Chlorogenic Acid Reduces Liver in Ammation and Fibrosis Through Inhibition of Toll-Like Receptor 4 Signaling Pathway

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Toxicology 303 (2013) 107–114

Contents lists available at SciVerse ScienceDirect

Toxicology
journal homepage: www.elsevier.com/locate/toxicol

Chlorogenic acid reduces liver inflammation and fibrosis through inhibition of


toll-like receptor 4 signaling pathway
Haitao Shi, Lei Dong ∗ , Jiong Jiang, Juhui Zhao, Gang Zhao, Xiaoyan Dang, Xiaolan Lu, Miao Jia
Department of Gastroenterology, Second Affiliated Hospital of Xi’an Jiaotong University, No. 157 Xiwu Road, Xi’an, Shaanxi 710004, China

a r t i c l e i n f o a b s t r a c t

Article history: Chlorogenic acid (CGA) is a type of polyphenol with anti-inflammatory, antioxidant activities. Our previ-
Received 23 September 2012 ous studies showed CGA could efficiently inhibit carbon tetrachloride (CCl4 )-induced liver fibrosis in rats.
Received in revised form 28 October 2012 However, the specific underlying mechanism remains unclear. The aim of this study is to investigate the
Accepted 30 October 2012
effects of CGA on liver inflammation and fibrosis induced by CCl4 and whether they are related to inhibi-
Available online 9 November 2012
tion of toll-like receptor 4 (TLR4) signaling pathway. Male Sprague-Dawley (SD) rats were administrated
CCl4 together with or without CGA for 8 weeks. Histopathological and biochemical analyses were carried
Keywords:
out. The mRNA and protein expression levels of proinflammatory and profibrotic mediators were detected
Chlorogenic acid
Liver inflammation
by RT-PCR and Western blot, respectively. The levels of serum proinflammatory cytokines were detected
Liver fibrosis by ELISA. CGA significantly attenuated CCl4 -induced liver damage and symptoms of liver fibrosis, accom-
TLR4 panied by reduced serum transaminase levels, collagen I and ␣-smooth muscle actin (␣-SMA) expression.
As compared with the CCl4 -treated group, the expression levels of TLR4, myeloid differentiation factor
88 (MyD88), inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) were reduced in the
treatment group of CCl4 and CGA, whereas bone morphogenetic protein and activin membrane-bound
inhibitor (Bambi) expression was increased. CGA also suppressed CCl4 induced nuclear factor-␬B (NF-
␬B) activation. Moreover, the hepatic mRNA expression and serum levels of tumor necrosis factor-␣
(TNF-␣), interleukin-6 (IL-6) and interleukin-1␤ (IL-1␤) were significantly increased in CCl4 -treated rats
and attenuated by co-treatment with CGA. Our data indicate that CGA can efficiently inhibit CCl4 -induced
liver fibrosis in rats and the protective effect may be due to the inhibition of TLR4/MyD88/NF-␬B signaling
pathway.
© 2012 Elsevier Ireland Ltd. All rights reserved.

Liver fibrosis is a wound-healing process in injured liver factors which play important roles in liver inflammation, acceler-
caused by various agents, including infection with hepatitis ating the liver fibrosis (Pinzani and Macias-Barragan, 2010).
viruses, chronic alcohol abuse, metabolic or autoimmune diseases Lipopolysaccharides (LPS) is the major constituent of outer
(Friedman, 2003; Kawada, 2011). Upon liver injury, quiescent membrane in gram-negative bacteria. It activates a variety of
hepatic stellate cells (HSCs) are activated and produce excessive the mammalian cell types including monocytes/macrophages and
extracellular matrix (Bataller and Brenner, 2005; Friedman, 2003, endothelial cells through the toll like receptor 4 (TLR4) (Zhu and
2010). In chronic liver injury, the injured or damaged cells release Mohan, 2010). Previous study shows serum LPS levels are signifi-
a number of cytokines, which then stimulate the Kupffer cells cantly elevated in patients with chronic liver disease owing to the
to release more inflammatory mediators (Luckey and Petersen, increased intestinal mucosal permeability and bacterial transloca-
2001). Kupffer cells and activated HSCs have been implicated tion (Choi et al., 2011; Lin et al., 1995; Parlesak et al., 2000). Many
to secrete proinflammatory cytokines, chemokines and adhesion liver cells including Kupffer cells, HSCs and liver endothelial cells
(LECs) can express TLR4 and respond to LPS, which has an impor-
tant role in liver fibrosis (Aoyama et al., 2010). It is known that LPS
stimulates Kupffer cells to release a number of cytokines to activate
Abbreviations: CGA, chlorogenic acid; HSCs, hepatic stellate cells; LPS, HSCs and TLR4 signaling in LECs regulates angiogenesis, which are
lipopolysaccharides; TLR4, toll-like receptor 4; CCl4 , carbon tetrachloride; ␣-SMA, linked to the development of liver fibrosis (Seki and Brenner, 2008).
␣-smooth muscle actin; MyD88, myeloid differentiation factor 88; iNOS, inducible LPS sensitizes quiescent HSCs to TGF-␤ and Kupffer cell–mediated
nitric oxide synthase; COX-2, cyclooxygenase-2; Bambi, bone morphogenetic pro- activation (Seki et al., 2007), while for activated HSCs, it can pro-
tein and activin membrane-bound inhibitor; NF-␬B, nuclear factor-␬B; TNF-␣,
tumor necrosis factor-␣; IL-6, interleukin-6; IL-1␤, interleukin-1␤.
mote them to develop an inflammatory phenotype and make a
∗ Corresponding author. Tel.: +86 29 87679272; fax: +86 29 87678758. contribution to the inflammatory network in the liver during endo-
E-mail addresses: shihaitao7@163.com (H. Shi), Dong556@126.com (L. Dong). toxemia (Brun et al., 2005; Paik et al., 2003; Thirunavukkarasu et al.,

0300-483X/$ – see front matter © 2012 Elsevier Ireland Ltd. All rights reserved.
http://dx.doi.org/10.1016/j.tox.2012.10.025
108 H. Shi et al. / Toxicology 303 (2013) 107–114

2006). Besides Kupffer cell, HSC is another target for LPS-induced with each other so as to divide parenchyma into separate fragments; and grade 4,
liver injury and provide a direct link between inflammatory and the same as grade 3, except for the presence of thick septa (complete cirrhosis). To
avoid sampling errors, biopsies were obtained from equivalent lobes and the semi-
fibrotic liver injury (Seki et al., 2007).
quantitative grades were performed by independent histopathologists who were
Some epidemiological researches suggest that coffee consump- blinded to the treatment assignment. Each sample was observed at 100× magnifi-
tion may reduce the risk of liver fibrosis (Ruhl and Everhart, 2005; cation. The degree of liver damage was expressed as the mean of ten fields of view
Tverdal and Skurtveit, 2003). Our study, as well as one from others, on each slide.
shows that coffee can improve carbon tetrachloride (CCl4 )-induced
liver fibrosis in rats (Shi et al., 2010; Shin et al., 2010). However, 1.5. Reverse transcription-polymerase chain reaction (RT-PCR)

it is not clear which components of coffee are responsible for the Total RNA was extracted from liver tissue samples by the TRIzol kit (Invitrogen)
protective effect. Other than caffeine, kahweol and cafestol, cof- according to the manufacturer’s protocol. The RNA was then subjected to reverse
fee drink is also rich in chlorogenic acid (CGA). Caffeine, kahweol transcription using a RevertAidTM First Strand cDNA Synthesis Kit (Fermentas).
and cafestol have been shown to have hepatoprotective and anti- The mRNA expression levels of TLR4, myeloid differentiation factor 88 (MyD88),
inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), tumor necrosis
oxidant effects (Gressner et al., 2008; Lee et al., 2007). CGA is a
factor-␣ (TNF-␣), interleukin-6 (IL-6), interleukin-1␤ (IL-1␤), bone morphogenetic
type of polyphenol with anti-inflammatory, antioxidant, antiobe- protein and activin membrane-bound inhibitor (Bambi), ␣-smooth muscle actin (␣-
sity and antimicrobial activities (Feng et al., 2005; Kono et al., 1997; SMA), collagen I were measured by RT-PCR. The primer sequences used in PCR are
Xu et al., 2010; Zhang et al., 2010). A recent study showed that CGA shown in Table 1. The annealing temperatures and the thermal cycles for each tar-
can protect mice from LPS-induced hepatotoxicity (Xu et al., 2010). get gene were as follows: TLR4 (56 ◦ C, 30 cycles), MyD88 (54 ◦ C, 30cycles), iNOS
(56 ◦ C, 30 cycles), COX-2 (54 ◦ C, 30 cycles), TNF-␣ (56 ◦ C, 34 cycles), IL-6 (56 ◦ C, 30
Another study showed that CGA can inhibit LPS-induced inflamma- cycles), IL-1␤ (53 ◦ C, 30 cycles), Bambi (57 ◦ C, 30 cycles), ␣-SMA (53 ◦ C, 30 cycles)
tory cytokines release in RAW264.7 cells (Shan et al., 2009). And and collagen-I (53 ◦ C, 30 cycles), ␤-actin (56 ◦ C, 30 cycles). PCR products were run
our previous study showed that CGA can efficiently inhibit CCl4 - on a 2% agarose gel stained with ethidium bromide recorded on polaroid film, and
induced liver fibrosis in rats (Shi et al., 2009). However, the specific the bands quantified by densitometry. The expression levels of all the transcripts
were normalized to that of ␤-actin mRNA in the same tissue samples.
underlying mechanism remains unclear, but it may be related to its
anti-inflammatory activities. This study is to investigate the effects
1.6. Western blot
of CGA on liver inflammation and fibrosis induced by CCl4 and
whether they are related to inhibition of TLR4 signaling pathway. The cytosolic and nuclear protein extracts from rat liver were performed as
described previously (Choi et al., 2009). Samples of 50 ␮g of protein were mixed
1. Materials and methods with gelloading buffer, boiled for 5 min, and loaded on 8% or 10% polyacrylamide
gels. Electrophoresis was then carried out and the proteins transferred to nitrocellu-
1.1. Reagents lose membranes. Non-specific antibody binding was blocked by preincubation of the
membranes in 1× Tris-buffered saline (TBS) containing 5% skimmed milk for 2 h at
CGA was purchased from Sigma–Aldrich (St. Louis, USA). Anti-␣-SMA was from room temperature. The membranes were incubated overnight at 4 ◦ C with primary
Epitomic (Burlingame, CA, USA). Anti-TLR4, Bambi and Lamin B were from Pro- antibodies against rat TLR4 (1:500), MyD88 (1:500), NF-␬B (1:400), I␬B–␣ (1:500), p-
teinTech Group (Chicago, IL, USA). Anti-MyD88, I␬B-␣, p-I␬B-␣, and ␤-actin were I␬B–␣ (1:500), iNOS (1:500), COX-2 (1:500), Bambi (1:800), ␣-SMA (1:1000), Lamin
from Bioworld Technology (St. Louis Park, MN, USA). Anti-NF-␬B was from Abcam B (1:1000), and ␤-actin (1:1000) in 1× TBS containing 5% skimmed milk. After wash-
(Cambridge, UK). Anti-COX-2, iNOS and horseradish-peroxidase (HRP)-conjugated ing, they were incubated for 2 h at room temperature with anti-rabbit IgG at a 1:4000
secondary antibodies were purchased from Santa Cruz Biotechnology (Santa Cruz, dilution. Bands were visualized using a SuperSignal Substrate Chemiluminescence
CA, USA). The SuperSignal Substrate Chemiluminescence Kit was from Pierce (Rock- Kit.
ford, USA). All other chemicals used in the experiment were of analytical grade.
1.7. Determination of cytokine levels.
1.2. Animals and experimental treatments
Specific and sensitive enzymelinked immunoassays (ELISA) obtained from R&D
Thirty-two SD rats weighing (220–250 g) were housed with free access to food Systems (Minneapolis, MN, U.S.A.) were used to determine concentrations of TNF-␣,
and water throughout the experiments. The rats were maintained in a controlled IL-6, and IL-1␤ in serum.
environment at 21 ± 2 ◦ C and 50 ± 5% relative humidity under a 12-h dark/light cycle
and were acclimatized for at least 1 week prior to use. The rats were randomly
1.8. Statistical analysis
divided into four groups (8 rats per group): (1) control group: given the appropri-
ate vehicles (distilled water and olive oil); (2) CGA group: given CGA (60 mg/kg,
The experimental results were expressed as the mean ± SD. Statistical signifi-
dissolved in distilled water, intragastric administration once daily); (3) CCl4 group:
cance among the groups was analyzed by one-way analysis of variance with the
given CCl4 [3 mL/kg, dissolved in olive oil (40%, V/V), intraperitoneal injection twice
Student–Newman–Keuls test. Histopathological data from the liver were analyzed
weekly]; (4) CCl4 + CGA group: given CCl4 along with CGA (dosages and treatments
by the Kruskal–Wallis non-parametric test, followed by the Mann–Whitney U-test.
as mentioned for CGA group and CCl4 group, respectively). No rats died during the
P < 0.05 was considered significant.
experimental period. At the end of 8 weeks and 24 h after the last dose of CCl4 the
rats were anesthetized with chloral hydrate and blood was drawn from inferior caval
vein prior to the excision of organ tissues. Serum was collected by centrifugation at 2. Results
13,000 rpm for 15 min at 4 ◦ C and stored at −70 ◦ C for liver function test and ELISA
analysis. Liver was rapidly excised and stored in liquid nitrogen for later analysis.
The experiment was performed in accordance with the guidelines of the Animal 2.1. Effects of CGA on general features and liver function
Care Committee of Xi’an Jiaotong University.
There was no difference in body weight and liver index [Liver-
1.3. Liver function test weight (g)/body-weight (100 g)] between control group and CGA
Alanine transaminase (ALT), aspartate transaminase (AST) activities and albu-
control group. CCl4 -treated rats showed significantly decreased
min were measured using a biochemistry analyzer (Olympus AU2700, Japan). body weight (P < 0.01) and increased liver index (P < 0.01). How-
ever, CCl4 and CGA co-treatment have no effect on body weight
1.4. Histological examinations and liver index compared with CCl4 treatment alone. For assess-
ments of hepatocellular damage, the serum transaminase (AST
A portion of each liver was fixed in 10% formalin, processed by routine histo-
logical procedures, embedded in paraffin, and cut into 5-␮m sections. The samples and ALT) activities and albumin level were measured. There was
were stained with hematoxylin and eosin (H&E) for histopathological examination no difference in serum transaminase activities and albumin level
and with Masson’s trichrome for assessment of fibrosis. Sections were examined in between control group and CGA control group. After administra-
a blinded manner under a microscope (Leica TCS SP, German). Fibrosis was graded tion for 8 weeks, a dramatic increase in liver enzyme activities
according to the method of (Ruwart et al., 1989) as follows: grade 0, normal liver
and no fibrosis; grade 1, increase in collagen without septa formation; grade 2, for-
and decrease in albumin level were observed in CCl4 -treated rats
mation of incomplete septa from the portal tract to the central vein (septa that do compared with controls (P < 0.01). CCl4 and CGA co-treatment sig-
not interconnect with each other); grade 3, complete but thin septa interconnecting nificantly decreased ALT and AST activities (P < 0.01) and increased
H. Shi et al. / Toxicology 303 (2013) 107–114 109

Table 1
Primer sequences for RT-PCR.

Target genes Sequences PCR product (bp)

␤-actin Forward 5 - CTATCGGCAATGAGCGGTTCC -3 147


Reverse 5 -TGTGTTGGCATAGAGGTCTTTACG -3
␣-SMA Forward 5 -GGCTTCTCTATCTACCTTCC-3 167
Reverse 5 -ACATTCACAGTTGTGTGCTA-3
Collagen I Forward 5 -ACAGGCGAACAAGGTGACAGAG-3 159
Reverse 5 -GCCAGGAGAACCAGCAGAGC-3
TLR4 Forward 5 - GGCATCATCTTCATTGTCCTTG -3 111
Reverse 5 - AGCATTGTCCTCCCACTCG -3
MyD88 Forward 5 - TACATACGCAACCAGCAG -3 160
Reverse 5 - GGCAGTAGCAGATGAAGG -3
iNOS Forward 5 -CTCACTGTGGCTGTGGTCACCTA-3 101
Reverse 5 -GGGTCTTCGGGCTTCAGGTTA-3
COX-2 Forward 5 - ACCAGCAGTTCCAGTATCAG -3 158
Reverse 5 - AGCAAGTCCGTGTTCAAGG -3
TNF-␣ Forward 5 - CCACCACGCTCTTCTGTCTAC -3 148
Reverse 5 - GCTACGGGCTTGTCACTCG -3
IL-6 Forward 5 -CTTCCAGCCAGTTGCCTTCTTG-3 109
Reverse 5 -TGGTCTGTTGTGGGTGGTATCC-3
IL-1␤ Forward 5 -AATCTCACAGCAGCATCTC-3 189
Reverse 5 -AGCAGGTCGTCATCATCC-3
Bambi Forward 5 - CTCAGGACAAGGAAACAGATACC -3 173
Reverse 5 - TTCGCAGCATTCGCAAGG -3

Table 2
Body weight, liver weight and liver function tests in different treatment group.

Control CGA CCl4 CCl4 + CGA

Body weight (g) 418 ± 17.9 424 ± 21.9 367 ± 10.8* 375 ± 14.3
Liver weight (g) 14.0 ± 1.03 14.1 ± 1.15 13.9 ± 0.69 13.7 ± 0.47
Liver index 3.35 ± 0.26 3.31 ± 0.18 3.78 ± 0.21* 3.67 ± 0.18
ALT (U/L) 48.2 ± 6.0 47.6 ± 8.6 165.1 ± 21.8* 99.7 ± 23.4#
AST (U/L) 39.1 ± 4.1 37.5 ± 7.9 140.6 ± 22.1* 103.7 ± 15.3#
Albumin (g/L) 31.7 ± 2.1 32.2 ± 2.5 20.0 ± 3.4* 23.6 ± 2.8##

Results are mean ± SD (n = 8). Liver index: liver-weight (g)/body-weight (100 g).
*
P < 0.01 as compared with control group.
#
P < 0.01 as compared with CCl4 group.
##
P < 0.05 as compared with CCl4 group.

albumin level (P < 0.05), suggesting that CGA can ameliorate liver 2.3. Effects of CGA on the expression of ˛-SMA and collagen I
injury induced by CCl4 administration (Table 2).
In order to evaluate liver fibrosis at the molecular level, we
2.2. Effects of CGA on liver histopathologic changes detected the expression of ␣-smooth muscle actin (␣-SMA), a
marker of hepatic stellate cell activation, and collagen I, the
The histopathological changes in liver were examined with H&E major component of extracellular matrix in fibrotic liver. The
and Masson’s trichrome staining. Liver tissue samples from the con- expression of ␣-SMA and collagen I were significantly elevated in
trol and CGA control rats showed an integrity lobular structure with CCl4 -treated rats compared with the control group (P < 0.01), and
clear central veins and radiating hepatic cords, without necrosis, these inductions were significantly inhibited by CCl4 and CGA co-
inflammation or fibrosis development (Fig. 1A–D). In contrast, CCl4 - treatment (P < 0.01)(Fig. 2A and B), which suggested CGA could
treated rats showed severe changes in liver morphology, including inhibit the activation of quiescent HSCs and extracellular matrix
necrosis, obvious collagen deposition, formation of pseudo-lobules deposition.
and infiltration of inflammatory cells in liver interstitial (Fig. 1E
and F). However, treatment with CGA significantly alleviated the
2.4. Effects of CGA on the TLR4 signaling pathway
degree of liver fibrogenesis and formation of pseudo-lobulus, which
accompanied with little inflammatory cell infiltration (Fig. 1G and
It has been demonstrated earlier that TLR4 signaling pathway
H, Table 3).
plays an important role in the progression of liver inflammation
and fibrosis, so we detected the effect of CGA on the TLR4 signaling
Table 3 pathway in order to elucidate the precise molecular antiinflamma-
Effect of CGA on the pathologic grading of hepatic fibrosis rats induced by CCl4 .
tion and antifibrosis and mechanism of CGA. After CCl4 treatment,
Group Severity score of fibrosis Mean rank the mRNA and protein levels of TLR4, MyD88, iNOS, COX-2 were
0 1 2 3 4 significantly increased compared to the control group, while the
level of Bambi was decreased (Fig. 3A–D). The expression of pro-
Control 8 0 0 0 0 8.5
CGA 8 0 0 0 0 8.5
inflammatory cytokines in the liver including TNF-␣, IL-6 and IL-␤
CCl4 0 0 1 2 5 27.75* were also up-regulated in CCl4 -treated rats (Fig. 3E). Moreover, CCl4
CCl4 + CGA 0 2 4 2 0 21.25# treatment could reduced I␬B␣ protein level and increased p-I␬B␣
Each group consists of 8 rats and figures represent number of rat per grade. and the nuclear translocation of NF-␬B (Fig. 4A and B). All of these
*
P < 0.01 as compared with control group. results suggested TLR4/NF-␬B signaling pathway was activated in
#
P < 0.01 as compared with CCl4 group. the liver of CCl4 -treated rats. However, CGA treatment could inhibit
110 H. Shi et al. / Toxicology 303 (2013) 107–114

Fig. 1. Effect of CGA on the histological changes in the liver of the CCl4 -treated rats as shown by hematoxylin and eosin staining (histopathology; original magnification
100×) and Masson’s trichrome staining (assessment of fibrosis; original magnification 100×). (A–D) H&E staining; (A) control group:normal lobular architecture and cell
structure, (B) CGA control group: normal lobular architecture and cell structure, (C) CCl4 -treated group: extensive hepatocellular damage with the presence of inflammatory
cell infiltration, and centrizonal necrosis, (D) CCl4 and CGA co-treated group: mild inflammation and minimal hepatocellular necrosis. (E–H) Masson’s trichrome staining,
(E) control group: no significant collagen deposition, (F): no significant collagen deposition, (G) CCl4 group: substantial collagen deposition in the periportal and pericellular
areas and (H) CCl4 and CGA co-treated group: less collagen deposition was observed than that in rats treated with CCl4 alone.
H. Shi et al. / Toxicology 303 (2013) 107–114 111

Fig. 2. Effects of CGA on the expression of ␣-SMA and collagen I. (A) ␣-SMA and collagen I mRNA. (B) ␣-SMA protein. The expression levels of ␣-SMA and collagen I were
significantly higher in the CCl4 -treated group as compared to the control group and CGA-treated group (P < 0.01). ␤-actin served as the internal control. * P < 0.01 as compared
with the control group; # P < 0.01 as compared with the CCl4 group.

the activation of TLR4 signaling pathway, including the expres- Currently, we do not have strong evidence that endogenous
sion of TLR4, MyD88, iNOS and COX-2 decreased, Bambi increased, TLR4 ligands are involved in liver fibrosis. It was shown that
TNF-␣, IL-6 and IL-␤ down-regulated, I␬B␣ protein increased, the HMGB1 stimulates HSCs proliferation and expression of a-SMA
nuclear translocation of NF-␬B and p-I␬B␣ reduced. in vitro (Kao et al., 2008). However, the roles of exogenous ligand
LPS in chronic liver diseases have been study for many years. TLR4
2.5. Effects of CGA on the serum proinflammatory cytokines levels and its ligand LPS mediate their effects in liver fibrosis through
different mechanisms. In Kupffer cell, TLR4 activation produces
To determine whether CGA suppresses inflammation caused by pro-inflammatory cytokines (for example, TNF-␣, IL-1␤ and IL-6)
CCl4 , we examined the serum levels of TNF-␣, IL-6 and IL-1␤. TNF- and ROS, resulting in hepatocyte damage, leukocyte infiltration
␣, IL-6 and IL-1␤ levels in serum of CCl4 rats were significantly and secretion of pro-fibrogenic cytokines. In HSCs, there are at
higher than those in the control group. In contrast, CGA treatment least two roles of TLR4 signaling. First, TLR4-stimulated HSCs
attenuate these cytokines levels, suggesting that CGA ameliorated produce various chemokines and adhesion molecules (IL-6, IL-8,
the CCl4 -induced inflammatory response. However, CGA per se had MCP-1, MIP2, ICAM-1 and VCAM-1) to recruit Kupffer cells and/or
no effect on the serum proinflammatory cytokines levels (Fig. 2F). circulating macrophages by the site of HSCs (Brun et al., 2005; Paik
et al., 2003; Thirunavukkarasu et al., 2006). Second, the activation
3. Discussion of TLR4 signaling enhances TGF-␤ signaling in HSCs through
downregulating Bambi (Seki et al., 2007). Bambi is the TGF-␤1
The current study shows that the administration of CGA effec- pseudoreceptor which is downregulated by a TLR4-MyD88-NF-␬B
tively attenuated CCl4 -induced histological changes, including dependent pathway, thereby sensitizing HSCs to TGF-␤1 signaling
destruction of the structure of the hepatic lobules, formation of (Sekiya et al., 2004a, b). Regulation of Bambi by TLR4 signaling
pseudo-lobules, liver necrosis and inflammation. Meanwhile, lev- provides a link between pro-inflammatory and profibrogenic
els of serum transaminase and albumin were improved. Moreover, signals. In addition, liver endothelial cells (LECs) can also express
collagen I and ␣-SMA expression and TLR4 signaling pathway acti- TLR4. TLR4 pathway in LECs regulates angiogenesis through its
vation were inhibited. Taken together, our results suggest that CGA MyD88 pathway and this process is linked to the development of
can protect against inflammation response and ameliorate liver liver fibrosis (Jagavelu et al., 2010).
fibrosis in a rat model induced by CCl4. LPS-triggered activation of NF-␬B can induce the expression of
TLR4, one of the members of the TLR protein family, is essen- COX-2, iNOS, TNF-␣, IL-6 and IL-1␤ in Kupffer cell or activated HSC.
tial for LPS-mediated signaling. After binding to TLR4, LPS triggers The protein expression of iNOS is upregulated in kinds of acute
two critical intracellular signaling pathways, including MyD88- and chronic inflammation and plays a role in the pathogenesis of
dependent and MyD88-independent signaling cascades (Lu et al., liver fibrosis (Aram et al., 2008; McKim et al., 2003; Sanz-Cameno
2008). The MyD88-dependent pathway signals through activa- et al., 2002). Cyclooxygenase-2 (COX-2) is a crucial enzyme in the
tion of i␬B kinase (IKK), which in turn leads to activation of biosynthesis of prostaglandins and is inducible by a variety of pro-
transcription factor NF-␬B, and controls the expression of pro- inflammatory stimuli, such as cytokines and lipopolysaccharide. In
inflammatory cytokines and other immune-related genes. The the liver, COX-2 and prostaglandins production has been impli-
MyD88-independent pathway activates interferon regulatory fac- cated in inflammation, matrix remodeling, fibrosis progress and
tor 3 (IRF3) and induces the expression of interferon IFN-␤ and development of hepatocellular carcinoma (Martinez et al., 2003).
IFN-responsive genes. Besides its natural exogenous ligand LPS, Experimental evidence shows that selective pharmacologic inhibi-
there are endogenous ligands that are also able to bind and acti- tion of COX-2 could be useful to the treatment of liver fibrosis and
vate TLR4, including HMGB1, hyaluronan, and heat shock protein cancer (Breinig et al., 2007; Kim et al., 2008). TNF-␣, IL-6 and IL-
60 (Kao et al., 2008). There is rapidly increasing knowledge about 1␤, documented as “proinflammatory cytokines”, can induce liver
the relationship between TLR4 signaling and the mechanisms of inflammation and promote fibrosis (Choi et al., 1994; Kayano and
human diseases, which makes TLR4 signaling inhibition appear to Okita, 2000; Simeonova et al., 2001; Tiggelman et al., 1995).
be a promising strategy for the prevention of many inflammatory It was shown that CCl4 administration elevated the mRNA lev-
diseases (O’Neill, 2003). els of TLR4 and MyD88, leading to activation of NF-␬B and thus
112 H. Shi et al. / Toxicology 303 (2013) 107–114

Fig. 3. Effect of CGA on the TLR4 signaling pathway. mRNA expression of TLR4(A), MyD88(A). iNOS(C), COX-2(C), Bambi(C), TNF-␣(E), IL-6(E) and IL-␤(E) were detected by
RT-PCR. Protein expression of TLR4(B), MyD88(B). iNOS(D), COX-2(D) and Bambi(D) were measured by Western blot. ␤-actin served as the internal control. TNF-␣(F), IL-6(F)
and IL-1␤(F) levels in serum were detected by ELISA. The average expression of TLR4, MyD88, iNOS, COX-2, TNF-␣, IL-6 and IL-␤ was significantly higher in the CCl4 -treated
group as compared to the control group and CGA co-treated group (P < 0.01). The mRNA and protein expression of Bambi was significantly lower in the CCl4 group as compared
to the control group (P < 0.01) and it was significantly higher in the CGA co-treated group as compared to the CCl4 -treated group (P < 0.01). TNF-␣, IL-6 and IL-1␤ levels in
serum of CCl4 rats were significantly higher than those in the control group and CGA co-treatment could attenuate these cytokine levels. * P < 0.01 as compared with the
control group; # P < 0.01 as compared with the CCl4 group.

increasing the expression of proinflammatory cytokines such as monocytes treated with LPS. In the present study, CGA administra-
iNOS, COX-2, TNF-␣, IL-6, and IL-1␤ in the liver of the experimental tion decreased the hepatic mRNA expression of proinflammatory
rats. CGA is known to inhibit the biosynthesis of proinflam- cytokines, including iNOS, COX-2, TNF-␣, IL-6 and IL-1␤, and their
matory cytokines such as TNF-␣ and interleukin-6 (IL-6) in human upstream signaling molecules, such as TLR4 and MyD88. Apart
H. Shi et al. / Toxicology 303 (2013) 107–114 113

Fig. 4. Western blot analyses of NF-␬B activation. The NF-␬B protein in the nuclear and p-I␬B␣ protein were significantly higher in the CCl4 group as compared to the
control group and CGA co-treated group (P < 0.01). The level of I␬B␣ protein was significantly lower in the CCl4 group as compared to the control group (P < 0.01) and it was
significantly higher in the CGA co-treated group as compared to the CCl4 -treated group (P < 0.01). Lamin B and ␤-actin served as the internal control. * P < 0.01 as compared
with the control group; # P < 0.01 as compared with the CCl4 group.

from these, it was shown that CCl4 can reduce the expression of Acknowledgments
Bambi, and the treatment of CGA can reverse the reduction. Based
on these results, we speculate that in CCl4 -treated rats, CGA can This work was supported by National Natural Science Founda-
attenuate hepatic fibrosis, probably through the suppression of the tion of China (Nos. 81200310 and 81070328). The authors thank
TLR4-mediated proinflammatory signaling cascades. Hao Sun and Huaijie Wang for their language help and writing assis-
In population studies, greater coffee intake has been associ- tance, and also thank Yaping Liu for her help in the experimental
ated with lower risk of cirrhosis. A prospective study conducted research.
by Klatsky et al. (1993) owed subjects who drank 4 or more cups
of coffee per day had about a five-fold lower risk of cirrhosis References
than non-coffee drinkers. Another large prospective study of par-
ticipants with advanced hepatitis C-related liver disease showed Aoyama, T., Paik, Y.H., Seki, E., 2010. Toll-like receptor signaling and liver fibrosis.
3 or more cups of coffee per day consumption was associated Gastroenterol. Res. Pract., Article ID:192543.
Aram, G., Potter, J.J., Liu, X.P., Torbenson, M.S., Mezey, E., 2008. Lack of inducible
with lower rate of disease progression (Freedman et al., 2009).
nitric oxide synthase leads to increased hepatic apoptosis and decreased fibro-
Meanwhile, Tverdal and Skurtveit (2003) provided that the risk sis in mice after chronic carbon tetrachloride administration. Hepatology 47,
of death from hepatic cirrhosis was distinctively lower amongst 2051–2058.
Bataller, R., Brenner, D.A., 2005. Liver fibrosis. J. Clin. Invest. 115, 209–218.
people drinking 3 or more cups of coffee per day, compared
Breinig, M., Schirmacher, P., Kern, M.A., 2007. Cyclooxygenase-2 (COX-2) – a thera-
with persons drinking less than 2 cups. One cup of instant cof- peutic target in liver cancer? Curr. Pharm. Des. 13, 3305–3315.
fee (200 ml) contains 100–160 mg of CGA (Olthof et al., 2003) Brun, P., Castagliuolo, I., Pinzani, M., Palu, G., Martines, D., 2005. Exposure to bacterial
and Coffee drinkers are said to ingest 500–1000 mg of CGA daily cell wall products triggers an inflammatory phenotype in hepatic stellate cells.
Am. J. Physiol. Gastrointestinal Liver Physiol. 289, G571–G578.
(3–7 cups) (Olthof et al., 2001). In our study, the dose of CGA Choi, H.K., Pokharel, Y.R., Lim, S.C., Han, H.K., Ryu, C.S., Kim, S.K., Kwak, M.K., Kang,
(60 mg/kg) used for rats is equivalent to 576 mg of CGA and K.W., 2009. Inhibition of liver fibrosis by solubilized coenzyme Q10: role of Nrf2
3–4 cups of coffee for a 60-kg person, closing to daily intake of activation in inhibiting transforming growth factor-beta 1 expression. Toxicol.
Appl. Pharmacol. 240, 377–384.
coffee drinkers. Therefore coffee’s antifibrotic effect may be rel- Choi, I., Kang, H.S., Yang, Y., Pyun, K.H., 1994. IL-6 induces hepatic inflammation and
evant to CGA. Thus we proposed that a long-term consumption collagen-synthesis in-vivo. Clin. Exp. Immunol. 95, 530–535.
of coffee or other vegetable which contains substantial amount Choi, Y., Jeon, W.K., Hwang, S.J., Kim, B.I., Sohn, C.I., Park, D.I., Cho, Y.K., Kim, H.J.,
Park, J.H., 2011. The role of the gut barrier function in the pathophysiology of
of CGA may prevent or retard the progression of liver fibro- viral liver cirrhosis. Hepato-Gastroenterol. 58, 1244–1247.
sis. Feng, R.T., Lu, Y.J., Bowman, L.L., Qian, Y., Castranova, V., Ding, M., 2005. Inhibition of
In conclusion, the current study demonstrates that admin- activator protein-1, NF-kappa B, and MAPKs and induction of phase 2 detoxifying
enzyme activity by chlorogenic acid. J. Biol. Chem. 280, 27888–27895.
istration of CGA can reduce liver inflammation and fibrosis.
Freedman, N.D., Everhart, J.E., Lindsay, K.L., Ghany, M.G., Curto, T.M., Shiffman, M.L.,
The protective effect of CGA may be due to the inhibition of Lee, W.M., Lok, A.S., Di Bisceglie, A.M., Bonkovsky, H.L., Hoefs, J.C., Dienstag, J.L.,
TLR4/MyD88/NF-␬B signaling pathway. This finding provides a new Morishima, C., Abnet, C.C., Sinha, R., Grp, H.-C.T., 2009. Coffee intake is associated
with lower rates of liver disease progression in chronic hepatitis C. Hepatology
insight for understanding the anti-inflammatory effect of CGA. Our
50, 1360–1369.
observations suggest that anti-inflammatory action of CGA is one Friedman, S.L., 2003. Liver fibrosis–from bench to bedside. J. Hepatol. 38, S38–S53.
of the possible mechanisms to improve liver fibrosis. Since TLR4 Friedman, S.L., 2010. Evolving challenges in hepatic fibrosis. Nat. Rev. Gastroenterol.
signaling pathway exists in HSC, Kupffer and endothelial cells, & Hepatol. 7, 425–436.
Gressner, O.A., Lahme, B., Rehbein, K., Siluschek, M., Weiskirchen, R., Gressner, A.M.,
which one of them is the target of CGA is not clear; in vitro study is 2008. Pharmacological application of caffeine inhibits TGF-beta-stimulated con-
needed in the future. nective tissue growth factor expression in hepatocytes via PPAR gamma and
SMAD2/3-dependent pathways. J. Hepatol. 49, 758–767.
Jagavelu, K., Routray, C., Shergill, U., O’Hara, S.P., Faubion, W., Shah, V.H., 2010.
Endothelial cell toll-like receptor 4 regulates fibrosis-associated angiogenesis
Conflict of interest statement in the liver. Hepatology 52, 590–601.
Kao, Y.H., Jawan, B., Goto, S., Hung, C.T., Lin, Y.C., Nakano, T., Hsu, L.W., Lai, C.Y.,
Tai, M.H., Chen, C.L., 2008. High-mobility group box 1 protein activates hepatic
The authors declare no conflict of interest. stellate cells in vitro. Transplant. Proc. 40, 2704–2705.
114 H. Shi et al. / Toxicology 303 (2013) 107–114

Kawada, N., 2011. Evolution of hepatic fibrosis research. Hepatol. Res. 41, 199–208. Sanz-Cameno, P., Medina, J., Garcia-Buey, L., Garcia-Sanchez, A., Borque, M.J.,
Kayano, K., Okita, K., 2000. Does IL-6 regulate liver fibrosis/cirrhosis directly and Martin-Vilchez, S., Gamallo, C., Jones, E.A., Moreno-Otero, R., 2002. Enhanced
indirectly? J. Gastroenterol. 35, 250–251. intrahepatic inducible nitric oxide synthase expression and nitrotyrosine accu-
Kim, S.M., Park, K.C., Kim, H.G., Han, S.J., 2008. Effect of selective cyclooxygenase-2 mulation in primary biliary cirrhosis and autoimmune hepatitis. J. Hepatol. 37,
inhibitor meloxicam on liver fibrosis in rats with ligated common bile ducts. 723–729.
Hepatol. Res. 38, 800–809. Seki, E., Brenner, D.A., 2008. Toll-like receptors and adaptor molecules in liver dis-
Klatsky, A.L., Armstrong, M.A., Friedman, G.D., 1993. Coffee, tea, and mortality. Ann. ease: update. Hepatol. 48, 322–335.
Epidemiol. 3, 375–381. Seki, E., De Minicis, S., Osterreicher, C.H., Kluwe, J., Osawa, Y., Brenner, D.A., Schwabe,
Kono, Y., Kobayashi, K., Tagawa, S., Adachi, K., Ueda, A., Sawa, Y., Shibata, H., 1997. R.F., 2007. TLR4 enhances TGF-beta signaling and hepatic fibrosis. Nat. Med. 13,
Antioxidant activity of polyphenolics in diets - rate constants of reactions of 1324–1332.
chlorogenic acid and caffeic acid with reactive species of oxygen and nitrogen. Sekiya, T., Adachi, S., Kohu, K., Yamada, T., Higuchi, O., Furukawa, Y., Naka-
BBA-Gen. Subjects 1335, 335–342. mura, Y., Nakamura, T., Tashiro, K., Kuhara, S., Ohwada, S., Akiyama, T.,
Lee, K.J., Choi, J.H., Jeong, H.G., 2007. Hepatoprotective and antioxidant effects of the 2004a. Identification of BMP and activin membrane-bound inhibitor (BAMBI),
coffee diterpenes kahweol and cafestol on carbon tetrachloride-induced liver an inhibitor of transforming growth factor-beta signaling, as a target of
damage in mice. Food Chem. Toxicol. 45, 2118–2125. the beta-catenin pathway in colorectal tumor cells. J. Biol. Chem. 279,
Lin, R.S., Lee, F.Y., Lee, S.D., Tsai, Y.T., Lin, H.C., Lu, R.H., Hsu, W.C., Huang, C.C., 6840–6846.
Wang, S.S., Lo, K.J., 1995. Endotoxemia in patients with chronic liver-diseases- Sekiya, T., Oda, T., Matsuura, K., Akiyama, T., 2004b. Transcriptional regulation of the
relationship to severity of liver-diseases, presence of esophagel-varices, and TGF-beta pseudoreceptor BAMBI by TGF-beta signaling. Biochem. Biophys. Res.
hyperdynamic circulation. J. Hepatol. 22, 165–172. Commun. 320, 680–684.
Lu, Y.C., Yeh, W.C., Ohashi, P.S., 2008. LPS/TLR4 signal transduction pathway. Shan, J.H., Fu, J., Zhao, Z.H., Kong, X.Q., Huang, H., Luo, L., Yin, Z.M., 2009. Chloro-
Cytokine 42, 145–151. genic acid inhibits lipopolysaccharide-induced cyclooxygenase-2 expression in
Luckey, S.W., Petersen, D.R., 2001. Activation of Kupffer cells during the course of RAW264.7 cells through suppressing NF-kappa B and JNK/AP-1 activation. Int.
carbon tetrachloride-induced liver injury and fibrosis in rats. Exp. Mol. Pathol. Immunopharmacol. 9, 1042–1048.
71, 226–240. Shi, H.Y., Dong, L., Bai, Y.H., Zhao, J.H., Zhang, Y., Zhang, L., 2009. Chlorogenic acid
Martinez, O.N., Ricote, G.C., Monzon, C.G., 2003. Role of cyclooxygenase-2 in the against carbon tetrachloride-induced liver fibrosis in rats. Eur. J. Pharmacol. 623,
pathogenesis of chronic liver diseases. Med. Clin. 121, 743–748. 119–124.
McKim, S.E., Gabele, E., Isayama, F., Lambert, J.C., Tucker, L.M., Wheeler, M.D., Connor, Shi, H.Y., Dong, L., Zhang, Y., Bai, Y.H., Zhao, J.H., Zhang, L., 2010. Protective effect
H.D., Mason, R.P., Doll, M.A., Hein, D.W., Arteel, G.E., 2003. Inducible nitric oxide of a coffee preparation (Nescafe pure (R)) against carbon tetrachloride-induced
synthase is required in alcohol-induced liver injury: studies with knockout mice. liver fibrosis in rats. Clin. Nutr. 29, 399–405.
Gastroenterol. 125, 1834–1844. Shin, J.W., Wang, J.H., Kang, J.K., Son, C.G., 2010. Experimental evidence for the pro-
O’Neill, L.A., 2003. Therapeutic targeting of toll-like receptors for inflammatory and tective effects of coffee against liver fibrosis in SD rats. J. Sci. Food Agric. 90,
infectious diseases. Curr. Opin. Pharmacol. 3, 396–403. 450–455.
Olthof, M.R., Hollman, P.C.H., Buijsman, M., van Amelsvoort, J.M.M., Katan, M.B., Simeonova, P.P., Gallucci, R.M., Hulderman, T., Wilson, R., Kommineni, C., Rao, M.,
2003. Chlorogenic acid, quercetin-3-rutinoside and black tea phenols are exten- Luster, M.I., 2001. The role of tumor necrosis factor-alpha in liver toxicity, inflam-
sively metabolized in humans. J. Nutr. 133, 1806–1814. mation, and fibrosis induced by carbon tetrachloride. Toxicol. Appl. Pharmacol.
Olthof, M.R., Hollman, P.C.H., Katan, M.B., 2001. Chlorogenic acid and caffeic acid are 177, 112–120.
absorbed in humans. J. Nutr. 131, 66–71. Thirunavukkarasu, C., Watkins, S.C., Gandhi, C.R., 2006. Mechanisms of endotoxin-
Paik, Y.H., Schwabe, R.F., Bataller, R., Russo, M.P., Jobin, C., Brenner, D.A., 2003. Toll- induced NO, IL-6, and TNF-alpha production in activated rat hepatic stellate
like receptor 4 mediates inflammatory signaling by bacterial lipopolysaccharide cells: Role of p38 MAPK. Hepatology 44, 389–398.
in human hepatic stellate cells. Hepatol. 37, 1043–1055. Tiggelman, A.M.B., Boers, W., Linthorst, C., Sala, M., Chamuleau, R.A.F., 1995.
Parlesak, A., Schafer, C., Schutz, T., Bode, J.C., Bode, C., 2000. Increased intestinal per- Collagen-synthesis by human liver (myo)fibroblasts in culture-evidence for
meability to macromolecules and endotoxemia in patients with chronic alcohol a regulatory role of IL-1-beta, IL-4, TGF-beta and IFN-gamma. J. Hepatol. 23,
abuse in different stages of alcohol-induced liver disease. J. Hepatol. 32, 742–747. 307–317.
Pinzani, M., Macias-Barragan, J., 2010. Update on the pathophysiology of liver fibro- Tverdal, A., Skurtveit, S., 2003. Coffee intake and mortality from liver cirrhosis. Ann.
sis. Expert Rev. Gastroenterol. Hepatol. 4, 459–472. Epidemiol. 13, 419–423.
Ruhl, C.E., Everhart, J.E., 2005. Coffee and tea consumption are associated with a Xu, Y.X., Chen, J.W., Yu, X.A., Tao, W.W., Jiang, F.R., Yin, Z.M., Liu, C., 2010. Protective
lower incidence of chronic liver disease in the United States. Gastroenterol. 129, effects of chlorogenic acid on acute hepatotoxicity induced by lipopolysaccha-
1928–1936. ride in mice. Inflamm. Res. 59, 871–877.
Ruwart, M.J., Wilkinson, K.F., Rush, B.D., Vidmar, T.J., Peters, K.M., Henley, K.S., Appel- Zhang, X., Huang, H., Yang, T.T., Ye, Y., Shan, J.H., Yin, Z.M., Luo, L., 2010. Chlorogenic
man, H.D., Kim, K.Y., Schuppan, D., Hahn, E.G., 1989. The integrated value of acid protects mice against lipopolysaccharide-induced acute lung injury. Injury-
serum procollagen-III peptide over time predicts hepatic hydroxyproline con- Int. J. Care Injured 41, 746–752.
tent and stainable collagen in a model of dietary cirrhosis in the rat. Hepatology Zhu, J.K., Mohan, C., 2010. Toll-like receptor signaling pathways-therapeutic oppor-
10, 801–806. tunities. Mediat. Inflamm., Article ID:781235.

You might also like