Subscriber access provided by UNIV AUTONOMA DE COAHUILA UADEC
Ecotoxicology and Human Environmental Health
Integration of metabolomics and lipidomics reveals metabolic mechanisms of triclosan-induced toxicity in human hepatocytes Hongna Zhang, Xiaojian Shao, Hongzhi Zhao, Xiaona Li, Juntong Wei, Chunxue Yang, and Zongwei Cai Environ. Sci. Technol., Just Accepted Manuscript • DOI: 10.1021/acs.est.8b07281 • Publication Date (Web): 09 Apr 2019 Downloaded from http://pubs.acs.org on April 9, 2019
Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.
is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.
Page 1 of 38
Environmental Science & Technology
1 2
Integration of metabolomics and lipidomics reveals
3
metabolic mechanisms of triclosan-induced toxicity in
4
human hepatocytes
5 6 7
Hongna Zhang, Xiaojian Shao, Hongzhi Zhao, Xiaona Li, Juntong Wei,
8
Chunxue Yang, Zongwei Cai*
9 10
State Key Laboratory of Environmental and Biological Analysis, Department of
11
Chemistry, Hong Kong Baptist University, Hong Kong Special Administrative Region
12 13 14 15
* Corresponding author (Z.W. Cai)
16
Phone: +852-34117070;
17
Fax: +852-34117348;
18
E-mail:
[email protected] 19
ACS Paragon Plus Environment
Environmental Science & Technology
20
TOC/Abstract art Lipidomics
Metabolomics
Amino acid metabolism Dysregulated lipids Purine metabolism Energy metabolism Toxicity
OH
Cl O
Oxidative (Decreased in cancer) stress
Cl
Cl
Triclosan
(Increased in cancer) Detoxification O
Cl
HSO3/C6H9O6
O
Cl
21
Human liver cells Cl
Triclosan Sulfate/Glucuronide
ACS Paragon Plus Environment
Page 2 of 38
Page 3 of 38
Environmental Science & Technology
22
ABSTRACT: Triclosan (TCS), an extensively used antimicrobial agent, has raised
23
considerable concern due to its hepatocarcinogenic potential. However, previous
24
hepatotoxicity studies primarily focused on the activation of specific intracellular
25
receptors, the underlying mechanisms still warrant further investigation at the
26
metabolic level. Herein, we applied metabolomics in combination with lipidomics to
27
unveil TCS-related metabolic responses in human normal and cancerous hepatocytes.
28
Endogenous and exogenous metabolites were analyzed for the identification of
29
metabolic biomarkers and biotransformation products. In L02 normal cells, TCS
30
exposure induced the up-regulation of purine metabolism and amino acid metabolism,
31
caused lipid accumulation and disturbed energy metabolism. These metabolic disorders
32
in turn enhanced the overproduction of reactive oxygen species (ROS), leading to the
33
alteration of antioxidant enzyme activities, down-regulation of endogenous
34
antioxidants and peroxidation of lipids. TCS-induced oxidative stress is thus considered
35
to be one crucial factor for hepatotoxicity. However, in HepG2 cancer cells, TCS
36
underwent fast detoxification through phase II metabolism, accompanied by the
37
enhancement of energy metabolism and elevation of antioxidant defense system, which
38
contributed to the potential effects of TCS on human hepatocellular carcinoma
39
development. These different responses of metabolism between normal and cancerous
40
hepatocytes provide novel and robust perspectives for revealing the mechanisms of
41
TCS-triggered hepatotoxicity.
42 43
Keywords: triclosan, hepatotoxicity, metabolomics, lipidomics, oxidative stress
ACS Paragon Plus Environment
Environmental Science & Technology
44
■ INTRODUCTION
45
Triclosan [2,4,4´-trichloro-2´-hydroxydiphenyl ether, TCS] has been manufactured
46
as an antimicrobial agent for over 50 years. The chemical is extensively employed in
47
consumer and personal care products such as toothpastes, soaps, detergents, textiles,
48
plastics, and paints.1 The widespread application of TCS has caused contamination and
49
potential toxicity in environmental media and biological species.2,3 Beginning in 2017,
50
TCS is banned from use in human hygiene biocidal products by the European
51
Commission,3 and in over-the-counter consumer antiseptic washing products by the
52
U.S. Food and Drug Administration.4 However, these restrictions don’t regulate all
53
TCS applications in product formulation. TCS remains one of the most commonly used
54
antimicrobials worldwide, its human health risks still warrant further evaluation.
55
Humans are exposed to TCS mainly through dermal contact with personal care
56
products and devices,5 as well as ingestion of contaminated food and drinking water.6,7
57
The frequent exposure of TCS has resulted in its wide detection in human body fluids,
58
including blood, urine, amniotic fluid, and human milk.8-11 The epidemiologic study
59
has demonstrated associations between TCS exposure and allergic sensitization.12
60
Through in vitro and/or in vivo models, TCS was found to be implicated with endocrine
61
disruption,13 impaired muscle contraction14, and carcinogenesis.15,16 In vivo studies
62
showed that TCS could cause liver cell proliferation and fibrotic responses, functioning
63
as a liver tumor promoter.15,17 The effects of liver tumor promotion has been attributed
64
to the activation of the nuclear receptor constitutive androstane receptor (CAR) through
65
xenobiotic receptor screening assay.15 Moreover, Ashrap et al.18 found that TCS could
ACS Paragon Plus Environment
Page 4 of 38
Page 5 of 38
Environmental Science & Technology
66
be transformed into more lipophilic and bioactive metabolites with much higher CAR
67
activation than the parent compound in humans. As one pathogenic mechanism
68
associated with liver fibrosis, oxidative stress is suspected as one of the underlying
69
factors for TCS-mediated liver carcinogenesis.19,20 Ma et al.21 demonstrated that TCS
70
treatment in human cancer cells significantly reduced the levels of global DNA
71
methylation, which is considered as a biomarker of cancer progression. However, the
72
links between TCS-medicated receptor activation and physiological effects of
73
hepatotoxicity remain largely unknown, and need to be further identified through the
74
evaluation of downstream metabolic processes.
75
Mass spectrometry (MS)-based metabolomics is an efficient strategy in toxic risk
76
assessment by monitoring small molecule metabolites within biological systems in
77
response to external stressors.22 As a branch of metabolomics, lipidomics is also a
78
powerful method for integral investigation of biological responses based on the lipid
79
profiling in biological samples.23 Up to now, TCS-triggered responses of hepatocellular
80
endogenous metabolites have not been well elucidated. The combined application of
81
metabolomics and lipidomics is expected to promote comprehensive and deep
82
understanding of the metabolic mechanisms of TCS-induced hepatotoxicity.
83
Biotransformation is an organism’s important defense against xenobiotics. TCS is
84
generally considered to be rapidly detoxified through glucuronidation and sulfonation
85
by phase II metabolism in humans.24 To accurately comprehend the liver toxicity of
86
TCS, it is also essential to evaluate TCS biotransformation in biological models.
87
Therefore, the objective of this study was to explore the metabolic mechanisms of
ACS Paragon Plus Environment
Environmental Science & Technology
Page 6 of 38
88
TCS-induced hepatotoxicity, especially the carcinogenic potential. The human normal
89
hepatocyte L02 cell line and the hepatocarcinoma HepG2 cell line were chosen as the
90
model systems. These two cell models have been widely applied for hepatotoxicity
91
studies of xenobiotics to predict the potential human health risks.25,26 L02 hepatocytes
92
were selected due to the normal liver-specific functions. Enhanced cell proliferation
93
was observed in HepG2 cells after TCS treatment,15 it is thus desirable for the
94
mechanism investigation of TCS effects on the human hepatocellular carcinoma (HCC)
95
development. We firstly performed TCS impact on cell viability to explore its
96
cytotoxicity. MS-based metabolomics and lipidomics approaches were applied to
97
evaluate the cellular metabolic responses to TCS exposure. To achieve a better
98
understanding of the involved toxic mechanisms, the biotransformation of TCS was
99
also investigated. The reactive oxygen species (ROS) generation and anti-oxidative
100
responses in TCS-treated cells were further determined to evaluate the oxidative
101
stresses. The obtained results were expected to provide new insights into the
102
mechanisms of TCS-induced hepatotoxicity by comparing the metabolic differences in
103
normal and cancer liver cells.
104 105 106
■ MATERIALS AND METHODS Chemicals and Materials. TCS was purchased from TCI (purity ≥ 98.0%, Tokyo 13C
107
Chemical Industry, Japan). Stable isotope-labeled standard of
108
Wellington Laboratories Inc. (Guelph, Ontario, Canada). Dulbecco's modified eagle
109
medium (DMEM) high glucose cell culture medium, fetal bovine serum (FBS),
ACS Paragon Plus Environment
12-TCS
was from
Page 7 of 38
Environmental Science & Technology
110
penicillin-streptomycin, 0.25% trypsin-EDTA, and phosphate-buffered saline (PBS)
111
were from GibcoTM (Thermo Fisher Scientific, Waltham, MA). All the other chemicals
112
used were of analytical grade or better. Solvents, including methanol (MeOH),
113
acetonitrile (ACN), isopropanol (IPA), and methyl tert-butyl ether (MTBE) were of
114
HPLC grade (Duksan, Seoul, Korea). Purified water obtained by a Millipore water
115
purification system (Synergy® UV, Bedford, MA) were used throughout the experiment.
116
Cell Culture and Cell Viability Assay. Human normal hepatic cell line L02, and
117
human hepatocarcinoma cell line HepG2 were purchased from the Shanghai Cell Bank
118
of Type Culture Collection of the Chinese Academy of Sciences. Cells were cultured
119
in DMEM supplemented with 10% FBS and 1% penicillin-streptomycin, and
120
maintained at 37℃ in a humidified atmosphere of 5% CO2. To examine cell viability,
121
L02 and HepG2 cells were seeded at a density of 2 × 104/well in 96-well plates. After
122
attachment for 24 h, the cells were exposed to TCS in serum-free DMEM containing
123
1% penicillin-streptomycin for 48 h. The exposure groups were implemented with 0,
124
0.01, 0.1, 0.5, 1, 2.5, 5, 10, 15, and 20 μM TCS dissolved in DMSO (0.05%, v/v). Cell
125
viability was assessed using the CellTiter 96® AQueous One Solution Cell Proliferation
126
Assay (Promega, Madison, WI). The solution reagent contains a tetrazolium compound
127
of MTS [3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulphenyl)-
128
2H-tetrazolium, inner salt]. At post-treatment time, 20 μL of the MTS solution reagent
129
was added to 100 μL of culture media in each well and incubated for 2 h at 37℃. The
130
absorbance was recorded at 490 nm using a Victor X3 multilabel plate reader (Perkin
131
Elmer, Waltham, MA). The results were expressed as a percentage of cell viability
ACS Paragon Plus Environment
Environmental Science & Technology
132
normalized to the control group.
133
Metabolomics Sample Preparation and Metabolite Extraction. Ten replicates
134
were performed for the control and TCS-treated groups. For each sample, 1 × 106 cells
135
were seeded in the 6-cm dish in fresh medium for 24 h. The cells were then exposed to
136
TCS in serum-free medium based on the cell viability results (0, 1, 2.5 μM for L02
137
cells; 0, 1, 10 μM for HepG2 cells). After 48 h, the culture medium was removed, the
138
cells were quickly rinsed with PBS, and harvested by trypsin digestion. Cell pellets
139
collected from each dish were rapidly quenched by 500 μL of chilled MeOH/H2O (4:1,
140
v/v) and conserved in liquid nitrogen in 2.0 mL Eppendorf tubes. The cell disruption
141
was conducted by freeze-thawing with liquid nitrogen.
142
For the metabolite extraction, the supernatants were collected after sample
143
centrifugation (15,000 g, 10 min, 4℃), and dried at 4°C using a Max-Up (NB-504CIR)
144
IR vacuum concentrator (N-Biotek Inc., GyeongGi-Do, Korea). The residuals were re-
145
dissolved in MeOH/H2O (1:1, v/v) which contained 1 μg/mL of 4-chloro-phenylalanine
146
as the internal standard (IS), the solvent amount was proportional to cell number that
147
was counted in prior. The samples were vortexed, and followed by centrifugation
148
(15,000 g, 10 min, 4℃) for supernatant collection. The quality control (QC) sample was
149
prepared by pooling 20 μL of solution from each sample in all groups. For the extraction
150
of extracellular TCS and its biotransformation products, the culture medium from each
151
dish was transferred into a 2.0 mL Eppendorf tube and centrifuged at 1,000 g for 5 min.
152
Supernatant (500 μL, containing 10 ng of 13C12-TCS) was then extracted as described
153
in the Supporting Information (SI) before the analysis of TCS biotransformation
ACS Paragon Plus Environment
Page 8 of 38
Page 9 of 38
154
Environmental Science & Technology
products.
155
Lipidomics Sample Preparation and Lipid Extraction. The sample preparation
156
method of lipidomics was the same as that of metabolomics as described above. After
157
the cell disruption, lipids were extracted based on the method of Matyash et al.27 with
158
some modifications. Briefly, 1.2 mL of MTBE was added to the disrupted cells,
159
followed by vortexing extraction for 10 min. Phase separation was then induced by
160
adding 200 μL of water. After 10 min of incubation at room temperature, the samples
161
were centrifuged (15,000 g, 10 min, 4 ℃) for organic phase collection. The lower phase
162
was re-extracted with 400 μL of the solvent mixture, which obtained by mixing
163
MTBE/MeOH/water (12:4:3, v/v/v) and collecting the upper phase. The combined
164
organic phases were dried at 4℃ using the vacuum concentrator and stored at −80℃.
165
Before instrumental analysis, the residuals were reconstituted in ACN/IPA/H2O
166
(65:30:5,
167
phosphocholine as the IS, the solvent amount was proportional to cell number that was
168
counted in prior. The supernatant was then collected after sample vortex and
169
centrifugation (15,000 g, 10 min, 4℃). The QC sample was prepared by mixing 20 μL
170
of solution from each sample in all groups.
v/v/v)
containing
5
μg/mL
of
1,2-dinonadecanoyl-sn-glycero-3-
171
Instrumental Analysis. The metabolimics samples were analyzed using a Thermo
172
Scientific ultra-high performance liquid chromatography system (UHPLC) coupled to
173
a Q Exactive™ Focus Hybrid Quadrupole-Orbitrap™ Mass Spectrometer (QE Orbitrap
174
MS). The instrumental conditions were modified according to the method of Xiang et
175
al.28 The lipidomics samples were analyzed using a Thermo Scientific UHPLC coupled
ACS Paragon Plus Environment
Environmental Science & Technology
176
to an Orbitrap Fusion™ Tribrid™ Mass Spectrometer (Orbitrap Fusion MS). The
177
instrumental conditions were optimized based on the method of Bird et al.29 Details of
178
chromatographic conditions and MS parameters are summarized in Tables S1 and S2.
179
Samples of the control and TCS-treated groups were analyzed successively. The blank
180
and QC samples were injected at the beginning and at the end of the sample run, the
181
QC sample was also analyzed every five samples.
182
Data Processing and Analysis. The Xcalibur Software v.4.1 (Thermo) was used for
183
data acquisition and pretreatment. For the metabolimics samples, chromatography peak
184
alignment and metabolite extraction were achieved using the software R. For lipid peak
185
acquisition and alignment, the software LipidSearch (Thermo) was utilized. Detailed
186
parameters of R package and LipidSearch are available in the SI. Metabolic features
187
with disturbed signals in blank samples and coefficient of variation (CV) > 30% in QC
188
samples were excluded to eliminate the potential contamination. The qualified
189
metabolic peaks were imported into software SIMCA-P (Version 13.0, Umetrics, Umea,
190
Sweden) for the partial least squares discriminant analysis (PLS-DA). The
191
differentiating metabolites between the control and exposure groups were selected
192
based on the variable importance in projection (VIP) scores in PLS-DA (VIP > 1), as
193
well as P-value (P < 0.05) and fold change (FC, FC >1.2 or < 0.8). The P values were
194
calculated in comparison of the control using Student’s t-test. Metabolites were
195
structurally identified by matching MS/MS fragments with data in the databases of
196
METLIN (http://metlin.scripps.edu/) and Human Metabolome Database (HMDB,
197
http://www.hmdb.ca/), and the software Lipidsearch. Metabolite biomarkers were
ACS Paragon Plus Environment
Page 10 of 38
Page 11 of 38
Environmental Science & Technology
198
further confirmed through the comparison of authentic standards. Pathway analysis
199
based on the identified metabolites was carried out using MetaboAnalyst according to
200
the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway database
201
(www.genome.jp/kegg/).
202
TCS Biotransformation Product Identification and Quantification. According to
203
metabolomics data operated in negative ionization mode, the biotransformation
204
products of TCS in cell extracts were identified based on the absence of peak intensity
205
in control group but with abundance in exposure groups. Another important feature is
206
the exhibition of several peak clusters in MS due to the presence of chlorine atoms.
207
Their structures were further elucidated through MS/MS analysis. Standards of
208
triclosan sulfate (TCSS) and triclosan glucuronide (TCSG) were synthesized as
209
described in the SI, the 1H-NMR of TCSS is shown in Figure S1. TCS, TCSS and TCSG
210
in the samples were quantified using a Thermo Scientific UHPLC coupled to a TSQ
211
Quantiva™ Triple Quadrupole Mass Spectrometer (UHPLC–MS/MS). Electrospray
212
ionization was operated in negative mode, and multiple reaction monitoring (MRM)
213
was used for the analysis. The hydroxylated metabolites of TCS were identified by
214
UHPLC–Orbitrap Fusion MS. Detailed information of the above instrumental methods
215
are shown in Tables S3 and S4. Quality control and quality assurance are provided in
216
the SI.
217
Relevant Metabolic Enzyme Activities and Intracellular Ammonia Level. To
218
further explore the toxicity mechanisms, TCS-treated cells were lysed, then the
219
supernatant was collected after centrifugation (15,000 g, 5 min, 4℃) for further analysis.
ACS Paragon Plus Environment
Environmental Science & Technology
220
The specific assay kits were used to measure the activities of xanthine oxidase (XOD,
221
Nanjing Jiancheng Bioengineering Institute, China) and succinate dehydrogenase
222
(SDH, Sigma-Aldrich, Milwaukee, WI), following the manufacturer instructions. The
223
results were normalized to the protein content. For the evaluation of ammonia
224
production, TCS-treated cells were rinsed with PBS and snap frozen in liquid nitrogen.
225
Intracellular ammonia was quantified with the Ammonia Assay Kit (Sigma-Aldrich
226
Chemical, Milwaukee, WI) following the recommended manufacturer’s protocol.
227
ROS Detection and Oxidative Stress Marker Determination. Cellular production
228
of ROS was measured using the molecular probe 2′,7′-dichlorodihydrofluorescein
229
diacetate (H2DCF-DA, Life technologies, Eugene, OR). After TCS exposure for 48 h,
230
L02 and HepG2 cells were incubated with 10 µM H2DCF-DA for 30 min at 37°C, then
231
washed three times with serum-free medium before the fluorescence record.
232
Fluorescence intensities were analyzed by the software ImageJ (National Institutes of
233
Health, Bethesda, MD). For the determination of other oxidative stress markers, the
234
specific assay kits (Beyotime Institute of Biotechnology, Beijing, China) were adopted
235
to determine the level of malondialdehyde (MDA), and the activities of superoxide
236
dismutase (SOD) and catalase (CAT) in cell lysate. The assays were performed
237
according to the manufacturer instructions, and the determinations were normalized to
238
the protein content. The relative contents of glutathione (GSH) and taurine were
239
calculated according to the metabolomics results.
240 241
■ RESULTS AND DISCUSSION
ACS Paragon Plus Environment
Page 12 of 38
Page 13 of 38
Environmental Science & Technology
242
Cytotoxicity Assay. After 48 h of incubation with TCS, cell viabilities were
243
determined to obtain the optimal exposure concentrations for metabolomics and
244
lipidomics studies. No difference in cell viability was observed with FBS
245
concentrations ranged 0–10% for L02 and HepG2 hepatocytes (Figure S2), thus serum
246
free medium was used to eliminate the combination of TCS with proteins in FBS. As
247
shown in Figure 1A, the toxicity of TCS was unobvious for both hepatocytes when TCS
248
concentration was less than 1 μM. Then L02 cell viability decreased significantly in a
249
dose-dependent manner. A decrease in HepG2 cell viability was observed at
250
concentrations above 5 μM. We chose 2.5 μM for L02 cells and 10 μM for HepG2 cells
251
as high-dose exposure concentrations, because the corresponding cell viabilities were
252
around 80% for both cell types. Besides, 1 μM of TCS was set as the low-dose exposure
253
concentration. It should also be noted that the median lethal dose (LD50) of TCS in
254
normal L02 cells was about 5 μM (Figure 1A). This TCS value and all our treatment
255
levels were within those detected in human urine, which have been reported to contain
256
TCS as high as 13 μM.30,31
257
Potential Metabolite Biomarkers and Perturbed Metabolic Pathways. Global
258
metabolomics was applied to profile the metabolic changes of hepatic cells in response
259
to TCS exposure. The PLS-DA models demonstrated satisfactory explanation, fitness,
260
and prediction power for L02 cells (Figure 1B-C) and HepG2 cells (Figure 1E-F).
261
Distinct differences were observed between the control group and TCS-treated groups,
262
suggesting that TCS induced conspicuous perturbation of intracellular metabolites. The
263
volcano plot screening showed that TCS affected the hepatic cells in a dose-dependent
ACS Paragon Plus Environment
Environmental Science & Technology
264
manner (Figure S3). For the same TCS concentration of 1 μM, more metabolite features
265
were significantly changed in L02 cells than in HepG2 cells (Figures S3A and S3C),
266
indicating that TCS induced more severe alterations of metabolic profiles in normal
267
hepatocytes. This result was consistent with the higher cytotoxicity of TCS in L02 cells.
268
Among the significantly changed endogenous metabolites, 42 of them in L02 cells and
269
45 in HepG2 cells were structurally identified, with 22 metabolites in common (Tables
270
S5 and S6). Those biomarkers mainly included amino acids, peptides, nucleosides,
271
nucleotides, carboxylic acids, phospholipids, and acylcarnitines. Based on the KEGG
272
pathway database, the significantly changed metabolites upon TCS intervention were
273
highly associated with the perturbation of alanine, aspartate and glutamate metabolism;
274
purine metabolism; and tricarboxylic acid (TCA) circle in both hepatocytes. In addition,
275
taurine and hypotaurine metabolism in L02 cells, and glutathione metabolism in HepG2
276
cells were also significantly impacted by TCS exposure (Figures 1D and 1G). The
277
details regarding pathway perturbation were further discussed as follows.
278
TCS Exposure Associates with the Disruption of Amino Acid Metabolism and
279
Purine Metabolism. In normal and cancerous hepatocytes, the most relevant metabolic
280
pathway affected by TCS exposure was alanine, aspartate and glutamate metabolism.
281
The contents of downstream metabolites in this pathway were significantly up-
282
regulated (Figure 2). In comparison with the high-dose and control groups, the FCs of
283
pyruvate, fumarate, and succinate in L02 cells were 1.50, 2.00, and 1.76, respectively.
284
Elevated levels of pyruvate (FC 1.44), fumarate (FC 1.92), and α-ketoglutarate (FC
285
2.46) were observed in HepG2 cells. These results revealed the possible up-regulation
ACS Paragon Plus Environment
Page 14 of 38
Page 15 of 38
Environmental Science & Technology
286
of this metabolic pathway. It should be mentioned that the above metabolites could also
287
be generated during other amino acid metabolism. Moreover, these metabolic
288
intermediates can also serve as a source of carbon skeletons for the synthesis of amino
289
acids. The significantly changed metabolite biomarkers were also involved in
290
phenylalanine metabolism, cysteine and methionine metabolism, and tyrosine
291
metabolism for both hepatocytes (Tables S5 and S6). Therefore, all 20 amino acids
292
were further selected and structurally identified using the standards (Figures S4 and
293
S5). According to their FCs, the intracellular contents of 17 amino acids decreased in
294
TCS-treated L02 cells with 10 of them significantly decreased in dose-dependent
295
manner, indicating the acceleration of amino acid metabolism. Additionally, 12 amino
296
acids showed decreasing tendency for low or/and high TCS exposure in HepG2 cells,
297
these may due to the promoted import of extracellular nutrients into the cancer cells.
298
Oxidative deamination of amino acids in the liver is the main source of endogenous
299
ammonia. The generation of ammonia occurs mainly via the α-amino nitrogen of
300
glutamate. Glutamate levels decreased in L02 cells and increased in HepG2 cells after
301
TCS treatment (Figures S4 and S5), indicating the possible disturbance of ammonia
302
formation. Alanine production through partial amino acid catabolism is an important
303
indicator for ammonia detoxification.26 The down-regulation of alanine in liver cells
304
implied the potential accumulation of ammonia. Another important evidence for the
305
overproduction of ammonia is the elevated levels of N-acetylglutamate, which is an
306
essential allosteric activator of carbamoyl phosphate synthetase I (CPS1), the rate-
307
limiting enzyme of the urea cycle (Figure S6A). When the hepatic ammonia
ACS Paragon Plus Environment
Environmental Science & Technology
308
concentration tends to increase, it would activate glutaminase to enhance
309
intramitochondrial N-acetylglutamate level; these in turn, by activating CPS1, will
310
actually convert ammonia to urea for excretion.32 In this study, TCS treatment induced
311
significant increase of N-acetylglutamate in L02 cells (FC 1.49) and HepG2 cells (FC
312
5.98. Moreover, through the quantification of ammonia levels, we directly
313
demonstrated the elevation of intracellular ammonia in TCS-treated cells (Figure S6B).
314
These results clearly showed that TCS exposure led to the up-regulation of amino acid
315
metabolism in human hepatocytes.
316
TCS intervention was also considered to be highly responsible for the perturbation
317
of purine metabolism in normal and cancerous hepatocytes. Purine metabolism is at the
318
downstream of nucleic acid degradation. In TCS-treated L02 cells, all the identified
319
downstream intermediate levels significantly increased (Figures 2A and 2B),
320
suggestive of the up-regulation of this metabolic pathway. It should be stressed that,
321
hypoxanthine and xanthine are the substrates of XOD which produces hydrogen
322
peroxide (H2O2). XOD activity in L02 cells was significantly enhanced after high-dose
323
TCS exposure (Figure 2A). Therefore, the up-regulation of purine metabolism was a
324
critical source of ROS in normal hepatocytes. However, in TCS-treated HepG2 cells,
325
the concentration of hypoxanthine decreased, while no significant change was observed
326
for xanthine (Figures 2A and 2C).
327
TCS Exposure Associates with Lipid Metabolism Perturbation. It’s noteworthy
328
that some metabolites were not listed under the metabolic pathway database. The levels
329
of several acylcarnitines, including palmitoylcarnitine (C16), tetradecanoylcarnitine
ACS Paragon Plus Environment
Page 16 of 38
Page 17 of 38
Environmental Science & Technology
330
(C14), 2-methylbutyroylcarnitine (C5) in L02 cells, and isobutyrylcarnitine (C4) in
331
HepG2 cells drastically increased after TCS exposure (Tables S5 and S6, Figure S7).
332
The accumulation of even-chain C14 and C16 was associated with incomplete fatty
333
acid β-oxidation within the mitochondrial matrix, odd-chain C5 arose from amino acid
334
catabolism, whereas C4 could be produced from both fatty acids and amino acids.33
335
Additionally, three metabolites in L02 cells, and five in HepG2 cells were involved in
336
the glycerophospholipid metabolism. Glycerophosphorylcholine is one form of choline
337
storage in the cytosol, its content significantly decreased in TCS-treated liver cells.
338
Choline, the main source of phosphatidylcholines (PCs), showed opposite change
339
tendency between L02 and HepG2 cells. As the oxidative metabolites of PCs, LPC(16:0)
340
was up-regulated in L02 cells, while LPC(14:0) and LPC(16:0) were down-regulated
341
in HepG2 cells. Same trends were observed for LPEs, the breakdown products of
342
phosphatidylethanolamines (PEs). Changes of these biomarkers demonstrated that TCS
343
induced intercellular lipid dysregulation, the malfunction between normal and
344
cancerous hepatocytes could be different.
345
These abnormalities of lipid metabolism were further investigated through
346
lipidomics analysis. PLS-DA score plots and volcano plots showed TCS treatment
347
induced significant perturbation of intracellular lipids (Figure S8). Abundance of the
348
identified lipids displayed significant up-regulation in triglyceride (TG), diacylglycerol
349
(DG), PC, PE, phosphatidylglycerol (PG), LPC, LPE, ceramide (Cer), and
350
sphingomyelin (SM) as well as remarkable down-regulation in polyphosphoinositide
351
(PI) in L02 cells (Figure S9A). The subclass lipids of DG, PC, PE, and SM significantly
ACS Paragon Plus Environment
Environmental Science & Technology
352
increased, whereas TG, PG, PI, LPC, LPE, and Cer decreased in HepG2 cells (Figure
353
S9B). The lipid biomarkers were further selected based on VIP scores, P-value and
354
FCs. The results are shown in the heat maps of Figure 3. Of note, the change trends of
355
LPC and LPE were consistent with those obtained from metabolomics. It should also
356
be pointed out that PC (16:0/16:0) was up-regulated in both cells, showing different
357
change tendency from its precursor LPC(16:0).
358
In TCS-treated L02 cells, the lipid biomarkers were generally up-regulated except
359
several individual PCs (Figure 3A). A dose-dependent increase of TGs occurred,
360
indicating that TCS induced TG accumulation in normal hepatocytes, which is a
361
hallmark feature of the pathogenesis of nonalcoholic fatty liver disease in humans.34 As
362
two main components of phospholipids, PC and PE contents were significantly
363
enhanced, indicating the disruption of cell membrane homeostasis. It should be noted
364
that the PC class showed an increase trend although several individual PCs were down-
365
regulated (Figure S9A and 3A), this could result from the up-regulation of the more
366
abundant PCs. LPC level was dramatically up-regulated, serving as an important
367
evidence of the hepatic lipotoxicity.35 PG is a precursor for the synthesis of cardiolipin,
368
an important component of the inner mitochondrial membrane. The remarkable
369
increase of PG levels suggested that TCS treatment disturbed the structure of
370
mitochondrial membrane. The SM cycle, specifically the conversion of SM to Cer, has
371
been regarded as a key signaling pathway involved in apoptosis. In this study, TCS-
372
associated up-regulation of Cer was an important signal for the activation of apoptotic
373
cascade. Moreover, high abundance of Cer could enhance the production of ROS by
ACS Paragon Plus Environment
Page 18 of 38
Page 19 of 38
Environmental Science & Technology
374
directly acting on the mitochondrial respiratory electron chain.36 Therefore, TCS
375
treatment triggered lipid deposition in human normal hepatocytes. This result was
376
consistent with those obtained in zebrafish embryos and tadpole hepatocytes, which
377
TCS induced lipid droplet accumulation through the reduction of fatty acid β-
378
oxidation.37,38 In TCS-treated HepG2 cells, the depletion of TG could arise from the
379
increased energy demand, and/or the decreased fatty acid synthesis. Moreover, defects
380
on Cer generation and SM metabolism were also observed (Figure S9B, Figure 3B). It
381
has been reported that malignant cells with low Cer levels are resistant to apoptosis,
382
and Cer content in cancer cells might be involved in the pathogenesis of tumor growth.39
383
In our study, the decrease of Cer level in HepG2 cells provided an evidence for the
384
apoptotic resistant features in response to TCS exposure, which might contribute to
385
TCS-associated human HCC development.
386
TCS Exposure Associates with Energy Metabolism Disruption. Based on the
387
metabolomics analysis, disorders of energy metabolism were observed in TCS-treated
388
hepatocytes (Figure 4). Glucose was supplied in cell culture media as the major energy
389
source, it could be converted into pyruvate and subsequently lactate by the pathway of
390
glycolysis. In L02 and HepG2 cells, pyruvate and lactate levels presented increasing
391
tendencies with the increase of TCS doses, suggestive of the up-regulation of glycolysis
392
in the cytoplasm. It is important to note that, in oncology, the majority of cancer cells
393
have been observed to predominantly produce energy by glycolysis.40,41 Another key
394
feature of the metabolic profile of cancer cells is the increased glutaminolysis for energy
395
production.42,43 Elevated levels of glutamine and glutamate were observed in TCS-
ACS Paragon Plus Environment
Environmental Science & Technology
396
treated HepG2 cells (Table S6 and Figure S5), indicating the energy production from
397
glutamine metabolism should also be enhanced. Therefore, TCS-induced energy
398
enhancement may contribute to the promotion of cancer cell proliferation. Additionally,
399
as discussed above, TCS caused a significant acceleration of amino acid metabolism
400
and disturbance of fatty acid β-oxidation in liver cells. Pantothenic acid is a component
401
of the acyl group carrier coenzyme A (CoA-SH), its intracellular contents significantly
402
increased after TCS exposure. These changes could elevate the generation of acetyl-
403
CoA, which was then oxidized in the TCA cycle.
404
Mitochondrial TCA cycle is the hub of cellular energy metabolism. Constituents of
405
the TCA cycle, including citrate, fumarate, and malate were significantly up-regulated
406
in TCS-treated hepatocytes. In addition, the level of succinate increased in L02 cells,
407
and α-ketoglutarate increased in HepG2 cells. Metabolite biomarkers observed in L02
408
cells also include flavin adenine dinucleotide (FAD) and nicotinamide adenine
409
dinucleotide (NAD+). Playing vital roles in energy metabolism, NAD+ and FAD accept
410
hydride equivalents to generate reducing equivalents, NADH and FADH2, respectively,
411
which are then furnished to the mitochondrial electron transport chain to fuel oxidative
412
phosphorylation for energy production. It has been extensively proved that TCS is a
413
mitochondrial uncoupler that disrupted membrane potential and inhibited the activity
414
of SDH (complex II), resulting in the enhancement of ROS production.44-46 In this study,
415
the decreases of SDH activity in TCS-treated cells were also observed (Figure S10). In
416
addition, lipidomics analysis demonstrated that TCS treatment disturbed the
417
mitochondrial membrane homeostasis. Succinate, the substrate of complex II,
ACS Paragon Plus Environment
Page 20 of 38
Page 21 of 38
Environmental Science & Technology
418
significantly increased in L02 cells, which could result from the complex II activity
419
changes and enhanced energy metabolism. Moreover, succinate has been identified as
420
an oncometabolite, its accumulation in mitochondria is considered to be associated with
421
tumor progression.47 Mitochondria ROS generated at the electron transport chain
422
during oxidative phosphorylation is the major source of the cellular oxidative
423
burden.48,49 Therefore, TCS-induced up-regulation of TCA cycle could lead to the
424
accumulation of mitochondrial ROS, which further enhanced oxidative stresses in
425
normal hepatocytes.
426
Biotransformation of TCS in Hepatic Cells. After exposure for 48 h, TCS was
427
detected in L02 cells, with the FC of 3.48 between high and low exposure groups (Table
428
1, Figure S11). However, TCS content was below the detection limit in HepG2 cells,
429
even that the high-dose exposure group was in the initial concentration of 10 μM. In
430
the culture medium, the mass composition of TCS was 12.7% in low and 10.2% in high
431
exposure groups for L02 cells, while only about 1% of TCS remained for HepG2 cells
432
(Figure S12). These results demonstrated the fast biotransformation of TCS in
433
cancerous hepatocytes. In cell extracts, sulfate and glucuronide conjugated metabolites
434
of TCS were identified (Table 1). The structures of TCSS and TCSG were further
435
elucidated and confirmed using the synthesized standards (Figures S13 and S14). In
436
L02 cells, FCs (High/Low) of TCSS and TCSG were 3.79 and 7.54, respectively, this
437
could be explained by the preference generation of TCSG than TCSS in humans.24 After
438
TCS exposure, intracellular and extracellular TCSS and TCSG were further monitored
439
using UHPLC-MS/MS. TCSG/TCSS peak area ratio in the extracellular culture media
ACS Paragon Plus Environment
Environmental Science & Technology
440
was much higher than that in intracellular extracts for both hepatocytes (Figure S15),
441
demonstrating that TCS was mainly transformed into TCSG and excreted by the liver
442
cells. Moreover, TCSG/TCSS peak area ratio in culture media of HepG2 cells was
443
much higher than that in L02 cells (> 15 folds), further demonstrating the rapid phase
444
II metabolism of TCS in cancerous hepatocytes. Therefore, the lower toxicity of TCS
445
to HepG2 cells was partly attributed to its fast biotransformation into conjugated
446
metabolites, and excretion for detoxification.
447
It should be pointed out that the upstream target of TCS has been demonstrated to be
448
the nuclear receptor CAR, the regulated genes of which are members of the CYP2B,
449
CYP2C, and CYP3A subfamilies.15,18 Under the catalysis of these cytochrome P450
450
enzymes, TCS underwent the rapid phase I metabolism to form hydroxylated
451
metabolites (OH-TCS).18 In this study, no OH-TCS was detected in the samples after
452
48 h of TCS exposure. However, sulfate and glucuronide conjugates of OH-TCS, OH-
453
TCSS and OH-TCSG, were identified in both extracellular and intracellular extracts
454
(Figure S16), indicating the generation of OH-TCS. Furthermore, the structure of OH-
455
TCS was elucidated in the extracts of hepatic cells treated with TCS for 3 h, directly
456
demonstrating the fast phase I metabolism of TCS (Figure S17). Formation of ROS, in
457
particular, superoxide anion radical and H2O2, can emerge following the breakdown or
458
uncoupling of the P450 catalytic cycle.49,50
459
ROS Generation and Oxidative Stress Markers. Based on above analysis, we
460
hypothesized that ROS was excessively generated within the hepatic cells, altered the
461
intracellular redox status and exacerbated metabolic dysfunction. To verify the
ACS Paragon Plus Environment
Page 22 of 38
Page 23 of 38
Environmental Science & Technology
462
overproduction of ROS, real-time generation of ROS was imaged using the fluorescent
463
probe of H2DCF-DA. The fluorescence signal intensities of TCS-treated groups were
464
significantly stronger than that of the control group in L02 and HepG2 cells (Figure
465
5A), demonstrating that TCS induced the excessive accumulation of ROS and created
466
a prooxidant environment in hepatocytes. This result was consistent with those obtained
467
in mouse models.15 MDA is the end product of lipid peroxidation and can serve as a
468
sensitive diagnostic index of oxidative injury. MDA content in TCS-treated L02 cells
469
was significantly elevated in a concentration-dependent manner, similar to the trends
470
of ROS accumulation within the cells (Figure 5B). These results provide solid evidence
471
that TCS treatment led to oxidative stresses and lipid peroxidation in normal
472
hepatocytes. However, TCS exposure did not induce MDA content changes in HepG2
473
cells (P > 0.05, Figure 5B), which indicated that serious oxidative damage did not occur
474
in cancerous hepatocytes. As for the antioxidant enzymes, SOD activities increased,
475
while CAT activities decreased in response to TCS exposure in both liver cells (Figures
476
5C and 5D), which could lead to the intracellular accumulation of H2O2. GSH and
477
taurine are endogenous antioxidants that protect against oxidative stress in liver,50,51
478
their contents significantly decreased in TCS-treated L02 cells (Figures 5E and 5F).
479
These results clearly demonstrated a dose-dependent variation of cellular oxidative
480
status in normal liver cells. In HepG2 cells, GSH content was dramatically elevated,
481
indicating the enhancement of anti-oxidative defense system in response to TCS
482
treatment. According to the metabolic pathway analysis, TCS induced significant up-
483
regulation of glutathione metabolism in HepG2 cells (Figure 1G), suggestive of a
ACS Paragon Plus Environment
Environmental Science & Technology
484
positive anti-oxidative response. Therefore, TCS-induced oxidative stress was one
485
crucial factor that caused cytotoxicity in normal hepatocytes. The elevation of anti-
486
oxidative defense system in TCS-treated cancer cells contributed to the HCC
487
development.
488
Insights into Toxicity Mechanism. This study provides direct evidence for the
489
metabolic mechanisms of toxicity induced by environmental dose-related TCS in
490
human hepatocytes. On the basis of the obtained results, the possible mechanisms of
491
TCS hepatotoxicity are deduced as follows. As a xenobiotic, TCS activates the nuclear
492
receptor CAR and is metabolized by cytochrome P450 enzymes,15,18 which triggers the
493
oxidative stress. As the adaptive responses, purine metabolism and amino acid
494
metabolism are up-regulated, lipid metabolism and energy metabolism are disturbed.
495
These metabolic disorders in turn enhance the overproduction of ROS mainly from
496
XOD activity and mitochondrial dysfunction, then exacerbate oxidative damage. The
497
potential mechanisms of TCS effects on the human HCC development include the up-
498
regulation of energy metabolism, elevation of antioxidant defense system, and fast
499
detoxification through phase II metabolism. Moreover, this study also showed that
500
metabolomics is a powerful strategy for the simultaneous identification of endogenous
501
and exogenous metabolites in biological samples. The information presented here is
502
useful to improve current understanding of TCS-induced hepatotoxicity. Future
503
research will include TCS-related metabolomics study in human body fluids for further
504
mechanism elucidation.
505
ACS Paragon Plus Environment
Page 24 of 38
Page 25 of 38
Environmental Science & Technology
506
■ ASSOCIATED CONTENT
507
Supporting Information
508
Additional information as noted in the text. This material is available free of charge via
509
the Internet at http://pubs.acs.org.
510
■ AUTHOR INFORMATION
511
Corresponding Author
512
* Phone: +852-34117070. Fax: +852-34117348. E-mail:
[email protected].
513
Notes
514
The authors declare no competing financial interest.
515
■ ACKNOWLEDGMENTS
516
We thank the National Natural Science Foundation of China (21777010 and 21806134),
517
and General Research Fund (12301518) and Collaborative Research Fund (C2014-14E)
518
of Hong Kong Research Grants Council for financial support.
519
■ REFERENCES
520 521 522 523
(1) Halden, R. U. On the need and speed of regulating triclosan and triclocarban in the United States. Environ. Sci. Technol. 2014, 48 (7), 3603–3611. (2) Montaseri, H.; Forbes, P. B. C. A review of monitoring methods for triclosan and its occurrence in aquatic environments. Trac-Trend. Anal. Chem. 2016, 85, 221–231.
524
(3) Halden, R. U.; Lindeman, A. E.; Aiello, A. E.; Andrews, D.; Arnold, W. A.; Fair,
525
P.; Fuoco, R. E.; Geer, L. A.; Johnson, P. I.; Lohmann, R.; McNeill, K.; Sacks, V. P.;
526
Schettler, T.; Weber, R.; Zoeller, R. T.; Blum, A. The Florence statement on triclosan
527
and triclocarban. Environ. Health Perspect. 2017, 125 (6), 064501.
ACS Paragon Plus Environment
Environmental Science & Technology
528
(4) FDA. Safety and effectiveness of consumer antiseptics; topical antimicrobial drug
529
products for over-the-counter human use. Final rule. Fed. Reg. 2016, 81 (172), 61106–
530
61130.
531
(5) Moss, T.; Howes, D.; Williams, F. M. Percutaneous penetration and dermal
532
metabolism of triclosan (2,4,4′-trichloro-2′-hydroxydiphenyl ether). Food Chem.
533
Toxicol. 2000, 38 (4), 361–370.
534
(6) Wu, X.; Ernst, F.; Conkle, J. L.; Gan, J. Comparative uptake and translocation of
535
pharmaceutical and personal care products (PPCPs) by common vegetables. Environ.
536
Int. 2013, 60, 15–22.
537
(7) Li, X.; Ying, G. G.; Su, H. C.; Yang, X. B.; Wang, L. Simultaneous determination
538
and assessment of 4-nonylphenol, bisphenol A and triclosan in tap water, bottled water
539
and baby bottles. Environ. Int. 2010, 36 (6), 557–562.
540
(8) Allmyr, M.; Adolfsson-Erici, M.; McLachlan, M. S.; Sandborgh-Englund, G.
541
Triclosan in plasma and milk from Swedish nursing mothers and their exposure via
542
personal care products. Sci. Total Environ. 2006, 372 (1), 87–93.
543
(9) Philippat, C.; Wolff, M. S.; Calafat, A. M.; Ye, X.; Bausell, R.; Meadows, M.;
544
Stone, J.; Slama, R.; Engel, S. M. Prenatal exposure to environmental phenols:
545
Concentrations in amniotic fluid and variability in urinary concentrations during
546
pregnancy. Environ. Health Perspect. 2013, 121 (10), 1225–1231.
547
(10) Pycke, B. F. G.; Geer, L. A.; Dalloul, M.; Abulafia, O.; Jenck, A. M.; Halden, R.
548
U. Human fetal exposure to triclosan and triclocarban in an urban population from
549
Brooklyn, New York. Environ. Sci. Technol. 2014, 48 (15), 8831–8838.
ACS Paragon Plus Environment
Page 26 of 38
Page 27 of 38
Environmental Science & Technology
550
(11) Wu, C.; Li, J.; Xia, W.; Li, Y.; Zhang, B.; Zhou, A.; Hu, J.; Li, C.; Zhao, H.;
551
Jiang, M.; Hu, C.; Liao, J.; Huo, W.; Chen, X.; Xu, B.; Lu, S.; Cai, Z.; Xu, S. The
552
association of repeated measurements of prenatal exposure to triclosan with fetal and
553
early-childhood growth. Environ. Int. 2018, 120, 54–62.
554
(12) Savage, J. H.; Matsui, E. C.; Wood, R. A.; Keet, C. A. Urinary levels of triclosan
555
and parabens are associated with aeroallergen and food sensitization. J. Allergy Clin.
556
Immun. 2012, 130 (2), 453–460.
557
(13) Ahn, K. C.; Zhao, B.; Chen, J.; Cherednichenko, G.; Sanmarti, E.; Denison, M.
558
S.; Lasley, B.; Pessah, I. N.; Kultz, D.; Chang, D. P. Y.; Gee, S. J.; Hammock, B. D. In
559
vitro biologic activities of the antimicrobials triclocarban, its analogs, and triclosan in
560
bioassay screens: Receptor-based bioassay screens. Environ. Health Perspect. 2008,
561
116 (9), 1203–1210.
562
(14) Cherednichenko, G.; Zhang, R.; Bannister, R. A.; Timofeyev, V.; Li, N.; Fritsch,
563
E. B.; Feng, W.; Barrientos, G. C.; Schebb, N. H.; Hammock, B. D.; Beam, K. G.;
564
Chiamvimonvat, N.; Pessah, I. N. Triclosan impairs excitation–contraction coupling
565
and Ca2+ dynamics in striated muscle. Proc. Natl. Acad. Sci. U.S.A. 2012, 109 (35),
566
14158–14163.
567
(15) Yueh, M.-F.; Taniguchi, K.; Chen, S.; Evans, R. M.; Hammock, B. D.; Karin, M.;
568
Tukey, R. H. The commonly used antimicrobial additive triclosan is a liver tumor
569
promoter. Proc. Natl. Acad. Sci. U.S.A. 2014, 111 (48), 17200–17205.
570
(16) Yang, H.; Wang, W.; Romano, K. A.; Gu, M.; Sanidad, K. Z.; Kim, D.; Yang, J.;
571
Schmidt, B.; Panigrahy, D.; Pei, R.; Martin, D. A.; Ozay, E. I.; Wang, Y.; Song, M.;
ACS Paragon Plus Environment
Environmental Science & Technology
572
Bolling, B. W.; Xiao, H.; Minter, L. M.; Yang, G.-Y.; Liu, Z.; Rey, F. E.; Zhang, G. A
573
common antimicrobial additive increases colonic inflammation and colitis-associated
574
colon tumorigenesis in mice. Sci. Transl. Med. 2018, 10 (443), eaan4116.
575
(17) Rodricks, J. V.; Swenberg, J. A.; Borzelleca, J. F.; Maronpot, R. R.; Shipp, A. M.
576
Triclosan: A critical review of the experimental data and development of margins of
577
safety for consumer products. Crit. Rev. Toxicol. 2010, 40 (5), 422–484.
578
(18) Ashrap, P.; Zheng, G.; Wan, Y.; Li, T.; Hu, W.; Li, W.; Zhang, H.; Zhang, Z.;
579
Hu, J. Discovery of a widespread metabolic pathway within and among phenolic
580
xenobiotics. Proc. Natl. Acad. Sci. U.S.A. 2017, 114 (23), 6062–6067.
581 582 583 584
(19) Yueh, M. F.; Tukey, R. H., Triclosan: A widespread environmental toxicant with many biological effects. Annu. Rev. Pharmacol.Toxicol. 2016, 56, 251–272. (20) Parola, M.; Robino, G. Oxidative stress-related molecules and liver fibrosis. J. Hepatol. 2001, 35 (2), 297–306.
585
(21) Ma, H. M.; Zheng, L. J.; Li, Y. H.; Pan, S. X.; Hu, J. J.; Yu, Z. Q.; Zhang, G.;
586
Sheng, G. Y.; Fu, J. M. Triclosan reduces the levels of global DNA methylation in
587
HepG2 cells. Chemosphere 2013, 90 (3), 1023–1029.
588 589 590 591
(22) Nicholson, J. K.; Lindon, J. C. Systems biology: Metabonomics. Nature 2008, 455 (7216), 1054–1056. (23) Blanksby, S. J.; Mitchell, T. W. Advances in mass spectrometry for lipidomics. Ann. Rev. Anal. Chem. 2010, 3 (1), 433–465.
592
(24) Arbuckle, T. E.; Marro, L.; Davis, K.; Fisher, M.; Ayotte, P.; Bélanger, P.; Dumas,
593
P.; Leblanc, A.; Bérubé, R.; Gaudreau, É. Exposure to free and conjugated forms of
ACS Paragon Plus Environment
Page 28 of 38
Page 29 of 38
Environmental Science & Technology
594
bisphenol A and triclosan among pregnant women in the MIREC cohort. Environ.
595
Health Perspect. 2015, 123 (4), 277–284.
596
(25) Zou, W.; Chen, C.; Zhong, Y.; An, J.; Zhang, X.; Yu, Y.; Yu, Z.; Fu, J. PI3K/Akt
597
pathway mediates Nrf2/ARE activation in human L02 hepatocytes exposed to low-
598
concentration HBCDs. Environ. Sci. Technol. 2013, 47 (21), 12434–12440.
599
(26) Geng, N.; Zhang, H.; Zhang, B.; Wu, P.; Wang, F.; Yu, Z.; Chen, J. Effects of
600
short-chain chlorinated paraffins exposure on the viability and metabolism of human
601
hepatoma HepG2 cells. Environ. Sci. Technol. 2015, 49 (5), 3076–3083.
602
(27) Matyash, V.; Liebisch, G.; Kurzchalia, T. V.; Shevchenko, A.; Schwudke, D.
603
Lipid extraction by methyl-tert-butyl ether for high-throughput lipidomics. J. Lipid Res.
604
2008, 49 (5), 1137–1146.
605
(28) Xiang, L.; Zhang, H.; Wei, J.; Tian, X. Y.; Luan, H.; Li, S.; Zhao, H.; Cao, G.;
606
Chung, A. C. K.; Yang, C.; Huang, Y.; Cai, Z. Metabolomics studies on db/db diabetic
607
mice in skeletal muscle reveal effective clearance of overloaded intermediates by
608
exercise. Anal.Chim. Acta 2018, 1037, 130–139.
609
(29) Bird, S. S.; Marur, V. R.; Sniatynski, M. J.; Greenberg, H. K.; Kristal, B. S.
610
Lipidomics profiling by high-resolution LC−MS and high-energy collisional
611
dissociation fragmentation: Focus on characterization of mitochondrial cardiolipins and
612
monolysocardiolipins. Anal. Chem. 2011, 83 (3), 940–949.
613
(30) Calafat, A. M.; Ye, X.; Wong, L. Y.; Reidy, J. A.; Needham, L. L. Urinary
614
concentrations of triclosan in the US population: 2003−2004. Environ. Health Perspect.
615
2008, 116 (3), 303–307.
ACS Paragon Plus Environment
Environmental Science & Technology
616
(31) Stacy, S. L.; Eliot, M.; Etzel, T.; Papandonatos, G.; Calafat, A. M.; Chen, A. M.;
617
Hauser, R.; Lanphear, B. P.; Sathyanarayana, S.; Ye, X. Y.; Yolton, K.; Braun, J. M.
618
Patterns, variability, and predictors of urinary triclosan concentrations during
619
pregnancy and childhood. Environ. Sci. Technol. 2017, 51 (11), 6404–6413.
620 621
(32) Brosnan, J. T. Glutamate, at the interface between amino acid and carbohydrate metabolism. J. Nutr. 2000, 130 (4), 988S–990S.
622
(33) Koves, T. R.; Ussher, J. R.; Noland, R. C.; Slentz, D.; Mosedale, M.; Ilkayeva,
623
O.; Bain, J.; Stevens, R.; Dyck, J. R. B.; Newgard, C. B.; Lopaschuk, G. D.; Muoio, D.
624
M. Mitochondrial overload and incomplete fatty acid oxidation contribute to skeletal
625
muscle insulin resistance. Cell Metab. 2008, 7 (1), 45–56.
626
(34) Donnelly, K. L.; Smith, C. I.; Schwarzenberg, S. J.; Jessurun, J.; Boldt, M. D.;
627
Parks, E. J. Sources of fatty acids stored in liver and secreted via lipoproteins in patients
628
with nonalcoholic fatty liver disease. J. Clin. Invest. 2005, 115 (5), 1343–1351.
629
(35) Han, M. S.; Park, S. Y.; Shinzawa, K.; Kim, S.; Chung, K. W.; Lee, J. H.; Kwon,
630
C. H.; Lee, K. W.; Lee, J. H.; Park, C. K. Lysophosphatidylcholine as a death effector
631
in the lipoapoptosis of hepatocytes. J. Lipid Res. 2008, 49 (1), 84–97.
632
(36) Andrieu-Abadie, N.; Gouazé, V.; Salvayre, R.; Levade, T. Ceramide in apoptosis
633
signaling: Relationship with oxidative stress. Free Radical Bio. Med. 2001, 31 (6), 717–
634
728.
635 636 637
(37) Ho, J. C. H.; Hsiao, C. D.; Kawakami, K.; Tse, W. K. F. Triclosan (TCS) exposure impairs lipid metabolism in zebrafish embryos. Aquat. Toxicol. 2016, 173, 29–35. (38) Chai, L.; Chen, A.; Luo, P.; Zhao, H.; Wang, H. Histopathological changes and
ACS Paragon Plus Environment
Page 30 of 38
Page 31 of 38
Environmental Science & Technology
638
lipid metabolism in the liver of Bufo gargarizans tadpoles exposed to triclosan.
639
Chemosphere 2017, 182, 255–266.
640
(39) Selzner, M.; Bielawska, A.; Morse, M. A.; Rüdiger, H. A.; Sindram, D.; Hannun,
641
Y. A.; Clavien, P.-A. Induction of apoptotic cell death and prevention of tumor growth
642
by ceramide analogues in metastatic human colon cancer. Cancer Res. 2001, 61 (3),
643
1233–1240.
644
(40) Warburg, O. On the origin of cancer cells. Science 1956, 123 (3191), 309–314.
645
(41) Gatenby, R. A.; Gillies, R. J. Why do cancers have high aerobic glycolysis? Nat.
646 647 648 649 650
Rev. Cancer 2004, 4, 891–899. (42) Altman, B. J.; Stine, Z. E.; Dang, C. V. From Krebs to clinic: Glutamine metabolism to cancer therapy. Nat. Rev. Cancer 2016, 16, 619–634. (43) Daye, D.; Wellen, K. E. Metabolic reprogramming in cancer: Unraveling the role of glutamine in tumorigenesis. Semin. Cell Dev. Biol. 2012, 23 (4), 362–369.
651
(44) Teplova, V. V.; Belosludtsev, K. N.; Kruglov, A. G. Mechanism of triclosan
652
toxicity: Mitochondrial dysfunction including complex II inhibition, superoxide release
653
and uncoupling of oxidative phosphorylation. Toxicol. Lett. 2017, 275, 108–117.
654
(45) Weatherly, L. M.; Shim, J.; Hashmi, H. N.; Kennedy, R. H.; Hess, S. T.; Gosse,
655
J. A. Antimicrobial agent triclosan is a proton ionophore uncoupler of mitochondria in
656
living rat and human mast cells and in primary human keratinocytes. J. Appl.Toxicol.
657
2016, 36 (6), 777–789.
658
(46) Newton, A. P. N.; Cadena, S. M. S. C.; Rocha, M. E. M.; Carnieri, E. G. S.;
659
Martinelli de Oliveira, M. B. Effect of triclosan (TRN) on energy-linked functions of
ACS Paragon Plus Environment
Environmental Science & Technology
660 661 662 663 664 665 666
rat liver mitochondria. Toxicol. Lett. 2005, 160 (1), 49–59. (47) Gogvadze, V.; Orrenius, S.; Zhivotovsky, B. Mitochondria in cancer cells: What is so special about them? Trends Cell Biol. 2008, 18 (4), 165–173. (48) Balaban, R. S.; Nemoto, S.; Finkel, T. Mitochondria, oxidants, and aging. Cell 2005, 120 (4), 483–495. (49) Morgan, M. J.; Liu, Z.-g. Crosstalk of reactive oxygen species and NF-κB signaling. Cell Res. 2010, 21, 103–115.
667
(50) Valko, M.; Rhodes, C. J.; Moncol, J.; Izakovic, M.; Mazur, M. Free radicals,
668
metals and antioxidants in oxidative stress-induced cancer. Chem-Biol. Interact. 2006,
669
160 (1), 1–40.
670 671
(51) Bouckenooghe, T.; Remacle, C.; Reusens, B. Is taurine a functional nutrient? Curr. Opin. Clin. Nutr. Metab. Care 2006, 9 (6), 728–733.
ACS Paragon Plus Environment
Page 32 of 38
Page 33 of 38
672
Environmental Science & Technology
Table 1. TCS and its phase II metabolites identified in cell extracts based on the metabolomics data Compound RT (min) TCS
16.13
TCSS
13.80
TCSG
11.99
Major measured mass (m/z)
Molecular structure
∆ppm
Mass pattern
FC (L02) High/Low a
FC (HepG2) High/Low a
286.9440 288.9412 366.9007 368.8976 462.9761 464.9728
[C12H6O235Cl3][C12H6O235Cl237Cl][C12H6SO535Cl3][C12H6SO535Cl237Cl][C18H14O835Cl3][C12H14O835Cl237Cl]-
0.35
–b –b 286.9438 288.9410 286.9440, 175.0239, 113.0228 288.9406, 175.0239, 113.0229
3.48 3.48 3.79 3.79 7.54 7.00
–c –c 11.4 11.0 7.14 7.17
0.01 0.22
673
a Peak
674
b
No high abundant product ion was observed within the detection range (> m/z 50) of our instrument, TCS was confirmed by the RT with standard.
675
c
No signal was observed.
intensity ratio between high/low exposure groups, the theoretical value was 2.5 for L02 cells and 10 for HepG2 cells.
ACS Paragon Plus Environment
Environmental Science & Technology
676
Page 34 of 38
FIGURES (B)
Control
1μM
2.5μM
QC
(C)
Control
2.5μM
1μM
QC
(D)
Alanine, aspartate and glutamate metabolism
Negative mode (–)
Positive mode (+)
(A)
OH
Cl
TCA cycle
O Cl
Cl
Taurine and hypotaurine metabolism
L02
Purine metabolism
TCS 120
L02 HepG2
80 60 40
(E)
Control
1μM
10μM
QC
Control
1μM
10μM
QC
Negative mode (–)
Positive mode (+)
(F)
(G) Alanine, aspartate and glutamate metabolism
20 0
TCA cycle
Purine metabolism
0 0.01 0.1 1 2.5 5 10 15 20
Glutathione metabolism
TCS concentration (M)
R2X = 0.559, R2Y = 0.879, Q2 = 0.832
HepG2
Cell viability (%)
R2X = 0.555, R2Y = 0.900, Q2 = 0.873
R2X = 0.461, R2Y = 0.817, Q2 = 0.769
100
R2X = 0.725, R2Y = 0.869, Q2 = 0.832
677 678
Figure 1. (A) The cell viability of L02 and HepG2 hepatocytes after TCS exposure. Data are the mean ± SD of n = 6, * P < 0.05, ** P < 0.01 (in
679
comparison to control group via Student’s t‐Test). (B-G) PLS-DA score plots of metabolomics data and pathway analysis in TCS-treated cells.
680
The PLS-DA parameters in (B), (C), (E), and (F) were based on two principal components (PCs) of the control and exposure groups.
ACS Paragon Plus Environment
Page 35 of 38
Environmental Science & Technology
681 682
Figure 2. Changes of identified metabolite biomarkers in response to TCS exposure in the pathway of alanine, aspartate and glutamate metabolism
683
and purine metabolism. Data are the mean ± SD of n = 10, * P < 0.05, ** P < 0.01 (in comparison to control group via Student’s t‐Test); for XOD
684
activity, n = 4.
ACS Paragon Plus Environment
Environmental Science & Technology
(A) L02
Control
TCS-1μM
TCS-2.5μM
(B) HepG2
Page 36 of 38
Control
TCS-1μM
TCS-10μM
685 686
Figure 3. Heatmap analysis of the identified lipid biomarkers in (A) L02 cells, and (B) HepG2 cells. The values were based on FCs of the lipid
687
peak intensity in comparison of the control.
ACS Paragon Plus Environment
Environmental Science & Technology
2.5
FC (Lactate)
ADP
Amino acid catabolism
2.5
1.0 0.5 0.0
ATP
Amino Acids
1.5
0
0
1M 2.5M
L02
Pyruvate
1M 10M
HepG2
Lactate CoA-SH
CoA-SH
2.5 2.0
0
1M 2.5M
0
L02
3.5 2.5 1.5
3.5
α-ketoglutarate
Fumarate
1.0
1M 10M
HepG2
NADH
0
1M 2.5M
NAD+
TCA Circle
0
Malate
HepG2
2.0
1.0
Isocitrate
Control TCS-Low TCS-High
3.0
Control TCS-Low TCS-High
3.0 2.0 1.5 1.0 0.5 0.0
0
1M 10M
HepG2
NAD+ + CoA-SH
2.5 2.0
0.5 1M 2.5M
0
L02
2.5 2.0
FC (FAD)
α-ketoglutarate Succinyl-CoA Fumarate Oxaloacetate
NADH + CO2
1M 10M
HepG2
FADH2
Succinyl-CoA FAD
Control TCS-Low TCS-High
2.0
GDP + Pi
1.5
3.0
1.0
2.5
0.5 0.0
Succinate
0
1M 2.5M
L02
Control TCS-Low TCS-High
2.0
GTP + CoA-SH
Control TCS-Low TCS-High
Control TCS-Low TCS-High
1.5 1.0 0.5 0.0
1.5
0
1M 2.5M
L02
1.0 0.5 0.0
1.5
0
1M 2.5M
L02
1.0 0.5 0.0
2.5
FC (NADH)
0
FC (Succinate)
0.0
2.5
+
FC (Fumarate)
1M 10M
1.5
0.5 0.0
2.0
NAD+
1.0
1M 10M
HepG2
L02
NADH
0
1M 2.5M
0
2.5
0.0
1.5
0.0
FC (NAD )
FC (Malate)
3.0
0.5
0.5
Oxaloacetate
Control TCS-Low TCS-High
Control TCS-Low TCS-High
3.0
FC (Citrate)
Acetyl-CoA
3.5
Citrate 3.5
1.0
Pantothenic Acid
NADH + CO2
Fatty Acids
1.5
L02
NAD+ +
Fatty acid β-oxidation
Control TCS-Low TCS-High
2.0
FC (α -ketoglutarate)
Glucose
Control TCS-Low TCS-High
2.0
Glycolysis
FC (Pantothenic acid)
Page 37 of 38
Q
0
1M 2.5M
L02
C
Electron transport chain
ROS ATP
688
mitochondrial inner membrane
Oxidative Phosphorylation
689
Figure 4. Disturbance of energy metabolism in liver cells after exposure to TCS. Data
690
are the mean ± SD of n = 10, * P < 0.05, ** P < 0.01 (in comparison to control group
691
via Student’s t‐Test).
ACS Paragon Plus Environment
Environmental Science & Technology
692 693
Figure 5. TCS-induced oxidative damage and anti-oxidative responses in liver cells. (A) ROS generation, (B) MDA content, (C) SOD activity,
694
(D) CAT activity, (E) GSH level, and (F) taurine level. Data are the mean ± SD of n = 6 (A), n= 4 (B, C, D) and n = 10 (E, F), * P < 0.05, ** P