sphingosine 1-phosphate receptor regulation of n-cadherin...

13
Sphingosine 1-phosphate receptor regulation of N-cadherin mediates vascular stabilization Ji-Hye Paik, 1 Athanasia Skoura, 1 Sung-Suk Chae, 1 Ann E. Cowan, 2 David K. Han, 1 Richard L. Proia, 3 and Timothy Hla 1,4 1 Center for Vascular Biology, Department of Cell Biology, and 2 Center for Biomedical Imaging Technology, University of Connecticut Health Center, Farmington, Connecticut 06030-3501, USA; 3 Genetics of Disease and Development Branch, National Institutes of Diabetes, Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, Maryland 20892, USA Vascular stabilization, a process by which nascent vessels are invested with mural cells, is important in angiogenesis. Here we describe the molecular basis of vascular stabilization regulated by sphingosine 1-phosphate (S1P), a platelet-derived lipid mediator. S1P 1 receptor-dependent cell-surface trafficking and activation of the cell–cell adhesion molecule N-cadherin is essential for interactions between endothelial and mural cells. Endothelial cell S1P 1 /G i /Rac pathway induces microtubule polymerization, resulting in trafficking of N-cadherin to polarized plasma membrane domains. S1P treatment modulated the phosphorylation of N-cadherin as well as p120-catenin and induced the formation of cadherin/catenin/actin complexes containing novel regulatory and trafficking factors. The net result of endothelial cell S1P 1 receptor activation is the proper trafficking and strengthening of N-cadherin-dependent cell–cell adhesion with mural cells. Perturbation of N-cadherin expression with small interfering RNA profoundly attenuated vascular stabilization in vitro and in vivo. S1P-induced trafficking and activation of N-cadherin provides a novel mechanism for the stabilization of nascent blood vessels by mural cells and may be exploited to control angiogenesis and vascular diseases. [Keywords: sphingosine 1-phosphate; cadherin; angiogenesis; vascular stabilization; endothelial cells; pericytes] Supplemental material is available at http://www.genesdev.org. Received June 2, 2004; revised version accepted August 11, 2004. Vascular stabilization, which is defined as the invest- ment of mural cells (pericytes and vascular smooth muscle cells [VSMC]) to the endothelial cell (EC) layer of nascent vessels, is critical for vascular development, an- giogenesis, and the maintenance of the established vas- culature (Hirschi and D’Amore 1997; Jain 2003). Muta- tion of vascular regulatory genes (growth factors, recep- tors, signaling molecules) causes cardiovascular system failure and embryonic lethality, which are often due to abnormal mural cell investment of nascent vessels (Yan- copoulos et al. 2000). During the development of the retina, abnormal mural cell coverage of the nascent blood vessels leads to compensatory angiogenesis and dysfunctional retinal development (Risau 1997; Benja- min et al. 1998). In the adult, vasotoxic stresses (hypoxia, hyperglycemia) induce mural cell loss from established microvessels, leading to abnormal vascular permeability, uncontrolled angiogenesis, and ultimately tissue de- struction (Benjamin et al. 1998). This is exemplified prominently in diabetic retinopathy, in which angio- genic vessels fail to stabilize properly, eventually leading to blindness (Engerman 1989; Hammes et al. 2002). Moreover, tumor angiogenesis, a process critical for the growth of solid tumors, also requires vascular stabiliza- tion (Bergers et al. 2003). Inhibition of vascular stabili- zation leads to attenuated tumor angiogenesis and re- duced growth rate of tumors. Therefore, vascular stabi- lization (also known as maturation) is considered to be an important process in development, physiology, and pathology (Jain 2003). Despite the importance of this phenomenon, molecu- lar mechanisms involved are poorly understood. EC/mu- ral cell coculture studies have identified PDGF-BB and TGF- as important regulators of this process (Hirschi et al. 1998). However, our understanding of vascular stabi- lization at the cellular and molecular levels is limited. The S1P 1 receptor (also known as EDG-1), a G-protein- coupled receptor (GPCR) for the bioactive lipid S1P, was originally cloned from vascular EC as an inducible gene (Hla and Maciag 1990; Lee et al. 1998). Previous work has implicated this receptor as a major regulator of EC func- 4 Corresponding author. E-MAIL [email protected]; FAX (860) 679-1201. Article published online ahead of print. Article and publication date are at http://www.genesdev.org/cgi/doi/10.1101/gad.1227804. 2392 GENES & DEVELOPMENT 18:2392–2403 © 2004 by Cold Spring Harbor Laboratory Press ISSN 0890-9369/04; www.genesdev.org Cold Spring Harbor Laboratory Press on June 2, 2021 - Published by genesdev.cshlp.org Downloaded from

Upload: others

Post on 26-Jan-2021

1 views

Category:

Documents


0 download

TRANSCRIPT

  • Sphingosine 1-phosphate receptorregulation of N-cadherin mediatesvascular stabilizationJi-Hye Paik,1 Athanasia Skoura,1 Sung-Suk Chae,1 Ann E. Cowan,2 David K. Han,1 Richard L. Proia,3

    and Timothy Hla1,4

    1Center for Vascular Biology, Department of Cell Biology, and 2Center for Biomedical Imaging Technology, University ofConnecticut Health Center, Farmington, Connecticut 06030-3501, USA; 3Genetics of Disease and Development Branch,National Institutes of Diabetes, Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, Maryland20892, USA

    Vascular stabilization, a process by which nascent vessels are invested with mural cells, is important inangiogenesis. Here we describe the molecular basis of vascular stabilization regulated by sphingosine1-phosphate (S1P), a platelet-derived lipid mediator. S1P1 receptor-dependent cell-surface trafficking andactivation of the cell–cell adhesion molecule N-cadherin is essential for interactions between endothelial andmural cells. Endothelial cell S1P1/Gi/Rac pathway induces microtubule polymerization, resulting in traffickingof N-cadherin to polarized plasma membrane domains. S1P treatment modulated the phosphorylation ofN-cadherin as well as p120-catenin and induced the formation of cadherin/catenin/actin complexes containingnovel regulatory and trafficking factors. The net result of endothelial cell S1P1 receptor activation is theproper trafficking and strengthening of N-cadherin-dependent cell–cell adhesion with mural cells. Perturbationof N-cadherin expression with small interfering RNA profoundly attenuated vascular stabilization in vitro andin vivo. S1P-induced trafficking and activation of N-cadherin provides a novel mechanism for the stabilizationof nascent blood vessels by mural cells and may be exploited to control angiogenesis and vascular diseases.

    [Keywords: sphingosine 1-phosphate; cadherin; angiogenesis; vascular stabilization; endothelial cells; pericytes]

    Supplemental material is available at http://www.genesdev.org.

    Received June 2, 2004; revised version accepted August 11, 2004.

    Vascular stabilization, which is defined as the invest-ment of mural cells (pericytes and vascular smoothmuscle cells [VSMC]) to the endothelial cell (EC) layer ofnascent vessels, is critical for vascular development, an-giogenesis, and the maintenance of the established vas-culature (Hirschi and D’Amore 1997; Jain 2003). Muta-tion of vascular regulatory genes (growth factors, recep-tors, signaling molecules) causes cardiovascular systemfailure and embryonic lethality, which are often due toabnormal mural cell investment of nascent vessels (Yan-copoulos et al. 2000). During the development of theretina, abnormal mural cell coverage of the nascentblood vessels leads to compensatory angiogenesis anddysfunctional retinal development (Risau 1997; Benja-min et al. 1998). In the adult, vasotoxic stresses (hypoxia,hyperglycemia) induce mural cell loss from establishedmicrovessels, leading to abnormal vascular permeability,

    uncontrolled angiogenesis, and ultimately tissue de-struction (Benjamin et al. 1998). This is exemplifiedprominently in diabetic retinopathy, in which angio-genic vessels fail to stabilize properly, eventually leadingto blindness (Engerman 1989; Hammes et al. 2002).Moreover, tumor angiogenesis, a process critical for thegrowth of solid tumors, also requires vascular stabiliza-tion (Bergers et al. 2003). Inhibition of vascular stabili-zation leads to attenuated tumor angiogenesis and re-duced growth rate of tumors. Therefore, vascular stabi-lization (also known as maturation) is considered to bean important process in development, physiology, andpathology (Jain 2003).

    Despite the importance of this phenomenon, molecu-lar mechanisms involved are poorly understood. EC/mu-ral cell coculture studies have identified PDGF-BB andTGF-� as important regulators of this process (Hirschi etal. 1998). However, our understanding of vascular stabi-lization at the cellular and molecular levels is limited.

    The S1P1 receptor (also known as EDG-1), a G-protein-coupled receptor (GPCR) for the bioactive lipid S1P, wasoriginally cloned from vascular EC as an inducible gene(Hla and Maciag 1990; Lee et al. 1998). Previous work hasimplicated this receptor as a major regulator of EC func-

    4Corresponding author.E-MAIL [email protected]; FAX (860) 679-1201.Article published online ahead of print. Article and publication date areat http://www.genesdev.org/cgi/doi/10.1101/gad.1227804.

    2392 GENES & DEVELOPMENT 18:2392–2403 © 2004 by Cold Spring Harbor Laboratory Press ISSN 0890-9369/04; www.genesdev.org

    Cold Spring Harbor Laboratory Press on June 2, 2021 - Published by genesdev.cshlp.orgDownloaded from

    http://genesdev.cshlp.org/http://www.cshlpress.com

  • tion in vitro, including the regulation of cell survival,migration, and morphogenesis (Lee et al. 1999, 2001). Incultured EC, S1P interaction with its receptors S1P1 andS1P3 induced the small GTPase Rho- and Rac-dependentassembly of VE-cadherin-based adherens junctions (Leeet al. 1999). Global deletion of the S1p1 gene results in afully penetrant embryonic lethality at embryonic days12.5–14.5 (E12.5–E14.5) of gestation. The phenotype ofthe animals included aberrant mural cell ensheathmentof the dorsal aorta and other vessels (Liu et al. 2000). Inaddition, tissue-specific knock-out of S1P1 in endothelialcells phenocopies the global knock-out phenotype,whereas VSMC-specific knock-out did not induce em-bryonic lethality (Allende et al. 2003). These data indi-cate that signaling of S1P1 in the endothelial cells regu-lates the mural cell coverage of nascent vessels in transand that signaling in mural cells is dispensable for de-velopmental vascular stabilization.

    In the present report, we provide a mechanistic basisfor S1P1-induced vascular maturation.

    Results

    S1P1 receptor is required for EC–mural cell interaction

    The phenotype of the S1P1 knock-out mice suggests thatEC interaction with mural cells may be deficient (Liu etal. 2000). We therefore tested whether S1P would modu-late EC–mural cell interactions in an in vitro coculturesystem (Hirschi et al. 1998). First, we isolated and estab-lished murine embryonic EC (MEEC) lines from S1p1

    −/−,S1p1

    +/−, and S1p1+/+ mouse embryos as described (Fusco

    et al. 1988; Balconi et al. 2000). When MEEC were co-cultured with fluorescently labeled 10T1/2 cells (a mes-enchymal cell line with pericyte properties), S1P induced

    (three- to fourfold after 0.5 h) 10T1/2 cell binding towild-type and heterozygous but not with S1p1

    −/− MEEC(Fig. 1a,c). Fluorescence micrographs show that S1Ptreatment induced 10T1/2 cell interaction with wild-type MEEC via podlike processes (Fig. 1a,b). Morphologi-cally, this resembles pericytes interacting with EC inpostcapillary venules (Allt and Lawrenson 2001). Thiseffect was specific, as related lipids that are not agonistsfor the S1P1 receptor failed to induce this interaction(Fig. 1d). Cell–cell interaction between S1p1

    −/− MEECand 10T1/2 cells was restored upon expression of theS1P1 receptor in S1p1

    −/− MEEC (Fig. 1e). These data sug-gest that S1P interaction with the S1P1 receptor on en-dothelial cells is critical for interaction with 10T1/2 cells.

    We also tested the effect of other regulators of vascularmaturation such as PDGF-BB, angiopoietins 1 and 2, andVEGF. Only S1P was effective in this cell–cell interac-tion assay, suggesting that it acts in a specific manner toinduce cell–cell interaction between EC and mural cells(Supplementary Fig. 1a). In addition, several additionallines of VSMC were tested to rule out the possibility thatthis effect is a peculiarity of the 10T1/2 cells. Two dif-ferent VSMCs with distinctive phenotypes, specificallythat from adult aorta and embryonic aorta, responded toS1P-induced adhesion to wild-type but not to S1p1

    −/−

    MEEC (Supplementary Fig. 1b). The adult VSMC and10T1/2 cells do not express detectable levels of the S1P1receptor whereas the embryonic VSMC express this re-ceptor constitutively (Kluk and Hla 2001). Likewise, hu-man umbilical vein endothelial cells (HUVEC) also be-haved similarly to the wild-type MEEC. These data sug-gest that S1P interaction with S1P1 receptor on ECregulates EC/mural cell interaction in trans, regardlessof the status of this receptor on mural cells and supportthat the in vitro cell–cell interaction system faithfullyrecapitulates an aspect of vascular maturation.

    Figure 1. Regulation of EC/mural cell interaction by theS1P1 receptor. (a) Regulation of cell–cell interaction be-tween MEEC and 10T1/2 cells by S1P1. Vybrant-labeled10T1/2 cells were incubated with adherent MEECs (ei-ther +/+ or −/− S1P1) in the presence or absence of S1P.Cocultures were fixed and stained with VE-cadherin (red)and imaged. Bar, 40 µm. (b) Three-dimensional recon-struction of z-series with IMARIS software of adherentwild-type MEEC cocultured with 10T1/2 cells in thepresence of S1P. Note the formation of extended cellularprocesses on 10T1/2 (green) that make contacts withMEEC (red). (c) Quantitation of cell–cell interaction be-tween 10T1/2 cells and MEEC of different genotypes(n = 7; *, P < 0.01). (d) Effect of different lipids (100 nM)on MEEC and 10T1/2 interaction. (Sph) Sphingosine;(LPA) lysophosphatidic acid; (DHS) dihydrosphingosine;(C1P) ceramide-1-phosphate; (Cer) ceramide. Results rep-resent mean ± S.E. of triplicate determinations from arepresentative experiment that was repeated three times(one-way ANOVA [P < 0.01], Bonferroni’s multiple com-parison posttest; **, P < 0.005). (e) Rescue of cell–cell in-

    teraction by the expression of S1P1 receptor. S1p1−/− MEEC were transduced with either �-gal or S1P1 adenovirus and interaction with

    10T1/2 cells was conducted as described. Inset shows the immunoblot analysis of S1P1 of adenoviral-transduced MEEC (n = 5, *,P < 0.05).

    Mechanism of vascular maturation

    GENES & DEVELOPMENT 2393

    Cold Spring Harbor Laboratory Press on June 2, 2021 - Published by genesdev.cshlp.orgDownloaded from

    http://genesdev.cshlp.org/http://www.cshlpress.com

  • N-cadherin is necessary for S1P1 receptor-inducedEC–mural cell interaction

    N-cadherin, which is expressed in both EC and muralcells, was proposed to be important for the interactionsbetween these two cell types (Dejana 2004). Specifically,injection of anti-N-cadherin antibodies into the ven-tricles of chick embryonic brain induced vascular defects(Gerhardt et al. 2000). We utilized the small interferingRNA (siRNA) approach to inhibit N-cadherin expressionin EC to determine the requirement for this adhesionmolecule in S1P-induced cell–cell interaction. As shownin Figure 2a, the siRNA for N-cadherin but not thescrambled counterpart potently blocked its expression inEC. Treatment of EC with the N-cadherin siRNA butnot the scrambled siRNA nor the irrelevant E-cadherinsiRNA inhibited the interaction between EC and 10T1/2cells (Fig. 2b). Additional experiments were done to con-firm the specificity of the siRNA for N-cadherin. First,chicken N-cadherin or N-cadherin-GFP, which have dis-tinct sequences at the siRNA-binding site, was not si-lenced (Supplementary Fig. 2). When chicken N-cad-herin-GFP was transfected into endothelial cells, its ex-pression and function (as assayed by N-cadherin beadbinding; see below) was not suppressed by the siRNA forN-cadherin. In contrast, murine N-cadherin-GFP trans-fection was fully suppressed in a sequence-specific man-ner (Supplementary Fig. 3). S1P-induced EC and 10T1/2adhesion was strongly inhibited by chelation of extracel-lular calcium but not by the inhibitors of integrins suchas the RGD peptide or echistatin (Fig. 2b). This confirmsthe requirement for calcium-dependent cell–cell adhe-

    sion. On the other hand, integrin-dependent mecha-nisms appear to be dispensable.

    Immunofluorescence of endogenous N-cadherin in ECshows that it is excluded from endothelial intercellularjunctions and that it is diffusely localized in vesicle-likestructures in the cytoplasm. In addition, when N-cad-herin-GFP-expressing HUVEC were cocultured with10T1/2 cells in the presence of S1P, N-cadherin-GFP wasconcentrated at filopodia-like structures from endothe-lial cells that dynamically attach to 10T1/2 cells (Supple-mentary Movie 1). When N-cadherin-GFP was expressedin 10T1/2 cells, the arrangement of N-cadherin-contain-ing structures was much less dynamic, although it waspresent in the intercellular contact sites (SupplementaryMovie 2). These observations suggest that S1P signalingvia the S1P1 receptor in EC induced N-cadherin-depen-dent adhesive contacts between EC and mural cells.

    Trafficking of N-cadherin to polarized microdomainson EC cell surface induced by S1P1/Racsignaling pathway

    Mechanisms involved in S1P1 regulation of N-cadherinlocalization and function were further investigated. Weused latex beads coated with N-cadherin ectodomain-Fcchimera (NcadFc) to measure N-cadherin-dependent ad-hesion in EC (Lambert et al. 2000). Binding of NcadFc-coated beads to EC increased markedly upon S1P addi-tion (Fig. 3a,b). In contrast, S1P treatment did not alterthe binding of beads coated with CD31/PECAM anti-body or a control mouse IgG to EC. These data indicatethat N-cadherin is translocated to the cell surface uponactivation of the S1P1 receptor and suggest that the cell-surface-localized N-cadherin is active in binding to otherN-cadherin molecules in trans.

    To further demonstrate the cell-surface trafficking ofN-cadherin upon S1P treatment, a cell-surface biotinyla-tion experiment was conducted, followed by avidin af-finity selection and immunoblot analysis. Significantlymore N-cadherin (but not CD31 or VE-cadherin) was bio-tinylated on the cell surface after S1P treatment of EC(Fig. 3c), suggesting that S1P induces the trafficking ofendogenous N-cadherin to the EC surface.

    Cadherin-dependent junction formation was shown tobe regulated by RhoGTPases (Dejana 2004). In HUVEC,we showed that S1P treatment induced the activity ofboth RhoA and Rac1 GTPases (Paik et al. 2001). Simi-larly, in wild-type MEEC, RhoA and Rac1 were rapidlyactivated after S1P addition. S1p1

    −/− MEEC, however, ex-hibited only RhoA activation (Supplementary Fig. 4),suggesting that S1P1 receptor is important in the activa-tion of the Rac1 GTPase.

    We further investigated the role of RhoGTPases in N-cadherin dependent adhesion. S1P-induced NcadFc beadbinding to EC was potently inhibited by dominant nega-tive Rac (N17Rac) but not by dominant negative Rho(N19Rho) (Fig. 3d). This finding was confirmed by down-regulating Rac1 expression. siRNA for Rac1 potently si-lenced Rac1 protein expression (Supplementary Fig. 5).Interestingly, diminution in Rac1 levels led to the inhi-

    Figure 2. S1P-induced EC/mural cell interaction requires N-cadherin. (a) Silencing of N-cadherin by siRNA. The siRNA forN-cadherin was chosen in a conserved region from murine andhuman sequences. Human umbilical vein endothelial cells(HUVEC) or MEEC were treated with siRNAs ([NCDi] N-cad-herin siRNA; [Scr] scrambled siRNA), cell extracts were pre-pared and probed for the expression of N-cadherin or actin in animmunoblot assay. (b) Requirement for N-cadherin for EC/mu-ral cell interaction. Wild-type MEEC and 10T1/2 cells were as-sayed in the cell–cell adhesion assay as described. MEEC werepretreated with siRNA for N-cadherin (NCDi) or scrambled(Scr), 48 h prior to the initiation of the assay or were incubatedwith EGTA, Echistatin, or RGD during the assay. Results rep-resent mean ± S.E. of triplicate determinations from a represen-tative experiment that was repeated three times (one-wayANOVA [P < 0.01], Bonferroni’s multiple comparison posttest;**, P < 0.005).

    Paik et al.

    2394 GENES & DEVELOPMENT

    Cold Spring Harbor Laboratory Press on June 2, 2021 - Published by genesdev.cshlp.orgDownloaded from

    http://genesdev.cshlp.org/http://www.cshlpress.com

  • bition of S1P-induced membrane ruffles and increasedstress fiber formation (Supplementary Fig. 6). Concomi-tantly, S1P-induced NcadFc-bead binding to the cell sur-face was inhibited (Supplementary Fig. 7). These dataindicate that Rac activation by S1P1 is essential. More-over, time lapse microscopic monitoring of N-cadherin-GFP showed that N-cadherin-GFP was dynamicallytranslocated to plasma membrane domains in a polarizedmanner. This was potently inhibited by N17Rac but notby N19Rho (Supplementary Movies 3–6).

    N-cadherin trafficking to the plasma membrane wasfurther investigated in EC treated or not with S1P usingconfocal immunofluorescence imaging of fixed cells. Asshown in Figure 4b, endogenously expressed N-cadherinwas dramatically redistributed into polarized microdo-mains on the EC plasma membrane. This localizationcorrelated with the asymmetric nature of NcadFc-beadbinding to the EC surface upon S1P stimulation (Fig. 3a,blue arrowheads), suggesting that N-cadherin on suchdomains is active. Optical sectioning and reconstructionof the z-axis of EC stained with N-cadherin antibodyclearly showed a polarized distribution of N-cadherin onone side of the apical cell surface (Fig. 4b). This findingsuggests the existence of specific domains containing N-cadherin on the EC surface.

    Microtubule-dependent trafficking of N-cadherinto polarized microdomains on the endothelialcell surface

    We further investigated the mechanisms involved inS1P1-induced N-cadherin translocation to the endothe-lial cell surface microdomains. Recently, trafficking ofN-cadherin–kinesin complex along the microtubule(MT) cytoskeleton has been reported (Mary et al. 2002;Chen et al. 2003). Therefore, we investigated whetherS1P regulated MT polymerization in EC. MT growth inEC was monitored by visualizing the incorporation ofthe EB1-GFP protein (a specific marker for growing tips)onto growing ends of MT (Piehl et al. 2004). S1P-stimu-lated HUVEC displayed marked increases in the fre-quency and persistency of MT growth (SupplementaryMovies 7, 8). S1P-induced increases in MT polymeriza-tion and protrusion of MT growth to the cell peripherywere strongly inhibited by the dominant negative mu-tant of the small GTPase Rac (N17Rac) (SupplementaryMovie 9). As expected, nocodazole treatment completelyinhibited MT polymerization in EC (SupplementaryMovie 10). These observations suggest that the activa-tion of the GTPase Rac by S1P is required for efficientMT polymerization in EC.

    Immunofluorescence imaging of endogenous N-cad-herin in EC shows that nocodazole treatment blockedpolarized cell-surface translocation (Fig. 4c) suggestingthat S1P-induced MT polymerization is necessary for N-cadherin trafficking to the cell surface. In addition N-cadherin-bead binding to the EC cell surface was com-pletely inhibited by nocodazole in a time-dependentmanner (Fig. 5a).

    The involvement of the actin cytoskeleton was alsoinvestigated, as it is known that the S1P1/Gi/Rac path-

    Figure 4. S1P induces polarized distribution of N-cadherin onthe apical endothelial cell surface. HUVEC were immunos-tained for endogenous N-cadherin, and optical sections of z-axisof cells were reconstructed as described. (a–d) XY view and z-stack view (boxes below) of pointed lines (blue arrowheads).N-cadherin is shown in green, nuclei are in blue (DAPI), andplasma membrane in red (DiI-C18). (b) Note that S1P stimula-tion dramatically induced polarized N-cadherin distribution.This was attenuated by nocodazole treatment (2 µM, 1 h, c) butnot by cytochalasin D (1 µM, 10 min, d).

    Figure 3. S1P induces Rac1-dependent binding of NcadFcbeads to EC. (a) Binding of NcadFc-beads to HUVEC surface inthe absence or presence of S1P stimulation. Plasma membraneswere labeled in red (DiI-C18) and N-cadherin and NcadFc-beadswere stained in green (NCD). Note that blue arrows indicatepolarized binding of NcadFc-beads to EC surface. Pink arrowspoint to dorsal ruffles that are devoid of NcadFc-bead binding.Bar, 20 µm. (b) Beads coated with irrelevant mouse IgG (IgG),CD31-IgG, or NcadFc were used in the binding assay with S1P-treated HUVEC. The number of beads bound to the cell surfacewas quantified as described (**, P < 0.001). (c) HUVEC weretreated or not with S1P, and cell-surface proteins (N-cad, CD31,VE-cad) were biotinylated and recovered by affinity isolationwith Streptavidin-Sepharose. Immunoblot analysis with spe-cific antibodies ([NB] nonbiotinylated; [WCL] whole-cell lysate)is shown. (d) Binding of NcadFc-beads to S1P-treated HUVECexpressing N17Rac or N19Rho or vector (pTet) was quantifiedas above (**, P < 0.001).

    Mechanism of vascular maturation

    GENES & DEVELOPMENT 2395

    Cold Spring Harbor Laboratory Press on June 2, 2021 - Published by genesdev.cshlp.orgDownloaded from

    http://genesdev.cshlp.org/http://www.cshlpress.com

  • way induces the assembly of the cortical actin network(Lee et al. 1999; Paik et al. 2001). Depolymerization ofactin filaments by cytochalasin D significantly reducedNcadFc-bead binding to the EC cell surface, suggestingthe critical role of actin polymerization in supportingN-cadherin-dependent adhesion (Fig. 5a). However, S1P-induced N-cadherin trafficking to polarized plasmamembrane domains was not inhibited by cytochalasinD, even though the morphology of the EC was altered(Fig. 4d). These data suggest that the actin cytoskeletonis not involved in the trafficking of N-cadherin after S1Ptreatment, even though ultimate binding of N-cadherinmolecules requires the formation of F-actin.

    When EC were treated with nocodazole or cytochala-sin D for the duration of the assay, interaction of EC and10T1/2 cells was significantly inhibited (Fig. 5b). Asshown in Supplementary Movie 10, acute treatment (10min) of EC with nocodazole efficiently blocked MTpolymerization. However, 50 min after S1P treatment,acute nocodazole treatment did not appreciably inhibit

    EC–10T1/2 interactions or NcadFc-bead binding, sug-gesting that once N-cadherin trafficking is set into mo-tion and sufficient N-cadherin accumulates on theplasma membrane, inhibition of the MT cytoskeleton isunable to block the junction assembly between EC and10T1/2 (Fig. 5a,b).

    S1P1-induced N-cadherin activation involves complexformation and phosphorylation

    Cadherin-dependent stable cell–cell interaction requiresthe formation of protein complexes, consisting of cad-herin, �-catenin, �-catenin, actin filaments, and otherregulatory factors (Dejana 2004). S1P induced the forma-tion of N-cadherin/�-catenin/�-catenin complexes inwild-type MEEC (Fig. 6a). This response was specificallyinduced by S1P1 as S1p1

    −/− MEEC or adenoviral-medi-ated expression of S1P2 or S1P3 in N-cadherin-expressingCHO cells did not show increases in cadherin/catenincomplex formation upon S1P stimulation (data notshown).

    To further define the molecular basis of localizationand function of N-cadherin, we identified protein com-plexes associated with N-cadherin in S1P-treated EC byimmunoprecipitation of N-cadherin and LC/MS-MSanalysis of associated polypeptides. As expected, �-�-p120-catenin and actin were found in the complex (Fig.6a,b). Additional proteins were also identified in the N-cadherin complex as detailed below. N-cadherin bindingwith MT-associated protein (MAP)4 may suggest a mo-lecular basis for MT-dependent N-cadherin trafficking.Interaction with sorting nexin9 and COPII-like proteinsmay indicate a novel mechanism of N-cadherin traffick-ing via vesicular machinery. RACK1 has previously beenshown to associate with protein tyrosine phosphatase,which modulates cadherin functions (Hellberg et al.2002) and thus suggests a regulatory mechanism via ty-rosine phosphorylation. Indeed, we observed changes intyrosine phosphorylation of N-cadherin/p120-catenin byS1P stimulation (Fig. 6c). Specific interactions betweenN-cadherin and interacting proteins were further verifiedby coimmunoprecipitation in the presence of cytochala-

    Figure 5. N-cadherin-mediated adhesion between EC/muralcells requires S1P-induced MT polymerization and actin cyto-skeleton. (a) Binding of NcadFc-beads in the presence of noco-dazole (noc, 2 and 10 µM, 1 h and 10 min, respectively), cyto-chalasinD (cytD, 1 µM, 10 min), and S1P. Bead binding wasquantified as described and expressed as fold (±S.D.) control. (b)10T1/2 cell adhesion to MEEC in the presence of S1P and no-codazole or cytochalasinD as in a. (c) After 1 h stimulation withS1P, HUVEC were washed and NcadFc-beads were placed overthe cells in the presence of nocodazole (2 µM for 1 h) alone(noc-S1P) or with S1P (noc + S1P). Number of bound beads wasexpressed fold (±S.D.) over number of beads bound to control(noc-S1P) cells.

    Figure 6. S1P regulation of N-cadherin multi-protein complex in EC. (a) MEECs were treatedwith S1P for various times, cell extracts wereprepared and immunoprecipitated with �-catenin. Immunoprecipitates were immunoblot-ted with antibodies for �- and �-catenin, and N-cadherin (NCD). Total amount of N-cadherinand �-catenin were also assessed in the extracts.(b) Identification of the components of multipro-tein complexes of N-cadherin in HUVEC. Large-scale immunoprecipitation of N-cadherin wasconducted from S1P-treated HUVEC extracts andanalyzed by SDS-PAGE and Coomassie blue staining. Specific regions of the gel were cut, and protein identities were determined bymass spectrometry. (MAP) Microtubule-associated protein; (TSP) thrombospondin; (MVP) major vault protein. (c) S1P-stimulated (1 h)HUVEC lysates were immunoprecipitated for p120 catenin (p120) or NCD and blotted for phosphotyrosine (pY) or phosphoserine/threonine (pST). Numbers are expressed as fold increases over nonstimulated conditions after normalization with the loading control(N = 3).

    Paik et al.

    2396 GENES & DEVELOPMENT

    Cold Spring Harbor Laboratory Press on June 2, 2021 - Published by genesdev.cshlp.orgDownloaded from

    http://genesdev.cshlp.org/http://www.cshlpress.com

  • sin D and nocodazole. Binding of N-cadherin withcortactin was dependent on actin cytoskeleton as it wasnot observed in the presence of cytochalasin D. How-ever, coimmunoprecipitation of RACK1 with N-cad-herin was observed even in the presence of cytochalasinD (data not shown). The significance of other coprecipi-tated proteins, such as major vault protein (MVP), leu-kophysin, thrombospondin (TSP1), and the translationfactor Tiar1 is not known at present. These data suggestthe complexity in the regulation of N-cadherin traffick-ing and adhesiveness in EC by multiple protein–proteininteractions.

    Previously cadherin/catenin phosphorylation eventswere shown to modulate the strength of cadherin-depen-dent junctions (Gottardi and Gumbiner 2001). As shownin Figure 5c, S1P stimulation potentiates the binding ofNcadFc-beads to EC even in the presence of nocodazole,suggesting there are additional activation steps inducedby S1P1 in addition to the trafficking to the plasma mem-brane. Therefore we explored the hypothesis that S1Pstimulation might alter the phosphorylation of proteinsin the cadherin complex and thereby alter its adhesive-ness. Phosphorylation of the N-cadherin-binding proteinp120-catenin (especially the p100 isoform) was modu-lated by S1P treatment of HUVEC. Specifically, tyrosinephosphorylation was enhanced whereas serine/threo-nine phosphorylation was attenuated (Fig. 6c). In addi-tion, tyrosine-phosphorylated N-cadherin was also in-creased by S1P treatment. However, �-catenin phos-phorylation was not altered significantly (data notshown). These data suggest that alterations in p120-catenin and N-cadherin phosphorylation are likely tocontribute to the changes in S1P-induced N-cadherin ad-hesivity.

    Aberrant localization of N-cadherin in S1p1−/− mice

    To obtain in vivo correlates of our findings, we sought todetermine if N-cadherin localization in the vascular sys-tem is altered in the S1p1

    −/− mouse embryo. Sections ofthe dorsal aorta were imaged by confocal immunofluo-rescence microscopy. As expected, defective mural cellcoverage of the dorsal aorta was observed in S1p1

    −/− em-bryos (Liu et al. 2000). N-cadherin is expressed in EC,mural cells, and surrounding mesenchymal cells in wild-type embryos (Fig. 7). Aberrant N-cadherin localizationwas observed in S1p1

    −/− embryos, compared to wild-typecounterparts (Fig. 7a–d). Specifically, N-cadherin local-ization in the intima is shifted from the basal membraneto a more lateral pattern in EC. A significant decrease inthe ratio of fluorescence intensity at EC/mural cellboundaries versus that at EC/EC boundaries was ob-served in S1p1

    −/− embryos when compared to wild-typecounterparts (Fig. 7e). This mislocalization of N-cad-herin was also observed in S1p1

    −/− MEEC in vitro, inwhich the N-cadherin signal was stronger at EC/ECjunctions compared to that of wild-type MEEC (Fig. 7f).Together these data support the notion that S1P1 signal-ing in EC is essential to induce the formation of N-cad-

    herin-based junctions between EC and VSMC in vivo.This model is shown schematically in Figure 7g.

    Silencing of S1P1 or N-cadherin with siRNAsuppresses neovessel stabilization

    Based on the hypothesis that N-cadherin function iscritical for vessel stability, we explored whether suppres-sion of its expression blocks the formation of maturevessels. We used the siRNA technology (Elbashir et al.2001) to determine the requirement for S1P1/N-cadherinpathway.

    The role of S1P1/N-cadherin in neovessel formationand stability was tested in the rat aortic ring angiogen-esis assay (Nicosia and Ottinetti 1990). Ex vivo treat-ment with VEGF and S1P induced new vessel sproutsthat are invested with �-smooth muscle actin positivecells (Fig. 8a–c). These cells also expressed desmin, amarker for mural cells (data not shown). Treatment withN-cadherin siRNA strongly suppressed the expression ofN-cadherin but not the related VE-cadherin protein (Fig.8d). Inhibition of N-cadherin expression profoundly sup-pressed neovessel maturation without inhibiting vesselsprouting or proliferation of cells of the vessel wall (Fig.8a–c).

    Figure 7. Mislocalization of N-cadherin in dorsal aorta ofS1p1

    −/− mouse embryos. E12.5 embryos (wild-type in b,d;S1p1

    −/− in a,c) were sectioned (caudal–rostral) and EC were vi-sualized by immunofluorescence confocal microscopy with PE-CAM-1 (red, a–d). (a,b) Similarly, mural cells were visualized by�-smooth muscle actin (green) staining. Note that aberrant mu-ral cell coverage is apparent in S1p1

    −/− dorsal aorta. (c,d) N-cadherin-expressing cells were also visualized (green). Enhancedaccumulation of N-cadherin at EC/EC contact sites comparedwith EC/VSMC contacts was observed in S1p1

    −/− mice (indi-cated by arrowheads; boxed regions are shown as higher-mag-nification images in the right panel). Note that only the areasthat possess VSMC underneath the EC are imaged for N-cad-herin analysis. Bars: b, 50 µm; d, 20 µm. (e) Ratio of the fluo-rescence intensity of N-cadherin staining on EC/pericyte junc-tions (EP) to EC/EC (EE). Data represent mean ± S.D. of mea-surements from 10 cells per section from 14 independentsections derived from 14 embryos. (f) Differential localization ofN-cadherin in wild-type and S1p1

    −/− MEEC cells in culture.Note more EC/EC junctional N-cadherin staining (arrowheads)in S1p1

    −/− MEEC. Bar, 5 µm. (g) Schematic representation ofaberrant N-cadherin localization on S1p1

    −/− vasculature.

    Mechanism of vascular maturation

    GENES & DEVELOPMENT 2397

    Cold Spring Harbor Laboratory Press on June 2, 2021 - Published by genesdev.cshlp.orgDownloaded from

    http://genesdev.cshlp.org/http://www.cshlpress.com

  • To test the in vivo relevance of our mechanistic find-ings, we used the subcutaneous implants of Matrigelsupplemented with growth factors in the mouse as an invivo model (Lee et al. 1999; Ancellin et al. 2002). Weshowed previously that the formation of mature neoves-sels in the subcutaneous implants of Matrigel plugs re-quired S1P signaling via the S1P1 receptor. Inclusion ofsiRNA for S1P1 but not the scrambled counterpart in theMatrigel plug markedly reduced transcript expression invivo in cells that migrated into the Matrigel (Fig. 9a). Asexpected, neovessel formation was strongly suppressedby S1P1 siRNA (Fig. 9b, E1i). The siRNA for N-cadherinpotently down-regulated the expression of the corre-sponding protein in vivo, as determined by immunoblotanalysis of Matrigel plug extracts (Fig. 9c).

    Immunohistochemical analysis showed that siRNAfor N-cadherin did not suppress neovessel formation;rather it profoundly inhibited the acquisition of�-smooth muscle actin positive mural cells (Fig. 9b,NCDi). Immunoblot analysis showed the decrease in thetotal amount of �-smooth muscle actin in the Matrigelplugs. Structural alterations of neovessels and apparentextravasation of blood cells were observed in N-cadherinsiRNA-treated Matrigel sections. Endothelial cells ofnascent vessel were disorganized, phenocopying theS1p1

    −/− embryonic vasculature (Fig. 9b). These observa-tions further confirm that S1P1 expression is critical forneovessel formation and pericyte ensheathment. Impor-tantly, N-cadherin expression is required for vessel sta-bilization in vivo.

    Discussion

    This work defines the mechanism by which S1P1 recep-tor regulates vascular stabilization. Isolated EC from

    S1p1−/− mice failed to interact with pericyte-like 10T1/2

    cells even after treatment with S1P. The coculture sys-tem, which recapitulates the in vivo phenotype allowedthe deconvolution of molecular mechanisms. The effectof S1P in the coculture assay was specific, as PDGF,VEGF, and angiopoietin 1 and 2 were inactive, consistentwith the fact that these factors signal via a different classof receptors. Indeed, knock-out mice for these factorsexhibit defects in vascular stabilization distinct fromthat of the S1p1

    −/− mice; for example, PDGF-receptor-�knock-out mice showed reduced numbers of pericytes inbrain microvessels, which contrasts sharply with theS1p1

    −/− mice (Hellstrom et al. 1999, 2001).In the present study, we show that S1P interaction

    with S1P1 receptor in EC is critical for the regulatedinteraction between EC and mural cells. We observedthat S1P-induced EC–mural cell interaction in vitro isnot influenced by the expression level of S1P1 in muralcells but is absolutely dependent on endothelial S1P1.This agrees with the data from EC-specific deletion ofS1P1, which phenocopies the global S1P1 knock-outmice, whereas a vascular smooth-muscle-specific knock-out remains viable throughout development (Allende etal. 2003). These observations strongly argue for S1P1-dependent EC signaling in trans to the mural cells duringvascular stabilization.

    A salient finding of this work is that N-cadherin is acritical mediator of cell–cell adhesion between EC andmural cells. N-cadherin, a widely expressed cell–cell ad-hesion molecule, was previously implicated in EC–mu-ral cell interaction. (Hellstrom et al. 1999, 2001). How-ever, mechanisms by which N-cadherin regulate vascu-lar stabilization are not well understood. In the presentstudy, we show that activation of N-cadherin by theS1P1-mediated Gi/Rac1 signaling pathway induces MT

    Figure 8. Inhibition of vascular sprout maturationby N-cadherin RNAi. (a) Rat aortic rings were cul-tured in collagen gels in the presence or absence ofVEGF/S1P. Some cultures were transfected repeat-edly with siRNA for N-cadherin (NCDi) orscrambled control (scr) as described. After 7 d, vas-cular sprouts were quantified by analysis of phasecontrast images as described. Microvessel densitieswere quantitated by the NIH image software andpresented as fold increase over that of control cul-ture. Data represent a representative example froman experiment that was repeated two times. (b) Vas-cular maturation of neovessels was assessed by im-munofluorescence microscopy of sections of aorticrings in collagen gel. PECAM (red), �-smoothmuscle actin (green), and an overlay is shown. Notethat siRNA for N-cadherin (NCDi, 1 µM) inhibitedmicrovessel stabilization. Bar, 100 µm. Nonspecificstaining of elastin fibers was seen with both anti-bodies. However, immunostaining of neovesselsprouts was specific. (c) Pericyte ensheathment ofmicrovessels was reconstructed in three dimensions with z-series of PECAM (red) and �-smooth muscle actin (green) images obtainedfrom representative fields. Note that suppression of N-cadherin dramatically abolished pericyte coverage of microvessels. Bar, 10 µm.(d) Protein extracts from rat aorta treated with siRNA were immunoprecipitated for N-cadherin (NCD). VE-cadherin (VECD) levelswere not altered in the whole-cell lysate (WCL).

    Paik et al.

    2398 GENES & DEVELOPMENT

    Cold Spring Harbor Laboratory Press on June 2, 2021 - Published by genesdev.cshlp.orgDownloaded from

    http://genesdev.cshlp.org/http://www.cshlpress.com

  • polymerization and N-cadherin multiprotein complextrafficking to polarized plasma membrane microdo-mains.

    Our study defines two essential steps in the formationof S1P1-induced N-cadherin-based EC–mural cell junc-tions. The first step involves N-cadherin trafficking tothe EC surface. S1P induced the activation of the smallGTPase Rac, which is essential for proper trafficking ofN-cadherin via the MT cytoskeleton network. Weshowed the MT polymerization in response to S1P bymonitoring the dynamics of EB1-GFP, a MT tip-bindingprotein. The mechanism by which Rac1 stimulates MTpolymerization is thought to involve the activation ofthe protein kinase p65PAK and phosphorylation of stath-min, a MT destabilizing protein (Daub et al. 2001).

    Second, we found that S1P increases N-cadherin-beadbinding to cells even in the presence of nocodazole, sug-gesting that S1P1 may regulate the adhesive activity ofN-cadherin. It has been proposed that anchoring of thecadherin C terminus to the actin cytoskeleton may sta-bilize cadherin-mediated adhesion (Watabe et al. 1994).We show that the GPCR S1P1-induced �-, �-, and p120-catenin association with N-cadherin, which may furthersupport cadherin-dependent adhesion. Moreover, clus-tering of cadherin through the juxtamembrane domain

    (JMD) may be regulated by S1P1 signaling. Requirementof JMD in the regulation of cadherin adhesive activityhas been postulated (Yap et al. 1998). p120-catenin, origi-nally identified as a Src substrate that binds to the JMDof cadherin, may influence the dimerization or cluster-ing of cadherin (Ozawa 2003). Interestingly, the angio-genic factor VEGF induces the decrease in serine/threo-nine phosphorylation but the increase in tyrosine phos-phorylation of p120-catenin in EC (Esser et al. 1998;Wong et al. 2000). We show that S1P regulates the phos-phorylation of p120-catenin in a similar manner. Fur-ther, we also show that N-cadherin tyrosine phosphory-lation is induced by S1P. Indeed, p120-catenin has beenshown to be strongly tyrosine phosphorylated at earlytimes of calcium-induced keratinocyte differentiation,which leads to increased interaction between p120-catenin and cadherin (Calautti et al. 1998). Our findingthat the active signaling via the S1P1 GPCR is needed forN-cadherin activation/trafficking suggest a novelmechanism in the regulation of cell–cell adhesion andvascular stabilization.

    A related question is how N-cadherin activation in ECby S1P1 signaling activates the mural cell N-cadherin intrans. It has been previously demonstrated that whencells are attached to the substrata coated with a highdensity of the adhesive ectodomain of Xenopus C-cad-herin, forced clustering of the ectodomain occurs, whichin turn significantly strengthened adhesiveness (Yap etal. 1997a,b). Similarly, such clustering and activation oflocal N-cadherin on the EC surface by S1P1 may induceclustering of N-cadherin on neighboring mural cells.This possibility needs to be tested further by sensitivebiophysical methods.

    Most interestingly, we have discovered that asymmet-ric distribution of N-cadherin is induced by S1P1 signal-ing in EC. It should be pointed out that apical localiza-tion of N-cadherin in cultured endothelial cells does notimply a luminal distribution in vivo. Our data simplyimply that N-cadherin trafficking to polarized domainson the plasma membrane is a receptor-mediated event. Arecent study has demonstrated that N-cadherin is one ofthe earliest proteins to be expressed asymmetrically dur-ing gastrulation, and inhibition of N-cadherin leads torandomized looping of the heart, suggesting the criticalrole of this adhesion receptor in asymmetry determina-tion (Garcia-Castro et al. 2000). However, active polar-ization of cadherin molecules upon extracellular stimu-lation has not been reported. MT has been recognized asan important determinant of polarity in different celltypes. For example, neuronal polarity is determined bytranslocation of PAR3 to distal tips of axons by KIF3A, aplus-end-directed MT motor protein (Nishimura et al.2004). Epithelial polarization is often accompanied by adramatic change in MT organization. The polarized de-livery of apical (luminal) membrane proteins is influ-enced by MT dynamics (Mays et al. 1994). We proposethat asymmetric distribution of N-cadherin may providea proper spatial cue in vascular maturation. Indeed,many sporadic vascular malformation diseases includingKlippel-Trenaunay and Sturge-Weber syndromes have

    Figure 9. N-cadherin is required for neovessel maturation invivo. (a) Silencing of S1P1 expression in vivo by siRNA. Matrigelplugs containing FGF-2 were supplemented with siRNA forS1P1 or a scrambled siRNA. Total RNA from Matrigel plugs wasanalyzed by a Northern blot. (E1i) S1P1 siRNA; (scr) scrambledsiRNA. Band intensities were normalized by GAPDH level. (b)Histological analysis of Matrigel sections. Anti-�-smoothmuscle actin (SM-�-actin) and anti-PECAM staining of plug sec-tions from parallel sections. (CON) No FGF; (FGF) bFGF alone;(scr) scrambled siRNA 0.5 µM + FGF; (E1i) S1P1/EDG-1 siRNA0.5 µM + FGF; (NCDi) N-cadherin siRNA 0.5 µM + FGF. Bar, 50µm. Note that S1P1 siRNA dramatically inhibited neovesselformation, and N-cadherin siRNA abolished mural cell cover-age of PECAM-positive neovessels. (Arrowheads) DisorganizedEC. Data represent a representative example from an experi-ment that was repeated two times. (c) Protein extracts fromMatrigel plugs were examined for the expression of N-cadherin,SM-�-actin, and �-actin.

    Mechanism of vascular maturation

    GENES & DEVELOPMENT 2399

    Cold Spring Harbor Laboratory Press on June 2, 2021 - Published by genesdev.cshlp.orgDownloaded from

    http://genesdev.cshlp.org/http://www.cshlpress.com

  • been linked to asymmetric (left–right body axis) devel-opment of symptoms, suggesting potential correlationbetween vascular development and regulation of asym-metry (Cohen 2001). Indeed, those patients are shown toexhibit clear asymmetry of cerebral and cerebellar hemi-spheres where N-cadherin and S1P1 are known to behighly expressed.

    Potent inhibitory effects of N-cadherin siRNA in theMatrigel plug assay in vivo as well as the rat aortic ringassay ex vivo show that N-cadherin is a critical compo-nent of neovessel stabilization. The absence of �-smoothmuscle actin positive cells around N-cadherin-depletedneovessels may provide clues how N-cadherin functionsduring the maturation of angiogenic sprouts. AlthoughN-cadherin was implicated in breast cancer cell migra-tion in vitro (Hazan et al. 2000), down-regulation of N-cadherin by RNAi did not impede migration of 10T1/2cells or VSMC toward PDGF or serum (data not shown).Proliferation of VSMC in the rat aortic ring was also notaffected, suggesting that N-cadherin may primarily playa role in EC/mural cell interaction. It is possible thatcell–cell interaction is necessary for the coordinated mu-ral cell migration along the EC of neovessels as “longi-tudal migration” proposed previously (Carmeliet et al.1999). Alternatively, EC/mural cell contact may initiatethe expression of �-smooth muscle actin within perivas-cular mesenchymal cells. Indeed, activation of TGF-� bycell–cell contact in EC/10T1/2 cells was reported as apotential mechanism of mural cell differentiation (Hirs-chi et al. 1998). The fate of mural cells after N-cadherinengagement warrants further investigation.

    S1P is secreted abundantly by platelets in the adult(Yatomi et al. 2001). Although the origin of S1P in theembryo is unknown, recent data suggest that vascularEC can generate S1P in the extracellular environment bythe action of the sphingosine kinase ecto enzyme (An-cellin et al. 2002; Sanchez et al. 2003). Thus, plasma-borne S1P can regulate EC/mural cell interaction, andmay also be required for physiological maintenance ofthe vascular tree. In addition, S1P induces NO produc-tion in EC by Akt-dependent phosphorylation of the en-dothelial cell nitric oxide synthase enzyme (Morales-Ruiz et al. 2001). Therefore, an important physiologicalfunction of S1P may be to promote vascular maturationand to maintain vascular homeostasis. In this context, ithas been appreciated for several decades that platelet-derived molecules are important for the preservation ofEC integrity, which includes proper regulation of vascu-lar tone, permeability, and prevention of thrombosis(Gimbrone et al. 1969). Indeed, an experimental im-munomodulator FTY720, which is phosphorylated bythe sphingosine kinase-2 enzyme into a S1P receptoragonist, profoundly inhibits vascular permeability invivo (Sanchez et al. 2003). Although previous studieshave emphasized the role of S1P and its receptors in pro-moting EC barrier integrity, the mechanisms identifiedare limited to EC junction assembly (Lee et al. 1999;Garcia et al. 2001; Schaphorst et al. 2003). These recentdata, together with the mechanistic information pro-vided in this study, demonstrate that S1P1 receptor sig-

    naling in EC is a critical modulator of vascular stabili-zation in developmental and physiological contexts.Modulation of this pathway to induce vascular stabili-zation in a regulated manner may prove useful in variouspathological conditions in which aberrant EC/pericyteinteractions occur, for example, in cancer, diabetic reti-nopathy, sepsis, and so on. In addition, this pathwaycould be used to coax the formation of new collateralvessels in ischemic tissues such as the heart and thelimb with the aim of reestablishing blood flow.

    Materials and methods

    EC/mural cell adhesion assay

    Mouse embryonic endothelial cells were isolated from E12.5embryos and immortalized with polyoma middle T carryingretroviruses as described (Carmeliet et al. 1999). For in vitroadhesion (Hirschi et al. 1998), 10T1/2 cells were prelabeled withVybrant (Molecular Probe) cell tracker and incubated with ad-herent MEEC for 1 h either in the presence or absence of 100 nMS1P, and trypsinized under mild conditions (1× Hank’s balancedsalt solution, 0.01% trypsin, 1 mM CaCl2) to preserve cell sur-face cadherin (Gavard et al. 2004). Labeled cells were incubatedwith adherent MEEC for 1 h either in the presence or absence of100 nM S1P, other lipids, inhibitors (2 mM EGTA, 100 nMEchistatin, 0.3 mM GRGDTP RGD peptide) or cytokines (10ng/mL PDGF-BB, 50 ng/mL VEGF, 0.6 g/mL Ang1 and 2). Un-bound cells were washed and adherent cells on the plates werequantified with a fluorimeter. Alternatively, fully adherent10T1/2 cells (containing multiple fine processes as shown inFig. 1b) were imaged and scored after fluorescence microscopy.The results were subjected to statistical analysis by Student’s ttest unless specified.

    Inhibition of gene expression by siRNA

    Inhibition of S1P1/EDG-1 or N-cadherin expression by siRNAwas performed using specific siRNA reagents as described (El-bashir et al. 2001). The siRNAs used are mouse S1P1/EDG-1-(sense CUGACUUCAGUGGUGUUCAdTdT and antisenseUGAACACCACUGAAGUCAGdTdT), N-cadherin siRNA(UGUCAAUGGGGUUCUCCACdTdT and GUGGAGAACCCCAUUGACAdTdT), E-cadherin siRNAs (CCUGGUUCAGAUCAAAUCCdTdT and GGAUUUGAUCUGAACCAGGdTdT), and scrambled siRNA (CAUGCGGAUUCGGAUUUUCdTdT and GAAAAUCCGAAUCCGCAUGdTdT). Oligo-nucleotides were synthesized by Dharmacon Research Inc., de-protected, and duplexed as described (Elbashir et al. 2001). Forthe silencing of Rac1, SMARTpool Rac1 siRNA from Dharma-con was used at 200 nM. Target cells were transfected withsiRNA complexed with Oligofectamine reagent (Invitrogen) and2 d after transfection, gene expression was examined by North-ern or Western blot analysis. For testing the specificity of N-cadherin siRNA, HEK293 cells transiently expressing mouse(Shan et al. 2000) or chicken N-cadherin were transfected with200 nM N-cadherin siRNA for 48 h and analyzed. For NcadFc-bead binding, mouse N-cadherin-GFP (provided by Dr.Gauthier, Centre de Recherche de Biochemie Macromolecu-laire, Centre National de la Recherche Scientifique Unite Pro-pre de Recherche, Montpellier, France) or chicken N-cadherin-GFP (provided by Dr. Eppenberger, Institute of Cell Biology,Federal Institute of Technology (ETH), Zurich, Switzerland)

    Paik et al.

    2400 GENES & DEVELOPMENT

    Cold Spring Harbor Laboratory Press on June 2, 2021 - Published by genesdev.cshlp.orgDownloaded from

    http://genesdev.cshlp.org/http://www.cshlpress.com

  • were transfected into HUVEC along with 200 nM N-cadherinsiRNA.

    Matrigel plug assay

    siRNAs were mixed with Matrigel and implanted subcutane-ously as described before. Some plugs contained FGF-2 to stimu-late angiogenesis. One week after injection, plugs were removedand analyzed by immunohistochemistry (Lee et al. 1999). Alter-natively, protein or RNA was extracted from plugs and geneexpression was examined by Western or Northern analysis (Paiket al. 2001).

    Immunoprecipitation and immunoblotting

    Cells were lysed as described (Lee et al. 1999). One hundredmicrograms of cell lysate were incubated with anti-�-catenin orN-cadherin antibodies (Santa Cruz Biotechnology, Inc.) and im-mune complexes were captured with precleared protein Gbeads. To exclude the possibility of actin filament-mediatedaggregation of proteins, cells were treated with cytochalasin D(1 µM) for 10 min before lysis and cytochalasin D was alsoincluded in the lysis buffer. For the detections of either tyrosineor serine/threonine phosphorylation on cadherins and catenins,500 µg of lysates (prepared in CHAPS buffer with 1 mM sodiumorthovanadate, 1 mM of sodium fluoride, 10 mM of �-glycero-phosphate) were immunoprecipitated and probed with anti-phosphotyrosine (clone 4G10, Upstate Biotechnology) or anti-phosphoserine/threonine (clone 22a, BD Pharmingen) antibod-ies. Cadherin antibodies and immune complexes were capturedwith precleared protein G beads. For surface biotinylation, cellswere exposed to 2 mg/mL sulfo-NHS-biotin (Pierce) for 10 minon ice. Biotinylated proteins were pulled down with Streptavi-din beads.

    Protein identification by µ-capillary liquid chromatographyand tandem mass spectrometry

    Characterization of N-cadherin associated multimeric proteincomplexes was performed by large-scale immunoprecipitationof N-cadherin from S1P-treated HUVEC cells (1 × 107 cells or 3mg of total cell lysate). Briefly, cells were lysed (1% TritonX-100, 1% CHAPS-containing lysis buffer), immunoprecipi-tated with 3 µg N-cadherin antibody, and immunoprecipitatedproteins were then resolved by SDS-PAGE and visualized byCoomassie blue staining. Protein bands were then excised, de-hydrated with acetonitrile, and in-gel digested with sequencing-grade Trypsin. Peptides from each of the protein bands werethen extracted with a buffer containing 5% formic acid and 50%acetonitrile, dried, and analyzed by a µ-capillary LC-MS/MSprocedure using a Finnigan LCQ-DECA ion-trap tandem massspectrometer. Uninterpreted MS/MS spectra were comparedagainst a nonredundant human protein database using theSEQUEST algorithm (Sadygov et al. 2002). Protein coverage wasanalyzed by the INTERACT program and probabilities of theproteins were calculated by Peptide Prophet Software (Han et al.2001). Only conclusively identified proteins with multiple high-scoring peptides are shown.

    Immunofluorescence and imaging

    Mouse embryos (E12.5) were fixed in 4% formaldehyde, soakedin 0.3 M sucrose overnight, and cryostat sectioned. Five-micronsections were stained with PECAM/CD31 (1:100, Pharmingen),�-smooth muscle actin (Sigma), or N-cadherin (BD Biosciences)followed by Alexa 488 goat anti-mouse and Texas red goat anti-

    rat in 5% nonfat dry milk in PBS. Confocal images were ob-tained using a Zeiss LSM510 confocal microscope. Quantitationof N-cadherin localization was performed using the MetaMorphsoftware. A constant background value was subtracted fromeach image. Intensities were quantified by averaging fluores-cence intensity along a line drawn following the EC/EC or EC/VSMC boundary on confocal images.

    For N-cadherin-GFP localization, HUVEC were transientlytransfected with pNcad-GFP (Mary et al. 2002) (kindly providedby Dr. Gauthier) and live-cell imaging was done by scanning at1-min intervals using a Zeiss LSM510 confocal microscope.Time series images were stored as a movie file at 8 frames/sec.For EB1-GFP (kindly provided by Dr. Piehl, Department of Bio-logical Sciences, Lehigh University, Bethlehem, PA) (Piehl et al.2004), transfected HUVEC were scanned at 15-sec intervals andstored as 15 frames/sec. For coculture imaging one cell type waslabeled with FM4-64 (Molecular Probes) to distinguish it fromthe other cell types. For the study of dorsal ruffle formation cellswere fixed and stained with antibodies followed by incubationwith DiI-C18 (1 µg/mL; Molecular Probes, Inc.) for 30 min. Co-localization of N-cadherin (mouse monoclonal; BD Biosciences)with connexin43 (rabbit polyclonal; Zymed) or Tie2 (rabbitpolyclonal; Santa Cruz Biotechnology, Inc.) was performed us-ing Alexa 546 (goat anti-mouse) and Alex 488 (goat anti-rabbit).Optical z-slices (0.5 µm) were collected and viewed using theLSM510 browser.

    N-cadherin–Fc-bead adhesion assay

    pCECH-NcadFc (construct encoding chicken N-cadherinectodomain-mouse IgG Fc chimera) was kindly provided by Dr.Mege (Signalisation et Differenciation Cellulaires dans les Sys-tems Nerveux et Musculaire, U440 INSERM/UPMC, Institutdu Fer a Moulin, Paris, France) (Lambert et al. 2000). NcadFcprotein was collected from AIM-V medium of HEK293 cellstransfected with pCECH-NcadFc using protein G beads. Protein(200 µg) was eluted and rebound to goat anti-mouse Fc IgG (200µg, Jackson Immunoresearch)-coated polystyrene latex sulfatebeads (1 µm, 2 × 109; Interfacial Dynamics). Beads were placedon top of cells for 1 h in the presence or absence of S1P. Noco-dazole (10 µM for 10 min or 2 µM for 1 h) or cytochalasin D (1µM for 10 min) were incubated with the cells before fixation andanalysis. N17Rac, N19Rho, and pTet adenoviruses were giftsfrom Dr. Sessa (Department of Pharmacology, Yale UniversitySchool of Medicine, New Haven, CT) (Gratton et al. 2001). HU-VECs were infected with dominant negative viruses (100 M.O.I)along with pTet (5 M.O.I) in the presence of 2 µg/mL tetracy-cline. Cells were recovered and induced to express N17Rac orN19Rho by removing tetracycline overnight. Beads were visu-alized by labeling with Alexa conjugated anti-mouse Fc anti-bodies and their numbers were quantified on a single cell basiswith the ImageJ version 1.30 software.

    Ex vivo rat aorta model of angiogenesis andvessel maturation

    Aortic rings obtained from 5- to 10-week-old Sprague-Dawleymale rats (Charles River Laboratories) were embedded in type Icollagen gels and transferred to 24 wells, each containing 0.5 mLserum-free endothelial basal medium as described (Nicosia andOttinetti 1990). The growth medium was changed three times aweek starting from day 3. siRNA against N-cadherin was com-plexed with liposome and overlaid on aortic rings embedded incollagen gel as 1 µM every 2 d. As a control scrambled siRNAwas used. After 96 h, cultures were treated with 5-bromo-2�-deoxy-uridine (BrdU), for 1 h and gels were embedded in OCT

    Mechanism of vascular maturation

    GENES & DEVELOPMENT 2401

    Cold Spring Harbor Laboratory Press on June 2, 2021 - Published by genesdev.cshlp.orgDownloaded from

    http://genesdev.cshlp.org/http://www.cshlpress.com

  • and cryosectioned. Ten-micron sections were stained forPECAM, �-smooth muscle actin, or anti-BrdU and images werescanned using a Zeiss LSM510 confocal microscope. Z-serialimages were three dimensionally reconstructed using IMARISsoftware (Bitplane).

    Acknowledgments

    We are grateful to Bill Sessa for Rho and Rac adenoviral con-structs, Bruce White for chicken N-cadherin cDNA, LawrenceShapiro for mouse N-cadherin cDNA, Rene-Marc Mege for theN-cadherin-Fc construct, Cécile Gauthier-Rouvière and HansEppenberger for N-cadherin GFP, and Michelle Piehl for EB1-GFP construct. We also thank Henry Furneaux and PhilipZamore for advice on siRNA experiments, Richard Mains for rataorta explant experiments, and Guo-Hua Fong and Julie D. Sabafor helpful comments. This work is supported by NIH grantsHL67330 and HL70694 to T.H.

    References

    Allende, M.L., Yamashita, T., and Proia, R.L. 2003. G-protein-coupled receptor S1P1 acts within endothelial cells to regu-late vascular maturation. Blood 102: 3665–3667.

    Allt, G. and Lawrenson, J.G. 2001. Pericytes: Cell biology andpathology. Cells Tissues Organs 169: 1–11.

    Ancellin, N., Colmont, C., Su, J., Li, Q., Mittereder, N., Chae,S.S., Stefansson, S., Liau, G., and Hla, T. 2002. Extracellularexport of sphingosine kinase-1 enzyme. Sphingosine 1-phos-phate generation and the induction of angiogenic vascularmaturation. J. Biol. Chem. 277: 6667–6675.

    Balconi, G., Spagnuolo, R., and Dejana, E. 2000. Development ofendothelial cell lines from embryonic stem cells: A tool forstudying genetically manipulated endothelial cells in vitro.Arterioscler. Thromb. Vasc. Biol. 20: 1443–1451.

    Benjamin, L.E., Hemo, I., and Keshet, E. 1998. A plasticity win-dow for blood vessel remodelling is defined by pericyte cov-erage of the preformed endothelial network and is regulatedby PDGF-B and VEGF. Development 125: 1591–1598.

    Bergers, G., Song, S., Meyer-Morse, N., Bergsland, E., and Hana-han, D. 2003. Benefits of targeting both pericytes and endo-thelial cells in the tumor vasculature with kinase inhibitors.J. Clin. Invest. 111: 1287–1295.

    Calautti, E., Cabodi, S., Stein, P.L., Hatzfeld, M., Kedersha, N.,and Paolo Dotto, G. 1998. Tyrosine phosphorylation and srcfamily kinases control keratinocyte cell–cell adhesion. J.Cell Biol. 141: 1449–1465.

    Carmeliet, P., Lampugnani, M.G., Moons, L., Breviario, F.,Compernolle, V., Bono, F., Balconi, G., Spagnuolo, R., Oos-tuyse, B., Dewerchin, M., et al. 1999. Targeted deficiency orcytosolic truncation of the VE-cadherin gene in mice impairsVEGF-mediated endothelial survival and angiogenesis. Cell98: 147–157.

    Chen, X., Kojima, S., Borisy, G.G., and Green, K.J. 2003. p120catenin associates with kinesin and facilitates the transportof cadherin–catenin complexes to intercellular junctions. J.Cell Biol. 163: 547–557.

    Cohen Jr., M.M. 2001. Asymmetry: Molecular, biologic, em-bryopathic, and clinical perspectives. Am. J. Med. Genet.101: 292–314.

    Daub, H., Gevaert, K., Vandekerckhove, J., Sobel, A., and Hall,A. 2001. Rac/Cdc42 and p65PAK regulate the microtubule-destabilizing protein stathmin through phosphorylation atserine 16. J. Biol. Chem. 276: 1677–1680.

    Dejana, E. 2004. Endothelial cell–cell junctions: Happy to-gether. Nat. Rev. Mol. Cell Biol. 5: 261–270.

    Elbashir, S.M., Harborth, J., Lendeckel, W., Yalcin, A., Weber,K., and Tuschl, T. 2001. Duplexes of 21-nucleotide RNAsmediate RNA interference in cultured mammalian cells. Na-ture 411: 494–498.

    Engerman, R.L. 1989. Pathogenesis of diabetic retinopathy. Dia-betes 38: 1203–1206.

    Esser, S., Lampugnani, M.G., Corada, M., Dejana, E., and Risau,W. 1998. Vascular endothelial growth factor induces VE-cad-herin tyrosine phosphorylation in endothelial cells. J. CellSci. 111: 1853–1865.

    Fusco, A., Portella, G., Grieco, M., Tajana, G., Di Minno, G.,Polli, N., and Pinto, A. 1988. A retrovirus carrying the poly-omavirus middle T gene induces acute thrombocythemicmyeloproliferative disease in mice. J. Virol. 62: 361–365.

    Garcia, J.G., Liu, F., Verin, A.D., Birukova, A., Dechert, M.A.,Gerthoffer, W.T., Bamberg, J.R., and English, D. 2001. Sphin-gosine 1-phosphate promotes endothelial cell barrier integ-rity by Edg-dependent cytoskeletal rearrangement. J. Clin.Invest. 108: 689–701.

    Garcia-Castro, M.I., Vielmetter, E., and Bronner-Fraser, M.2000. N-Cadherin, a cell adhesion molecule involved in es-tablishment of embryonic left–right asymmetry. Science288: 1047–1051.

    Gavard, J., Lambert, M., Grosheva, I., Marthiens, V., Irinopou-lou, T., Riou, J.F., Bershadsky, A., and Mege, R.M. 2004.Lamellipodium extension and cadherin adhesion: Two cellresponses to cadherin activation relying on distinct signal-ling pathways. J. Cell Sci. 117: 257–270.

    Gerhardt, H., Wolburg, H., and Redies, C. 2000. N-cadherin me-diates pericytic–endothelial interaction during brain angio-genesis in the chicken. Dev. Dyn. 218: 472–479.

    Gimbrone Jr., M.A., Aster, R.H., Cotran, R.S., Corkery, J., Jandl,J.H., and Folkman, J. 1969. Preservation of vascular integrityin organs perfused in vitro with a platelet-rich medium. Na-ture 221: 33–36.

    Gottardi, C.J. and Gumbiner, B.M. 2001. Adhesion signaling:How �-catenin interacts with its partners. Curr. Biol.11: R792–R794.

    Gratton, J.P., Morales-Ruiz, M., Kureishi, Y., Fulton, D., Walsh,K., and Sessa, W.C. 2001. Akt down-regulation of p38 sig-naling provides a novel mechanism of vascular endothelialgrowth factor-mediated cytoprotection in endothelial cells.J. Biol. Chem. 276: 30359–30365.

    Hammes, H.P., Lin, J., Renner, O., Shani, M., Lundqvist, A.,Betsholtz, C., Brownlee, M., and Deutsch, U. 2002. Pericytesand the pathogenesis of diabetic retinopathy. Diabetes51: 3107–3112.

    Han, D.K., Eng, J., Zhou, H., and Aebersold, R. 2001. Quantita-tive profiling of differentiation-induced microsomal proteinsusing isotope-coded affinity tags and mass spectrometry.Nat. Biotechnol. 19: 946–951.

    Hazan, R.B., Phillips, G.R., Qiao, R.F., Norton, L., and Aaron-son, S.A. 2000. Exogenous expression of N-cadherin in breastcancer cells induces cell migration, invasion, and metastasis.J. Cell Biol. 148: 779–790.

    Hellberg, C.B., Burden-Gulley, S.M., Pietz, G.E., and Brady-Kalnay, S.M. 2002. Expression of the receptor protein-tyro-sine phosphatase, PTPmu, restores E-cadherin-dependentadhesion in human prostate carcinoma cells. J. Biol. Chem.277: 11165–11173.

    Hellstrom, M., Kalen, M., Lindahl, P., Abramsson, A., and Bet-sholtz, C. 1999. Role of PDGF-B and PDGFR-� in recruit-ment of vascular smooth muscle cells and pericytes duringembryonic blood vessel formation in the mouse. Develop-

    Paik et al.

    2402 GENES & DEVELOPMENT

    Cold Spring Harbor Laboratory Press on June 2, 2021 - Published by genesdev.cshlp.orgDownloaded from

    http://genesdev.cshlp.org/http://www.cshlpress.com

  • ment 126: 3047–3055.Hellstrom, M., Gerhardt, H., Kalen, M., Li, X., Eriksson, U.,

    Wolburg, H., and Betsholtz, C. 2001. Lack of pericytes leadsto endothelial hyperplasia and abnormal vascular morpho-genesis. J. Cell Biol. 153: 543–553.

    Hirschi, K.K. and D’Amore, P.A. 1997. Control of angiogenesisby the pericyte: Molecular mechanisms and significance.EXS 79: 419–428.

    Hirschi, K.K., Rohovsky, S.A., and D’Amore, P.A. 1998. PDGF,TGF-�, and heterotypic cell–cell interactions mediate endo-thelial cell-induced recruitment of 10T1/2 cells and theirdifferentiation to a smooth muscle fate. J. Cell Biol.141: 805–814.

    Hla, T. and Maciag, T. 1990. An abundant transcript induced indifferentiating human endothelial cells encodes a polypep-tide with structural similarities to G-protein-coupled recep-tors. J. Biol. Chem. 265: 9308–9313.

    Jain, R.K. 2003. Molecular regulation of vessel maturation. Nat.Med. 9: 685–693.

    Kluk, M.J. and Hla, T. 2001. Role of the sphingosine 1-phos-phate receptor EDG-1 in vascular smooth muscle cell pro-liferation and migration. Circ. Res. 89: 496–502.

    Lambert, M., Padilla, F., and Mege, R.M. 2000. Immobilizeddimers of N-cadherin-Fc chimera mimic cadherin-mediatedcell contact formation: Contribution of both outside-in andinside-out signals. J. Cell Sci. 113: 2207–2219.

    Lee, M.J., Van Brocklyn, J.R., Thangada, S., Liu, C.H., Hand,A.R., Menzeleev, R., Spiegel, S., and Hla, T. 1998. Sphingo-sine-1-phosphate as a ligand for the G protein-coupled recep-tor EDG-1. Science 279: 1552–1555.

    Lee, M.J., Thangada, S., Claffey, K.P., Ancellin, N., Liu, C.H.,Kluk, M., Volpi, M., Sha’afi, R.I., and Hla, T. 1999. Vascularendothelial cell adherens junction assembly and morphogen-esis induced by sphingosine-1-phosphate. Cell 99: 301–312.

    Lee, M.J., Thangada, S., Paik, J.H., Sapkota, G.P., Ancellin, N.,Chae, S.S., Wu, M., Morales-Ruiz, M., Sessa, W.C., Alessi,D.R., et al. 2001. Akt-mediated phosphorylation of the Gprotein-coupled receptor EDG-1 is required for endothelialcell chemotaxis. Mol. Cell 8: 693–704.

    Liu, Y., Wada, R., Yamashita, T., Mi, Y., Deng, C.X., Hobson,J.P., Rosenfeldt, H.M., Nava, V.E., Chae, S.S., Lee, M.J., et al.2000. Edg-1, the G protein-coupled receptor for sphingosine-1-phosphate, is essential for vascular maturation. J. Clin.Invest. 106: 951–961.

    Mary, S., Charrasse, S., Meriane, M., Comunale, F., Travo, P.,Blangy, A., and Gauthier-Rouviere, C. 2002. Biogenesis ofN-cadherin-dependent cell–cell contacts in living fibroblastsis a microtubule-dependent kinesin-driven mechanism. Mol.Biol. Cell 13: 285–301.

    Mays, R.W., Beck, K.A., and Nelson, W.J. 1994. Organizationand function of the cytoskeleton in polarized epithelial cells:A component of the protein sorting machinery. Curr. Opin.Cell Biol. 6: 16–24.

    Morales-Ruiz, M., Lee, M.J., Zollner, S., Gratton, J.P., Scotland,R., Shiojima, I., Walsh, K., Hla, T., and Sessa, W.C. 2001.Sphingosine 1-phosphate activates Akt, nitric oxide produc-tion, and chemotaxis through a Gi protein/phosphoinositide3-kinase pathway in endothelial cells. J. Biol. Chem.276: 19672–19677.

    Nicosia, R.F. and Ottinetti, A. 1990. Growth of microvessels inserum-free matrix culture of rat aorta. A quantitative assayof angiogenesis in vitro. Lab. Invest. 63: 115–122.

    Nishimura, T., Kato, K., Yamaguchi, T., Fukata, Y., Ohno, S.,and Kaibuchi, K. 2004. Role of the PAR-3–KIF3 complex inthe establishment of neuronal polarity. Nat. Cell Biol.6: 328–334.

    Ozawa, M. 2003. p120-independent modulation of E-cadherinadhesion activity by the membrane-proximal region of thecytoplasmic domain. J. Biol. Chem. 278: 46014–46020.

    Paik, J.H., Chae, S., Lee, M.J., Thangada, S., and Hla, T. 2001.Sphingosine 1-phosphate-induced endothelial cell migrationrequires the expression of EDG-1 and EDG-3 receptors andRho-dependent activation of � v�3- and �1-containing inte-grins. J. Biol. Chem. 276: 11830–11837.

    Piehl, M., Tulu, U.S., Wadsworth, P., and Cassimeris, L. 2004.Centrosome maturation: Measurement of microtubulenucleation throughout the cell cycle by using GFP-taggedEB1. Proc. Natl. Acad. Sci. 101: 1584–1588.

    Risau, W. 1997. Mechanisms of angiogenesis. Nature 386: 671–674.

    Sadygov, R.G., Eng, J., Durr, E., Saraf, A., McDonald, H., Mac-Coss, M.J., and Yates III, J.R. 2002. Code developments toimprove the efficiency of automated MS/MS spectra inter-pretation. J. Proteome Res. 1: 211–215.

    Sanchez, T., Estrada-Hernandez, T., Paik, J.H., Wu, M.T., Ven-kataraman, K., Brinkmann, V., Claffey, K., and Hla, T. 2003.Phosphorylation and action of the immunomodulatorFTY720 inhibits vascular endothelial cell growth factor-in-duced vascular permeability. J. Biol. Chem. 278: 47281–47290.

    Schaphorst, K.L., Chiang, E., Jacobs, K.N., Zaiman, A., Natara-jan, V., Wigley, F., and Garcia, J.G. 2003. Role of sphingo-sine-1 phosphate in the enhancement of endothelial barrierintegrity by platelet-released products. Am. J. Physiol. LungCell Mol. Physiol. 285: L258–L267.

    Shan, W.S., Tanaka, H., Phillips, G.R., Arndt, K., Yoshida, M.,Colman, D.R., and Shapiro, L. 2000. Functional cis-het-erodimers of N- and R-cadherins. J. Cell Biol. 148: 579–590.

    Watabe, M., Nagafuchi, A., Tsukita, S., and Takeichi, M. 1994.Induction of polarized cell–cell association and retardationof growth by activation of the E-cadherin–catenin adhesionsystem in a dispersed carcinoma line. J. Cell Biol. 127: 247–256.

    Wong, E.Y., Morgan, L., Smales, C., Lang, P., Gubby, S.E., andStaddon, J.M. 2000. Vascular endothelial growth factorstimulates dephosphorylation of the catenins p120 and p100in endothelial cells. Biochem. J. 346: 209–216.

    Yancopoulos, G.D., Davis, S., Gale, N.W., Rudge, J.S., Wiegand,S.J., and Holash, J. 2000. Vascular-specific growth factors andblood vessel formation. Nature 407: 242–248.

    Yap, A.S., Brieher, W.M., and Gumbiner, B.M. 1997a. Molecularand functional analysis of cadherin-based adherens junc-tions. Annu. Rev. Cell Dev. Biol. 13: 119–146.

    Yap, A.S., Brieher, W.M., Pruschy, M., and Gumbiner, B.M.1997b. Lateral clustering of the adhesive ectodomain: A fun-damental determinant of cadherin function. Curr. Biol.7: 308–315.

    Yap, A.S., Niessen, C.M., and Gumbiner, B.M. 1998. The juxta-membrane region of the cadherin cytoplasmic tail supportslateral clustering, adhesive strengthening, and interactionwith p120ctn. J. Cell Biol. 141: 779–789.

    Yatomi, Y., Ozaki, Y., Ohmori, T., and Igarashi, Y. 2001. Sphin-gosine 1-phosphate: Synthesis and release. ProstaglandinsOther Lipid Mediat. 64: 107–122.

    Mechanism of vascular maturation

    GENES & DEVELOPMENT 2403

    Cold Spring Harbor Laboratory Press on June 2, 2021 - Published by genesdev.cshlp.orgDownloaded from

    http://genesdev.cshlp.org/http://www.cshlpress.com

  • 10.1101/gad.1227804Access the most recent version at doi: 18:2004, Genes Dev.

    Ji-Hye Paik, Athanasia Skoura, Sung-Suk Chae, et al. vascular stabilizationSphingosine 1-phosphate receptor regulation of N-cadherin mediates

    Material

    Supplemental

    http://genesdev.cshlp.org/content/suppl/2004/10/04/gad.1227804.DC1

    References

    http://genesdev.cshlp.org/content/18/19/2392.full.html#ref-list-1

    This article cites 57 articles, 32 of which can be accessed free at:

    License

    ServiceEmail Alerting

    click here.right corner of the article or

    Receive free email alerts when new articles cite this article - sign up in the box at the top

    Cold Spring Harbor Laboratory Press

    Cold Spring Harbor Laboratory Press on June 2, 2021 - Published by genesdev.cshlp.orgDownloaded from

    http://genesdev.cshlp.org/lookup/doi/10.1101/gad.1227804http://genesdev.cshlp.org/content/suppl/2004/10/04/gad.1227804.DC1http://genesdev.cshlp.org/content/18/19/2392.full.html#ref-list-1http://genesdev.cshlp.org/cgi/alerts/ctalert?alertType=citedby&addAlert=cited_by&saveAlert=no&cited_by_criteria_resid=protocols;10.1101/gad.1227804&return_type=article&return_url=http://genesdev.cshlp.org/content/10.1101/gad.1227804.full.pdfhttp://genesdev.cshlp.org/cgi/adclick/?ad=55564&adclick=true&url=https%3A%2F%2Fhorizondiscovery.com%2Fen%2Fcustom-synthesis%2Fcustom-rna%3Futm_source%3DCSHL_RNA%26utm_medium%3Dbanner%26utm_campaign%3Dcustom_synth%26utm_term%3Doligos%26utm_content%3Djan21http://genesdev.cshlp.org/http://www.cshlpress.com