Journal Pre-proof Vancomycin pretreatment attenuates acetaminophen-induced liver injury through 2hydroxybutyric acid Ningning Zheng, Yu Gu, Ying Hong, Lili Sheng, Linlin Chen, Feng Zhang, Zimiao Weng, Weidong Zhang, Zean Zhang, Wei Jia, Houkai Li PII:
S2095-1779(19)30567-2
DOI:
https://doi.org/10.1016/j.jpha.2019.11.003
Reference:
JPHA 500
To appear in:
Journal of Pharmaceutical Analysis
Received Date: 9 July 2019 Revised Date:
11 October 2019
Accepted Date: 5 November 2019
Please cite this article as: N. Zheng, Y. Gu, Y. Hong, L. Sheng, L. Chen, F. Zhang, Z. Weng, W. Zhang, Z. Zhang, W. Jia, H. Li, Vancomycin pretreatment attenuates acetaminophen-induced liver injury through 2-hydroxybutyric acid, Journal of Pharmaceutical Analysis (2019), doi: https://doi.org/10.1016/ j.jpha.2019.11.003. This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain. © 2019 Xi'an Jiaotong University. Production and hosting by Elsevier B.V. All rights reserved.
1
Vancomycin pretreatment attenuates acetaminophen-induced liver
2
injury through 2-hydroxybutyric acid
3
Ningning Zheng1, Yu Gu1, Ying Hong1, Lili Sheng1, Linlin Chen1, Feng Zhang1,
4
Zimiao Weng2, Weidong Zhang1,3, Zean Zhang4, Wei Jia5,6*, Houkai Li1*
5
1. Institute of Interdisciplinary Integrative Medicine Research, Shanghai
6
University of Traditional Chinese Medicine, Shanghai 201203, China
7
2. Dalian Medical University, Dalian 116044, China
8
3. School of Pharmacy, Second Military Medical University, Shanghai 200433,
9
China
10
4. Center for Drug Safety Evaluation and Research, Shanghai University of
11
Traditional Chinese Medicine, Shanghai 201203, China
12
5. University of Hawaii Cancer Center, Honolulu, Hawaii 96813, USA
13
6. Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational
14
Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital,
15
Shanghai 200233, China
16
*Correspondence author:
17
Wei Jia, University of Hawaii Cancer Center, Honolulu, Hawaii 96813, USA;
18
[email protected];
19
Houkai Li, Shanghai University of Traditional Chinese Medicine, Shanghai
20
201203, China;
[email protected];
21 22 1
23
Abstract
24
Liver injury caused by acetaminophen (AP) overdose is a leading public
25
health problem. Although AP-induced liver injury is well recognized as the
26
formation of N-acetyl-p-benzoquinone (NAPQI), a toxic metabolite of AP,
27
resulting in cell damage, emerging evidence indicates that AP-induced liver
28
injury
29
microbiota-involved mechanism remains largely unknown. In our study, we
30
found that vancomycin (Vac) pretreatment (100 mg/kg, twice a day for 4 days)
31
attenuated AP-induced liver injury, altered the composition of gut microbiota,
32
and changed serum metabolic profile. Moreover, we identified Vac
33
pretreatment elevated cecum and serum 2-hydroxybutyric acid (2-HB), which
34
ameliorated AP-induced cell damage and liver injury in mice by reducing AP
35
bioavailability and elevating GSH levels. Our current results reveal the novel
36
role of 2-HB in protecting AP-induced liver injury and add new evidence for gut
37
microbiota in affecting AP toxicity.
is
also
associated
with
gut
microbiota.
However,
the
gut
38 39
Key words: Liver injury; Gut microbiota; Acetaminophen; Vancomycin;
40
2-Hydroxybutyric acid
41 42
1. Introduction
43
Acetaminophen (AP) is a widely used over-the-counter medication for
44
relief of pain and fever. However, liver damage caused by AP overdose is a 2
45
leading public health problem. AP-induced hepatotoxicity accounts for 46% of
46
all acute liver failure in the United States [1] and 40-70% of all cases in Great
47
Britain and Europe [2]. About 85% of AP undergoes phase Ⅱ conjugation to
48
sulfated and glucuronidated metabolism in liver, and 10% of AP undergoes
49
phaseⅠoxidation to N-acetyl-p-benzoquinone (NAPQI), which is catalyzed by
50
CYP2E1, CYP1A2, CYP3A4 and normally conjugated with glutathione (GSH)
51
to nontoxic metabolites. Overdose of AP increases NAPQI production resulting
52
in depletion of GSH, as well as binding with cellular proteins leading to cell
53
injury [3-5].
54
Gut microbiota usually modulates the oral drug bioavailability or half-life
55
by altering the capacity of drug-metabolizing enzymes or expression of genes
56
involved in drug metabolism in host tissues [6]. Clayton et al. [7] have
57
observed that individuals with a high level of pre-dose urinary p-cresol, a
58
microbial metabolite, show low post-dose urinary ratios of sulfated AP to
59
glucuronic AP, suggesting that gut microbiota might affect the metabolism of
60
AP. Moreover, Lee et al. have evaluated the effects of gut microbiota on AP
61
metabolism in antibiotic-treated rats. They find antibiotic-treated rats have a
62
higher level of AP glutathione conjugates in blood than untreated rats,
63
suggesting the impacts of gut microbiota on AP metabolism [8]. Moreover,
64
Gong et al. [9] have demonstrated that AP-induced rhythmic variation in
65
hepatotoxicity is at least partially due to the production of gut microbial
66
metabolite,1-phenyl-1,2-propanedione, that depletes hepatic GSH levels. 3
67
Such evidence suggest that microbial metabolites also play important role in
68
affecting
69
mechanism remains largely unexplored due to the extremely complex
70
relationship between gut microbiota and host.
AP-induced
hepatotoxicity.
However,
the
microbiota-related
71
In our present study, we observed that vancomycin (Vac) pretreatment
72
attenuated AP-induced hepatotoxicity, as well as the alteration of gut
73
microbiota composition, convincing the microbial impacts on AP-induced
74
hepatotoxicity. Furthermore, untargeted and targeted metabolomics revealed
75
Vac pretreatment increased serum levels of 2-hydroxybutyric acid (2-HB),
76
which attenuated AP-induced hepatotoxicity both in vitro and in vivo.
77 78
2. Materials and Methods
79
2.1 Animals, cell lines and reagents
80
Male SD and Wistar rats (160-180 g) and C57 mice (18-22 g) were
81
purchased from Shanghai SLAC laboratory animal company and housed at
82
constant temperature (24 ± 2℃) with a 12 h light-dark cycle. BRL3A (ATCC
83
number: CRL-1442) and AML12 (ATCC number: CRL-2254) cells were
84
purchased from ATCC (Manassas, MA, USA). Vac was purchased from Lilly
85
Company (USA). AP, 2-HB, phenacetin were purchased from Sigma-Aldrich
86
(St. Louis, MO, USA). Sodium (±)-2-hydroxybutyrate-2,3,3-d3 (d3-2-HB) was
87
purchased from C/D/N Isotopes Inc (Canada). ALT, AST kits were purchased
88
from Nanjing Jiancheng Bioengineering Institute (China). GSH & GSSG and 4
89
CCK-8 kits were purchased from Beyotime Biotechnology (China). Gut
90
bacterial DNA extraction kit was purchased from QIAGEN company (Germany).
91
TRIzol reagent, reverse transcript enzyme and SYBR Green master mix were
92
purchased from Invitrogen (Carlsbad, CA). CYP2E1 (Cat: ab28146,
93
Lot:GR324351-11), CYP3A4 (Cat: ab3527, Lot: GR3210692-3), NQO-1 (Cat:
94
ab2346, Lot: GR3550-44) primary antibodies were purchased from Abcam
95
(USA). Collagenase type I (Cat: LS004196 ) was purchased from Washington
96
Biochemical Corporation. Percoll (Cat: P1644) was purchased from
97
Sigma-Aldrich.
98 99
2.2 Animal experiments and sample collection
100
The animal experiments were conducted under the Guidelines for Animal
101
Experiment of Shanghai University of Traditional Chinese Medicine and the
102
protocol was approved by the institutional Animal Ethics Committee. First, to
103
distinguish the gut microbial impacts on AP-induced acute liver injury, male SD
104
rats were divided into three groups as following: Con group (n=6), AP (i.g)
105
group (n=14) and AP (i.p) group (n=8). After overnight fasting, rats were
106
administered with single-dose of AP either orally (2.0 g/kg) or intraperitoneal
107
injection (1.0 g/kg) respectively, and sacrificed 24 h after AP administration. In
108
the second animal experiment, SD rats were divided into four groups as
109
following: Con group (n=6), Vac group (n=6), AP group (n=29) and Vac_AP
110
group (n=21). Vac and Vac_AP groups were orally given Vac (200 mg/kg/d) [10] 5
111
for continued 4 days prior to the administration of a single dose of AP (2.0
112
g/kg). After 24 h, all rats were anesthetized with 2% pentobarbital sodium and
113
samples were collected and stored at −80 °C for subsequent analysis.
114
Meanwhile, a paralleled experiment was conducted on Wistar rats by oral
115
gavage of 1.0 g/kg of AP based on our previous dosage experiment (n=20).
116
For the pharmacokinetic study of AP, serum samples in AP and Vac_AP
117
groups were collected at different timepoints (n=4-8).
118 119
2.3 Biochemical and liver histological analysis
120
Serum ALT, AST, liver and cellular GSSG/GSH were analyzed according
121
to the instructions. Liver tissues were maintained in 10% neutral formalin, then
122
embedded in paraffin and liver tissues were sliced at 5 μm for hematoxylin
123
and eosin (H&E) staining. The degree of liver injury was scored according to
124
reference [11].
125 126
2.4 Untargeted metabolomics analysis on serum sample with GC-MS
127
Serum samples stored at −80 °C were thawed and vortexed for 5 s at
128
room temperature. A total of 30 µL of serum sample was added to tube
129
containing 10 µL of internal standard (0.1 mg/mL dulcitol), and vortexed for 5 s.
130
Then, 120 µL of ice-cold methanol/chloroform (3:1) was added, the resulting
131
mixture was vortexed for 30 s and placed at −20 °C for 20 min before
132
centrifugation at 16 000 g and 4 °C for 15 min. Quality control (QC) sample 6
133
was prepared in parallel by pooling a small part of serum from each sample
134
and analyzed with the same procedure. The supernatant was utilized for
135
untargeted metabolomics analysis with GC-MS. The instrumental analysis and
136
data preprocessing was described as our previous method [12].
137 138
2.5 Quantitation of AP and 2-HB with targeted metabolomics in serum
139
and cecum samples
140
For serum samples, 5 µL of d3-2-HB (20 µg/mL, internal standard for
141
2-HB) and 5 µLof phenacetin (2 µg/mL, internal standard for AP) were added
142
to 25 uLof serum sample, and 165 µL of methanol was added into each
143
sample to precipitate protein. After votex for 1 min, the samples were
144
centrifuged (14000 rpm, 4 °C, 10 min) and the supernatant was transferred
145
into tubes for further analysis. For 2-HB calibration curve, 25 µL of 2-HB
146
standard solutions (125 ng/mL, 250 ng/mL, 500 ng/mL, 1 µg/mL, 2.5 µg/mL, 5
147
µg/mL,10 µg/mL, 50 µg/mL, 100 µg/mL, 200 µg/mL) were mixed with 5 µL of
148
d3-2-HB (20 µg/mL) and 170 µL of methanol and prepared with the same
149
method. For AP calibration curve, 2.5 µL of AP standard solutions (10 ng/mL,
150
20 ng/mL, 100 ng/mL, 500 ng/mL, 1 µg/mL, 2 µg/mL, 5 µg/mL,10 µg/mL, 20
151
µg/mL, 50 µg/mL, 100 µg/mL, 200 µg/mL, 500 µg/mL) were mixed with 22.5 µL
152
of blank serum, 5 µL of phenacetin (2 µg/mL) and 170 µL of methanol and
153
prepared with the same method.
154
A total of 50 mg cecum content of each mouse was weighed and added 7
155
with 400 µL of water. The samples were grinded thoroughly and centrifuged
156
(14000 rpm, 4 °C, 10 min) . 100 µL of supernatant was transferred into the tube
157
and 295 µL of methanol and 5 µL of d3-2-HB (20 µg/mL) were added, after
158
votex for 1 min, the samples were centrifuged (14000 rpm, 4 °C, 10 min).The
159
supernatant was transferred into a new tube and concentrated to dry under
160
nitrogen. The dried sample was dissolved in 100ul of methanol, after votex for
161
2 min and centrifuged at 14000 rpm for 2 min, the supernatant was transferred
162
into tubes for further analysis. For calibration curve of 2-HB, 100 µL of
163
standard solution (31.25, 62.5, 125, 250, 500, 1000 ng/mL) was mixed with 5
164
µL of d3-2-HB (20 µg/mL) and 295 µL methanol and prepared with the same
165
method.
166
The quantitation of AP and 2-HB was analyzed with LC20AD (Shimadzu)
167
coupled with AB Sciex Triple Quadrupole 4500. AP and 2-HB was separated
168
through a C18 column (5 µm, 2 × 25 mm). The mobile phase was water with
169
0.1% formic acid (mobile phase A) and acetonitrile (mobile phase B). At a flow
170
rate of 0.25 mL/min, a gradient was used as follows: 0-1.0 min:3% B, 1.0-1.1
171
min: 3% B to 50% B , 1.1-2 min: 50% B , 2.0-2.1 min: 50% B to 3% B, 2.1-2.9
172
min: 3% B. The column was maintained at 40 ℃, and the samples were kept
173
at 4 °C in an auto sampler during the entire analysis. The response of AP and
174
phenacetin were measured in positive mode ([M+H]+). The Q1 Mass (Da) / Q3
175
Mass (Da) for AP and phenacetin was 152.0 / 110.1 and 180.1 / 110.0,
176
respectively. The response of 2-HB and d3-2-HB was measured in negative 8
177
mode ([M-H]-). The Q1 Mass (Da) / Q3 Mass (Da) for 2-HB and d3-2-HB was
178
103.0 / 57.1 and 106.0 / 59.1, respectively. Data were acquired and processed
179
with Analyst 1.5.2 system software (AB SCIEX™, Foster City, CA, USA).
180 181
2.6 Gut microbiota analysis with 16S rRNA gene sequencing
182
Gut bacterial DNA extraction and 16S rRNA gene sequencing were
183
conducted according to the method previously published [12]. Briefly, bacterial
184
DNA of cecum contents was extracted with bacteria DNA stool mini kit
185
following the manufacturer’s instructions. The extracted DNA samples were
186
used as template for amplification of the V3 region of 16S rRNA gene. The
187
PCR amplification, pyrosequencing of PCR amplicons and quality control were
188
performed on Ion PGM™ System according to reference [13]. Raw fastq data
189
was quality-filtered for further analysis. Operational taxonomic units (OTUs)
190
were delineated at 97% similarity level with Mothur software. Raw fastq files
191
were subsequently deposited to the Sequence Read Archive database under
192
the accession number PRJNA546452.
193 194
2.7 Preparation of primary hepatocytes from mice
195
Mouse primary hepatocytes were isolated by two-step collagenase
196
perfusion protocol [14]. Briefly, mice were anaesthetized with 1% pentobarbital
197
sodium solution by intraperitoneal injection. Then, the mouse liver was
198
perfused with perfusion medium I via portal vein for 10 min. Next, the liver was 9
199
perfused with medium containing collagenase type I for 10 min. After adequate
200
digestion, the liver was dissected in sterile dish and passed through a 100 µm
201
filter followed by centrifugation for 2 min at 600 rpm. The supernatant was
202
discarded and hepatocytes pellet was resuspended in DMEM medium. The
203
above washing step was repeated for three times. Then the hepatocytes were
204
purified with percoll regent and then seeded on plates with 10% FBS DMEM
205
for further experiments.
206 207
2.8 Cell viability test under the treatment of 2-HB
208
1 x 104 cells per well were seeded on a 96 well plate. After 24 h, cells were
209
incubated with or without 2-HB (5~40 mM) for 24 h or 48 h. Cell viability was
210
measured with CCK-8 kit, with the addition of CCK-8 regent (10%) into the
211
medium following the incubation at 37 ℃ for 1 h. The absorbance was
212
measured at 450 nm (the main wavelength) and 600 nm (the secondary
213
wavelength) and the viability was calculated as the percentage of control.
214 215
2.9 The protective effects of 2-HB on AP-induced cell injury
216
First, the IC50 of AP in BRL3A, AML12 and mouse primary hepatocytes
217
were evaluated. Different concentrations of AP (3.125, 6.25, 12.5, 25, 50 mM)
218
was incubated with cells for 24 h or 48 h. The cell viability was analyzed with
219
CCK-8 kit. And these cell lines were separately treated with AP at its IC50
220
dosage and different concentrations of 2-HB. BRL3A cells were treated with 9 10
221
mM of AP and 2-HB (5, 10,20,40 mM) for 48 h. AML12 cells were treated with
222
14 mM of AP and 2-HB (5, 10, 20, 40 mM) for 48 h. Mice primary liver cells
223
were treated with 17 mM of AP and 2-HB (5, 10,20,40 mM) for 24 h. Cell
224
viability was measured with CCK-8 kit and calculated as the percentage of
225
control group.
226 227
2.10 Functional investigation of 2-HB in AP-induced acute liver injury in
228
mice
229
To study the impact of 2-HB on AP-induced liver injury, male C57 mice
230
were divided into four groups as following: Con group (n=5), 2-HB group (n=6),
231
AP group (n=7) and 2-HB+AP group (n=7). 2-HB (250 mg/kg) were
232
intraperitoneally injected into C57 mice in 2-HB and 2-HB+AP groups for 4
233
days, and 30 min after the last injection, 400 mg/kg of AP were orally
234
administrated to mice in AP and 2-HB+AP groups. After 24 h, all mice were
235
anesthetized with 2% pentobarbital sodium and samples were collected and
236
stored at −80 °C for subsequent analysis. For the pharmacokinetic study of AP,
237
serum samples in AP and 2-HB+AP groups were collected at the time points of
238
0, 10 min, 0.5 h, 1 h, 3 h, 8 h, and 24 h. Three mice in AP or 2-HB+AP group at
239
every time point were used in this study and each serum sample was analyzed
240
twice with instrument (n=6).
241 242
2.11
β-GD activity assay in cecum content 11
243
A total of 100 mg of cecum contents from each rat of Con, Vac, AP and
244
Vac_AP was homogenized with 100 µL of lysis buffer and grinded with
245
magnetic beads for 1 min. After centrifuged at 8000 g for 10 min (4 °C), the
246
supernatant was transferred to another clean tube for further analysis. Total
247
protein concentration of each sample was assayed and the samples were
248
adjusted to the same protein concentration before β-glucuronidase (β-GD)
249
assay. The assay was performed based on articles previously published [15].
250
Briefly. P-nitrophenyl-b-D-glucuronide acid (PNPG) was used as the probe
251
substrate of β-GD. The incubation mixture with a total volume of 0.1 mL was
252
consisted of 48 µL of 0.2 M phosphate buffer (pH 6.8), 50 µL of sample and 2
253
µL of PNPG (200 mM, final concentration). PNPG hydrolysis was performed at
254
37 °C for 30 min. The signal of the hydrolytic metabolite of PNPG (PNP) was
255
detected at 405 nm every 1 min, by a Synergy H1 Hybrid Multi-Mode
256
Microplate Reader (BioTek, USA). The reaction rate was calculated based on
257
the OD405 and used for the comparison of β-GD activity between groups.
258 259
2.12 Real-time RT-PCR analysis on gene expression
260
Total RNA of liver tissue was extracted using TRIzol reagent (Invitrogen,
261
Carlsbad, CA) and quantified by UV absorption (Colibri, Titertek Berthold).
262
Following quantification, reverse transcript reaction was performed with a
263
reverse transcript enzyme (Life technologies, Invitrogen) according to the
264
manufacturer’s instructions. Quantitative amplification by PCR was carried out 12
265
using SYBR Green Master Mix regent by Biorad CFX Connect system. Cycle
266
numbers of both 18s (as an internal control) and cDNAs of interest were used
267
to calculated relative expression levels of the genes of interest.
268 269
2.13 Western blot
270
Liver protein was extracted with a commercial lysis buffer (Shanghai
271
beyotime Biotechnology Company) added with 1% of phosphatase inhibitor
272
cocktail 2 and phosphatase inhibitor cocktail 3 (Sigma) according to
273
well-established protocols. The concentration of total protein was determined
274
with Pierce™ BCA protein assay kit (Thermo Scientific). Then the samples
275
were adjusted to the same concentration and mixed with loading buffer, and
276
heated at 100℃ for 10 min. A total of 40 µg protein was loaded into each lane
277
and separated by 10% SDS-PAGE gel, and then transferred to a PVDF
278
membrane (Millipore). The membranes were blocked with 10% milk at room
279
temperature for 90 min and then washed with TBST (20 mM Tris-HCl, 137mM
280
NaCl, and 0.1% Tween20, pH7.5) for three times at 10 min interval, following
281
the incubation with primary antibodies for NQO-1 (Abcam), CYP2E1 (Abcam)
282
and CYP3A4 (Abcam) overnight at 4 ℃. The membranes were washed with
283
TBST for three times, and incubated with the HRP-conjugated secondary
284
antibodies for 2 h at room temperature. The membranes were exposed with
285
SuperSignal West Pico Chemiluminescent Substrate (Thermo Fisher Scientific,
286
USA) and the bands were quantified with Amersham Imager 600 system 13
287
(General Electric Company, USA).
288 289
2.14 Liver CYP450 enzymatic activity assay
290
S9 fractions were prepared from livers to determine the activities of
291
CYP2E1 and CYP1A2 according to previous method with some modifications
292
[16]. Briefly, 250 mg of liver tissue in each group (n=5-10) was mixed with 1 mL
293
of 100 mM potassium phosphate containing 0.15M potassium chloride
294
(pH=7.4) and grinded with magnetic beads for 1 min. After centrifuged at 9000
295
g for 20 min, the supernatant was transferred to another clean tube and tested
296
for the total protein concentration with BCA method. Each assay contained 99
297
µL of 0.1 M potassium phosphate buffer (pH 7.4), a NADPH-generating system
298
(containing 0.1 M glucose 6-phosphate, 10 mM NADP+, and 10 IU/mL
299
glucose-6-phosphate dehydrogenase, 40 mM MgCl2), 20 µl of S9 fraction and
300
1 µL of probe substrate. Following a 3 min preincubation period in the absence
301
of probe substrate, the reaction mixture was incubated for 30 min at 37 °C, and
302
the reaction was stopped by adding 100 µl acetonitrile. LC20AD (Shimadzu)
303
coupled with AB Sciex Triple Quadrupole 4500 system was utilized to measure
304
the
305
(acetaminophen).
metabolites
of
CYP2E1
(6-hydroxy-chlorzoxazone)
and
CYP1A2
306 307 308
2.15 PICRUSt analysis with 16S rRNA gene sequencing data Phylogenetic
Investigation
of
Communities 14
by
Reconstruction
of
309
Unobserved States (PICRUSt) analysis was performed with 16S rRNA gene
310
sequencing data[17]. This method predicts the gene family abundance from
311
the phylogenetic information with an estimated accuracy of 0.8. The OTU table
312
was used as the input for metagenome imputation and was first rarefied to an
313
even sequencing depth prior to the PICRUSt analysis. Next, the resulting OTU
314
table was normalized by 16S rRNA gene copy number. The gene content was
315
predicted for each individual. Then the predicted functional composition
316
profiles were collapsed into level 3 of KEGG (Kyoto Encyclopedia of Genes
317
and Genomes) database pathways.
318 319
2.16 Statistical analysis
320
Data are shown as means ± SEM unless otherwise noted. Statistical
321
significance was determined with the unpaired two-tailed Student’s t test. The
322
statistical significance of hepatic steatosis scores was evaluated with
323
non-parametric Kruskal-Wallis test followed by the MannWhitney U test. p<
324
0.05 was considered statistically significant.
325 326
3. Results
327
3.1 Vac pretreatment attenuates AP-induced acute liver injury in rats
328
First of all, to confirm the impact of gut microbiota on AP-induced liver
329
injury, male SD rats were administered with single-dose of AP either orally (2.0
330
g/kg) or through intraperitoneal injection (1.0 g/kg). We found that serum ALT 15
331
and AST levels were significantly elevated in both oral administration and
332
intraperitoneal injection groups compared to control group. However, dramatic
333
inter-individual variation in serum ALT and AST was only present in orally
334
administrated rats (Figure 1A), but not those under intraperitoneal injection
335
(Figure 1B). Given the fact that the absorption process of intraperitoneal
336
injection is absent of impact from gut microbiota, high inter-individual variation
337
of AP-induced liver injury in orally administrated rats was probably due to gut
338
microbial influence on AP pharmacokinetics. Then, the microbial impact on
339
AP-induced liver injury was investigated in male SD rats pretreated with or
340
without Vac for 4 consecutive days prior to a single-dose AP (2.0 g/kg)
341
administration. After 24 h of AP treatment, we found that pretreatment of Vac
342
significantly attenuated serum ALT and AST elevation compared to that of AP
343
alone (Figure 1C), which was convinced by liver histological analysis (Figure
344
1D). Meanwhile, a paralleled experiment was conducted in Wistar rats, which
345
showed consistent results with that observed in SD rats (Supplementary
346
Figure 1A and B). These results indicated that Vac pretreatment could
347
attenuate AP-induced acute liver injury in rats.
348 349
3.2 Vac pretreatment decreases the bioavailability of AP and ratio of
350
GSSG/GSH
351 352
To investigate whether the attenuation of AP-induced liver injury by Vac pretreatment
was
associated
with 16
changes
of
AP
bioavailability,
353
pharmacokinetics of AP were compared between AP and Vac_AP groups
354
either in SD or Wistar rats. The results showed that serum concentrations of
355
AP at the 2nd ,4th, 8th, 24th h and AUC0-t were significantly lower in Vac_AP
356
group than AP group in either SD or Wistar rats (Figure 2A, Supplementary
357
Figure 1C). Since the serum concentration of AP is partially dependent on the
358
extent of AP reabsorption in intestinal tract through deglucuronidation of
359
conjugated AP by bacteria-derived β-GD [18, 19], we therefore expected
360
whether Vac pretreatment altered the activity of β-GD or not. The activity of
361
β-GD was measured in cecum contents. The results showed that the activity of
362
β-GD was significantly inhibited by Vac pretreatment (Figure 2B), suggesting
363
that the reduced serum concentration of AP in Vac-pretreated rats was
364
probably associated with the inhibition of bacterial β-GD activity.
365
Since the AP-induced liver injury is partly due to depletion of GSH [5], the
366
ratio of GSSG/GSH was compared between AP and Vac_AP groups. We
367
found that the ratio of GSSG/GSH was dramatically reduced in Vac_AP rats
368
compared to AP rats (Figure 2C), suggesting that Vac pretreatment increased
369
levels of GSH, rather than its oxidative product GSSG. In line with the
370
increased levels of GSH, Vac pretreatment resulted in up-regulated mRNA
371
expression of Nqo-1 and Gclc gene, and down-regulated Tnf-α and Il-1β
372
(Figure 2D).
373
In addition, the protein expression of CYP2E1 and CYP3A4, and enzyme
374
activity of CYP2E1 and CYP1A2 was measured in liver tissue, which regulate 17
375
the formation of toxic NAPQI from AP [20, 21]. There were no significant
376
differences in CYP3A4 protein expression and CYP1A2 activity between the
377
two groups (Figure 2E and F, Supplementary Figure 2), except for the
378
up-regulated activity of CYP2E1 in Vac_AP group (Figure 2G and H,
379
Supplementary Figure 2). Collectively, these results suggested that the
380
protection of Vac pretreatment against AP-induced liver injury may not due to
381
the reduction of NAPQI product, but associated with decreased bioavailability
382
of AP and increased ratio of GSH/GSSG.
383 384
3.3 Vac pretreatment elevates 2-HB in both cecum and serum
385
In addition to the pharmacokinetics change, previous study indicates that
386
endogenous metabolite derived from gut microbiota may also contribute to
387
AP-induced hepatotoxicity[9] . We therefore investigated the mechanism
388
underlying the protective effect of Vac pretreatment against AP-induced liver
389
injury using GC-MS-based untargeted metabolomics on serum samples. First
390
of all, we observed that Vac treatment not only significantly altered the
391
metabolic profile at baseline, but also in AP treated-rats (Figure 3A),
392
suggesting that Vac treatment could on one hand alter the metabolism of host,
393
on the other hand, reverse the endogenous metabolism in AP-treated rats.
394
Then, differential metabolites among groups were determined with the double
395
criteria of VIP>1.0 in OPLS-DA model and p<0.05 in Student’s t test, in which a
396
total of 13 differential metabolites were identified and visualized with a 18
397
heatmap (Figure 3B). These 13 differential metabolites were further uploaded
398
to MetaboAnalyst (https://www.metaboanalyst.ca/) for pathway enrichment
399
analysis, and 9 pathways were enriched with criteria of p< 0.05 including
400
pathways involved in propanoate metabolism, amino acids metabolism (i.e.
401
phenylalanine and tyrosine, beta-alanine, asparate, alanine, cysteine,
402
malate-asparate shuttle and glucose-alanine cycle) and urea cycle (Figure
403
3C). Propanoate metabolism pathway was the most significant pathway with
404
the lowest p value, which included 2-HB, beta-alanine, 2-ketoglutaric acid and
405
glutamic acid. All these four metabolites were increased by AP treatment
406
compared to Con group, whereas 2-HB was one of the most significantly
407
altered metabolites (Supplementary Table 1). Furthermore, we quantified the
408
concentrations of 2-HB in both cecum content and serum with or without AP
409
treatment by using targeted metabolomics based on LC-MS/MS. The results
410
showed that Vac pretreatment significantly elevated 2-HB levels in cecum
411
content with or without AP treatment (Figure 3D), suggesting gut-derived 2-HB
412
was stimulated in Vac. Consistent with this finding, Vac pretreatment also
413
increased levels of 2-HB in serum before AP treatment (Figure 3E). However,
414
serum 2-HB level was lower in Vac_AP than AP group 24 h after AP treatment,
415
which was in line with the untargeted metabolomics result (Figure 3E).
416
To test whether the increased concentration of 2-HB by Vac treatment was
417
due to altered gut microbiome, we compared the gut microbiota profiles by
418
using 16S rRNA sequencing between AP and Vac_AP groups. The PCoA 19
419
showed that AP and Vac_AP group were clearly separated (Figure 3F). At the
420
phylum level, the relative abundance of Firmicutes and Bacteroides were
421
dramatically decreased, while the abundance of Proteobacteria and
422
Fusobacteria were greatly enriched in Vac_AP group (Figure 3G). Then, a
423
total of 50 genera with relatively higher abundance was chosen and compared
424
between AP and Vac_AP groups, in which the relative abundance of 9
425
gram-negative and 16 gram-positive bacteria genera were either significantly
426
increased or decreased by Vac pretreatment (Supplementary Figure 3).
427
Functional annotation of gut bacteria showed that propanoate metabolism
428
pathway was significantly enriched in Vac_AP group than AP group (Figure
429
3H), which is consistent with the observation of serum metabolomics (Figure
430
3C). In addition, since 2-HB is generated from 2-oxobutanoate by lactate
431
dehydrogenase (LDH) (from Kyoto Encyclopedia of Genes and Genomes
432
database, https://www.kegg.jp/), which is also present in some kinds of gut
433
microbiota [22-25], we specially analyzed the abundance of LDH-expressing
434
bacteria. Interestingly, we observed that the relative abundance of
435
LDH-expressing bacteria was also increased in Vac_AP group (Figure 3I).
436
Alltogether, our results suggest that increased 2-HB by Vac treatment may due
437
to altered gut microbiota.
438 439 440
3.4 2-HB attenuates AP-induced cellular toxicity To investigate the biological function of 2-HB, different hepatocytes were 20
441
treated with 2-HB at a series of concentrations including rat-derived liver cell
442
line BRL3A, mice-derived liver cell line AML12 and mice primary hepatocytes.
443
Interestingly, 2-HB treatment enhanced cell viability to different extent in
444
AML-12 and mice primary hepatocytes, but not BRL3A (Supplementary
445
Figure 4A-C). We then asked whether 2-HB could influence AP-induced cell
446
toxicity. The IC50 of AP on these three cell lines were determined first
447
(Supplementary Figure 4D-F), which were used for subsequent experiments
448
with or without addition of 2-HB for 24 or 48 h. We found that addition of 2-HB
449
significantly attenuated the AP-induced cell toxicity regardless of species
450
difference in cell origin (Figure 4A-C). Representative photographs of BRL3A
451
cells treated with AP for 48 h in the presence or absence of 2-HB co-culture
452
were presented in Figure 4D. In line with the cell viability results, AP treatment
453
resulted in a large portion of cells dead and floating, which was obviously
454
improved by 2-HB (Figure 4D). Furthermore, we observed that 2-HB addition
455
decreased the ratio of GSSG/GSH in mice primary hepatocytes compared to
456
AP alone (Figure 4E). Our current results indicated that 2-HB protected
457
AP-induced cell toxicity in either rat- or mouse-derived cell lines.
458 459
3.5 2-HB treatment protects AP-induced acute liver injury in mice
460
Given the observed evidence, we hypothesized that the attenuation on
461
AP-induced liver injury by Vac pretreatment was probably due to 2-HB
462
production. We observed the impact of 2-HB on AP-induced liver injury in a 21
463
group of male C57 mice, which were orally given with single-dose of AP (400
464
mg/kg) following 3 days of 2-HB (250 mg/kg per day, i.p) treatment. Results
465
showed that AP treatment induced significant liver injury in mice, whereas
466
2-HB pretreatment effectively protected mice from severe liver injury (Figure
467
5A and B). Moreover, we found that 2-HB decreased the peak concentration of
468
serum AP and reduced AUC0-t (Figure 5C). In addition, the serum
469
concentration of 2-HB was also detected before and after AP administration.
470
We observed that intraperitoneally injection of 2-HB definitely increased serum
471
levels of 2-HB, especially within the 1st h after AP treatment (Figure 5D).
472
Moreover, 2-HB pretreatment also inhibited mRNA expression of genes
473
involved in mediating inflammation (Tnf-α, Il-1β) (Figure 6A), cell proliferation
474
process (Btg2, Ccng1) and DNA damage induced genes (Ddit4l, Gadd45α)
475
(Figure 6C) that were stimulated by AP treatment, as well as the regulation of
476
genes in anti-oxidative pathways such as Nqo-1, Txnrd1, Car3 (Figure 6B)
477
and down-regulation of genes related with hepatotoxicity (Krt8, Krt18) (Figure
478
6C). In addition, 2-HB did not alter the protein expression of hepatic CYP2E1
479
or NQO-1 in AP-treated mice (Figure 6D, Supplementary Figure 5). Overall,
480
these data highlighted that 2-HB was protective against AP-induced liver injury
481
in mice.
482 483 484
4. Discussion In our current study, we demonstrated that Vac pretreatment attenuated 22
485
AP-induced liver injury in rats, and altered the composition of gut microbiota.
486
Then, our untargeted metabolomics revealed the significant metabolic change
487
by Vac pretreatment. Moreover, we found 2-HB was elevated in serum by Vac
488
pretreatment, which could reduce AP-induced toxicity in either rat- or
489
mouse-derived cell lines or primary hepatocytes, as well as acute liver injury in
490
mice.
491
AP-induced liver injury has been extensively investigated so far; however,
492
the underlying mechanism is not fully elucidated. In addition to the
493
conventional theory of NAPQI production [3, 5], emerging evidence has
494
indicated that the extent of AP-induced liver injury is dependent on microbial
495
composition because of the extensive co-metabolism on xenobiotic between
496
gut microbiota and host [7, 8]. Previously, Jeremy et al. demonstrate that the
497
character of baseline metabolic profile is predictive for the extent of
498
AP-induced liver injury [11], in which microbial metabolites such as taurine are
499
involved. Moreover, the variation of urine p-cresol in human subject influences
500
the metabolism of AP [7]. Recently, the impact of gut microbiota on AP-induced
501
liver injury is associated with changes of GSH levels [9] or alteration of AP
502
metabolism [8, 11]. In our current study, we first observed that obvious
503
inter-individual variation in levels of serum ALT and AST existed in AP orally
504
treated rats, but not in those with intraperitoneal injection. It is reasonable that
505
the bioavailability of orally administrated xenobiotics is apt to be impacted by
506
more factors than intraperitoneal injection, in particular gut microbiota [6]. We 23
507
further found that Vac pretreatment decreased the levels of serum ALT, AST
508
and the extent of liver necrosis caused by AP in either SD or Wistar rats. Vac is
509
a potent antibiotic against gram-positive bacteria and with limited systemic
510
impacts due to non-absorptive character in intestine [10, 26, 27]. As a result,
511
Vac is frequently used for investigating the role of gut microbiota in various
512
studies [28-30]. Our results convinced that alteration of gut microbiota was a
513
critical factor in determining the extent of AP-induced liver injury. Moreover, we
514
found that Vac pretreatment significantly decreased the bioavailability of AP.
515
Since the bioavailability of AP is influenced by intestinal absorption of AP,
516
which is deglucuronidated by microbiota produced β-GD [18, 19], we showed
517
that Vac pretreatment greatly depleted the activity of β-GD in cecum contents.
518
Therefore, it is possible that the depletion of intestinal β-GD reduces the
519
absorption of AP. Nevertheless, the change of AP bioavailability cannot fully
520
explain the reduced hepatotoxicity by Vac pretreatment. Moreover, the
521
expression of CYP2E1 protein was even up-regulated in Vac pretreated group,
522
which implied that the protective effect of Vac on AP-induced liver injury was
523
not due to reduced NAPQI production.
524
Metabolomics is increasingly applied to investigate the underlying
525
mechanisms of drug activity/toxicity because metabolomics is a unique
526
approach for quantitatively measurement of the time-related metabolic
527
response of living systems to pathophysiological stimuli or genetic modification
528
simultaneously [31, 32]. Gong et al. [9] reveals that gut microbiota determine 24
529
the diurnal variation of AP-induced liver injury through microbial metabolite,
530
1-phenyl-1,2-propanedione by using untargeted metabolomics. In our current
531
study, an untargeted GC-MS-based metabolomics was adopted to test
532
whether there are metabolites that are involved in attenuation of AP-induced
533
liver injury by Vac pretreatment. Our results showed that the metabolic profile
534
of AP was very different from others. Then, a total of 13 differential metabolites
535
were identified between Con and AP group that were restored by Vac
536
pretreatment,
537
AP-induced hepatotoxicity. Moreover, propanoate metabolism was one of the
538
most significantly altered metabolic pathways enriched with these differential
539
metabolites, in which metabolite 2-HB attracted our attention because of its
540
involvement in propanoate metabolism and dramatic fold change between Con
541
and AP groups. Consistently, our targeted metabolomics analysis convinced
542
the elevation of 2-HB by Vac pretreatment in cecum content with or without AP
543
treatment, suggesting that 2-HB production was associated with gut microbiota.
544
Our results showed serum 2-HB level was lower in Vac_AP than AP group
545
after 24 h of AP treatment. Oxidative stress or detoxification of xenobiotics in
546
the liver can dramatically increase the rate of hepatic glutathione synthesis.
547
Under such metabolic stress conditions, homocysteine is diverted from the
548
transmethylation pathway (which forms methionine) into the transsulfuration
549
pathway (which forms cystathionine). alpha-ketobutyrate is produced when
550
cystathionine is cleaved into cysteine that is incorporated into glutathione, and
suggesting
that
these
25
metabolites
were
relevant
with
551
2-HB is formed via a reaction catalyzed by LDH from alpha-ketobutyrate
552
(http://www.hmdb.ca/metabolites/HMDB0000008). So the elevation of serum
553
2-HB in AP group after 24 h of AP treatment may be a stress response
554
because of the upregulation of hepatic glutathione synthesis.
555
Meanwhile, 16S rRNA gene sequencing data indicated that Vac treatment
556
obviously altered the composition of gut microbiota. PICRUSt is a
557
computational
558
reconstruction algorithm to predict which gene families are present and then
559
combines gene families to estimate the composite metagenome, thus can
560
predict the functional composition of a metagenome using a database of
561
reference genomes [17]. Based on the PICRUSt analysis of 16S rRNA gene
562
sequencing data, the abundance of propanoate metabolism pathway involved
563
in 2-HB producing was upregulated in Vac_AP group. At genus level, Vac
564
treatment also upregulated the relative abundance of some LDH expressing
565
bacteria such as Lactobacillus, Clostridium, Fusobacterium, Desulfovibrio
566
[22-25]. As a result, we speculated that the attenuation on AP-induced liver
567
injury by Vac pretreatment might be associated with the increase of 2-HB.
approach
which
uses
an
extended
ancestral-state
568
In line with our hypothesis, our results showed that 2-HB significantly
569
improved cell viability under AP treatment in cell lines from either rats or mouse
570
such as rat-derived BRL3A cells, mouse-derived AML12 cells and mouse
571
primary hepatocytes, suggesting that the hepatoprotective effect of 2-HB is
572
trans-species. Cellular GSH is critical for the detoxification of AP through 26
573
conjugation with the toxic product NAPQI of AP. The accumulated NAPQI will
574
deplete cellular GSH resulting to oxidative stress-induced liver injury [33, 34].
575
We found 2-HB reduced the AP-induced upregulation of liver GSSG/GSH in
576
mouse primary hepatocytes that is consistent with its protective effect against
577
AP-induced cell toxicity. Given the higher inter-individual variation of
578
AP-induced liver injury among rats and observed trans-species protection of
579
2-HB against AP-induced toxicity in cell lines, the biological function of 2-HB
580
was further validated in AP-treated mice, instead of rats. We observed that
581
2-HB prevented AP-induced liver injury in mice. Interestingly, 2-HB also
582
decreased the Cmax and AUC0-t of AP, as well as regulated the expression of
583
genes involved in inflammation, oxidative stress, cell proliferation and DNA
584
damage process. Nevertheless, we are not quite clear about the link between
585
2-HB and AP bioavailability based on our current results. Further investigation
586
will be conducted for explaining their potential relationship.
587
In summary, our current study demonstrates that Vac pretreatment
588
attenuates AP-induced liver injury, alters serum metabolic and gut microbiota
589
profiles in rats. The identified metabolite 2-HB has trans-species protective
590
effect against AP-induced cell toxicity in either rat- or mouse-derived cells,
591
which is further validated in mice. The protective effect of 2-HB is associated
592
with the reduction of AP bioavailability and increase of GSH levels. This study
593
convinces the impacts of gut microbiota on AP-induced hepatotoxicity, and
594
sheds light on the potential to prevent AP-induced hepatotoxicity by 27
595
modulating gut microbiota with antibiotic or other approaches such as prebiotic
596
or probiotic if specific functional bacteria was determined in the future.
597 598
Author contributions
599
Houkai Li and Wei Jia supervised the whole study and revised the
600
manuscript; Ningning Zheng conducted all of the in vivo and in vitro
601
experiments and data analysis; Yu Gu helped in targeted metabolomics of
602
serum and cecum samples with LC-MS/MS; Ying Hong, Lili Sheng and
603
Weidong Zhang helped in study design and data analysis; Linlin Chen helped
604
in the animal experiments; Feng Zhang helped in analysis of hepatic CYP450
605
enzymatic activity; Zimiao Weng helped in the cecum β-GD activity assay;
606
Zean Zhang helped in histological analysis of liver tissue. All authors have read
607
and approved the final version of the manuscript.
608 609
Acknowledgments
610
This work was funded by National Natural Science Foundation of China
611
(No. 81873059 & 81673662), & National Key Research and Development
612
Program of China (No. 2017YFC1700200), & Shuguang Scholar (16SG36) at
613
Shanghai Institutions of Higher Learning from Shanghai Municipal Education
614
Commission.
615 616
Conflicts of interest 28
The authors declare that there are no conflicts of interest.
617 618 619
References
620
1.
G. Ostapowicz, R. J. Fontana, F. V. Schiodt, et al., Results of a
621
prospective study of acute liver failure at 17 tertiary care centers in the
622
United States. Ann. Intern. Med. 137(2002) 947-54.
623
2.
376(2010) 190-201.
624 625
3.
L. F. Prescott, Kinetics and metabolism of paracetamol and phenacetin. Br. J. Clin. Pharmacol. 10 Suppl 2(1980) 291S-298S.
626 627
W. Bernal, G. Auzinger, A. Dhawan, et al., Acute liver failure. Lancet.
4.
C. K. Gelotte, J. F. Auiler, J. M. Lynch, et al., Disposition of
628
acetaminophen at 4, 6, and 8 g/day for 3 days in healthy young adults.
629
Clin. Pharmacol. Ther. 81(2007) 840-8.
630
5.
J. R. Mitchell, D. J. Jollow, W. Z. Potter, et al., Acetaminophen-induced
631
hepatic necrosis. IV. Protective role of glutathione. J. Pharmacol. Exp.
632
Ther. 187(1973) 211-7.
633
6.
xenobiotic metabolism. Pharmacol. Res. 69(2013) 21-31.
634 635
H. J. Haiser, P. J. Turnbaugh, Developing a metagenomic view of
7.
T. A. Clayton, D. Baker, J. C. Lindon, et al., Pharmacometabonomic
636
identification of a significant host-microbiome metabolic interaction
637
affecting human drug metabolism. Proc. Natl. Acad. Sci. U S A.
638
106(2009) 14728-33. 29
639
8.
S. H. Lee, J. H. An, H. J. Lee, et al., Evaluation of pharmacokinetic
640
differences of acetaminophen in pseudo germ-free rats. Biopharm.
641
Drug. Dispos. 33(2012) 292-303.
642
9.
S. Gong, T. Lan, L. Zeng, et al., Gut microbiota mediates diurnal
643
variation of acetaminophen induced acute liver injury in mice. J. Hepatol.
644
69(2018) 51-59.
645
10.
I. K. Yap, J. V. Li, J. Saric, et al., Metabonomic and microbiological
646
analysis of the dynamic effect of vancomycin-induced gut microbiota
647
modification in the mouse. J. Proteome. Res. 7(2008) 3718-28.
648
11.
T. A. Clayton, J. C. Lindon, O. Cloarec, et al., Pharmaco-metabonomic
649
phenotyping and personalized drug treatment. Nature. 440(2006)
650
1073-7.
651
12.
X. He, N. Zheng, J. He, et al., Gut Microbiota Modulation Attenuated the
652
Hypolipidemic Effect of Simvastatin in High-Fat/Cholesterol-Diet Fed
653
Mice. J. Proteome. Res. 16(2017) 1900-1910.
654
13.
X. Zhang, Y. Zhao, M. Zhang, et al., Structural changes of gut
655
microbiota during berberine-mediated prevention of obesity and insulin
656
resistance in high-fat diet-fed rats. PLoS. One. 7(2012) e42529.
657
14.
Q. Wang, L. Jiang, J. Wang, et al., Abrogation of hepatic ATP-citrate
658
lyase protects against fatty liver and ameliorates hyperglycemia in leptin
659
receptor-deficient mice. Hepatology. 49(2009) 1166-75.
660
15.
Z. M. Weng, P. Wang, G. B. Ge, et al., Structure-activity relationships of 30
661
flavonoids as natural inhibitors against E. coli beta-glucuronidase. Food.
662
Chem. Toxicol. 109(2017) 975-983.
663
16.
J. H. Jo, S. Kim, T. W. Jeon, et al., Investigation of the Regulatory
664
Effects of Saccharin on Cytochrome P450s in Male ICR Mice. Toxicol.
665
Res. 33(2017) 25-30.
666
17.
M. G. Langille, J. Zaneveld, J. G. Caporaso, et al., Predictive functional
667
profiling of microbial communities using 16S rRNA marker gene
668
sequences. Nat. Biotechnol. 31(2013) 814-21.
669
18.
N. Watari, M. Iwai,N. Kaneniwa, Pharmacokinetic study of the fate of
670
acetaminophen and its conjugates in rats. J. Pharmacokinet. Biopharm.
671
11(1983) 245-72.
672
19.
C. P. Siegers, K. Rozman,C. D. Klaassen, Biliary excretion and
673
enterohepatic circulation of paracetamol in the rat. Xenobiotica.
674
13(1983) 591-6.
675
20.
C. J. Patten, P. E. Thomas, R. L. Guy, et al., Cytochrome P450
676
enzymes involved in acetaminophen activation by rat and human liver
677
microsomes and their kinetics. Chem. Res. Toxicol. 6(1993) 511-8.
678
21.
K. E. Thummel, C. A. Lee, K. L. Kunze, et al., Oxidation of
679
acetaminophen to N-acetyl-p-aminobenzoquinone imine by human
680
CYP3A4. Biochem. Pharmacol. 45(1993) 1563-9.
681 682
22.
S. Chaillou, M. C. Champomier-Verges, M. Cornet, et al., The complete genome sequence of the meat-borne lactic acid bacterium Lactobacillus 31
sakei 23K. Nat. Biotechnol. 23(2005) 1527-33.
683 684
23.
M. Monot, C. Boursaux-Eude, M. Thibonnier, et al., Reannotation of the
685
genome sequence of Clostridium difficile strain 630. J. Med. Microbiol.
686
60(2011) 1193-9.
687
24.
V. Kapatral, I. Anderson, N. Ivanova, et al., Genome sequence and
688
analysis of the oral bacterium Fusobacterium nucleatum strain ATCC
689
25586. J. Bacteriol. 184(2002) 2005-18.
690
25.
J. F. Heidelberg, R. Seshadri, S. A. Haveman, et al., The genome
691
sequence of the anaerobic, sulfate-reducing bacterium Desulfovibrio
692
vulgaris Hildenborough. Nat. Biotechnol. 22(2004) 554-9.
693
26.
Dis. 42 Suppl 1(2006) S3-4.
694 695
R. C. Moellering, Jr., Vancomycin: a 50-year reassessment. Clin. Infect.
27.
N. J. Pultz, U. Stiefel,C. J. Donskey, Effects of daptomycin, linezolid,
696
and vancomycin on establishment of intestinal colonization with
697
vancomycin-resistant
698
extended-spectrum-beta-lactamase-producing Klebsiella pneumoniae
699
in mice. Antimicrob. Agents. Chemother. 49(2005) 3513-6.
700
28.
enterococci
and
A. Zarrinpar, A. Chaix, Z. Z. Xu, et al., Antibiotic-induced microbiome
701
depletion alters metabolic homeostasis by affecting gut signaling and
702
colonic metabolism. Nat. Commun. 9(2018) 2872.
703 704
29.
I. Hwang, Y. J. Park, Y. R. Kim, et al., Alteration of gut microbiota by vancomycin and bacitracin improves insulin resistance via glucagon-like 32
peptide 1 in diet-induced obesity. FASEB. J. 29(2015) 2397-411.
705 706
30.
S. Fujisaka, J. Avila-Pacheco, M. Soto, et al., Diet, Genetics, and the
707
Gut Microbiome Drive Dynamic Changes in Plasma Metabolites. Cell.
708
Rep. 22(2018) 3072-3086.
709
31.
J. K. Nicholson, J. C. Lindon,E. Holmes, 'Metabonomics': understanding
710
the metabolic responses of living systems to pathophysiological stimuli
711
via multivariate statistical analysis of biological NMR spectroscopic data.
712
Xenobiotica. 29(1999) 1181-9.
713
32.
J. K. Nicholson, J. Connelly, J. C. Lindon, et al., Metabonomics: a
714
platform for studying drug toxicity and gene function. Nat. Rev. Drug.
715
Discov. 1(2002) 153-61.
716
33.
H. Jaeschke, T. R. Knight,M. L. Bajt, The role of oxidant stress and
717
reactive nitrogen species in acetaminophen hepatotoxicity. Toxicol. Lett.
718
144(2003) 279-88.
719
34.
H. Jaeschke, M. R. Mcgill,A. Ramachandran, Oxidant stress,
720
mitochondria, and cell death mechanisms in drug-induced liver injury:
721
lessons learned from acetaminophen hepatotoxicity. Drug. Metab. Rev.
722
44(2012) 88-106.
723 724
Figure legends
725
Figure 1 Vac pretreatment attenuates AP-induced acute liver injury in rats.
726
(A and B) Serum ALT and AST levels. i.g and i.p represent oral gavage or 33
727
intraperitoneal injection with a single dose of 2.0 g/kg or 1.0 g/kg of AP,
728
respectively (n = 6-14). (C and D) Serum ALT, AST, hepatic H&E staining, and
729
histology score in SD rats treated with Vac (100 mg/kg, twice a day) and/or with
730
AP (i.g, 2.0 g/kg). n = 5-6 in Con or Vac group. n = 29 in AP and 20 in Vac_AP
731
groups. Data were mean ± S.E.M. The magnification of H&E staining
732
photograph is 200х. * p < 0.05; ** p < 0.01.
733
Figure 2. Vac pretreatment decreases the bioavailability of AP and ratio
734
of GSSG/GSH. (A) The serum concentrations of AP in SD rats at different time
735
points (n = 4). (B) The activity of β-glucuronidase (β-GD) in cecum. n = 5 in
736
Con or Vac group, and 10 in AP or Vac_AP group. (C and D) Hepatic
737
GSSG/GSH and mRNA expression of genes (n = 6-8). (E and G) The hepatic
738
protein levels of CYP3A4 and CYP2E1 (n = 6) as well as (F and H) enzymic
739
activity of CYP1A2 and CYP2E1 measured with LC-MS/MS (n = 10 in Con or
740
Vac group and 20 in AP or Vac_AP group). Data were mean ± S.E.M. * p <
741
0.05, ** p < 0.01.
742
Figure 3. Vac pretreatment elevates 2-HB in both cecum and serum. (A)
743
PLS-DA plot of serum metabolites (n = 5). (B) Differential metabolites among
744
groups (n = 5). (C) Pathway analysis based on the 13 differential metabolites
745
in B. The quantitation of 2-HB in cecum contents (D) and serum (E) of rats 24 h
746
after AP gavage (n = 5-8). (F-G) PCoA analysis at OTU level and column
747
diagram at phylum level based on 16S rRNA gene sequencing (n = 5). (H) The
748
abundance of propanoate metabolism pathway based on PICRUSt analysis of 34
749
16S rRNA gene sequencing (n = 5). (I) Heatmap of the relative abundance of
750
LDH-producing bacteria at genus level (n = 5). Data were mean ± S.E.M. * p <
751
0.05; ** p < 0.01.
752
Figure 4. 2-HB attenuates AP-induced cellular toxicity. (A) BRL3A cells, (B)
753
AML12 cells, and (C) mouse primary hepatocytes were treated with AP alone
754
or in combination with 5-40 mM 2-HB for indicated periods, and cell viability
755
was measured with CCK-8 kit (n=6). (D) Representative photographs of
756
BRL3A cells treated with AP alone or in combination with 5-40 mM 2-HB for 48
757
h (The magnification of photographs is 100х). (E) Cellular GSSG/GSH was
758
analyzed in primary hepatocytes treated with AP alone or in combination of 5
759
mM 2-HB for 24 h (n = 3). Data were mean ± S.E.M. * p < 0.05, ** p < 0.01.
760
Figure 5. 2-HB treatment protects AP-induced acute liver injury in mice.
761
Mice were intraperitoneally injected with 2-HB (250 mg/kg, once a day) for 4
762
days and administrated with a single dose of AP (400mg/kg,) 30 min after the
763
last injection of 2-HB. (A-B) Serum ALT, AST, and liver histology in mice 24 h
764
after AP treatment (n = 5-7). * p< 0.05, ** p < 0.01. (C-D) Serum concentrations
765
of AP and 2-HB quantified with LC-MS/MS in mice treated with AP and/or 2-HB
766
(n = 6). * p< 0.05, ** p < 0.01 compared to the same time point between the
767
two groups. Data were mean ± S.E.M.
768
Figure 6. 2-HB normalizes gene expression. (A) Inflammation related genes.
769
(B) Oxidative stress related genes. (C) Cell proliferation and DNA damage
770
induced genes. (D) Hepatic CYP2E1 and NQO-1 protein levels. n = 5-7 in A-D. 35
771
Data were mean ± S.E.M. * p < 0.05, ** p < 0.01.
772 773
Supplementary Figure 1. Vac pretreatment attenuates AP-induced acute
774
liver injury and the bioavailability of AP in Wistar rats. (A-B) Serum ALT,
775
AST, hepatic H&E staining, and histology score in rats orally gavaged with Vac
776
(100 mg/kg, twice a day) and/or AP (1.0g/kg). n = 5 in Con or Vac group, n = 20
777
in AP or Vac_AP group. Data were mean ± S.E.M. The magnification of H&E
778
staining photograph is 200х. * p < 0.05; ** p < 0.01. (C) The concentration of
779
serum AP in rats at different time points, n = 4.
780
Supplementary Figure 2. Raw data. Original uncropped scan images of
781
Figures 2E and 2G.
782
Supplementary Figure 3. Vac alters gut microbiota composition in rats.
783
Heatmap analysis of the top 50 genera in AP and Vac_AP group. n = 5. * p <
784
0.05, ** p < 0.01 with Wilcoxon rank-sum test.
785
Supplementary Figure 4. (A-C) Cell viability of BRL3A cells, AML12 cells, and
786
C57 mice primary hepatocytes treated with 5-40 mM 2-HB for indicated time.
787
(D-F) The IC50 values of AP in BRL3A, AML12 and C57 mice primary
788
hepatocytes determined by non-linear regression. n = 6 in A-F.
789
Supplementary Figure 5. Raw data. Original uncropped scan images of
790
Figure 6D.
791
Supplementary Table 1 Common differential metabolites among groups.
792 36
Highlights 1. Vac pretreatment attenuated AP-induced liver injury in rats. 2. Vac pretreatment elevated metabolite 2-HB both in cecum and serum. 3. 2-HB attenuated the AP-induced hepatotoxicity both in vitro and in vivo.