ArticlePDF Available

Taurocholate Induces Cyclooxygenase-2 Expression via the Sphingosine 1-phosphate Receptor 2 in a Human Cholangiocarcinoma Cell Line

Authors:

Abstract

Cholangiocarcinoma (CCA) is a rare, but highly malignant primary hepatobiliary cancer with a very poor prognosis and limited treatment options. Our recent studies reported that conjugated bile acids (CBAs) promote the invasive growth of CCA via activation of sphingosine 1-phosphate receptor 2 (S1PR2). Cyclooxygenase-2 (COX-2)-derived prostaglandin E2 (PGE2) is the most abundant prostaglandin in various human malignancies including CCA. Previous studies have indicated that COX-2 was highly expressed in CCA tissues, and the survival rate of CCA patients was negatively associated with high COX-2 expression levels. It has also been reported that CBAs induce COX-2 expression, whereas free bile acids inhibit COX-2 expression in CCA mouse models. However, the underlying cellular mechanisms and connection between S1PR2 and COX-2 expression in CCA cells have still not been fully elucidated. In the current study, we examined the role of S1PR2 in conjugated bile acid (taurocholate, (TCA))-induced COX-2 expression in a human HuCCT1 CCA cell line and further identified the potential underlying cellular mechanisms. The results indicated that TCA-induced invasive growth of human CCA cells was correlated with S1PR2-medated up-regulation of COX-2 expression and PGE2 production. Inhibition of S1PR2 activation with chemical antagonist (JTE-013) or down-regulation of S1PR2 expression with gene-specific shRNA not only reduced COX-2 expression, but also inhibited TCA-induced activation of EGFR and the ERK1/2/Akt-NF-κB signaling cascade. In conclusion, S1PR2 plays a critical role in TCA-induced COX-2 expression and CCA growth and may represent a novel therapeutic target for CCA.
JBC/2015/668277-R1 Bile acid and S1PR2-mediated signaling in CCA
1
Taurocholate Induces Cyclooxygenase-2 Expression via the Sphingosine 1-phosphate
Receptor 2 in a Human Cholangiocarcinoma Cell Line
Runping Liu1, 2, Xiaojiaoyang Li1, Lan Luo3, Xiaoyan Qiang1,2, Phillip B. Hylemon2,4,
Zhenzhou Jiang1, 3*, Luyong Zhang1, 3 * and Huiping Zhou2,4*,
1Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing,
Jiangsu, China
2Department of Microbiology and Immunology, Virginia Commonwealth University,
Richmond, Virginia, USA
3Jiangsu Center for Pharmacodynamics Research and Evaluation, Nanjing, Jiangsu, China
2McGuire Veterans Affairs Medical Center, Richmond, Virginia, USA
*Running title: Bile acid and S1PR2-mediated signaling in CCA
To whom correspondence should be addressed: Huiping Zhou, Department of Microbiology
and Immunology and McGuire Veterans Affairs Medical Center, Richmond, VA, USA 23298.
Tel:804-828-6817; Fax:804-828-0676; Email:hzhou@vcu.edu. Or Luyong Zhang, Jiangsu
Center for Drug Screening, China Pharmaceutical University, Nanjing, Jiangsu, China
210009. Tel: 86-25-83271500; Fax: 86-25-83271500; Email: lyzhang@cpu.edu.cn. Or
Zhenzhou Jiang, Jiangsu Center for Pharmacodynamics Research and Evaluation, China
Pharmaceutical University, Nanjing, Jiangsu, China 210009. Tel: 86-25-83271142; Fax: 86-
25-83271142; Email: beaglejiang@cpu.edu.cn.
Keywords: Bile acids, Sphingosine 1-phosphate, Biliary cancer, NF-B, EGFR
Background: Cyclooxygenase-2 (COX-2)
and sphingosine 1-phosphate receptor 2
(S1PR2) are highly expressed in human
cholangiocarcinoma (CCA) and taurocholate
(TCA) promotes CCA cell growth via S1PR2.
Results: TCA-mediated activation of S1PR2
contributed to COX-2 expression and CCA
cell growth.
Conclusion: S1PR2 plays a critical role in
TCA-induced COX-2 expression and CCA
growth.
Significance: S1PR2 represents a novel
therapeutic target for CCA.
ABSTRACT
Cholangiocarcinoma (CCA) is a rare, but
highly malignant primary hepatobiliary
cancer with a very poor prognosis and
limited treatment options. Our recent studies
reported that conjugated bile acids (CBAs)
promote the invasive growth of CCA via
activation of sphingosine 1-phosphate
receptor 2 (S1PR2). Cyclooxygenase-2
(COX-2)-derived prostaglandin E2 (PGE2) is
the most abundant prostaglandin in various
human malignancies including CCA.
Previous studies have indicated that COX-2
was highly expressed in CCA tissues, and
the survival rate of CCA patients was
negatively associated with high COX-2
expression levels. It has also been reported
that CBAs induce COX-2 expression, while
free bile acids inhibit COX-2 expression in
CCA mouse models. However, the
underlying cellular mechanisms and
http://www.jbc.org/cgi/doi/10.1074/jbc.M115.668277The latest version is at
JBC Papers in Press. Published on October 30, 2015 as Manuscript M115.668277
Copyright 2015 by The American Society for Biochemistry and Molecular Biology, Inc.
at VIVA, VA Commonwealth Univ on December 15, 2015http://www.jbc.org/Downloaded from
JBC/2015/668277-R1 Bile acid and S1PR2-mediated signaling in CCA
2
connection between S1PR2 and COX-2
expression in CCA cells have still not been
fully elucidated. In the current study, we
examined the role of S1PR2 in conjugated
bile acid (taurocholate, TCA)-induced COX-
2 expression in a human HuCCT1 CCA cell
line and further identified the potential
underlying cellular mechanisms. The results
indicated that TCA-induced invasive growth
of human CCA cells was correlated with
S1PR2-medated upregulation of COX-2
expression and PGE2 production. Inhibition
of S1PR2 activation with chemical
antagonist (JTE-013) or downregulation of
S1PR2 expression with gene-specific
shRNA not only reduced COX-2 expression,
but also inhibited TCA-induced activation of
EGFR and ERK1/2/Akt-NF-B signaling
cascade. Conclusion: S1PR2 plays a critical
role in TCA-induced COX-2 expression and
CCA growth and may represent a novel
therapeutic target for CCA.
Cholangiocarcinoma (CCA) is the most
common fatal biliary malignancy and
accounts to 10% to 15% of primary hepatic
cancer originating from the epithelial lining of
bile ducts (1-4). Recent epidemiological
studies indicate that the incidence of CCA is
increasing worldwide, although the risk
factors differ geographically (3). Unlike other
gastrointestinal carcinomas, CCA is
relatively rare and has not been extensively
studied. Certain environment-linked risk
factors, such as liver fluke infection and toxic
chemicals, may play a critical role in the
disease development and progression of
CCA in some patients. Recent studies have
identified several interacting molecular
pathways as vital promoting factors in the
development of CCA , such as cholestasis,
chronic inflammation, oxidative stress, and
dysregulation of the cell injury repair
processes (1-3,5,6). However, the role of
conjugated bile acids (CBA) in CCA
pathogenesis remain largely unknown.
The current available therapeutic options are
mainly limited to surgical resection or liver
transplantation (7).
Cyclooxygenase-2 (COX-2)-derived
prostaglandins (PG) have been implicated in
various carcinogenesis including
cholangiocarcinogenesis (8,9).
Overexpression of COX-2 promoted
cholangiocarcinoma cell growth and survival,
while depletion or pharmacological inhibition
of COX-2 reduced CCA cell proliferation (9-
11). COX-2-derived PGE2 is the most
abundant prostanoid and mediates many
physiological and pathological effects via
activating different G protein coupled
receptors (GPCRs) EP1-4. Both enhanced
expression of COX-2 and increased
production of PGE2 have been associated
with various cancers including CCA (12-14).
Occurrence of CCA is often associated with
chronic cholestasis, and the accumulation of
bile acids, especially conjugated bile acids,
in the liver and bile duct is linked to the
disease progression of CCA. It also has
been reported that CBAs induce COX-2
expression via the epidermal growth factor
receptor (EGFR) and NF-B pathways in
CCA cells (15,16). In CCA patients, the
composition of taurine- and glycine-
conjugated cholic acids is significantly
increased when compared to patients with
benign biliary diseases (17). Our recent
studies showed that CBAs promoted the
proliferation and invasion of both rodent and
human CCA cells through the activation of
sphingosine 1-phosphate receptor 2
(S1PR2)(18). S1PR2 was the predominant
S1PRs expressed in CCA cell and tissues.
Although several recent studies reported
that S1PR2-mediated signaling pathways
were involved in promoting tumor growth and
at VIVA, VA Commonwealth Univ on December 15, 2015http://www.jbc.org/Downloaded from
JBC/2015/668277-R1 Bile acid and S1PR2-mediated signaling in CCA
3
progression, the underlying
cellular/molecular mechanisms remain to be
elucidated (19). Whether conjugated bile
acid-mediated activation of S1PR2
contributes to the upregulation of COX-2
expression and PGE2 production in CCA
cells has not been determined and is the
major focus of this study.
In the current study, we examined the
effect of TCA on COX-2 expression and
elucidated the role of S1PR2 in TCA-induced
COX-2 expression and further identified
potential signaling pathways in human CCA
cells.
EXPERIMENTAL PROCEDURES
Materials-S1P and JTE-013 were
purchased from Cayman Chemical (Boston,
MA, USA). pcDNA3.1-3xHA-tagged human
S1PR2 (N-terminus) was obtained from
UMR cDNA Resource Center (Rolla, MO);
Celecoxib, U0126 and MK-2206 were
purchased from Selleck Chemicals (Boston,
MA, USA). Taurocholate (TCA) and dimethyl
sulfoxide (DMSO), monoclonal anti-HA
antibody were obtained from Sigma-Aldrich
(St. Louis, MO, USA). BD Biocoat Matrigel
Invasion Chamber was purchased from BD
biosciences (USA). A QIAzol lysis reagent
was from QIAGEN Sciences (Valencia, CA,
USA). Thermo Hygreen supermix reagent
was purchased from Thermo (Waltham, MA,
USA). RIPA lysis buffer and nuclear protein
isolation kit were obtained from Beyotime
(Nanjing, Jiangsu, China). Bio-Rad protein
assay reagent and 4x sample buffer were
from BIO-RAD (Hercules, CA, USA). Primary
antibodies against COX-2, p-ERK1/2, ERK1,
ERK2, p-Akt, Akt, p-IKKα/β, IKKα/β, p-NF-κB
p65, NF-κB p65, p-EGFR, EGFR, β-actin
and lamin B, as well as HRP-conjugated
secondary antibodies for Western blot were
from Santa Cruz Biotechnology (Santa Cruz,
CA, USA). Alexa Fluor® 488 conjugated
secondary antibodies for
immunofluorescence, High-capacity cDNA
Reverse Transcription Kit, Lipofectamine
2000, cell cultures medium and supplement
components were all from Life Technologies
(Grand Island, NY, USA).
Cell culture and treatment-The human
HuCCT1 CCA cell line was obtained from
Guangzhou Jenniobio Biotechnology Co.,
Ltd (Guangzhou, China) and cultured in
RPMI1640 medium, supplemented with 10%
FBS, 2 mM of L-glutamine, and 50 μg/mL of
gentamicin. TCA was dissolved in serum-
free culture medium. S1P, JTE-013,
Celecoxib, U0126, MK-2206 were prepared
in DMSO to desired concentrations. Drugs
were delivered by directly adding into cell
culture medium. For the cell invasion assay,
drugs were added into medium in the upper
chamber of the matrigel pre-coated transwell.
Transfection of shRNA for down-
regulating S1PR2-The stem loop sequences
of short hairpin RNA (shRNA) specifically
targeting human S1PR2 and the scrambled
control shRNA plasmid were gifts from Dr.
Murthy Karnam (Department of Physiology
and Biophysics, Virginia Commonwealth
University, Richmond, VA). HuCCT1 cells
were transiently transfected with the control
shRNA or S1PR2 shRNA plasmids using
Lipofectamine 2000, as described previously
(20).
Western blot analysis-Total cell proteins
were lysed with RIPA lysis buffer (Beyotime,
China). Nuclear and cytosol proteins were
isolated using a nuclear protein isolation Kit
(Beyotime, China). 50 µg of the proteins
were resolved on a 10% SDS-PAGE gel and
transferred to Nitrocellulose membranes.
Membranes were blocked for 1 h at room
temperature with 5% BSA in TBS buffer and
at VIVA, VA Commonwealth Univ on December 15, 2015http://www.jbc.org/Downloaded from
JBC/2015/668277-R1 Bile acid and S1PR2-mediated signaling in CCA
4
then incubated with specific primary
antibodies for overnight at 4C.
Immunoreactive bands were detected using
horseradish peroxidaseconjugated
secondary antibodies and ECL reagents
(Thermo, USA). Images were recorded
using Bio-Rad ChemiDoc XRS imaging
system (Bio-Rad, USA) and further analyze
using Quantity One (Bio-Rad, USA).
RNA isolation and real-time RT-PCR-
Total cellular RNA was isolated using TRIzol
Reagent (QIAGEN, Valencia, CA, USA) and
reverse transcribed into first-strand cDNA
using the High-capacity cDNA Reverse
Transcription Kit (Life Technologies) as
described previously(18). IL-6, TNF-α,
matrix metalloproteinase-2 (MMP-2) and
MMP-9 mRNA levels were determined by
real-time PCR using Thermo Hygreen
supermix reagents and normalized using
GAPDH as an internal control as described
previously(18,20). Primer sequences will be
provided upon request.
Enzyme-linked Immunosorbent Assays
(ELISA) of PGE2-Following different
treatment, cell culture medium was collected
and centrifuged to remove cell debris. PGE2
levels in the supernatant were determined
using a high sensitivity ELISA kit (Enzo Life
Sciences, USA) according to the
manufacturer’s instruction. Safire2
microplate reader (Tecan, Switzerland) was
used to detect the absorbance at 405 nM.
Cell invasion assay-HuCCT1 cells and
HuCCT1 cells transfected with control
shRNA or S1PR2 shRNA were seeded in the
upper chamber of the BD Biocoat Matrigel
Invasion Chamber (BD Bioscience, USA)
and cultured in complete medium with
1%FBS. Cells were pretreated with JTE-013
(10 μM) or Celecoxib (40 μM) or DMSO
(vehicle control) for 1 h, then treated with
TCA (100 μM), or S1P (100 nM), or DMSO
and incubated at 37°C for 24 h. At the end of
treatment, noninvasive cells were removed
from the top surface of upper chamber
membrane. Invasive cells that migrated to
lower surface were fixed with 3.7%
paraformaldehyde and stained with 0.1%
crystal violet solution. Invasive cells were
counted and the invasion index was
analyzed as previously described (18).
Immunofluorescence staining of NF-
B p65-
HuCCT1 cells were seeded into 96-well
plates with glass bottom (Matrical bioscience,
USA) and cultured overnight in serum-free
medium. After treatment with TCA or S1P
with or without JTE-013, cells were fixed with
3.7% formaldehyde in PBS for 45 min,
permeabilized with 0.1% Triton-X-100 for 3
min, and blocked with 3% BSA for 45 min.
Cells were then incubated with rabbit anti-
NF-B p65 antibody (1:100 dilution)
overnight at 4C, followed by incubation with
Alexa Fluor-488 labeled goat anti-rabbit
secondary antibody for 30 min at RT. After
washing with PBS, staining of the
intracellular and nuclear NF-B p65 was
visualized under an Olympus IX81 motorized
inverted fluorescence microscope with a 60
x oil objective (Olympus, Japan).
Detection of S1PR2 internalization-HEK293
cells were plated into 6-well plates with
coverslips. Cells were cultured overnight to
60% confluence and transfected with 1 µg of
plasmid DNA (pcDNA3.1-3xHA-tagged
S1PR2) using PolyJetTMDNA In Vitro
Transfection Reagent according to
manufactural instruction. After 24 h of
transfection, cells were cultured in serum-
free medium for overnight, then stimulated
with TCA (100 µM), or S1P (100 nM) or
vehicle control for 10 mins. Cells were fixed
with 3.7% formaldehyde in PBS for 15 mins.
at VIVA, VA Commonwealth Univ on December 15, 2015http://www.jbc.org/Downloaded from
JBC/2015/668277-R1 Bile acid and S1PR2-mediated signaling in CCA
5
The expression and localization of S1PR2
were monitored using immunofluorescence
staining with anti-HA antibody and Alexa
Fluor-488 labeled goat anti-mouse
secondary antibody. The images were
recorded using Zeiss Axio Imager A1
fluorescence microscope with a 100 x oil
objective (Jenamed, Carl Zeiss, Germany).
Statistical analysis-All data were expressed
as the mean ± standard error of the mean.
One-way ANOVA and student’s t-test were
employed to analyze the differences
between sets of data. Statistical analysis
was performed using Prism 5.0 (GraphPad,
San Diego, CA) as described previously
(18,20). A value of p<0.05 was considered
statistically significant.
RESULTS
TCA induces COX-2 expression and
chronic inflammation via activation of
S1PR2- COX-2 is a key enzyme involved in
production of prostaglandins and has been
implicated in various cell transformations
including cholangiocytes (21-25). Previous
studies reported that CBAs induced COX-2
expression and promoted growth in human
CCA cells in culture (15,16). Our recent
studies showed that conjugated bile acid
(TCA) promoted invasive cell growth via
activation of S1PR2 in both rat and human
CCA cell lines (18). However, whether
activation of S1PR2 also contributes to
conjugated bile acid-mediated expression of
COX-2 and prostaglandin (PG) synthesis
remained unknown. Therefore, we first
examined the effect of TCA on COX-2
expression in human HuCCT1 cells. As
shown in Fig.1A-B, TCA significantly
increased COX-2 protein levels in a time-
dependent manner, peaking at 8 h. TCA-
induced COX-2 expression was also dose-
dependent (Fig.1C-D). In order to define the
role of S1PR2 in TCA-induced COX-2
expression, a chemical antagonist of S1PR2,
JTE-013, was used. As shown in Fig.2A-B,
both S1P- and TCA-induced COX-2
expression was markedly inhibited by JTE-
013. Similarly, as shown in Fig. 2C-E, down-
regulation of S1PR2 using a S1PR2 shRNA
significantly blocked TCA-induced COX-2
expression.
PGE2, the final enzymatic product of
COX-2, has been implicated in various
chronic inflammation-related cell
transformation including CCA (14). Previous
study from Dr. Wu’s lab reported that COX-
2-derived PGE2 regulates human CCA cell
growth (8). Our study also showed that TCA
significantly increased PGE2 production in
HuCCT1 cells (Fig. 2F). Furthermore,
TCA-induced PGE2 production was
significantly inhibited by JTE-013. However,
both S1P and TCA had no effect on COX-1
protein expression in HuCCT1 cells at
protein level (Fig.3A-B). In addition, we
examined the effect of S1P and TCA on the
mRNA expression of two inflammatory
cytokines, IL-6 and TNF-α. As shown in Fig.
3C-D, both S1P and TCA significantly
induced mRNA expression of IL-6 and TNF-
α, which was attenuated by JTE-013. These
results suggest that S1PR2 may be a key
regulator in TCA-mediated COX-2
expression, PGE2 production and chronic
inflammation.
TCA enhances invasion of HuCCT1
cells through S1PR2-dependent COX-2
activation-Although several studies have
reported that CBAs promote CCA cell growth
and invasion, the contribution of TCA-
induced S1PR2 activation and COX-2
expression on CCA cell invasion has not
been examined (15,26,27). Our recent
studies showed that S1P- and TCA-induced
at VIVA, VA Commonwealth Univ on December 15, 2015http://www.jbc.org/Downloaded from
JBC/2015/668277-R1 Bile acid and S1PR2-mediated signaling in CCA
6
invasive growth was blocked by inhibition of
S1PR2 activation in rat CCA cells (18). To
further verify the effect of TCA on the
invasion of human CCA cells, HuCCT1 cells
were plated in the upper chamber of Matrigel
pre-coated transwell inserts and treated with
TCA (100 μM), S1P (100 nM, as a positive
control), or vehicle control, in the presence
or absence of JTE-013 (10 μM). Similar to
our findings in rat CCA cells, the invasion
indexes of TCA and S1P groups were
significantly increased in human HuCCT1
cells, which were significantly inhibited by
JTE-013 (Fig. 4A-B). Down-regulation of
S1PR2 expression using a shRNA also
blocked TCA-induced increase of invasion
index (Fig. 4C-D). In order to further define
the role of TCA-induced S1PR2 activation
and COX-2 expression in CCA cell invasion,
a specific chemical inhibitor of COX-2,
Celecoxib, was used. As shown in Fig. 4E-F,
in the presence of Celecoxib (40 µM), TCA-
induced increase of the invasion index was
also markedly inhibited in HuCCT1 cells.
These results suggest that TCA promotes
CCA cell invasion via activation of S1PR2,
increased COX-2 expression, and synthesis
of PGE2.
TCA activates NF-
B via activation of
S1PR2- The transcription factor NF-B is a
well-known evolutionarily conserved
signaling molecule with many biological
activities. NF-B can be activated by cell
signaling pathways which activate IB
kinase (IKKα/β). Activated IKKα/β further
phosphorylates IB and leads to degradation
of IB and nuclear translocation of NF-B
p65 (28). In addition, IKKα/β are also
involved in the direct phosphorylation of NF-
B p65. Phosphorylation of NF-B p65 not
only enhances the efficiency of DNA binding,
but also provides an additional interaction
site for transcriptional co-activator CBP/p300
(29). Previous studies have shown that bile
acids activate NF-B signaling pathways in
cancer cells (15,30). In order to determine
whether TCA also can activate NF-B
signaling pathways, we examined the
protein levels of phosphorylated IKKα/β (p-
IKKα/β) and phosphorylated NF-B p65 (p-
NF-B p65). As shown in Fig. 5, TCA
significantly increased protein levels of p-
IKKα/β and p-NF-B p65 in both a time-
dependent and dose-dependent manner. In
addition, TCA significantly increased nuclear
translocation of NF-B p65 (Fig. 6). To
further determine whether the NF-B
activation depends on TCA-mediated
activation of S1PR2, we examined the effect
of JTE-013 on TCA-induced activation of NF-
B in HuCCT1 cells. As shown in Fig. 7 and
Fig.8, JTE-013 not only inhibited the TCA-
induced increase of p-IKKα/β and p-NF-B
p65, but also blocked TCA-induced nuclear
translocation of NF-B p65.
TCA activates EGF receptor via S1PR2-
Previous studies reported that bile acids
transactivate the EGF receptor (EGFR) in
cholangiocytes via a TGF-alpha-dependent
mechanism (31). Bile acid-induced
activation of EGFR/ERK1/2 also induced
COX-2 expression in CCA cells (16). In
human CCA, EGFR expression level is
significantly associated with tumor
progression and poor prognosis (32). To
further understand the relationship between
S1PR2 and EGFR/ERK1/2 activation, we
first examined the effect of TCA on the level
of EGFR phosphorylation in HuCCT1 cells.
As shown in Fig. 9, TCA rapidly and dose-
dependently induced phosphorylation of
EGFR. The presence of JTE-013 or
downregulation of S1PR2 using gene-
specific shRNA completely blocked TCA-
induced activation of EGFR, but had no
effect on total protein expression (Fig.10).
at VIVA, VA Commonwealth Univ on December 15, 2015http://www.jbc.org/Downloaded from
JBC/2015/668277-R1 Bile acid and S1PR2-mediated signaling in CCA
7
One of the most common mechanisms of
GPCR-mediated activation of EGFR is the
release of the mature EGFR ligands from
transmembrane precursors by matrix
metalloproteinases (MMPs). MMP2 and
MMP9 are members of the MMP family
reported to mediate EGFR activation
through GPCRs (33). We further examined
whether activation of S1PR2 had any effect
on the expression of MMP2 and MMP9 in
HuCCT1 cells. As shown in Fig.11, both
TCA and S1P significantly increased the
mRNA levels as well as the protein levels of
MMP2 and MMP9, but these increases were
blocked by JTE-013.
Role of S1PR2 in TCA-mediated ERK1/2
and Akt activation in HuCCT1 cells-Bile acid-
induced EGFR activation is linked to the
activation of ERK1/2 and Akt (34). Our
previous studies also showed that TCA
induced ERK1/2 and AKt activation via
S1PR2 (18,20). To confirm the role of
S1PR2 activation in TCA-induced ERK1/2
and Akt activation in HuCCT1 cells, we
examined the protein levels of p-ERK1/2 and
p-Akt in TCA-treated HuCCT1 cells. As
shown in Fig.12A-B, TCA rapidly induced
ERK1/2 and Akt activation in a time-
dependent manner, peaking at 4 h. Similar
to TCA-induced EGFR activation, TCA also
dose-dependently induced ERK1/2 and Akt
activation (Fig.12C-D). Furthermore, both
TCA- and S1P-induced ERK1/2 and Akt
activation was markedly inhibited by JTE-
013 (Fig.12E-F).
Effect of TCA-induced ERK1/2 and Akt
activation on COX-2 expression-It has been
well documented that COX-2 expression is
regulated by ERK1/2 and Akt in various cell
types (8,35-38). To further delineate the role
of S1PR2-mediated ERK1/2 and Akt
activation in TCA-induced COX-2
expression in HuCCT1 cells, selective
chemical inhibitors of MEK1/2 and Akt were
used. As shown in Fig.13A-B, in the
presence of U0126 (10 µM), both TCA- and
S1P-induced ERK1/2 activation was
completely blocked. TCA- and S1P-induced
COX-2 expression was also significantly
inhibited by U0126 (Fig. 14A-B). Similarly,
both TCA- and S1P-induced Akt activation
and COX-2 expression were markedly
inhibited by MK2206, a highly selective
inhibitor of Akt1/2/3 (Figs.13C-D and 14C-D).
S1P and TCA induce S1PR2 internalization-
Our previous studies and current study
strongly indicated that TCA activates S1PR2.
In order to delineate whether TCA is able to
directly interact with S1PR2, we examined
the effect of TCA on S1PR2 internalization in
HEK293 cells. Cells were transfected with
HA-tagged human S1PR2 and then treated
with TCA (100 μM) or S1P (100 nM) or
vehicle control for 10 mins at 37°C. To stop
the ligand-mediated internalization process,
cells were fixed with 3.7% formaldehyde at
4°C. The expression and localization of
HA-S1PR2 were monitored by
immunofluorescence staining using specific
anti-HA antibody. As shown in Fig.15, both
TCA and S1P rapidly induced S1PR2
internalization. These results suggest that
TCA directly interact with S1PR2 as
previously suggest from molecular modeling
experiments (20).
DISCUSSION
CCA is very difficult to diagnose at
early stages due to its anatomic location.
Delayed diagnosis precludes many CCA
patients from surgical intervention, the
treatment option with the best prognosis (1).
Concurrently, the outcomes of current
conventional chemotherapies and radiation
are also very poor (3). Therefore,
at VIVA, VA Commonwealth Univ on December 15, 2015http://www.jbc.org/Downloaded from
JBC/2015/668277-R1 Bile acid and S1PR2-mediated signaling in CCA
8
identification of novel therapeutic targets for
CCA is extremely urgent. Although
significant amount of effort has been put in
understanding CCA development and
progression during last two decades, the
etiopathogenesis of this cancer are still
largely unknown.
Accumulation of bile acids in bile
ducts and liver is associated with an
increased incidence of CCA (39). However,
the exact role of bile acids in
cholangiocarcinogenesis is still unclear.
Several studies suggested that bile acid-
mediated activation of the nuclear receptor
FXR (farnesoid X receptor) and the GPCR,
TGR5, might be involved in regulating the
proliferation and survival of cholangiocytes
(40). However, our recent studies also
identified that conjugated bile acid-mediated
activation of S1PR2 plays a critical role in
invasive growth of CCA cells in culture (18).
Impaired bile flow is often accompanied with
chronic inflammation and high basal level of
COX-2 expression in the biliary tracts (1),
while the underlying cellular mechanisms
are still not fully identified. Inflammation is
a key stimulator of carcinogenesis,
promoting tumor cell invasion and migration
in various cancers including CCA (41-43).
Recently, there has been significant
progress in identifying the critical role of
COX-2 plays in tumorgenesis of various
cancers including CCA (4,9,44), and PGE2,
the enzymatic product of COX-2, has been
identified as a strong promoter for
mitogenesis, cell proliferation, and invasion
of CCA (9,45,46).
The principal findings of the current
study directly link conjugated bile acid-
mediated activation of S1PR2 to both COX-
2 expression and PGE2 production in human
CCA cells. The results indicated that TCA-
induced activation of S1PR2 is responsible
for activation of EGFR/ ERK1/2/Akt signaling
pathways, which further activates NF-B and
increases COX-2 expression in CCA cells. A
schematic diagram of potential pathways
involved in TCA-induced COX-2 expression
in CCA cells is illustrated in Fig.16. The
current results provide novel mechanisms
underlying conjugated bile acid-mediated
chronic inflammation in CCA.
S1PR2 is one of the five S1PRs
identified so far and has been implicated in
various physiological and pathological
conditions (19). A recent study reported
that extracellular S1P induced COX-2
expression and PGE2 production via
activation of S1PR2, as well as subsequent
activation of ERK1/2 in rat renal mesangial
cells (47). It also has been reported that
S1PR2 signaling is involved in macrophage
retention and inflammatory secretions in the
atherosclerotic lesions (48). However, the
role of S1PR2 in tumor growth and
progression is controversial in different types
of cancers, probably due to its coupling to
different G proteins in different types of cells
(19). In this study, we demonstrated that
TCA dose-dependently induced expression
of COX-2 via activation of S1PR2 in human
HuCCT1 cells. The protein level of COX-2
was significantly increased even at a
concentration of 25 µM (Fig.1). Inhibition of
S1PR2 activation by a chemical antagonist
or down-regulation of S1PR2 with gene-
specific shRNA completely blocked TCA-
induced COX-2 expression and CCA cell
invasion (Figs.2 and 4). Previous studies
reported that bile acids induced COX-2
expression via activation of EGFR/MAPK
signaling pathways in CCA cells (16). We
also have reported that bile acids activated
MAPK via EGFR in hepatocytes (34). In this
study, we found that TCA also significantly
at VIVA, VA Commonwealth Univ on December 15, 2015http://www.jbc.org/Downloaded from
JBC/2015/668277-R1 Bile acid and S1PR2-mediated signaling in CCA
9
activated EGFR/MAPK signaling pathways
in HuCCT1 cells (Fig.10). A recent study
reported that taurolithocholic acid (TLCA)
promotes CCA cell growth through the
activation of the muscarinic acetylcholine
receptor and the EGFR/ERK1/2 signaling
pathway(49). However, the concentration of
TLCA is relative low compared to conjugated
primary bile acids in CCA as cholestasis
decreases the formation of secondary bile
acids. Clinical studies have shown that the
concentrations of conjugated primary bile
acids (cholic and chenodeoxycholic acid) are
significantly higher in the bile of CCA
patients than in those with the benign
disease (17). In addition, we have reported
that S1PR2 can be activated by TCA, and
S1PR2 is the predominant S1PR expressed
in CCA cells and human tumor tissues
(18,20). Our current study further confirmed
that TCA rapidly induced S1PR2
internalization, indicating that TCA may
directly activates S1PR2 (Fig.15). Therefore,
S1PR2 appears to play a key role in
conjugated bile acid-mediated COX-2
expression and PGE2 synthesis in CCA cells.
It has been shown that activation of
Src kinase or GPCR-mediated activation of
Gi protein can increase the expression of
MMPs, which play an important role in
forming active EGFR ligands (16,31,33,46).
Similarly, in this study, we also found that
both S1P and TCA significantly increased
the expression of MMP2 and MMP9 in
HuCCT1 cells, which was markedly inhibited
by JTE-013 (Fig. 11). These results suggest
that TCA-induced activation of S1PR2
contributes to the increase of MMPs and
subsequent activation of EGFR/ERK/Akt
signaling pathways. In addition, S1PR2 can
also directly activate ERK and Akt directly
through Gαi (18). It has been well
established that activation of ERK/Akt
signaling pathways is essential for activation
of NF-B. NF-B is a key transcription factor
involved in the regulation of various
inflammatory factors including COX-2.
Consistent with previous reports, we also
found that TCA-induced
S1PR2/EGFR/ERK/Akt activation was linked
to NF-B activation in CCA cells (Figs 5-10),
and JTE-013 inhibited not only TCA-/S1P-
induced NF-B activation, but also the
nuclear translocation of NF-B. By using
specific chemical inhibitors of MEK1 and Akt,
we further demonstrated that TCA-induced
activation of S1PR2/ERK1/2/Akt is
responsible for COX-2 expression in CCA
cells (Fig.14). Inhibition of ERK1/2/Akt also
blocked TCA-induced nuclear translocation
of NF-kB (data not shown). Furthermore,
S1P- and TCA-induced expression of
inflammatory cytokines (IL-6 and TNF-α)
was also blocked by JTE-013 (Fig.3).
In summary, as illustrated in Fig.16,
we determined that S1PR2 is a key player in
TCA-induced COX-2 expression in HuCCT1
cells. Activation of S1PR2 by TCA activates
ERK1/2 and Akt signaling pathways via G
proteins and EGFR. Activated ERK1/2 and
Akt further leads to the activation of NF-B
and subsequent COX-2 expression and
PGE2 synthesis. Our study provides further
evidence indicating that S1PR2 represents a
novel therapeutic target for CCA.
Acknowledgement: This study was supported by National Natural Science Fondation of
China Grants (81320108029 to LZ; 81070245, 81270489 to HZ); China National 12th Five-
year Plan Major Project of Science and Technologies for Signifiant Drug Discovery”
at VIVA, VA Commonwealth Univ on December 15, 2015http://www.jbc.org/Downloaded from
JBC/2015/668277-R1 Bile acid and S1PR2-mediated signaling in CCA
10
(2012ZX09504001-001 to LZ), National Institutes of Heath Grant R01 DK-057543-11 (to PBH
and HZ), VA Merit Awards (to HZ); VCU Massey Cancer Pilot grant (A35362 to HZ).
Conflict of interest: The authors declare no competing financial interest with the contents of
this article.
Author contributions: RL, PBH, ZJ, LZ and HZ conceived the original ideas, designed the
study, analyzed the data and wrote the manuscript; RL, XL, XQ and LL carried out the
experiments and data analysis. ZJ, LZ and HZ contributed equally.
References
1. Gores, G. J. (2003) Cholangiocarcinoma: current concepts and insights. Hepatology
(Baltimore, Md.) 37, 961-969
2. Sirica, A. E. (2005) Cholangiocarcinoma: molecular targeting strategies for
chemoprevention and therapy. Hepatology (Baltimore, Md.) 41, 5-15
3. Al-Bahrani, R., Abuetabh, Y., Zeitouni, N., and Sergi, C. (2013) Cholangiocarcinoma: risk
factors, environmental influences and oncogenesis. Annals of clinical and laboratory science
43, 195-210
4. Yeh, C. N., Chiang, K. C., Juang, H. H., JH, S. P., Yu, C. S., Lin, K. J., Yeh, T. S., and
Jan, Y. Y. (2013) Reappraisal of the therapeutic role of celecoxib in cholangiocarcinoma. PloS
one 8, e69928
5. Kiguchi, K. (2014) Molecular aspects of cholangiocarcinoma. Journal of hepato-biliary-
pancreatic sciences 21, 371-379
6. Aleksandrova, K., Boeing, H., Nothlings, U., Jenab, M., Fedirko, V., Kaaks, R., Lukanova,
A., Trichopoulou, A., Trichopoulos, D., Boffetta, P., Trepo, E., Westhpal, S., Duarte-Salles, T.,
Stepien, M., Overvad, K., Tjonneland, A., Halkjaer, J., Boutron-Ruault, M. C., Dossus, L.,
Racine, A., Lagiou, P., Bamia, C., Benetou, V., Agnoli, C., Palli, D., Panico, S., Tumino, R.,
Vineis, P., Bueno-de-Mesquita, B., Peeters, P. H., Gram, I. T., Lund, E., Weiderpass, E.,
Quiros, J. R., Agudo, A., Sanchez, M. J., Gavrila, D., Barricarte, A., Dorronsoro, M., Ohlsson,
B., Lindkvist, B., Johansson, A., Sund, M., Khaw, K. T., Wareham, N., Travis, R. C., Riboli, E.,
and Pischon, T. (2014) Inflammatory and metabolic biomarkers and risk of liver and biliary
tract cancer. Hepatology (Baltimore, Md.) 60, 858-871
7. Blechacz, B., Komuta, M., Roskams, T., and Gores, G. J. (2011) Clinical diagnosis and
staging of cholangiocarcinoma. Nature reviews. Gastroenterology & hepatology 8, 512-522
8. Han, C., and Wu, T. (2005) Cyclooxygenase-2-derived prostaglandin E2 promotes human
cholangiocarcinoma cell growth and invasion through EP1 receptor-mediated activation of the
epidermal growth factor receptor and Akt. The Journal of biological chemistry 280, 24053-
24063
9. Wu, T. (2005) Cyclooxygenase-2 and prostaglandin signaling in cholangiocarcinoma.
Biochimica et biophysica acta 1755, 135-150
10. Nzeako, U. C., Guicciardi, M. E., Yoon, J. H., Bronk, S. F., and Gores, G. J. (2002) COX-
2 inhibits Fas-mediated apoptosis in cholangiocarcinoma cells. Hepatology (Baltimore, Md.)
35, 552-559
at VIVA, VA Commonwealth Univ on December 15, 2015http://www.jbc.org/Downloaded from
JBC/2015/668277-R1 Bile acid and S1PR2-mediated signaling in CCA
11
11. Sirica, A. E., Lai, G. H., Endo, K., Zhang, Z., and Yoon, B. I. (2002) Cyclooxygenase-2
and ERBB-2 in cholangiocarcinoma: potential therapeutic targets. Seminars in liver disease
22, 303-313
12. Reader, J., Holt, D., and Fulton, A. (2011) Prostaglandin E2 EP receptors as therapeutic
targets in breast cancer. Cancer metastasis reviews 30, 449-463
13. Surh, I., Rundhaug, J., Pavone, A., Mikulec, C., Abel, E., and Fischer, S. M. (2011)
Upregulation of the EP1 receptor for prostaglandin E2 promotes skin tumor progression.
Molecular carcinogenesis 50, 458-468
14. Jongthawin, J., Chusorn, P., Techasen, A., Loilome, W., Boonmars, T., Thanan, R.,
Puapairoj, A., Khuntikeo, N., Tassaneeyakul, W., Yongvanit, P., and Namwat, N. (2014) PGE2
signaling and its biosynthesis-related enzymes in cholangiocarcinoma progression. Tumour
biology : the journal of the International Society for Oncodevelopmental Biology and Medicine
35, 8051-8064
15. Dai, J., Wang, H., Dong, Y., Zhang, Y., and Wang, J. (2013) Bile acids affect the growth
of human cholangiocarcinoma via NF-kB pathway. Cancer investigation 31, 111-120
16. Yoon, J. H., Higuchi, H., Werneburg, N. W., Kaufmann, S. H., and Gores, G. J. (2002)
Bile acids induce cyclooxygenase-2 expression via the epidermal growth factor receptor in a
human cholangiocarcinoma cell line. Gastroenterology 122, 985-993
17. Jusakul, A., Khuntikeo, N., Haigh, W. G., Kuver, R., Ioannou, G. N., Loilome, W., Namwat,
N., Bhudhisawasdi, V., Pugkhem, A., Pairojkul, C., and Yongvanit, P. (2012) Identification of
biliary bile acids in patients with benign biliary diseases, hepatocellular carcinoma and
cholangiocarcinoma. Asian Pacific journal of cancer prevention : APJCP 13 Suppl, 77-82
18. Liu, R., Zhao, R., Zhou, X., Liang, X., Campbell, D. J., Zhang, X., Zhang, L., Shi, R., Wang,
G., Pandak, W. M., Sirica, A. E., Hylemon, P. B., and Zhou, H. (2014) Conjugated bile acids
promote cholangiocarcinoma cell invasive growth through activation of sphingosine 1-
phosphate receptor 2. Hepatology (Baltimore, Md.) 60, 908-918
19. Adada, M., Canals, D., Hannun, Y. A., and Obeid, L. M. (2013) Sphingosine-1-phosphate
receptor 2. The FEBS journal 280, 6354-6366
20. Studer, E., Zhou, X., Zhao, R., Wang, Y., Takabe, K., Nagahashi, M., Pandak, W. M.,
Dent, P., Spiegel, S., Shi, R., Xu, W., Liu, X., Bohdan, P., Zhang, L., Zhou, H., and Hylemon,
P. B. (2012) Conjugated bile acids activate the sphingosine-1-phosphate receptor 2 in primary
rodent hepatocytes. Hepatology (Baltimore, Md.) 55, 267-276
21. Bocca, C., Ievolella, M., Autelli, R., Motta, M., Mosso, L., Torchio, B., Bozzo, F., Cannito,
S., Paternostro, C., Colombatto, S., Parola, M., and Miglietta, A. (2014) Expression of Cox-2
in human breast cancer cells as a critical determinant of epithelial-to-mesenchymal transition
and invasiveness. Expert opinion on therapeutic targets 18, 121-135
22. Fukazawa, E. M., Baiocchi, G., Soares, F. A., Kumagai, L. Y., Faloppa, C. C., Badiglian-
Filho, L., Coelho, F. R., Goncalves, W. J., Costa, R. L., and Goes, J. C. (2014) Cox-2, EGFR,
and ERBB-2 expression in cervical intraepithelial neoplasia and cervical cancer using an
automated imaging system. International journal of gynecological pathology : official journal of
the International Society of Gynecological Pathologists 33, 225-234
23. Generali, D., Buffa, F. M., Deb, S., Cummings, M., Reid, L. E., Taylor, M., Andreis, D.,
Allevi, G., Ferrero, G., Byrne, D., Martinotti, M., Bottini, A., Harris, A. L., Lakhani, S. R., and
at VIVA, VA Commonwealth Univ on December 15, 2015http://www.jbc.org/Downloaded from
JBC/2015/668277-R1 Bile acid and S1PR2-mediated signaling in CCA
12
Fox, S. B. (2014) COX-2 expression is predictive for early relapse and aromatase inhibitor
resistance in patients with ductal carcinoma in situ of the breast, and is a target for treatment.
British journal of cancer 111, 46-54
24. Ishii, Y., Sasaki, T., Serikawa, M., Minami, T., Okazaki, A., Yukutake, M., Ishigaki, T.,
Kosaka, K., Mouri, T., Yoshimi, S., Shimizu, A., Tsuboi, T., and Chayama, K. (2013) Elevated
expression of cyclooxygenase-2 and microsomal prostaglandin E synthase-1 in primary
sclerosing cholangitis: iotamplications for cholangiocarcinogenesis. International journal of
oncology 43, 1073-1079
25. Aishima, S., Mano, Y., Tanaka, Y., Kubo, Y., Shirabe, K., Maehara, Y., and Oda, Y. (2013)
Different roles of inducible nitric oxide synthase and cyclooxygenase-2 in carcinogenesis and
metastasis of intrahepatic cholangiocarcinoma. Human pathology 44, 1031-1037
26. Dai, J., Wang, H., Shi, Y., Dong, Y., Zhang, Y., and Wang, J. (2011) Impact of bile acids
on the growth of human cholangiocarcinoma via FXR. Journal of hematology & oncology 4,
41
27. Han, C., Leng, J., Demetris, A. J., and Wu, T. (2004) Cyclooxygenase-2 promotes human
cholangiocarcinoma growth: evidence for cyclooxygenase-2-independent mechanism in
celecoxib-mediated induction of p21waf1/cip1 and p27kip1 and cell cycle arrest. Cancer
research 64, 1369-1376
28. Ghosh, S., and Hayden, M. S. (2012) Celebrating 25 years of NF-kappaB research.
Immunological reviews 246, 5-13
29. Hayden, M. S., and Ghosh, S. (2004) Signaling to NF-kappaB. Genes & development 18,
2195-2224
30. Glinghammar, B., Inoue, H., and Rafter, J. J. (2002) Deoxycholic acid causes DNA
damage in colonic cells with subsequent induction of caspases, COX-2 promoter activity and
the transcription factors NF-kB and AP-1. Carcinogenesis 23, 839-845
31. Werneburg, N. W., Yoon, J. H., Higuchi, H., and Gores, G. J. (2003) Bile acids activate
EGF receptor via a TGF-alpha-dependent mechanism in human cholangiocyte cell lines.
American journal of physiology. Gastrointestinal and liver physiology 285, G31-36
32. Yoshikawa, D., Ojima, H., Iwasaki, M., Hiraoka, N., Kosuge, T., Kasai, S., Hirohashi, S.,
and Shibata, T. (2008) Clinicopathological and prognostic significance of EGFR, VEGF, and
HER2 expression in cholangiocarcinoma. British journal of cancer 98, 418-425
33. Roelle, S., Grosse, R., Aigner, A., Krell, H. W., Czubayko, F., and Gudermann, T. (2003)
Matrix metalloproteinases 2 and 9 mediate epidermal growth factor receptor transactivation
by gonadotropin-releasing hormone. The Journal of biological chemistry 278, 47307-47318
34. Rao, Y. P., Studer, E. J., Stravitz, R. T., Gupta, S., Qiao, L., Dent, P., and Hylemon, P. B.
(2002) Activation of the Raf-1/MEK/ERK cascade by bile acids occurs via the epidermal
growth factor receptor in primary rat hepatocytes. Hepatology (Baltimore, Md.) 35, 307-314
35. Eo, S. H., Cho, H. S., and Kim, S. J. (2014) Resveratrol regulates type II collagen and
COX-2 expression via the ERK, p38 and Akt signaling pathways in rabbit articular
chondrocytes. Experimental and therapeutic medicine 7, 640-648
36. Ma, Y., Li, H., Yue, Z., Guo, J., Xu, S., Xu, J., Jia, Y., Yu, N., Zhang, B., Liu, S., Liu, M.,
Shao, W., Chen, S., and Liu, P. (2014) Cryptotanshinone attenuates cardiac fibrosis via
at VIVA, VA Commonwealth Univ on December 15, 2015http://www.jbc.org/Downloaded from
JBC/2015/668277-R1 Bile acid and S1PR2-mediated signaling in CCA
13
downregulation of COX-2, NOX-2, and NOX-4. Journal of cardiovascular pharmacology 64,
28-37
37. Kim, H. G., Kim, Y. R., Park, J. H., Khanal, T., Choi, J. H., Do, M. T., Jin, S. W., Han, E.
H., Chung, Y. H., and Jeong, H. G. (2014) Endosulfan induces COX-2 expression via NADPH
oxidase and the ROS, MAPK, and Akt pathways. Archives of toxicology
38. Mercau, M. E., Astort, F., Giordanino, E. F., Martinez Calejman, C., Sanchez, R., Caldareri,
L., Repetto, E. M., Coso, O. A., and Cymeryng, C. B. (2014) Involvement of PI3K/Akt and p38
MAPK in the induction of COX-2 expression by bacterial lipopolysaccharide in murine
adrenocortical cells. Molecular and cellular endocrinology 384, 43-51
39. Lozano, E., Sanchez-Vicente, L., Monte, M. J., Herraez, E., Briz, O., Banales, J. M., Marin,
J. J., and Macias, R. I. (2014) Cocarcinogenic effects of intrahepatic bile acid accumulation in
cholangiocarcinoma development. Molecular cancer research : MCR 12, 91-100
40. Xia, X., Francis, H., Glaser, S., Alpini, G., and LeSage, G. (2006) Bile acid interactions
with cholangiocytes. World journal of gastroenterology : WJG 12, 3553-3563
41. Mathema, V. B., and Na-Bangchang, K. (2015) Current insights on cholangiocarcinoma
research: a brief review. Asian Pacific journal of cancer prevention : APJCP 16, 1307-1313
42. Ghouri, Y. A., Mian, I., and Blechacz, B. (2015) Cancer review: Cholangiocarcinoma.
Journal of carcinogenesis 14, 1.doi:10.4103/1477-3163.151940.
43. Rizvi, S., and Gores, G. J. (2014) Molecular pathogenesis of cholangiocarcinoma.
Digestive diseases 32, 564-569
44. Nunez Martinez, O., Clemente Ricote, G., and Garcia Monzon, C. (2003) [Role of
cyclooxygenase-2 in the pathogenesis of chronic liver diseases]. Medicina clinica 121, 743-
748
45. Zhang, L., Jiang, L., Sun, Q., Peng, T., Lou, K., Liu, N., and Leng, J. (2007) Prostaglandin
E2 enhances mitogen-activated protein kinase/Erk pathway in human cholangiocarcinoma
cells: involvement of EP1 receptor, calcium and EGF receptors signaling. Mol Cell Biochem
305, 19-26
46. Sun, B., Rong, R., Jiang, H., Zhang, H., Wang, Y., Bai, X., Zhang, M., Ma, J., Xia, S., Shu,
W., Zhang, L., and Leng, J. (2013) Prostaglandin E2 receptor EP1 phosphorylate CREB and
mediates MMP2 expression in human cholangiocarcinoma cells. Mol Cell Biochem 378, 195-
203
47. Volzke, A., Koch, A., Meyer Zu Heringdorf, D., Huwiler, A., and Pfeilschifter, J. (2014)
Sphingosine 1-phosphate (S1P) induces COX-2 expression and PGE2 formation via S1P
receptor 2 in renal mesangial cells. Biochimica et biophysica acta 1841, 11-21
48. Skoura, A., Michaud, J., Im, D. S., Thangada, S., Xiong, Y., Smith, J. D., and Hla, T. (2011)
Sphingosine-1-phosphate receptor-2 function in myeloid cells regulates vascular inflammation
and atherosclerosis. Arterioscler Thromb Vasc Biol 31, 81-85
49. Amonyingcharoen, S., Suriyo, T., Thiantanawat, A., Watcharasit, P., and Satayavivad, J.
(2015) Taurolithocholic acid promotes intrahepatic cholangiocarcinoma cell growth via
muscarinic acetylcholine receptor and EGFR/ERK1/2 signaling pathway. International journal
of oncology 46, 2317-2326
FOOTNOTES
at VIVA, VA Commonwealth Univ on December 15, 2015http://www.jbc.org/Downloaded from
JBC/2015/668277-R1 Bile acid and S1PR2-mediated signaling in CCA
14
To whom correspondence may be addressed: Huiping Zhou, Department of Microbiology and
Immunology and McGuire Veterans Affairs Medical Center, Richmond, VA, USA 23298.
Tel:804-828-6817; Fax:804-828-0676; Email:hzhou@vcu.edu. Or Luyong Zhang, Jiangsu
Center for Drug Screening, China Pharmaceutical University, Nanjing, Jiangsu, China 210009.
Tel: 86-25-83271500; Fax: 86-25-83271500; Email: lyzhang@cpu.edu.cn. Or Zhenzhou Jiang,
Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical
University, Nanjing, Jiangsu, China 210009. Tel: 86-25-83271142; Fax: 86-25-83271142;
Email: beaglejiang@cpu.edu.cn.
The abbreviations used are: Akt, protein kinase B; CBA: conjugated bile acid; CCA,
Cholangiocarcinoma; COX-2, cyclooxygenase-2; EGF, epidermal growth factor; EGFR,
epidermal growth factor receptor; EP, Prostaglandin E receptor, ERK, extracellular regulated
protein kinases; IκB, Inhibitor of κB; IKK α/β, IκB kinase α/β; IL-6, interleukin-6, MMP-2/9,
matrix metalloproteinases 2/9; NF-B, Nuclear factor kappa-light-chain-enhancer of activated
B cells; PI3K, phosphatidylinositide 3-kinases; PGE2, prostaglandin E2; S1P, sphingosine 1-
phosphate; S1PR2, sphingosine 1-phosphate receptor 2; TCA, taurocholate; TLCA;
taurolithocholic acid; TNF-α, tumor necrosis factor-α
FIGURE 1. The effect of TCA on COX-2 expression in HuCCT1 cells. Cells were cultured
in serum-free medium overnight and then treated with either 100 μM TCA for different time
periods (0-24 h) or different concentrations of TCA (0, 25, 50, 100 or 200 μM) for 8h. At the
end of each treatment, cells were harvested and protein levels of COX-2 were detected by
Western blot analysis. (A and C) Representative images of the immunoblots for COX-2 and
actin are shown. (B and D) Relative densities of COX-2 were analyzed using Quantity One
software using actin as a loading control. Values represent the mean ± S.E. of three
independent experiments. Statistical significance relative to the vehicle control: **p<0.01, ***p
<0.001.
FIGURE 2. The role of S1PR2 in TCA-induced COX-2 expression in HuCCT1 cells. Cells
were cultured in serum-free medium overnight and then treated with S1P (100 nM) or TCA
(100 μM) with or without JTE-013 (10 μM) for 8h. At the end of treatment, total protein was
isolated to determine COX-2 protein levels using Western blot analysis. (A) Representative
images of the immunoblots for COX-2 and actin are shown. (B) Relative densities of COX-2
were determined using actin as a loading control. Values represent the mean ± S.E. of three
independent experiments. Statistical significance relative to the vehicle control: *p<0.05,
***p<0.001; Statistical significance relative to corresponding treatment group without JTE-013:
#p<0.05, ###p<0.001. (C) Cells were transfected with either a control scrambled shRNA or
S1PR2-specifc shRNA expression vector for 48h. Relative mRNA levels of S1PR2 were
detected by real-time RT-PCR and normalized using GAPDH as an internal control as
described in “Methods. (D) Cells were treated with vehicle control or TCA (100 μM) for 8 h
after transfection with control or S1PR2 shRNA for 48 h. Protein levels of COX-2 was
determined by Western blot analysis. Representative images of the immunoblots for COX-2
and actin are shown. (E) Relative densities of COX-2 were determined using actin as a loading
at VIVA, VA Commonwealth Univ on December 15, 2015http://www.jbc.org/Downloaded from
JBC/2015/668277-R1 Bile acid and S1PR2-mediated signaling in CCA
15
control. Values represent the mean ± S.E. of three independent experiments. Statistical
significance relative to the vehicle control: *p<0.05; Statistical significance relative to the TCA-
treated group with control shRNA: #p<0.05. (F) Cells were pretreated with JTE-013 (10 μM)
for 1 h before being treated with TCA (100 μM) for 8 hours. At the end of the treatment, cell
culture medium was collected for detection of PGE2 secretion as described in “Methods”.
Values represent the mean ± S.E. of three independent experiments. Statistical significance
relative to the vehicle control: **p<0.01; relative to the TCA treatment group, #p<0.05.
Figure 3. (A-B) Effect of TCA on COX-1 expression. HuCCT1 cells were cultured in serum-
free medium overnight and then treated with S1P (100 nM) or TCA (100 μM) with or without
JTE-013 (10 μM) for 8h. At the end of treatment, total proteins were isolated. The protein level
of COX-1 was detected by Western blot analysis. (A) Representative images of immunoblots
for COX-1 and Actin are shown. (B) Relative protein levels of COX-1 were analyzed using
Actin as a loading control. Values represent the mean ± S.E. of three independent experiments.
(C-D) Effect of TCA on inflammatory cytokines expression. HuCCT1 cells were cultured
in serum-free medium overnight and then treated with S1P (100 nM) or TCA (100 μM) with or
without JTE-013 (10 μM) for 8h. At the end of treatment, total cellular RNA was isolated.
Relative mRNA levels of (A) IL-6 and (B) TNF-α were determined by real-time RT-PCR and
normalized using GAPDH as an internal control. Values represent the mean ± S.E. of three
independent experiments. Statistical significance relative to the vehicle control, *p<0.05,
**p<0.01, ***p<0.001; relative to S1P or TCA treatment group: #p<0.05, ##p<0.01, ###p<0.001.
FIGURE 4. The effect of S1PR2 activation and COX-2 expression on TCA-induced
invasiveness of HuCCT1 cells. (A and B): Cells were cultured inside of the matrigel pre-
coated transwell inserts and pretreated with JTE-013 (10 μM) for 1 hour and then treated with
either S1P (100 nM) or TCA (100 μM) for 24 h. (C and D): Cells were transfected with either
control shRNA or S1PR2 shRNA for 48h. Transfected cells were cultured inside of the matrigel
pre-coated transwell inserts and then treated with TCA (100 μM) for 24 h. (E and F): Cells
were cultured inside of the matrigel pre-coated transwell inserts and treated with TCA (100
μM) for 24 h with or without Celecoxib (40 µM). At the end of treatment, the number of invasive
cells and invasive index were analyzed as described in Methods. Representative Images for
each treatment from three independent experiments are shown. Statistical significance
relative to the vehicle control: *p<0.05, **p<0.01, ***p<0.001; relative to the corresponding
S1P or TCA treatment group: #p<0.05, ##p<0.01, ###p<0.001.
FIGURE 5. The effect of TCA on activation of IKKα/β-NF-B pathways in HuCCT-1 cells.
(A-C) Time course of TCA-induced activation of IKKα/β-NF-B pathways. Cells were
cultured in serum-free medium overnight and then treated with TCA (100 μM) for different
treatment periods (0-24 h). The protein levels of phosphorylated IKKα/β (p-IKKα/β), total
IKKα/β, phosphorylated NF-B p65 (p-NF-B p65) and total NF-B p65 were determined by
Western blot analysis. (D-F) TCA-induced dose-dependent activation of IKKα/β-NF-B
pathways. Cells were cultured in serum-free medium overnight and then treated with
different concentrations of TCA (0, 25, 50, 100 or 200 μM) for 4 h. At the end of each treatment,
cells were harvested, and total protein was isolated. The protein levels of p-IKKα/β, total
IKKα/β, p-NF-B p65 and total NF-B p65 were determined by Western blot analysis. (A
at VIVA, VA Commonwealth Univ on December 15, 2015http://www.jbc.org/Downloaded from
JBC/2015/668277-R1 Bile acid and S1PR2-mediated signaling in CCA
16
and D) Representative images of p-IKKα/β, total IKKα/β, p-NF-B p65 and total NF-B p65
are shown. (B-C and E-F) Relative protein levels of p-IKKα/β/ IKKα/β and p-NF-B p65/NF-
B p65 were determined. Values represent the mean ± S.E. of three independent experiments.
Statistical significance relative to vehicle control: *p<0.05, **p<0.01, ***p<0.001.
Figure 6. The effect of TCA on nuclear translocation of NF-B. HuCCT1 cells were
cultured in serum-free medium for overnight and then treated with TCA (100 μM) for different
treatment periods (0, 2, 4, 8 or 24 h). At the end of each treatment, cytosol and nuclear proteins
were isolated as described in “Methods”. Protein levels of NF-B p65 were determined by
Western Blot analysis. Lamin B and actin were used as loading controls for nuclear and cytosol
protein, respectively. (A) Representative images of the immunoblots for NF-B p65, lamin B
and actin are shown. (B) Relative protein levels of NF-B p65 in the nucleus and cytosol.
Values represent the mean ± S.E. of three independent experiments. Statistical significance
relative to the vehicle control: ***p<0.001. (C-D) Immunofluorescence staining of NF-B p65.
HuCCT1 cells were cultured in serum-free medium overnight and treated with either 100 μM
TCA for different treatment periods (0, 2, 4, or 8 h) or different concentrations of TCA (0, 25,
50, or 100 μM) for 4 h. At the end of treatment, immunofluorescence staining was performed
to detect subcellular location of NF-B p65. Representative images for each group are shown.
FIGURE 7. The effect of chemical antagonist of S1PR2 on S1P- and TCA-induced
activation of IKKα/β- NF-κB pathway. HuCCT1 cells were cultured in serum-free medium
overnight and pre-treated with JTE-013 (10 µM) for 1 h, and then treated with S1P (100 nM)
or TCA (100 μM) for 4h. (A and B) Total protein was isolated, and protein levels of p-IKKα/β,
total IKKα/β, p-NF-B p65 and total NF-B p65 were determined by Western blot analysis.
The relative densities of p-IKKα/β/total IKKα/β and p-NF-B p65/total NF-Bp65 were
determined. (C and D) Cytosol and nuclear proteins were isolated as described in “Methods”.
Protein levels of NF-κB p65, lamin B and actin were determined by Western blot analysis.
Lamin B and actin were used as a loading control for nuclear and cytosol protein, respectively.
(A and C) Representative images of the immunoblots for p-IKKα/β, total IKKα/β, p-NF-B p65
and total NF-B p65 are shown. (B) Relative protein levels of p-IKKα/β/total IKKα/β and p-NF-
B p65/total NF-B p65. (D) Relative protein levels of nuclear NF-B p65/lamin B and cytosol
NF-B p65/actin. Values represent the mean ± S.E. of three independent experiments.
Statistical significance relative to the vehicle control: *p<0.05, **p<0.01, ***p<0.001; relative
to S1P or TCA treatment group: #p<0.05, ##p<0.01, ###p<0.001.
Figure 8. The effect of JTE-013 on S1P- and TCA-induced nuclear translocation of NF-
B. HuCCT1 cells were cultured in serum-free medium overnight and then treated with either
100 μM TCA for different treatment periods (0, 2, 4, 8 or 24 h) or different concentrations of
TCA (0, 25, 50, or 100 μM) for 4 h. At the end of treatment, immunofluorescence staining was
performed to detect subcellular location of NF-B p65. Representative images for each group
are shown.
Figure 9. The effect of TCA on EGFR transactivation in HuCCT1 cells. HuCCT1 cells
were cultured in serum-free medium overnight and then treated with either 100 μM TCA for
different time periods (0, 0.17, 0.5, 1, 2, 4 or 8 h), or with different concentrations of TCA (0,
at VIVA, VA Commonwealth Univ on December 15, 2015http://www.jbc.org/Downloaded from
JBC/2015/668277-R1 Bile acid and S1PR2-mediated signaling in CCA
17
25, 50, 100 or 200 μM) for 15 min. At the end of treatment, total protein was isolated. Protein
levels of phosphorylated EGFR (p-EGFR), total EGFR (t-EGFR) were detected by Western
Blot analysis. (A and C) Representative images of the immunoblots for p-EGFR and t-EGFR
are shown. (B and D) Relative protein levels of p-EGFR/t-EGFR were calculated from three
independent experiments. Values represent the mean ± S.E. of three independent
experiments. Statistical significance relative to the vehicle control: *p<0.05, **p<0.01,
***p<0.001; relative to S1P or TCA treatment group: ##p<0.01, ###p<0.001.
FIGURE 10. The role of S1PR2 in TCA-induced EGFR transactivation in HuCCT1 cells.
(A-B) Cells were cultured in serum-free medium and pretreated with JTE-013 (10μM) for 1h,
and then treated with S1P (100 nM) or TCA (100 μM) for 15 min. (C-D) HuCCT1 cells were
transfected with control shRNA or S1PR2 shRNA for 48 h and then treated with TCA (100 μM)
for 15 min. At the end of treatment, total protein was isolated. Protein levels of p-EGFR and
t-EGFR were detected by Western Blot analysis. (A and C) Representative images of the
immunoblots for p-EGFR and t-EGFR are shown. (B and D) Relative protein levels of p-
EGFR/t-EGFR were calculated from three independent experiments. Values represent the
mean ± S.E. of three independent experiments. Statistical significance relative to the vehicle
control: **p<0.01, ***p<0.001; relative to the S1P or TCA treatment group: ##p<0.01,
###p<0.001.
Figure 11. The effect of TCA on MMP-2 and MMP9 expression. HuCCT1 cells were
cultured in serum-free medium overnight and then treated with S1P (100 nM) or TCA (100 μM)
with or without JTE-013 (10 μM) for 8h. At the end of treatment, total cellular RNA and protein
lysates were isolated. Relative mRNA levels of (A) MMP-2 and (B) MMP-9 were determined
by real-time RT-PCR and normalized using GAPDH as an internal control. Protein levels of
MMP2 and MMP9 were detected by Western Blot analysis. Representative images of the
immunoblots for MMP2 and MMP9 are shown (C and E). Relative protein levels of MMP2/Actin
and MMP9/Actin were calculated from three independent experiments. Values represent the
mean ± S.E. of three independent experiments. Statistical significance relative to the vehicle
control: **p<0.01, ***p<0.001; relative to S1P or TCA treatment group: #p<0.05, ##p<0.01.
Figure 12. The effect of TCA on ERK and Akt activation in HuCCT1 cells. (A-B) Time
course of TCA-induced ERK and Akt activation. HuCCT1 cells were cultured in serum-free
medium overnight and then treated with TCA (100 μM) for different treatment periods (0, 0.5,
1, 2, 4, 8 or 24 h) (C-D) TCA-induced dose-dependent activation of ERK and Akt.
HuCCT1 cells were cultured in serum-free medium overnight and then treated with different
concentrations of TCA (0, 25, 50, or 100 μM) for 1 h. (E-F) The effect of JTE-013 on TCA-
/S1P-induced ERK and Akt activation. HuCCT1 cells were cultured in serum-free medium
overnight and pre-treated with JTE-013 (10 μM) for 1 hour and then treated with S1P (100 nM)
or TCA (100 μM) for 1 h. At the end of each treatment, cells were harvested and total protein
was isolated. The protein levels of phosphorylated ERK1/2 (p-ERK), total ERK1/2 (t-ERK),
phosphorylated Akt1/2/3 (p-Akt) and total Akt1/2/3 (t-Akt) were detected by Western blot
analysis. (A, C and E) Representative images of the immunoblots for p-ERK, t-ERK, p-Akt,
and t-Akt are shown. (B, D and F) Relative levels of p-ERK/t-ERK and p-Akt/t-Akt. Values
represent the mean ± S.E. of three independent experiments. Statistical significance relative
at VIVA, VA Commonwealth Univ on December 15, 2015http://www.jbc.org/Downloaded from
JBC/2015/668277-R1 Bile acid and S1PR2-mediated signaling in CCA
18
to the vehicle control: *p<0.05, **p<0.01, ***p<0.001; relative to S1P or TCA treatment group:
##p<0.01, ###p<0.001.
Figure 13. Inhibition of TCA-induced ERK1/2 and Akt activation by chemical inhibitors
in HuCCT1 cells. HuCCT1 cells were cultured in serum-free medium overnight and
pretreated with a selective inhibitor of MEK1/2, U0126 (10 μM) or a selective inhibitor of Akt,
MK2206 (3 μM) for 1 h and then treated with S1P (100 nM) or TCA (100 μM) for 1 h. At the
end of treatment, total protein was isolated. Protein levels of p-ERK1/2, total ERK1/2, p-Akt,
and t-Akt were detected by Western blot analysis. (A and C) Representative images of the
immunoblots for p-ERK1/2, t-ERK1/2, p-Akt, and t-Akt are shown. (B and D) Relative protein
levels of p-ERK1/2/t-ERK1/2 and p-Akt/t-Akt were calculated from three independent
experiments. Values represent the mean ± S.E. of three independent experiments. Statistical
significance relative to vehicle control: **p<0.01, ***p<0.001; relative to S1P or TCA treatment
group: ##p<0.05, ###p<0.001.
FIGURE 14 . Effect of TCA-induced ERK1/2 and Akt activation on COX-2 expression in
HuCCT1 cells. Cells were cultured in serum-free medium overnight and pretreated with a
selective inhibitor of MEK1/2, U0126 (10 μM) for 1 h and then treated with S1P (100 nM) or
TCA (100 μM) for 1 h. At the end of treatment, total protein was isolated and protein levels of
COX-2 and actin were detected by Western blot analysis. (A and C) Representative images
of the immunoblots for COX-2 and actin are shown. (B and D) Relative protein levels of COX-
2/Actin were calculated from three independent experiments. Values represent the
mean ± S.E. of three independent experiments. Statistical significance relative to the vehicle
control: *p<0.05, **p<0.01; relative to the S1P or TCA treatment group: #p<0.05, ###p<0.01.
Figure 15. Effect of TCA and S1P on S1PR2 internalization. HEK293 cells were transfected
with 3xHA-tagged human S1PR2 as described in “Methods”. Cells were stimulated with
vehicle control or TCA (100 µM) or S1P (100 nM) for 10 mins, the internalization process was
stopped by fixing with 3.7% formaldehyde at 4C. The localization of S1PR2 was monitored
by immunofluorescence staining. Representative images of each treatment group are shown.
Figure 16. Proposed signaling pathways involved in TCA-induced COX-2 expression in
human CCA cells. Activation of S1PR2 by TCA directly activates ERK1/2/AKt via activated
G proteins or indirectly activates ERK1/2/Akt via EGFR. Activated ERK1/2/Akt further activate
NF-B and COX-2 expression by inducing phosphorylation of IKKα/β and IB, ubiquitination
and degradation of p-IB, and nuclear translocation of NF-B.
at VIVA, VA Commonwealth Univ on December 15, 2015http://www.jbc.org/Downloaded from
Figure 1
A
COX-2
Actin
0 0.5 1 2 4 8 24
TCA (100 μM)
Time (h)
0 25 50 100 200
TCA (μM)
COX-2
Actin
C
Time ( 8 h)
00.5 1 2 4824
0.0
2.0
4.0
6.0
** **
***
Time (h)
Relative protein levels
of COX-2
B
025 50 100 200
0.0
1.0
2.0
3.0
4.0
** **
***
TCA (M)
Relative protein levels
of COX-2
D
at VIVA, VA Commonwealth Univ on December 15, 2015http://www.jbc.org/Downloaded from
COX-2
Actin
S1P - + - - + -
TCA - - + - - +
JTE-013
A
COX-2
Actin
Con TCA Con TCA
Control
shRNA
S1PR2
shRNA
D
Control shRNA S1PR2 shRNA
0.0
0.5
1.0
1.5
**
Reltive mRNA levels
of S1PR2
C
B
0.0
1.0
2.0
3.0
4.0
5.0
*
***
####
- + - - + -
- - + - - +
- - - + + +
JTE-013
TCA
S1P
Relative protein levels
of COX-2
F
Control JTE-013 TCA TCA+JTE-013
0
50
100
150
200
**
#
PGE2
(pg/ml)
E
0.0
1.0
2.0
3.0
4.0
5.0
*
#
ControlControl TCA TCA
Control shRNA S1PR2 shRNA
Relative protein leve ls
of COX-2
Figure 2
at VIVA, VA Commonwealth Univ on December 15, 2015http://www.jbc.org/Downloaded from
0.0
1.0
2.0
3.0
4.0
** **
## #
- + - - + -
- - + - - +
- - - + + +JTE-013
TCA
S1P
Relative mRNA levels
of TNF-
Figure 3
C D
0
1
2
JTE-013
TCA
S1P - + - - + -
- - + - - +
- - - + + +
Relative protein levels
of COX-1
COX-1
Actin
S1P - + - - + -
TCA - - + - - +
JTE-013
A B
at VIVA, VA Commonwealth Univ on December 15, 2015http://www.jbc.org/Downloaded from
JTE-013
S1P+JTE-013
TCA+JTE-013
Control
TCA
S1P
A
0.0
1.0
2.0
3.0
***
##
#
JTE-013
TCA
S1P - - + + - -
- - - - + +
- + - + - +
Relative inv as ion inde x
B
Control TCA
Control TCA
Control
shRNA
S1PR2
shRNA
C
Control
TCA
Control
TCA
0.0
1.0
2.0
3.0
**
Control shRNA S1PR2 shRNA
###
Relative inv as ion inde x
D
Control TCA
Celecoxib Celecoxib+TCA
E
0.0
1.0
2.0
3.0
TCA
Celecoxib
- - + +
- + - +
***
##
Relative inv as ion inde x
F
Figure 4
at VIVA, VA Commonwealth Univ on December 15, 2015http://www.jbc.org/Downloaded from
Figure 5
TCA (100μM)
p-NF-κB p65
NF-κB p65
p-IKKα/β
IKKα/β
0 0.5 1 2 4 8 24
Time (h)
A
0
0.5
1
2
4
8
24
0.0
0.5
1.0
1.5
2.0
2.5
Time (h)
** *** ***
Relative protein levels of
p-IKK//IKK/
0
0.5
1
2
4
8
24
0.0
0.5
1.0
1.5
2.0
** **
Time (h)
Relative protein levels of
p-NF-Bp65 /NF-Bp65
B
Time (4h)
p-IKKα/β
IKKα/β
0 25 50 100 200
TCA (μM)
p-NF-κB p65
NF-κB p65
D
0
25
50
100
200
0.0
1.0
2.0
3.0
**
***
***
TCA (M)
***
Relative protein levels
of p-IKK //IKK/
0
25
50
100
200
0.0
1.0
2.0
3.0
4.0
TCA (M)
*
**
Relative protein levels of
p-NF-Bp65/NF-Bp65
E
CF
at VIVA, VA Commonwealth Univ on December 15, 2015http://www.jbc.org/Downloaded from
Figure 6
0
2
4
8
12
24
0
1
2
3
4
Time (h)
*** ***
Relative protein levels
of NF-B in nuclear
0
2
4
8
12
24
0.0
0.5
1.0
1.5
Time (h)
Relative protein levels
of NF-B in cytosol
B
D
025 50 100
TCA (μM)
Time (4h)
Cytosol
NF-B p65
Actin
TCA (100μM)
A
NF-B p65
Lamine B
Nuclear
0 2 4 8 12 24
Time (h)
024 8
TCA (100μM)
C
Time (h)
Hochest Rabbit IgG
Negative control
at VIVA, VA Commonwealth Univ on December 15, 2015http://www.jbc.org/Downloaded from
p-NF-κB p65
NF-κB p65
p-IKKα/β
IKKα/β
S1P - + - - + -
TCA - - + - - +
JTE-013
Figure 7
A
0.0
0.5
1.0
1.5
2.0
2.5
** **
## ##
- + - - + -
- - + - - +
- - - + + +JTE-013
TCA
S1P
Relative protein levels of
p-IKK//IKK/
0.0
1.0
2.0
3.0
**
##
- + - - + -
- - + - - +
- - - + + +JTE-013
TCA
S1P
Relative protein levels of
p-NF-B p65/NF-B p65
B
Nuclear
Lamin B
NF-κB p65
NF-κB p65
Actin
Cytosol
S1P - + - - + -
TCA - - + - - +
JTE-013
C
0.0
1.0
2.0
3.0
**
***
## ###
- + - - + -
- - + - - +
- - - + + +JTE-013
TCA
S1P
Relative protein levels of
NF-B p65 in nucleus
0.0
0.5
1.0
1.5
- + - - + -
- - + - - +
- - - + + +JTE-013
TCA
S1P
Relative protein levels of
NF-B p65 in c ytosol
D
at VIVA, VA Commonwealth Univ on December 15, 2015http://www.jbc.org/Downloaded from
JTE-013 S1P+JTE-013 TCA+JTE-013Control TCAS1P
Figure 8
at VIVA, VA Commonwealth Univ on December 15, 2015http://www.jbc.org/Downloaded from
Figure 9
A
00.17 0.5 1248
0.0
1.0
2.0
3.0
4.0
Time (h)
*
*** *** **
Relative protein levels of
p-EGFR/t-EGFR
B
C
t-EGFR
p-EGFR
0 25 50 100 200
TCA (μM)
Time (15 min)
0
25
50
100
200
0.0
2.0
4.0
6.0
TCA (M)
**
***
Relative protein levels of
p-EGFR/t-EGFR
D
t-EGFR
p-EGFR
0 0.17 0.5 1 2 4 8
TCA (100 μM)
Time (h)
at VIVA, VA Commonwealth Univ on December 15, 2015http://www.jbc.org/Downloaded from
Figure 10
t-EGFR
p-EGFR
S1P - + - - + -
TCA - - + - - +
JTE-013
A
0.0
1.0
2.0 ***
**
### ##
- + - - + -
- - + - - +
- - - + + +
JTE-013
S1P
TCA
Relative protein levels of
p-EGFR/t-EGFR
B
C
0.0
1.0
2.0
3.0
ControlControl TCA TCA
Control shRNA S1PR2 shRNA
**
##
Relative protein levels of
p-EGFR/t-EGFR
D
t-EGFR
p-EGFR
Control TCA Control TCA
Control
shRNA
S1PR2
shRNA
at VIVA, VA Commonwealth Univ on December 15, 2015http://www.jbc.org/Downloaded from
A
0
1
2
3
**
***
#
##
JTE-013
TCA
S1P - + - - + -
- - + - - +
- - - + + +
Relative mRNA levels
of MMP-2
0
1
2
3
4
**
***
##
##
JTE-013
TCA
S1P - + - - + -
- - + - - +
- - - + + +
Relative mRNA levels
of MMP-9
C
B
D
Figure 11
MMP-2
S1P - + - - + -
TCA - - + - - +
JTE-013
Actin
0
1
2
3
4
JTE-013
TCA
S1P - + - - + -
- - + - - +
- - - + + +
**
#
Relative protein
levels of MMP-2
E F
MMP-9
Actin
0
1
2
3
JTE-013
TCA
S1P - + - - + -
- - + - - +
- - - + + +
**
*
#
Relative protein
levels of MMP-9
S1P - + - - + -
TCA - - + - - +
JTE-013
at VIVA, VA Commonwealth Univ on December 15, 2015http://www.jbc.org/Downloaded from
Figure 12
A
p-ERK1/2
t-ERK1/2
0 0.5 1 2 4 8 24
TCA (100 μM)
p-Akt
t-Akt
Time (h)
C
p-ERK1/2
t-ERK1/2
Time (1 h)
p-Akt
t-Akt
0 25 50 100 200
TCA (μM)
p-
ERK1/2
t-ERK1/2
S1P - + - - + -
TCA - - + - - +
JTE-013
p-Akt
t-Akt
E
0
0.5
1
2
4
8
24
0.0
0.5
1.0
1.5
2.0
*** ** ***
Time (h)
Relative protein levels of
p-ERK1/2/t-ERK1/2
0
0.5
1
2
4
8
24
0
1
2
3
Time (h)
**
***
*
Relative protein levels of
p-Akt/t-Akt
B
0
25
50
100
200
0.0
0.5
1.0
1.5
2.0
2.5
**
TCA (M)
Relative protein levels of
p-ERK1/2/t-ERK1/2
0
25
50
100
200
0.0
1.0
2.0
3.0
TCA (M)
**
Relative protein levels of
p-Akt/t-Akt
D
0.0
1.0
2.0
3.0
**
***
## ###
- + - - + -
- - + - - +
- - - + + +
JTE-013
TCA
S1P
Relative protein levels of
p-Akt/t-Akt
0.0
1.0
2.0
3.0
*
**
##
##
Relative protein levels of
p-ERK1/2/t-ERK1/2
F
at VIVA, VA Commonwealth Univ on December 15, 2015http://www.jbc.org/Downloaded from
p-ERK1/2
t-ERK1/2
S1P - + - - + -
TCA - - + - - +
U0126
A
0.0
1.0
2.0
3.0
** ***
*** ### ###
- + - - + -
- - + - - +
- - - + + +U0126
TCA
S1P
Relative protein levels of
p-ERK1/2/t-ERK1/2
B
C
Figure 13
p-Akt
t-Akt
S1P - + - - + -
TCA - - + - - +
MK2206
0.0
1.0
2.0
3.0
*** **
###
##
- + - - + -
- - + - - +
- - - + + +
MK-2206
TCA
S1P
Relative protein levels of
p-Akt/t-Akt
D
at VIVA, VA Commonwealth Univ on December 15, 2015http://www.jbc.org/Downloaded from
0.0
1.0
2.0
3.0
4.0
**
#
- + - - + -
- - + - - +
- - - + + +
U0126
TCA
S1P
Relative protein levels
of COX-2/Actin
B
Figure 14
A
COX-2
Actin
S1P - + - - + -
TCA - - + - - +
U0126
C
0.0
1.0
2.0
3.0
4.0
*
**
#
##
- + - - + -
- - + - - +
- - - + + +
MK-2206
TCA
S1P
Relative protein levels
of COX-2/Actin
D
COX-2
Actin
S1P - + - - + -
TCA - - + - - +
MK2206
at VIVA, VA Commonwealth Univ on December 15, 2015http://www.jbc.org/Downloaded from
Control TCA S1P
Figure 15
FITC
DAPI
at VIVA, VA Commonwealth Univ on December 15, 2015http://www.jbc.org/Downloaded from
MMPs
Pro-EGF EGF
EGFR
PLCβ
S1PR2
ERK1/2 and Akt
TCA JTE-013
Transcription
Coactivator
Activated
IKKK
Iκ-B kinase
(IKK) Activated IKK-
I-B NF-BMigration of NF-B
into nucleus
Ubiquitylation and
degradation of p-I-B
In proteasome
COX-2
Figure 16
at VIVA, VA Commonwealth Univ on December 15, 2015http://www.jbc.org/Downloaded from
Zhou
Zhenzhou Jiang, Luyong Zhang and Huiping
Xiaoyan Qiang, Phillip B. Hylemon,
Runping Liu, Xiaojiaoyang Li, Lan Luo,
Cholangiocarcinoma Cell Line
Receptor 2 in a Human 1-phosphateExpression via the Sphingosine
Taurocholate Induces Cyclooxygenase-2
Gene Regulation:
published online October 30, 2015J. Biol. Chem.
10.1074/jbc.M115.668277Access the most updated version of this article at doi:
.JBC Affinity SitesFind articles, minireviews, Reflections and Classics on similar topics on the
Alerts:
When a correction for this article is posted When this article is cited
to choose from all of JBC's e-mail alertsClick here
http://www.jbc.org/content/early/2015/10/30/jbc.M115.668277.full.html#ref-list-1
This article cites 0 references, 0 of which can be accessed free at
at VIVA, VA Commonwealth Univ on December 15, 2015http://www.jbc.org/Downloaded from
... Taurocholate, a blood metabolite, previously lacked clear evidence for its specific impact on telomere length. However, given the significant role of bile acids in cell signaling and lipid metabolism [68], we hypothesized that taurocholate might influence telomere length by regulating cell growth, differentiation, apoptosis [69][70][71], and altering intracellular fatty acid and cholesterol levels [28,30,72]. Additionally, taurocholate was primarily synthesized and metabolized in the liver, and liver dysfunction could lead to abnormal bile acid metabolism, subsequently affecting telomere length [73]. ...
Article
Full-text available
Background Telomere length refers to the protective cap at the end of chromosomes, and it plays a crucial role in many diseases. The objective of this study is to explore the relationship between blood metabolites and telomere length, aiming to identify novel biological factors that influence telomere length. Methods In this study, we extracted genome-wide association study (GWAS) data for blood metabolites from a sample of 7824 Europeans. Additionally, GWAS data for telomere length were obtained from the Open GWAS database (GWAS ID: ieu-b-4879). The primary analysis of this study utilized the random inverse variance weighted (IVW) method. Complementary analyses were also conducted using the MR-Egger and weighted median approaches. Sensitivity analyses were performed to assess the robustness of the findings. These included the Cochran Q test, MR-Egger intercept test, MR-PRESSO, and leave-one-out analysis. To investigate the possibility of reverse causation, reverse MR analysis was conducted. Additionally, multivariable MR was utilized to evaluate the direct effect of metabolites on telomere length. Results The results suggested a potential association between 15-methylpalmitate, taurocholate, levulinate, and X-12712 and telomere length. MVMR analysis further showed that 15-methylpalmitate, taurocholate, and levulinate can directly influence telomere length, regardless of other metabolites. Conclusions This study suggests that 15-methylpalmitate, taurocholate, and levulinate are likely factors correlated with telomere length. These findings will contribute to the development of strategies for protecting telomeres, preventing related diseases, and establishing a new biological foundation for achieving healthy aging.
... It has been suggested that conjugated BAs promote cell growth through S1PR2 in CCA [13], and the accumulation of conjugated BAs attributed to the bile duct obstruction results in the activation of S1PR2, which further activates the extracellular signal-regulated protein kinase 1/2 signaling pathway [14]. However, the role of S1PR2 in cancer is controversial, demonstrating that this receptor can not only promote tumorigenesis but also inhibit the motility of cancer cells and tumor angiogenesis [15][16][17]. To date, there have been few studies on S1PR2 in extrahepatic CCA, which have been carried out on human CCA cell lines or mice [14,18,19]. ...
Article
Full-text available
Background and aims: In biliary epithelial cells, two bile acid receptors, sphingosine 1-phosphate receptor 2 (S1PR2) and Takeda G protein-coupled receptor 5 (TGR5) have been reported to trigger cell proliferation, as well as neoplastic cell invasiveness. In this study, we aimed to investigate the clinical significance of S1PR2/ TGR5 expression in extrahepatic cholangiocarcinoma (CCA) patients. Methods: Patients who underwent surgical resection of extrahepatic CCA at Korea University Guro Hospital between 2002 and 2018 were included. Data on immunohistochemical staining and H-score of S1PR2 and TGR5 were evaluated using digital image analysis. Results: A total of 115 cases of invasive CCA were analyzed. The H-score of S1PR2 showed a decrease in invasive CCA (p=0.052) but that of TGR5 showed a significant increase (p=0.02). Overall survival and disease-free survival were significantly lower in the low S1PR2 expression group (p<0.05) than in the control group; however, TGR5 expression was not significant (p=0.096). In multivariate analysis, low S1PR2 was only significant for poor prognosis. Conclusion: Low S1PR2 level was the only independent poor prognostic factor in patients with resected extrahepatic CCA.
... This hypothesis assumes that substances formed during the decomposition of TCA lead to CRC. However, our results showed that TCA also has a direct effect on colon cells, as shown in esophageal cancer or cholangiocarcinoma [40,41]. ...
Preprint
Full-text available
An increased incidence of colorectal cancer (CRC) has been observed among cholecystectomized patients. However, the exact mechanism is not fully understood. Therefore, this study aimed to compare the effects of hepatic and gallbladder bile on normal and cancerous colon cells and to separately examine the effects of the most common bile acids (BAs). The concentrations of BAs in hepatic and gallbladder bile were determined using high-performance liquid chromatography. The effects of bile and individual BAs (100, 300, 500, and 1000 µM) on the viability, proliferation, and metastatic potential of a normal cell line (FHC) and two adenocarcinoma (Caco-2 and FHC-116) cell lines were investigated using different cell assays. No difference was observed in the effects of hepatic and gallbladder bile on the proliferation of normal and adenocarcinoma cells. Among the BAs, taurine-conjugated BAs, especially taurocholic acid, time-dependently increased the proliferation of normal and adenocarcinoma cells and the metastatic potential of cancer cells. The study results showed that the increased risk of CRC in cholecystectomized patients is not due to bile composition changes, but elevated levels of taurine-conjugated BAs may contribute to the development or progression of CRC.
... Moreover, it inhibited S1P-mediated stimulation of ERK1/2 in MDA-MB-453 breast cancer cells expressing S1P 2 and S1P 4 and improved chemotherapy efficacy in Ewing sarcoma (Pyne and Pyne 2011;Marmonti et al. 2020). In cholangiocarcinoma, it prevented disease progression through COX-2 inhibition (Liu et al. 2015). ...
Chapter
Sphingolipids, particularly their two main bioactive metabolites ceramide and sphingosine-1-phosphate (S1P), play a key role in cancer death and survival. Ceramides induce cancer cell death through apoptosis, autophagy, or necroptosis. S1P on the other hand inhibits apoptosis and stimulates proliferation, migration, metastasis, and drug resistance via receptor-dependent or receptor-independent pathways. Modulating the cellular sphingolipidome, through targeting specific enzymes or metabolites, is emerging as a promising pharmacological intervention that could limit cancer progression and improve disease outcomes. In this chapter, we highlight new pharmacological tools that have the potential to modulate key sphingolipid enzymes and metabolites to be used in cancer treatment.
... Mice deficient in either S1pr2 or Sphk2 develop rapid steatosis on a high-fat diet [83]. In cholangiocarcinoma cell lines, conjugated bile acids activated S1PR2 and upregulated cyclooxygenase 2, which led to invasive growth and bile duct proliferation [84,85]. The TCA-mediated activation of ERK1/2 and AKT in cholangiocytes was inhibited by S1PR2 antagonists or siRNA knockdown, which also inhibited cholangiocyte proliferation and migration ( Figure 2C) [32]. ...
Article
Full-text available
Biliary fibrosis is the driving pathological process in cholangiopathies such as primary biliary cholangitis (PBC) and primary sclerosing cholangitis (PSC). Cholangiopathies are also associated with cholestasis, which is the retention of biliary components, including bile acids, in the liver and blood. Cholestasis may worsen with biliary fibrosis. Furthermore, bile acid levels, composition and homeostasis are dysregulated in PBC and PSC. In fact, mounting data from animal models and human cholangiopathies suggest that bile acids play a crucial role in the pathogenesis and progression of biliary fibrosis. The identification of bile acid receptors has advanced our understanding of various signaling pathways involved in regulating cholangiocyte functions and the potential impact on biliary fibrosis. We will also briefly review recent findings linking these receptors with epigenetic regulatory mechanisms. Further detailed understanding of bile acid signaling in the pathogenesis of biliary fibrosis will uncover additional therapeutic avenues for cholangiopathies.
Article
Full-text available
Bile acids, once considered mere dietary surfactants, now emerge as critical modulators of macronutrient (lipid, carbohydrate, protein) metabolism and the systemic pro-inflammatory/anti-inflammatory balance. Bile acid metabolism and signaling pathways play a crucial role in protecting against, or if aberrant, inducing cardiometabolic, inflammatory, and neoplastic conditions, strongly influencing health and disease. No curative treatment exists for any bile acid influenced disease, while the most promising and well-developed bile acid therapeutic was recently rejected by the FDA. Here, we provide a bottom-up approach on bile acids, mechanistically explaining their biochemistry, physiology, and pharmacology at canonical and non-canonical receptors. Using this mechanistic model of bile acids, we explain how abnormal bile acid physiology drives disease pathogenesis, emphasizing how ceramide synthesis may serve as a unifying pathogenic feature for cardiometabolic diseases. We provide an in-depth summary on pre-existing bile acid receptor modulators, explain their shortcomings, and propose solutions for how they may be remedied. Lastly, we rationalize novel targets for further translational drug discovery and provide future perspectives. Rather than dismissing bile acid therapeutics due to recent setbacks, we believe that there is immense clinical potential and a high likelihood for the future success of bile acid therapeutics.
Article
Background Ductular reactivity is central to the pathophysiology of cholangiopathies. Mechanisms underlying the reactive phenotype activation by exogenous inflammatory mediators and bile acids are poorly understood. Methods Using human extrahepatic cholangiocyte organoids (ECOs) we developed an injury model emulating the cholestatic microenvironment with exposure to inflammatory mediators and various pathogenic bile acids. Moreover, we explored roles for the bile acid activated Sphingosine-1-phosphate receptor 2 (S1PR2) and potential beneficial effects of therapeutic bile acids UDCA and norUDCA. Results Synergistic exposure to bile acids (taurocholic acid, glycocholic acid, glycochenodeoxycholic acid) and TNF-α for 24 hours induced a reactive state as measured by ECO diameter, proliferation, lactate dehydrogenase activity and reactive phenotype markers. While NorUDCA and UDCA treatments given 8 hours after injury induction both suppressed reactive phenotype activation and most injury parameters, proliferation was improved by NorUDCA only. Extrahepatic cholangiocyte organoid stimulation with S1PR2 agonist sphingosine-1-phosphate reproduced the cholangiocyte reactive state and upregulated S1PR2 downstream mediators; these effects were suppressed by S1PR2 antagonist JET-013 (JET), downstream mediator extracellular signal-regulated kinase 1/2 inhibitor, and by norUDCA or UDCA treatments. JET also partially suppressed reactive phenotype after bile acid injury. Conclusions We developed a novel model to study the reactive cholangiocyte state in response to pathological stimuli in cholestasis and demonstrated a contributory role of S1PR2 signaling in both injury and NorUDCA/UDCA treatments. This model is a valuable tool to further explore the pathophysiology of human cholangiopathies.
Article
Full-text available
The farnesoid-X receptor (FXR), a member of the nuclear hormone receptor superfamily, can be activated by bile acids (BAs). BAs binding to FXR activates BA signaling which is important for maintaining BA homeostasis. FXR is differentially expressed in human organs and exists in immune cells. The dysregulation of FXR is associated with a wide range of diseases including metabolic disorders, inflammatory diseases, immune disorders, and malignant neoplasm. Recent studies have demonstrated that FXR influences tumor cell progression and development through regulating oncogenic and tumor-suppressive pathways, and, moreover, it affects the tumor microenvironment (TME) by modulating TME components. These characteristics provide a new perspective on the FXR-targeted therapeutic strategy in cancer. In this review, we have summarized the recent research data on the functions of FXR in solid tumors and its influence on the TME, and discussed the mechanisms underlying the distinct function of FXR in various types of tumors. Additionally, the impacts on the TME by other BA receptors such as takeda G protein-coupled receptor 5 (TGR5), sphingosine-1-phosphate receptor 2 (S1PR2), and muscarinic receptors (CHRM2 and CHRM3), have been depicted. Finally, the effects of FXR agonists/antagonists in a combination therapy with PD1/PD-L1 immune checkpoint inhibitors and other anti-cancer drugs have been addressed.
Article
Full-text available
Cholangiocytes are exposed to high concentrations of bile acids at their apical membrane. A selective transporter for bile acids, the Apical Sodium Bile Acid Cotransporter (ASBT) (also referred to as Ibat; gene name Slc10a2) is localized on the cholangiocyte apical membrane. On the basolateral membrane, four transport systems have been identified (t-ASBT, multidrug resistance (MDR)3, an unidentified anion exchanger system and organic solute transporter (Ost) heteromeric transporter, Ostα-Ostβ. Together, these transporters unidirectionally move bile acids from ductal bile to the circulation. Bile acids absorbed by cholangiocytes recycle via the peribiliary plexus back to hepatocytes for re-secretion into bile. This recycling of bile acids between hepatocytes and cholangiocytes is referred to as the cholehepatic shunt pathway. Recent studies suggest that the cholehepatic shunt pathway may contribute in overall hepatobiliary transport of bile acids and to the adaptation to chronic cholestasis due to extrahepatic obstruction. ASBT is acutely regulated by an adenosine 3', 5’-monophosphate (cAMP)-dependent translocation to the apical membrane and by phosphorylation-dependent ubiquitination and proteasome degradation. ASBT is chronically regulated by changes in gene expression in response to biliary bile acid concentration and inflammatory cytokines. Another potential function of cholangiocyte ASBT is to allow cholangiocytes to sample biliary bile acids in order to activate intracellular signaling pathways. Bile acids trigger changes in intracellular calcium, protein kinase C (PKC), phosphoinositide 3-kinase (PI3K), mitogen-activated protein (MAP) kinase and extracellular signal-regulated protein kinase (ERK) intracellular signals. Bile acids significantly alter cholangiocyte secretion, proliferation and survival. Different bile acids have differential effects on cholangiocyte intracellular signals, and in some instances trigger opposing effects on cholangiocyte secretion, proliferation and survival. Based upon these concepts and observations, the cholangiocyte has been proposed to be the principle target cell for bile acids in the liver.
Article
Full-text available
Cholangiocarcinoma (CCA) is a malignant cancer of the biliary tract and its occurrence is associated with chronic cholestasis which causes an elevation of bile acids in the liver and bile duct. The present study aimed to investigate the role and mechanistic effect of bile acids on the CCA cell growth. Intrahepatic CCA cell lines, RMCCA-1 and HuCCA-1, were treated with bile acids and their metabolites to determine the growth promoting effect. Cell viability, cell cycle analysis, EdU incorporation assays were conducted. Intracellular signaling proteins were detected by western immunoblotting. Among eleven forms of bile acids and their metabolites, only taurolithocholic acid (TLCA) concentration dependently (1-40 µM) increased the cell viability of RMCCA-1, but not HuCCA-1 cells. The cell cycle analysis showed induction of cells in the S phase and the EdU incorporation assay revealed induction of DNA synthesis in the TLCA-treated RMCCA-1 cells. Moreover, TLCA increased the phosphorylation of EGFR, ERK 1/2 and also increased the expression of cyclin D1 in RMCCA-1 cells. Furthermore, TLCA-induced RMCCA-1 cell growth could be inhibited by atropine, a non-selective muscarinic acetylcholine receptor (mAChR) antagonist, AG 1478, a specific EGFR inhibitor, or U 0126, a specific MEK 1/2 inhibitor. These results suggest that TLCA induces CCA cell growth via mAChR and EGFR/EKR1/2 signaling pathway. Moreover, the functional presence of cholinergic system plays a certain role in TLCA-induced CCA cell growth.
Article
Full-text available
Cholangiocarcinoma (CCA) is the most common biliary tract malignancy. CCA is classified as intrahepatic, perihilar or distal extrahepatic; the individual subtypes differ in their biologic behavior, clinical presentation, and management. Throughout the last decades, CCA incidence rates had significantly increased. In addition to known established risk factors, novel possible risk factors (i.e. obesity, hepatitis C virus) have been identified that are of high importance in developed countries where CCA prevalence rates have been low. CCA tends to develop on the background of inflammation and cholestasis. In recent years, our understanding of the molecular mechanisms of cholangiocarcinogenesis has increased, thereby, providing the basis for molecularly targeted therapies. In its diagnostic evaluation, imaging techniques have improved, and the role of complementary techniques has been defined. There is a need for improved CCA biomarkers as currently used ones are suboptimal. Multiple staging systems have been developed, but none of these is optimal. The prognosis of CCA is considered dismal. However, treatment options have improved throughout the last two decades for carefully selected subgroups of CCA patients. Perihilar CCA can now be treated with orthotopic liver transplantation with neoadjuvant chemoradiation achieving 5-year survival rates of 68%. Classically considered chemotherapy-resistant, the ABC-02 trial has shown the therapeutic benefit of combination therapy with gemcitabine and cisplatin. The benefits of adjuvant treatments for resectable CCA, local ablative therapies and molecularly targeted therapies still need to be defined. In this article, we will provide the reader with an overview over CCA, and discuss the latest developments and controversies.
Article
Full-text available
Colangiocarcinoma (CCA) is a progressively fatal disease which generally occurs due to malignant transformation of hepatic biliary cholangiocytes. The incidence of CCA has been increasing worldwide and there is an urgent requirement for effective diagnosis and treatment strategies against this devastating disease. Different factors including liver-fluke infestation, viral hepatitis, exogenous nitrosamine-mediated DNA damage, and chronic inflammation have been linked to CCA genesis. However, the risk factors and underlying complex mechanisms leading to development of CCA are not sufficiently understood to devise an effective targeted treatment therapy. In this review, we summarize currently known epidemiological and pathological aspects of the disease and briefly describe various potential biomarkers and experimental anticancer phytochemicals related to CCA research. In addition, we also sum up recent findings that link chronic inflammation of hepatic biliary cholangiocytes with CCA. The collective information concisely presented in this article would provide useful insights into the current understanding of this cancer.
Article
Full-text available
Bile acids are implicated as aetiological factors in many types of gastrointestinal tract cancer including cholangiocarcinoma (CCA). Alterations in bile acid concentrations may affect the pathogenesis of these different types of cancer. Our aim was to determine the bile acid profile in gallbladder bile from patients who underwent liver resection. Thirty-seven patients with cholangiocarcinoma, 5 with hepatocellular carcinoma, and 7 with benign biliary diseases were studied. High pressure liquid chromatography was used to analyze conjugated and unconjugated bile acids. CCA patients with low (≤2 mg/dl) and high (>2 mg/dl) levels of total serum bilirubin had significantly higher total bile acid and conjugated bile acid concentrations than the benign biliary disease group. Markedly elevated levels of cholic and chenodeoxycholic acid were found in CCA cases with high levels of total serum bilirubin. Concentrations of total bile acids and primary bile acid were correlated with serum cholesterol, bilirubin and ALP in CCA. Notably, correlation of the carcinoembryonic antigen, a tumor marker, was found with level of total bile acids and chenodeoxycholic acid. These findings suggest a different pattern of bile acid concentration in cancer patients compared to patients with benign biliary diseases. Thus, accumulation of certain bile acids may be involved in carcinogenesis.
Article
Full-text available
Endosulfan (1,4,5,6,7,7-hexachloro-8,9,10-trinorborn-5-en-2,3-ylenebismet-hylene) is correlated with endocrine disruption, reproductive, and immune dysfunctions. Recently, endosulfan was shown to have an effect on inflammatory pathways, but its influence on cyclooxygenase-2(COX-2) expression is unclear. This study investigated the effects of COX-2 and molecular mechanisms by endosulfan in murine macrophage RAW 264.7 cells. Endosulfan significantly induced COX-2 protein and mRNA levels, as well as COX-2 promoter-driven luciferase activity and the production of prostaglandin E2, a major COX-2 metabolite. Transfection experiments with several human COX-2 promoter constructs revealed that endosulfan activated NF-κB, C/EBP, AP-1, and CREB. Moreover, Akt and mitogen-activated protein kinases (MAPK) were significantly activated by endosulfan. Moreover, endosulfan increased production of the ROS and the ROS-producing NAPDH-oxidase (NOX) family oxidases, NOX2, and NOX3. Endosulfan-induced Akt/MAPK pathways and COX-2 expression were attenuated by DPI, a specific NOX inhibitor, and the ROS scavenger N-acetylcysteine. These results demonstrate that endosulfan induces COX-2 expression via NADPH oxidase, ROS, and Akt/MAPK pathways. These findings provide further insight into the signal transduction pathways involved in the inflammatory effects of endosulfan.
Article
Full-text available
Background: Stratification of patients for treatment of ductal carcinoma in situ (DCIS) is suboptimal, with high systemic overtreatment rates. Methods: A training set of 95 tumours from women with pure DCIS were immunostained for proteins involved in cell survival, hypoxia, growth factor and hormone signalling. A generalised linear regression with regularisation and variable selection was applied to a multiple covariate Cox survival analysis with recurrence-free survival 10-fold cross-validation and leave-one-out iterative approach were used to build and test the model that was validated using an independent cohort of 58 patients with pure DCIS. The clinical role of a COX-2-targeting agent was then tested in a proof-of-concept neoadjuvant randomised trial in ER-positive DCIS treated with exemestane 25 mg day−1±celecoxib 800 mg day−1. Results: The COX-2 expression was an independent prognostic factor for early relapse in the training (HR 37.47 (95% CI: 5.56–252.74) P=0.0001) and independent validation cohort (HR 3.9 (95% CI: 1.8–8.3) P=0.002). There was no significant interaction with other clinicopathological variables. A statistically significant reduction of Ki-67 expression after treatment with exemestane±celecoxib was observed (P<0.02) with greater reduction in the combination arm (P<0.004). Concomitant reduction in COX-2 expression was statistically significant in the exemestane and celecoxib arm (P<0.03) only. Conclusions: In patients with DCIS, COX-2 may predict recurrence, aiding clinical decision making. A combination of an aromatase inhibitor and celecoxib has significant biological effect and may be integrated into treatment of COX2-positive DCIS at high risk of recurrence.
Article
Evidence is accumulating that bile acids induce apoptosis in colonic cells. Therefore, it becomes important to study the underlying molecular mechanisms and the role of this phenomenon in tumor promotion. Minutes after exposure of HCT 116 and HT-29 cells to deoxycholate (DCA), DNA damage, measured using the COMET assay, was evident. Caspase-3 was rapidly activated in HCT 116 cells exposed to DCA, whereas in HT-29 cells, caspase-3 activation was delayed. Using transient transfections with reporter constructs, we showed that the transcription factors activator protein-1 (AP-1) and NF-kB were increased in HCT 116 cells, in a dose-dependent fashion, by DCA COX-2 promoter activity was also induced by DCA and using mutant COX-2 promoter plasmids, we showed that the ability of DCA to induce promoter activity was partly dependent upon a functional NF-kB and C/EBP site, and completely dependent on a functional c-AMP response element site. DNA damage thus appears to be the initiating event in DCA-induced apoptosis. In conclusion, the bile acid, DCA, has a major impact on apoptotic mechanisms in colonic cells and this may be contributing to its effect as a tumor promoter.
Article
It has become increasingly apparent of late that inflammation plays an integral role in a spectrum of malignancies including cholangiocarcinoma (CCA). Primary sclerosing cholangitis with chronic inflammation is the most common risk factor for CCA in the Western world. Recent work has highlighted that inflammatory pathways are essential in carcinogenesis and tissue invasion and migration. Inflammation advances carcinogenesis by induction of DNA damage, evasion of apoptosis, promotion of cell proliferation, and neoangiogenesis. CCA is characterized by the presence of a desmoplastic stroma consisting of cancer-associated fibroblasts, tumor-associated macrophages, and tumor-infiltrating lymphocytes. This rich inflammatory milieu is vital to the cancer ecosystem, and targeting its components represents an attractive therapeutic option. © 2014 S. Karger AG, Basel.