New Insights into the Cytotoxic Mechanism of ... - ACS Publications

Feb 13, 2016 - E-mail: [email protected]., *Phone/Fax: 86-411-8437-9562. ... The obtained metabolomic results also provided some new evidence and ...
0 downloads 0 Views 1MB Size
Subscriber access provided by University of Otago Library

Article

New Insights into the Cytotoxic Mechanism of Hexabromocyclododecane (HBCD) from a Metabolomic Approach Feidi Wang, Haijun Zhang, Ningbo Geng, Baoqin Zhang, Xiaoqian Ren, and Jiping Chen Environ. Sci. Technol., Just Accepted Manuscript • DOI: 10.1021/acs.est.5b03678 • Publication Date (Web): 13 Feb 2016 Downloaded from http://pubs.acs.org on February 13, 2016

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

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

Page 1 of 27

Environmental Science & Technology

1

New

Insights

into

the

Cytotoxic

Mechanism

2

Hexabromocyclododecane (HBCD) from a Metabolomic Approach

3

Feidi Wang,†,‡ Haijun Zhang,†,* Ningbo Geng,†,‡ Baoqin Zhang,† Xiaoqian

4

Ren,†,‡ Jiping Chen†,*

5



6

Chinese Academy of Sciences, Dalian, 116023, China

7



Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics,

University of Chinese Academy of Sciences, Beijing 100049, China

8 9 10 11 12

of

TOC/Abstract Art

13 14 15 16 17 18

*Corresponding Authors

19

Phone: +86-411-8437-9972, fax: +86-411-8437-9562; e-mail: [email protected].

20

Phone/fax: +86-411-8437-9562; e-mail: [email protected].

ACS Paragon Plus Environment

Environmental Science & Technology

21

ABSTRACT

22

The toxic effects of hexabromocyclododecane (HBCD) are complex, and the underlying

23

toxicological mechanisms are still not completely understood. In this study, a

24

pseudo-targeted metabolomic approach based on the UHPLC/Q-Trap MS system was

25

developed to assess the HBCD-intervention-related metabolic alteration in HepG2 cells. In

26

addition, some physiologic indicators and relevant enzyme activities were measured. HBCD

27

exposure obviously impaired metabolic homeostasis and induced oxidative stress, even at

28

an environmentally relevant dose (0.05 mg/L). Metabolic profiling and multivariate

29

analysis indicated that the main metabolic pathways perturbed by HBCD included amino

30

acid metabolism, protein biosynthesis, fatty acid metabolism and phospholipid metabolism.

31

HBCD suppressed the cell uptake of amino acids, mainly through inhibition of the activity

32

of membrane transport protein Na+/K+-ATPase. HBCD down-regulated glycolysis and

33

β-oxidation of long-chain fatty acids, causing a large decrease of ATP production. As a

34

result, the across-membrane transport of amino acids was further inhibited. Meanwhile,

35

HBCD induced a significant increase of total phospholipids, mainly through the remodeling

36

of phospholipids from the increased free fatty acids. The obtained metabolomic results also

37

provided some new evidence and clues regarding the toxicological mechanisms of HBCD

38

that contribute to obesity, diabetes, nervous system damage, and developmental disorders.

39

INTRODUCTION

40

Hexabromocyclododecane (HBCD) is a brominated cyclic alkane used primarily as an

41

additive flame retardant in polystyrene-based materials including resins and fabrics.1, 2 It

42

has been listed as a new persistent organic pollutant (POP) in Annex A of the Stockholm

43

Convention in 2013,3 but with specific exemptions for expanded and extruded polystyrene

44

foams in buildings until 2024.4 Because of its persistence and lipophilicity, HBCD can

45

accumulate in the human body through a combination of diet, dust ingestion and indoor air

46

inhalation.5 As a result, HBCD has been frequently detected in human blood and breast ACS Paragon Plus Environment

Page 2 of 27

Page 3 of 27

Environmental Science & Technology

47

milk.6, 7

48

The acute toxicity of HBCD appears to be low,5 and it lacks significant genotoxic

49

potential to an organism.1, 8 However, the subacute and subchronic effects of HBCD are

50

manifest.9, 10 Hepatic toxicity and thyroid toxicity of HBCD were universally observed in in

51

vivo animal studies.11,

52

hyperplasia,14 cytochrome P450 enzyme induction,11 and thyroid hormone disruption.12

53

Furthermore, potential reproductive effects,15 nervous system damage16 and developmental

54

toxicity17 were also observed in response to HBCD exposure.

12

The adverse effects include liver weight increase,13 thyroid

55

Accumulating evidence shows that HBCD can induce endocrine disruption,13 metabolic

56

dysfunction and obesity,18 even at environmentally relevant doses. HBCD has the ability to

57

interact with the constitutive androstane receptor (CAR) and pregnane X receptor (PXR).19

58

An in vivo study indicated that oral exposure to HBCD gave rise to a dose-dependent

59

decrease of trabecular bone mineral density in female rats with a BenchMark Dose Lower

60

confidence bound (BMDL) of 0.056 mg/kg body weight/day.20 Another in vivo study found

61

that the body and liver weight were markedly increased in high-fat diet mice treated with

62

HBCD at a medium dose of 35 and a high dose of 700 µg/kg body weight/week, paralleled

63

by the disruption of lipid and glucose homeostasis.18 These low-dose effects raise serious

64

concerns for human health based on human body burdens of HBCD, particularly for the

65

occupational exposure setting.

66

In the past decade, significant advances have been made toward understanding

67

mechanisms underlying the adverse effects of HBCD. As a xenobiotic, HBCD is first

68

metabolized by cytochrome P450 enzymes, and thereby reactive oxygen species (ROS) are

69

generated.21 Overproduction of ROS likely results in oxidative damage to lipids, proteins

70

and DNA.21 Meanwhile, HBCD-induced activation of the PI3K/Akt pathway and

71

Nrf2-ARE pathway provide a cytoprotective process that responds to HBCD-induced ROS

72

generation.22, 23 HBCD can induce CYP2B and CYP3A enzymes through interaction with ACS Paragon Plus Environment

Environmental Science & Technology

73

CAR/PXR receptors, which may account for the disruption of the thyroid hormone axis.19

74

Several in vitro studies also indicated the inhibitory effect of HBCD on the

75

sarcoplasmic/endoplasmic reticulum Ca2+-ATPase, which is accompanied by an

76

intracellular Ca2+ increase and mitochondrial dysfunction.24, 25 Gene expression profiles in

77

rat livers revealed that HBCD exposure disrupted several specific pathways, such as

78

PPAR-mediated regulation of lipid metabolism, triacylglycerol metabolism, cholesterol

79

biosynthesis, and phase I and II pathways.9 Furthermore, untargeted metabolomics based on

80

in vitro study revealed that exposure to HBCD for 72 h could significantly induce the

81

metabolic changes.26 However, the metabolic pathways intervened by HBCD exposure

82

require further confirmation, and there still remains a lack of metabolomic evidence

83

supporting the underlying mechanisms of HBCD toxicity.

84

The biological events induced by HBCD appear to be complex, and the underlying

85

toxicological mechanisms of HBCD are not completely elucidated. To better understand the

86

mode of action of HBCD, metabolomic evidence is required. Metabolomics aiming to

87

systematically study small molecule metabolites is a method of understanding metabolic

88

regulation.27 It can offer a perspective on how mechanistic biochemistry relates to the

89

phenotypic state of an organism because metabolites serve as direct signatures of

90

biochemical activity.28 The intrinsic physiological responses of an organism after HBCD

91

exposure are reflected and propagated by its metabolism, and conversely, the variation in

92

metabolic fingerprints can reveal and verify the mechanisms that underlie the toxicity of

93

HBCD.29 In this study, the cytological effects of HBCD exposure on HepG2 cells were

94

examined, and a pseudo-targeted metabolomic analysis was performed to investigate

95

HBCD-intervention-related metabolic alteration. The obtained results are expected to

96

provide a better understanding of metabolic disturbances induced by HBCD exposure and

97

provide new evidence and clues concerning the toxicological mechanisms of HBCD from a

98

metabolomic perspective. ACS Paragon Plus Environment

Page 4 of 27

Page 5 of 27

Environmental Science & Technology

99

EXPERIMENTAL SECTION

100

Cell Culture, Exposure Conditions and Sample Collection. Human hepatoma

101

HepG2 cell was adopted as an in vitro model for the study of cytotoxic mechanism. HepG2

102

cell has high stability and unlimited life-span, and it retains a wide range of human

103

liver-specific functions.30 In addition, its molecular expression and biological phenotypes

104

have been extensively characterized.30 The test HepG2 cells were provided by China

105

Infrastructure of Cell Line Resources (Shanghai, China), and these cells were maintained in

106

Dulbecco’s Modified Eagle Medium (DMEM basic (1X), Gibco-BRL) supplemented with

107

10% fetal bovine serum (FBS, Gibco-BRL) and 1% Penicillin–Streptomycin (Beyotime,

108

China) under humidified air containing 5% CO2 at 37 °C. Cells in the logarithmic phase of

109

growth were rinsed with phosphate-buffered saline, trypsinized, and then seeded in culture

110

plates for exposure experiments.

111

HepG2 cells were exposed to a reagent-grade HBCD formula (purity: > 95%, Aladdin

112

Industrial Corp.) with varying concentrations in the culture medium. HBCD stock solutions

113

were made up in ACS grade dimethyl sulfoxide (DMSO; Amresco, Solon, OH, USA).

114

After cells were seeded and grown to 80% confluency, HBCD stock solution was

115

incorporated into the cell culture medium. The final DMSO content in the culture medium

116

was 0.05% (v/v), and background control cells were treated with only 0.05% DMSO.

117

Cell viability was first tested using a thiazolyl blue tetrazolium bromide (MTT, Amresco,

118

Solon, OH, USA) assay to ascertain the appropriate exposure conditions for metabolomic

119

study. HepG2 cells were exposed to a series of HBCD concentrations with incubation

120

durations of 1, 2 and 3 days. The details of the cell viability assays are shown in the

121

Supporting Information. According to the results of MTT assay (see Supporting

122

Information), an exposure time of 1 day was adopted for metabolomic study; and three

123

HBCD exposure doses relating to different effects on cell viability, the lowest dose with

124

observable inhibition effect (0.05 mg/L), a middle dose without observable effect (1 mg/L) ACS Paragon Plus Environment

Environmental Science & Technology

Page 6 of 27

125

and the highest dose with observable stimulation effect (10 mg/L), were selected. The

126

exposure dose of 0.05 mg/L is comparable with the maximum serum concentration of

127

HBCD (856 ng/g lipid) in individuals occupationally exposed.7

128

In the metabolomic study, HepG2 cells were seeded in 6-well plates at about 3 × 105

129

cells/well and incubated with HBCD at concentrations of 0.05, 1 and 10 mg/L for 24 h.

130

After exposure, 200 µL of culture medium was transferred to an Eppendorf tube for

131

analysis of extracellular metabolites, and then the remaining culture medium was removed.

132

Cells were rinsed by gently dispensing ultrapure water to the surface twice, aspirating

133

rapidly, and then quenched by liquid nitrogen freezing. The quenched cells adhering to

134

plates, together with the culture medium in Eppendorf tubes, were stored at –80 °C and

135

then extracted within 7 days.

136

Metabolomic Analysis. To analyze the intracellular metabolites, 1 mL of ultrapure

137

water was added to each well, followed by ultrasonic disruption in an ice-water bath for 3

138

min, and then the suspension containing cell fragments was transferred into an Eppendorf

139

tube. The freeze-drying of the samples was then performed simultaneously for the disrupted

140

cells containing intracellular metabolites and collected culture medium containing

141

extracellular metabolites. Soon afterwards, metabolites in the freeze-dried samples were

142

dissolved in 0.5 mL of 80% methanol, vortexed for 20 min, and centrifuged for 20 min at

143

13,000 × g and 8 °C. Finally, the supernatant was filtered by an organic phase filter and

144

transferred to a vial for metabolite analysis. Six internal standards (L-phenylalanine-d5,

145

octanoyl

146

1,2-diheptadecanoyl-sn-glycero-3-phosphoethanolamine,

147

nonadecanoic acid) served as quality controls for sample preparation and instrumental

148

analysis.

(8,8,8-D3)-L-carnitine,

1-lauroyl-2-hydroxy-sn-glycero-3-phosphocholine, hendecanoic

acid

and

149

Metabolomic analysis adopted a pseudo-targeted approach, which can display better

150

repeatability and wider linear range than the traditional Q-TOF MS-based untargeted ACS Paragon Plus Environment

Page 7 of 27

Environmental Science & Technology

151

metabolomic method.27, 31 In brief, the extracts from HepG2 cells and culture medium were

152

analyzed by a Waters Acquity Ultra Performance liquid chromatography coupled online to

153

an ABI Q-Trap 5500 (AB SCIEX, USA) system (UHPLC/Q-Trap MS) operated in multiple

154

reaction monitoring (MRM) mode, for which the MRM ion pairs were acquired from the

155

cell extracts through untargeted tandem MS using an Agilent 1290 Infinity Ultra

156

Performance liquid chromatography system coupled online to an Agilent 6540 UHD

157

Q-TOF MS (Agilent, Santa Clara, CA) system (UHPLC/ Q-TOF MS). The same condition

158

of liquid chromatography was performed on both UHPLC/Q-Trap MS and UHPLC/Q-TOF

159

MS. A total of 241 ion pairs with defined parameters were analyzed in the mode of positive

160

electrospray ionization (ESI+) with an Acquity UPLC BEH C8 column (2.1 mm × 100 mm,

161

1.7 µm, Waters, USA). In ESI– mode, an Acquity UPLC HSS T3 column (2.1 mm × 100

162

mm, 1.8 µm, Waters, USA) was used for the chromatographic separation of 59 ion pairs.

163

The details of instrumental analysis are shown in Supporting Information.

164

Sequence analysis of intracellular metabolites and extracellular metabolites were

165

performed. To ensure data quality for metabolic profiling, pooled quality control (QC)

166

samples were prepared by mixing all of the samples. During analysis of the sample

167

sequence, 7 replicates of the QC samples were inserted into the analytical sequence.

168

Validations of the method, including precision, stability, and recovery, were carried out.

169

The results indicated that this metabolomics method is reliable (see Supporting

170

Information).

171

Data Processing. MultiQuant software (3.0.1, AB SCIEX) was used for data processing

172

by analyzing the extracted ion chromatograms of the Q-Trap MS data. After the peak

173

alignment and the removal of the missing values, ion peak areas across ESI+ and ESI–

174

modes were normalized to internal standards and then merged into one data set. The

175

merged data set of each sample was further normalized to the sum of peak areas to balance

ACS Paragon Plus Environment

Environmental Science & Technology

176

the difference in cell number. A one-way analysis of variance (ANOVA) was performed

177

using SPSS PASW Statistics software (SPSS Inc., Chicago, IL), and P values of < 0.05

178

were considered as significantly different from the control. Principal component analysis

179

(PCA) and partial least squares discriminate analysis (PLS-DA) were applied with unit

180

variance (UV) scaling using SIMCA-P11.5 software (Umetrics, Sweden), and hierarchical

181

cluster analysis (HCA) was conducted using the MeV software package (version 4.8.1).

182

The correlation network was constructed using the Cytoscape software package (version

183

2.8.2). The metabolite set enrichment analysis and pathway analysis were based on

184

MetaboAnalyst, a web service for metabolomics data analysis.

185

Determination of Physiologic Indicators and Relevant Enzyme Activities. To

186

further understand and verify the results of the metabolomic study, ROS generation, ATP

187

level, several oxidative stress markers, together with activities of some crucial enzymes

188

regulating glycolytic pathway, transport across cellular membranes, β-oxidation of fatty

189

acid and phospholipid metabolism, were determined. The selected oxidative stress markers

190

included the levels of reduced glutathione (GSH) and malondialdehyde (MDA) as well as

191

the activities of superoxide dismutase (SOD) and catalase (CAT). Intracellular ROS levels

192

in HepG2 cells were assayed using a fluorescence microscope with 2,7-dichlorofuorescin

193

diacetate (DCFH-DA), according to the instructions of the ROS detection kit manufacturer

194

(Nanjing Jiancheng Bioengineering Institute, China). The specific assay kits (Nanjing

195

Jiancheng Bioengineering Institute, China) and enzyme-linked immune sorbent assay

196

(ELISA) were adopted to determine the oxidative stress biomarkers and the activities of

197

relevant metabolic enzymes, and the method details are shown in Supporting Information.

198

RESULTS AND DISCUSSION

199

Basic Physiological Effects Induced by HBCD. Intracellular ROS plays an

200

important role in cell signaling and homeostasis. In this study, HBCD exposure gave rise to

ACS Paragon Plus Environment

Page 8 of 27

Page 9 of 27

Environmental Science & Technology

201

a linear increase of intracellular ROS with increasing doses, and the increases were

202

significant at all three exposure doses (0.05, 1 and 10 mg/L) compared with the control

203

group (Figure S3). The over-production of ROS resulted in an observable oxidative stress

204

response primarily characterized by the elevated activity of CAT (Figure S4). Moreover,

205

exposure to a high-dose of HBCD caused a reduction in SOD activity and an increase of

206

GSH levels (Figure S4). The MDA level, as a lipid peroxidation indicator, was also

207

significantly altered by HBCD intervention (Figure S4).

208

Cell viability was examined based on five exposure doses (0.05, 0.5, 1, 5 and 10 mg/L)

209

for 24, 48 and 72 h. As shown in Figure S1, HBCD induced a dose- and time-dependent

210

variation of cell viability. After 1 day exposure, cell viability was decreased at a dose of

211

0.05 mg/L, whereas the viability of cells in the group with exposure dose of 1 mg/L was

212

similar to that of control group. Subsequently, cell viability increased at doses of ≥ 1.0

213

mg/L with a maximum at a dose of 10 mg/L. The increased cell viability was also observed

214

for HepG2/C3A cell exposed to HBCD at concentration of 5 µmol/L (equivalent to 3.2

215

mg/L) for 1 day.32 Exposure to HBCD at doses of ≤ 5 mg/L for 2 days significantly

216

stimulated cell proliferation. However, HBCD produced statistically significant cell

217

lethality at all doses after exposure for 3 days. Two previous in vitro studies observed an

218

obvious inhibitory effect of HBCD on HepG2 cell at concentrations of 10–100 µmol/L

219

(equivalent to 6.4–64.2 mg/L) after exposure for 3 days. 25, 33

220

Metabolic Profiling and Multivariate Analysis. A pseudo-targeted metabolomic

221

analysis was performed to profile the changes of intracellular metabolites in HepG2 cells

222

induced by HBCD intervention. A total of 300 intracellular metabolites were detected.

223

Among them, 133 metabolites were identified qualitatively using quasimolecular ions, and

224

86 metabolites were further confirmed by authentic standard samples. PLS-DA was

225

performed on the normalized data sets for all detected intracellular metabolites. The

ACS Paragon Plus Environment

Environmental Science & Technology

226

PLS-DA score plot showed a clear separation between HBCD-exposed groups and a

227

control group along the component 1 direction (Figure 1). Meanwhile, three

228

HBCD-exposed groups were clearly separated from each other along the component 2

229

direction. This result indicated that HBCD caused a significant metabolic perturbation.

230 231 232 233 234

Component 2 (14.3%)

20

10

10 mg/L 0

control 1 mg/L -10

0.05 mg/L -20 -20

-15

-10 -5 0 5 Component 1 (32.8%)

10

15

235 236

Figure 1. PLS-DA score plot of metabolites in HepG2 cells exposed to HBCD at various doses

237

for 24 h. R2 = 0.977, Q2 = 0.920.

238 239

A one-way ANOVA was performed to find the differential metabolites among different

240

treatment settings. A total of 115 intracellular metabolites showed significant changes (P
0.76 were identified quantitatively and

243

are listed in Table S1. Subsequently, hierarchical clustering was used to arrange these

244

identified metabolites based on their relative contents across groups. A heat map showed

245

five dose-response trajectories (Figure S6). Compared with the control, almost all amino

246

acids presented a significant decrease in all three HBCD-exposed groups; whereas, most of

247

the phospholipids and fatty acids displayed a significant increase in low- and middle-dose

248

groups.

249

Correlation Network of Differential Metabolites and Disturbance of Metabolic

250

Pathway. The involved pathways of each differential intracellular metabolite were

ACS Paragon Plus Environment

Page 10 of 27

Page 11 of 27

Environmental Science & Technology

251

ascertained

252

http://www.genome.jp/kegg/), and the results are listed in Table S2. To investigate the

253

latent relationships among the differential metabolites, a correlation network diagram was

254

constructed according to a comprehensive pair-wise computation of Pearson correlations

255

between the differential metabolites involved in the same metabolic pathway. As shown in

256

Figure 2, the differential metabolites were mainly bridged by fatty acids. All phospholipids,

257

including phosphatidyl choline (PC), lysophosphatidyl choline (LysoPC), phosphatidyl

258

ethanolamine (PE), lysophosphatidyl ethanolamine (LysoPE), sphingomyelin (SM) and

259

lysosphingomyelin (LysoSM), were highly up-regulated by HBCD exposure either at low-

260

and middle-doses or at a high dose. The similar up-regulation was also observed for fatty

261

acids. However, the levels of most amino acids were highly down-regulated by low- and

262

middle-dose HBCD. Unlike long-chain acylcarnitine (C18:0) and medium-chain

263

acylcarnitine (C8:0), short-chain acylcarnitine (SC-AC, C2–6) presented a high

264

down-regulation at a high dose of HBCD.

by

an

online

database

of

metabolic

pathways

(KEGG,

265

Metabolic pathway perturbation induced by HBCD intervention was further studied

266

using MetaboAnalyst on the basis of the significantly different intracelluar metabolites.

267

Pathway analysis was conducted to reveal the most relevant pathways influenced by

268

different exposure

269

(www.hmdb.ca/) ID that changed significantly upon HBCD intervention, considered to be

270

highly responsible for the perturbation of amino acid metabolism (Figure S7). The most

271

relevant pathways influenced by HBCD were Alanine, aspartate and glutamate metabolism;

272

glycine, serine and threonine metabolism; and arginine and proline metabolism.

273

Furthermore, enrichment analysis indicated that protein biosynthesis, the urea cycle,

274

pantothenate and COA biosynthesis, glutathione metabolism and ammonia recycling were

275

also disturbed (Figure S8).

doses. There were 88 metabolites with identified

ACS Paragon Plus Environment

HMDB

Environmental Science & Technology

Page 12 of 27

276

277 278

Figure 2. Metabolic correlation networks of the differential metabolites and related pathways.

279

FA:

280

lysosphingomyelin; PC: phosphatidyl choline; LysoPC: lysophosphatidyl choline; PE:

281

phosphatidyl ethanolamine; LysoPE: lysophosphatidyl ethanolamine.

fatty

acid;

SC-AC:

short

chain

acylcarnitine;

SM:

sphingomyelin;

LysoSM:

282 283

Perturbation in Amino Acid Transport and Metabolism. Amino acids are the

284

essential building blocks of proteins, energy sources, metabolite precursors and signaling

285

molecules in all living cells.34 In this study, 15 types of amino acids were supplied in

286

DMEM culture medium for HepG2 cell growth. Exposure to HBCD for 1 day did not give

287

rise to a significant variation in the extracellular total contents of amino acids (Figure 3).

288

Only histidine, isoleucine, leucine and threonine in culture medium of low- or middle-dose

289

groups showed slight but significant differences from that of control group (Figure S10).

290

However, the intracellular total contents of amino acids in the three exposure groups were

291

all down-regulated significantly (P < 0.01), and the maximum down-regulation (45.2% of

292

control) was observed at a low exposure dose (Figure 3). Except tryptophan in high-dose

293

group, all intracellular amino acid nutrients in three exposure groups were largely and

294

significantly decreased when compared with a control group (Figure S11). These results

295

suggested a possible inhibitory effect of HBCD on amino acid transport.

296

Membrane transport proteins, mainly the SLC-family, mediate the transfer of amino

ACS Paragon Plus Environment

Page 13 of 27

Environmental Science & Technology

297

acids across cell membranes.34 The transport via most of SLC-family is dependent on the

298

Na+-gradient, which is driven by Na+/K+-ATPase and ATP.34 The activity of

299

Na+/K+-ATPase and ATP levels in HepG2 cells were further determined. As shown in

300

Figure 3, HBCD exposure led to a significant reduction in Na+/K+-ATPase activity in a

301

dose-dependent manner, and the high-dose exposure group showed a reduction of 65.7% in

302

Na+/K+-ATPase activity compared with the control group. This result suggested a direct

303

inhibitory effect. Meanwhile, the levels of ATP were down-regulated significantly by

304

HBCD exposure (Figure 3). The maximum down-regulation (73.8% of control) was

305

observed at a middle exposure dose (Figure 3). Evidently, the significant inhibition of

306

Na+/K+-ATPase activity and the large decrease of ATP levels both caused the suppressed

307

transport of amino acids across the membrane, as indicated by the large down-regulation of

308

intracellular amino acids.

309

A

previous

study

found

that

HBCD

had

the

potential

to

inhibit

the

310

sarcoplasmic-endoplasmic reticulum Ca2+-ATPase by affecting ATP binding and the E2 to

311

E1 transition step.35 In this study, a HBCD-induced decrease of Ca2+-ATPase was also

312

observed (Figure S5). Ca2+-ATPase and Na+/K+-ATPase are both P-type ATPases, which

313

can catalyze the decomposition of ATP into ADP and a free phosphate ion. Therefore, the

314

molecular mechanisms by which HBCD inhibits the Na+/K+-ATPase should be similar to

315

that of Ca2+-ATPase, i.e., altering ATP binding and affecting the E2 to E1 transition step of

316

Na+/K+-ATPase.

317

A map of amino acid-related metabolic pathways was constructed according to KEGG

318

pathways (Figure S9). The levels of all intracellular amino acids were down-regulated

319

significantly after exposure to HBCD at three doses, with the exception of aspartate. The

320

significant down-regulation of amino acid metabolism inevitably affected protein

321

biosynthesis, urea cycle, pantothenate and CoA biosynthesis, glutathione metabolism and

322

ammonia recycling, as indicated by the enrichment analysis (Figure S8). ACS Paragon Plus Environment

Environmental Science & Technology

Page 14 of 27

323

327

40

SLC

100

Na+

80

110

Extracellular glucose

100

328

Na+

Glucose

60 120

50

Amino acids

90

Glucose SGLT +

Na+

Glucose

Na

GLUT

Glucose

Relative abundance (%)

326

Relative content (%)

325

120

Cell outside

324

Amino acids

∑ Extracellular amino acids

Cell intside

140

∑ Intracellular amino acids

30 20

* *

* *

* *

10 0.0015 0.0010

Intracellular glucose

0.0005

330 331 332

150 100 50

Na+/K+-ATPase *

* *

* *

+

Na+

Na

ATP

Na+/K+ -ATPase +

K

0 10 control 0.05 1 HBCD dose (mg/L)

ADP + Pi

Relative content (%)

329

Relative activity (%)

0.00000

150 100 50

ATP * *

* *

* *

0 10 control 0.05 1 HBCD dose (mg/L)

333

Figure 3. Effects of HBCD exposure on the transport of amino acids and glucose across the

334

membrane of HepG2 cells. SLC: amino acid transporter SLC-family; SGLT: glucose transporter

335

SGLT-family; GLUT: glucose transporter GLUT-family. Significant differences were indicated in

336

comparison of the control by T-test. *, P < 0.05; **, P < 0.01.

337 338

Cysteine is an important precursor of cystine, GSH and redox-sensitive proteins, which

339

are necessary antioxidants in cells.36 In this study, the contents of cysteine in three exposure

340

groups were significantly decreased by more than 50% compared with that of a control

341

group. The large down-regulation of cysteine inevitably inhibited the biosynthesis of these

342

antioxidants; otherwise, their content in cells might remarkably increase, responding to the

343

physiological need to reduce the H2O2 to H2O and increase the cellular capability to

344

eliminate the redundant ROS. In addition, the large decrease of neurotransmitters (aspartate,

345

taurine, glycine and alanine) and neurotransmitters-related metabolites (e.g., tryptophan,

346

tyrosine, valine and serotonin) might induce noticeable effects of HBCD on the functioning

347

of the nervous system.

348

Perturbation in Glucose Transport and Glycolysis. The transport of glucose across

349

the plasma membrane is mediated by two groups of glucose transporters: (I) the

ACS Paragon Plus Environment

Page 15 of 27

Environmental Science & Technology

350

Na+-dependent glucose transporter SGLT members37 and (II) the Na+-independent glucose

351

transporter GLUT family.38 These two groups of transporters are both partly expressed in

352

the liver.37, 38 Therefore, the HBCD-induced decrease of Na+/K+-ATPase activity would

353

inevitably affect the glucose transport. In this study, the intracellular glucose content in the

354

three exposure groups only presented a slight decrease, but did not show a significant

355

difference from that of the control (Figure 3). Meanwhile, the extracellular contents of

356

glucose in the three exposure groups also showed no significant differences from those of

357

the control group (Figure 3). This implies that the Na+-independent transport mediated by

358

the GLUT family should be predominant. A previous study has indicated that GLUT1 and

359

GLUT9 proteins are the major contributors to glucose influx in HepG2 cells.39

360

The slight decrease of intracellular glucose content might partly result from the

361

glycolysis inhibition produced by HBCD exposure. In order to test the hypothesis, we

362

further determined the activities of two rate-limiting enzymes of glycolysis, hexokinase

363

(HK) and phosphofructokinase (PFK), which catalyze the conversion of glucose to and the

364

conversion of fructose-6-phosphate to fructose-1,6-bisphosphate, respectively. It was found

365

that exposure to a middle-dose or high-dose of HBCD led to significant reductions of

366

49.4% and 82.1%, respectively, in PFK activity when compared with the control group

367

(Figure S5). However, only slight increases of HK activity were induced by exposure to a

368

middle-dose and high-dose of HBCD (Figure S5). On the whole, glycolysis was inhibited.

369

The inhibitory effect can be further verified by the large down-regulation of lactate content

370

(above 45% of control) in the three exposure groups, though the activities of lactate

371

dehydrogenase (LDH) were only slightly changed (Figure S5). The inhibition of glycolysis

372

partly accounted for the large reduction of ATP levels in the HepG2 cells exposed to

373

HBCD (Figure 3).

374

Perturbation in β-oxidation of Free Fatty Acids. The free fatty acids in the

ACS Paragon Plus Environment

Environmental Science & Technology

375

cytoplasm are first activated to acylcarnitines before they are carried into the mitochondria

376

and peroxisome where fatty acid β-oxidation occurs.40 As shown in Figure 4, HBCD

377

exposure significantly increased the total content of intracellular long-chain fatty acids

378

(LC-FA, C14–22) by more than 24%, and the total content of LC-AC were also up-regulated

379

significantly in the low- and middle-dose exposure groups. This result clearly indicated an

380

inhibitory effect of HBCD on the metabolism of LC-FA. The activities of two rate-limiting

381

enzymes for β-oxidation, long-chain acyl-CoA synthetase (LCACS) and long-chain

382

acyl-CoA dehydrogenase (LCACD), were further determined. Compared with control

383

group, the activities of LCACD in the three exposure groups were not changed significantly

384

(Figure S5), while the activities of LCACS showed significant decreases of above 45% in

385

three exposure groups (Figure 4). The large reduction in the activity of LCACS could be

386

responsible for the significant suppression of LC-FA β-oxidation. The decreased activity of

387

LCACS could mainly result from the HBCD-induced down-regulation in the expression of

388

genes relevant to LCACS, such as ACSL1, ACSL3, ACSL4 and ACSL5, which has been

389

indicated by the hepatic gene expression profiles in rats exposed to HBCD.9

390

Mitochondria are responsible for the oxidation of the major portion of fatty acids, while

391

very long-chain fatty acids (VLC-FA, C>22) are exclusively oxidized in the peroxisome.41 In

392

this study, the level of intracellular VLC-FA was increased significantly (P < 0.01) only in

393

HepG2 cells exposed to a low-dose of HBCD (Figure 4). Furthermore, MC-AC, as an

394

activated product of medium-chain fatty acids, also showed slightly increased in the three

395

exposure groups compared with the control group (Figure 4). However, the total contents of

396

the SC-AC were largely down-regulated by the low-dose and high-dose HBCD, suggesting

397

an accelerated oxidation of SC-FA (Figure 4). On the whole, the β-oxidation of fatty acids

398

was down-regulated mainly due to the inhibited oxidation of LC-FA, which predominates

399

over free fatty acids in HepG2 cells. The down-regulation of fatty acid β-oxidation is

ACS Paragon Plus Environment

Page 16 of 27

Page 17 of 27

Environmental Science & Technology

400

another important reason for the energy deficiency in cells, as indicated by the lower ATP

401

levels.

0.10

VLC-FA

0.08

40

* *

30

0.06

405 406 407

* *

* *

* *

20

0.003 0.002

* *

*

0.001 0.000

LC-acyl-CoA

SC-AC

LC-AC

VLC-acyl--CoA VLC-AC

80 60 40

LC-FA

LCACS

* *

* *

* *

20

SC-AC

LC-acyl-CoA

MC-FA

Cytosol

0.2

VLC-FA

VLC-AC MC-AC VLC-acyl-CoA MC-acyl-CoA

LC-AC

0

O.M.

Inter-membrane space

0.6

0.05

0.4

MC-AC

I.M.

0.04

* *

* *

SC-AC

0.03

SC-acyl-CoA

0.02

409

Peroxisome lumen

SC-acyl-CoA

100 LC- AC

SC-AC

408

VLC-acyl--CoA β-oxidation

0.02

Relative activity (%)

404

Relative abundance (%)

0.04

LC-FA

VLCACS

403

b

0.12

MCACS

a

LCACS

402

0.0 control 0.05

1

10

control 0.05

1

LC-AC

MC-AC

LC-acyl-CoA β-oxidation

Mitochondrial matrix

MC-acyl-CoA

10

TAC

Acetyl-CoA

HBCD dose (mg/L)

410

Figure 4. Effects of HBCD exposure on fatty acid (FA) metabolism. a) Relative abundances; b)

411 412

Metabolic pathways. AC: acylcarnitine; VLC: very long-chain; MC: medium-chain; SC:

413

long-chain acyl-CoA synthetase; MCACS: medium-chain acyl-CoA synthetase. O.M.: out

414

membrane; I.M.: inner membrane. Significant differences were indicated in comparison of the

415

control by T-test. *, P < 0.05; **, P < 0.01.

short-chain;

VLCACS:

very

long-chain

acyl-CoA synthetase;

LCACS:

416 417

Perturbation in Phospholipid Metabolism. Phospholipids are a major component of

418

all cell membranes and are also involved in key regulatory functions within cells.42 As

419

shown in Figure 5, the metabolism of intracellular phospholipid was obviously disturbed. In

420

the glycerophospholipid pathway, the major membrane component PC and its hydrolysis

421

product lysoPC were significantly up-regulated at all three exposure doses, and the total

422

contents of lysoPC metabolites, glycerophosphocholine and choline, also showed

423

significant increases in the three exposure groups compared with the control group (Figure

424

5). Moreover, HBCD elevated lysoPE levels at low and middle exposure doses, but PE

ACS Paragon Plus Environment

Environmental Science & Technology

Page 18 of 27

425

levels were only slightly increased in the HBCD exposure groups without significant

426

differences when compared to the control group (Figure 5). The slight increase of PE could

427

be attributed to the inhibited biosynthesis of its direct precursor, phosphatidylserine,

428

resulting from the large decrease of L-serine in the cells exposed to HBCD. Furthermore,

429

SM and lysoSM in the sphingolipid pathway were both significantly increased in the cells

430

exposed to a low-dose and middle-dose of HBCD (Figure 5).

433 434

30 20

3.0 LysoPC * * * *

2 * *

10

GP-C

* *

1

Choline (C) * * * *

2.0 1.5

0

14 PC * *

0.8 * *

8

* *

CDP-C

Phospho-C

0.6

GP: Glycerophosphate

PC: Phosphatidyl choline

PS: Phosphatidyl serine

CDP: Cytidine diphosphate

PLA1: Phospholipase A1

PE: Phosphatidyl ethanolamine

PLA2: Phospholipase A2

S1P: Sphingosine-1-phosphate

AA: Arachidonic acid

SM

LysoSM * *

10 * *

* *

8

0.4

6

SM: Sphingomyelin

12

1.0

12 10

2.5

* *

4

1

435 PE

150 Phospho-E

S1P

Sphingosine

0.4

436 0.3

437

CDP-E

1.0

3 LysoPE

2

*

0.8

* *

0.6 GP-E

438

1 control 0.05 1 10 HBCD dose (mg/L)

Ethanolamine (E)

0.4

L-serine

* *

* *

* *

Ceramide

0.2 control 0.05 1 10 HBCD dose (mg/L)

* *

Phospholipids containing AA

PLA1

100 Relative activity (%)

PS

* *

* *

2

6

0.2

0.5

3 * *

PLA2

432

Relative abundance (%)

431

50

150

8

PLA2

100

AA

*

6

* *

*

50

4

0 control 0.05 1 10 HBCD dose (mg/L)

control 0.05 1 10 HBCD dose (mg/L)

2

439 440

Figure 5. Metabolic changes of phospholipids after exposure to HBCD. Significant differences

441

were indicated in comparison to the control by T-test. *, P < 0.05; **, P < 0.01.

442 443

Phospholipids can be hydrolyzed by phospholipase A1 (PLA1) and phospholipase A2

444

(PLA2) to form free fatty acids and lysophospholipids, which maintain the homeostasis

445

between phospholipids and free fatty acids. The activity of PLA1 was determined to be

446

almost stable in different treatments, and the activity of PLA2 was only decreased

447

significantly in cells exposed to a high-dose HBCD (Figure 5). These results implied that

448

HBCD seems not to have the potential to disturb the mass balance between phospholipids

ACS Paragon Plus Environment

Page 19 of 27

Environmental Science & Technology

449

and free fatty acids at low and middle exposure doses. Therefore, the elevated

450

phospholipids levels in cells should mainly result from the inversion of increased free fatty

451

acids due to the significant inhibition of HBCD on the β-oxidation of fatty acids. A

452

previous in vitro study found that H2O2-induced neuronal cell injury led to the remodeling

453

of phospholipids mediated by PLA2, and the arachidonic acid (AA) level increased prior to

454

the increase in phospholipids.43,

455

phospholipids containing AA were also observed in cells exposed to HBCD (Figure 5). The

456

up-regulation of AA could further inhibit the production of ATP through uncoupling the

457

oxidative phosphorylation in mitochondria.45 In addition, AA is a key inflammatory

458

intermediate. The elavated AA could provide an evidence for the liver weight increase

459

indcued by HBCD exposure, which was observed in in vivo study using Wistar rats.13

460

Proposed Mechanism for HBCD Toxicity. Our data suggest that the basic cytotoxic

461

mechanism of HBCD could be related to an excessive suppression of energy metabolism

462

(Figure 6). First, HBCD adsorbed on the cell membrane directly inhibits the activities of

463

membrane transport protein P-type ATPases, mainly the Na+/K+-ATPase and Ca2+-ATPase.

464

As a result, the cell uptake of amino acids and glucose is suppressed, and the intracellular

465

concentrations of Na+ and Ca2+ would be enhanced. The suppression of amino acid

466

transport across the membrane led to the down-regulation of intracellular amino acids and

467

further inhibits protein biosynthesis. The biosynthesis inhibition of GSH and

468

redox-sensitive protein, resulting from the large down-regulation of intracellular cysteine,

469

will restrict the ability of cells to eliminate the redundant ROS. When HBCD enters into

470

cells, it not only suppresses glycolysis by inhibiting the activity of enzyme PFK, but also

471

decreases the β-oxidation of LC-FA mainly by down-regulating the gene expression

472

relevant to enzyme LCACS.9 The down-regulation of both glycolysis and fatty acid

473

β-oxidation result in a large decrease of ATP production, which further inhibits the

44

In this study, the proportional increases of AA and

ACS Paragon Plus Environment

Environmental Science & Technology

Page 20 of 27

474

across-membrane transport of amino acids and glucose. Moreover, the significant inhibitory

475

effect of HBCD on the β-oxidation of LC-FA induces the overall up-regulation of free fatty

476

acids in cells, which further increases the total phospholipids through the remodeling of

477

phospholipids from free fatty acids, mainly mediated by the enzyme PLA2.

478

479 480

Figure 6. Proposed mechanism for HBCD-induced cytotoxicity based on the metabolomic

481

approach. LCACS: long-chain acyl-CoA synthetase; PFK: phosphofructokinase; HK:

482

hexokinase;

483

adenosine-diphosphate; ROS: reactive oxygen species; GSH: reduced glutathione.

LDH:

lactate

dehydrogenase;

ATP:

adenosine-triphosphate;

ADP:

484 485

ASSOCIATED CONTENT

486

Supporting Information

487

Detailed description on cell viability assay, instrumental analysis, the repeatability of

488

metabolic profiling analysis, ROS measurement, oxidative stress marker determination,

ACS Paragon Plus Environment

Page 21 of 27

Environmental Science & Technology

489

relevant metabolic enzyme activities and total protein; results of data processing;

490

HBCD-induced perturbation of extracellular and intracellular nutrients.

491

AUTHOR INFORMATION

492

Corresponding Authors

493

* Phone: +86-411-8437-9972, fax: +86-411-8437-9562; e-mail: [email protected].

494

* Phone/fax: +86-411-8437-9562; e-mail: [email protected].

495

Notes

496

The authors declare no competing financial interest

497

ACKNOWLEDGMENTS

498

The authors thank the National Natural Science Foundation of China (Grant No. 21337002)

499

and the National Basic Research Program (Grant No. 2015CB453100) for the financial

500

support.

501

REFERENCES

502 503

(1) Birnbaum, L. S.; Staskal, D. F. Brominated flame retardants: cause for concern? Environ. Health Perspect. 2004, 112, (1), 9−17. DOI: 10.1289/ehp.6559.

504

(2) Alaee, M.; Arias, P.; Sjodin, A.; Bergman, A. An overview of commercially used

505

brominated flame retardants, their applications, their use patterns in different

506

countries/regions and possible modes of release. Environ. Int. 2003, 29, (6), 683−689. DOI:

507

10.1016/S0160-4120(03)00121-1.

508

(3)

UNEP.

Stockholm

Convention

on

Persistent

Organic

Pollutants.

509

http://chm.pops.int/TheConvention/POPsReviewCommittee/ReportsandDecisions/tabid/33

510

09/Default.aspx. (accessed December 12, 2014).

511

(4)

Koch,

C.;

Schmidt-Kotters,

T.;

Rupp,

R.;

Sures,

B.

Review

of

512

hexabromocyclododecane (HBCD) with a focus on legislation and recent publications

513

concerning toxicokinetics and -dynamics. Environ. Pollut. 2015, 199C, 26−34. DOI:

514

10.1016/j.envpol.2015.01.011. ACS Paragon Plus Environment

Environmental Science & Technology

515

(5)

Page 22 of 27

Marvin, C. H.; Tomy, G. T.; Armitage, J. M.; Arnot, J. A.; McCarty, L.; Covaci,

516

A.;

517

environmental fate and toxicology and implications for global management. Environ. Sci.

518

Technol. 2011, 45, (20), 8613−8623. DOI: 10.1021/es201548c.

519

Palace,

(6)

V.

Hexabromocyclododecane:

current

understanding

of

chemistry,

Kakimoto, K.; Akutsu, K.; Konishi, Y.; Tanaka, Y. Time trend of

520

hexabromocyclododecane in the breast milk of Japanese women. Chemosphere 2008, 71,

521

(6), 1110−1114. DOI: 10.1016/j.chemosphere.2007.10.035.

522

(7)

Thomsen, C.; Molander, P.; Daae, H. L.; Janak, K.; Froshaug, M.; Liane, V. H.;

523

Thorud, S.; Becher, G.; Dybing, E. Occupational exposure to hexabromocyclododecane at

524

an industrial plant. Environ. Sci. Technol. 2007, 41, (15), 5210−5216. DOI:

525

10.1021/es0702622.

526

(8)

European Commission.Risk Assessment Hexabromocyclododecane, CAS no.:

527

25637-99-4,

528

R044_0805_env_hh_final_ECB.doc. Office for Official Publications of the European

529

Communities. http://ecb.jrc.ec.europa.eu/esis.

530

(9)

EINECS

no.:

247-148-4,

Final

Draft

May

2008;

Canton, R. F.; Peijnenburg, A. A.; Hoogenboom, R. L.; Piersma, A. H.; van der

531

Ven, L. T.; van den Berg, M.; Heneweer, M. Subacute effects of hexabromocyclododecane

532

(HBCD) on hepatic gene expression profiles in rats. Toxicol. Appl. Pharmacol. 2008, 231,

533

(2), 267−272. DOI: 10.1016/j.taap.2008.04.013.

534

(10) Zhang, H. J.; Kuo, Y. Y.; Gerecke, A. C.; Wang, J. Co-release of

535

hexabromocyclododecane (HBCD) and Nano- and microparticles from thermal cutting of

536

polystyrene

537

DOI: 10.1021/es302559v.

foams.

Environ.

Sci.

Technol.

2012,

46,

(20),

10990−10996.

538

(11) Ronisz, D.; Finne, E. F.; Karlsson, H.; Forlin, L. Effects of the brominated flame

539

retardants hexabromocyclododecane (HBCDD), and tetrabromobisphenol A (TBBPA), on

ACS Paragon Plus Environment

Page 23 of 27

Environmental Science & Technology

540

hepatic enzymes and other biomarkers in juvenile rainbow trout and feral eelpout. Aquat.

541

Toxicol. 2004, 69, (3), 229−245. DOI: 10.1016/j.aquatox.2004.05.007.

542

(12) Schriks, M.; Zvinavashe, E.; Furlow, J. D.; Murk, A. J. Disruption of thyroid

543

hormone-mediated Xenopus laevis tadpole tail tip regression by hexabromocyclododecane

544

(HBCD) and 2,2',3,3',4,4',5,5',6-nona brominated diphenyl ether (BDE206). Chemosphere

545

2006, 65, (10), 1904−1908. DOI: 10.1016/j.chemosphere.2006.07.077.

546

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

547

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

548

28-day

549

hexabromocyclododecane in Wistar rats. Toxicol. Sci. 2006, 94, (2), 281−292. DOI:

550

10.1093/toxsci/kfl113.

551 552

oral

(14) Data

dose

toxicity

Summary

study

and

test

enhanced

plan

for

to

detect

endocrine

hexabromocyclododecane

effects

of

(HBCD);

AR201-13459A; American Chemistry Council: Arlington, VA, 2001.

553

(15) Marteinson, S. C.; Bird, D. M.; Letcher, R. J.; Sullivan, K. M.; Ritchie, I. J.;

554

Fernie, K. J. Dietary exposure to technical hexabromocyclododecane (HBCD) alters

555

courtship, incubation and parental behaviors in American kestrels (Falco sparverius).

556

Chemosphere 2012, 89, (9), 1077−1083. DOI: 10.1016/j.chemosphere.2012.05.073.

557

(16) Reistad, T.; Fonnum, F.; Mariussen, E. Neurotoxicity of the pentabrominated

558

diphenyl ether mixture, DE-71, and hexabromocyclododecane (HBCD) in rat cerebellar

559

granule

560

10.1007/s00204-006-0099-8.

cells

in

vitro.

Arch.

Toxicol.

2006,

80,

(11),

785−796.

DOI:

561

(17) Saegusa, Y.; Fujimoto, H.; Woo, G. H.; Inoue, K.; Takahashi, M.; Mitsumori, K.;

562

Hirose, M.; Nishikawa, A.; Shibutani, M. Developmental toxicity of brominated flame

563

retardants, tetrabromobisphenol A and 1,2,5,6,9,10-hexabromocyclododecane, in rat

564

offspring after maternal exposure from mid-gestation through lactation. Reprod. Toxicol.

565

2009, 28, (4), 456−467. DOI: 10.1016/j.reprotox.2009.06.011. ACS Paragon Plus Environment

Environmental Science & Technology

Page 24 of 27

566

(18) Yanagisawa, R.; Koike, E.; Win-Shwe, T. T.; Yamamoto, M.; Takano, H.

567

Impaired lipid and glucose homeostasis in hexabromocyclododecane-exposed mice fed a

568

high-fat

569

10.1289/ehp.1307421.

diet.

Environ.

Health

Perspect.

2014,

122,

(3),

277−283.

DOI:

570

(19) Germer, S.; Piersma, A. H.; van der Ven, L.; Kamyschnikow, A.; Fery, Y.;

571

Schmitz, H. J.; Schrenk, D. Subacute effects of the brominated flame retardants

572

hexabromocyclododecane and tetrabromobisphenol A on hepatic cytochrome P450 levels

573

in rats. Toxicology 2006, 218, (2-3), 229−236. DOI: 10.1016/j.tox.2005.10.019.

574

(20) van der Ven, L. T.; van de Kuil, T.; Leonards, P. E.; Slob, W.; Lilienthal, H.;

575

Litens, S.; Herlin, M.; Hakansson, H.; Canton, R. F.; van den Berg, M.; Visser, T. J.; van

576

Loveren, H.; Vos, J. G.; Piersma, A. H. Endocrine effects of hexabromocyclododecane

577

(HBCD) in a one-generation reproduction study in Wistar rats. Toxicol. Lett. 2009, 185, (1),

578

51−62. DOI: 10.1016/j.toxlet.2008.12.003.

579

(21) Zhang, X.; Yang, F. X.; Zhang, X. L.; Xu, Y.; Liao, T.; Song, S. B.; Wang, J. W.

580

Induction of hepatic enzymes and oxidative stress in Chinese rare minnow (Gobiocypris

581

rarus) exposed to waterborne hexabromocyclododecane (HBCDD). Aquat. Toxicol. 2008,

582

86, (1), 4−11. DOI: 10.1016/j.aquatox.2007.07.002.

583

(22) Zou, W.; Chen, C.; Zhong, Y. F.; An, J.; Zhang, X. Y.; Yu, Y. X.; Yu, Z. Q.; Fu, J.

584

M. PI3K/Akt pathway mediates Nrf2/ARE activation in human L02 hepatocytes exposed to

585

low-concentration HBCDs. Environ. Sci. Technol. 2013, 47, (21), 12434−12440.

586

DOI: 10.1021/es401791s.

587

(23) An, J.; Wang, X.; Guo, P. P.; Zhong, Y. F.; Zhang, X. Y.; Yu, Z. Q.

588

Hexabromocyclododecane

589

hepatocellular carcinoma cells to cisplatin through the phosphatidylinositol 3-kinase/protein

590

kinase

591

10.1016/j.toxlet.2014.06.025.

B

pathway.

and

polychlorinated

Toxicol.

Lett.

biphenyls

2014,

229,

ACS Paragon Plus Environment

increase

(1),

resistance

265−272.

of

DOI:

Page 25 of 27

Environmental Science & Technology

592

(24) Al-Mousa, F.; Michelangeli, F. Some commonly used brominated flame retardants

593

cause Ca2+-ATPase inhibition, beta-amyloid peptide release and apoptosis in SH-SY5Y

594

neuronal cells. PloS one 2012, 7, (4), e33059. DOI: 10.1371/journal.pone.0033059.

595

(25) An, J.; Chen, C.; Wang, X.; Zhong, Y. F.; Zhang, X. Y.; Yu, Y. X.; Yu, Z. Q.

596

Oligomeric proanthocyanidins alleviate hexabromocyclododecane-induced cytotoxicity in

597

HepG2 cells through regulation on ROS formation and mitochondrial pathway. Toxicol. In

598

Vitro. 2014, 28, (2), 319−326. DOI: 10.1016/j.tiv.2013.11.009.

599

(26) Zhang, J. K.; Abou-Elwafa Abdallah, M.; Williams, T. D.; Harrad, S.; Chipman, J.

600

K.; Viant, M. R., Gene expression and metabolic responses of HepG2/C3A cells exposed to

601

flame retardants and dust extracts at concentrations relevant to indoor environmental

602

exposures. Chemosphere 2016, 144, 1996-2003. DOI: 10.1016/j.chemosphere.2015.10.014.

603

(27) Chen, S.; Kong, H.; Lu, X.; Li, Y.; Yin, P. Y.; Zeng, Z. D.; Xu, G. W.

604

Pseudotargeted metabolomics method and its application in serum biomarker discovery for

605

hepatocellular carcinoma based on ultra high-performance liquid chromatography/triple

606

quadrupole

607

DOI: 10.1021/ac4016787.

608 609

mass

spectrometry.

Anal.

Chem.

2013,

85,

(17),

8326−8333.

(28) Patti, G. J.; Yanes, O.; Siuzdak, G., Innovation: Metabolomics: the apogee of the omics trilogy. Nat. Rev. Mol. Cell Biol. 2012, 13, (4), 263-269. DOI: 10.1038/nrm3314.

610

(29) Geng, N. B.; Zhang, H. J.; Zhang, B. Q.; Wu, P.; Wang, F. D.; Yu, Z. K.; Chen, J. P.

611

Effects of short-chain chlorinated paraffins exposure on the viability and metabolism of

612

human hepatoma HepG2 cells. Environ. Sci. Technol. 2015, 49, (5), 3076−3083.

613

DOI: 10.1021/es505802x.

614

(30) Iyer, V. V.; Yang, H.; Ierapetritou, M. G.; Roth, C. M. Effects of glucose and

615

insulin on HepG2-C3A cell metabolism. Biotechnol. Bioeng. 2010, 107, (2), 347-356. DOI:

616

10.1002/bit.22799.

ACS Paragon Plus Environment

Environmental Science & Technology

Page 26 of 27

617

(31) Li, Y.; Ruan, Q.; Li, Y. L.; Ye, G. Z.; Lu, X.; Lin, X. H.; Xu, G. W., A novel

618

approach to transforming a non-targeted metabolic profiling method to a pseudo-targeted

619

method using the retention time locking gas chromatography/mass spectrometry-selected

620

ions

621

10.1016/j.chroma.2012.01.076.

monitoring.

J.

Chromatogr.

A

2012,

1255,

228-236.

DOI:

622

(32) Zhang, J. K.; Williams, T. D.; Abdallah, M. A.; Harrad, S.; Chipman, J. K.; Viant,

623

M. R. Transcriptomic and metabolomic approaches to investigate the molecular responses

624

of human cell lines exposed to the flame retardant hexabromocyclododecane (HBCD).

625

Toxicol. In Vitro 2015, 29, (8), 2116-2123. DOI: 10.1016/j.tiv.2015.08.017.

626

(33) Hu, X. Z.; Hu, D. C.; Xu, Y. Effects of tetrabrominated diphenyl ether and

627

hexabromocyclododecanes in single and complex exposure to hepatoma HepG2 cells.

628

Environ. Toxicol. Pharmacol. 2009, 27, (3), 327−337. DOI: 10.1016/j.etap.2008.11.014.

629

(34) Fotiadis, D.; Kanai, Y.; Palacin, M. The SLC3 and SLC7 families of amino acid

630

transporters.

631

10.1016/j.mam.2012.10.007.

Mol.

Aspects

Med.

2013,

34,

(2-3),

139−158.

DOI:

632

(35) Al-Mousa, F.; Michelangeli, F. The sarcoplasmic-endoplasmic reticulum

633

Ca(2+)-ATPase (SERCA) is the likely molecular target for the acute toxicity of the

634

brominated flame retardant hexabromocyclododecane (HBCD). Chem. Biol. Interact. 2014,

635

207, 1−6. DOI: 10.1016/j.cbi.2013.10.021.

636 637 638 639 640 641

(36) Reczek, C. R.; Chandel, N. S. ROS-dependent signal transduction. Curr. Opin. Cell Biol. 2015, 33, 8−13. DOI: 10.1016/j.ceb.2014.09.010. (37) Wright, E. M. Glucose transport families SLC5 and SLC50. Mol. Aspects Med. 2013, 34, (2-3), 183−196. DOI: 10.1016/j.mam.2012.11.002. (38) Mueckler, M.; Thorens, B. The SLC2 (GLUT) family of membrane transporters. Mol. Aspects Med. 2013, 34, (2-3), 121−138. DOI: 10.1016/j.mam.2012.07.001.

ACS Paragon Plus Environment

Page 27 of 27

Environmental Science & Technology

642

(39) Takanaga, H.; Chaudhuri, B.; Frommer, W. B., GLUT1 and GLUT9 as major

643

contributors to glucose influx in HepG2 cells identified by a high sensitivity intramolecular

644

FRET glucose sensor. Biochim. Biophys. Acta 2008, 1778, (4), 1091-1099. DOI:

645

10.1016/j.bbamem.2007.11.015.

646

(40) Zammit, V. A. Carnitine acyltransferases: functional significance of subcellular

647

distribution and membrane topology. Prog. Lipid Res. 1999, 38, (3), 199−224. DOI:

648

10.1016/S0163-7827(99)00002-8.

649 650 651 652

(41) Bartlett, K.; Eaton, S. Mitochondrial beta-oxidation. Eur. J. Biochem. 2004, 271, (3), 462−469. DOI: 10.1046/j.1432-1033.2003.03947.x. (42) Vance, J. E.; Vance, D. E. Phospholipid biosynthesis in mammalian cells. Biochem. Cell Biol. 2004, 82, (1), 113−128. DOI: 10.1139/o03-073.

653

(43) Peterson, B.; Stovall, K.; Monian, P.; Franklin, J. L.; Cummings, B. S. Alterations

654

in phospholipid and fatty acid lipid profiles in primary neocortical cells during

655

oxidant-induced cell injury. Chem. Biol. Interact. 2008, 174, (3), 163−176. DOI:

656

10.1016/j.cbi.2008.05.028.

657

(44) Hermansson,

M.;

Hokynar,

K.;

Somerharju,

P.

Mechanisms

of

658

glycerophospholipid homeostasis in mammalian cells. Prog. Lipid Res. 2011, 50, (3),

659

240−257. DOI: 10.1016/j.plipres.2011.02.004.

660

(45) Farooqui, A. A.; Horrocks, L. A.; Farooqui, T. Glycerophospholipids in brain:

661

their metabolism, incorporation into membranes, functions, and involvement in

662

neurological

663

10.1016/S0009-3084(00)00128-6.

disorders.

Chem.

Phys.

Lipids

2000,

664 665

ACS Paragon Plus Environment

106,

(1),

1−29.

DOI: