)

38.87±3.38

39.47±4.76

40.70±13.66

37.16±8.82

TSH (IU/L)

2.67±0.85

1.98±0.88

2.44±0.81

2.63±0.99

TRH (mIU/L)

3.40±1.26

5.38±1.44

5.90±1.61*

5.19±1.43

TG (ng/L)

32.54±7.51

40.31±6.14

62.58±18.96*

73.69±0.87*

TPO (µg/L)

0.05±0.03

0.15±0.08

0.26±0.11*

0.33±0.05*

Female

25

ACS Paragon Plus Environment

Environmental Science & Technology

590

Data are expressed as the mean ± SD, n=7.

591

* Significantly different from the control at P < 0.05.

26

ACS Paragon Plus Environment

Page 26 of 32

Page 27 of 32

Environmental Science & Technology

592

Table 3. Histopathological changes in the thyroid of SD rats exposed to IAA for 28 days at

593

different doses (mg/kg bw).

Organ findings

Male Control

6

Female 12

24

Control

6

Male + Female 12

24

Control

6

12

24

Vacuolization

0/7a

0/7 0/7 1/7

0/7

0/7 0/7 0/7

0/14b

0/14 0/14 1/14

Smaller follicles

1/7

2/7 3/7 4/7

1/7

1/7 2/7 4/7

2/14

3/14 5/14 8/14*

Depleted follicles

2/7

3/7 2/7 3/7

1/7

2/7 2/7 2/7

3/14

5/14 4/14 5/14

594

a Incidence: Affected rats/total examined (n=7).

595

b Incidence: Affected rats/total examined (n=14).

596

* Significantly different from the control at P < 0.05.

27

ACS Paragon Plus Environment

Environmental Science & Technology

Fig. 1 A

B

C

ACS Paragon Plus Environment

Page 28 of 32

Page 29 of 32

Environmental Science & Technology

Fig. 2 A

B

C

D

ACS Paragon Plus Environment

Environmental Science & Technology

Fig. 3

ACS Paragon Plus Environment

Page 30 of 32

Page 31 of 32

Environmental Science & Technology

Fig. 4

ACS Paragon Plus Environment

Environmental Science & Technology

TOC Art 46x26mm (600 x 600 DPI)

ACS Paragon Plus Environment

Page 32 of 32