ArticlePDF Available

Sphingosine-1-phosphate promotes lymphangiogenesis by stimulating S1P1/G(i)/PLC/Ca2+ signaling pathways

Authors:

Abstract and Figures

The lymphatic system plays pivotal roles in mediating tissue fluid homeostasis and immunity, and excessive lymphatic vessel formation is implicated in many pathological conditions, which include inflammation and tumor metastasis. However, the molecular mechanisms that regulate lymphatic vessel formation remain poorly characterized. Sphingosine-1-phosphate (S1P) is a potent bioactive lipid that is implicated in a variety of biologic processes such as inflammatory responses and angiogenesis. Here, we first report that S1P acts as a lymphangiogenic mediator. S1P induced migration, capillary-like tube formation, and intracellular Ca(2+) mobilization, but not proliferation, in human lymphatic endothelial cells (HLECs) in vitro. Moreover, a Matrigel plug assay demonstrated that S1P promoted the outgrowth of new lymphatic vessels in vivo. HLECs expressed S1P1 and S1P3, and both RNA interference-mediated down-regulation of S1P1 and an S1P1 antagonist significantly blocked S1P-mediated lymphangiogenesis. Furthermore, pertussis toxin, U73122, and BAPTA-AM efficiently blocked S1P-induced in vitro lymphangiogenesis and intracellular Ca(2+) mobilization of HLECs, indicating that S1P promotes lymphangiogenesis by stimulating S1P1/G(i)/phospholipase C/Ca(2+) signaling pathways. Our results suggest that S1P is the first lymphangiogenic bioactive lipid to be identified, and that S1P and its receptors might serve as new therapeutic targets against inflammatory diseases and lymphatic metastasis in tumors.
S1P-induced lymphangiogenesis was mediated through the S1P1. (A) HLECs were infected with retroviruses carrying S1P1 and S1P3 shRNA expression vectors in pRS plasmid, after which stably transfected cells were obtained by selection with puromycin. pRS plasmid was used as control. After a 4-hour incubation with 100 ng/mL S1P, migrated HLECs were stained and counted in 3 random fields ( XI ). HLECs were laid on a GFR Matrigel-coated 24-well plate and incubated with 100 ng/mL S1P for 6 hours. Two randomly chosen fields per well were photographed and the total tube area was analyzed using Scion Image ( f ). (B,C) Effect of S1P1 antagonist or S1P1 control molecule on the S1P-induced migration (B) and capillary-like tube formation (C) of HLECs was examined as described in panel A. (D,E) Ten days after subcutaneous injection of Matrigel in C57BL/6 mice (5 mice per group), the Matrigel was removed, fixed, embedded in paraffin, sectioned at 4 ␮ m, and immunostained using antibodies specific for Prox-1 (green) and podoplanin (red). (D) Representative photographs of untreated control mice, and mice treated with S1P (0.4 ␮ g) in the absence or presence of S1P1 antagonist (100 ␮ M) or S1P1 control molecule (100 ␮ M). Scale bars represent 50 ␮ m. (E) The number of Prox-1 ϩ /podoplanin ϩ lymphatic vessels per field was counted. All values are expressed as means plus or minus SD (A-C) and means plus or minus SEM (E). In panels A-C, data are representative of 3 independent experiments with similar results. ** indicates a statistically significant difference ( P Ͻ .01).
… 
Content may be subject to copyright.
doi:10.1182/blood-2007-11-125203
Prepublished online June 9, 2008;
2008 112: 1129-1138
Chi-Bom Chae and Yong Song Gho
Chang Min Yoon, Bok Sil Hong, Hyung Geun Moon, Seyoung Lim, Pann-Ghill Suh, Yoon-Keun Kim,
S1P1/Gi/PLC/Ca2+ signaling pathways
Sphingosine-1-phosphate promotes lymphangiogenesis by stimulating
http://bloodjournal.hematologylibrary.org/content/112/4/1129.full.html
Updated information and services can be found at:
(2497 articles)Hemostasis, Thrombosis, and Vascular Biology Articles on similar topics can be found in the following Blood collections
http://bloodjournal.hematologylibrary.org/site/misc/rights.xhtml#repub_requests
Information about reproducing this article in parts or in its entirety may be found online at:
http://bloodjournal.hematologylibrary.org/site/misc/rights.xhtml#reprints
Information about ordering reprints may be found online at:
http://bloodjournal.hematologylibrary.org/site/subscriptions/index.xhtml
Information about subscriptions and ASH membership may be found online at:
Copyright 2011 by The American Society of Hematology; all rights reserved.
Washington DC 20036.
by the American Society of Hematology, 2021 L St, NW, Suite 900,
Blood (print ISSN 0006-4971, online ISSN 1528-0020), is published weekly
For personal use only. by guest on June 10, 2013. bloodjournal.hematologylibrary.orgFrom
HEMOSTASIS, THROMBOSIS,AND VASCULAR BIOLOGY
Sphingosine-1-phosphate promotes lymphangiogenesis by stimulating
S1P1/Gi/PLC/Ca2signaling pathways
Chang Min Yoon,1Bok Sil Hong,1Hyung Geun Moon,1Seyoung Lim,1Pann-Ghill Suh,1Yoon-Keun Kim,1Chi-Bom Chae,2
and Yong Song Gho1
1Division of Molecular and Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea; and 2Institute of Biomedical Science and
Technology, Konkuk University, Seoul, Republic of Korea
The lymphatic system plays pivotal roles in
mediating tissue fluid homeostasis and im-
munity, and excessive lymphatic vessel for-
mation is implicated in many pathological
conditions, which include inflammation and
tumor metastasis. However, the molecular
mechanisms that regulate lymphatic vessel
formation remain poorly characterized.
Sphingosine-1-phosphate (S1P) is a potent
bioactive lipid that is implicated in a variety
of biologic processes such as inflammatory
responses and angiogenesis. Here, we first
report that S1P acts as a lymphangiogenic
mediator. S1P induced migration, capillary-
like tube formation, and intracellular Ca2
mobilization, but not proliferation, in human
lymphatic endothelial cells (HLECs) in vitro.
Moreover, a Matrigel plug assay demon-
strated that S1P promoted the outgrowth of
new lymphatic vessels in vivo. HLECs ex-
pressed S1P1 and S1P3, and both RNA
interference–mediated down-regulation of
S1P1 and an S1P1 antagonist significantly
blocked S1P-mediated lymphangiogenesis.
Furthermore, pertussis toxin, U73122, and
BAPTA-AM efficiently blocked S1P-induced
in vitro lymphangiogenesis and intracellular
Ca2mobilization of HLECs, indicating that
S1P promotes lymphangiogenesis by stimu-
lating S1P1/Gi/phospholipase C/Ca2signal-
ing pathways. Our results suggest that S1P
is the first lymphangiogenic bioactive lipid
to be identified, and that S1P and its recep-
tors might serve as new therapeutic targets
against inflammatory diseases and lym-
phatic metastasis in tumors. (Blood. 2008;
112:1129-1138)
Introduction
Sphingosine-1-phosphate (S1P) has been implicated in a wide
spectrum of biologic processes, including the promotion of cell
growth and survival, migration and differentiation, platelet aggrega-
tion, inflammatory responses, and angiogenesis.1S1P is generated
by the phosphorylation of sphingosine through a process mediated
by sphingosine kinase 1 (SphK1) and SphK2. S1P acts both
intracellularly as a second messenger2and extracellularly as a
ligand for a family of G-protein–coupled S1P receptors.3S1P1
couples stringently to the Giprotein family, whereas S1P2 and
S1P3 couple to the Gi,G
q, and G12/13 protein families. Multiple
interconnections of S1P signaling through S1P1 and S1P3 induce
vascular endothelial cell proliferation, migration, morphogenesis,
cytoskeletal reorganization, and adherens junction assembly,
whereas signaling via S1P2 negatively regulates S1P-mediated
multiple responses of vascular endothelial cells.4The defective
vascular maturation observed in S1P1-deficient mice highlights a
fundamental role for S1P signaling on vasculogenesis.5Neutraliza-
tion of the action of extracellular S1P shows significant inhibition
of angiogenesis, tumor growth, metastasis, and lymphocyte trans-
migration, indicating that S1P is an important pathological regula-
tor of inflammation and angiogenesis.6-8
Lymphatic vessels play important roles in mediating tissue fluid
homeostasis and immunity.9Although lymphangiogenesis, the
formation of new lymphatic vessels from preexisting vessels,
occurs under physiological and pathological conditions, including
chronic inflammation10 and tumor metastasis,11 the molecular
mechanisms that regulate lymphatic vessel formation remain
largely uncharacterized. Vascular endothelial growth factor C
(VEGF-C), the first identified lymphangiogenic factor, is a se-
creted, proteolytically processed glycoprotein that activates VEGF
receptors 2 and 3 on lymphatic endothelial cells.12,13 VEGF-C is
overexpressed in many primary tumors and induces lymphangiogen-
esis and angiogenesis around tumor tissues, as well as promotes
tumor metastasis via newly formed lymphatic or blood vessels.14,15
In addition, VEGF-C is up-regulated in inflammatory cells and so
induces lymphangiogenesis and angiogenesis during chronic
inflammation.10
VEGF-A, hepatocyte growth factor (HGF), angiopoietin-1,
platelet-derived growth factor BB (PDGF-BB), and insulin-like
growth factor 1/2 (IGF1/2), all of which are proangiogenic factors,
can also give rise to in vivo lymphangiogenesis.16-19 However,
relatively less interest has been focused on the involvement of
bioactive lipid molecules compared with that of growth factors in
lymphangiogenesis. Because S1P is a potent proangiogenic and
inflammatory bioactive lipid molecule,20,21 we hypothesized that
S1P could affect lymphangiogenesis. In this report, we show that
S1P induced the migration and capillary-like tube formation of
lymphatic endothelial cells in vitro and lymphangiogenesis in vivo.
Real-time polymerase chain reaction (PCR) analysis revealed that
human lymphatic endothelial cells (HLECs) expressed S1P1 and
S1P3. S1P-induced lymphangiogenesis was significantly inhibited
by pertussis toxin (PTX), RNA interference–mediated down-
regulation of S1P1, and an S1P1 antagonist, indicating the involve-
ment of Giprotein activation coupled with S1P1. In addition, the
inhibition of S1P-induced in vitro lymphangiogenesis by U73122
and BAPTA-AM demonstrated that phospholipase C (PLC)/Ca2
Submitted November 20, 2007; accepted May 27, 2008. Prepublished online as
Blood First Edition paper, June 9, 2008; DOI 10.1182/blood-2007-11-125203.
The publication costs of this article were defrayed in part by page charge
payment. Therefore, and solely to indicate this fact, this article is hereby
marked ‘‘advertisement’’ in accordance with 18 USC section 1734.
© 2008 by The American Society of Hematology
1129BLOOD, 15 AUGUST 2008 VOLUME 112, NUMBER 4
For personal use only. by guest on June 10, 2013. bloodjournal.hematologylibrary.orgFrom
was an important signaling pathway. Moreover, we found that S1P
induced intracellular Ca2mobilization and formation of actin
stress fibers in a PTX/PLC-dependent manner in HLECs. Our
findings strongly suggest that S1P is the first lymphangiogenic
bioactive lipid and that S1P secreted from inflammatory cells or
tumor cells may induce lymphangiogenesis during tumor growth,
metastasis, and inflammation.
Methods
Materials
S1P, heparin, FITC-dextran (2000 kDa) and sulfinipyrazone were pur-
chased from Sigma-Aldrich (St Louis, MO); S1P1 selective antagonist
(R-isomer) and S1P1 control molecule (S-isomer) were acquired from
Avanti Polar Lipids (Alabaster, AL); L-NAME, LY294002, and BAPTA-AM
were obtained from BioMol Research Laboratories (Plymouth Meeting,
PA); PTX was purchased from Calbiochem (La Jolla, CA); PD98059,
SB203580, and U73122 were acquired from Alexis Biochemicals (San
Diego, CA); recombinant human VEGF-C and basic fibroblast growth
factor (bFGF) were obtained from R&D Systems (Minneapolis, MN);
human fibronectin, growth factor–reduced (GFR)–Matrigel, and Matrigel
were purchased from BD Biosciences (San Jose, CA); medium 199, fetal
bovine serum (FBS), penicillin/streptomycin, human endothelial serum–
free medium (HE-SFM), fura-2/AM, lipofectamine 2000, AlexaFluor594-
streptavidin, AlexaFluor488 donkey anti–mouse and anti–rabbit, and Alex-
aFluor594 donkey anti–rat IgG antibodies were obtained from Invitrogen
(Carlsbad, CA). Diff-Quick solution was purchased from Baxter Healthcare
(McGraw Park, IL); One Step SYBR reverse-transcription (RT)–PCR kit
was acquired from TaKaRa Bio (Tokyo, Japan); hamster antibody 8.1.1 was
purchased from Developmental Studies Hybridoma Bank (Iowa City, IA);
biotin-labeled anti–hamster IgG antibody, normal mouse, rat, rabbit IgGs
were obtained from Santa Cruz Biotechnology (Santa Cruz, CA); rabbit
anti–Prox-1 antibody was acquired from RELIATech (Braunschweig,
Germany); rat anti–lymphatic vascular endothelial hyaluronan receptor-1
(LYVE-1) antibody was kindly gifted by Dr Gou Young Koh (Korea
Advanced Institute of Science and Technology, Daejeon, Republic of
Korea); mouse anti–human podoplanin antibody (D2-40) was obtained
from Signet Laboratories (Dedham, MA); rabbit anti–phospho-histone-H3
(PH3) antibody was obtained from Upstate Biotechnology (Lake Placid,
NY); shRNAs in pRS plasmid against S1P1 and S1P3 were acquired from
OriGene (Rockville, MD); HLECs and human dermal lymphatic microvas-
cular endothelial cells (HMVECs-dLy) were purchased from ScienCell
(San Diego, CA) and Lonza (Walkersville, MD), respectively.
Cell culture
Human umbilical vein endothelial cells (HUVECs) were isolated from
freshly delivered umbilical cords as described previously22 and main-
tained in medium 199 supplemented with 20% FBS, 3 ng/mL bFGF,
5 U/mL heparin, 100 U/mL penicillin, and 0.1 mg/mL streptomycin.
HLECs were maintained in endothelial cell medium supplemented with
5% FBS, endothelial cell growth supplement (ScienCell), 100 U/mL
penicillin, and 0.1 mg/mL streptomycin. HMVECs-dLy were cultured in
EGM-2 MV medium (Lonza). Cells were maintained at 37°C in a
humidified 5% CO2atmosphere. Cells from 4 to 5 passages were used
for the experiments in this study.
Migration assay
A migration assay was performed in a 48-well microchemotaxis chamber
(Neuro Probe, Cabin John, MD) as described previously.23 Polycarbonate
membranes with 12-m pores (Neuro Probe) were coated with 1 g/mL
human fibronectin in double-distilled water and then dried for 1 hour. Cells
were harvested and resuspended in HE-SFM containing medium only or
with 100 ng/mL PTX, 10 M LY294002, 1 mM L-NAME, 10 M PD98059,
25 M SB203580, 30 M BAPTA-AM, 5 M U73122, S1P1 antagonist,
or S1P1 control molecule. The bottom chamber was loaded with
3104cells, and the filter membrane was laid over the cells. The
microchamber was then inverted and incubated at 37°C for 2 hours. After
reinverting the chamber to its upright position, the upper wells were then
loaded with HE-SFM containing the indicated concentration of S1P or
100 ng/mL S1P with 100 ng/mL PTX, 10 M LY294002, 1 mM L-NAME,
10 M PD98059, 25 M SB203580, 30 M BAPTA-AM, 5 M U73122,
S1P1 antagonist, or S1P1 control molecule. The chamber was then
reincubated at 37°C for 4 hours, and the filter membrane was fixed and
stained using Diff-Quick solution. The cells that migrated through the filter
membrane were quantified by counting 3 random fields of each well using a
Nikon Eclipse TS100 microscope (Nikon, Tokyo, Japan) equipped with a
Plan Fluor 20/0.50 DIC M/N2 objective lens and COOLPIX 995 digital
camera (Nikon). Experiments were carried out in triplicate and repeated
independently 3 times.
Proliferation assay
HLECs and HMVECs-dLy were plated at 2.5 104cells/well on a
fibronectin-coated 48-well plate. After starvation in HE-SFM for 6 hours,
the cells were incubated with S1P or VEGF-C for 24 hours. The cells were
treated with 0.5 Ci (0.0185 MBq) [3H]-thymidine per well and further
incubated for 12 hours. The radioactivity of incorporated [3H]-thymidine
was determined in a liquid scintillation counter. Experiments were carried
out in triplicate and repeated 3 times.
Capillary-like tube formation assay
The tube formation assay was performed as described previously.23 Cells
(2 104cells/well) in 0.4 mL HE-SFM with the indicated concentration of
S1P, 500 ng/mL VEGF-C, or 100 ng/mL S1P with 100 ng/mL PTX, 10 M
LY294002, 1 mM L-NAME, 10 M PD98059, 25 M SB203580, 30 M
BAPTA-AM, 5 M U73122, S1P1 antagonist, or S1P1 control molecule
were plated on a GFR Matrigel-coated 24-well plate. After a 6-hour
incubation, the cells were fixed with Diff-Quick solution, and 2 randomly
chosen fields per well were visualized and acquired using a Nikon Eclipse
TS100 microscope equipped with a 10/0.25 Ph1 ADL objective lens and
COOLPIX 995 digital camera, and processed using Adobe Photoshop 7.0
(Adobe Systems, San Jose, CA). Total tube area was analyzed using Scion
Image software (Frederick, MD). Analyses of the test samples were
performed in duplicate and independent experiments were repeated 3 times.
Immunocytochemistry
HLECs and HMVECs-dLy were fixed with 4% paraformaldehyde and
incubated with anti–Prox-1, anti–LYVE-1, or anti-podoplanin antibody. For
immunostaining of Prox-1, fixed cells were permeabilized with 0.2% Triton
X-100. Isotype-matched control IgGs were used for negative staining. After
treatment of AlexaFluor-conjugated secondary antibodies, cells were coun-
terstained with Hoechst. Five randomly chosen fields were visualized using
AxioSkop2 Plus Fluorescent microscope (Carl Zeiss, Jena, Germany)
equipped with an Achroplan 20/0.45 objective lens and images were
acquired using an AxioVision 4.0 software (Carl Zeiss) and processed using
Adobe Photoshop 7.0.
Real-time RT-PCR
PCR primers for human S1P1, S1P2, S1P3, and -actin were designed
using the Primer3 program (Whitehead Institute, http://biotools.umass-
med.edu/bioapps/primer3_www.cgi). The primers used are shown in Table
1. After phosphate-buffered saline washing, subconfluent HLECs were
trypsinized and centrifuged. Cell pellets were used for isolation of total
RNA via RNease Mini Kit (QIAGEN, Valencia, CA). For real-time
RT-PCR, total RNA (100 ng) was amplified with a One Step SYBR RT-PCR
kit using a LightCycler 2.0 PCR system (Roche Diagnostics, Mannheim,
Germany). This experiment was repeated twice.
Matrigel plug assay, immunostaining, and lymphangiography
C57BL/6 (male, 5 weeks old, 5 mice per group) mice were subcutaneously
injected with 0.5 mL Matrigel containing 0.4 g S1P or 1 g VEGF-C. The
1130 YOON et al BLOOD, 15 AUGUST 2008 VOLUME 112, NUMBER 4
For personal use only. by guest on June 10, 2013. bloodjournal.hematologylibrary.orgFrom
mice were killed at 10 days after implantation, and the Matrigel was
removed, fixed in 4% paraformaldehyde, and embedded in paraffin.
Cross-sections of paraffin-embedded Matrigel were stained with hematoxy-
lin and eosin (H&E) and images were visualized using an Olympus BX51
microscope (Olympus, Tokyo, Japan) equipped with an UPlanFL N
100/1.30 oil objective lens and acquired using analysis LS Research
software (Soft Imaging System, Munster, Germany). Lymphatic vessels
were immunostained with anti–Prox-1, anti–LYVE-1, or anti-podoplanin
antibody. Proliferating nuclei in lymphatic vessels were immunostained
with rabbit anti-PH3 antibody. For lymphangiography, at 10 days after
implantation, FITC-dextran (2000 kDa, 10 mg/mL) was injected intrader-
mally apart from Matrigel, and lymphatic vessel networks were analyzed.
All images were visualized using an FV1000 Olympus Confocal micro-
scope (Olympus) equipped with an UPlanSApo 20/0.75 objective lens
and acquired using FV1000-ASW 1.5 software (Olympus). This experiment
was independently repeated twice. Animal study protocols were approved
by the Institutional Animal Care and Use Committee at Pohang University
of Science and Technology.
RNA interference–mediated down-regulation of S1P1 and S1P3
The shRNA retroviral pRS plasmids for S1P1 and S1P3 were transfected
into mouse packaging PT67 cells (Clontech, Palo Alto, CA) using
lipofectamine 2000 and cultured with puromycin (Clontech). For negative
control, empty pRS plasmid was used. After infection with retrovirus,
HLECs were cultured with puromycin. The shRNA sequences against S1P1
and S1P3 are as follows: S1P1, 5-GTACTTCCTGGTGTTAGCTGTGCT-
CAACT-3; S1P3, 5-TCACCACCGTGCTCTTCTTGGTCATCTGC-3.
Measurement of intracellular free calcium mobilization
Intracellular Ca2mobilization in HLECs was determined with the
fluorescent Ca2indicator fura-2/AM as described previously.24 Briefly,
HLECs were incubated with fura-2/AM at a final concentration of 3 Min
HE-SFM at 37°C for 30 minutes. After loading, the cells were washed twice
with Ca2-free Locke solution (158.4 mM NaCl, 5.6 mM KCl, 1.2 mM
MgCl2, 0.2 mM EGTA, 5 mM HEPES, and 10 mM glucose, pH 7.3) to
remove extracellular dye. Sulfinipyrazone was added to both the loading
medium and the washing solution to a final concentration of 250 Mto
prevent dye leakage. Approximately 1.5 105cells of the cell suspension
were transferred to a quartz cuvette and placed in a thermostatically
controlled cell holder at 37°C, in which the cell suspension was continu-
ously stirred, and exposed to 100 ng/mL S1P with 100 ng/mL PTX, 10 M
LY294002, 1 mM L-NAME, 10 M PD98059, 25 M SB203580, 30 M
BAPTA-AM, or 5 M U73122. LY294002, L-NAME, PD98059, SB203580,
BAPTA-AM, and U73122 were preincubated with the cells for 30 minutes
prior to exposure to S1P, and PTX was preincubated for 6 hours.
Fluorescence ratios were taken by dual excitation at 340 and 380 nm, and
emission at 500 nm by the alternative wavelength time scanning method.
Experiments were repeated 3 times.
Statistical analyses
Student ttest was used to calculate Pvalues based on comparisons with the
appropriate control samples tested at the same time.
Results
S1P induced in vitro lymphangiogenesis
Lymphangiogenesis is a complex cellular process that occurs via
proliferation, migration, and differentiation of lymphatic endothe-
lial cells. To investigate whether S1P had in vitro lymphangiogenic
activity, we performed migration, proliferation, and capillary-like
tube formation assays in HLECs, lymphatic endothelial cells
derived from lymph nodes. S1P significantly induced the migration
of HLECs in a dose-dependent manner over the migration in the
presence of medium alone (Figure 1A), whereas the presence of
S1P at concentrations up to 200 ng/mL did not show any effect on
the proliferation of HLECs (Figure 1B). VEGF-C, a potent
lymphangiogenic factor, also induced the migration and prolifera-
tion of HLECs (Figure 1A,B). Although S1P had no mitogenic
activity on HLECs, the migratory activity of S1P (100 ng/mL) was
much higher than that of VEGF-C (500 ng/mL). We next examined
the ability of S1P to promote the capillary-like tube formation of
HLECs on GFR Matrigel. In HLECs, the presence of S1P at
20 ng/mL, 100 ng/mL, and 200 ng/mL caused 1.7 plus or minus
0.3-fold, 2.1 plus or minus 0.2-fold, and 2.2 plus or minus 0.1-fold
increases in tube area, respectively, compared with the negative
control containing only the medium (Figure 1C). VEGF-C
(500 ng/mL) also stimulated tube formation (2.4 0.3-fold
increase).
To further confirm that S1P has in vitro lymphangiogenic
activity, we examined the ability of S1P to promote the migration
and capillary-like tube formation of HMVECs-dLy, lymphatic
endothelial cells derived from dermal skins. S1P significantly
promoted the migration and tube formation of HMVECs-dLy in a
dose-dependent manner (Figure 1D,E), whereas the presence of
S1P at concentrations up to 200 ng/mL did not show any effect on
the proliferation of HMVECs-dLy (data not shown). We next
characterized HLECs and HMVECs-dLy with lymphatic endothe-
lial cell–specific marker proteins by immunocytochemistry. More
than 95% of HLECs and HMVECs-dLy expressed Prox-1, LYVE-1,
and podoplanin (Figure 1F). These results suggest that S1P exerts
in vitro lymphangiogenic activity by promoting migration and
differentiation, but not proliferation, of human lymphatic endothe-
lial cells.
S1P promoted in vivo lymphangiogenesis
Since S1P stimulated migration and differentiation of HLECs and
HMVECs-dLy in vitro, we performed a Matrigel plug assay to
investigate whether S1P had in vivo lymphangiogenic activity.
Matrigels containing S1P (0.4 g) or VEGF-C (1 g) were subcu-
taneously injected into C57BL/6 mice; after 10 days, the mice were
killed and Matrigels were extracted. H&E staining and immunostain-
ing of podoplanin revealed that both S1P and VEGF-C induced
pronounced lymphatic vessel formation (Figure 2A-C), whereas
lower numbers of lymphatic vessels were observed in the control.
The podoplanin-positive lymphatic vessels were also positive for
Prox-1, another lymphatic endothelial cell–specific marker, and
sprouting lymphatic vessels were observed in S1P- and VEGF-C–
treated Matrigel (Figure 2D). In the VEGF-C– and S1P-treated
mice, respectively, 70.6% plus or minus 12.5% and 53.8% plus or
Table 1. Primer sets of human S1P receptors and -actin
Gene Sequence Product size, bp
S1P1
Forward 5-TGCGGGAAGGGAGTATGTTT-3
60Reverse 5-CGATGGCGAGGAGACTGAAC-3
S1P2
Forward 5-GCCTCTCTACGCCAAGCATTA-3
107Reverse 5-TTGAGCGGACCACGCAGTA-3
S1P3
Forward 5-TGATTGTGGTGAGCGTGTTCA-3
68Reverse 5-GGCCACATCAATGAGGAAGAG-3
-actin
Forward 5-TCTACAATGAGCTGCGTGTG-3
127Reverse 5-ATGGCTGGGGTGTTGAAG-3
LYMPHANGIOGENICACTIVITY OF S1P 1131BLOOD, 15 AUGUST 2008 VOLUME 112, NUMBER 4
For personal use only. by guest on June 10, 2013. bloodjournal.hematologylibrary.orgFrom
minus 6.8% of LYVE-1–positive lymphatic vessels were stained by
PH3, a marker of cell proliferation (Figure 2E,F). Furthermore,
FITC-dextran microlymphography revealed that both S1P and
VEGF-C were able to induce the growth of functional lymphatic
vessels (Figure 2G). These observations suggest that S1P is a
potent lymphangiogenic lipid molecule in vivo and that the
lymphangiogenic activity of S1P is comparable to that of VEGF-C,
a potent lymphangiogenic growth factor.
S1P stimulated lymphangiogenesis via the S1P1/Gipathway
S1P1, S1P2, and S1P3 are widely expressed subtypes in vascular
endothelial cells,25-27 and S1P stimulates proliferation and migra-
tion of vascular endothelial cells via S1P1 and S1P3.20,28 To
investigate whether S1P-induced lymphangiogenesis was mediated
by the activation of S1P receptors, we performed real-time PCR
analysis to identify the expression pattern of S1P receptors in
HLECs. Real-time PCR analysis revealed that both HLECs and
HUVECs expressed S1P1 and S1P3, but not S1P2 (Figure 3A).
Although it has been reported that VEGF induces S1P1 receptors in
endothelial cells,29 real-time PCR analysis revealed that VEGF-C
did not affect the expression of S1P1 and S1P3 in HLECs (data not
shown). Because 2 distinctive S1P receptors were expressed on
HLECs, we next carried out migration and tube formation assays to
determine which receptor was mainly involved in S1P-induced
lymphangiogenesis. S1P-induced migration and tube formation of
HLECs were almost completely blocked by treatment with 100 ng/
mL PTX, a Giprotein–specific inhibitor, whereas treatment with
PTX in the control did not affect HLECs (Figure 3B,C). Both S1P1
Figure 1. S1P induced migration and the formation
of capillary-like tube structure of human lymphatic
endothelial cells. (A) After 4 hours of incubation with
S1P or VEGF-C, migrated HLECs were stained and
counted in 3 random fields. (B) S1P or VEGF-C was
added to serum-starved HLECs for 24 hours, followed
by additional incubation for 12 hours with 0.5 Ci
(0.0185 MBq) [3H]-thymidine in HE-SFM. Results are
expressed as the percentage [3H]-thymidine incorpora-
tion of the control versus S1P- or VEGF-C–treated
HLECs. (C) HLECs were laid on a 24-well, GFR
Matrigel-coated plate and incubated with S1P or
VEGF-C for 6 hours. Two randomly chosen fields per
well were photographed and the total tube area was
analyzed using Scion Image. (D,E) Effect of S1P on the
migration (D) and capillary-like tube formation (E) of
HMVECs-dLy, lymphatic endothelial cells derived from
dermal skins, was examined as described in panels A
and C, respectively. (F) Cultured HLECs and HMVECs-
dLy were fixed, and immunostained using antibodies
against Prox-1 (green), LYVE-1 (red), and podoplanin
(green). The nuclei were counterstained by Hoechst
(blue). Scale bars represent 50 m. Note that HLECs
and HMVECs-dLy were not immunolabeled by isotype-
matched control IgG antibodies (data not shown). All
values are expressed as means (SD). Data are
representative of 3 independent experiments with simi-
lar results. * and ** indicate statistically significant
differences (P.05 and P.01, respectively).
1132 YOON et al BLOOD, 15 AUGUST 2008 VOLUME 112, NUMBER 4
For personal use only. by guest on June 10, 2013. bloodjournal.hematologylibrary.orgFrom
and S1P3 convert external signals into intracellular signals via
heterotrimeric G proteins coupled to them; S1P1 is coupled with
the Giprotein, but S1P3 is coupled with the Gqand G12/13 proteins
as well as the Giprotein.4Therefore, our results suggest that the
activation of Giproteins coupled to S1P1 may be involved in
S1P-induced migration and differentiation of HLECs.
We next examined the effect of specific knockdown of S1P1 and
S1P3 by RNA interference on S1P-induced migration and differen-
tiation of HLECs. Down-regulation of S1P1 significantly inhibited
S1P-induced migration and tube formation of HLECs, whereas
down-regulation of S1P3 did not (Figure 4A). To further confirm
that S1P promotes lymphangiogenesis by stimulating S1P1, we
investigated the effect of an S1P1 antagonist30 on S1P-induced in
vitro and in vivo lymphangiogenesis. The S1P1 antagonist blocked
S1P-induced migration and tube formation of HLECs in a dose-
dependent manner, whereas an inactive S1P1 control molecule did
not (Figure 4B,C). Furthermore, Matrigel plug assays showed that
the S1P1 antagonist almost completely blocked S1P-induced
lymphatic vessel formation, whereas the inactive S1P1 control
molecule did not (Figure 4D,E). These results strongly suggest that
the activation of Giproteins coupled to S1P1 may be involved in
S1P-induced migration and differentiation of HLECs in vitro and
lymphangiogenesis in vivo.
S1P-induced lymphangiogenesis was mediated via the
PLC/Ca2pathway
After activation of the Giprotein complex by an upstream receptor
such as S1P1, the Giprotein complex is dissociated into Giand
Gi␤␥ subunits to activate downstream signaling molecules. To
investigate downstream signaling events involved in the S1P-
induced lymphangiogenesis of HLECs, we performed migration
and tube formation assays with LY294002, L-NAME, PD98059,
SB203580, BAPTA-AM, and U73122, which are specific inhibi-
tors for phosphatidylinositol 3-kinase (PI3K), nitric oxide synthase
(NOS), p44/42 mitogen-activated protein kinase (MAPK), p38
MAPK, intracellular Ca2chelator, and PLC, respectively. Approxi-
mately 78.3% and 85.2% of S1P-induced migration of HLECs was
efficiently blocked when 100 ng/mL S1P was applied to HLECs
with 30 M BAPTA-AM and 5 M U73122, respectively (Figure
Figure 2. S1P promoted in vivo lymphangiogen-
esis. Ten days after subcutaneous injection of Matrigel
containing none, VEGF-C (1 g), or S1P (0.4 g), in
C57BL/6 mice (5 mice per group), the Matrigel was
removed, fixed, embedded in paraffin, sectioned at
4m, and immunostained. (A) Cross-sections of Matri-
gel were stained by H&E. Scale bars represent 50 m.
(B,C) Lymphatic vessels in Matrigel were immuno-
stained for podoplanin (red). Representative photo-
graphs and the number of podoplanin-positive lym-
phatic vessels per field are shown in panels B and C,
respectively. Scale bars represent 200 m. (D) Lym-
phatic vessels in Matrigel were immunostained for
Prox-1 (green) and podoplanin (red). Arrows show
sprouting lymphatic vessels. Scale bars represent
50 m. (E,F) Proliferating lymphatic endothelial cells in
Matrigel were double immunostained for PH3 (green)
and LYVE-1 (red). Representative photographs are
shown in panel E. Scale bars represent 20 m. The
number of LYVE-1() or LYVE-1/PH3(f) vessels
per field were counted (F). (G) Uptake of injected
FITC-dextran (2000 kDa) into newly formed lymphatic
vessels was visualized using confocal microscope.
Scale bars represent 100 m. All values are expressed
as means (SEM). ** indicates statistically significant
difference (P.01).
LYMPHANGIOGENICACTIVITY OF S1P 1133BLOOD, 15 AUGUST 2008 VOLUME 112, NUMBER 4
For personal use only. by guest on June 10, 2013. bloodjournal.hematologylibrary.orgFrom
5A). Furthermore, S1P-induced capillary-like tube formation of
HLECs was completely inhibited by treatment with 30 M
BAPTA-AM and 5 M U73122 (Figure 5B). However, treatment
with the inhibitors for PI3K, NOS, p44/42 MAPK, and p38 MAPK
did not show any inhibitory effect on S1P-induced migration and
tube formation of HLECs. All these results suggest that PLC and
intracellular Ca2mobilization, but not NOS, PI3K, p44/42
MAPK, and p38 MAPK, are major signaling pathways involved in
S1P-induced lymphangiogenesis.
Increased mobilization of intracellular Ca2by S1P was
mediated through the Gi/PLC/Ca2pathway
The activation of PLC by extracellular S1P evokes the induction of
robust Ca2mobilization in HUVECs, which is important to the
S1P-induced migration of vascular endothelial cells.31,32 Since we
demonstrated the involvement of S1P-mediated PLC activation and
intracellular Ca2release in the migration and capillary-like tube
formation of HLECs, we investigated whether the S1P-mediated
increase in intracellular Ca2release was dependent on the Gi/PLC
pathway in HLECs. The application of S1P to fura-2/AM-loaded
HLECs caused a dramatic increase in Ca2release into the cytosol
in a dose-dependent manner (data not shown). The increase in Ca2
influx by 100 ng/mL S1P was significantly inhibited by treatment
with 100 ng/mL PTX (78.9% 14.0%), 30 M BAPTA-AM
(80.3% 7.3%), and 5 M U73122 (81.3% 4.7%), but treat-
ment with the signaling inhibitors for PI3K, NOS, p44/42 MAPK,
and p38 MAPK did not show any inhibitory effect on Ca2
mobilization (Figure 5C). These results suggest that S1P-induced
migration and differentiation of HLECs may be mediated by
intracellular Ca2release following Gi/PLC activation.
Discussion
The lymphatic system has important roles in regulating tissue fluid
balance for homeostasis and facilitating interstitial protein trans-
port and immunologic function through lymph nodes. Clinical
evidence suggests that the dissemination of malignant tumors to
regional lymph nodes via the lymphatic vessels is important in
tumor metastasis and that chronic inflammation causes lymphangio-
genesis and lymphedema, although the molecular mechanisms
regulating lymphangiogenesis are largely unclear. In addition to
VEGF-C, a potent lymphangiogenic growth factor, VEGF-A,
bFGF, HGF, angiopoietin-1, IGF-1/2, and PDGF-BB, previously
known as proangiogenic factors, have lymphangiogenic activity.
However, no previous studies have examined the effects of
bioactive lipid molecules, including S1P, on lymphangiogenesis. In
this report, we provide evidence that S1P induces in vitro and in
vivo lymphangiogenesis by stimulating the migration and differen-
tiation of lymphatic endothelial cells via a S1P1/Gi/PLC/Ca2
signaling pathway. To the best of our knowledge, this study is the
first to establish that S1P is a lymphangiogenic lipid mediator.
During angiogenesis and lymphangiogenesis, the migration and
proliferation of endothelial cells are important procedures to
maintain net vascular structure by replenishing new endothelial
cells in the empty space caused by the migration of endothelial
cells toward stimuli. Several reports have supported the conclusion
that S1P has potent migratory effects on vascular endothelial
cells.21,33 In our study, the S1P (100 ng/mL)–induced migration of
HLECs was approximately 5.3-fold higher than that induced by
VEGF-C (500 ng/mL). This result indicates that the activity of S1P
to induce migration of lymphatic endothelial cells was similar to
that for vascular endothelial cells, in which S1P has 3- to 10-fold
greater effects than induction by VEGF-A or bFGF.34 Furthermore,
we also demonstrated that S1P (0.4 g/0.5 mL in Matrigel) can
promote pronounced lymphangiogenesis in vivo, similar to VEGF-C
Figure 3. S1P-induced in vitro lymphangiogenesis was mediated through the
S1P1/Giprotein. (A) Total RNA (100 ng) from HUVECs () or HLECs (f) was
amplified using primers for S1P receptors and -actin. For quantification, the targets
were normalized to -actin as an internal standard. (B) After a 2-hour preincubation
with 100 ng/mL PTX, HLECs were treated with 100 ng/mL S1P and 100 ng/mL PTX
for an additional 4 hours. The migrated HLECs were stained and counted in 3 random
fields. (C) HLECs were laid on a GFR Matrigel-coated 24-well plate and incubated
with 100 ng/mL S1P and 100 ng/mL PTX for 6 hours. Two randomly chosen fields per
well were photographed and the total tube area was analyzed using Scion Image. All
values are expressed as means (SD). Data are representative of 3 independent
experiments with similar results. NS and ** indicate no significant difference and a
statistically significant difference (P.01), respectively.
1134 YOON et al BLOOD, 15 AUGUST 2008 VOLUME 112, NUMBER 4
For personal use only. by guest on June 10, 2013. bloodjournal.hematologylibrary.orgFrom
(1 g/0.5 mL in Matrigel), suggesting that S1P might be an
important regulator of lymphangiogenesis. Although the physiolog-
ical concentration of S1P has been reported to be 0.2 g/mL in
serum, levels of S1P are elevated in pathological conditions:
0.4 g/mL in synovial fluids from rheumatoid arthritis patients and
0.9 g/mL in ascites of ovarian cancer patients.35,36 However, we
do not know the local concentration of S1P in tissues.
Although S1P directly induces the proliferation of human aortic
endothelial cells (HAECs) and bovine aortic endothelial cells,21,28,33
S1P (40 ng/mL) showed less potent mitogenic activities on these
cells (approximately 1.3-fold increase of proliferation in HAECs
and bovine aortic endothelial cells), suggesting that the mitogenic
activity of S1P is less potent than that of VEGF-A (10 ng/mL). In
this study, we found that S1P, up to 200 ng/mL, did not induce the
Figure 4. S1P-induced lymphangiogenesis was mediated through the S1P1. (A) HLECs were infected with retroviruses carrying S1P1 and S1P3 shRNA expression
vectors in pRS plasmid, after which stably transfected cells were obtained by selection with puromycin. pRS plasmid was used as control. After a 4-hour incubation with
100 ng/mL S1P,migrated HLECs were stained and counted in 3 random fields (). HLECs were laid on a GFR Matrigel-coated 24-well plate and incubated with 100 ng/mL S1P
for 6 hours. Two randomly chosen fields per well were photographed and the total tube area was analyzed using Scion Image (f). (B,C) Effect of S1P1 antagonist or S1P1
control molecule on the S1P-induced migration (B) and capillary-like tube formation (C) of HLECs was examined as described in panel A. (D,E) Ten days after subcutaneous
injection of Matrigel in C57BL/6 mice (5 mice per group), the Matrigel was removed, fixed, embedded in paraffin, sectioned at 4 m, and immunostained using antibodies
specific for Prox-1 (green) and podoplanin (red). (D) Representative photographs of untreated control mice, and mice treated with S1P (0.4 g) in the absence or presence of
S1P1 antagonist (100 M) or S1P1 control molecule (100 M). Scale bars represent 50 m. (E) The number of Prox-1/podoplaninlymphatic vessels per field was counted.
All values are expressed as means plus or minus SD (A-C) and means plus or minus SEM (E). In panels A-C, data are representative of 3 independent experiments with similar
results. ** indicates a statistically significant difference (P.01).
LYMPHANGIOGENICACTIVITY OF S1P 1135BLOOD, 15 AUGUST 2008 VOLUME 112, NUMBER 4
For personal use only. by guest on June 10, 2013. bloodjournal.hematologylibrary.orgFrom
proliferation of HLECs and HMVECs-dLy, but lymphatic vessels
formed by S1P were proliferative in in vivo Matrigel plug assay.
The discrepancies in in vitro and in vivo results might be explained
as follows. During angiogenesis and lymphangiogenesis, migration
is very earlier cellular response than proliferation by angiogenic or
lymphangiogenic stimuli, and is induced by lower concentrations
than those required for cell proliferation.37 Lymphatic endothelial
cells can be dedifferentiated and stimulated to proliferate when
contact inhibition is broken by migration. It is well known that
soluble forms of E-selectin, vascular cell adhesion molecule-1, and
intercellular adhesion molecule-1 show angiogenic responses in
vascular endothelial cells by induction of migration and differentia-
tion without any in vitro mitogenic activity of vascular endothelial
cells.22,38 Another possible explanation is that lymphatic endothe-
lial cells can be proliferative by lymphangiogenic factors secreted
from leukocytes recruited by S1P-induced migration. It has been
known that S1P promotes trafficking of various kinds of leukocytes
that secret lymphangiogenic factors such as VEGF-C/-D. However,
the precise mechanisms by which S1P induces in vivo proliferation
of lymphatic endothelial cells remain to be elucidated.
Although S1P receptors are coupled with various G proteins,
such as Gi,G
q, and G12/13, S1P1 is coupled with only the Giprotein;
other S1P receptors can bind to various G proteins. We identified 2
kinds of S1P receptors, S1P1 and S1P3, in HLECs. S1P-mediated
migration and tube formation were completely blocked by the
application of PTX. Since PTX specifically binds to the Giprotein
and inhibits the dissociation of the Giheterotrimer protein complex
into 2 subunits, our results suggest that the Giprotein is an
important signaling mediator of lymphangiogenesis induced by
extracellular S1P. Although S1P3 can also bind to the Giprotein,
PTX shows partial inhibition of the responsiveness mediated by
S1P3/Gi, but complete inhibition of that mediated by S1P1/Gi.We
further showed that shRNA-mediated down-regulation of S1P1
significantly blocked S1P-induced migration and differentiation of
HLECs, whereas down-regulation of S1P3 did not. Moreover, an
S1P1 selective antagonist significantly blocked S1P-induced in
vitro and in vivo lymphangiogenesis, whereas an inactive S1P1
control molecule did not. These results strongly suggest that
S1P-induced lymphangiogenesis is mediated mainly by activation
of the Giprotein coupled to S1P1.
S1P effectively activates PI3K, p44/42 MAPK, p38 MAPK, and
PLC in vascular endothelial cells, and these activations are
dramatically inhibited by treatment with PTX.21,39 However, PI3K
and p44/42 MAPK signaling pathway are less important to
S1P-induced vascular endothelial cell migration.21,28,40-42 Further-
more, whether the involvement of the p38 MAPK pathway is
important to S1P-induced vascular endothelial cell migration
remains unclear.21,28 In this study, we found that inhibition of PLC
activation and chelation of intracellular Ca2ions by treatment
with U73122 and BAPTA-AM resulted in a significant reduction in
S1P-induced migration and tube formation, as well as Ca2influx
to lymphatic endothelial cells. This suggests that Ca2ions
generated by active PLC are an important second messenger in
S1P-induced migration and differentiation of lymphatic endothelial
cells. Furthermore, application of inhibitors for PI3K, p44/42
MAPK, and p38 MAPK did not affect S1P-induced migration and
differentiation of HLECs, suggesting that PI3K, p44/42 MAPK,
and p38 MAPK are not involved in S1P-induced lymphangiogenesis.
Our present data strongly suggest that extracellular S1P pro-
motes new lymphatic vessel formation with the following mecha-
nisms (Figure 6). Extracellular S1P binds to and stimulates its
receptor S1P1, which is expressed on HLECs, resulting in the
Figure 5. S1P-induced in vitro lymphangiogenesis was mediated through the
PLC/Ca2pathway. (A)After a 4-hour incubation with 100 ng/mL S1P containing 10 M
LY294002, 1 mM L-NAME, 10 M PD98059, 25 M SB203580, 30 M BAPTA-AM, or
5M U73122, the migrated HLECs were stained and counted in 3 random fields. (B) HLECs
were laid on a GFR Matrigel-coated 24-well plate and incubated with 100 ng/mL S1P
containing 10 M LY294002, 1 mM L-NAME, 10 M PD98059, 25 M SB203580, 30 M
BAPTA-AM, or 5 M U73122 for 6 hours. Two randomly chosen fields per well were
photographed and the total tube area was analyzed using Scion Image. (C) HLECs were
loaded with fura-2/AM for 30 minutes. The cells were resuspended in Ca2-free Locke
solution, transferred to a quartz cuvette, and exposed to 100 ng/mL S1P with 100 ng/mL
PTX, 10 M LY294002, 1 mM L-NAME, 10 M PD98059, 25 M SB203580, 30 M
BAPTA-AM, or 5 M U73122. Relative intracellular Ca2influx was calculated from the
tracing. All values are expressed as means (SD). Data are representative of 3 independent
experiments with similar results. ** indicates a statistically significant difference (P.01).
1136 YOON et al BLOOD, 15 AUGUST 2008 VOLUME 112, NUMBER 4
For personal use only. by guest on June 10, 2013. bloodjournal.hematologylibrary.orgFrom
activation of coupled Giproteins. Dissociation of active heterotri-
meric Giproteins from activated S1P1 stimulates PLC. Activated
PLC stimulates the release of intracellular Ca2that causes the
migration and differentiation of HLECs, resulting in lymphangio-
genesis. S1P is generated by the phosphorylation of sphingosine,
which is mediated by SphK1 and SphK2. In many primary tumor
cells or inflammatory cells, the expression or activity of SphK1,
also known as oncogenic kinase,43 is up-regulated by many
angiogenic and inflammatory cytokines such as PDGF, epidermal
growth factor, tumor necrosis factor-, and interleukin-1.44 Al-
though it remains unclear how increased intracellular S1P is
secreted to the extracellular milieu in tumor-associated cells or
inflammatory cells, extracellular S1P can be generated by exported
SphK1 in HUVECs.45,46 Hence, the possibility exists that extracel-
lular S1P secreted from tumor-surrounded stromal cells, platelets,
or inflammatory cells,47 as well as tumor cells, can activate vascular
and lymphatic endothelial cells; this activation induces angiogen-
esis and lymphangiogenesis for tumor cell survival and tumor
metastasis to regional lymph nodes.
In this study, we demonstrated that S1P has in vitro and in vivo
lymphangiogenic activity and identified S1P as the first lipid
lymphangiogenic factor. This report may allow us to categorize
S1P, a bioactive lipid molecule, as a new lymphangiogenic factor.
Because several bioactive lipids, including lysophosphatidic acid,
sphingosylphosphorylcholine, gangliosides, and sphingomyelin,
have proangiogenic activity,48 investigations of other bioactive
lymphangiogenic lipids may be warranted to clarify the mecha-
nisms of lymphangiogenesis. In addition, because chronic inflam-
mation and cancer are highly related to each other, coordinated
regulation of lymphangiogenesis and inflammation by S1P sug-
gests that they may be pharmacological targets for development of
novel anti-inflammatory and antitumor metastatic therapies.
Acknowledgments
We thank Mr Hee-Yeoul Park and Ms Kyung Young Ji (Pohang
University of Science and Technology, Pohang, Republic of Korea)
for immunohistologic and confocal analysis, respectively.
This work was supported by the Korea Science and Engineering
Foundation (KOSEF, Daejeon, Republic of Korea) grant funded by the
Korea government (MOST, no. R15-2004-033-05001-0), supported by
a grant of the National R&D Program for Cancer Control (Goyang,
Republic of Korea), Ministry of Health & Welfare, Republic of Korea
(0320380-2), and supported by the Korea Basic Science Institute
(Daejeon, Republic of Korea) K-MeP (T27021; Y.S.G.). B.S.H., H.G.M.,
and S.L. were recipients of Brain Korea 21 fellowships.
Authorship
Contribution: C.M.Y. designed research, performed research, ana-
lyzed data, and drafted the paper; B.S.H. performed research,
analyzed data, and drafted the paper; H.G.M. and S.L. performed
research and analyzed data; P.-G.S. and Y.-K.K. designed and
advised research; C.-B.C. designed research, advised research, and
reviewed the paper; Y.S.G. designed research, advised research,
and drafted the paper.
Conflict-of-interest disclosure: The authors declare no compet-
ing financial interests.
Correspondence: Yong Song Gho, Division of Molecular and
Life Sciences, Pohang University of Science and Technology,
Pohang 790-784, Republic of Korea; e-mail: ysgho@postech.ac.kr.
References
1. Hla T. Signaling and biological actions of sphin-
gosine 1-phosphate. Pharmacol Res. 2003;47:
401-407.
2. Spiegel S, Foster D, Kolesnick R. Signal trans-
duction through lipid second messengers. Curr
Opin Cell Biol. 1996;8:159-167.
3. Pyne S, Pyne N. Sphingosine 1-phosphate sig-
nalling via the endothelial differentiation gene
family of G-protein-coupled receptors. Pharmacol
Ther. 2000;88:115-131.
4. Taha TA, Argraves KM, Obeid LM. Sphingosine-
1-phosphate receptors: receptor specificity ver-
sus functional redundancy. Biochim Biophys Acta.
2004;1682:48-55.
5. Liu Y, Wada R, Yamashita T, et al. Edg-1, the G
protein-coupled receptor for sphingosine-1-
phosphate, is essential for vascular maturation.
J Clin Invest. 2000;106:951-961.
6. Visentin B, Vekich JA, Sibbald BJ, et al. Validation
of an anti-sphingosine-1-phosphate antibody as a
potential therapeutic in reducing growth, invasion,
and angiogenesis in multiple tumor lineages.
Cancer Cell. 2006;9:225-238.
7. LaMontagne K, Littlewood-Evans A, Schnell C, et
al. Antagonism of sphingosine-1-phosphate re-
ceptors by FTY720 inhibits angiogenesis and tu-
mor vascularization. Cancer Res. 2006;66:221-
231.
8. Wei SH, Rosen H, Matheu MP, et al. Sphingosine
1-phosphate type 1 receptor agonism inhibits
transendothelial migration of medullary T cells to
lymphatic sinuses. Nat Immunol. 2005;6:1228-
1235.
9. Swartz MA. The physiology of the lymphatic sys-
tem. Adv Drug Deliv Rev. 2001;50:3-20.
10. Baluk P, Tammela T,Ator E, et al. Pathogenesis
of persistent lymphatic vessel hyperplasia in
chronic airway inflammation. J Clin Invest. 2005;
115:247-257.
11. Cao Y. Opinion: emerging mechanisms of tumour
lymphangiogenesis and lymphatic metastasis.
Nat Rev Cancer. 2005;5:735-743.
12. Joukov V, Pajusola K, Kaipainen A, et al.A novel
vascular endothelial growth factor, VEGF-C, is a
ligand for the Flt4 (VEGFR-3) and KDR
(VEGFR-2) receptor tyrosine kinases. EMBO J.
1996;15:290-298.
13. Joukov V, Sorsa T, Kumar V, et al. Proteolytic pro-
cessing regulates receptor specificity and activity
of VEGF-C. EMBO J. 1997;16:3898-3911.
14. Jeltsch M, Tammela T, Alitalo K, Wilting J. Gen-
esis and pathogenesis of lymphatic vessels. Cell
Tissue Res. 2003;314:69-84.
Figure 6. Plausible mechanism for S1P-mediated lymphangiogenesis. Extracel-
lular S1P, which is derived from tumor or inflammatory cells, binds to its receptor
S1P1 and stimulates the activation of coupled Giproteins. Dissociation of active
heterotrimeric Giproteins from activated S1P1 stimulates PLC, which causes the
release of intracellular Ca2, resulting in the stimulation of HLEC migration and
differentiation. Extracellular S1P activates lymphatic endothelial cells to induce
lymphangiogenesis, as well as vascular endothelial cells to induce angiogenesis, for
tumor metastasis or an immune response.
LYMPHANGIOGENICACTIVITY OF S1P 1137BLOOD, 15 AUGUST 2008 VOLUME 112, NUMBER 4
For personal use only. by guest on June 10, 2013. bloodjournal.hematologylibrary.orgFrom
15. Jeltsch M, Kaipainen A, Joukov V, et al. Hyperpla-
sia of lymphatic vessels in VEGF-C transgenic
mice. Science. 1997;276:1423-1425.
16. Cursiefen C, Chen L, Borges LP, et al. VEGF-A
stimulates lymphangiogenesis and hemangio-
genesis in inflammatory neovascularization via
macrophage recruitment. J Clin Invest. 2004;113:
1040-1050.
17. Kajiya K, Hirakawa S, Ma B, Drinnenberg I, Det-
mar M. Hepatocyte growth factor promotes lym-
phatic vessel formation and function. EMBO J.
2005;24:2885-2895.
18. Morisada T, Oike Y, Yamada Y, et al. Angiopoi-
etin-1 promotes LYVE-1-positive lymphatic vessel
formation. Blood. 2005;105:4649-4656.
19. Bjorndahl M, Cao R, Nissen LJ, et al. Insulin-like
growth factors 1 and 2 induce lymphangiogenesis
in vivo. Proc Natl Acad Sci U S A. 2005;102:
15593-15598.
20. Lee MJ, Thangada S, Claffey KP, et al. Vascular
endothelial cell adherens junction assembly and
morphogenesis induced by sphingosine-1-
phosphate. Cell. 1999;99:301-312.
21. Lee OH, Kim YM, Lee YM, et al. Sphingosine
1-phosphate induces angiogenesis: its angio-
genic action and signaling mechanism in human
umbilical vein endothelial cells. Biochem Biophys
Res Commun. 1999;264:743-750.
22. Gho YS, Kleinman HK, Sosne G. Angiogenic ac-
tivity of human soluble intercellular adhesion
molecule-1. Cancer Res. 1999;59:5128-5132.
23. Kim CW, Lee HM, Lee TH, Kang C, Kleinman HK,
Gho YS. Extracellular membrane vesicles from
tumor cells promote angiogenesis via sphingomy-
elin. Cancer Res. 2002;62:6312-6317.
24. Kim YH, Song M, Oh YS, et al. Inhibition of phos-
pholipase C-beta1-mediated signaling by O-Glc-
NAc modification. J Cell Physiol. 2006;207:689-
696.
25. Kluk MJ, Hla T. Signaling of sphingosine-1-
phosphate via the S1P/EDG-family of G-protein-
coupled receptors. Biochim Biophys Acta. 2002;
1582:72-80.
26. Tamama K, Okajima F. Sphingosine 1-phosphate
signaling in atherosclerosis and vascular biology.
Curr Opin Lipidol. 2002;13:489-495.
27. Takuwa Y. Subtype-specific differential regulation
of Rho family G proteins and cell migration by the
Edg family sphingosine-1-phosphate receptors.
Biochim Biophys Acta. 2002;1582:112-120.
28. Kimura T, Watanabe T, Sato K, et al. Sphingosine
1-phosphate stimulates proliferation and migra-
tion of human endothelial cells possibly through
the lipid receptors, Edg-1 and Edg-3. Biochem J.
2000;348(pt 1):71-76.
29. Igarashi J, Erwin PA, Dantas AP, Chen H, Michel
T. VEGF induces S1P1 receptors in endothelial
cells: Implications for cross-talk between sphin-
golipid and growth factor receptors. Proc Natl
Acad Sci U S A. 2003;100:10664-10669.
30. Sanna MG, Wang SK, Gonzalez-Cabrera PJ, et
al. Enhancement of capillary leakage and restora-
tion of lymphocyte egress by a chiral S1P1 antag-
onist in vivo. Nat Chem Biol. 2006;2:434-441.
31. Lee OH, Lee DJ, Kim YM, et al. Sphingosine
1-phosphate stimulates tyrosine phosphorylation
of focal adhesion kinase and chemotactic motility
of endothelial cells via the G(i) protein-linked
phospholipase C pathway. Biochem Biophys Res
Commun. 2000;268:47-53.
32. Butler J, Lana D, Round O, LaMontagne K. Func-
tional characterization of sphingosine 1-
phosphate receptor agonist in human endothelial
cells. Prostaglandins Other Lipid Mediat. 2004;
73:29-45.
33. Wang F, Van Brocklyn JR, Hobson JP, et al.
Sphingosine 1-phosphate stimulates cell migra-
tion through a G(i)-coupled cell surface receptor:
potential involvement in angiogenesis. J Biol
Chem. 1999;274:35343-35350.
34. English D, Kovala AT, Welch Z, et al. Induction of
endothelial cell chemotaxis by sphingosine
1-phosphate and stabilization of endothelial
monolayer barrier function by lysophosphatidic
acid, potential mediators of hematopoietic angio-
genesis. J Hematother Stem Cell Res. 1999;8:
627-634.
35. Hong G, Baudhuin LM, Xu Y. Sphingosine-1-
phosphate modulates growth and adhesion of
ovarian cancer cells. FEBS Lett. 1999;460:513-
518.
36. Kitano M, Hla T, Sekiguchi M, et al. Sphingosine
1-phosphate/sphingosine 1-phosphate receptor 1
signaling in rheumatoid synovium: regulation of
synovial proliferation and inflammatory gene ex-
pression. Arthritis Rheum. 2006;54:742-753.
37. Casscells W. Migration of smooth muscle and
endothelial cells: critical events in restenosis. Cir-
culation. 1992;86:723-729.
38. Koch AE, Halloran MM, Haskell CJ, Shah MR,
Polverini PJ. Angiogenesis mediated by soluble
forms of E-selectin and vascular cell adhesion
molecule-1. Nature. 1995;376:517-519.
39. Morales-Ruiz M, Lee MJ, Zollner S, et al. Sphin-
gosine 1-phosphate activates Akt, nitric oxide
production, and chemotaxis through a Gi protein/
phosphoinositide 3-kinase pathway in endothelial
cells. J Biol Chem. 2001;276:19672-19677.
40. Panetti TS, Nowlen J, Mosher DF. Sphingosine-1-
phosphate and lysophosphatidic acid stimulate
endothelial cell migration. Arterioscler Thromb
Vasc Biol. 2000;20:1013-1019.
41. Katkade V, Soyombo AA, Isordia-Salas I, et al.
Domain 5 of cleaved high molecular weight
kininogen inhibits endothelial cell migration
through Akt. Thromb Haemost. 2005;94:606-614.
42. Rikitake Y, Hirata K, Kawashima S, et al. Involve-
ment of endothelial nitric oxide in sphingosine-1-
phosphate-induced angiogenesis. Arterioscler
Thromb Vasc Biol. 2002;22:108-114.
43. Xia P, Gamble JR, Wang L, et al. An oncogenic
role of sphingosine kinase. Curr Biol. 2000;10:
1527-1530.
44. Doll F, Pfeilschifter J, Huwiler A. The epidermal
growth factor stimulates sphingosine kinase-1
expression and activity in the human mammary
carcinoma cell line MCF7. Biochim Biophys Acta.
2005;1738:72-81.
45. Ancellin N, Colmont C, Su J, et al. Extracellular
export of sphingosine kinase-1 enzyme: sphin-
gosine 1-phosphate generation and the induction
of angiogenic vascular maturation. J Biol Chem.
2002;277:6667-6675.
46. Venkataraman K, Thangada S, Michaud J, et al.
Extracellular export of sphingosine kinase-1a
contributes to the vascular S1P gradient. Bio-
chem J. 2006;397:461-471.
47. Zhi L, Leung BP, MelendezAJ. Sphingosine ki-
nase 1 regulates pro-inflammatory responses
triggered by TNFalpha in primary human mono-
cytes. J Cell Physiol. 2006;208:109-115.
48. Dyatlovitskaya EV, Kandyba AG. Sphingolipids in
tumor metastases and angiogenesis. Biochemis-
try (Mosc). 2006;71:347-353.
1138 YOON et al BLOOD, 15 AUGUST 2008 VOLUME 112, NUMBER 4
For personal use only. by guest on June 10, 2013. bloodjournal.hematologylibrary.orgFrom
... Sphingosine-1-phosphate: Sphingosine-1-phosphate (S1P) acts as a lymphangiogenic mediator in LECs. It induces migration, sprouting, capillary-like tube formation, and intracellular calcium mobilization in cultured human LECs in vitro and in a Matrigel plug assay in vivo [28]. In a murine model of breast cancer metastasis [29], S1P, suppressed by SK1-I, the specific sphingosine kinase 1 (a critical role in producing S1P and mediating tumor-induced lymphangiogenesis) inhibitor, reduced metastases to lymph nodes and lungs, and decreased overall tumor burden. ...
Article
Full-text available
The tumor microenvironment is a complex network of cells, extracellular matrix, and signaling molecules that plays a critical role in tumor progression and metastasis. Lymphatic and blood vessels are major routes for solid tumor metastasis and essential parts of tumor drainage conduits. However, recent studies have shown that lymphatic endothelial cells (LECs) and blood endothelial cells (BECs) also play multifaceted roles in the tumor microenvironment beyond their structural functions, particularly in hepatocellular carcinoma (HCC). This comprehensive review summarizes the diverse roles played by LECs and BECs in HCC, including their involvement in angiogenesis, immune modulation, lymphangiogenesis, and metastasis. By providing a detailed account of the complex interplay between LECs, BECs, and tumor cells, this review aims to shed light on future research directions regarding the immune regulatory function of LECs and potential therapeutic targets for HCC.
... At present, many S1P actions are mediated through the subtypes of S1P G protein-coupled receptors, which comprise S1P receptors 1-5. Through S1P receptor 1 (S1PR1), S1P plays an important role in regulating vascular remodeling, improving endothelial function, maintaining vascular wall permeability, and lymphangiogenesis [15,17,18]. The clinical role and molecular mechanism of the S1P pathway in hypoxia affecting cardiovascular function remain unclear. ...
Article
Full-text available
Background : Vascular failure (VF) and heart failure (HF) are extremely harmful and are the primary causes of hypoxia. Our previous results have shown that the sphingosine-1-phosphate (S1P) pathway was involved in regulating intermittent hypoxia–induced vascular defection, but the clinical role and molecular mechanism of the S1P pathway remain unclear. Methods : Normalized relative expression values and differentially expressed genes were downloaded in GSE145221 from the Gene Expression Omnibus dataset. WGCNA was used to construct a gene co-expression network. The Spearman correlation matrix was used to identify the top 500 highly correlated genes with the S1P pathway genes. R package clusterProfiler was used to perform Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses on the WGCNA modules. Homer software was utilized to identify regulatory motifs in the promoter and gene body regions of S1P pathway genes. An intermittent hypoxic injury cell model was induced by chronic intermittent hypoxia (CIH). ROS and TUNEL staining and Western blot were used to detect cell apoptosis and reactive oxygen species. Results : The transcriptional regulatory regions of S1P pathway genes were enriched with hypoxia-inducible factor 1-alpha, which indicated the close connection between the S1P pathway and the CIH process. In vitro, we confirmed that the endothelial cell apoptosis induced by CIH could be reversed by exogenous addition of S1P. Conclusions : This study elucidated the mechanism of the S1P pathway in regulating cardiovascular injury caused by CIH and provided a new strategy for early intervention in people with cardiovascular dysfunction induced by hypoxia.
... As described above, the concentration of S1P is nearly zero in interstitial fluid, while it is high in blood and lymph, and LECs are the major source of lymph S1P. S1P can promote LEC sprouting in vitro in an S1PR1-dependent manner [160]. The study by Surya et al. [161] proposed a model in which S1P and FSS act synergistically during the development and remodeling of the lymphatic system. ...
Article
Full-text available
The ever-growing research on lymphatic biology has clearly identified lymphatic vessels as key players that maintain human health through their functional roles in tissue fluid homeostasis, immunosurveillance, lipid metabolism and inflammation. It is therefore not surprising that the list of human diseases associated with lymphatic malfunctions has grown larger, including issues beyond lymphedema, a pathology traditionally associated with lymphatic drainage insufficiency. Thus, the discovery of factors and pathways that can promote optimal lymphatic functions may offer new therapeutic options. Accumulating evidence indicates that aside from biochemical factors, biomechanical signals also regulate lymphatic vessel expansion and functions postnatally. Here, we review how mechanical forces induced by fluid shear stress affect the behavior and functions of lymphatic vessels and the mechanisms lymphatic vessels employ to sense and transduce these mechanical cues into biological signals.
Article
Full-text available
In recent years, newly emerging therapies, such as immune checkpoint inhibitors and antibody-drug conjugates, have further improved outcomes for breast cancer patients. However, recurrent and metastatic breast cancer often eventually develops resistance to these drugs, and cure is still rare. As such, the development of new therapies for refractory breast cancer that differ from conventional mechanisms of action is necessary. Sphingosine-1-phosphate (S1P) is a key molecule with a variety of bioactive activities, including involvement in cancer cell proliferation, invasion, and metastasis. S1P also contributes to the formation of the cancer microenvironment by inducing surrounding vascular- and lymph-angiogenesis and regulating the immune system. In this article, we outline the basic mechanism of action of S1P, summarize previous findings on the function of S1P in cancer cells and the cancer microenvironment, and discuss the clinical significance of S1P in breast cancer and the therapeutic potential of targeting S1P signaling.
Article
Full-text available
Lymphatic vessels, comprising the secondary circulatory system in human body, play a multifaceted role in maintaining homeostasis among various tissues and organs. They are tasked with a serious of responsibilities, including the regulation of lymph absorption and transport, the orchestration of immune surveillance and responses. Lymphatic vessel development undergoes a series of sophisticated regulatory signaling pathways governing heterogeneous-origin cell populations stepwise to assemble into the highly specialized lymphatic vessel networks. Lymphangiogenesis, as defined by new lymphatic vessels sprouting from preexisting lymphatic vessels/embryonic veins, is the main developmental mechanism underlying the formation and expansion of lymphatic vessel networks in an embryo. However, abnormal lymphangiogenesis could be observed in many pathological conditions and has a close relationship with the development and progression of various diseases. Mechanistic studies have revealed a set of lymphangiogenic factors and cascades that may serve as the potential targets for regulating abnormal lymphangiogenesis, to further modulate the progression of diseases. Actually, an increasing number of clinical trials have demonstrated the promising interventions and showed the feasibility of currently available treatments for future clinical translation. Targeting lymphangiogenic promoters or inhibitors not only directly regulates abnormal lymphangiogenesis, but improves the efficacy of diverse treatments. In conclusion, we present a comprehensive overview of lymphatic vessel development and physiological functions, and describe the critical involvement of abnormal lymphangiogenesis in multiple diseases. Moreover, we summarize the targeting therapeutic values of abnormal lymphangiogenesis, providing novel perspectives for treatment strategy of multiple human diseases.
Article
Full-text available
The vascular and lymphatic systems are both comprised of a series of structurally distinct vessels lined with an inner layer of endothelial cells that function to provide a semi-permeable barrier to blood and lymph. Regulation of the endothelial barrier is critical for maintaining vascular and lymphatic barrier homeostasis, and its disruption is involved in accelerating various diseases, including systemic inflammation, acute organ failure, and cancer metastasis. One of the regulators of endothelial barrier function and integrity is sphingosine-1-phosphate (S1P), a bioactive sphingolipid metabolite secreted into the blood by erythrocytes, platelets, and endothelial cells and into the lymph by lymph endothelial cells. Binding of S1P to its G protein-coupled receptors, known as S1PR1-5, regulates its pleiotropic functions. This review outlines the structural and functional differences between vascular and lymphatic endothelium and describes current understanding of the importance of S1P/S1PR signaling in regulation of barrier functions. Most studies thus far have been primarily focused on the role of the S1P/S1PR1 axis in vasculature and have been summarized in several excellent reviews and thus, we will only discuss new perspectives on the molecular mechanisms of action of S1P and its receptors. Much less is known about the responses of the lymphatic endothelium to S1P and the functions of S1PRs in lymph endothelial cells and this is a major focus of this review. We also discuss current knowledge related to signaling pathways and factors regulated by the S1P/S1PR axis that control lymphatic endothelial cell junctional integrity. Gaps and limitations in current knowledge are highlighted together with the need to further understand the role of S1P receptors in the lymphatic system.
Article
Background: S1P (sphingosine-1-phosphate) has been reported to possess vasodilatory properties, but the underlying pathways are largely unknown. Methods: Isolated mouse mesenteric artery and endothelial cell models were used to determine S1P-induced vasodilation, intracellular calcium, membrane potentials, and calcium-activated potassium channels (KCa2.3 and KCa3.1). Effect of deletion of endothelial S1PR1 (type 1 S1P receptor) on vasodilation and blood pressure was evaluated. Results: Mesenteric arteries subjected to acute S1P stimulation displayed a dose-dependent vasodilation response, which was attenuated by blocking endothelial KCa2.3 or KCa3.1 channels. In cultured human umbilical vein endothelial cells, S1P stimulated immediate membrane potential hyperpolarization following activation of KCa2.3/KCa3.1 with elevated cytosolic Ca2+. Further, chronic S1P stimulation enhanced expression of KCa2.3 and KCa3.1 in human umbilical vein endothelial cells in dose- and time-dependent manners, which was abolished by disrupting either S1PR1-Ca2+ signaling or downstream Ca2+-activated calcineurin/NFAT (nuclear factor of activated T-cells) signaling. By combination of bioinformatics-based binding site prediction and chromatin immunoprecipitation assay, we revealed in human umbilical vein endothelial cells that chronic activation of S1P/S1PR1 promoted NFATc2 nuclear translocation and binding to promoter regions of KCa2.3 and KCa3.1 genes thus to upregulate transcription of these channels. Deletion of endothelial S1PR1 reduced expression of KCa2.3 and KCa3.1 in mesenteric arteries and exacerbated hypertension in mice with angiotensin II infusion. Conclusions: This study provides evidence for the mechanistic role of KCa2.3/KCa3.1-activated endothelium-dependent hyperpolarization in vasodilation and blood pressure homeostasis in response to S1P. This mechanistic demonstration would facilitate the development of new therapies for cardiovascular diseases associated with hypertension.
Article
Full-text available
The recently identified vascular endothelial growth factor C (VEGF-C) belongs to the platelet-derived growth factor (PDGF)/VEGF family of growth factors and is a ligand for the endothelial-specific receptor tyrosine kinases VEGFR-3 and VEGFR-2. The VEGF homology domain spans only about one-third of the cysteine-rich VEGF-C precursor. Here we have analysed the role of post-translational processing in VEGF-C secretion and function, as well as the structure of the mature VEGF-C. The stepwise proteolytic processing of VEGF-C generated several VEGF-C forms with increased activity towards VEGFR-3, but only the fully processed VEGF-C could activate VEGFR-2. Recombinant 'mature' VEGF-C made in yeast bound VEGFR-3 (KD = 135 pM) and VEGFR-2 (KD = 410 pM) and activated these receptors. Like VEGF, mature VEGF-C increased vascular permeability, as well as the migration and proliferation of endothelial cells. Unlike other members of the PDGF/VEGF family, mature VEGF-C formed mostly non-covalent homodimers. These data implicate proteolytic processing as a regulator of VEGF-C activity, and reveal novel structure–function relationships in the PDGF/VEGF family.
Article
Full-text available
Sphingolipid signaling pathways have been implicated in many critical cellular events. Sphingosine-1-phosphate (SPP), a sphingolipid metabolite found in high concentrations in platelets and blood, stimulates members of the endothelial differentiation gene (Edg) family of G protein–coupled receptors and triggers diverse effects, including cell growth, survival, migration, and morphogenesis. To determine the in vivo functions of the SPP/Edg signaling pathway, we disrupted the Edg1 gene in mice. Edg1–/– mice exhibited embryonic hemorrhage leading to intrauterine death between E12.5 and E14.5. Vasculogenesis and angiogenesis appeared normal in the mutant embryos. However, vascular maturation was incomplete due to a deficiency of vascular smooth muscle cells/pericytes. We also show that Edg-1 mediates an SPP-induced migration response that is defective in mutant cells due to an inability to activate the small GTPase, Rac. Our data reveal Edg-1 to be the first G protein–coupled receptor required for blood vessel formation and show that sphingolipid signaling is essential during mammalian development.
Article
Angiogenesis, the sprouting of new blood vessels from pre-existing ones, and the permeability of blood vessels are regulated by vascular endothelial growth factor (VEGF) via its two known receptors Flt1 (VEGFR-1) and KDR/Flk-1 (VEGFR-2). The Flt4 receptor tyrosine kinase is related to the VEGF receptors, but does not bind VEGF and its expression becomes restricted mainly to lymphatic endothelia during development. In this study, we have purified the Flt4 ligand, VEGF-C, and cloned its cDNA from human prostatic carcinoma cells. While VEGF-C is homologous to other members of the VEGF/platelet derived growth factor (PDGF) family, its C-terminal half contains extra cysteine-rich motifs characteristic of a protein component of silk produced by the larval salivary glands of the midge, Chironomus tentans. VEGF-C is proteolytically processed, binds Flt4, which we rename as VEGFR-3 and induces tyrosine autophosphorylation of VEGFR-3 and VEGFR-2. In addition, VEGF-C stimulated the migration of bovine capillary endothelial cells in collagen gel. VEGF-C is thus a novel regulator of endothelia, and its effects may extend beyond the lymphatic system, where Flt4 is expressed.
Article
Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid that has recently been shown to bind cell surface S1P receptors (previously called endothelial differentiation gene (Edg) receptors), which are members of the G-protein-coupled family of receptors. Signaling via S1P is a complex process, as cells usually express a number of these receptors on their surfaces. Many of the S1P receptors share common G-proteins, invoking the question of how these receptors are specific in their actions. This review describes the coupling pathways of S1P receptors, and highlights the in vitro and in vivo evidence for the “uniqueness” of each receptor in activating downstream signaling pathways, taking the effect of S1P on migration as an example.
Article
Angiogenesis, the formation of new blood vessels, is an important component of restoration of hematopoiesis after BMT, but the mediators involved in hematopoietic angiogenesis have not been identified. We examined the influence of the lipid growth factors, phosphatidic acid (PA), lysophos- phatidic acid (LPA), and sphingosine 1-phosphate (S1P), on several angiogenic properties of en- dothelial cells, including migration and stabilization of vascular barrier integrity. In a previous study, PA was found to disrupt the permeability of established endothelial monolayers, an early event in the angiogenic response that liberates cells for subsequent mobilization. In the present study, both PA and LPA weakly induced the chemotactic migration of endothelial cells from an established monolayer. The chemotactic response induced by PA and LPA was similar in intensity to that ob- served with optimal levels of the known protein endothelial cell chemoattractants, basic fibroblast growth factor (bFGF) and vascular endothelial growth factor (VEGF). A markedly greater chemo- tactic response was effected by nanomolar concentrations of S1P, indicating that this platelet-de- rived factor plays an important role in a key aspect of angiogenesis, chemotactic migration of en- dothelial cells. The chemotactic response to S1P was completely inhibited by preincubation of endothelial cells with antisense oligonucleotides to the high-affinity S1P receptor, Edg-1. In addi- tion, chemotaxis of endothelial cells to S1P was inhibited by preincubation of cells with specific in- hibitors of tryosine kinases, but inhibitors of phosphatidylinositol 3 9 kinase had little effect. Finally, LPA effectively stabilized endothelial monolayer barrier function, a late event in angiogenesis. Thus, the phospholipid growth factors, PA, S1P, and LPA, display divergent and potent effects on angio- genic properties of endothelial cells and giogenic differentiation of endothelial cells potentially act in tandem to effectively induce neovascularization. These mediators may thus exert important roles in restoration of hematopoiesis, as they facilitate blood vessel formation at sites of transplanted stem cells, allowing the progeny of engrafted progenitors to move from marrow sinusoids to the periph- eral vasculature.
Article
Sphingosine 1-phosphate (S1P) is a bioactive sphingolipid metabolite abundantly stored in platelets and released upon platelet activation. Recently, S1P has been postulated for its potential roles in angiogenesis. In this study, we provided several lines of evidence showing that S1P has angiogenic activity. In vitro, S1P stimulated DNA synthesis and chemotactic motility of human umbilical vein endothelial cells (HUVECs) in a dose-dependent manner, reaching a near maximum at 1 μM. S1P also significantly induced tube formation of HUVECs on Matrigel. Matrigel plug assay in mice revealed that S1P promotes angiogenesis in vivo. In addition, exposure of HUVECs to S1P led to rapid activation of extracellular signal-regulated kinases (ERKs) and p38 mitogen-activated protein kinase (p38 MAPK) in a pertussis toxin (PTX)-sensitive manner. Notably, HUVEC migration and tube formation in response to S1P were completely blocked by pretreatment with PTX. Further, the MEK inhibitor U0126 markedly inhibited S1P-induced tube formation but S1P-induced migration was not affected by inhibition of ERK and p38 MAPK. Taken together, these results indicate that S1P induces angiogenesis predominantly via Gi protein-coupled receptors in endothelial cells and suggest that S1P may act as an important modulator of platelet-induced angiogenesis.