Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

REVIEW  FOR  THE  2008  SATO  AWARD Circ J 2009; 73: 2183 – 2191

Vascular Integrity Mediated by Vascular Endothelial


Cadherin and Regulated by Sphingosine 1-Phosphate
and Angiopoietin-1

Naoki Mochizuki, MD

Development of blood vessels is coordinated by angiogenesis and stabilization of vascular endothelial cells (ECs).
The vascular network is established during embryogenesis to supply oxygen and nutrients to the tissues and organs.
However, after cardiac or peripheral ischemia is caused by occlusion of the vessels, new vessels must be formed
to rescue the ischemic tissues. Many angiogenic growth factors and chemokines are produced in the ischemic
tissue to induce angiogenic sprouting of preexisting vessels. Branched vessels must be again restabilized to form
mature vessels that deliver blood to the tissues. To this end, vascular EC–cell adhesion is tightly regulated by
cell–cell adhesion molecules and extracellular stimuli that activate G protein-coupled receptors and receptor
tyrosine kinases exclusively expressed on vascular ECs. This review spotlights the recent studies of vascular
endothelial cadherin and of sphingosine 1-phosphate signaling and angiopoietin-Tie signaling.   (Circ J 2009;
73: 2183 – 2191)
Key Words: Angiogenesis; Angiopoietin; Sphingosine 1-phosphate; Stabilization; Vascular endothelial cadherin

not only relax/contract but also loosen/tighten the cell–cell


How is Vascular Integrity Regulated? adhesions, which are constituted by endothelial-specific and
Blood Vessels Regulate Delivery of Blood and Nutrition common adhesion molecules. Structural characteristics of
by Controlling Blood Flow and Permeability vascular ECs determine the supply of the molecules in the
Neovascularization is required for developmental (physio- blood vessels across the monolayer of ECs.9 How the mole-
logical) and pathological angiogenesis.1 The latter is found cules pass through this barrier has been extensively studied.
in the ischemic heart, peripheral arterial disease and tumors. The concentration of molecules inside or outside of the
Thus it is important to assist neovascularization in ischemic monolayer is determined by transcellular transport and
diseases and to block angiogenesis in cancer.2 The vascular paracellular permeability.10 The transcellular transport of
network of the body maintains the circulation of blood and the molecules occurs in the fenestrated capillaries, whereas
nutrients necessary for the peripheral tissues and organs by the paracellular pathway is via the continuous capillaries.
relaxing and constricting autonomously or non-autono- The surface of the endothelial cellular that consists of
mously. Many of the molecules produced by endothelial cells glycocalyx sieves the molecules that will be subjected to
(ECs) and vascular smooth muscle cells (VSMCs), which the transcellular or paracellular pathway.11 The presence of
affect relaxation or contraction of blood vessels in both basement membrane beneath the endothelial monolayer
an autocrine and paracrine manner, have been identified. affects the diffusion of the molecules that pass the endo-
Among them, sphingosine 1-phosphate (S1P) released from thelial layer. ECs are attached to the extracellular matrix
vascular ECs stimulates S1P receptors on the ECs to induce (ECM) via integrin receptors to form an EC–ECM adhe-
vascular relaxation and those on the VSMCs to induce vas- sion,12 which is involved in both the transcellular and para-
cular contraction.3 Vascular ECs produce nitric oxide (NO) cellular pathways. In clear contrast, the cell–cell adhesions
to induce the relaxation of VSMCs. The NO-dependent directly control the paracellular pathway (Figure 1).
cGMP-PKG signal modulated by phosphodiesterase in
VSMCs is central to vascular relaxation.4,5 On the other Roles of Adhesion Molecules Expressed on Vascular ECs
hand, ECs synthesize vascular contraction factors, including in Extravasation of Blood Cells
the metabolites of arachidonic acids.6 Vascular contraction The intercellular adhesions between vascular ECs mainly
and relaxation are mainly regulated by the activity myosin constitute adherens junctions and tight junctions. Vascular
light-chain kinase (MLC-K) controlled by the intracellular ECs express endothelial-specific adhesion molecules: vas-
increase/decrease in Ca2+ and Rho kinase in the signaling cular endothelial cadherin (VE-cad), and platelet and endo-
provoked by G protein-coupled receptors (GPCR).7 Recent- thelial adhesion molecule-1 (PECAM-1).13–15 In addition
ly, it has been also shown that oxidative stress affects vas- to these adhesive molecules, junctional adhesion molecule
cular cells.8 In order to deliver nutrients and blood, vessels (JAM), claudin-5 and nectin are involved in the formation

Received September 8, 2009; accepted September 14, 2009; released online October 19, 2009
Department of Structural Analysis, National Cardiovascular Center Research Institute, Suita, Japan
Mailing address:  Naoki Mochizuki, MD, Department of Structural Analysis, National Cardiovascular Center Research Institute, 5-7-1
Fujishirodai, Suita 565-8565, Japan.   E-mail: nmochizu@ri.ncvc.go.jp
All rights are reserved to the Japanese Circulation Society.  For permissions, please e-mail: cj@j-circ.or.jp

Circulation Journal Vol.73, December 2009


2184 MOCHIZUKI N

Figure 1.   Two types, continuous and fenestrated, of endothelial cells (ECs) regulate the paracellular and transcellular
transport of molecules across the monolayer of the endothelium. Glycocalyx expressed on both continuous ECs and
fenestrated ECs lines the luminal surface of the endothelium and limits the passage of molecules by their charge, suggest-
ing the glycocalyx is the first stage in regulation of permeability by sieving the molecules. An electron microscopic view
of the glycocalyx detected by alcian blue-lanthanium nitrate staining of rat coronary artery. Beneath the endothelium,
basement membrane (BM) forms the extracellular matrix (ECM) –endothelial cell adhesion to stabilize the endothelial
cells to the ECM. Furthermore, pericytes/vascular smooth muscle cells and podocytes in continuous ECs and fenestrated
glomerular ECs support the ECs by respectively forming cell–cell contacts and by secreting angiopoietin-1.

Figure 2.   Schema of how leukocytes extra-


vasate across the endothelium and the intercel-
lular adhesion molecules of the endothelium.
Local inflammation requires an accumulation
of leukocytes across the endothelium through
several steps: attachment of leukocytes to the
endothelium, rolling of leukocytes on the endo-
thelium, firm attachment of leukocytes to the
endothelium via integrin, and transmigration
via cell–cell adhesion molecules. Cell–cell
junction molecules not only participate in the
extravasation of blood cells but also directly
regulate loosening and tightening of the cell–
cell adhesion. Assembly of cortical actin aug-
ments the accumulation of VE-cadherin (VE-
cad), but not PECAM-1, at the cell–cell contact
by being linked to VE-cad through β-ctn and
α-ctn. JAM, junctional adhesion molecule;
PECAM-1, platelet and endothelial cell adhe-
sion molecule-1, p120ctn, p120 catenin;β-ctn,
β-catenin;α-ctn,α-catenin.

of cell–cell contacts.16–18 VE-cad constitutes adherens junc- FoxO-1 and β-ctn complex outside of the nucleus.19
tion, whereas claudin-5, JAM and ESAM form tight junc- PECAM-1 localizes in the region outside of the adherens
tions. Recently, VE-cad was shown to transcriptionally and tight junctions20 (Figure 2).
upregulate claudin-5 by inducing the nuclear export of β- The interendothelial cell–cell adhesions regulate the
catenin (β-ctn), which is mediated by the formation of a transendothelial migration of blood cells into the intersti-

Circulation Journal Vol.73, December 2009


Stabilization of Endothelial Cell–Cell Contact (2008 Sato Award) 2185

Figure 3.   Stabilization (indicated by black font) and destabilization (indicated by red font) is controlled by receptor
tyrosine kinases and G protein-coupled receptors expressed on the vascular endothelial cells. Activation of sphingosine 1-
phosphate receptor 1 (S1P1) and Tie2 enhances cell–cell adhesions, whereas that of VEGF receptor and PAR-1 receptor
destabilizes the cell–cell adhesion. Gi-mediated signal downstream of S1P1 exerts a vasculoprotective action via endo-
thelial nitric oxide synthase, while Gq- and G12/13-induced signal promotes the contraction of endothelial cells, thereby
physically dissociating cell–cell contacts. MLC-PPase, myosin light chain phosphatase; CaM-K, calmodulin kinase;
MLC-K, myosin light chain kinase.

tial cells, as well as permeability. There are several steps bin stimulation, and it then signals to Gq and G12/13 proteins
observed in leukocyte recruitment to ECs: initial attachment to increase permeability.30 Gq and G12/13 activation leads to
and rolling of leukocytes through their interaction with increased intracellular Ca2+, which enhances activation of
integrin (leukocytes) and p- and e-selectins (ECs), then calmodulin kinase (CaM-K)/MLC-K, leading to activation
firm attachment and subsequent recruitment of leukocytes of Rho–Rho kinase to reduce MLC phosphatase. Both
to the cell–cell junction mediated by the interaction between signals result in actin-myosin-dependent contraction of
integrins on the leukocytes and ICAM-1/VCAM-1 on the vascular ECs.31 Bradykinin produced from kininogen in the
ECs and transmigration mediated by several molecules blood acts on the Gq/i protein-coupled B1 and B2 receptors
expressed on the interendothelial junction.21 This inter- expressed on ECs to increase permeability.32 In contrast to
action has become a target for reducing inflammation.22 thrombin, bradykinin induces vascular permeability inde-
The homophilic association of PECAM-1 contributes to pendently of MLC-K and Rho kinase.33 Histamine released
transendothelial migration after the initial attachment of from mast cells also transiently increases permeability, com-
the leukocytes to ECs.23 The engagement of integrins with pared with thrombin, by activating H1 receptors expressed
ICAM-1 results in phosphorylation of VE-cad, thereby on ECs.34 Platelet-activating factor augments vascular per-
weakening the VE-cad-mediated cell–cell junctions.24 Thus, meability independently of the actin–myosin contraction,
VE-cad works as the gate keeper against leukocytes, which similar to bradykinin33 (Figure 3).
induce inflammation (Figure 2).
In this review, I focus on vascular integrity controlled VE-Cad Regulates Barrier Function in ECs
by the VE-cad-mediated cell–cell adhesion, angiopoietin-1 VE-cad and N-cadherin belonging to the classical cadherin
(Ang1)–Tie2 signaling and the S1P-mediated signal as family that are expressed on the endothelium and undergo
representative cell adhesion molecules, tyrosine kinase Ca2+-dependent homophilic trans- and cis-interactions. VE-
receptor-mediated signals and GPCR-mediated signals, cad accumulates at the interendothelial cell–cell junctions,
respectively. The atrial natriuretic peptide family of mole- whereas N-cadherin is mostly located between ECs and
cules is essential for vascular integrity by activating their smooth muscle cells.35,36 In contrast, there is a report of
receptors and exerting guanylate cyclase activity. The localization of N-cadherin at the interendothelial junc-
beneficial effects of natriuretic peptides on the vasculature tions.37 Similar to other classical cadherins, VE-cad is a
have been reviewed.25,26 single-membrane-spanning molecule that contains 5 extra-
cellular cadherin domains, a transmembrane domain and a
cytoplasmic domain that binds p120 catenin (p120ctn) and
Control of Vascular Integrity β-ctn at the juxtamembrane domain and the carboxy ter-
Permeability Regulating Factors minal domain, respectively.38
Increased vascular permeability is found in inflammation, The expression of VE-cad is regulated by many steps:
hypoxia and thrombosis. Under these conditions, vascular shedding, endocytosis, synthesis, transport and stabiliza-
permeability is either enhanced or reduced by extracellular tion at the cell–cell contacts (Figure 4). The extracellular
stimuli that activate cell surface receptors, tyrosine kinases domain of VE-cad is detected in the culture medium from
and GPCRs. Vascular endothelial growth factor (VEGF) is apoptotic ECs, indicating shedding of VE-cad.39 Recently,
a vascular permeability increasing factor,27 whereas Ang1 the metalloprotease-disintegrin, ADAM9, was reported to
activates Tie2 receptor tyrosine kinase and counteracts the potentially cleave VE-cad.40
VEGF–VEGFR-2 signal to maintain vascular integrity.28,29 VEGF induces endocytosis of VE-cad through the phos-
Several permeability-increasing factors are induced phorylation of VE-cad at Ser 665. Activation of Rac by
through GPCR activation. Thrombin is concentrated in Vav2 in VEGF-stimulated cells leads to PAK-dependent
clots and released into the blood to activate PAR-1, which is phosphorylation of Ser at 665, resulting in endocytosis of
activated when its amino-terminus is cleaved upon throm- VE-cad in a manner dependent onβ-arrestin2.41 Src tyrosine

Circulation Journal Vol.73, December 2009


2186 MOCHIZUKI N

Figure 4.   Expression of vascular endothelial cadherin (VE-cad) at the cell–cell contact depends on shedding, endocytosis,
synthesis, transport and stabilization of VE-cad. Shedding, endocytosis and reduced transport decrease the expression of
VE-cad at the cell–cell contact, whereas accelerated transport, synthesis and stabilization results in increased expression at
the cell–cell contact. Therefore, the balance of these regulatory steps determines the expression of VE-cad and the mainte-
nance of vascular integrity.

kinase is involved in the Vav2–Rac–PAK signal-mediated p120ctn determines a set point mechanism that regulates
phosphorylation of VE-cad. Although phosphorylation of cadherin expression levels by controlling the internaliza-
the cytoplasmic domain of VE-cad is accompanied by an tion and degradation of VE-cad.51 Reduction of p120ctn
increase in vascular permeability in vitro and in vivo,42,43 it by siRNA results in decreased expression of endogenous
has not been explored whether tyrosine-phosphorylated VE-cad. Forced expression of p120ctn reduces the clathrin-
VE-cad is prone to endocytosis. Src is also involved in mediated endocytosis of VE-cad.52 The essential role of the
tyrosine phosphorylation of VE-cad.44 association between p120ctn and VE-cad is supported by
VE-cad associates with VEGFR-2 (Flk-1) and reduces evidence that overexpression of the juxtamembrane domain
the internalization of VEGFR-2.45 The significance of the of VE-cad reduces VE-cad expression and barrier function.53
association of VE-cad with VEGFR-2 is confirmed by mice Furthermore, p120ctn regulates the transendothelial migra-
lacking the entire VE-cad and those lacking its cytoplasmic tion of leukocytes by altering the tyrosine phosphorylation
domain,46 although the precise mechanism of VEGF-medi- of VE-cad.54 Conversely, phosphorylation of VE-cad at
ated internalization of VE-cad has yet to be clarified. Tyr658 and Tyr731 inhibits binding of p120ctn to VE-cad.44
The VE-cad promoter has been isolated and is used as Phosphorylation of the 2 tyrosines correlates with a reduc-
an endothelial-specific promoter.47 There are 2 conserved tion in the barrier function of ECs. Another study reports
Ets binding sites in the 5’ region (within −200 bp) of the that Golgi-associated cPLA2α regulates the transport of
transcription initiation site of the VE-cad gene (CDH5). VE-cad, as well as claudin-5, from the Golgi apparatus to
Chromatin immunoprecipitation assay confirms that Erg is the cell–cell junction.55
capable of binding these 2 sites. Perturbation of Erg using Theβ-ctn binding to E-cadherin is well known as linking
antisense oligonucleotides reduces the expression of VE- link E-cadherin to actin via α-catenin (α-ctn) to stabilize
cad in HUVECs, indicating the involvement of Erg in the E-cadherin-mediated cell–cell adhesion.56 Thus, similar to
transcriptional regulation of VE-cad.48 There are 6 poten- E-cadherin, VE-cad is thought to be linked to actin fibers to
tial and putative hypoxia response elements, suggesting maintain the integrity of endothelial adhesions. Depletion
the regulation of VE-cad expression by hypoxia-inducible of β-ctn in the ECs of mice results in defective vascular
factors (HIF-1α and HIF-2α).49 Although CDH5 is not patterning, suggesting the critical role of β-ctn in interen-
responsive to hypoxia, HIF-2α, but not HIF-1α, activates dothelial adhesions.57 Therefore, stabilization of VE-cad
the promoter of VE-cad. Serum response factor (SRF) is depends on the link between VE-cad and actin. However,
also reported to be required for the expression of VE-cad, this classical model is challenged by the Nelson58 and Weis
as confirmed by the depletion of SRF in the vascular ECs groups.59 They claim thatα-ctn does not connect actin fibers
of mice.50 SRF binds to CArG box found in the regulatory to E-cadherin and propose a dynamic model instead of the
regions of CDH5. classical static model. In addition, phosphorylation ofβ-ctn
The cytoplasmic domain of VE-cad is important for the as well as VE-cad affects the binding of β-ctn to VE-cad.
trafficking and stabilization of VE-cad because it provides There are several phosphatases that dephosphorylate the
the binding sites for p120ctn and β-ctn. The amount of molecules participating in the regulation of cell–cell adhe-

Circulation Journal Vol.73, December 2009


Stabilization of Endothelial Cell–Cell Contact (2008 Sato Award) 2187

Figure 5.   Fluorescence recovery after photobleaching technique of analyzing the mobility of vascular endothelial
cadherin (VE-cad) at the cell–cell contact. Fluorescence recovery at the region of interest (cell–cell contact marked by the
yellow circle) after photobleaching of VE-cad tagged with green fluorescent protein (VE-cad–GFP) can be detected by a
confocal microscope. The mobile and immobile fractions can be deduced from the graph (Top) of the plot of raw data
(Middle). The decrease in the mobile fraction denoted by the red arrow and red line reflects the increased stabilization of
VE-cad–GFP at the cell–cell contact. We can compare the dynamics of VE-cad to PECAM-1 by constructing plasmids
expressing VE-cad–GFP and PECAM-1 tagged with GFP (PECAM-1–GFP) and their chimeric molecules (Bottom).

sions. Thrombin induces phosphorylation of SHP2 and its further demonstrate that the cAMP–Epac (another GEF for
dissociation of VE-cad, resulting in the phosphorylation of Rap1) –Rap1 signal stabilizes the VE-cad-mediated cell–cell
β-ctn and a reduction of the amount of β-ctn that binds to adhesion.67,68 Although cAMP-increasing stimuli are known
VE-cad.60 The requirement of vascular endothelial protein to stabilize the cell–cell adhesion via protein kinase A (PKA),
tyrosine phosphatase (VE-PTP) has been demonstrated.61 Epac instead of PKA is critical for cAMP-induced stabili-
Downregulation of VE-PTP enhances EC permeability and zation. Accumulation of VE-cad at the cell–cell contacts is
inhibits VE-cad adhesion. The essential substrate of VE-PTP increased by an Epac-specific activator, 8-pCPT-2’-O-Me-
in VE-cad-dependent cell–cell adhesions is plakoglobin, cAMP (known as 007), as well as forskolin in vitro and in
notβ-ctn. DEP-1/CD148 is reported to be required for VE- vivo.67,69 007 not only reduces EC permeability, but also
cad-dependent inhibition of VEGFR-2 activation.62 inhibits transendothelial migration of leukocytes.70
The cellular actin cytoskeleton that determines cell shape Although the downstream effectors of Rap1 in the stabili-
is regulated by the Rho-family GTPases (Rho, Rac and zation of VE-cad are not fully understood, several potential
Cdc42). Indeed, Rho-family GTPases are involved in signaling pathways have been suggested. Cerebral cavern-
cadherin-mediated cell–cell adhesion by controlling the ous malformation (CCM) is associated with defective endo-
actin-based cytoskeleton.63 Besides the Rho-family GTPases, thelial junctions. There are 3 genes related to CCM: CCM1,
Rap1, a small GTPase, is well known for stabilizing the CCM2 and CCM3.71,72 CCM1/KRIT1 (K-Rev1 interaction
VE-cad-mediated cell–cell adhesion.64,65 trapped gene 1), a Rap1-binding protein, is reported to be
involved in Epac1/Rap1-regulated permeability of EC–cell
Stabilization of VE-Cad at the Cell–Cell Contact junctions.73 KRIT1 localizes at the cell–cell contacts and
by Rap1-Mediated Signal associatesβ-ctn, thereby being indirectly connected to VE-
We have previously shown that MAGI-1 binding to β-ctn cad. CCM2 functions as a scaffold protein and forms com-
recruits PDZ-GEF1, a guanine nucleotide exchange factor plexes with CCM1 and CCM3.74 CCM1 might connect the
(GEF) for Rap1, thereby activating Rap1 at the interendothe- heart-of-glass (Heg)-mediated signal to CCM2 to maintain
lial junctions upon engagement of VE-cad.66 My and others vascular structure, as well as VE-cad-β-ctn.75 Target disrup-

Circulation Journal Vol.73, December 2009


2188 MOCHIZUKI N

Figure 6.   Angiopoietin-1 (Ang1) secreted from pericytes induces the trans-association of Tie2 at the cell–cell contact
and subsequently activates the Akt–MEF2–KLF2 signal. KLF2 promotes the expression of endothelial nitric oxide synthase
and reduces the expression of VCAM-1. Therefore, the Ang1–Tie2 signal is anti-inflammation and pro-quiescence (Upper).
Sphingosine 1-phosphate (S1P) is produced from sphingosine (Sph) in the plasma membrane by sphingosine kinase
(Sph-K) in the cytoplasm. Thus, to activate sphingosine 1-phosphate receptor 1 (S1P1) that governs vascular stability, S1P
is released outside the cell from the cytoplasm. Spinster2 (Spns2) functions as a S1P transporter. Thus, Spns2 is likely to
participate in S1P-mediated signaling in a paracrine or autocrine manner to control vascular integrity (Lower).

tion of Ccm2 in mice results in failure of lumen formation skolin induces cortical actin bundling parallel to the cell–
and embryonic lethality. Whitehead et al showed activation cell contacts. The mobile fraction of VE-cad was decreased
of RhoA in CCM2-depleted cells and alteration of cell–cell upon forskolin stimulation, while that of PECAM-1 was
junctional structure because of the activation of RhoA.74 unchanged, suggesting a significant link between actin and
Therefore, the Rap1-CCMs signal is proposed to regulate cell adhesion molecules for the stabilization of adhesion
the cell–cell adhesion through modification of the RhoA molecules at the cell–cell contacts when the formation of
signaling that controls the actin cytoskeleton. cortical actin bundling is augmented (unpublished data).
Rap1 has been demonstrated to regulate actin cytoskeleton The precise molecular mechanism of how Rap1 regulates
via signaling to Rho-family GTPases. Rap1 is capable of acti- the stabilization of VE-cad at the cell–cell contact needs to
vating Rac and determining the localization of GFEs, Vav2 be clarified (Figure 5).
and Tiam1 for the Rho-family GTPases.76 Cell–cell contact-
dependent engagement of nectin induces the activation of Ang1/Tie2-Mediated Integrity of Vascular ECs
Rap1 and subsequent activation of Rac and Cdc42 via Vav2 Ang1 activates Tie2 receptor tyrosine kinase and counter-
and FRG, respectively.15 The role of affadin, which can bind acts the VEGF-VEGFR-2 signal to maintain vascular integ-
to Rap1, nectin, and zonula occludens-1 (ZO-1), has not been rity.28,29 Another angiopoietin, Ang2, is thought to compete
clearly investigated in the Rap1-mediated signal, although its with Ang1 and induce angiogenesis.80 Ang1 strengthens the
role in the EC–ECM has been characterized.77 Rho-family PECAM-1- and VE-cad-mediated intercellular junctions
GTPases are regulated by other molecules localized at the by inducing the dephosphorylation of PECAM-1 and VE-
cell–cell contacts. p120ctn and RhoA mutually inhibit their cad and by inhibiting TNF-α-induced transmigration of
functions by binding each other.78 p120ctn balances the leukocytes.81 We and others have demonstrated that Ang1
activity of Rho and Rac by binding to p190RhoGAP at the localizes Tie2 to the intercellular junctions by bridging
cell–cell contacts when p190RhoGAP is recruited to the Tie2.82,83 Trans-associated Tie2 preferentially activates
cell–cell contacts upon growth factor stimulation.79 AKT and reduces permeability. VE-PTP is also recruited to
In the light of cortical actin-dependent stabilization of the cell–cell junctions when stimulated by Ang1, suggest-
VE-cad, how Rap1 regulates VE-cad stabilization at the ing a potential role in reducing the permeability mediated
cell–cell contacts has been investigated. Although the static by enhanced accumulation of VE-cad. Antibody against
linkage of cadherin and actin is challenged by the Nelson58 VE-PTP can mimic depletion of VE-PTP and induces acti-
and Weis groups,59 the importance of the link between vation of Tie2. Thus, the association of VE-PTP with Tie2
cadherin and actin cytoskeleton has not been contradicted. appears to be very important for the regulation of endothelial
The importance of the link between VE-cad and actin can stabilization and proliferation.84 We further extended our
be claimed by comparing VE-cad with PECAM-1, which is studies of the function of trans-associated Tie2 at the cell–
not linked to actin. Thus, my group used a fluorescence cell contacts. Analysis of gene expression by cDNA array
recovery after photobleaching (FRAP) experiment using enabled identification of the responsible molecules that
VE-cad or PECAM-1 tagged with green fluorescence protein promote vascular quiescence, 1 of them being Krüppel-like
(GFP) (VE-cad-GFP and PECAM-1-GFP, respectively) to factor 2 (KLF2).82 KLF2 is a zinc finger transcription factor
monitor the dynamics at the junctions. We found that for- that belongs to the KLF family and is expressed in vascular

Circulation Journal Vol.73, December 2009


Stabilization of Endothelial Cell–Cell Contact (2008 Sato Award) 2189

ECs.85 The importance of KLF2 for the stability of blood mutant Spns2 (R153S) and found that Spns2 functioned as
vessels is confirmed by deletion of KLF2.86 One can specu- a transporter of S1P. Collectively, S1P released from vas-
late that KLF2 is a vascular protective transcription factor cular ECs via Spns2 might activates S1P1 expressed on
because it upregulates endothelial NO synthase (eNOS), vascular ECs to maintain vascular integrity (Figure 6).
natriuretic peptides and downregulates VCAM-1, E-selectin,
VEGFR-2, and endothelin-1.
My group delineated the signaling downstream of acti- Conclusion
vated Tie2 at the cell–cell contacts.87 Upregulation of KLF2 Vascular EC integrity is important for protecting the vascu-
is dependent on MEF2, but not ERK5, which is regulated lature from thrombosis, atherosclerosis and abnormal angio-
by the phosphatidylinositol 3-kinase (PI3K)-AKT signal. genesis. Because the ideal vasculature is organized during
Attachment of monocytes to cultured ECs stimulated with embryonic development, established blood vessels need to
VEGF was impeded by Ang1. This inhibition was reversed be maintained to keep blood flowing smoothly around the
by the depletion of KLF2, indicating that the inhibitory effect body. In this review, I have focused on the molecules that
of Ang1 on inflammatory cell adhesion to ECs is regulated are central to the stability of vascular ECs. Other vascular
by KLF2. Therefore, we conclude that Ang1-dependent sta- stabilization factors besides Ang1 and S1P will be identified
bility of ECs depends on KLF2 (Figure 6). to extend our knowledge of vascular integrity. Destabiliza-
tion of cell–cell contacts is required for preexisting vessels
S1P Regulates Integrity of ECs to promote the angiogenesis that should be re-stabilized
S1P is thought to be released from activated platelets and once ischemia is improved. Therefore, it is necessary to
erythrocytes.88,89 Its receptors, S1P1-S1P5, are GPCR and understand the stabilization and destabilization cycle in the
expressed in various tissues, including vascular ECs and vasculature under both pathological and physiological con-
VSMCs.90 S1P1 is coupled to Gi and induces PI3K-depen- ditions. We, as cardiologists, are expected to develop new
dent eNOS and Rac activation, whereas S1P2 and S1P3 are therapeutic strategies to rescue and reverse the damaged
coupled to Gi, Gq, and G12/13.91 Activation of Gq and that heart by regenerative medicine. We need to think about the
of G12/13 results in phosphorylation of MLC and subsequent regeneration of healthy vasculature, as well as the myocar-
actin–myosin-dependent contraction via the phospholipase dium, to regenerate the heart. Thus, it is important for us to
C-β(PLCβ)-Ca2+/CaM kinase signal and RhoA–Rho kinase instigate understanding of how we can control both regen-
signal, respectively.91 eration and maintenance of preexisting tissues.
Vascular integrity controlled by S1P is dependent on
Gi–Rac–PAK signaling that induces re-arrangement of the
Acknowledgments
actin cytoskeleton.92 S1P-mediated enhancement of the
ECs barrier function is explained by the phosphorylation of The research described here and done by Dr Mochizuki’s group has been
supported in part by grants from the Ministry of Education, Science,
cortactin and its binding to MLC-K.93 ZO-1 is essential for Sports and Culture of Japan; the Ministry of Health, Labour, and Welfare
the S1P-regulated barrier integrity of ECs, as evidenced by of Japan; the Program for the Promotion of Fundamental Studies in Health
the fact that depletion of ZO-1 leads to loss of the S1P- Sciences of the National Institute of Biomedical Innovation; Astra-Zeneca
enhanced barrier function.94 The association of VE-cad with Research Grant; Research Grants in the Natural Sciences from the
β-ctn-focal adhesion kinase upon S1P stimulation regulates Mitsubishi Foundation; and Takeda Science Foundation.
I am grateful to emeritus professors Hisakazu Yasuda and Kazuo
the cortical actin rearrangement that stabilizes the cell–cell Nagashima at Hokkaido University School of Medicine for teaching me
junction.95 Plasma S1P-less mice in which sphingosine the importance of basic medical science; Professors Michiyuki Matsuda
kinases (I and II) are knocked out show vascular leakage, (Kyoto University) and Kenji Kangawa (Director general at National
suggesting the mechanism of S1P1-regulated vascular integ- cardiovascular Center Research Institute) for making me rapt in signaling
and imaging; and many collaborators and friends, including Professors
rity of ECs in vivo.96 Thus, maintenance of vascular EC Hirofumi Sawa and Naomasa Makita, for their continuous encouragement.
integrity is regulated by S1P without activation of platelets. Finally, I thank the members of my laboratory, who study basic cardiovas-
From which cells is S1P released? S1P is synthesized cular medicine with me.
from sphingosine localized in the plasma membrane by
sphingosine kinase I and II in the cytoplasm.97 Therefore, References
S1P produced in the cells must be released from inside to    1. Carmeliet P. Angiogenesis in health and disease. Nat Med 2003; 9:
outside of the cells via transporters that activate S1P recep- 653 – 660.
tors. There are 3 ABC transporters, ABCC1, ABCA1, and    2. Carmeliet P, Jain RK. Angiogenesis in cancer and other diseases.
Nature 2000; 407: 249 – 257.
ABCA7, which might function as the S1P transporter.98–100    3. Saba JD, Hla T. Point-counterpoint of sphingosine 1-phosphate
It is still controversial whether these transporters function in metabolism. Circ Res 2004; 94: 724 – 734.
vivo to maintain vascular integrity. My group recently iden-    4. Balligand JL, Feron O, Dessy C. eNOS activation by physical forces:
tified a novel S1P transporter, Spinster2 (Spns2), by screen- From short-term regulation of contraction to chronic remodeling of
ing zebrafish mutants that show cardia bifida.101 The Spns cardiovascular tissues. Physiol Rev 2009; 89: 481 – 534.
   5. Tsutsui M, Shimokawa H, Otsuji Y, Ueta Y, Sasaguri Y, Yanagihara
family consists of Spns1, Spns2 and Spns3. The mutants N. Nitric oxide synthases and cardiovascular diseases: Insights from
showing cardia bifida harbored a R153S mutation of Spns2, genetically modified mice. Circ J 2009; 73: 986 – 993.
indicating that it was the gene responsible. Stainier’s group    6. Vanhoutte PM, Feletou M, Taddei S. Endothelium-dependent con-
previously reported that deletion of S1P2 causes cardia bifida tractions in hypertension. Br J Pharmacol 2005; 144: 449 – 458.
   7. Loirand G, Guerin P, Pacaud P. Rho kinases in cardiovascular physi-
in both zebrafish102 and another mutant fish. By analyzing ology and pathophysiology. Circ Res 2006; 98: 322 – 334.
the latter, they also demonstrated that the gene responsible    8. Higashi Y, Noma K, Yoshizumi M, Kihara Y. Endothelial function
for the phenotype was a mutation of Spns2.103 We assumed and oxidative stress in cardiovascular diseases. Circ J 2009; 73:
that Spns2 might function as a S1P transporter because it 411 – 418.
   9. Dejana E. Endothelial cell–cell junctions: Happy together. Nat Rev
appears to span the membrane 12 times according to its Mol Cell Biol 2004; 5: 261 – 270.
amino acid sequence. Therefore, we investigated the export   10. Mehta D, Malik AB. Signaling mechanisms regulating endothelial
activity of S1P using cells expressing wild-type Spns2 and permeability. Physiol Rev 2006; 86: 279 – 367.

Circulation Journal Vol.73, December 2009


2190 MOCHIZUKI N

  11. Chappell D, Jacob M, Paul O, Rehm M, Welsch U, Stoeckelhuber N-cadherins in human endothelial cells: VE-cadherin competes
M, et al. The glycocalyx of the human umbilical vein endothelial with N-cadherin for junctional localization. J Cell Biol 1998; 140:
cell: An impressive structure ex vivo but not in culture. Circ Res 1475 – 1484.
2009; 104: 1313 – 1317.   37. Luo Y, Radice GL. N-cadherin acts upstream of VE-cadherin in con-
  12. Davis GE, Senger DR. Endothelial extracellular matrix: Biosynthe- trolling vascular morphogenesis. J Cell Biol 2005; 169: 29 – 34.
sis, remodeling, and functions during vascular morphogenesis and   38. Lampugnani MG, Corada M, Caveda L, Breviario F, Ayalon O,
neovessel stabilization. Circ Res 2005; 97: 1093 – 1107. Geiger B, et al. The molecular organization of endothelial cell to cell
  13. Dejana E, Tournier-Lasserve E, Weinstein BM. The control of junctions: Differential association of plakoglobin, beta-catenin, and
vascular integrity by endothelial cell junctions: Molecular basis and alpha-catenin with vascular endothelial cadherin (VE-cadherin).
pathological implications. Dev Cell 2009; 16: 209 – 221. J Cell Biol 1995; 129: 203 – 217.
  14. Vestweber D. VE-cadherin: The major endothelial adhesion molecule   39. Herren B, Levkau B, Raines EW, Ross R. Cleavage of beta-catenin
controlling cellular junctions and blood vessel formation. Arterio- and plakoglobin and shedding of VE-cadherin during endothelial
scler Thromb Vasc Biol 2008; 28: 223 – 232. apoptosis: Evidence for a role for caspases and metalloproteinases.
  15. Newman PJ, Newman DK. Signal transduction pathways mediated Mol Biol Cell 1998; 9: 1589 – 1601.
by PECAM-1: New roles for an old molecule in platelet and vascular   40. Guaiquil V, Swendeman S, Yoshida T, Chavala S, Campochiaro PA,
cell biology. Arterioscler Thromb Vasc Biol 2003; 23: 953 – 964. Blobel CP. ADAM9 is involved in pathological retinal neovascular-
  16. Takai Y, Miyoshi J, Ikeda W, Ogita H. Nectins and nectin-like mole- ization. Mol Cell Biol 2009; 29: 2694 – 2703.
cules: Roles in contact inhibition of cell movement and proliferation.   41. Gavard J, Gutkind JS. VEGF controls endothelial-cell permeability
Nat Rev Mol Cell Biol 2008; 9: 603 – 615. by promoting the beta-arrestin-dependent endocytosis of VE-cad-
  17. Furuse M, Tsukita S. Claudins in occluding junctions of humans herin. Nat Cell Biol 2006; 8: 1223 – 1234.
and flies. Trends Cell Biol 2006; 16: 181 – 188.   42. Lambeng N, Wallez Y, Rampon C, Cand F, Christe G, Gulino-Debrac
  18. Weber C, Fraemohs L, Dejana E. The role of junctional adhesion D, et al. Vascular endothelial-cadherin tyrosine phosphorylation in
molecules in vascular inflammation. Nat Rev Immunol 2007; 7: angiogenic and quiescent adult tissues. Circ Res 2005; 96: 384 – 391.
467 – 477.   43. Potter MD, Barbero S, Cheresh DA. Tyrosine phosphorylation of
  19. Taddei A, Giampietro C, Conti A, Orsenigo F, Breviario F, Pirazzoli VE-cadherin prevents binding of p120- and beta-catenin and main-
V, et al. Endothelial adherens junctions control tight junctions by tains the cellular mesenchymal state. J Biol Chem 2005; 280: 31906 – 
VE-cadherin-mediated upregulation of claudin-5. Nat Cell Biol 2008; 31912.
10: 923 – 934.   44. Weis S, Shintani S, Weber A, Kirchmair R, Wood M, Cravens A, et
  20. Schnittler HJ. Structural and functional aspects of intercellular junc- al. Src blockade stabilizes a Flk/cadherin complex, reducing edema
tions in vascular endothelium. Basic Res Cardiol 1998; 93(Suppl 3): and tissue injury following myocardial infarction. J Clin Invest 2004;
30 – 39. 113: 885 – 894.
  21. Rao RM, Yang L, Garcia-Cardena G, Luscinskas FW. Endothelial-   45. Lampugnani MG, Orsenigo F, Gagliani MC, Tacchetti C, Dejana E.
dependent mechanisms of leukocyte recruitment to the vascular wall. Vascular endothelial cadherin controls VEGFR-2 internalization and
Circ Res 2007; 101: 234 – 247. signaling from intracellular compartments. J Cell Biol 2006; 174:
  22. Hsieh YH, Huang SS, Wei FC, Hung LM. Resveratrol attenuates 593 – 604.
ischemia–reperfusion-induced leukocyte–endothelial cell adhesive   46. Carmeliet P, Lampugnani MG, Moons L, Breviario F, Compernolle
interactions and prolongs allograft survival across the MHC barrier. V, Bono F, et al. Targeted deficiency or cytosolic truncation of the
Circ J 2007; 71: 423 – 428. VE-cadherin gene in mice impairs VEGF-mediated endothelial sur-
  23. Muller WA. Mechanisms of transendothelial migration of leukocytes. vival and angiogenesis. Cell 1999; 98: 147 – 157.
Circ Res 2009; 105: 223 – 230.   47. Gory S, Vernet M, Laurent M, Dejana E, Dalmon J, Huber P. The
  24. Turowski P, Martinelli R, Crawford R, Wateridge D, Papageorgiou vascular endothelial-cadherin promoter directs endothelial-specific
AP, Lampugnani MG, et al. Phosphorylation of vascular endothe- expression in transgenic mice. Blood 1999; 93: 184 – 192.
lial cadherin controls lymphocyte emigration. J Cell Sci 2008; 121:   48. Birdsey GM, Dryden NH, Amsellem V, Gebhardt F, Sahnan K,
29 – 37. Haskard DO, et al. Transcription factor Erg regulates angiogenesis
  25. Kuhn M. Structure, regulation, and function of mammalian mem- and endothelial apoptosis through VE-cadherin. Blood 2008; 111:
brane guanylyl cyclase receptors, with a focus on guanylyl cyclase-A. 3498 – 3506.
Circ Res 2003; 93: 700 – 709.   49. Le BA, Lionneton F, Mattot V, Lelievre E, Caetano B, Spruyt N, et
  26. Gardner DG, Chen S, Glenn DJ, Grigsby CL. Molecular biology of al. HIF-2alpha specifically activates the VE-cadherin promoter inde-
the natriuretic peptide system: Implications for physiology and hyper- pendently of hypoxia and in synergy with Ets-1 through two essen-
tension. Hypertension 2007; 49: 419 – 426. tial ETS-binding sites. Oncogene 2007; 26: 7480 – 7489.
  27. Keck PJ, Hauser SD, Krivi G, Sanzo K, Warren T, Feder J, et al.   50. Franco CA, Mericskay M, Parlakian A, Gary-Bobo G, Gao-Li J,
Vascular permeability factor, an endothelial cell mitogen related to Paulin D, et al. Serum response factor is required for sprouting angio-
PDGF. Science 1989; 246: 1309 – 1312. genesis and vascular integrity. Dev Cell 2008; 15: 448 – 461.
  28. Nambu H, Nambu R, Oshima Y, Hackett SF, Okoye G, Wiegand S,   51. Xiao K, Allison DF, Buckley KM, Kottke MD, Vincent PA, Faundez
et al. Angiopoietin 1 inhibits ocular neovascularization and break- V, et al. Cellular levels of p120 catenin function as a set point for
down of the blood–retinal barrier. Gene Ther 2004; 11: 865 – 873. cadherin expression levels in microvascular endothelial cells. J Cell
  29. Thurston G, Rudge JS, Ioffe E, Zhou H, Ross L, Croll SD, et al. Biol 2003; 163: 535 – 545.
Angiopoietin-1 protects the adult vasculature against plasma leakage.   52. Xiao K, Garner J, Buckley KM, Vincent PA, Chiasson CM, Dejana
Nat Med 2000; 6: 460 – 463. E, et al. p120-Catenin regulates clathrin-dependent endocytosis of
  30. Tiruppathi C, Minshall RD, Paria BC, Vogel SM, Malik AB. Role VE-cadherin. Mol Biol Cell 2005; 16: 5141 – 5151.
of Ca2+ signaling in the regulation of endothelial permeability. Vasc   53. Iyer S, Ferreri DM, DeCocco NC, Minnear FL, Vincent PA. VE-
Pharmacol 2002; 39: 173 – 185. cadherin-p120 interaction is required for maintenance of endo-
  31. Lum H, Malik AB. Regulation of vascular endothelial barrier func- thelial barrier function. Am J Physiol Lung Cell Mol Physiol 2004;
tion. Am J Physiol 1994; 267: L223 – L241. 286: L1143 – L1153.
  32. Moldobaeva A, Wagner EM. Heterogeneity of bronchial endothe-   54. Alcaide P, Newton G, Auerbach S, Sehrawat S, Mayadas TN, Golan
lial cell permeability. Am J Physiol Lung Cell Mol Physiol 2002; DE, et al. p120-Catenin regulates leukocyte transmigration through
283: L520 – L527. an effect on VE-cadherin phosphorylation. Blood 2008; 112: 2770 – 
  33. Adamson RH, Zeng M, Adamson GN, Lenz JF, Curry FE. PAF- and 2779.
bradykinin-induced hyperpermeability of rat venules is independent   55. Regan-Klapisz E, Krouwer V, Langelaar-Makkinje M, Nallan L,
of actin-myosin contraction. Am J Physiol Heart Circ Physiol 2003; Gelb M, Gerritsen H, et al. Golgiassociated cPLA2{alpha} regulates
285: H406 – H417. endothelial cell–cell junction integrity by controlling the trafficking
  34. van Nieuw Amerongen GP, Draijer R, Vermeer MA, van Hinsbergh of transmembrane junction proteins. Mol Biol Cell 2009 [Epub ahead
VW. Transient and prolonged increase in endothelial permeability of print].
induced by histamine and thrombin: Role of protein kinases, calcium,   56. Gates J, Peifer M. Can 1000 reviews be wrong? Actin, alpha-
and RhoA. Circ Res 1998; 83: 1115 – 1123. Catenin, and adherens junctions. Cell 2005; 123: 769 – 772.
  35. Sabatini PJ, Zhang M, Silverman-Gavrila R, Bendeck MP, Langille   57. Cattelino A, Liebner S, Gallini R, Zanetti A, Balconi G, Corsi A, et
BL. Homotypic and endothelial cell adhesions via N-cadherin deter- al. The conditional inactivation of the beta-catenin gene in endothe-
mine polarity and regulate migration of vascular smooth muscle lial cells causes a defective vascular pattern and increased vascular
cells. Circ Res 2008; 103: 405 – 412. fragility. J Cell Biol 2003; 162: 1111 – 1122.
  36. Navarro P, Ruco L, Dejana E. Differential localization of VE- and   58. Drees F, Pokutta S, Yamada S, Nelson WJ, Weis WI. Alpha-catenin

Circulation Journal Vol.73, December 2009


Stabilization of Endothelial Cell–Cell Contact (2008 Sato Award) 2191

is a molecular switch that binds E-cadherin-beta-catenin and regu- Kasminkas L, et al. Angiopoietin-1 is an antipermeability and anti-
lates actin-filament assembly. Cell 2005; 123: 903 – 915. inflammatory agent in vitro and targets cell junctions. Circ Res 2000;
  59. Yamada S, Pokutta S, Drees F, Weis WI, Nelson WJ. Deconstruct- 87: 603 – 607.
ing the cadherin–catenin–actin complex. Cell 2005; 123: 889 – 901.   82. Fukuhara S, Sako K, Minami T, Noda K, Kim HZ, Kodama T, et al.
  60. Ukropec JA, Hollinger MK, Salva SM, Woolkalis MJ. SHP2 asso- Differential function of Tie2 at cell–cell contacts and cell–substra-
ciation with VE-cadherin complexes in human endothelial cells is tum contacts regulated by angiopoietin-1. Nat Cell Biol 2008; 10:
regulated by thrombin. J Biol Chem 2000; 275: 5983 – 5986. 513 – 526.
  61. Nottebaum AF, Cagna G, Winderlich M, Gamp AC, Linnepe R,   83. Saharinen P, Eklund L, Miettinen J, Wirkkala R, Anisimov A,
Polaschegg C, et al. VE-PTP maintains the endothelial barrier via Winderlich M, et al. Angiopoietins assemble distinct Tie2 signalling
plakoglobin and becomes dissociated from VE-cadherin by leuko- complexes in endothelial cell–cell and cell-matrix contacts. Nat Cell
cytes and by VEGF. J Exp Med 2008; 205: 2929 – 2945. Biol 2008; 10: 527 – 537.
  62. Grazia LM, Zanetti A, Corada M, Takahashi T, Balconi G, Breviario   84. Winderlich M, Keller L, Cagna G, Broermann A, Kamenyeva O,
F, et al. Contact inhibition of VEGF-induced proliferation requires Kiefer F, et al. VE-PTP controls blood vessel development by bal-
vascular endothelial cadherin, beta-catenin, and the phosphatase ancing Tie-2 activity. J Cell Biol 2009; 185: 657 – 671.
DEP-1/CD148. J Cell Biol 2003; 161: 793 – 804.   85. Atkins GB, Jain MK. Role of Kruppel-like transcription factors in
  63. Fukata M, Kaibuchi K. Rho-family GTPases in cadherin-mediated endothelial biology. Circ Res 2007; 100: 1686 – 1695.
cell–cell adhesion. Nat Rev Mol Cell Biol 2001; 2: 887 – 897.   86. Kuo CT, Veselits ML, Barton KP, Lu MM, Clendenin C, Leiden JM.
  64. Dejana E, Orsenigo F, Lampugnani MG. The role of adherens junc- The LKLF transcription factor is required for normal tunica media
tions and VE-cadherin in the control of vascular permeability. J Cell formation and blood vessel stabilization during murine embryogene-
Sci 2008; 121: 2115 – 2122. sis. Genes Dev 1997; 11: 2996 – 3006.
  65. Kooistra MR, Dube N, Bos JL. Rap1: A key regulator in cell–cell   87. Sako K, Fukuhara S, Minami T, Hamakubo T, Song H, Kodama T, et
junction formation. J Cell Sci 2007; 120: 17 – 22. al. Angiopoietin-1 induces Kruppel-like factor 2 expression through
  66. Sakurai A, Fukuhara S, Yamagishi A, Sako K, Kamioka Y, Masuda a phosphoinositide 3-kinase/AKT-dependent activation of myocyte
M, et al. MAGI-1 is required for Rap1 activation upon cell–cell enhancer factor 2. J Biol Chem 2009; 284: 5592 – 5601.
contact and for enhancement of vascular endothelial cadherin-medi-   88. Yatomi Y, Ohmori T, Rile G, Kazama F, Okamoto H, Sano T, et al.
ated cell adhesion. Mol Biol Cell 2006; 17: 966 – 976. Sphingosine 1-phosphate as a major bioactive lysophospholipid that
  67. Fukuhara S, Sakurai A, Sano H, Yamagishi A, Somekawa S, is released from platelets and interacts with endothelial cells. Blood
Takakura N, et al. Cyclic AMP potentiates vascular endothelial 2000; 96: 3431 – 3438.
cadherin-mediated cell–cell contact to enhance endothelial barrier   89. Schaphorst KL, Chiang E, Jacobs KN, Zaiman A, Natarajan V,
function through an Epac-Rap1 signaling pathway. Mol Cell Biol Wigley F, et al. Role of sphingosine-1 phosphate in the enhancement
2005; 25: 136 – 146. of endothelial barrier integrity by platelet-released products. Am J
  68. Kooistra MR, Corada M, Dejana E, Bos JL. Epac1 regulates integ- Physiol Lung Cell Mol Physiol 2003; 285: L258 – L267.
rity of endothelial cell junctions through VE-cadherin. FEBS Lett   90. Anliker B, Chun J. Lysophospholipid G protein-coupled receptors.
2005; 579: 4966 – 4972. J Biol Chem 2004; 279: 20555 – 20558.
  69. Adamson RH, Ly JC, Sarai RK, Lenz JF, Altangerel A, Drenckhahn   91. Takuwa Y, Okamoto Y, Yoshioka K, Takuwa N. Sphingosine-1-
D, et al. Epac/Rap1 pathway regulates microvascular hyperperme- phosphate signaling and biological activities in the cardiovascular
ability induced by PAF in rat mesentery. Am J Physiol Heart Circ system. Biochim Biophys Acta 2008; 1781: 483 – 488.
Physiol 2008; 294: H1188 – H1196.   92. Garcia JG, Liu F, Verin AD, Birukova A, Dechert MA, Gerthoffer
  70. Wittchen ES, Worthylake RA, Kelly P, Casey PJ, Quilliam LA, WT, et al. Sphingosine 1-phosphate promotes endothelial cell barrier
Burridge K. Rap1 GTPase inhibits leukocyte transmigration by integrity by Edg-dependent cytoskeletal rearrangement. J Clin Invest
promoting endothelial barrier function. J Biol Chem 2005; 280: 2001; 108: 689 – 701.
11675 – 11682.   93. Dudek SM, Jacobson JR, Chiang ET, Birukov KG, Wang P, Zhan X,
  71. Liquori CL, Berg MJ, Siegel AM, Huang E, Zawistowski JS, Stoffer et al. Pulmonary endothelial cell barrier enhancement by sphingosine
T, et al. Mutations in a gene encoding a novel protein containing a 1-phosphate: Roles for cortactin and myosin light chain kinase.
phosphotyrosine-binding domain cause type 2 cerebral cavernous J Biol Chem 2004; 279: 24692 – 24700.
malformations. Am J Hum Genet 2003; 73: 1459 – 1464.   94. Lee JF, Zeng Q, Ozaki H, Wang L, Hand AR, Hla T, et al. Dual roles
  72. Bergametti F, Denier C, Labauge P, Arnoult M, Boetto S, Clanet M, of tight junction-associated protein, zonula occludens-1, in sphingo-
et al. Mutations within the programmed cell death 10 gene cause sine 1-phosphate-mediated endothelial chemotaxis and barrier integ-
cerebral cavernous malformations. Am J Hum Genet 2005; 76: 42 –  rity. J Biol Chem 2006; 281: 29190 – 29200.
51.   95. Sun X, Shikata Y, Wang L, Ohmori K, Watanabe N, Wada J, et al.
  73. Glading A, Han J, Stockton RA, Ginsberg MH. KRIT-1/CCM1 is a Enhanced interaction between focal adhesion and adherens junction
Rap1 effector that regulates endothelial cell cell junctions. J Cell proteins: Involvement in sphingosine 1-phosphate-induced endothe-
Biol 2007; 179: 247 – 254. lial barrier enhancement. Microvasc Res 2009; 77: 304 – 313.
  74. Whitehead KJ, Chan AC, Navankasattusas S, Koh W, London NR,   96. Camerer E, Regard JB, Cornelissen I, Srinivasan Y, Duong DN,
Ling J, et al. The cerebral cavernous malformation signaling pathway Palmer D, et al. Sphingosine-1-phosphate in the plasma compartment
promotes vascular integrity via Rho GTPases. Nat Med 2009; 15: regulates basal and inflammation-induced vascular leak in mice.
177 – 184. J Clin Invest 2009; 119: 1871 – 1879.
  75. Kleaveland B, Zheng X, Liu JJ, Blum Y, Tung JJ, Zou Z, et al.   97. Hla T, Venkataraman K, Michaud J. The vascular S1P gradient-
Regulation of cardiovascular development and integrity by the heart cellular sources and biological significance. Biochim Biophys Acta
of glass-cerebral cavernous malformation protein pathway. Nat Med 2008; 1781: 477 – 482.
2009; 15: 169 – 176.   98. Mitra P, Oskeritzian CA, Payne SG, Beaven MA, Milstien S, Spiegel
  76. Arthur WT, Quilliam LA, Cooper JA. Rap1 promotes cell spreading S. Role of ABCC1 in export of sphingosine-1-phosphate from mast
by localizing Rac guanine nucleotide exchange factors. J Cell Biol cells. Proc Natl Acad Sci USA 2006; 103: 16394 – 16399.
2004; 167: 111 – 122.   99. Sato K, Malchinkhuu E, Horiuchi Y, Mogi C, Tomura H, Tosaka M,
  77. Zhang Z, Rehmann H, Price LS, Riedl J, Bos JL. AF6 negatively et al. Critical role of ABCA1 transporter in sphingosine 1-phosphate
regulates Rap1-induced cell adhesion. J Biol Chem 2005; 280: release from astrocytes. J Neurochem 2007 [Epub ahead of print].
33200 – 33205. 100. Kobayashi N, Nishi T, Hirata T, Kihara A, Sano T, Igarashi Y, et al.
  78. Anastasiadis PZ, Moon SY, Thoreson MA, Mariner DJ, Crawford Sphingosine 1-phosphate is released from the cytosol of rat platelets
HC, Zheng Y, et al. Inhibition of RhoA by p120 catenin. Nat Cell in a carrier-mediated manner. J Lipid Res 2006; 47: 614 – 621.
Biol 2000; 2: 637 – 644. 101. Kawahara A, Nishi T, Hisano Y, Fukui H, Yamaguchi A, Mochizuki
  79. Wildenberg GA, Dohn MR, Carnahan RH, Davis MA, Lobdell NA, N. The sphingolipid transporter spns2 functions in migration of
Settleman J, et al. p120-catenin and p190RhoGAP regulate cell–cell zebrafish myocardial precursors. Science 2009; 323: 524 – 527.
adhesion by coordinating antagonism between Rac and Rho. Cell 102. Kupperman E, An S, Osborne N, Waldron S, Stainier DY. A sphin-
2006; 127: 1027 – 1039. gosine-1-phosphate receptor regulates cell migration during verte-
  80. Mitsuma W, Kodama M, Hirono S, Ito M, Ramadan MM, Tanaka brate heart development. Nature 2000; 406: 192 – 195.
K, et al. Angiopoietin-1, angiopoietin-2 and tie-2 in the coronary 103. Osborne N, Brand-Arzamendi K, Ober EA, Jin SW, Verkade H,
circulation of patients with and without coronary collateral vessels. Holtzman NG, et al. The spinster homolog, two of hearts, is required
Circ J 2007; 71: 343 – 347. for sphingosine 1-phosphate signaling in zebrafish. Curr Biol 2008;
  81. Gamble JR, Drew J, Trezise L, Underwood A, Parsons M, 18: 1882 – 1888.

Circulation Journal Vol.73, December 2009

You might also like