You are on page 1of 10

Biochemistry and Molecular Biology

Sphingosine-1-Phosphate Mediates Fibrosis in Orbital


Fibroblasts in Graves’ Orbitopathy
JaeSang Ko,1 Min Kyoung Chae,1 Joon H. Lee,2 Eun Jig Lee,3 and Jin Sook Yoon1
1
Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul, Korea
2
Myung-gok Eye Research Institute, Konyang University College of Medicine, Seoul, Korea
3Department of Endocrinology, Severance Hospital, Institute of Endocrine Research, Yonsei University College of Medicine, Seoul,

Korea

Correspondence: Jin Sook Yoon, PURPOSE. To investigate the effect of sphingosine-1-phosphate (S1P) on fibrosis in orbital
Department of Ophthalmology, Sev- fibroblasts in Graves’ orbitopathy (GO).
erance Hospital, Institute of Vision
Research, Yonsei University College METHODS. Orbital fibroblasts were cultured from orbital adipose/connective tissues of patients
of Medicine, 50–1 Yonsei-ro, Seo- with GO and healthy control subjects. Effects of treatment with TGF-b and cigarette smoke
daemun-gu, Seoul 03722, Korea; extract (CSE) on S1P receptor (S1PR) messenger RNA (mRNA) and S1P expression were
yoonjs@yuhs.ac. evaluated by real-time polymerase chain reaction and Western blotting. To evaluate the role of
Submitted: September 4, 2016 S1P in fibrosis, cells were pretreated with W146 (S1PR1 antagonist); JTE013 (S1PR2
Accepted: April 4, 2017 antagonist); FTY720 (S1PR1 modulator); or 5C (sphingosine kinase-1 blocker) for 1 hour
Citation: Ko J, Chae MK, Lee JH, Lee
before stimulation with TGF-b, CSE, or IL-1b. Expression of fibrosis-related proteins (collagen
EJ, Yoon JS. Sphingosine-1-phosphate Ia, fibronectin, and a-smooth muscle actin [SMA]) and tissue remodeling–related proteins
mediates fibrosis in orbital fibroblasts (matrix metalloproteinases [MMPs] and tissue inhibitor of metalloproteinase [TIMP]-1) was
in Graves’ orbitopathy. Invest Oph- then evaluated by Western blotting.
thalmol Vis Sci. 2017;58:2544–2553. RESULTS. Expression levels of S1PR mRNA and S1P in GO orbital fibroblasts increased upon
DOI:10.1167/iovs.16-20684
TGF-b and CSE treatment. Treatment with S1PR blockers and 5C inhibited TGF-b and CSE-
induced expression of collagen Ia, fibronectin, and a-SMA, as well as IL-1b–induced
expression of MMP-1, MMP-2, MMP-9, and TIMP-1. Exogenous S1P treatment without
profibrotic stimulants upregulated collagen Ia, fibronectin, a-SMA, MMP-1, MMP-2, MMP-9,
and TIMP-1 expression in a dose-dependent manner.
CONCLUSIONS. Blocking of S1PR activity and inhibition of S1P synthesis led to decreased
expression of fibrosis and tissue remodeling–related proteins in primary cultures of orbital
fibroblasts derived from patients with GO. Thus, modulation of S1P activity might have
therapeutic potential in the suppression of fibrosis in GO.
Keywords: fibrosis, Graves’ orbitopathy, sphingosine-1-phosphate

raves’ orbitopathy (GO), an autoimmune component of isoforms of SphK, SphK1 and SphK2, the latter has approx-
G Graves’ disease, occurs in up to 50% of patients with
Graves’ disease.1 It is characterized by inflammation and
imately 10-fold lower specific activity than SphK1.7 Synthesis of
S1P occurs intracellularly in organelles and on the inner leaflet
swelling of orbital tissue, with fibrosis and adipogenesis being of the plasma membrane. It acts as an intracellular second
predominant features.2–4 Current evidence indicates orbital messenger, mediating calcium homeostasis and apoptosis.5,8
fibroblasts as the key effector cells in GO. Although it is widely Additionally, S1P is secreted extracellularly through the
known that inflammation and adipogenesis are key participants adenosine triphosphate–binding cassette transporter superfam-
in the pathogenesis of GO, little is known about its fibrogenic ily and reaches the outer leaflet of the plasma membrane,6
mechanism. where it binds to S1P receptors (S1PRs) and acts as an
Three major classes of lipids—glycerolipids, sphingolipids, autocrine and paracrine modulator.6,9,10 Although S1P has a
and sterols—constitute the double-layered surface membrane dual mechanism of action as an intracellular second and
of all eukaryotic cells.5 It was previously believed that lipids extracellular first messenger, its greatest effect is exerted
play exclusive roles in energy metabolism and membrane extracellularly through binding with S1PRs.11 The five S1PRs,
composition. Recent studies, however, have elucidated the S1PR1, -2, -3, -4, and -5, exhibit selective tissue expression,
concept of ‘‘bioactive lipids’’ and identified the biologically which is crucial for their biological function, and employ well-
active sphingosine-1-phosphate (S1P), which regulates diverse known G-protein coupled receptor intracellular signaling
cellular processes including cytoskeletal rearrangement, cell pathways to mediate their specific effect.6,9,12
survival and migration, and inflammation.5,6 Cellular concen- Emerging evidence indicates that S1P acts on several types
trations of S1P are largely controlled through S1P synthesis by of target cells and is engaged in fibrogenic processes in the
phosphorylation of sphingosine catalyzed by sphingosine liver,13,14 kidney,15 lung,16 and cardiac muscles.17 Occurrence
kinase (SphK) and, to a lesser extent, by S1P degradation of SphK and S1PR has also been reported in RPE and
catalyzed by S1P phosphatase or S1P lyase.7 Of the two conjunctival and corneal fibroblasts.18 In retinal pigment

Copyright 2017 The Authors


iovs.arvojournals.org j ISSN: 1552-5783 2544

This work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.

Downloaded From: https://iovs.arvojournals.org/ on 07/23/2018


Sphingosine-1-Phosphate Mediates Fibrosis in GO IOVS j May 2017 j Vol. 58 j No. 5 j 2545

TABLE. Clinical Characteristics of the Patients Included in This Study

Age, y Sex Duration of GO, y CAS Previous GO Treatment Surgical Treatment

Patients with GO
29 F 0.7 3/7 GC Decompression
34 F 1.6 1/7 GC Decompression
41 F 0.9 1/7 GC Decompression
44 M 1.6 2/7 GC Decompression
44 F 1.5 1/7 GC Decompression
52 M 3.4 3/7 None Decompression
57 F 3.1 1/7 GC Decompression
59 F 2.1 0/7 GC Decompression
Control subjects
30 F N/A 0 N/A Orbital wall fracture
36 F N/A 0 N/A Upper lid blepharoplasty
43 M N/A 0 N/A Evisceration
44 F N/A 0 N/A Upper lid blepharoplasty
49 F N/A 0 N/A Orbital wall fracture
52 F N/A 0 N/A Upper lid blepharoplasty
59 F N/A 0 N/A Upper lid blepharoplasty
64 F N/A 0 N/A Upper lid blepharoplasty
CAS, clinical activity score; GO, Graves’ orbitopathy; GC, glucocorticoids; N/A, not applicable.

epithelium, S1P engages in proliferation and profibrotic with GO received steroid treatment or radiotherapy for at least
protein expression.18 In a previous study, anti-S1P monoclonal 3 months before surgery. For control orbital tissues, we tried to
antibodies inhibited subretinal fibrosis in a murine model of match the clinical characteristics of non-GO patients and
laser-induced choroidal neovascularization.19 patients with GO; however, because of the limited number of
We previously demonstrated the expression S1PRs in GO donors, it was not possible to achieve perfect matching. The
orbital fibroblasts as well as the role of S1P in the institutional review board of the Severance Hospital, Yonsei
differentiation of orbital adipocytes.20 In the present study, University College of Medicine, Seoul, Korea, approved the
we aimed to determine whether S1P mediates fibrogenic study, and written informed consent was obtained from all
processes in GO orbital fibroblasts and whether blocking of participants after explanation of the nature and possible
S1P and S1PRs decreases fibrosis in GO. consequences of the study. This study followed the tenets of
the Declaration of Helsinki.
Primary cultures of orbital fibroblasts were established as
MATERIALS AND METHODS described in our previous study.20 Briefly, minced tissue was
placed directly in 1:1 DMEM:F12 medium with 20% FBS and
Reagents antibiotics. Upon growth of fibroblasts from the explants,
Dulbecco’s modified Eagle’s medium (DMEM); fetal bovine monolayers were passaged serially with trypsin/ethylenedi-
serum (FBS); penicillin; and gentamycin were purchased from aminetetraacetic acid solution, and cultures were maintained
Hyclone Laboratories, Inc. (Logan, UT, USA). Recombinant in DMEM with 10% FBS and antibiotics. Cells between the
human TGF-b and IL-1b were purchased from R&D Systems, second and fifth passages were used for analysis.
Inc. (Minneapolis, MN, USA). Sphingosine-1-phosphate,
FTY720, an S1PR1 modulator that causes internalization of Real-Time Polymerase Chain Reaction
S1PR1, and 5C, an SphK1 blocker, were purchased from Sigma-
Aldrich Corp. (St. Louis, MO, USA). We purchased W146, a Expression levels of S1PR1, -2, and -3 were evaluated by real-
trifluoroacetate salt that acts as an S1PR1 antagonist, and time PCR. Isolation of RNA and real-time PCR were performed
JTE013, an S1PR2 antagonist, from Cayman Chemical (Ann as described previously.20 All PCR reactions were performed in
Arbor, MI, USA). Cigarette smoke extract (CSE) was freshly triplicate, and all samples were normalized to glyceraldehyde-3-
prepared within an hour of each experiment from commer- phosphate dehydrogenase (GAPDH) expression levels. Expres-
cially available filtered cigarettes (Marlboro 20 class A sion levels were determined by the 2–DDCt method as fold
cigarettes [8.0 mg tar; 0.7 mg nicotine]; Philip Morris Korea, change of threshold cycle (Ct) value relative to the control.
Inc., Seoul, Korea) as described in our previous study.20 Amplification bands were quantified by densitometry and
normalized against corresponding GAPDH bands to control for
PCR variability. The primers used for amplification were as
Cell Culture Protocols
follows: human S1PR1, 5 0 -TGC GGG AAG GGA GTA TGT TT-3 0
Orbital adipose/connective tissue explants were obtained as (forward) and 5 0 -CCA TCC CCA CCA CAC TCA AC-3 0 (reverse);
surgical waste during decompression surgery from eight human S1PR2, 5 0 -GCC TCT CTA CGC CAA GCA TTA-3 0
patients with GO, and normal control tissues were harvested (forward) and 5 0 -TTG AGC GGA CCA CGC AGT A-3 0 (reverse);
during eyelid or orbital surgery from eight individuals with no human S1PR3, 5 0 -TGA TTG TGG TGA GCG TCT TCA-3 0
history or clinical evidence of thyroid disease or GO (Table). All (forward) and 5 0 -GGC CAC ATC AAT GAG GAA GAG-3 0
eight patients with GO had achieved stable euthyroidism at the (reverse); and GAPDH 5 0 -GCC AAG GTC ATC CAT GAC AAC-
time of surgery, and their clinical activity scores at the time of 3 0 (forward) and 5 0 -GTC CAC CAC CCT GTT GCT GTA-3 0
surgery were less than 4. Additionally, none of the patients (reverse).

Downloaded From: https://iovs.arvojournals.org/ on 07/23/2018


Sphingosine-1-Phosphate Mediates Fibrosis in GO IOVS j May 2017 j Vol. 58 j No. 5 j 2546

Wilcoxon rank-sum test using a statistical program (SPSS for


Windows, version 16; SPSS, Chicago, IL, USA). Values of P <
0.05 were considered significant.

RESULTS
Effect of TGF-b and CSE Treatment on S1PR1, -2,
and -3 mRNA Expression in Orbital Fibroblasts
To determine the effect of TGF-b and CSE treatment on S1PR1,
-2, and -3 mRNA expression, orbital fibroblasts derived from
individuals with and without GO were treated with TGF-b (5
ng/mL) or CSE (2.5%) for 16 hours. Levels of mRNA expression
of S1PR1, -2, and -3 were compared between treated and
untreated cells by real-time PCR analysis. Although TGF-b
induced a marked increase in S1PR1 mRNA expression in both
GO and non-GO orbital fibroblasts, the increment in non-GO
cells was lower compared to that in GO cells. Expression of
S1PR2 mRNA in TGF-b–treated GO cells was also significantly
increased (Fig. 1A). Treatment with CSE resulted in a marked
FIGURE 1. Effect of TGF-b and CSE treatment on mRNA expression of increase of S1PR1, -2, and -3 expression in both GO and non-
S1PR1–3 in orbital fibroblasts. Orbital fibroblasts of individuals with
GO orbital fibroblasts, with the degree of induction in GO cells
(black columns) and without (white columns) GO were treated with 5
ng/mL TGF-b or 2.5% CSE for 16 hours. We compared mRNA being higher compared to that in non-GO cells (Fig. 1B).
expression levels of S1PR1, -2, and -3 in (A) TGF-b– and (B) CSE-
treated cells with those in untreated cells by real-time PCR. Data in the Effect of TGF-b Treatment on S1P Expression in
columns indicate the mean relative fold of mRNA levels 6 SD of three
experiments (*P < 0.05 and **P < 0.01 versus untreated control cells).
Orbital Fibroblasts
The effect of TGF-b treatment on S1P expression in GO and
Western Blotting non-GO orbital fibroblasts was evaluated by Western blotting.
Dose- and time-dependent effects of TGF-b treatment on S1P
Western blot analysis of S1P, SphK1, collagen Ia, fibronectin, a- expression were evaluated by comparison of S1P expression
smooth muscle actin (SMA), matrix metalloproteinase (MMP)- levels among GO and non-GO orbital fibroblasts treated with
1, MMP-2, MMP-7, MMP-9, and tissue inhibitor of metal- different concentrations of TGF-b (0–20 ng/mL) for 24 hours or
loproteinase (TIMP)-1 was performed as described previous- 5 ng/mL TGF-b for different time periods (0–24 hours). While
ly.20 The relative quantity of protein in each immunoreactive treatment with 1–10 ng/mL TGF-b for 24 hours had no
band was quantified by densitometry and normalized to the influence on S1P expression in non-GO orbital fibroblasts, TGF-
quantity of b-actin in the same sample. Anti-S1P, anti-SphK1, b induced S1P expression in GO orbital fibroblasts in a dose-
anti-fibronectin, anti-MMP-7, and anti-TIMP-1 antibodies were dependent manner (Fig. 2A). Additionally, while treatment
purchased from Abcam (Cambridge, UK); anti-collagen Ia with 5 ng/mL TGF-b had no effect on S1P expression in non-
antibody from Pierce Biotechnology (Rockford, IL, USA); anti– GO orbital fibroblasts regardless of treatment time, it increased
a-SMA antibody from Dako Corporation (Carpinteria, CA, USA); S1P expression in GO orbital fibroblasts at treatment times ‡16
anti–MMP-2 and anti–MMP-9 antibodies from Cell Signaling hours (Fig. 2B).
Technology (Beverly, MA, USA), and anti–MMP-1 and anti–b-
actin antibodies from Santa Cruz Biotechnology (Dallas, TX, Effect of CSE Treatment on S1P Expression in
USA). Orbital Fibroblasts
Transfection With Short Interfering RNA (siRNA) The effect of CSE-induced oxidative stress on S1P expression in
GO and non-GO orbital fibroblasts was evaluated by Western
We purchased siRNA of SphK1 and negative control siRNA blotting. Dose- and time-dependent effects of CSE treatment on
from Santa Cruz Biotechnology. Approximately 80% confluent S1P expression were determined by comparison of S1P
orbital fibroblasts from patients with GO were prepared in 100- expression levels among GO and non-GO orbital fibroblasts
mm plates. Negative control siRNA or SphK1 siRNA was treated with different concentrations of CSE (0%–10%) for 24
transfected with a commercial reagent (Lipofectamine 2000; hours or 2.5% CSE for different time periods (0–24 hours).
Invitrogen, Carlsbad, CA, USA) in accordance with the Treatment with CSE for 24 hours upregulated S1P expression
manufacturer’s instructions. After transfection, cells were in both GO and non-GO orbital fibroblasts in a dose-dependent
incubated with or without 5 ng/mL TGF-b for 24 hours. manner (Fig. 3A). Additionally, treatment with 2.5% CSE
increased S1P expression in GO and non-GO orbital fibroblasts
Statistical Analysis in a time-dependent manner (Fig. 3B).

All experiments were performed with cells from at least three S1PR and SphK1 Blockers Attenuate Pro-Fibrotic
different samples, with the samples being assayed in duplicate
Proteins in GO Orbital Fibroblasts
or triplicate each time. For statistical analysis of the Western
blotting and real time-PCR results, mean values and standard Expression levels of profibrotic proteins—collagen Ia, fibro-
deviations were calculated for normalized measurements of nectin, and a-SMA—in TGF-b–stimulated GO orbital fibroblasts
each protein or mRNA from at least three different samples. with or without pretreatment with S1PR (W146, JTE013, or
Differences in evaluated variables between the experimental FTY720) or SphK1 (5C) blockers were evaluated by Western
and control groups were assessed by the Student’s t-test or blotting as described previously.21–24 While TGF-b–induced

Downloaded From: https://iovs.arvojournals.org/ on 07/23/2018


Sphingosine-1-Phosphate Mediates Fibrosis in GO IOVS j May 2017 j Vol. 58 j No. 5 j 2547

FIGURE 2. Effect of TGF-b treatment on S1P expression in orbital fibroblasts. Expression levels of S1P in orbital fibroblasts of individuals with (black
columns) and without (white columns) GO were evaluated by Western blotting after treatment with different doses of TGF-b for different time
periods. Expression of S1P in orbital fibroblasts treated with (A) different concentrations of TGF-b (0–20 ng/mL) for 24 hours or (B) 5 ng/mL TGF-b
for different time periods (0–24 hours). Data in the columns indicate the mean density ratios 6 SD of three experiments (*P < 0.05 versus untreated
control cells).

expression of collagen Ia and fibronectin was downregulated Exogenous S1P Promotes Profibrotic and Tissue-
by pretreatment with FTY720 or 5C, TGF-b–induced a-SMA Remodeling Protein Expression in GO Orbital
expression was downregulated by pretreatment with W146, Fibroblasts
JTE013, FTY720, or 5C (Fig. 4A). Upon investigating the effect
of S1PR and SphK1 blockers on oxidative stress–induced Collagen Ia, fibronectin, and a-SMA expression levels in GO
profibrotic protein expression using CSE, CSE-induced expres- orbital fibroblasts treated with varying concentration of S1P
sion of collagen Ia, fibronectin, and a-SMA was found to be were evaluated by Western blotting. Exogenous treatment with
downregulated by pretreatment with W146, JTE013, FTY720, S1P resulted in dose-dependent increases in collagen Ia,
and 5C (Fig. 4B). In addition, we evaluated the expression fibronectin, and a-SMA expression (Fig. 5A).
levels of profibrotic proteins in nonstimulated GO orbital To investigate the role of S1P in tissue remodeling in GO,
fibroblasts with or without pretreatment with S1PR (W146, MMP-1, MMP-2, MMP-9, MMP-7, and TIMP-1 expression levels
JTE013, or FTY720) or SphK1 (5C) blockers, the results of in GO orbital fibroblasts treated with different concentrations
which are presented in Supplementary Figure S1A. of S1P were evaluated by Western blotting. Expression levels of
MMP-1, MMP-2, MMP-9, and TIMP-1 were found to be
significantly increased upon treatment with 5 and 10 lM of
Silencing of SphK1 Expression by siRNA-Mediated S1P (Fig. 5B; Supplementary Fig. S3A).
Inhibition of TGF-b–Induced Profibrotic Protein
Expression S1PR and SphK1 Blockers Attenuate Tissue-
The levels of TGF-b–induced collagen Ia, fibronectin, and a- Remodeling Proteins in GO Orbital Fibroblasts
SMA expression in GO orbital fibroblasts transfected with Expression levels of MMP-1, MMP-2, MMP-9, MMP-7, and TIMP-
SphK1-targeted siRNA were compared with those in negative 1 in IL-1b–stimulated GO orbital fibroblasts with or without
control siRNA-transfected cells. Western blotting findings pretreatment with W146, JTE013, FTY720, or 5C were
revealed that transfection with SphK1 siRNA resulted in evaluated by Western blotting. We found that S1PR blockers
downregulation of SphK1 and S1P expression. In addition, and 5C inhibited the IL-1b–induced expression of MMP-1,
TGF-b–induced collagen Ia and a-SMA expression levels were MMP-2, MMP-9, and TIMP-1, although their specific actions of
significantly decreased in SphK1-knockdown cells (Supple- the inhibitors were varied (Fig. 6; Supplementary Fig. S3B).
mentary Fig. S2). Trifluoroacetate salt W146 inhibited IL-1b–induced expression

Downloaded From: https://iovs.arvojournals.org/ on 07/23/2018


Sphingosine-1-Phosphate Mediates Fibrosis in GO IOVS j May 2017 j Vol. 58 j No. 5 j 2548

FIGURE 3. Effect of CSE treatment on S1P expression in orbital fibroblasts. Expression levels of S1P in orbital fibroblasts of individuals with (black
columns) and without (white columns) GO were evaluated by Western blotting after treatment with different doses of CSE for different time
periods. Expression of S1P in orbital fibroblasts treated with (A) different concentrations of CSE (0%–10%) for 24 hours or (B) 2.5% CSE for different
time periods (0–24 hours). Data in the columns indicate the mean density ratios 6 SD of three experiments (*P < 0.05 versus untreated control
cells).

of MMP-2, MMP-9, and TIMP-1; JTE013 inhibited IL-1b–induced S1PR expression.10,11 For example, TGF-b1 upregulates SphK1
expression of MMP-1, MMP-9, and TIMP-1; FTY720 inhibited IL- expression in murine myoblasts in an Smad-dependent manner
1b–induced expression of MMP-1, MMP-2, MMP-9, and TIMP-1; and concomitantly modifies S1PR expression by downregulat-
and 5C inhibited IL-1b–induced expression of MMP-2 and ing S1PR1 and upregulating S1PR3 mRNA expression.28 In this
TIMP-1. Pretreatment with S1PR blockers or 5C had no effect report, we have demonstrated that TGF-b stimulates S1PR1,
on MMP-7 expression. In addition, the expression levels of and -2 mRNA expression and S1P protein expression in GO
tissue-remodeling proteins in nonstimulated GO orbital fibro- orbital fibroblasts. Our data indicate a connection between S1P
blasts with or without pretreatment with W146, JTE013, and TGF-b signaling in orbital fibroblasts.
FTY720, or 5C were evaluated, the results of which are The profibrotic potential of fibroblasts is characterized by
presented in Supplementary Figure S1B. their capability to synthesize ECM components such as the
collagen family of proteins and fibronectin. Alpha-SMA is a
marker that directs the differentiation of fibroblasts into
DISCUSSION myofibroblasts, which are key effector cells in fibrogenesis.29
Although pathogenesis of GO involves inflammation, adipo- The downregulation of TGF-b–induced collagen Ia and
genesis, and fibrosis, glucocorticoids remain the mainstay of fibronectin expression by FTY720 and 5C in the present study
medical treatment for GO. Glucocorticoids are primarily indicates that the S1P/S1PR1 pathway plays a role in mediating
effective in active inflammatory conditions and have a limited TGF-b–induced ECM synthesis. Additionally, given that
role in the treatment of fibrosis of orbital connective tissues.25 FTY720, 5C, and JTE013 exhibited an inhibitory effect on
Here, we investigated the role of S1P, a potential therapeutic TGF-b–induced a-SMA expression, both S1P/S1PR1 and S1P/
target, as a mediator of fibrosis in GO. S1PR2 pathways seem to serve as mediators of TGF-b–induced
As a central mediator of fibrotic diseases, TGF-b induces myofibroblast differentiation. In this study, FTY720 and W146
fibroblasts to synthesize extracellular matrix (ECM).26 It exhibited different results in the experiment on S1PR1
regulates the expression of profibrotic proteins—collagen I, inhibition, as demonstrated in Figure 4A. Trifluoroacetate salt
collagen IV, connective tissue growth factor, and fibronectin— W146 is a specific S1PR1 antagonist, competitive for S1P.30 On
through the Smad and rho/rho-associated protein kinase the other hand, FTY720 causes persistent activation of S1PR1,
pathways.10,26 Additionally, there is evidence of cross talk followed by internalization and downregulation of the receptor
between S1PRs and TGF-b–induced fibrosis.27 Not only does protein.31 The inconsistencies in results between the two
TGF-b regulate S1P production through modification of SphK compounds might be due to their different mechanisms of
expression and activity in fibrotic lesions, but it also modifies action.

Downloaded From: https://iovs.arvojournals.org/ on 07/23/2018


Sphingosine-1-Phosphate Mediates Fibrosis in GO IOVS j May 2017 j Vol. 58 j No. 5 j 2549

FIGURE 4. Effect of S1PR and SphK1 blockers on expression of TGF-b– or CSE-induced profibrotic proteins in orbital fibroblasts in GO. Confluent
orbital fibroblasts derived from individuals with GO were either untreated or pretreated with 10 lM W146, 10 lM JTE013, 1 lM FTY720, or 10 lM
5C for 1 hour prior to treatment with TGF-b (5 ng/mL) or CSE (2.5%) for 24 hours. Collagen-Ia, fibronectin, and a-SMA levels in (A) TGF-b– and (B)
CSE-treated cultured cells were assayed by Western blotting. Data in the columns indicate the mean relative density ratios 6 SD of three
experiments (*P < 0.05 and **P < 0.01 versus TGF-b or CSE-treated cells without pretreatment).

Cigarette smoking is the most important risk factor for the related protein expression, the S1P/S1PR1 and -2 pathway
development and deterioration of GO; smoking activates might be a part of the mechanism underlying the profibrotic
pathways associated with adipogenesis, inflammation, and effect of smoking in GO. Moreover, exogenous S1P treatment
fibrosis.32–34 Treatment with CSE induces oxidative stress in of GO orbital fibroblasts without TGF-b or CSE stimulation
orbital fibroblasts by generating reactive oxygen species.32,35 resulted in upregulation of collagen Ia, fibronectin, and a-SMA
Oxidative stress influences S1P expression: moderate levels of expression. Therefore, S1P also seems to have a direct effect on
oxidative stress increase S1P expression by activation of fibrosis in GO orbital fibroblasts, without stimulation by TGF-b
SphK1, and excessive oxidative stress decreases S1P expres- or CSE.
sion by inducing SphK1 degradation.36 Treatment with CSE Remodeling of ECM as well as excessive ECM production
also increases S1PR2, -3, -4, and -5 mRNA expression as well as are important factors in the process of tissue fibrosis.
SphK1 and SphK2 expression in THP-1 macrophages.37,38 Remodeling of ECM is largely affected by the activities of
Given that CSE treatment increased S1PR1, -2, and -3 mRNA proteinases that can degrade matrix, such as MMPs and their
expression in GO and non-GO orbital fibroblasts and upregu- inhibitor, TIMP.39,40 There is excellent evidence in both human
lated S1P expression in GO orbital fibroblasts (Figs. 1B, 3), the liver disease and animal models, indicating that hepatic fibrosis
S1P/S1PR pathway seems to play a role in response to is potentially reversible.41 Decline in TIMP levels, which tips
oxidative stress in GO orbital fibroblasts. Additionally, since the overall MMP–TIMP balance toward MMP, results in
W146, JTE013, FTY720, and 5C inhibited CSE-induced fibrosis- increased matrix degradation and net degradation of scar

Downloaded From: https://iovs.arvojournals.org/ on 07/23/2018


Sphingosine-1-Phosphate Mediates Fibrosis in GO IOVS j May 2017 j Vol. 58 j No. 5 j 2550

FIGURE 5. Effect of S1P on expression of profibrotic and tissue remodeling proteins in orbital fibroblasts in GO. Confluent orbital fibroblasts derived
from individuals with GO were treated with different concentrations of S1P (0–10 lM) for 16 hours. Expression levels of (A) collagen Ia, fibronectin,
and a-SMA and (B) MMP-2, MMP-9, MMP-7, and TIMP-1 were then evaluated by Western blotting. Data in the columns indicate the mean relative
density ratios 6 SD of three experiments (*P < 0.05 and **P < 0.01 versus untreated control cells).

tissue.42,43 However, very few studies have investigated tissue the ECM.39 However, to date, expression of tissue remodeling
remodeling in GO. Han et al.44 reported that IL-1b treatment of proteins in GO orbital fibroblasts has only been reported in
orbital fibroblasts increases TIMP-1 expression, thus disrupting relation to TIMP-1, MMP-2, and MMP-9.34,44,45 The present
the balance between MMPs and TIMP. Additionally, treatment results demonstrate that IL-1b stimulation can induce MMP-1
with antioxidants decreases the levels of MMPs and TIMP- expression in GO orbital fibroblasts and that S1P is involved in
1.34,45 The results of the present study demonstrated that IL-1b this induction process. It is possible that S1P-mediated
increases MMP-2, MMP-9, and TIMP-1 expression in GO orbital upregulation or S1PR blocker–mediated suppression of colla-
fibroblasts, which is concordant with the findings of previous gen Ia and MMP-1 expression are related; however, this aspect
studies.34,46 Since treatment with W146, JTE013, FTY720, and has not been investigated in this study. Further confirmation of
5C resulted in the downregulation of IL-1b–induced expression related pathways is necessary. Moreover, MMP expression
of MMP-1, MMP-2, MMP-9, and TIMP-1, we believe that S1P varies depending on the organ, and while some MMPs are
might act as a mediator of inflammation-induced tissue indeed antifibrotic, others might have profibrotic functions.40
remodeling. Additionally, since exogenous S1P treatment For example, in the kidney, MMP-9 has profibrotic activity,
increased the expression of MMP-1, MMP-2, MMP-9, and while MMP-2 has an antifibrotic function.47,48 Therefore,
TIMP-1, we assume that S1P also has a direct effect on detailed studies on elucidating the function of MMPs in orbital
stimulation of tissue remodeling. Matrix metalloproteinase-1, fibroblasts are required.
also known as interstitial collagenase, degrades fibrillar Orbital fibroblasts can be divided into two subsets on the
collagen types I, II, and III, which are major components of basis of surface expression of Thy-1, a surface glycoprotein.49

Downloaded From: https://iovs.arvojournals.org/ on 07/23/2018


Sphingosine-1-Phosphate Mediates Fibrosis in GO IOVS j May 2017 j Vol. 58 j No. 5 j 2551

demonstrated that S1P plays a role in TGF-b–induced


myofibroblast differentiation, which is exclusive to Thy-1þ
cells. Moreover, we have demonstrated in our previous report
that S1P is also involved in the adipogenesis of GO orbital
fibroblasts, which is exclusive to Thy-1 cells.20 Therefore, S1P
appears to play a role in the pathogenesis of GO in both Thy-1þ
and Thy-1 orbital fibroblasts.
Orbital tissues of patients with GO are continuously
exposed to proinflammatory and oxidative stress.1,51 Although
cultured orbital fibroblasts derived from individuals with and
without GO often exhibit phenotypic differences, primary
cultures of orbital fibroblasts might not directly reflect the in
vivo inflammatory or fibrotic conditions. Because of this
limitation, it is inevitable that profibrotic or proinflammatory
conditions are induced with stimulants to observe the effects
of certain chemicals. In the present study, some of the
evaluated S1PR blockers inhibited the expression of profibrotic
and tissue-remodeling proteins in nonstimulated cells. Howev-
er, in most cases, the inhibitory effects of S1P blockers in TGF-
b–, CSE-, or IL-1b–stimulated conditions were more prominent
than those in the nonstimulated condition.
In conclusion, our results provide evidence indicating that
S1P plays an important role in orbital tissue fibrosis in GO
through induction of fibrosis- and tissue remodeling–related
protein expression. These findings indicate that S1P blocking
agents might have therapeutic potential in the suppression of
fibrosis in GO.

Acknowledgments
Supported by a grant of the Korea Health Technology R&D Project
through the Korea Health Industry Development Institute (KHIDI),
funded by the Ministry of Health & Welfare, Republic of Korea
(grant number HI14C1324), and by the Bio & Medical Technology
Development Program of the NRF funded by the Korean
government, MSIP(2015M3A9E2067031).
Disclosure: J. Ko, None; M.K. Chae, None; J.H. Lee, None; E.J.
Lee, None; J.S. Yoon, None

References
1. Garrity JA, Bahn RS. Pathogenesis of Graves ophthalmopathy:
implications for prediction, prevention, and treatment. Am J
Ophthalmol. 2006;142:147–153.
2. Wiesweg B, Johnson KT, Eckstein AK, Berchner-Pfannschmidt
U. Current insights into animal models of Graves’ disease and
orbitopathy. Horm Metab Res. 2013;45:549–555.
3. Li H, Fitchett C, Kozdon K, et al. Independent adipogenic and
contractile properties of fibroblasts in Graves’ orbitopathy: an
in vitro model for the evaluation of treatments. PLoS One.
2014;9:e95586.
4. Dik WA, Virakul S, van Steensel L. Current perspectives on the
FIGURE 6. Effect of S1PR and SphK1 blockers on expression of IL-1b– role of orbital fibroblasts in the pathogenesis of Graves’
induced tissue remodeling proteins in orbital fibroblasts in GO. ophthalmopathy. Exp Eye Res. 2016;142:83–91.
Confluent orbital fibroblasts derived from individuals with GO were
5. Lahiri S, Futerman AH. The metabolism and function of
either untreated or pretreated with 10 lM W146, 10 lM JTE013, 1 lM
FTY720, or 10 lM 5C for 1 hour prior to treatment with IL-1b (10 ng/ sphingolipids and glycosphingolipids. Cell Mol Life Sci. 2007;
mL, 24 hours). Expression levels of MMP-2, MMP-9, MMP-7, and TIMP-1 64:2270–2284.
were then evaluated by Western blotting. Data in the columns indicate 6. Hannun YA, Obeid LM. Principles of bioactive lipid signalling:
the mean relative density ratios 6 SD of three experiments (*P < 0.05 lessons from sphingolipids. Nat Rev Mol Cell Biol. 2008;9:
and **P < 0.01 versus IL-1b–treated cells without pretreatment). 139–150.
7. Leclercq TM, Pitson SM. Cellular signalling by sphingosine
kinase and sphingosine 1-phosphate. IUBMB Life. 2006;58:
Each subset responds to different extracellular stimuli and 467–472.
differentiates into distinct cell types.46,50 Only Thy-1þ orbital 8. Spiegel S, Milstien S. Sphingosine-1-phosphate: an enigmatic
fibroblasts are capable of differentiation into myofibroblasts signalling lipid. Nat Rev Mol Cell Biol. 2003;4:397–407.
after TGF-b treatment, and only Thy1 cells differentiate into 9. Rosen H, Goetzl EJ. Sphingosine 1-phosphate and its
lipofibroblasts.50 In the present study, although we did not sort receptors: an autocrine and paracrine network. Nat Rev
orbital fibroblasts on the basis of Thy-1 expression, our findings Immunol. 2005;5:560–570.

Downloaded From: https://iovs.arvojournals.org/ on 07/23/2018


Sphingosine-1-Phosphate Mediates Fibrosis in GO IOVS j May 2017 j Vol. 58 j No. 5 j 2552

10. Schwalm S, Pfeilschifter J, Huwiler A. Sphingosine-1-phos- 29. Darby I, Skalli O, Gabbiani G. Alpha-smooth muscle actin is
phate: a Janus-faced mediator of fibrotic diseases. Biochim transiently expressed by myofibroblasts during experimental
Biophys Acta. 2013;1831:239–250. wound healing. Lab Invest. 1990;63:21–29.
11. Takuwa Y, Takuwa N, Sugimoto N. The Edg family G protein- 30. Gonzalez-Cabrera PJ, Jo E, Sanna MG, et al. Full pharmaco-
coupled receptors for lysophospholipids: their signaling logical efficacy of a novel S1P1 agonist that does not require
properties and biological activities. J Biochem. 2002;131: S1P-like headgroup interactions. Mol Pharmacol. 2008;74:
767–771. 1308–1318.
12. Hla T, Lee MJ, Ancellin N, Paik JH, Kluk MJ. Lysophospholi- 31. Mullershausen F, Zecri F, Cetin C, Billich A, Guerini D, Seuwen
pids–receptor revelations. Science. 2001;294:1875–1878. K. Persistent signaling induced by FTY720-phosphate is
13. Liu X, Yue S, Li C, Yang L, You H, Li L. Essential roles of mediated by internalized S1P1 receptors. Nature Chemical
sphingosine 1-phosphate receptor types 1 and 3 in human Biology. 2009;5:428–434.
hepatic stellate cells motility and activation. J Cell Physiol. 32. Kau HC, Wu SB, Tsai CC, Liu CJ, Wei YH. Cigarette smoke
2011;226:2370–2377. extract-induced oxidative stress and fibrosis-related genes
14. Li C, Jiang X, Yang L, Liu X, Yue S, Li L. Involvement of expression in orbital fibroblasts from patients with Graves’
sphingosine 1-phosphate (SIP)/S1P3 signaling in cholestasis- ophthalmopathy. Oxid Med Cell Longev. 2016;2016:4676289.
induced liver fibrosis. Am J Pathol. 2009;175:1464–1472. 33. Planck T, Shahida B, Parikh H, et al. Smoking induces
15. Shiohira S, Yoshida T, Sugiura H, Nishida M, Nitta K, Tsuchiya overexpression of immediate early genes in active Graves’
K. Sphingosine-1-phosphate acts as a key molecule in the ophthalmopathy. Thyroid. 2014;24:1524–1532.
direct mediation of renal fibrosis. Physiol Rep. 2013;1: 34. Yoon JS, Chae MK, Jang SY, Lee SY, Lee EJ. Antifibrotic effects
e00172. of quercetin in primary orbital fibroblasts and orbital fat tissue
16. Sobel K, Menyhart K, Killer N, et al. Sphingosine 1-phosphate cultures of Graves’ orbitopathy. Invest Ophthalmol Vis Sci.
(S1P) receptor agonists mediate pro-fibrotic responses in 2012;53:5921–5929.
normal human lung fibroblasts via S1P2 and S1P3 receptors 35. Yoon JS, Lee HJ, Chae MK, Lee SY, Lee EJ. Cigarette smoke
and Smad-independent signaling. J Biol Chem. 2013;288: extract-induced adipogenesis in Graves’ orbital fibroblasts is
14839–14851. inhibited by quercetin via reduction in oxidative stress. J
17. Gellings Lowe N, Swaney JS, Moreno KM, Sabbadini RA. Endocrinol. 2013;216:145–156.
Sphingosine-1-phosphate and sphingosine kinase are critical
36. Van Brocklyn JR, Williams JB. The control of the balance
for transforming growth factor-beta-stimulated collagen pro-
between ceramide and sphingosine-1-phosphate by sphingo-
duction by cardiac fibroblasts. Cardiovasc Res. 2009;82:303–
sine kinase: oxidative stress and the seesaw of cell survival
312.
and death. Comp Biochem Physiol B Biochem Mol Biol.
18. Swaney JS, Moreno KM, Gentile AM, Sabbadini RA, Stoller GL. 2012;163:26–36.
Sphingosine-1-phosphate (S1P) is a novel fibrotic mediator in
the eye. Exp Eye Res. 2008;87:367–375. 37. Barnawi J, Tran HB, Roscioli E, et al. Pro-phagocytic effects of
thymoquinone on cigarette smoke-exposed macrophages
19. Caballero S, Swaney J, Moreno K, et al. Anti-sphingosine-1- occur by modulation of the sphingosine-1-phosphate signal-
phosphate monoclonal antibodies inhibit angiogenesis and ling system. COPD. 2016;13:653–661.
sub-retinal fibrosis in a murine model of laser-induced
choroidal neovascularization. Exp Eye Res. 2009;88:367–377. 38. Barnawi J, Tran H, Jersmann H, et al. Potential link between
the sphingosine-1-phosphate (S1P) system and defective
20. Kim SE, Lee JH, Chae MK, Lee EJ, Yoon JS. The role of
alveolar macrophage phagocytic function in chronic obstruc-
sphingosine-1-phosphate in adipogenesis of Graves’ orbitop-
tive pulmonary disease (COPD). PLoS One. 2015;10:
athy. Invest Ophthalmol Vis Sci. 2016;57:301–311.
e0122771.
21. Ryu JM, Baek YB, Shin MS, et al. Sphingosine-1-phosphate-
39. Visse R, Nagase H. Matrix metalloproteinases and tissue
induced Flk-1 transactivation stimulates mouse embryonic
inhibitors of metalloproteinases: structure, function, and
stem cell proliferation through S1P1/S1P3-dependent beta-
arrestin/c-Src pathways. Stem Cell Res. 2014;12:69–85. biochemistry. Circ Res. 2003;92:827–839.
22. Kong Y, Wang H, Lin T, Wang S. Sphingosine-1-phosphate/S1P 40. Giannandrea M, Parks WC. Diverse functions of matrix
receptors signaling modulates cell migration in human bone metalloproteinases during fibrosis. Dis Model Mech. 2014;7:
marrow-derived mesenchymal stem cells. Mediators In- 193–203.
flamm. 2014;2014:565369. 41. Ramachandran P, Iredale JP. Liver fibrosis: a bidirectional
23. Beach JA, Aspuria PJ, Cheon DJ, et al. Sphingosine kinase 1 is model of fibrogenesis and resolution. QJM. 2012;105:813–
required for TGF-beta mediated fibroblastto- myofibroblast 817.
differentiation in ovarian cancer. Oncotarget. 2016;7:4167– 42. Issa R, Zhou X, Constandinou CM, et al. Spontaneous
4182. recovery from micronodular cirrhosis: evidence for incom-
24. Payne SG, Oskeritzian CA, Griffiths R, et al. The immunosup- plete resolution associated with matrix cross-linking. Gastro-
pressant drug FTY720 inhibits cytosolic phospholipase A2 enterology. 2004;126:1795–1808.
independently of sphingosine-1-phosphate receptors. Blood. 43. Iredale JP, Benyon RC, Pickering J, et al. Mechanisms of
2007;109:1077–1085. spontaneous resolution of rat liver fibrosis. Hepatic stellate
25. Bartalena L, Pinchera A, Marcocci C. Management of Graves’ cell apoptosis and reduced hepatic expression of metal-
ophthalmopathy: reality and perspectives. Endocr Rev. 2000; loproteinase inhibitors. J Clin Invest. 1998;102:538–549.
21:168–199. 44. Han R, Smith TJ. Induction by IL-1 beta of tissue inhibitor of
26. Leask A, Abraham DJ. TGF-beta signaling and the fibrotic metalloproteinase-1 in human orbital fibroblasts: modulation
response. FASEB J. 2004;18:816–827. of gene promoter activity by IL-4 and IFN-gamma. J Immunol.
27. Pyne NJ, Dubois G, Pyne S. Role of sphingosine 1-phosphate 2005;174:3072–3079.
and lysophosphatidic acid in fibrosis. Biochim Biophys Acta. 45. Kim H, Choi YH, Park SJ, et al. Antifibrotic effect of
2013;1831:228–238. Pirfenidone on orbital fibroblasts of patients with thyroid-
28. Cencetti F, Bernacchioni C, Nincheri P, Donati C, Bruni P. associated ophthalmopathy by decreasing TIMP-1 and colla-
Transforming growth factor-beta1 induces transdifferentiation gen levels. Invest Ophthalmol Vis Sci. 2010;51:3061–3066.
of myoblasts into myofibroblasts via up-regulation of sphin- 46. Kook KH, Choi YH, Kim YR, et al. Altered ganglioside
gosine kinase-1/S1P3 axis. Mol Biol Cell. 2010;21:1111–1124. expression modulates the pathogenic mechanism of thyroid-

Downloaded From: https://iovs.arvojournals.org/ on 07/23/2018


Sphingosine-1-Phosphate Mediates Fibrosis in GO IOVS j May 2017 j Vol. 58 j No. 5 j 2553

associated ophthalmopathy by increase in hyaluronic acid. 49. Hogg MG, Evans CH, Smith TJ. Leukoregulin induces
Invest Ophthalmol Vis Sci. 2011;52:264–273. plasminogen activator inhibitor type 1 in human orbital
fibroblasts. Am J Physiol. 1995;269:C359–C366.
47. Takamiya Y, Fukami K, Yamagishi S, et al. Experimental
diabetic nephropathy is accelerated in matrix metalloprotei- 50. Koumas L, Smith TJ, Feldon S, Blumberg N, Phipps RP. Thy-1
expression in human fibroblast subsets defines myofibroblas-
nase-2 knockout mice. Nephrol Dial Transplant. 2013;28:55–
tic or lipofibroblastic phenotypes. Am J Pathol. 2003;163:
62.
1291–1300.
48. Wang X, Zhou Y, Tan R, et al. Mice lacking the matrix 51. Hondur A, Konuk O, Dincel AS, Bilgihan A, Unal M,
metalloproteinase-9 gene reduce renal interstitial fibrosis in Hasanreisoglu B. Oxidative stress and antioxidant activity in
obstructive nephropathy. Am J Physiol Renal Physiol. 2010; orbital fibroadipose tissue in Graves’ ophthalmopathy. Curr
299:F973–F982. Eye Res. 2008;33:421–427.

Downloaded From: https://iovs.arvojournals.org/ on 07/23/2018

You might also like