You are on page 1of 9

Biomedicine & Pharmacotherapy 101 (2018) 599–607

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy


journal homepage: www.elsevier.com/locate/biopha

Curcumin protects against hepatic stellate cells activation and migration by T


inhibiting the CXCL12/CXCR4 biological axis in liver fibrosis:A study in
vitro and in vivo

Lifeng Qin, Jinmei Qin, Xiumei Zhen, Qian Yang, Liyi Huang
Department of Infectious Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China

A R T I C L E I N F O A B S T R A C T

Keywords: The C-X-C motif chemokine 12/C-X-C chemokine receptor type 4 (CXCL12/ CXCR4) biological axis plays an
Curcumin important role in the pathogenesis of liver fibrosis. Curcumin is known to have an anti-fibrosis effect, but the
Liver fibrosis specific mechanism needs to be elucidated. There is currently no evidence illustrating a connection between
Hepatic stellate cell curcumin and the CXCL12/CXCR4 axis in liver fibrosis. Here, we investigated the contribution of curcumin on
CXCL12/CXCR4 axis
CXCL12/ CXCR4 biological axis in liver fibrosis. Our results showed that curcumin remarkably improved hepatic
function and liver fibrosis, and the effects are similar as silymarin. The alleviation of liver fibrosis with curcumin
treatment was associated with a reduction of CXCL12, CXCR4, α-SMA and RhoA. In addition, curcumin mark-
edly inhibited the proliferation and migration of HSC-T6 cells. This study indicates that curcumin could protect
against hepatic stellate cells activation and migration by inhibiting the CXCL12/CXCR4 biological axis in liver
fibrosis.

1. Introduction colchicine, silymarin, and so on. They can reverse liver fibrosis or early
cirrhosis, and have been widely used in a clinical setting. Curcumin, a
Liver fibrosis, a repair response of the liver against various injuries, monomer extracted from the root of turmeric plants, is a kind of natural
is caused by the excess hyperplasia and deposition of extracellular phenolic antioxidant. It has been found that curcumin has many
matrix (ECM) in the liver. Liver fibrosis is a necessary stage by which all pharmacological effects such as anti-inflammatory, anti-lipid perox-
chronic liver injury develops into cirrhosis. Previous studies [1,2] have idation, anti-tumor and liver protection, etc. [5]. In recent years, cur-
shown that liver fibrosis is a reversible pathological process; therefore, cumin has attracted widespread attention with regard to anti-fibrosis,
looking for effective drugs to block or reverse the occurrence and but its pharmacological mechanism is not entirely clear. There is cur-
progress of liver fibrosis can avoid liver cirrhosis or liver cancer, which rently no evidence illustrating a connection between curcumin and the
can significantly improve the prognosis of patients and save medical CXCL12/CXCR4 axis in liver fibrosis. This study aims to verify the role
resources. Hepatic stellate cells (HSCs) are the cellular source of most of of CXCL12/CXCR4 axis in pathogenesis of liver fibrosis and investigate
the ECM, and the activation and migration of HSCs is the central link of the effect of curcumin on the CXCL12/CXCR4 axis in liver fibrosis in
liver fibrosis. These processes are primarily regulated by cytokines, vitro and in vivo.
especially chemokines [3,4].
The C-X-C motif chemokine 12/C-X-C chemokine receptor type 4 2. Materials and methods
(CXCL12/CXCR4) biological axis, mainly composed of chemokine
CXCL12 and its receptor CXCR4, has been a hot topic in recent years. 2.1. Chemical and reagents
The CXCL12/CXCR4 axis is involved in a variety of pathological and
physiological processes, such as inflammation, immunity, wound Carbon tetrachloride (CCl4) and Olive oil were purchased from
healing, tissue and organ fibrosis, etc.; in particular, it participates in Aladdin Biochemical Technology Co., Ltd (Shanghai, China). Curcumin
the pathogenesis of liver fibrosis. (purity: 98%) was purchased from Macklin Biochemical Technology
Chinese medicine has shown unique advantages and excellent pro- Co., Ltd (Shanghai, China). Silymarin was purchased from MADAUS
spects in the prevention and treatment of liver fibrosis, such as salvia, GmbH (Cologne, Germany). RIPA and PMSF were purchased from


Corresponding author.
E-mail address: huangliyi175@163.com (L. Huang).

https://doi.org/10.1016/j.biopha.2018.02.091
Received 22 November 2017; Received in revised form 6 February 2018; Accepted 20 February 2018
0753-3322/ © 2018 Elsevier Masson SAS. All rights reserved.
L. Qin et al. Biomedicine & Pharmacotherapy 101 (2018) 599–607

Beyotime Biotechnology Co., Ltd (Shanghai, China). RNA Extraction Kit Table 1
and PrimeScript RT Master Mix were purchased from Takara Primer sequences for polymerase chain reaction.
Biotechnology co., Ltd (TaKaRa, Shiga, Japan). All primers were syn-
Gene name Direction Sequences(5′→3′) Product Length
thesized by Takara Biotechnology co., Ltd (TaKaRa, Shiga, Japan). (bp)
Primary antibodies against CXCR4, Tubulin and horseradish peroxidase
(HRP)-conjugated anti-rabbit secondary antibodies were purchased CXCL12 Forward GAGCCAACGTCAAACATCTGAA 147
Reverse ACTTGTTTAAGGCTTTGTCCAGGTA
from Abcam (Cambridge, MA, USA); primary antibodies against
CXCR4 Forward ATTGTCCACGCCACCAACAG 119
CXCL12 were purchased from Cell Signaling Technology, Inc. (Danvers, Reverse ACATCGGCGAAGATGATGTCAG
MA, USA). Primary antibodies against alpha-smooth muscle actin (α- GAPDH Forward GGCACAGTCAAGGCTGAGAATG 143
SMA), ras homolog gene family (member A) (RhoA) and biotinylated Reverse ATGGTGGTGAAGACGCCAGTA
goat anti-rabbit secondary antibodies were purchased from Boster
Biological Technology Co., Ltd (Wuhan, China). Rat Collagen I (COL I),
hyaluronic acid (HA), laminin (LN) and procollagen type III (PCIII) Tokyo, Japan). The fibrotic area was checked with the Image J 1.44s
ELISA Kit were purchased from Shanghai Enzyme-linked Biotechnology software (National Institutes of Health, Bethesda, MD, United States)
Co., Ltd (Shanghai, China). Platelet-derived growth factor-BB (PDGF- [6]. The percentage of the fibrotic area was calculated by comparing
BB) and CXCL12α was purchased from R&D Systems, Inc. (Minneapolis, the collagen stained area to the total area.
MN, USA). The liver tissue sections were de-waxed, hydrated and subjected to
5% H2O2 for 10min. Then, slides were transferred to sodium citrate
2.2. Animals and experiment protocols buffer for heat-induced antigen retrieval. Sections were blocked and
incubated overnight at 4 °C with rabbit anti-α-SMA antibody (1:200) or
Sixty 8-week-old, male Sprague-Dawley rats weighing 200–220 g rabbit anti–RhoA antibody (1:150), respectively. All antibodies were
were purchased from the Experimental Animal Center of Guangxi diluted in tris buffered saline 5% bovine serum albumin, while nega-
Medical University (Nanning, China) (Animal Use Certificate Number: tive-control sections were incubated with Phosphate Buffered Saline
SYXK Gui 2014-0003), and housed in an environmentally controlled (PBS) instead. After that, the slides were incubated with biotinylated
room (23 ± 3 °C, 55 ± 10% humidity) on a 12 h light-dark cycle goat anti-rabbit secondary antibodies for 15 min at 37 °C according to
(06:00–18:00, light), with standard food and water provided ad libitum. the instructions. The immunoreactivities were visualized with 3,3′-
The animal studies were carried out in strict accordance with the diaminobenzidine tetrahydrochloride (DAB) solution, followed by
guidelines for the Care and Use of Laboratory Animals of Guangxi staining the nucleus with hematoxylin, dehydrating, and mounting. The
Medical University, Nanning, China. All efforts were made to minimize sections were analyzed under a light microscope. The Image-Pro Plus
the pain and discomfort of the animals. 6.0 system (Media Cybernetics Inc., Bethesda, MD, USA) was used for
After a week-long acclimation period, rats were randomly divided the semi-quantitative evaluation. Integrated optical density per area
into six groups according to random number table, including the fol- (IOD/area) was applied to indicate the level of corresponding protein
lowing groups: control, model, silymarin, curcumin low dose ((CUR-L)), expression [7].
curcumin middle dose ((CUR-M)) and curcumin high dose ((CUR-H))
(n = 10/group). Liver fibrosis was induced by the administration of 2.5. Cell culture and cell proliferation assay
1.5 ml of CCl4/olive oil (2:3, v/v) ∕ kg body weight by subcutaneous
injection. Rats in all groups except the control group were injected with The HSC-T6 cell line sample was purchased from Procell Life
an olive oil solution of CCl4 every three days for 8 weeks. Rats in the Science & Technology Co., Ltd (Wuhan, China). The cells were cultured
control group were injected with the same volume of saline for 8 weeks. in Dulbecco's modified Eagle's medium (DMEM) (Gibco, Thermo Fisher
The treatments were performed simultaneously with the establishment Scientific, Inc., Waltham, MA, USA) supplemented with 10% fetal bo-
of the fibrosis model. Rats in the silymarin group were given silymarin vine serum (FBS) (Tianjin Haoyang Biological Manufacture Co., Ltd.,
orally at 50 mg/kg every day for 8 weeks. Rats in the (CUR-L), (CUR-M) Tianjin, China), and incubated at 37 °C in 5% CO2 air. Following 2
and (CUR-H) groups were given curcumin orally at 50 mg/kg, 100 mg/ weeks of culture on plastic tissue-culture dishes, the cells were plated at
kg and 200 mg/kg, respectively, every day for 8 weeks. Rats in the a density of 2 × 104 cells/well in 96-well plates. The HSC-T6 cells were
model group were orally given the same volume of saline as curcumin activated with PDGF-BB. The concentration of PDGF-BB used for
every day for 8 weeks. treatment was 10 ng/ml for 24 h at 37 °C.
Cell proliferation was evaluated using a 3-(4,5-dimethyl-2-thia-
2.3. Measurement of functional liver levels zolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) assay. After HSC-
T6 cells were digested by trypsin (0.25%), single suspended state cells
At 48 h after the last treatment in the experiment, all rats were were cultured in DMEM medium supplemented with 10% FBS and then
weighed and killed after being anaesthetized by an intraperitoneal in- seeded in a 96-well plate with 180 μL (5 × 104 cells/mL) per well. After
jection of 10% chloral hydrate solution at 3 ml/kg. Immediately after 24–96 h incubation, cells were exposed to curcumin dissolved in 0.1%
blood sample collection, alanine aminotransferase (ALT), aspartate dimethyl sulfoxide (DMSO) at different concentrations (0, 10, 20, 40,
aminotransferase (AST), total bilirubin (TBIL) and albumin (ALB) levels 80, 160 μM) for 24 h. Untreated cells were used as controls. MTT so-
in serum were measured with an automated analyzer (LX20; Beckman lution (20 μL, 5 mg/mL, diluted in PBS) was added to each well and the
Coulter, Fullerton, United States) at the First Affiliated Hospital of plate was incubated at 37 °C in a 5% CO2 atmosphere for 4 h. Then, the
Guangxi Medical University (Nanning, China). supernatants were removed and 150 μL/well DMSO was added to dis-
solve formazan crystals. After 5 min of shaking dissolution, the absor-
2.4. Histopathology and immunohistochemistry bance at 490 nm was read using ELx800 Microplate Reader (BioTek
Instruments, Inc., Winooski, VT, USA).
Harvested liver tissues were fixed in 40 g/L formaldehyde, em- To carry out follow-up experiments, HSC-T6 cells were randomly
bedded in paraffin and sectioned at a thickness of 4 μm. Hematoxylin divided into four groups: the control group, the PDGF-BB group, the
and eosin (HE) staining and Masson’s trichrome staining were used for PDGF-BB+curcumin (20 μM) group and the PDGF-BB+curcumin
histological structure analysis and fibrosis area analysis, respectively. (40 μM) group. The concentration and use of PDGF-BB are the same as
Five random views of Masson trichrome-stained sections from each above. All cells except the control group were activated with PDGF-BB.
sample (n = 10/group) were captured by a light microscope (Olympus, Cells in the PDGF − BB + curcumin (20 μM) group and the

600
L. Qin et al. Biomedicine & Pharmacotherapy 101 (2018) 599–607

Fig. 1. Graphical abstract.

Table 2
Comparison of functional liver levels in serum (mean ± SD).

Group n ALT AST TBIL ALB

* * *
Model 10 156.31 ± 24.57 287.12 ± 33.89 34.78 ± 13.69 8.45 ± 2.64*
Silymarin 10 78.22 ± 8.45** 185.10 ± 26.78** 12.91 ± 3.56** 15.78 ± 1.67**
CUR-L 10 124.36 ± 17.54 258.61 ± 30.18 26.47 ± 10.37 10.30 ± 2.42
CUR-M 10 103.54 ± 12.48 219.18 ± 27.05 18.75 ± 7.25 13.47 ± 2.03
CUR-H 10 76.72 ± 8.36**,*** 188.34 ± 25.14**,*** 13.03 ± 3.74**,*** 15.89 ± 1.75**,***
Control 10 62.05 ± 12.23 152.33 ± 22.56 9.64 ± 2.03 16.63 ± 1.18

Note: ALT: Alanine aminotransferase; AST: Aspartate aminotransferase; TBIL: Total bilirubin; ALB: Albumin.
* P < 0.05 vs. control group.
** P < 0.05 vs. model group.
*** P > 0.05 vs. Silymarin group.

PDGF − BB + curcumin (40 μM) group were exposed to curcumin dis- instructions. Each sample was analyzed in triplicate.
solved in 0.1% DMSO at 20 μM and 40 μM for 24 h, respectively.
2.8. Western blot analysis
2.6. Reverse Transcription and Real time Quantitative Polymerase Chain
Reaction (RT-qPCR) Total protein from liver tissue samples and cells was extracted by
the standard procedure [8]. The BCA assay was used to determine the
RT-qPCR was used to assess the mRNA expression of molecules in- protein concentration. Equal amounts of protein were resolved by 15%
volved in the CXCL12/CXCR4 axis (CXCL12 and CXCR4). Total RNA sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE)
was extracted from the liver tissues of each rat using the RNA and transferred to a polyvinylidene fluoride (PVDF) membrane. The
Extraction Kit. First-strand cDNA synthesis was performed with 1 μg of membrane was blocked with 5% nonfat milk in TBS-Tween 20 (0.1%
total RNA. Moreover, cDNA samples were thereafter amplified in the TBST) at room temperature for 1 h. After a brief rinse, the membrane
ABI Prism 7500 Sequence Detection system (Applied Biosystems, was incubated overnight at 4 °C in TBST with corresponding primary
Massachusetts, United States) for 40 cycles (95 °C for 5 s, 60 °C for 30 s) antibodies including rabbit mAb of CXCL12 (1:1000), CXCR4 (1:1000),
with specific oligonucleotide primers (Takara Bio Inc., Shiga, Japan). α-SMA (1:1500), RhoA (1:1500) and Tubulin (1:2000). After that, the
Each sample was analyzed in triplicate, with glyceraldehyde 3-phos- membrane was incubated with horseradish peroxidase (HRP)-con-
phate dehydrogenase (GAPDH) used for normalization. The relative jugated anti-rabbit secondary antibodies (1:10000) for 1 h at room
quantification of target genes was determined using the 2−ΔΔCT temperature. Then the membrane was washed in TBST and protein was
method. Primers used in RT-qPCR analyzes are listed in Table 1. detected by enhanced chemical luminescence (ECL). Each sample was
analyzed in triplicate.
2.7. Enzyme linked immunosorbent assay (ELISA)
2.9. Transwell assay
The levels of HA, LN, and PCIII in serum or COL I in cell culture
supernatant were detected by ELISA according to the manufacturer’s The migration ability of the HSC-T6 cells was evaluated with 24-

601
L. Qin et al. Biomedicine & Pharmacotherapy 101 (2018) 599–607

Fig. 2. Histopathological change of liver tissue in different groups. (A) Representative sections stained with HE are shown for each group (original magnification, 200×). (B)
Representative sections stained with Masson’s trichrome are shown for each group (original magnification, 200×). (C) Quantitative analyzes of liver fibrosis were performed using
Masson stained sections. *p < 0.05, **p < 0.01 compared with control group;#p < 0.05, ##p < 0.01 compared with model group;+p < 0.05, ++p < 0.01 compared with sily-
marin group.

well Transwell chambers (Corning, Tewksbury, MA, USA). Cells were insert (pore size, 8 μm). The lower chamber was filled with 600 μL
randomly divided into four groups: the control group, the CXCL12 DMEM medium with 4% FBS and 100 ng/mL CXCL12α, and the lower
group, the CXCL12 + curcumin (20 μM) group and the chamber of control group was filled with 600 μL DMEM medium with
CXCL12 + curcumin (40 μM) group. Cells in the CXCL12 + curcumin 4% FBS. After 24 h of incubation, cells were removed from the upper
(20 μM) group and the CXCL12 + curcumin (40 μM) group were treated chamber of the filter, using a cotton swab. Cells on the underside were
with curcumin 20 μM and 40 μM respectively, and subsequently culti- washed with PBS and fixed with 5% glutaraldehyde 30 min, stained
vated for 24 h. Each group cells were harvested, and 1 × 104 cells in with 0.5% crystal violet 20 min. The films were cut along the edge with
100 μL of non-serum medium was placed in the upper chamber of an blade and sealed with a neutral gum, and counted in five randomly

602
L. Qin et al. Biomedicine & Pharmacotherapy 101 (2018) 599–607

Table 3
Comparison of liver fibrosis parameters in serum (mean ± SD).

Group n HA LN PCⅢ

Model 10 186.24 ± 26.42* 237.64 ± 35.12* 215.06 ± 30.18*


Silymarin 10 95.87 ± 12.97** 145.81 ± 21.12** 138.22 ± 17.62**
CUR-L 10 157.35 ± 21.89 198.63 ± 31.23 184.34 ± 25.73
CUR-M 10 112.69 ± 17.24 174.30 ± 26.47 151.03 ± 21.39
CUR-H 10 97.34 ± 13.23**,*** 143.56 ± 22.75**,*** 136.46 ± 18.55**,***
Control 10 45.04 ± 7.35 69.79 ± 13.48 77.53 ± 15.36

Note: HA: Hyaluronic acid; LN: Laminin; PCⅢ: Procollagen type Ⅲ.


* P < 0.05 vs. control group.
** P < 0.05 vs. model group.
*** P > 0.05 vs. Silymarin group.

Fig. 3. Effect of curcumin on the mRNA and protein expression of molecules of the CXCL12/CXCR4 axis. (A–B) The bar graph shows mean relative mRNA expression levels of CXCL12 and
CXCR4 in liver tissues. Each sample was repeated three times from each cluster. Data are normalized to GAPDH mRNA expression levels. (C) Representative Western blot analysis of
CXCL12 and CXCR4 protein expression levels in each group. ((DeE)) The bar graph represents mean relative protein expression levels of CXCL12 and CXCR4. Data are normalized to
tubulin protein expression levels. *p < 0.05, **p < 0.01 compared with control group;#p < 0.05, ##p < 0.01 compared with model group;+p < 0.05, ++p < 0.01 compared with
silymarin group.

selected fields under a phase contrast microscope. Assays were per- followed by the Student-Newman-Keuls post hoc test. P-values less than
formed in triplicate. 0.05 were considered statistically significant. All of the statistical ana-
lyses were performed with the statistical software package SPSS version
2.10. Statistical analysis 20.0 (SPSS Inc., Chicago, United States).

All data are expressed as the mean ± SD. Comparisons between


groups were performed using one-way analysis of variance (ANOVA)

603
L. Qin et al. Biomedicine & Pharmacotherapy 101 (2018) 599–607

Fig. 4. Effects of curcumin on α-SMA and RhoA expression and hepatic distribution. (A) Immunohistochemical staining of α-SMA (original magnification, 200×). (B)
Immunohistochemical staining of RhoA (original magnification, 200×). (C–D) Quantification of α-SMA and RhoA staining by digital image analysis. The ocular fields per specimen were
assessed as mean IOD, and data are presented as mean ± SD (n = 10).*p < 0.05, **p < 0.01 compared with control group;#p < 0.05, ##p < 0.01 compared with model
group;+p < 0.05, ++p < 0.01 compared with silymarin group.

3. Results model group.


Taken together, it was clearly indicated that curcumin obviously
3.1. Curcumin treatment reduces CCl4-induced liver injury in rats improved liver injury in liver fibrosis. Moreover, the effect was similar
as silymarin.
During feeding, the rats in the control group were lively and sen-
sitive to external stimuli, with soft and shiny hair, a strong appetite and
3.2. Curcumin treatment attenuates CCl4-induced liver fibrosis
a good mental state, while the rats in the model group were feeble and
inactive, with bleak hair, poor appetite and slow response to external
Compared with the control group, the levels of HA, LN and PCIII in
stimuli. In eight weeks, the body weight of rats in the control group
the serum of the model group were significantly increased (P < 0.05).
showed a remarkable upward trend, while the body weight of rats in
The levels of HA, LN and PCIII in the curcumin and silymarin treatment
the model group increased slowly. Moreover, the general condition of
groups were obviously lower than those in the model group
rats in the curcumin treatment groups and the silymarin group were
(P < 0.05), in a dose-dependent manner. When comparing the (CUR-
between the control and model groups (Fig. 1).
H) group with the silymarin group, there was no significant difference
Compared with the control group, the levels of ALT, AST and TBIL
between the two groups (P > 0.05) (Table 3).
in the serum of the model group were significantly increased
After livers were harvested, compared with the macropathology of
(P < 0.05) and ALB was significantly decreased (P < 0.05). The levels
the livers in the control group, the color of livers in the model group
of ALT, AST, TBIL and ALB in the curcumin treatment groups and si-
were darker, the surfaces were less smooth, the edge of the livers be-
lymarin group were obviously improved compared with the model
came blunt, and the texture was more real with a nodular sense. There
group (P < 0.05), and the changes showed a dose-dependent manner
were no significant changes in the volume of the liver. The macro-
in the curcumin treatment group. Compared (CUR-H) group with the
pathology in the curcumin treatment groups and silymarin group was
silymarin group, there was no significant difference between the two
between that of the control and model groups.
groups (P > 0.05) (Table 2).
Histological examination using Masson’s staining was performed to
Histological examination using hematoxylin-eosin (HE) were per-
show the extent of liver fibrosis. The control group showed the normal
formed to show the extent of liver damage (Fig. 2A). Liver tissue
architecture while the model group presented extensive liver bridging
samples from the control group showed normal lobular architecture
fibrosis and substantial collagen deposition. However, bridging fibrosis
with radiating hepatic cords pointing to the central veins, whereas the
and collagen were distinctly decreased by the curcumin or silymarin
model group exhibited fatty degeneration, ballooning changes of he-
treatment compared with the model group (Fig. 2B and C).
patocytes and necrosis. In contrast, the curcumin and silymarin treat-
The above results show that curcumin significantly improved liver
ment remarkably ameliorated the adipose degeneration of hepatocytes
fibrosis in a dose-dependent manner. Moreover, the effect was similar
and reduced the immigration of inflammatory cells compared with the
as silymarin.

604
L. Qin et al. Biomedicine & Pharmacotherapy 101 (2018) 599–607

3.3. Curcumin treatment inhibits the CXCL12/CXCR4 axis in vivo

As shown in Fig. 3, compared with the control group, the mRNA and
protein level of CXCL12 and CXCR4 were significantly increased in the
model group (P < 0.01). In the curcumin treatment groups, the mRNA
and protein expression levels of CXCL12 and CXCR4 were significantly
decreased in a dose-dependent manner. However, in the silymarin
group, there were no obvious changes in the mRNA and protein ex-
pression levels of CXCL12 and CXCR4 compared with the model group
(P > 0.05). These data indicate that CXCL12/CXCR4 axis is involved in
the pathogenesis of liver fibrosis, and curcumin effectively inhibit the
CXCL12/CXCR4 axis to improve liver fibrosis. However, silymarin has
no obvious effects on the CXCL12/CXCR4 axis.

3.4. Curcumin treatment suppresses the expression levels of α-SMA and


RhoA in vivo

α-SMA (an indicator of the activation of HSCs) and RhoA (a


downstream target of CXCL12/CXCR4 axis, which may be closely cor-
related with cells migration) were evaluated by immunohistochemical
staining. Compared with the control group, the model group sig-
nificantly increased α-SMA-positive and RhoA-positive cells in fibrotic
areas and vessel walls in liver sections, and the curcumin treatment
groups showed less α-SMA-positive and RhoA -positive cells compared
to the model group (Fig. 4A and B). Moreover, the expression level is
inversely proportional to the dosage of curcumin. However, there was
little change for RhoA-positive cells in the silymarin group. Semi-
quantitative analysis by Image-Pro Plus 6.0 confirmed the findings by
immunohistochemistry (Fig. 4C and D). The data suggest that curcumin
inhibit activation and migration of HSCs.

3.5. Curcumin treatment inhibits the expression levels of α-SMA, RhoA and
COL I and CXCL12/ CXCR4 axis in vitro

As shown in Fig. 5, treatment of HSC-T6 cells with 10 ng/ml PDGF-


BB for 24 h significantly increased CXCR4, α-SMA, RhoA and COL I
expressions (P < 0.01). Western blot analysis showed that curcumin
reduced PDGF-BB-stimulated CXCR4, α-SMA and RhoA production in a
concentration-dependent manner (Fig. 5A and B). ELISA showed that
PDGF-BB-induced COL I was also suppressed by curcumin in a con-
centration-dependent manner (Fig. 5C). These results suggest that
curcumin attenuates liver fibrosis by inhibiting activation of HSCs and
Fig. 5. Effect of curcumin on expression levels of CXCL12/CXCR4 axis and relevant CXCL12/ CXCR4/ RhoA axis.
molecules in HSC-T6 cells. (A) Representative Western blot analysis of CXCR4, α-SMA
and RhoA protein expression levels in curcumin treated HSC-T6 cells. (B) The bar graph
3.6. Curcumin treatment suppresses the proliferation of HSC-T6 cells
represents mean relative protein expression levels of CXCR4, α-SMA and RhoA. Data are
normalized to tubulin protein expression levels. (C) Elisa analysis of COL I protein ex-
pression levels in curcumin treated HSC-T6 cells.*p < 0.05, **p < 0.01 compared with As shown in Fig. 6, curcumin reduced proliferation of HSC-T6 cells
PDGF-BB only group. ##P < 0.01 compared with HSC-T6 treated with solvent but no in a concentration-dependent manner (0–160 μM) and significant sup-
PDGF-BB and curcumin. pression was observed at 20–160 μM (P < 0.05). The data confirmed
that curcumin effectively suppressed the proliferation of HSCs.

3.7. Curcumin treatment inhibits the migration of HSC-T6 cells

CXCL12/CXCR4/ RhoA axis may be closely correlated with cells


migration, so we investigated the effect of curcumin on migration of
HSC-T6 cells. Transwell assay indicated that CXCL12 prominently in-
creased migration of HSC-T6 cells in CXCL12 group compared with the
control group, moreover, curcumin notably inhibited HSC-T6 cells
migration in a concentration-dependent manner (Fig. 7). The results
clearly indicate that curcumin significantly inhibited HSC-T6 cells mi-
gration.

4. Discussion
Fig. 6. Effect of curcumin on proliferation rate of HSC-T6 cells. *p < 0.05, **
p < 0.01
compared with HSC-T6 treated with solvent but no curcumin. Curcumin, a kind of plant polyphenol extracted from the rhizome of
turmeric, is an active ingredient in curry. Its molecular formula is

605
L. Qin et al. Biomedicine & Pharmacotherapy 101 (2018) 599–607

Fig. 7. The effect of curcumin on the HSC-T6 cells migration ability. (Left) Representative pictures of transwell assay in each group. (Right) The bar graph shows the result of analysis of
five randomly selected fields. *p < 0.05, **p < 0.01 compared with CXCL12 only group. ##P < 0.01 compared with HSC-T6 treated with solvent but no CXCL12 and curcumin.

C21H20O6, with a molecular weight of 368.39. In traditional Chinese subject to biological, physical and chemical irritation or other injuries,
medicine, turmeric is mainly used for the treatment of abdominal pain, HSCs are activated into myofibroblast-like cells, showing proliferation,
jaundice and other related diseases. Pharmacological studies [5] on migration, and secretion of ECM. Therefore, activation and migration of
curcumin indicate that curcumin has a wide range of pharmacological HSCs are critical in pathogenesis of liver fibrosis. A large number of
effects, such as anti-inflammatory, anti-oxidation, lipid-lowering, anti- studies [19–22] confirmed that CXCL12/CXCR4 axis is activated in
tumor and other effects. Researches show that [9,10] curcumin has an various of organs fibrosis, especially liver fibrosis. Sawitza [23] et al.
anti-tissue/organ fibrosis effect, mainly in anti-liver fibrosis, pulmonary have shown that CXCL12/CXCR4 promotes HSCs migration and che-
fibrosis, renal fibrosis, pancreatic fibrosis, cystic fibrosis and so on. motaxis in liver fibrosis. Hong [24] et al. confirmed that activated HSCs
However, the pharmacological mechanism of curcumin anti-liver fi- express functional CXCR4, and that receptor engagement by its ligand,
brosis remains to be further elucidated. Our study showed that cur- CXCL12, leads to a pro-fibrogenic phenotype characterized by increased
cumin obviously improved liver injury and fibrosis. These findings are proliferative capacity and COL I and α-SMA production. These reports
consistent with the results of other studies [11,12]. Moreover, inter- suggest that CXCL12/CXCR4 axis is closely related to the pathogenesis
vention effect in the (CUR-H) group was similar to that for the silymarin of liver fibrosis. We showed that CXCL12/CXCR4 axis is involved in
group. Silymarin is the most commonly used drug to treat liver fibrosis liver fibrosis, which is consistent with above reports; and curcumin
or injury owing to remarkable effect, so curcumin have an excellent could effectively inhibit the CXCL12/CXCR4 axis to improve liver fi-
clinical application prospect. brosis, while silymarin has no obvious effects on the CXCL12/CXCR4
CXCL12, also known as stromal cell derived factor (SDF-1) or pre-B- axis. CXCL12/CXCR4 axis may be a target of curcumin, but not the
cell-growth-stimulating factor (PBSF), is a small molecule cytokine, target of silymarin. However, no previous studies reported the effect of
belonging to the CXC chemokine family. The human CXCL12 gene is silymarin on the CXCL12/CXCR4 axis in liver fibrosis, therefore, more
located on the long arm of chromosome 10 and the length of the studies are needed to confirm our results.
CXCL12 cDNA is 1776 bp. The human and rats CXCL12 genes show It is generally believed that the activation of HSCs is the central link
about 90% homology, with similar biological functions; therefore, rats of liver fibrosis, and α-SMA is a molecular marker of HSCs activation.
and HSC-T6 cells show good representation as study objects. CXCL12 Furthermore, increased proliferation is the main manifestation of HSCs
has different isomers: CXCL12a and CXCL12β, which are caused by activation [1,25]. In this study, we found that curcumin could effec-
hydrolyzing different parts of protein after translation from mRNA. tively inhibit activation of HSCs to play the role of anti-liver fibrosis.
CXCL12 can be expressed in many tissues, including the brain, thymus, This result is consistent with the results of Zhao et al. [11,26].
liver, heart, lung, kidney, bone marrow, spleen and so on [13]. CXCR4 Under induction and chemotaxis of inflammatory mediators re-
is the main receptor of CXCL12, which is highly conserved in evolution leased from injured hepatocytes, HSCs migrate to areas of inflammatory
and is a G protein-coupled receptor consisting of 352 amino acids. It has activation and then secreting ECM, which is an essential step in the
a standard seven-membrane structure, the extracellular domain (N- occurrence and progress of liver fibrosis. Studies [22,23] showed that,
terminal region) combines with a ligand, the intracellular domain CXCL12/CXCR4 biological axis is related to directed migration of HSCs.
coupling with G proteins and the C-terminal containing serine/threo- In the present study, we found that curcumin could inhibit HSC-T6 cells
nine. When involved in signal transduction, it can be phosphorylated. migration, which is consistent with report of Huang et al.[27].
CXCR4 is not only present on the cell membrane but also in the cyto- RhoA, a downstream molecule of the CXCL12/CXCR4 biological
plasm and nucleus. CXCR4 is expressed in most tissues and organs of axis, is mainly involved in the RhoA/ROCK pathway, which is closely
the body, which is widely involved in a variety of pathophysiological related to the activation, migration and adhesion of HSCs [28–30]. On
processes [14]. The chemokine CXCL12 interacts with its receptor the basis of above results, we concluded that curcumin may inhibit
CXCR4 to form a coupled molecular pair, also known as chemotaxis CXCL12/CXCR4 biological axis so that downstream RhoA/ROCK
axis, which is a very important signal transduction pathway in the pathway is downregulated, and thus activation and migration ability of
body. A variety of growth factors and cytokines act via this signal HSCs is suppressed. Therefore, curcumin could attenuate liver fibrosis
pathway to regulate the expression of target genes, generally involved by inhibiting CXCL12/CXCR4/RhoA axis.
in cell activation, proliferation, differentiation, migration, immune In this study, we used 50, 100 and 200 mg / kg curcumin as low,
modulation and other processes. CXCR4 combining with CXCL12 can medium and high doses to intervene rat model of liver fibrosis, and
cause the calcium signal to rise instantaneously to activate downstream used 20 and 40 μM curcumin as low and high concentration to inter-
signaling pathways, including RhoA/ROCK, ERK1/2, MAPK, JNK and vene HSC-T6 cell line, which are the most commonly used doses and
AKT and other signal pathways [15–18]. concentrations of curcumin in the animal and cell experiments [5].
Under normal circumstances, HSCs are in resting state. When liver is Curcumin 200 mg/kg/d for rats, converted to human use amount, is

606
L. Qin et al. Biomedicine & Pharmacotherapy 101 (2018) 599–607

equivalent to 31.75 mg/kg/d, which is roughly equal to the re- [10] A.S. Strimpakos, R.A. Sharma, Curcumin: preventive and therapeutic properties in
commended dose of health products of curcumin on the market. In laboratory studies and clinical trials, Antioxid. Redox Signal. 10 (3) (2008)
511–545.
previous studies, curcumin was administered orally by gavage at [11] Y. Zhao, X. Ma, J. Wang, X. He, Y. Hu, P. Zhang, R. Wang, R. Li, M. Gong, S. Luo,
1200 mg/kg/d in animal experiment [11], and curcumin was taken X. Xiao, Curcumin protects against CCl4-induced liver fibrosis in rats by inhibiting
orally at 8000 mg/kg/d for three months in patients with high-risk or HIF-1alpha through an ERK-dependent pathway, Molecules 19 (11) (2014)
18767–18780.
pre-malignant lesions [31], which exerted good effects with no ap- [12] Q. Yao, Y. Lin, X. Li, X. Shen, J. Wang, C. Tu, Curcumin ameliorates intrahepatic
parent adverse reactions, this owing to safe range of curcumin is large. angiogenesis and capillarization of the sinusoids in carbon tetrachloride-induced rat
Curcumin has great clinical application prospect, whereas application liver fibrosis, Toxicol. Lett. 222 (1) (2013) 72–82.
[13] E.W. Smith, A.M. Nevins, Q. Zhen, L. Yan, A.E. Getschman, S.L. Vankayala,
was limited by poor solubility and low bioavailability, therefore, im- M.T. Kemp, F.C. Peterson, R. Li, B.F. Volkman, Structure-based identification of
proving bioavailability of curcumin will be an important research field novel ligands targeting multiple sites within a chemokine-G protein–coupled re-
in the future. ceptor interface, J. Med. Chem. 59 (9) (2016).
[14] X. Sun, G. Cheng, M. Hao, J. Zheng, X. Zhou, J. Zhang, R.S. Taichman, K.J. Pienta,
J. Wang, CXCL12 / CXCR4 / CXCR7 chemokine axis and cancer progression, Cancer
5. Conclusion Metastasis Rev. 29 (4) (2010) 709–722.
[15] R.L. Lee, J. Westendorf, M.R. Gold, Differential role of reactive oxygen species in
In conclusion, the CXCL12/CXCR4 biological axis plays an im- the activation of mitogen-activated protein kinases and Akt by key receptors on B-
lymphocytes: CD40, the B cell antigen receptor, and CXCR4, J. Cell Commun.
portant role in the pathogenesis of liver fibrosis, which may be a target Signal. 1 (1) (2007) 33–43.
of curcumin in the treatment of liver fibrosis. In particular, curcumin [16] D.Y. Lu, C.H. Tang, W.L. Yeh, K.L. Wong, C.P. Lin, Y.H. Chen, C.H. Lai, Y.F. Chen,
could exert an anti-liver fibrotic effect by protecting against HSCs ac- Y.M. Leung, W.M. Fu, SDF-1alpha up-regulates interleukin-6 through CXCR4, PI3K/
Akt, ERK, and NF-kappaB-dependent pathway in microglia, Eur. J. Pharmacol. 613
tivation and migration via the CXCL12/CXCR4 biological axis. (1–3) (2009) 146–154.
[17] K. Princen, S. Hatse, K. Vermeire, E. De Clercq, D. Schols, Evaluation of SDF-1/
Conflicts of interest CXCR4-induced Ca2+ signaling by fluorometric imaging plate reader (FLIPR) and
flow cytometry, Cytometry A 51 (1) (2003) 35–45.
[18] J. Roland, B.J. Murphy, B. Ahr, V. Robert-Hebmann, V. Delauzun, K.E. Nye,
The authors declare no conflict of interest. C. Devaux, M. Biard-Piechaczyk, Role of the intracellular domains of CXCR4 in SDF-
1-mediated signaling, Blood 101 (2) (2003) 399–406.
[19] R.J. Phillips, M.D. Burdick, K. Hong, M.A. Lutz, L.A. Murray, Y.Y. Xue,
Acknowledgments J.A. Belperio, M.P. Keane, R.M. Strieter, Circulating fibrocytes traffic to the lungs in
response to CXCL12 and mediate fibrosis, J. Clin. Invest. 114 (3) (2004) 438–446.
This work was funded by the National Natural Science Foundation [20] H.K. Shu, Y. Yoon, S. Hong, K. Xu, H. Gao, C. Hao, E. Torres-Gonzalez, C. Nayra,
M. Rojas, H. Shim, Inhibition of the CXCL12/CXCR4-axis as preventive therapy for
of China (No. 81472002) and Natural Science Foundation of Guangxi
radiation-induced pulmonary fibrosis, PLoS One 8 (11) (2013) e79768.
(No. 2016GXNSFAA380312). [21] F. Tögel, J. Isaac, H.U. Zhuma, K. Weiss, C. Westenfelder, Renal SDF-1 signals
mobilization and homing of CXCR4-positive cells to the kidney after ischemic in-
References jury, Kidney Int. 67 (5) (2005) 1772–1784.
[22] A. Liepelt, F. Tacke, Stromal cell-derived factor-1 (SDF-1) as a target in liver dis-
eases, Am. J. Physiol. Gastrointest. Liver Physiol. 311 (2) (2016) G203–G209.
[1] V. Hernandez-Gea, S.L. Friedman, Pathogenesis of liver fibrosis, Annu. Rev. Pathol. [23] I. Sawitza, C. Kordes, S. Reister, D. Haussinger, The niche of stellate cells within rat
6 (2011) 425–456. liver, Hepatology 50 (5) (2009) 1617–1624.
[2] J. Zhao, Z. Zhang, Y. Luan, Z. Zou, Y. Sun, Y. Li, L. Jin, C. Zhou, J. Fu, B. Gao, Y. Fu, [24] F. Hong, A. Tuyama, T.F. Lee, J. Loke, R. Agarwal, X. Cheng, A. Garg, M.I. Fiel,
F.S. Wang, Pathological functions of interleukin-22 in chronic liver inflammation M. Schwartz, J. Walewski, A. Branch, A.D. Schecter, M.B. Bansal, Hepatic stellate
and fibrosis with hepatitis B virus infection by promoting T helper 17 cell recruit- cells express functional CXCR4: role in stromal cell-derived factor-1alpha-mediated
ment, Hepatology 59 (4) (2014) 1331–1342. stellate cell activation, Hepatology 49 (6) (2009) 2055–2067.
[3] K. Iwaisako, D.A. Brenner, T. Kisseleva, What’s new in liver fibrosis? The origin of [25] G.O. Elpek, Cellular and molecular mechanisms in the pathogenesis of liver fibrosis:
myofibroblasts in liver fibrosis, J. Gastroenterol. Hepatol. 27 (Suppl. 2) (2012) an update, World J. Gastroenterol. 20 (23) (2014) 7260–7276.
65–68. [26] C. Lu, W. Xu, S. Zheng, Nrf2 activation is required for curcumin to induce lipocyte
[4] U.E. Lee, S.L. Friedman, Mechanisms of hepatic fibrogenesis, Best Pract. Res. Clin. phenotype in hepatic stellate cells, Biomed. Pharmacother. 95 (2017) 1–10.
Gastroenterol. 25 (2) (2011) 195–206. [27] J.X. Huang, B.H. Zhu, D. He, L. Huang, K. Hu, B. Huang, Curcumine inhibits mi-
[5] A. Goel, A.B. Kunnumakkara, B.B. Aggarwal, Curcumin as "Curecumin": from gration and invasion of hepatic stellate cells by reducing MMP-2 expression and
kitchen to clinic, Biochem. Pharmacol. 75 (4) (2008) 787–809. activity, Zhonghua gan zang bing za zhi 17 (11) (2009) 835–838.
[6] M. Chevallier, S. Guerret, P. Chossegros, F. Gerard, J.A. Grimaud, A histological [28] L. Li, J.Y. Wang, C.Q. Yang, W. Jiang, Effect of RhoA on transforming growth factor
semiquantitative scoring system for evaluation of hepatic fibrosis in needle liver beta1-induced rat hepatic stellate cell migration, Liver Int. 32 (7) (2012)
biopsy specimens: comparison with morphometric studies, Hepatology 20 (2) 1093–1102.
(1994) 349–355. [29] M.M. Wyse, S. Goicoechea, R. Garcia-Mata, A.L. Nestor-Kalinoski, K.M. Eisenmann,
[7] C.T. Tu, J.S. Guo, M. Wang, J.Y. Wang, Antifibrotic activity of rofecoxib in vivo is mDia2 and CXCL12/CXCR4 chemokine signaling intersect to drive tumor cell
associated with reduced portal hypertension in rats with carbon tetrachloride-in- amoeboid morphological transitions, Biochem. Biophys. Res. Commun. 484 (2)
duced liver injury, J. Gastroenterol. Hepatol. 22 (6) (2007) 877–884. (2017) 255–261.
[8] B. Alvaro Mercadal, R. Imbert, I. Demeestere, C. Gervy, A. De Leener, Y. Englert, [30] C.R. Gandhi, Hepatic stellate cell activation and pro-fibrogenic signals, J. Hepatol.
S. Costagliola, A. Delbaere, AMH mutations with reduced in vitro bioactivity are 67 (5) (2017) 1104–1105.
related to premature ovarian insufficiency, Hum. Reprod. 30 (5) (2015) [31] A. Cheng, C. Hsu, J. Lin, M. Hsu, Y. Ho, T. Shen, J. Ko, J. Lin, B. Lin, W. MingShiang,
1196–1202. Phase I clinical trial of curcumin, a chemopreventive agent, in patients with high-
[9] A. Goel, S. Jhurani, B.B. Aggarwal, Multi-targeted therapy by curcumin: how spicy risk or pre-malignant lesions, Anticancer Res. 21 (4B) (2001) 2895–2900.
is it? Mol. Nutr. Food Res. 52 (9) (2008) 1010–1030.

607

You might also like