ArticlePDF Available

Caffeic acid prevents acetaminophen-induced liver injury by activating the Keap1-Nrf2 antioxidative defense system

Authors:

Abstract and Figures

Unlabelled: Acute liver failure induced by acetaminophen (APAP) overdose is the main cause of drug-induced liver injury (DILI). Caffeic acid (CA) is a phenolic compound from many natural products. This study aims to investigate the protective mechanism of CA in APAP-induced liver injury. The results of serum alanine/aspartate aminotransferases (ALT/AST), liver myeloperoxidase (MPO) activity, liver glutathione (GSH) and reactive oxygen species (ROS) levels demonstrated the protection of CA against APAP-induced liver injury. Liver histological observation provided further evidences of CA-induced protection. CA was found to reverse the APAP-induced decreased cell viability in human normal liver L-02 cells and HepG2 cells. CA also reduced the increased cellular ROS level induced by APAP in hepatocytes. The results of luciferase assay and Western-blot analysis showed that CA increased the transcriptional activation of nuclear factor erythroid 2-related factor 2 (Nrf2) in the presence of APAP. Nrf2 siRNA reduced the protection of CA against APAP-induced hepatotoxicity. CA also reversed the APAP-induced decreased mRNA and protein expression of heme oxygenase 1 (HO-1) and Nad(p)h: quinone oxidoreductase 1(NQO1). In addition, HO-1 inhibitor zinc protoporphyrin (ZnPP) and NQO1 inhibitor diminutol (Dim) reduced the protection of CA against APAP-induced hepatotoxicity. CA also decreased the expression of kelch-like ECH-associated protein-1(Keap1). Molecular docking indicated the potential interacting of CA with Nrf2 binding site in the Keap1 protein. CA had little effect on the enzymatic activity of cytochrome P450 (CYP) 3A4 and CYP2E1 in vitro. In conclusion, we demonstrated that CA prevented APAP-induced hepatotoxicity by decreasing Keap1 expression, inhibiting binding of Keap1 to Nrf2, and thus activating Nrf2 and leading to increased expression of antioxidative signals including HO-1 and NQO1.
Content may be subject to copyright.
Original Contribution
Caffeic acid prevents acetaminophen-induced liver injury by activating
the Keap1-Nrf2 antioxidative defense system
Chun Pang
a,c,1
, Zhiyong Zheng
a,1
, Liang Shi
a
, Yuchen Sheng
b
, Hai Wei
c
,
Zhengtao Wang
a
, Lili Ji
a,
n
a
Shanghai Key Laboratory of Complex Prescription and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica,
Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China
b
Center for Drug Safety Evaluation and Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
c
Center for Traditional Chinese Medicine and Systems Biology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
article info
Article history:
Received 23 August 2015
Received in revised form
14 December 2015
Accepted 19 December 2015
Available online 23 December 2015
Keywords:
Caffeic acid
Acetaminophen
Hepatotoxicity
Nrf2
Keap1
abstract
Acute liver failure induced by acetaminophen (APAP) overdose is the main cause of drug-induced liver
injury (DILI). Caffeic acid (CA) is a phenolic compound from many natural products. This study aims to
investigate the protective mechanism of CA in APAP-induced liver injury. The results of serum alanine/
aspartate aminotransferases (ALT/AST), liver myeloperoxidase (MPO) activity, liver glutathione (GSH) and
reactive oxygen species (ROS) levels demonstrated the protection of CA against APAP-induced liver in-
jury. Liver histological observation provided further evidences of CA-induced protection. CA was found to
reverse the APAP-induced decreased cell viability in human normal liver L-02 cells and HepG2 cells. CA
also reduced the increased cellular ROS level induced by APAP in hepatocytes. The results of luciferase
assay and Western-blot analysis showed that CA increased the transcriptional activation of nuclear factor
erythroid 2-related factor 2 (Nrf2) in the presence of APAP. Nrf2 siRNA reduced the protection of CA
against APAP-induced hepatotoxicity. CA also reversed the APAP-induced decreased mRNA and protein
expression of heme oxygenase 1 (HO-1) and NAD(P)H: quinone oxidoreductase 1(NQO1). In addition,
HO-1 inhibitor zinc protoporphyrin (ZnPP) and NQO1 inhibitor diminutol (Dim) reduced the protection
of CA against APAP-induced hepatotoxicity. CA also decreased the expression of kelch-like ECH-asso-
ciated protein-1(Keap1). Molecular docking indicated the potential interacting of CA with Nrf2 binding
site in the Keap1 protein. CA had little effect on the enzymatic activity of cytochrome P450 (CYP) 3A4 and
CYP2E1 in vitro. In conclusion, we demonstrated that CA prevented APAP-induced hepatotoxicity by
decreasing Keap1 expression, inhibiting binding of Keap1 to Nrf2, and thus activating Nrf2 and leading to
increased expression of antioxidative signals including HO-1 and NQO1.
&2015 Elsevier Inc. All rights reserved.
1. Introduction
Caffeic acid (CA) is a natural polyphenolic compound derived
from coffee, some fruits and traditional Chinese medicines [17].
CA and its analogs have shown a variety of pharmacological ac-
tivities including anti-inammation, anti-cancer and anti-virus [8].
In addition, this compound is easily absorbed through the gut
barrier [9], so enhancing its mode of action. Previous reports have
already shown that CA prevented liver reperfusion injury and
griseofulvin-, nickel-, or doxorubicin-induced hepatotoxicity; fur-
thermore its antioxidative capacity contributes to protecting liver
against injury [1013]. Previously, we demonstrated that CA, as a
main compound in medicinal herb Flos Lonicerae, prevented
acetaminophen (APAP)-induced hepatotoxicity in human normal
liver L-02 cells in vitro [7]. CA has also been reported to inhibit
APAP-induced liver injury in both mice and rats in vivo [14].
However, the precise mechanism involved in its protection
Contents lists available at ScienceDirect
journal homepage: www.elsevier.com/locate/freeradbiomed
Free Radical Biology and Medicine
http://dx.doi.org/10.1016/j.freeradbiomed.2015.12.024
0891-5849/&2015 Elsevier Inc. All rights reserved.
Abbreviations: APAP, acetaminophen; DILI, drug-induced liver injury; CA, Caffeic
acid; ALT/AST, alanine/aspartate aminotransferases; MPO, myeloperoxidase; GSH,
glutathione; ROS, reactive oxygen species; Nrf2, nuclear factor erythroid 2-related
factor 2; HO-1, heme oxygenase 1; NQO1, NAD(P)H: quinone oxidoreductase 1;
ZnPP, zinc protoporphyrin; Dim, diminutol; Keap1, kelch-like ECH-associated
protein-1; CYP, cytochrome P450; NAPQI, N-acetyl-p-benzoquinone imine; NAC,
N-acetylcysteine; H
2
DCFDA, 2ʹ-7ʹ-Dichlorodihydrouorescein diacetate; FBS, fetal
bovine serum; BSO,
L
-Buthionine-(S, R)-sulfoximine; H&E, haematoxylin and eosin;
MTT, 3-(4,5-dimethyl-thiazol-2-yl) 2,5-diphenyltetrazolium bromide; TRE, tran-
scription response element; DDTC, sodium diethyldithiocarbamate trihydrate; GCL,
glutamate-cysteine ligase; ARE, antioxidant-related elements; GR, glutathione
reductase
n
Corresponding author.
E-mail address: lichenyue1307@126.com (L. Ji).
1
These two authors contributed equally to this paper.
Free Radical Biology and Medicine 91 (2016) 236246
remains unclear.
Drug-induced liver injury (DILI) is a major clinical problem.
Acute liver failure induced by APAP overdose is common, and is
reported to be the main reason for DILI in the United States and
the United Kingdom [15,16]. Despite great efforts made over the
last 40 years, the mechanism of APAP-induced liver injury is still
not completely elucidated. However, it is widely accepted that
APAP can be metabolized by liver cytochrome P450 (CYP) into a
reactive metabolite named N-acetyl-p-benzoquinone imine (NAP-
QI), which depleted cellular glutathione (GSH) and disturbed cel-
lular redox balance, and thus led to oxidative stress-induced liver
injury [17,18]. The commonly used antidote for APAP detoxica-
tion is N-acetylcysteine (NAC), which is a precursor for cellular
GSH synthesis and also is a well-known antioxidant [19]. Although
NAC attenuates APAP-induced hepatotoxicity, some patients may
still develop liver injury despite administration of recommended
dosage [20]. Therefore, there is a need to nd a more effective and
safe drug for APAP detoxication.
The transcription factor nuclear factor erythroid 2-related fac-
tor 2 (Nrf2) regulates the constitutive and inducible expression of
a variety of genes involved in drug metabolism, detoxication, and
antioxidative defenses [21]. Kelch-like ECH-associated protein 1
(Keap1), an adapter subunit of Cullin 3-based E3 ubiquitin ligase,
regulates the degradation of Nrf2 [22]. The Keap1-Nrf2 system is
thought to play a critical role in liver oxidative injury, and has been
considered as a prospective target for liver disease [21,22]. Here,
we investigated CA-induced protection against APAP-induced liver
oxidative injury, and the involvement of the Keap1-Nrf2 signaling
pathway.
2. Materials and methods
2.1. Chemical compounds and reagents
CA was purchased from Beijing Aoke Biological technology Co.,
Ltd (Beijing, China), and the purity is over 98.5%. Kits for analysis of
alanine/aspartate aminotransferase (ALT/AST) and myeloperox-
idase (MPO) were purchased from Nanjing Jiancheng Bioengi-
neering Institute (Nanjing, China). Cignal
Reporter Assay kit for
Nrf2&Nrf1, RNeasy
s
Plus Mini kit and Attractene were bought from
Qiagen (Hilden, German). Duan-Glo
s
Luciferase Assay System was
purchased from Promega (Madison, WI). Vivid
s
CYP2E1, Vivid
s
CYP1A2, and Vivid
s
CYP3A4 kits, Opti-MEM
s
, lipofectamine
RNAiMAX, 2ʹ-7ʹ-Dichlorodihydrouorescein diacetate (H
2
DCFDA),
RPMI1640, and fetal bovine serum (FBS) were purchased from Life
Technology (Carlsbad, CA). NE-PER
s
nuclear and cytoplasmic ex-
traction reagents, and Pierce
s
BCA Protein Assay Kit were pur-
chased from ThermoFisher Scientic (Waltham, MA). Whole cell
protein extraction kit and enhanced chemiluminescence kit were
all obtained from Millipore (Darmstadt, Germany). Antibodies for
immunoblotting including anti-Actin, -Lamin B, and -Keap1 were
all purchased from Cell Signaling Technology (Danvers, MA) (all
1:1000 dilutions). Antibodies for immunoblotting including anti-
Nrf2, -HO-1, -GCLC, -GCLM, and -NQO1 were all bought from Santa
Cruz (Santa Cruz, CA) (all 1:200 dilutions). Peroxidase-conjugated
goat anti-rabbit immunoglobulin G (IgG) (H þL) and anti-mouse
IgG (HþL) were purchased from Jackson ImmunoResearch (West
Grove, PA). PrimeScript
s
RT Master Mix and SYBR
s
Premix Ex Taq
were bought from Takara (Shiga, Japan). Control siRNA and Nrf2
siRNA were both purchased from Santa Cruz (Santa Cruz, CA).
APAP, NAPQI,
L
-Buthionine-(S, R)-sulfoximine (BSO), ZnPP, Dim and
other reagents unless indicated were purchased from Sigma Che-
mical Co. (St. Louis, MO).
2.2. Animals and treatments
Specic pathogen-free male ICR mice (1620 g body weight)
were purchased from Shanghai Laboratory Animal Center of Chi-
nese Academy of Science (Shanghai, China). The animals were
supplied with standard laboratory diet and water ad libitum at a
temperature 2271°C with a 12 h lightdark cycle (6:0018:00)
and 6575% humidity. All animals were received humane care in
compliance with the institutional animal care guidelines approved
by the Experimental Animal Ethical Committee, Shanghai Uni-
versity of Traditional Chinese Medicine.
Forty mice were randomly divided into 4 groups: (1) vehicle
control, (2) APAP (400 mg/kg), (3) APAP (400 mg/kg) þCA (10 mg/
kg), and (4) APAP (400 mg/kg) þCA (30 mg/kg). Mice were pre-
administered orally with CA (10, 30 mg/kg per day) for 7 con-
secutive days. On the last day, mice were orally given a single dose
of APAP (400 mg/kg) after administration of CA for 1 h. Animals
were then killed 4 h after APAP intoxication, and plasma and liver
tissue were collected.
For analyzing the effects of CA alone, 24 ICR mice were ran-
domly divided into 3 groups: (1) vehicle control, (2) CA (10 mg/
kg), and (3) CA (30 mg/kg). Mice were administered orally with CA
(10, 30 mg/kg per day) for 7 consecutive days. Animals were then
killed 5 h after the last administration of CA, and plasma and liver
tissue were collected.
2.3. Analysis of serum ALT/AST activities
The blood samples obtained were kept at room temperature for
2 h. Serum was then collected after centrifugation at 840 gfor
15 min. Serum ALT and AST were measured with kits according to
the manufacturer's instructions.
2.4. Analysis of liver GSH amount
Liver GSH amount was determined according to our previous
reported method [23].
2.5. Analysis of liver MPO activity
Liver MPO enzymatic activity was determined according to the
manufacturer's instruction. Protein concentration was detected by
BCA kit and MPO activity was expressed as units/g protein.
2.6. Liver histological observation
Slices of mice livers were xed in 10% phosphate buffered
saline (PBS)formalin for at least 24 h and then embedded in
parafn for histological assessment of tissue damage. Samples
were subsequently sectioned (5
μ
m), stained with haematoxylin
and eosin (H&E), and then observed under a light microscope
(Olympus, Japan) to evaluate liver damage.
2.7. Cell culture
The L-02 cell line was derived from an adult human normal
liver [24] (Cell Bank, Type Culture Collection of Chinese Academy
of Sciences, Shanghai). Hepatoma-derived HepG2 cell line was
obtained from the American Type Culture Collection (Manassas,
VA). L-02 or HepG2 cells were cultured in RPMI1640 or MEM
supplemented with 10% [v/v] fetal bovine serum, 2 mM glutamine,
100 U/ml penicillin and 100 mg/ml streptomycin.
2.8. Cell viability assay
Cells were seeded into 96-well plates. After attachment, cells
C. Pang et al. / Free Radical Biology and Medicine 91 (2016) 236246 237
were pre-incubated with or without various inhibitors for 15 min,
and then incubated with or without CA for another 15 min, and
then incubated with or without APAP or its metabolic product
NAPQI for another 48 h. After treatment, cells were incubated with
500
μ
g/ml 3-(4,5-dimethyl-thiazol-2-yl) 2,5-diphenyltetrazolium
bromide (MTT) for 4 h. The formed formazan in surviving cells was
dissolved in 10% SDS5% iso-butanol0.01 M HCl as described
previously [25], and the optical density was measured at 570 nm
with 630 nm as a reference. Cell viability was calculated as the
percentage of control.
2.9. Measurement of liver and cellular ROS
Cellular ROS were measured using the probe H
2
DCFDA as de-
scribed in our previous published paper [25]. As for detecting liver
ROS level, cold liver homogenate were centrifuged (10,000 g,
15 min, 4 °C). The supernatants were incubated with 10
μ
M
H
2
DCFDA in the dark for 1 h, and then were transferred to a Black
wall with clear bottom 96-well plate. Fluorescence was im-
mediately read at excitation 485720 nm, emission 525 720 nm
using a spectrophotometer (BioTek Synergy H4, Winooski, VT).
Protein concentrations in supernatants were assayed by BCA kits,
and all the results were calculated as units of uorescence per
microgram of protein and presented as percentage of control (% of
control).
2.10. Nrf2&Nrf1 luciferase reporter assay
Nrf2&Nrf1 luciferase reporter assay was performed as de-
scribed in Cignal
Reporter Assay kit. Briey, L-02 cells (2.5 10
4
per well) were transfected with Nrf2/1 transcription response
element (TRE) containing construct diluted in Opti-MEM
s
using
Attractene reagent. After 24 h of transfection, cells were pre-
treated with various concentrations of CA for 15 min, and then
incubated with APAP (7.5 mM) for additional indicated time. Lu-
ciferase activities were measured using the Duan-Glo
s
Luciferase
Assay System, and the constitutively expressed Renilla luciferase
was used as an internal control.
2.11. Protein extraction
Cells were seeded into dishes. After attachment, cells were pre-
incubated with or without CA (10, 50
μ
M) for 15 min, and then
incubated with or without APAP for additional indicated time.
After treatment, cellular proteins were isolated using whole cell
protein extraction kit according to the manufacturer's instruction.
Cytosolic and nuclear proteins were isolated as described in
NE-PER
s
nuclear and cytoplasmic extraction kit. Protein con-
centration was detected by BCA Kits, and all the samples in the
same experiment were normalized to the equal protein
concentration.
2.12. Western-blot analysis
Protein samples were separated by SDS-PAGE gel electrophor-
esis and electrophoretically transferred to PVDF membrane, and
then the membranes were probed with appropriate combination
of primary and horseradish peroxidase-conjugated secondary an-
tibodies. Proteins in the membranes were visualized by enhanced
chemiluminescence kits. The protein bands were quantied by the
average ratios of integral optic density following normalization to
the expression of internal control
β
-actin or Lamin B, and the re-
sults were further normalized to control.
2.13. siRNA transfection
L-02 cells were cultured in 60 mm dishes (for Western-blot
analysis) or 96-well plates (for cell viability assay). Control siRNA
(no silencing) and Nrf2 siRNA were transfected into cells by using
lipofectamine RNAiMAX. For cell viability assay, cells were pre-
treated with CA (10, 50
μ
M) for 15 min, and then incubated with
APAP (7.5 mM) for another 48 h, and surviving cells were de-
termined as described above.
2.14. RNA isolation and cDNA synthesis
L-02 cells were seeded into dishes. After attachment, cells were
pre-incubated with or without CA for 15 min, and then incubated
with APAP for additional 36 h. After treatment, cellular total RNA
was isolated by using RNeasy
s
Plus Mini kits. The RNA content was
determined by measuring the optical density at 260 nm, and cDNA
was synthesized according to the instruction described in
PrimeScript
s
RT Master Mix kit.
2.15. Real-time PCR analysis
Real-time PCR was performed by using SYBR
s
Premix Ex Taq
kit. Relative expression of target genes was normalized to GAPDH,
analyzed by 2
ΔΔ
Ct
method and given as ratio compared with the
control. The primer sequences used in this study are shown in the
Supplementary table.
2.16. Molecular docking analysis
To investigate the possible binding mode of CA to Keap1 as the
potential inhibitor, the molecular docking analysis was performed
as described in our previous study [26]. The conrmation of CA is
generated using Comformational Search (MMFF94X force eld) in
MOE2013.
2.17. CYP450 metabolic enzyme activity assay
The effects of CA on CYP2E1, CYP1A2 and CYP3A4 enzymatic
activity were analyzed by using Vivid
s
CYP2E1, CYP1A2, CYP3A4
Kits. The CYP450 positive inhibitors used for CYP2E1 is sodium
diethyldithiocarbamate trihydrate (DDTC), for CYP1A2 is furafyl-
line, for CYP3A4 is ketoconazole, respectively. The analysis of re-
sults was performed with a Kinetic Assay Mode using the fol-
lowing equation: % inhibition (of positive inhibitor)¼(
1
XB
AB
)
100%; X,Aand Brespectively represent the rate of test compound
CA, solvent control and positive inhibitor control.
2.18. Statistical analysis
Data were expressed as means7standard error of the mean.
The signicance of differences between groups was evaluated by
one-way ANOVA with LSD post hoc test, and Po0.05 was con-
sidered as statistically signicant differences.
3. Results
3.1. CA prevented APAP-induced liver injury in vivo
Fig. 1A showed that APAP (400 mg/kg) induced the elevation of
serum ALT and AST (Po0.01), whereas CA (30 mg/kg) reduced the
APAP-induced increased ALT and AST (Po0.01, Po0.05). However,
CA (10 mg/kg) had no signicant inhibition on the APAP-induced
increased ALT and AST. Fig. 1B showed that CA (10, 30 mg/kg) both
C. Pang et al. / Free Radical Biology and Medicine 91 (2016) 236246238
Fig. 1. CA prevented APAP-induced liver injury in ICR mice: (A) serum ALT/AST activity; (B) liver GSH amount; (C) liver MPO activity; and (D) liver ROS level. Data are
expressed as means7SEM (n¼10). **Po0.01, ***Po0.001 compared to control;
#
Po0.05,
##
Po0.01 compared to APAP.
Fig. 2. Histological observation of the protection of CA against APAP-induced liver injury.Typical images were chosen from each experimental group (original magnication
100 ), and the magnied images were shown in lower left corner in each picture (original magnication 200 ): (A) vehicle control, (B) APAP (400 mg/kg), (C) APAP
(400 mg/kg)þCA (10 mg/kg), and (D) APAP (400 mg/kg) þCA (30 mg/kg).
C. Pang et al. / Free Radical Biology and Medicine 91 (2016) 236246 239
reversed APAP-induced decreased liver GSH amount (Po0.05). As
shown in Fig. 1C and D, APAP (400 mg/kg) increased liver MPO
activity and ROS level (Po0.01, Po0.001), whereas CA (30 mg/kg)
reduced the increased MPO activity (Po0.05) and ROS level
(Po0.01), and CA (10 mg/kg) also reduced the increased ROS level
(Po0.01). CA (10, 30 mg/kg) alone increased liver GSH amount
(Po0.001), but had no effect on serum ALT/AST, liver MPO activity
and ROS level in mice (Supplementary Fig. 1).
Furthermore, mice treated with APAP (400 mg/kg) showed
severe liver damage, indicated by intrahepatic hemorrhage, lym-
phocytes inltration and the destruction of liver structure
(Fig. 2B). After CA (10, 30 mg/kg) treatment, such phenome were
all ameliorated (Fig. 2C and D).
3.2. CA attenuated APAP-induced cytotoxicity and oxidative stress
injury in vitro
As a normal hepatic cell line, L-02 cells are commonly used in
hepatotoxicity studies induced by various hepatotoxicants [27,28].
As shown in Fig. 3A, APAP (7.5, 10 mM) decreased cell viability as
compared to control (Po0.001), whereas CA (5, 10, 25, 50
μ
M)
reversed the deceased cell viability induced by APAP (10 mM) in
L-02 cells (Po0.05, Po0.001). In addition, CA (25, 50
μ
M) also
reversed the decreased cell viability induced by APAP (7.5 mM) in
L-02 cells (Po0.05, Po0.001). Further results in HepG2 cells de-
monstrated that CA (25, 50
μ
M) reversed the decreased cell via-
bility induced by APAP (10 mM) (Fig. 3B) (Po0.01, Po0.001).
As shown in Fig. 3C, APAP (7.5 mM) increased cellular ROS level
in both L-02 and HepG2 cells (Po0.001). Whereas, CA (50
μ
M)
reduced enhanced ROS level induced by APAP in both L-02 and
HepG2 cells (Po0.01). CA (10
μ
M) also reduced the enhanced ROS
level induced by APAP in L-02 cells (Po0.01).
3.3. CA induced the transcriptional activation of Nrf2
When L-02 cells were treated with APAP (7.5 mM) for 4 h, the
expression of nuclear Nrf2 was increased, but this increase was
reduced after CA (50
μ
M) treatment (Fig. 4A). While, there was no
obvious alternation of the expression of cytosolic Nrf2. After cal-
culating the gray density of protein bands, the results clearly de-
monstrated that CA (50
μ
M) reduced the increased nuclear Nrf2
expression induced by APAP when cells were treated with APAP
for 4 h (Po0.05) (Supplementary Fig. 2A). However, when L-02
cells were treated with APAP (7.5 mM) for 18 h or 36 h, the in-
creased expression of nuclear Nrf2 was diminished. Pretreatment
with CA (50
μ
M) increased the expression of nuclear Nrf2 when
cells were incubated with APAP for 36 h. Furthermore, data from
calculating the gray density of protein bands evidenced that CA
(50
μ
M) enhanced nuclear Nrf2 expression when cells were in-
cubated with APAP for 36 h (Po0.05) (Supplementary Fig. 2A). CA
(50
μ
M) also enhanced the expression of nuclear Nrf2 in HepG2
cells when cells were incubated with APAP for 36 h (Fig. 4B). The
results of densitometric analysis of Nrf2 protein bands further
implied that nuclear Nrf2 expression was enhanced by CA in
HepG2 cells (Po0.05) (Supplementary Fig. 2B).
The luciferase reporter assay demonstrated that APAP (7.5 mM)
enhanced the transcriptional activation of Nrf2 after incubated
with cells for 4, 8, and 12 h (Po0.05, Po0.001), but CA (50
μ
M)
reduced the APAP-induced increased Nrf2 transcriptional activa-
tion when cells were incubated with APAP for 4, 8 h (Po0.05)
(Fig. 4C). By extending the incubation period, APAP-induced Nrf2
activation was reduced (Fig. 4C). However, CA (50
μ
M) increased
Nrf2 transcriptional activation when cells were incubated with
APAP for 36 h (Po0.01) (Fig. 4C).
In order to conrm the important role of Nrf2 in regulating the
protection of CA against APAP-induced hepatotoxicity, Nrf2 siRNA
was used to knock down the expression of Nrf2 in L-02 cells
(Fig. 4D). As compared with control siRNA, Nrf2 siRNA decreased
the CA-induced elevated cell viability after APAP intoxication
(Po0.05) (Fig. 4E).
3.4. HO-1 and NQO1 were involved in the protection of CA against
APAP-induced hepatotoxicity
NQO1, HO-1, and glutamate-cysteine ligase (GCL) are important
cellular antioxidative enzymes, and NQO1, HO-1, and catalytic or
modify subunits of GCL (GCLC or GCLM) are all Nrf2-regulated
downstream genes [22]. As shown in Fig. 5A, APAP (7.5 mM) en-
hanced mRNA expression of GCLc (Po0.05), but CA (10, 50
μ
M)
did not further increase the mRNA expression of GCLc (P40.05).
The results in Fig. 5A also demonstrated that APAP and APAP plus
CA (10, 50
μ
M) had no signicant effect on the mRNA expression
of GCLm (P40.05). Moreover, APAP (7.5 mM) decreased mRNA
expression of both NQO1 and HO-1 (Po0.001) (Fig. 5B). After CA
(50
μ
M) treatment, the decreased mRNA expression of both NQO1
and HO-1 induced by APAP was reversed (Po0.05, Po0.01)
Fig. 3. CA attenuated APAP-induced cytotoxicity and oxidative stress injury in
hepatocytes: (A) CA prevented the hepatotoxicity induced by APAP (7.5 mM and
10 mM) in L-02 cells. (B) CA prevented the hepatotoxicity induced by APAP
(10 mM) in HepG2 cells. (C) CA reduced the APAP-induced elevation of cellular ROS
level in both L-02 and HepG2 cells. Data are expressed as means 7SEM (n¼3).
***Po0.001 compared to control;
#
Po0.05,
##
Po0.01,
###
Po0.001 compared to
APAP.
C. Pang et al. / Free Radical Biology and Medicine 91 (2016) 236246240
(Fig. 5B). Further, the protein expression of NQO1, HO-1, GCLC, and
GCLM was observed. As shown in Fig. 5C and D, there was no
obvious difference between the protein expression of GCLC and
GCLM with or without APAP. CA also had no effect on GCLC and
GCLM protein expression. APAP (7.5 mM) decreased HO-1 protein
expression (Po0.001), whereas CA (25, 50
μ
M) reversed the de-
creased HO-1 expression induced by APAP (Po0.05) (Fig. 5C and
D). APAP had no signicant effect on NQO1 protein expression
(Fig. 5C and D). However, CA (25, 50
μ
M) signicantly increased
NQO1 protein expression when pre-incubated with cells before
APAP intoxication (Po0.05) (Fig. 5C and D). Next, the chemical
inhibitors for NQO1, HO-1, and GCL were used to further evaluate
the roles of those antioxidative enzymes in regulating the pro-
tection of CA against APAP-induced hepatotoxicity. As shown in
Fig. 5E, GCL inhibitor BSO (both 20 and 50
μ
M) had no obvious
effect on the protection of CA against APAP-induced cytotoxicity.
Results in Fig. 5E showed that CA (50
μ
M) prevented APAP-in-
duced cytotoxicity (Po0.001), but such protection was abrogated
by both Dim (Po0.01, Po0.001) and ZnPP (Po0.05, Po0.01)
(both 20 and 50
μ
M), that are HO-1 and NQO1 inhibitors,
respectively.
3.5. The effects of CA on Keap1 expression and molecular docking
analysis
The effect of CA on the expression of Keap1, an inhibitor of Nrf2
activation, was observed. As shown in Fig. 6A and B, no apparent
effect on keap1 expression was shown after APAP (7.5 mM)
treatment in L-02 and HepG2 cells. After cells were pretreated
with CA (50
μ
M), the Keap1 protein expression was signicantly
decreased in the above hepatocytes (Po0.05). Molecular docking
analysis was used to analyze the potential binding of CA to Keap1
kelch domain, as it is important to determine whether small
molecules act as direct inhibitors of the proteinprotein interac-
tion between Keap1 and Nrf2. The chemical structure of CA was
shown in Fig. 6C. The docking mode of CA in the binding site of
human Keap1 was illustrated in Fig. 6D (front view) and Fig. 6E
(top view). The three-dimension (Fig. 6F) and two-dimension
(Fig. 6G) interaction-map showed that CA had one H-benzene
interaction between the benzene rings of chromone and Arg415,
which facilitated CA anchored into the binding site. In addition,
the hydrogen atom of the hydroxyl at 3-position in CA formed
water-mediated hydrogen bonds with Ser508, and the carbonyl
group of CA could form H-bond with Gly603 and Ser363, which
Fig. 4. CA induced the transcriptional activation of Nrf2: (A) L-02 cells were pre-incubated with CA (10, 50 μM) for 15 min, and then incubated with APAP (7.5 mM) for the
indicated time. The translocation of Nrf2 from cytoplasm into nucleus was detected by immunoblotting using specic antibody. β-actin and lamin B were used as loading
control for cytoplasm or nucleus, respectively. (B) HepG2 cells were pre-incubated with CA (10, 50 μM) for 15 min, and then incubated with APAP (7.5 mM) for 36 h. The
translocation of Nrf2 from cytoplasm into nucleus was detected by immunoblotting using specic antibody. β-actin and lamin B were used as loading control for cytoplasm
or nucleus, respectively. (C) L-02 cells were transfected with Nrf2/1 transcription response element (TRE) containing construct, and pre-incubated with CA (10, 50 μM) for
15 min, and then incubated with APAP (7.5 mM) for the indicated time. Luciferase activities were measured by using a luciferase assay system. Nrf2/1 luciferase activity was
expressed as a fold induction of control cells. Data are expressed as means7SEM (n¼3). *P o0.05, ***Po0.0 01 compared to control;
#
Po0.05,
##
Po0.01 compared to APAP.
(D) L-02 cells were transiently transfected with Nrf2 siRNA and the protein expression of Nrf2 was detected by immunoblotting using specic antibody, and β-actin and
Lamin B were used as loading control. Results represent three independent experiments. (E) L-02 cells were transiently transfected with Nrf2 siRNA, and then cells were pre-
incubated with or without CA for 15 min, and then cells were further incubated with or without APAP for 48 h. Finally, cell viability was detected by MTT assay, and the
results were obtained by deducting APAP alone. Data are expressed as means 7SEM (n¼6). *Po0.1 versus control siRNA.
C. Pang et al. / Free Radical Biology and Medicine 91 (2016) 236246 241
will help to enhance the binding afnity.
3.6. The effects of CA on the metabolic activity of CYP2E1, CYP1A2,
and CYP3A4 in vitro and NAPQI-induced hepatotoxicity
CYP2E1, CYP1A2, and CYP3A4 are regarded as the main meta-
bolic enzymes for converting APAP into the toxic and reactive
metabolite NAPQI [17,18]. Next, we observed whether CA will af-
fect the metabolic activity of CYP2E1, CY1A2, and CYP3A4 in vitro.
CA appeared to have limited inhibition on CYP2E1 over a range of
concentrations (Fig. 7A). For example, only less than 20% inhibition
of the positive inhibitor was noted at CA (100
μ
M) (Po0.05). Re-
sults of Fig. 7B showed that only the highest concentration of CA
(100
μ
M) had weak inhibition on CYP3A4 metabolic activity in
vitro (less than 20% inhibition of the positive inhibitor) (Po0.05).
Fig. 7C showed that CA had no effect on CYP1A2 metabolic activity
in vitro. Meanwhile, we also observed whether CA could prevent
NAPQI-induced hepatotoxicity in vitro. The results of Fig. 7D
showed that NAPQI (200
μ
M) decreased cell viability in both L-02
(Po0.05) and HepG2 (Po0.001) cells, whereas CA (50
μ
M) re-
versed the NAPQI-induced decreased cell viability in both above
cells (Po0.05).
Fig. 5. HO-1 and NQO1 were involved in the protection of CA against APAP-induced hepatotoxicity:(A) The mRNA expression of GCLc and GCLm was detected by Real-time
PCR. Data are expressed as means7SEM (n¼4). *Po0.05 versus control. (B) The mRNA expression of HO-1 and NQO1 was detected by Real-time PCR. Data are expressed as
means7SEM (n¼4). ***P o0.001 versus control,
#
Po0.05,
##
Po0.01 versus APAP. (C) The protein expression GCLC, GCLM, HO-1, and NQO1 was detected by im-
munoblotting using specic antibody, and β-actin was used as loading control. Results represent at least three repeated experiments. (D) The quantitative densitometric
analysis of GCLC, GCLM, HO-1, and NQO1 proteins. Data are expressed as means7SEM (n¼4). ***Po0.0 01 versus control,
#
Po0.05 versus APAP. (E) L-02 cells were pre-
incubated with BSO, ZnPP or Dim for 15 min, and then incubated with or without CA (50 μM) for another 15 min, and then incubated with APAP (7.5 mM) for another 48 h.
After treatment, the surviving cells were determined by MTT assay. The results are expressed in percentage of control and presented as the means7SEM (n¼6). ***Po0.001
versus control;
###
Po0.001 versus APAP;
$
Po0.05,
$$
Po0.01,
$$$
Po0.001 versus APAP plus CA.
C. Pang et al. / Free Radical Biology and Medicine 91 (2016) 236246242
4. Discussion
Polyphenols are the most abundant antioxidants distributed in
plant-derived food products, and phenolic acids (mainly CA) is one
of the main classes of polyphenols [29]. The measurement of
serum ALT and AST activities demonstrated that CA (30 mg/kg)
provided the protection against APAP-induced liver injury in mice
(Fig. 1A), and further liver histological evaluation conrmed this
protection (Fig. 2). Although CA (10 mg/kg) did not reduce APAP-
induced increased ALT/AST activities (Fig. 1A), it appears that CA
(10 mg/kg)-treated livers are healthier than CA (30 mg/kg)-treated
livers in histological evaluation (Fig. 2). Meanwhile, CA (10 mg/kg)
also reversed the decreased liver GSH amount (Fig. 1B) and in-
creased ROS level (Fig. 1D) induced by APAP in mice. These results
indicate the amelioration of CA (10 mg/kg) against APAP-induced
liver injury. The above in vivo results evidenced the protection of
Fig. 6. The effects of CA on Keap1 expression and molecular docking analysis: (A) the protein expression Keap1 was detected by immunoblotting using specic antibody, and
β-actin was used as loading control. Results represent four repeated experiments. (B) The quantitative densitometric analysis of Keap1 protein. Data are expressed as
means7SEM (n¼4).
#
Po0.05 versus APAP. (C) The chemical structure of CA. (D) Front view of the docking mode of CA (Green) in the binding site of Keap1 (shown in ribbon
representation and colored by structure). (E) Top view of the docking mode of CA (Green) in the binding site of Keap1 (shown in ribbon representation and colored by
structure). (F) Representative amino acid residues surrounding CA (Red) in the binding pocket of Keap1. (G) Two-dimensional interaction map of CA and the human Keap1.
The arrows indicate potential interactions between amino acid residues and CA. (For interpretation of the references to color in this gure legend, the reader is referred to the
web version of this article.)
C. Pang et al. / Free Radical Biology and Medicine 91 (2016) 236246 243
CA against APAP-induced liver injury, which are consistent with
previous reported study [14]. CA (10, 30 mg/kg) alone had no
obvious effect on serum ALT/AST activities when mice were orally
given with CA for 7 consecutive days (Supplementary Fig. 1A). In
addition, cell viability of L-02 and HepG2 cells demonstrated that
different concentrations of CA prevented APAP-induced cytotoxi-
city (Fig. 3A and B). Furthermore, CA reduced the APAP-induced
increased liver MPO activity (Fig. 1C), but CA alone had no ap-
parent effect on this enzyme in mice (Supplementary Fig. 1B).
Enhanced liver MPO activity generally indicates the occurrence of
liver inammation [30]. Thus, the above results suggest the po-
tential inhibition of CA on APAP-induced hepatic inammation.
These above results demonstrated that CA prevented APAP-in-
duced hepatotoxicity in vivo and in vitro.
Cellular GSH is critical for the detoxication of APAP when
conjugating with its metabolic product NAPQI. Thus, the accu-
mulation of NAPQI will deplete cellular GSH, so generating oxi-
dative stress-induced liver injury [17,18,31]. Our results displayed
the depletion of liver GSH in mice after APAP treatment; however
this was reversed by CA (10, 30 mg/kg) (Fig. 1B). Meanwhile, CA
(10, 30 mg/kg) also enhanced liver GSH level when CA alone was
orally given to mice (Supplementary Fig. 1C), which may con-
tribute to its antioxidant capacity. CA (10, 30 mg/kg) reduced the
APAP-induced formation of liver ROS in mice in vivo (Fig. 1D), and
it also abrogated the formation of cellular ROS in L-02 and HepG2
cells induced by APAP (Fig. 3C). However, CA (10, 30 mg/kg) alone
had no obvious effect on the formation of liver ROS in mice
(Supplementary Fig. 1D). Again, these results implied the protec-
tion of CA against APAP-induced liver oxidative stress injury.
Nrf2 is an important antioxidative transcription factor, which
has been reported to play a key role in protecting against APAP-
induced liver injury [32]. Nrf2 knockout mice were more suscep-
tible to APAP-induced liver injury [33]. Moreover, a variety of re-
ports demonstrated that some nature products prevented APAP-
induced hepatotoxicity by inducing the transcriptional activation
of Nrf2, some examples being quercetin, salvianolic acid B, gin-
senoside Rg3, sauchinone, and oleanolic acid [26,3437]. Firstly,
our ndings showed that CA pretreatment enhanced the expres-
sion of Nrf2 in nucleus when cells were incubated with APAP for
36 h (Fig. 4A and B, Supplementary Fig. 2). Secondly, luciferase
reporter assay also suggested that CA-induced Nrf2 transcriptional
activation occurred when hepatocytes were incubated with APAP
for 36 h (Fig. 4C). Thirdly, Nrf2 siRNA signicantly reduced the
protection of CA against APAP-induced hepatotoxicity (Fig. 4D and
E). Thus, it could be proposed that Nrf2 plays a critical role in
regulating the protection of CA against APAP-induced hepato-
toxicity. Our results also showed that APAP weakly induced Nrf2
activation when hepatocytes were incubated with APAP for 4, 8,
and 12 h (Fig. 4C). This is consistent with previous reported study
where the APAP metabolic product NAPQI can directly activate
Nrf2 [38]. However, CA reduced APAP-induced the increased Nrf2
activation after hepatocytes were incubated with APAP for 4 and
8h (Fig. 4C), which may be due to the reduced production of
NAPQI as our further results showed that CA weakly inhibited
CYP2E1 and CYP3A4 enzymatic activity (Fig. 7A and B). There may
be other signals regulating such phenome, which requires future
investigation.
The transcription factor Nrf2 regulates the expression of a
series of antioxidative genes, including NQO1, HO-1, GCLC and
GCLM, when binding to antioxidant-related elements (ARE)
[22,39]. It appears that CA reversed the APAP-induced decrease of
mRNA and protein expression of NQO1 and HO-1 (Fig. 5BD). In
addition, ZnPP and Dim reduced the protection of CA against
APAP-induced hepatotoxicity (Fig. 5E). These observations suggest
that CA activates Nrf2, which then enhances the expression of
NQO1 and HO-1, and thus contributes to CA-induced protection
against APAP-induced hepatotoxicity. APAP and APAP plus CA had
no obvious effects on either mRNA or protein expression of GCLM
Fig. 7. The effects of CA on the metabolic activity of CYP2E1, CYP1A2, and CYP3A4 in vitro and NAPQI-induced hepatotoxicity:(A) Inhibition of CA on CYP2E1 enzymatic
activity. (B) Inhibition of CA on CYP3A4 enzymatic activity. (C) Inhibition of CA on CYP1A2 enzymatic activity. Data are expressed as means7SEM (n¼4). *Po0.05 versus
solvent control. (D) L-02 and HepG2 cells were pre-incubated with CA (25, 50 μM) for 15 min, and then incubated with NAPQI (200 μM) for another 48 h. Af ter treatment, the
surviving cells were determined by MTT assay. The results are expressed in percentage of control and presented as the means7SEM (n¼6). *Po0.05,***Po0.001 versus
control;
#
Po0.05 versus NAPQI.
C. Pang et al. / Free Radical Biology and Medicine 91 (2016) 236246244
(Fig. 5A, C and D). Although APAP weakly enhanced the mRNA ex-
pression of GCLc (Fig. 5A), it had no signicant effect on the protein
expression of GCLC (Fig. 5C and D). Similarly, CA did not further
enhance the APAP-induced increased mRNA expression of GCLc
(Fig. 5A). BSO, a specic inhibitor for GCL which is composed of GCLC
and GCLM [40], also had no effect on CA-induced protection against
APAP-induced hepatotoxicity (Fig. 5E). Thus, we conclude that GCL is
not engaged in CA-induced protection against APAP-induced hepa-
totoxicity. GCL is critical for the de novo biosynthesis of cellular GSH
[41], which is important for the detoxication of APAP. But why GCL
is not involved in the protection of CA against APAP-induced liver
injury? We think that CA may directly conjugate with NAPQI, which
will contribute to the maintenance of cellular GSH level. Otherwise,
some other important enzymes for regulating cellular GSH amount
such as glutathione reductase (GR) may be involved.
Keap1, an inhibitor of Nrf2, acts as an adapter for Cul3/Rbx1-
mediated degradation of Nrf2 [22]. Our results showed that CA
reduced the expression of Keap1 protein in the presence of APAP
(Fig. 6A and B), and this may contribute to the activation of Nrf2
induced by CA. Furthermore, the outcome of molecular docking
(Fig. 6DG) suggests that CA may interact with Keap1 and occupy
the Nrf2 binding site in the protein, and thus lead to the dis-
sociation of Keap1 from Nrf2, and so nally induce the transcrip-
tional activation of Nrf2.
It is reported that CYP2E1, CYP3A4 and CYP1A2 are the main
metabolic enzymes for converting APAP into NAPQI, which sub-
sequently depleted cellular GSH and so caused oxidative stress-
induced injury [42,43]. In this study, we further observed if CA-
induced protection against APAP-induced hepatotoxicity is due to
inhibition on CYP2E1, CYP3A4, and CYP1A2. The results showed
that CA displayed weak inhibition on the enzymatic activity of
CYP2E1 and CYP3A4 (both less than 20% inhibition of the positive
inhibitor) in vitro (Fig. 7A and B). However, CA had no inhibition on
the enzymatic activity of CYP1A2 in vitro (Fig. 7C). These above
results indicate that CA-induced inhibition on CYP2E1 and CYP3A4
may weakly contribute to the protection of CA against APAP-in-
duced hepatotoxicity, but it is not a major determinant. Further
results showed that CA also reversed NAPQI-induced hepatotoxi-
city in hepatocytes (Fig. 7D). The above results indicate that CA can
directly inhibit the hepatotoxicity induced by APAP's toxic meta-
bolite NAPQI, and CA-induced the weak inhibition on CYP2E1 and
CYP3A4 is not required for protection.
In summary, the present study demonstrates that CA induces
Nrf2 activation by decreasing the expression of its inhibitor pro-
tein Keap1 and blocking the binding of Nrf2 with Keap1, and then
leads to the enhanced expression of downstream antioxidative
enzymes including NQO1 and HO-1, and thus prevents APAP-in-
duced liver oxidative injury.
Acknowledgments
This work was nancially supported by National Natural Sci-
ence Foundation of China (81322053 and 81573679), Shu Guang
project supported by Shanghai Municipal Education Commission
and Shanghai Education Development Foundation (13SG43), and
State major science and technology special projects during the
12th ve year plan (2015ZX09501004-002-002).
Appendix A. Supplementary material
Supplementary data associated with this article can be found in
theonlineversionathttp://dx.doi.org/10.1016/j.freeradbiomed.2015.
12.024.
References
[1] M.R. Chowdhury, M.A. Saqor, N. Tabassum, M.A. Potol, H. Hossain, M.A. Alam,
Supplementation of Citrus maxima peel powder prevented oxidative stress,
brosis, and hepatic damage in carbon tetrachloride (CCl4) treated rats, Evid.
Based Complement. Altern. Med. (2015) 598179.
[2] T. Erk, J. Hauser, G. Williamson, M. Renouf, H. Steiling, F. Dionisi, E. Richling,
Structure- and doseabsorption relationships of coffee polyphenols, Biofactors
40 (1) (2014) 103112 .
[3] G. Haghi, A. Hatami, M. Mehran, H. Hosseini, Caffeic acid derivatives from
Bupleurum chinense, Res. Pharm. Sci. 9 (5) (2014) 323330.
[4] Y.D. Jeon, J.Y. Kee, D.S. Kin, Y.H. Han, S.H. Kim, S.J. Kim, Y.J. Um, S.H. Hong,
Effects of Ixeris dentate water extract and caffeic acid on allergic inammation
in vivo and in vitro, BMC Complement. Altern. Med. 15 (2015) 196.
[5] S. Lima Mdos, da Conceicao Prudencio Dutra M., I.M. Toaldo, L.C. Correa, G.
E. Pereira, D. de Oliveira, M.T. Bordiqnon-Luiz, J.L. Ninow, Phenolic com-
pounds, organic acids and antioxidant activity of grape juices produced in
industrial scale by different processes of maceration, Food Chem. 188 (2015)
384392.
[6] J.Y. Han, J.Y. Fan, Y. Horie, S. Miura, D.H. Cui, H. Ishii, T. Hibi, H. Tsuneki,
I. Kimura, Ameliorating effects of compounds derived from Salvia miltiorrhiza
root extract on microcirculatory disturbance and target organ injury by
ischemia and reperfusion, Pharmacol. Ther. 117 (2008) 280295.
[7] P. Jiang, Y.C. Sheng, Y.H. Chen, L.L. Ji, Z.T. Wang, Protection of Flos Lonicerae
against acetaminophen-induced liver injury and its mechanism, Environ.
Toxicol. Pharmacol. 38 (3) (2014) 991999.
[8] M. Touaibia, J. Jean-Francois, J. Doiron, Caffeic acid, a versatile pharmacophore:
an overview, Mini Rev. Med. Chem. 11 (2011) 695713.
[9] A. Scalbert, C. Morand, C. Manach, C. Remesy, Absorption and metabolism of
polyphenols in the gut and impact on health, Biomed. Phamacother. 56 (6)
(2002) 276282.
[10] H.N. Mu, Q. Li, C.S. Pan, Y.Y. Liu, L. Yan, B.H. Hu, K. Sun, X. Chang, X.R. Zhao, J.
Y. Fan, J.Y. Han, Caffeic acid attenuates rat liver reperfusion injury through
sirtuin 3-dependent regulation of mitochondrial respiratory chain, Free Radic.
Biol. Med., 85, (2015) 237249.
[11] M.C. Martinez, S.G. Afonso, A.M. Buzaleh, A. Batlle, Protective action of anti-
oxidants on hepatic damage induced by griseofulvin, Sci. World J. (2014)
982385.
[12] L. Pari, A. Prasath, Efcacy of caffeic acid in preventing nickel induced oxida-
tive damage in liver of rats, Chem. Biol. Interact. 173 (2008) 7783.
[13] A. Gokcimen, A. Cim, H.T. Tola, D. Bayram, A. Kocak, F. Ozquner, A. Ayata,
Protective effect of N-acetylcysteine, caffeic acid and vitamin E on doxorubicin
hepatotoxicity, Hum. Exp. Toxicol. 26 (2007) 519525.
[14] K.H. Janbaz, S.A. Saeed, A.H. Gilani, Studies on the protective effects of caffeic
acid and quercetin on chemical-induced hepatotoxicity in rodents, Phytome-
dicine 11 (2004) 424430.
[15] M. Blieden, L.C. Paramore, D. Shah, R. Ben-Joseph, A perspective on the epi-
demiology of acetaminophen exposure and toxicity in the United States, Ex-
pert Rev. Clin. Pharmacol. 7 (2014) 341348.
[16] W. Bernal, Changing patterns of causation and the use of transplantation in
the United Kingdom, Semin. Liver Dis. 23 (2003) 227237.
[17] H. Jaeschke, M.R. McGill, A. Ramachandran, Oxidant stress, mitochondria, and
cell death mechanisms in drug-induced liver injury: lessons learned from
acetaminophen hepatotoxicity, Drug Metab. Rev. 44 (2012) 88106 .
[18] H. Jaeschke, T.R. Knight, M.L. Bajt, The role of oxidant stress and reactive ni-
trogen species in acetaminophen hepatotoxicity, Toxicol. Lett. 144 (2003)
279288.
[19] K. Heard, J. Green, Acetylcysteine therapy for acetaminophen poisoning, Curr.
Pharm. Biotechnol. 13 (2012) 19171923.
[20] W. Klein-Schwartz, S. Doyon, Intravenous acetylcysteine for the treatment of
acetaminophen overdose, Expert Opin. Pharmacother. 12 (2011) 119130.
[21] A.M. Bataille, J.E. Manautou, Nrf2: a potential target for new therapeutics in
liver disease, Clin. Pharmacol. Ther. 92 (2012) 340348.
[22] J.W. Kaspar, S.K. Niture, A.K. Jaiswal, Nrf2:INrf2 (Keap1) signaling in oxidative
stress, Free Radic. Biol. Med. 47 (2009) 13041309.
[23] Q.N. Liang, Y.C. Sheng, P. Jiang, L.L. Ji, Y.Y. Xia, Y. Min, Z.T. Wang, The difference
of glutathione antioxidant system in newly weaned and young mice liver and
its involvement in isoline-induced hepatotoxicity, Arch. Toxicol. 85 (2011)
12671279.
[24] H.J. Yeh, T.H. Chu, T.W. Shen, Ultrastructure of continuously cultured adult
human liver cell, Acta Biol. Exp. Sin. 13 (1980) 361364.
[25] L.L. Ji, T.Y. Liu, Y. Chen, Z.T. Wang, Protective mechanisms of N-Acetyl-Cysteine
against pyrrolizidine alkaloid clivorine-induced hepatotoxicity, J. Cell. Bio-
chem. 108 (2009) 424432.
[26] L.L. Ji, Y.C. Sheng, Z.Y. Zheng, L. Shi, Z.T. Wang, The involvement of p62-Keap1-
Nrf2 antioxidant signaling pathway and JNK in the protection of natural a-
vonoid quercetin against hepatotoxicity, Free Radic. Biol. Med. 85 (2015)
1223.
[27] F. Xiao, Y. Li, L. Luo, Y. Xie, M. Zeng, A. Wang, H. Chen, C. Zhong, Role of mi-
tochondrial electron transport chain dysfunction in Cr (VI)-induced cytotoxi-
city in L-02 hepatocytes, Cell. Physiol. Biochem. 33 (2014) 10131025.
[28] G. Hu, H. Huang, L. Yang, C. Zhong, B. Xia, Y. Yang, J. Liu, D. Wu, Q. Liu,
Z. Zhuang, Down-regulation of Pol
η
expression leads to increased DNA da-
mage, apoptosis and enhanced S phase arrest in L-02 cells exposed to hy-
droquinone, Toxicol. Lett. 214 (2012) 209217.
C. Pang et al. / Free Radical Biology and Medicine 91 (2016) 236246 245
[29] H. Tapiero, K.D. Tew, G.N. Ba, G. Mathe, Polyphenols: do they play a role in the
prevention of human pathologies? Biomed. Pharmacother. 56 (2002)
200207.
[30] S.K. Ramaiah, H. Jaeschke, Role of neutrophils in the pathogenesis of acute
inammatory liver injury, Toxicol. Pathol. 35 (2007) 757766.
[31] L.P. James, P.R. Mayeux, J.A. Hinson, Acetaminophen-induced hepatotoxicity,
Drug Metab. Dispos. 31 (2003) 14991506.
[32] S. Gum, M.K. Cho, Recent updates on acetaminophen hepatotoxicity: the role
of Nrf2 in hepatoprotection, Toxicol. Res. 29 (2013) 165172 .
[33] A. Enomoto, K. Itoh, E. Nagayoshi, J. Haruta, T. Kimura, T. O'Connor, T. Harada,
M. Yamamoto, High sensitivity of Nrf2 knockout mice to acetaminophen he-
patotoxicity associated with decreased expression of ARE-regulated drug
metabolizing enzymes and antioxidant genes, Toxicol. Sci. 59 (2001) 169177.
[34] S.A. Reisman, L.M. Aleksunes, C.D. Klaassen, Oleanolic acid activates Nrf2 and
protects from acetaminophen hepatotoxicity vai Nrf2-dependent and Nrf2-
independent processes, Biochem. Pharmacol. 77 (2009) 12731282.
[35] H.Y. Kay, Y.W. Kim, Ryu da H., S.H. Sung, S.J. Hwang, S.G. Kim, Nrf2-mediated
liver protection by sauchinone, an antioxidant lignan, from acetaminophen
toxicity through the PKC
δ
-GSK3
β
pathway, Br. J. Pharmacol. 163 (2011)
16531665 .
[36] S.I. Gum, M.K. Cho, The amelioration of N-acetyl-p-benzoquinone imine
toxicity by ginsenoside Rg3: the role of Nrf2-mediated detoxication and
Mrp1/Mrp3 transports, Oxid. Med. Cell. Longev. (2013) 957947.
[37] M. Lin, X. Zhai, G. Wang, X. Tian, D. Gao, L. Shi, H. Wu, Q. Fan, J. Peng, K. Liu,
J. Yao, Salvianolic acid B protects against acetaminophen hepatotoxicity by
inducing Nrf2 and phase II detoxication gene expression via activation of the
PI3K and PKC signaling pathways, J. Pharmacol. Sci. 127 (2015) 203210.
[38] I.M. Copple, C.E. Goldring, R.E. Jenkins, A.J.L. Chia, L.E. Randle, J.D. Hayes, N.
R. Kitteringham, B.K. Park, The hepatotoxic metabolite of acetaminophen di-
rectly activates the Keap1-Nrf2 cell defense system, Hepatology 48 (20 08)
12921301.
[39] A.K. Jaiswal, Nrf2 signaling in coordinated activation of antioxidant gene ex-
pression, Free. Radic. Biol. Med. 36 (2004) 11991207.
[40] O.W. Grifth, Mechanism of actin, metabolism and toxicity of buthionine
sulfoximine and its higher homologs, potent inhibitors of glutathione synth-
esis, J. Biol. Chem. 257 (1982) 1370413712.
[41] O.W. Grifth, Biologic and pharmacologic regulation of mammalian glu-
tathione synthesis, Free. Radic. Biol. Med. 27 (1999) 922935.
[42] J.E. Laine, S. Auriola, M. Pasanen, R.O. Juvonen, Acetaminophen bioactivation
by human cytochrome P450 enzymes and animal microsomes, Xenobiotica 39
(2009) 1121.
[43] J.L. Raucy, J.M. Lasker, C.S. Lieber, M. Black, Acetaminophen activation by
human liver cytochromes P450IIE1 and P450IA2, Arch. Biochem. Biophys. 271
(1989) 270283.
C. Pang et al. / Free Radical Biology and Medicine 91 (2016) 236246246
... Initiating this process ultimately promotes the expression of various downstream detoxification proteins, which helps protect against APAP-induced liver injuries. The detoxification proteins regulated by the activated NRF2 pathway include GPx, GCL , GSTs, HO-1, NQO1, and UDP-glucuronosyltransferases (UGTs) (Guo et al., 2016;Pang et al., 2016;Yuan-jing et al., 2016;Li et al., 2018a;b, 2020Lin et al., 2018;Ning et al., 2018;Dkhil et al., 2019;Jiang et al., 2019;Luo et al., 2019;Ruan et al., 2019;Wu et al., 2019;Choi et al., 2021;HU et al., 2021;Jayasuriya et al., 2021;Rahman et al., 2021;Zhong et al., 2021;Dayalan Naidu et al., 2022;Gao et al., 2022;He et al., 2022;Zhang et al., 2022;An et al., 2023;Somade et al., 2023;Zhou et al., 2023). Collectively, all this evidence highlights the critical role of KEAP1 activity in determining NRF2 activation. ...
... Also, the presence of epicatechins and procyanidin in both plants which were proven to have anti-hepatotoxic activity by reducing TNF-α levels (Azadnasab et al. 2021). In addition to the presence of organic acids like cinnamic acid (Amany et al. 2016), syringic acid and vanillic acid (Itoh et al. 2010), O-coumaric acid (Cha et al. 2018), ascorbic acid (Cetin Ozturk et al. 2009), caffeic acid (Pang et al. 2016), ferulic acid (Esmat et al. 2022), and sinapic acid (Shin et al. 2013) in both plants which are all reported to have hepatoprotective activity through various mechanisms. ...
Article
Lampranthus glaucus and Lampranthus glaucoides are only reported to have significant cytotoxic activity against certain cancer cell lines with phytochemical investigation of their petroleum ether and the ethyl acetate extracts. Further investigation was suggested concerning their hepatoprotective activity and relating it to the metabolic profile of their defatted methanol extracts using LC-ESI/ MS analysis. Hepatoprotective activity was evaluated through assessment of three liver parameters as well as liver histopatholog-ical examination in thioacetamide-induced hepatotoxicity model. Sixty-eight and 26 phytochemicals were tentatively identified in L. glaucoides and L. glaucus, respectively, with phenolic compounds as the major class. Both plants showed significant inhibition of serum GPT and GOT levels, inhibition of tissue IL-1β and TNF-α levels and inhibition of tissue NF-κβ and caspase-3 gene expression proving hepatoprotective action. Liver treated with L. glaucoides showed lesion scoring range between negative to mild, whereas L. glaucus showed a range between mild to moderate.
... Plant polyphenols are structurally similar to PDA in that both are enriched with phenolic hydroxyl groups in their molecular structure . A metallicpolyphenol coating can be formed on the surface of a material by simply immersing the substance to be modi ed into a plant polyphenol bath together with metal ions as cross-linking agents (Pang et al. 2016). Compared to PDA, the plant polyphenol coating has the advantages of non-neurotoxicity, non-cytotoxicity, low cost, and good availability. ...
Preprint
Full-text available
Vat dyeing is the mainstream process of cotton fabric dyeing at present, but the high toxicity of the reducing agent and the high cost of dyeing waste liquid treatment make the process no longer meet the requirements of green and sustainable development of society. The photonic crystal structure color, which is made of nanoscale colloidal microspheres arranged in an orderly manner as structural units, is expected to be a new printing and dyeing technology, that provides a new way of thinking for dyeing cotton fabrics. However, the practical applications of photonic crystal structural colors on textile fabrics are hampered by their inadequate stability, muted color saturation, and weak mechanical properties. In this study, P(St-BA-MAA) nanospheres with a hard-core-soft-shell structure were prepared by soapless emulsion polymerization, and then P(St-BA-MAA)@PGA-Fe ³⁺ photonic composite inks were prepared by forming a dark-colored coating film on the surface of the microspheres through the complexation of gallic acid with Fe ³⁺ . It was found that the performance of the prepared P(St-BA-MAA)@PGA-Fe ³⁺ photonic composite ink was stable, and the dark metallic-polyphenol network film enabled the formation of vibrant non-iridescent structured colors on cotton fabrics, the soft shell of the seeded microspheres also significantly improved the adhesion between the ink and the fabrics, and the structural color fastness was greatly improved compared with the PS microspheres. The preparation of this photonic composite ink is facile, low-cost, and produces a bright color, providing practical value for the development of textile products that construct structural color effects on textile substrates.
... Nrf2 not only regulates liver detoxification, it regulates the expression of the oxidative stress kinase, thereby playing a pivotal function in oxidative stress in the liver. For liver illnesses caused by APAP, such as ALI, the Keap1-Nrf2 pathway has been identified as a possible therapeutic target [40]. Under nonstressed conditions, Keap1 and Nrf2 are tightly bound in the cytoplasm; however, when triggered by ROS or electrophiles, Nrf2 separates from Keap1 and translocates to the nucleus after gathering in the cytoplasm, where it recognizes and reacts to the antioxidant response element (ARE) to activate the antioxidant enzyme (NQO1) downstream [41][42][43]. ...
Article
Full-text available
Acetaminophen (APAP) abuse is a common public health problem which can cause severe liver damage. However, strategies for dealing with this situation safely and effectively are very limited. The goal of the current work was to evaluate the protection and potential molecular mechanisms of an ethanol extract from shoots of the wild vegetable shutou (Crateva unilocularis Buch.) (ECS) against APAP-induced liver damage in mice. Mice orally received ECS for seven days (300 or 600 mg/kg b.w. per day) before being intraperitoneally injected with APAP (250 mg/kg). Results exhibited that ECS obviously decreased the content of alkaline phosphatase, alanine aminotransferase, aspartate transaminase, and malondialdehyde (p < 0.05). Catalase and superoxide dismutase were notably restored (p < 0.05), and the content of reduced glutathione was obviously increased (p < 0.05). Moreover, ECS significantly inhibited the secretion of interleukin-1β and tumor necrosis factor-α (p < 0.05). Further analyses of the mechanisms showed that ECS may alleviate oxidative stress in the liver by increasing the expression of the nuclear factor erythroid-2-related factor 2 and NADH quinone oxidoreductase 1 proteins, and may suppress liver inflammation by inhibiting the expression of the phosphorylated-inhibitor kappa B alpha/inhibitor kappa B alpha, phosphorylated-nuclear factor κB/nuclear factor κB, and cyclooxygenase-2 proteins. Meanwhile, ECS inhibited hepatocyte apoptosis by enhancing B-cell lymphoma gene 2 and suppressing Bcl-2-associated X protein. In summary, ECS may be used as a dietary supplement to prevent the liver damage caused by APAP abuse.
... For instance, caffeic acid, a phenolic compound, can be used as an electrophile in addition to its nucleophilic nature, which can provide Nrf2 transcription [40]. In the study by Pang et al., it was demonstrated that caffeic acid increased the Nrf2 gene expressions, thereby activating Nrf2 and leading to increased expression of antioxidant signals and reduced cellular ROS levels including HO-1 and NQO1 to prevent acetaminophen-induced acute liver failure in human healthy liver L-02 cells and HepG2 cells [41]. Also, FA is a rich monomer ingredient with a methoxy group on the benzene ring, which makes it a strong ROS scavenger [7]. ...
Article
Full-text available
Liver as iron storage organ is particularly susceptible to oxidative stress-induced injury from excess iron. Thus, antioxidant therapies are often used to reverse oxidative damage and protect cells and tissues. This study investigated the protective effects of phenolic acids; ferulic acid (FA) and its metabolite, ferulic acid 4-O-sulfate disodium salt (FAS) against oxidative stress under iron overload conditions in mouse and HepG2 cells. Cells were exposed to FA or FAS and then treated with iron-induced oxidative stress complex of 50 μmol/L FAC and 20 μmol/L of 8-hydroxyquinoline 8HQ (8HQ-FAC). Iron dextran was injected intraperitoneally on alternate days for 10 days to induce the iron overload condition in BALB/c mice. The study revealed that the phenolic acids were protective against ROS production, lipid peroxidation and antioxidant depletion in HepG2 cells and liver tissues of BALB/c mice during iron-induced oxidative stress. The protective function of phenolic acids was achieved by the transcriptional activation of nuclear factor erythroid-2-related factor 2 (Nrf2) to regulate antioxidant genes. In conclusion, the study provides evidence that FA has the potential as a therapeutic agent against iron-related diseases such as T2D.
Article
Excessive acetaminophen (APAP) application is a major cause of drug-induced liver injury (DILI). Febuxostat (Feb), a drug for reducing uric acid (UA) levels, was demonstrated to relieve hepatic inflammation and reverse organ functions. However, the effect of Feb on APAP-induced DILI and its mechanisms have not been fully explored. In this study, Feb (10 mg/kg) was given to mice by gavage 1 h after APAP (300 mg/kg, i.g.) induction. Serum and liver samples were collected 12 or 3 h after APAP challenge. Feb treatment was found to remarkably improve APAP-induced DILI, as evidenced by reduced serum ALT, AST and UA levels, pathomorphology, inflammatory, and oxidative responses. Consistently, treatment with Feb also reduced the cell injury induced by APAP in LO2 cells. Mechanistically, Feb induced GPX4 expression, activated the Keap1/Nrf2 pathway, and inhibited the TLR4/NF-κB p65 pathway. Feb also inhibited glutathione (GSH) depletion and Jun N-terminal kinase (JNK) activation in the early injury phase. Notably, pretreatment with Feb for 3 days also revealed preventive effects against APAP-induced DILI in mice. Overall, our data revealed a potential health impact of Feb on APAP-mediated DILI in vivo and in vitro, suggesting that Feb might be a potential candidate for treating DILI.
Article
Full-text available
Citrus maxima peel is rich in natural phenolic compounds and has a long use in the traditional medicine. HPLC-DAD analysis on Citrus maxima peel powder exhibited the presence of various phenolic compounds such as caffeic acid and (−)-epicatechin. To determine the plausible hepatoprotective activity of Citrus maxima peel powder, we used carbon tetrachloride (CCl 4) treated rat model. Liver damage in rats was confirmed by measuring the AST, ALT, and ALP enzyme activities. In addition, lipid peroxidation products (MDA), nitric oxide, advanced protein oxidation products level (APOP), and catalase activities were also analyzed along with the histological profiling for the inflammatory cell infiltration, collagen, and iron deposition in liver. Dietary supplementation of Citrus maxima peel powder exhibited significant reduction of serum AST, ALT, and ALP activities in carbon tetrachloride treated rats. Moreover, Citrus maxima peel powder also showed a significant reduction of the oxidative stress markers (MDA, NO, and APOP level) and restored the catalase activity in CCl 4 treated rats. Histological examination of the liver section revealed reduced inflammatory cells infiltration, collagen, and iron deposition in CCl 4 treated rats. The results from this study demonstrated that Citrus maxima peel powder produced significant hepatoprotective action in CCl 4 administered rats.
Article
Full-text available
Ixeris dentata Nakai has been used for the treatment of mithridatism, calculous, indigestion, pneumonia, hepatitis, and tumors in Korea, China, and Japan. However, the effect of a water extract of Ixeris dentata (ID) and its molecular mechanism on allergic inflammation has not been elucidated. In this study, we attempted to evaluate the effects of ID and its major compound caffeic acid on allergic inflammation in vivo and in vitro. ID was applied to 2, 4-dinitrofluorobenzene (DNFB)-induced atopic dermatitis (AD)-like skin lesion mice and immune cell infiltration, cytokine production, and the activation of mitogen-activated protein kinases (MAPKs) were investigated. Moreover, the effect of ID on compound 48/80-induced anaphylactic shock was investigated in a mouse model. The human keratinocyte cell line (HaCaT cells) and human mast cells (HMC-1) were treated with ID or caffeic acid to investigate the effects on the production of chemokines and proinflammatory cytokines and on the activation of MAPKs. ID inhibited the serum levels of IgE and interleukin (IL)-1β in DNFB-induced AD-like skin lesion mouse models and suppressed anaphylactic shock in the mouse models. ID and caffeic acid inhibited the production of chemokines and adhesion molecules in HaCaT cells. In addition, ID reduced the release of tumor necrosis factor-α and IL-8 via the inhibition of MAPKs phosphorylation in HMC-1 cells. These results suggest that ID is a potential therapeutic agent for allergic inflammatory diseases, including dermatitis.
Article
Full-text available
Acetaminophen (APAP) is used drugs worldwide for treating pain and fever. However, APAP overdose is the principal cause of acute liver failure in Western countries. Salvianolic acid B (SalB), a major water-soluble compound extracted from Radix Salvia miltiorrhiza, has well-known antioxidant and anti-inflammatory actions. We aimed to evaluate the ability of SalB to protect against APAP-induced acute hepatotoxicity by inducing nuclear factor-erythroid-2-related factor 2 (Nrf2) expression. SalB pretreatment ameliorated acute liver injury caused by APAP, as indicated by blood aspartate transaminase levels and histological findings. Moreover, SalB pretreatment increased the expression of Nrf2, Heme oxygenase-1 (HO-1) and glutamate-l-cysteine ligase catalytic subunit (GCLC). Furthermore, the HO-1 inhibitor zinc protoporphyrin and the GCLC inhibitor buthionine sulfoximine reversed the protective effect of SalB. Additionally, siRNA-mediated depletion of Nrf2 reduced the induction of HO-1 and GCLC by SalB, and SalB pretreatment activated the phosphatidylinositol-3-kinase (PI3K) and protein kinase C (PKC) signaling pathways. Both inhibitors (PI3K and PKC) blocked the protective effect of SalB against APAP-induced cell death, abolishing the SalB-induced Nrf2 activation and decreasing HO-1 and GCLC expression. These results indicated that SalB induces Nrf2, HO-1 and GCLC expression via activation of the PI3K and PKC pathways, thereby protecting against APAP-induced liver injury. Copyright © 2015 Japanese Pharmacological Society. Production and hosting by Elsevier B.V. All rights reserved.
Article
Full-text available
In this study, caffeic acid (CA) and its three derivatives including 3-caffeoylquinic acid (3-CQA, neochlorogenic acid), 4-caffeoylquinic acid (4-CQA, cryptochlorogenic acid), and 5-caffeoylquinic acid (5-CQA, chlorogenic acid) were identified in Bupleurum chinense aerial parts using reverse-phase high-performance liquid chromatography (RP-HPLC) with photodiode array (PDA) detector, reference compounds and chemical reactions. Separation was performed on a C18 column using gradient elution with 4% (v/v) aqueous acetic acid and acetonitrile as mobile phase at ambient temperature. In addition, the flavonoid aglycones were characterized and quantified after acid hydrolysis of the plant material. The flavonols profile showed quercetin (0.36 g per 100 g), kaempferol (1.11 g per 100 g) and isorhamnetin (0.16 g per 100 g). Total phenolic and total flavonoid contents ranged from 7.3 to 18.7% and 0.58 to 2.72% in dry plant material, respectively.
Article
Full-text available
Backgroud: Hexavalent chromium [Cr(VI)] and its compounds (e.g. chromates), which are extensively used in numerous industrial processes including leather tanning and steel manufacturing, are considered as priority pollutants. There is growing evidence supporting that Cr(VI) could be a human carcinogen that induces primary liver cancer after oral exposure, and this sheds light on the importance of the investigation of Cr(VI)-induced hepatotoxicity. Although it is known that mitochondria are major targets for heavy metals, the mechanisms of electron transfer chain (ETC) dysfunction involved in Cr(VI)-induced cytotoxicity are unclear. Methods: In the present study, by using mitochondrial respiratory chain complex (MRCC) I inhibitor rotenone (ROT) and its substrates glutamate/malate (Glu/Mal), MRCC III inhibitor antimycin A (AA) and its substrate coenzyme Q (CoQ), and the antioxidant Vitamin C (Vit C), we aimed to elucidate the role of mitochondrial ETC dysfunction in Cr(VI)-induced cytotoxicity. Results: We found that Cr(VI) targeted and inhibited MRCC I and III to induce ETC dysfunction, which played an important role in Cr(VI)-induced cytotoxicity. Conclusion: Our current data provides novel important insight into the mechanisms of mitochondrial ETC dysfunction in Cr(VI)-induced cytotoxicity in the hepatocytes, and we will be poised to develop new methods in the prevention and treatment of liver diseases involving mitochondrial ETC dysfunction for the occupational exposure population. Copyright (C) 2014 S. Karger AG, Basel
Article
Sirtuin 3 (Sirt3) plays critical roles in regulating mitochondrial oxidative metabolism. However, whether Sirt3 is involved in liver ischemia and reperfusion (I/R) injury remain elusive. Caffeic acid (CA) is a natural antioxidant derived from Salvia miltiorrhiza. Whether CA protects liver I/R injury through regulating Sirt3 and mitochondrial respiratory chain (MRC) is unclear. This study investigated the effect of CA on liver I/R injury, microcirculatory disturbance and potential mechanism, particularly focusing on Sirt3 dependent MRC. The liver I/R of male Sprague-Dawley rats was established by occlusion of portal area vessels for 30min followed by 120min reperfusion. CA (15mg/kg/h) was continuously infused via femoral vein staring from 30min before ischemia. After I/R, Sirt3 expression and MRC activity decreased, acetylation of NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 9 and succinate dehydrogenase complex, subunit A, flavoprotein variant were provoked, and the liver microcirculatory disturbance and injury were observed. Treatment with CA attenuated liver injury, inhibited Sirt3 down expression, and upregulated MRC activity. CA attenuated rat liver microcirculatory disturbance and oxidative injury through regulation of Sirt3 and mitochondrial respiratory chain. Copyright © 2015. Published by Elsevier Inc.
Article
Quercetin, one of the most abundant dietary flavonoids, is reported to have protective function against various hepatotoxicants-induced hepatotoxicity. The present study aims to investigate the critical role of nuclear factor erythroid 2-related factor 2 (Nrf2) antioxidative signaling pathway in the protection of quercetin against hepatotoxicity. Quercetin prevented the cytotoxicity induced by a variety of hepatotoxicants including clivorine (Cliv), acetaminophen (APAP), ethanol, carbon tetrachloride (CCl4), and toosendanin (TSN) in human normal liver L-02 cells. Quercetin induced the nuclear translocation of Nrf2, along with the increased expression of the antioxidant responsive element (ARE)-dependent genes like catalytic or modify subunit of glutamate-cysteine ligase (GCLC/GCLM), and heme oxygenase-1 (HO-1). In addition, HO-1 inhibitor zinc protoporphyrin (ZnPP) and GCL inhibitor L-Buthionine-(S, R)-sulfoximine (BSO) both reduced the hepato-protection induced by quercetin. Quercetin had no effect on kelch-like ECH-associated protein-1(Keap1) expression, but molecular docking results indicated the potential interacting of quercetin with Nrf2 binding site in Keap1 protein. Quercetin increased the expression of p62, and p62 siRNA decreased quercetin-induced hepato-protection. Quercetin induced the activation of c-Jun N-terminal kinase (JNK) in hepatocytes. JNK inhibitor SP600125 and JNK siRNA both reduced quercetin-induced hepato-protection. SP600125 and JNK siRNA decreased the increased p62 expression induced by quercetin. In addition, SP600125 also decreased the increased mRNA and protein expression of GCLC, GCLM and HO-1 induced by quercetin. Taken together, our present study demonstrates that quercetin prevents hepatotoxicity by inducing p62 expression, inhibiting the binding of Keap1 to Nrf2, and thus leading to the increased expression of antioxidative genes dependent on Nrf2. Meanwhile, our study indicates that JNK plays some regulation in this process.
Article
This study aims to observe the protective action of Flos Lonicerae (FL) aqueous extract against acetaminophen (AP)-induced liver injury and its mechanism. Results show that FL decreases AP-increased serum alanine/aspartate transaminases (ALT/AST) activity, as well as total bilirubin (TB) amount, in mice. Histological evaluation of the liver further confirms the protection of FL against AP-induced hepatotoxicity. TdT-mediated biotin-dUTP nick-end labeling (TUNEL) assay shows that FL reduces AP-increased apoptotic cells. Furthermore, AP-decreased liver glutamate-cysteine ligase (GCL) enzymatic activity and glutathione (GSH) amount are both reversed by FL because of the increased expression of the catalytic subunit of GCL (GCLC) protein. The amount of chlorogenic acid (CGA), caffeic acid, and luteolin, the main active compounds in FL, is detected by high-performance liquid chromatography (HPLC). In addition, cell viability assay demonstrates that polyphenols in FL, such as CGA, caffeic acid, as well as isochlorogenic acids A, B, and C, can reverse AP-induced cytotoxicity. In conclusion, FL can prevent AP-induced liver injury by inhibiting apoptosis. The cellular antioxidant enzyme GCL is also involved in such protection. Polyphenols may be the main active hepato-protective ingredients in FL.