HE Printed in U.S.A. Analysis of Biological Effects and ... · Analysis of Biological Effects and...

10
Analysis of Biological Effects and Signaling Properties of Flt-1 (VEGFR-1) and KDR (VEGFR-2) A REASSESSMENT USING NOVEL RECEPTOR-SPECIFIC VASCULAR ENDOTHELIAL GROWTH FACTOR MUTANTS* Received for publication, March 8, 2000, and in revised form, August 29, 2000 Published, JBC Papers in Press, October 31, 2000, DOI 10.1074/jbc.M002016200 Hendrik Gille‡, Joe Kowalski‡, Bing Li§, Jennifer LeCouter‡, Barbara Moffat, Thomas F. Zionchecki, Nicolas Pelletieri, and Napoleone Ferrara‡** From the Departments of Molecular Oncology, §Protein Engineering, Protein Chemistry, and iPharmacokinetics and Metabolism, Genentech, Inc., South San Francisco, California 94080 Endothelial cells express two related vascular endo- thelial growth factor (VEGF) receptor tyrosine kinases, KDR (kinase-insert domain containing receptor, or VEGFR-2) and Flt-1 (fms-like tyrosine kinase, or VEGFR- 1). Although considerable experimental evidence links KDR activation to endothelial cell mitogenesis, there is still significant uncertainty concerning the role of indi- vidual VEGF receptors for other biological effects such as vascular permeability. VEGF mutants that bind to either KDR or Flt-1 with high selectivity were used to determine which of the two receptors serves to mediate different VEGF functions. In addition to mediating mi- togenic signaling, selective KDR activation was suffi- cient for the activation of intracellular signaling path- ways implicated in cell migration. KDR stimulation caused tyrosine phosphorylation of both phosphatidyl- inositol 3-kinase and phospholipase Cg in primary en- dothelial cells and stimulated cell migration. KDR-selec- tive VEGF was also able to induce angiogenesis in the rat cornea to an extent indistinguishable from wild type VEGF. We also demonstrate that KDR, but not Flt-1, stimulation is responsible for the induction of vascular permeability by VEGF. Angiogenesis is an important developmental process and is also critically involved in several pathological conditions such as rheumatoid arthritis, diabetic retinopathy, and growth of solid tumors (1, 2). VEGF 1 is a major mediator of normal and abnormal angiogenesis (1). VEGF is secreted by tumor cells and their surrounding stroma and is causally involved in the progression of the majority of solid tumors (1). Inhibition of VEGF activity using various inhibitors results in suppression of tumor growth in vivo (3– 6). VEGF is also up-regulated in ischemic tissues and has been implicated as a potential therapy for peripheral and myocardial ischemia (7). VEGF is expressed as at least four different splice isoforms (1). The most abundantly expressed variant is VEGF 165 , which contains a heparin-binding domain. Two VEGF receptor tyro- sine kinases, Flt-1 (VEGFR-1) and KDR (VEGFR-2), are ex- pressed on proliferating and quiescent endothelial cells (8). Although Flt-1 exhibits higher affinity for VEGF 165 , its func- tion in the adult vasculature is still poorly understood. Mice lacking the complete Flt-1 gene display an increased number of endothelial progenitors and vascular disorganization and die in utero at embryonic day 9 (9, 10). However, animals homozygous for a deletion of the cytoplasmic domain are fertile and do not display any obvious defects (11). Mice in which the flk-1/KDR gene has been inactivated also die at embryonic day 9. They are deficient in vasculogenesis and also lack blood island formation (12). Placenta growth factor (PlGF) shares 53% identity with the platelet-derived growth factor-like domain of VEGF (13). It binds Flt-1 with high affinity but is unable to interact with KDR (14). PlGF exerts minimal effects on cell growth and migration, suggesting that binding to Flt-1 alone is not suffi- cient to mediate these activities. In some instances, however, effects of PlGF on mitogenicity and MAP kinase activation have been reported (15). In recent studies, the effects of PlGF on endothelial cells have been compared with those of another VEGF-like molecule, VEGF orf . Members of the VEGF orf gene family have been isolated from the Orf parapoxyvirus, and the encoded proteins bind to KDR but not Flt-1 (16, 17). VEGF orf mediates migration of KDR-expressing PAE cells and corneal angiogenesis to an extent that is comparable with VEGF (17, 18). However, the residues mediating VEGF orf binding to KDR are unknown because the three basic amino acids comprising the major determinants of VEGF binding to the KDR receptor are absent in VEGF orf . In addition, VEGF orf does not possess the heparin-binding domain found on VEGF. These differences may complicate conclusions about VEGF function, drawn from studies using VEGF orf proteins. VEGF binding to KDR leads to receptor phosphorylation and activation of MAP kinases as well as tyrosine phosphorylation of PI3K and PLCg. Although Flt-1 becomes phosphorylated and can activate several signal- ing molecules when expressed in heterologous cells (19), it is unclear whether these signaling events also occur in endothe- lial cells in response to Flt-1 engagement. Recent studies have shown that KDR signaling results in endothelial nitric-oxide synthase up-regulation and activation (20). Nitric oxide plays a critical role in the VEGF-induced endothelial cell proliferation, migration, and tube formation, as well as increased vascular permeability, hypotension, and an- giogenesis in vivo (21–24). VEGF stimulates the formation of vasodilator prostaglandins, which have been implicated as me- * The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement” in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. ** To whom correspondence should addressed: Dept. of Molecular Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080. Tel.: 650-225-2968; Fax: 650-225-6327; E-mail: [email protected]. 1 The abbreviations used are: VEGF, vascular endothelial growth factor; PlGF, placenta growth factor; PLCg, phospholipase Cg; PI3K, phosphatidylinositol 3-kinase; HUVEC, human umbilical vein endothe- lial cell(s); PAE, porcine aortic endothelial cells; ERK, extracellular signal-regulated kinase; MAP, mitogen-activated protein. THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. 276, No. 5, Issue of February 2, pp. 3222–3230, 2001 © 2001 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in U.S.A. This paper is available on line at http://www.jbc.org 3222 by guest on April 3, 2020 http://www.jbc.org/ Downloaded from

Transcript of HE Printed in U.S.A. Analysis of Biological Effects and ... · Analysis of Biological Effects and...

Page 1: HE Printed in U.S.A. Analysis of Biological Effects and ... · Analysis of Biological Effects and Signaling Properties of Flt-1 (VEGFR-1) and KDR (VEGFR-2) A REASSESSMENT USING NOVEL

Analysis of Biological Effects and Signaling Properties ofFlt-1 (VEGFR-1) and KDR (VEGFR-2)A REASSESSMENT USING NOVEL RECEPTOR-SPECIFIC VASCULAR ENDOTHELIAL GROWTHFACTOR MUTANTS*

Received for publication, March 8, 2000, and in revised form, August 29, 2000Published, JBC Papers in Press, October 31, 2000, DOI 10.1074/jbc.M002016200

Hendrik Gille‡, Joe Kowalski‡, Bing Li§, Jennifer LeCouter‡, Barbara Moffat¶,Thomas F. Zionchecki, Nicolas Pelletieri, and Napoleone Ferrara‡**

From the Departments of ‡Molecular Oncology, §Protein Engineering, ¶Protein Chemistry, and iPharmacokinetics andMetabolism, Genentech, Inc., South San Francisco, California 94080

Endothelial cells express two related vascular endo-thelial growth factor (VEGF) receptor tyrosine kinases,KDR (kinase-insert domain containing receptor, orVEGFR-2) and Flt-1 (fms-like tyrosine kinase, or VEGFR-1). Although considerable experimental evidence linksKDR activation to endothelial cell mitogenesis, there isstill significant uncertainty concerning the role of indi-vidual VEGF receptors for other biological effects suchas vascular permeability. VEGF mutants that bind toeither KDR or Flt-1 with high selectivity were used todetermine which of the two receptors serves to mediatedifferent VEGF functions. In addition to mediating mi-togenic signaling, selective KDR activation was suffi-cient for the activation of intracellular signaling path-ways implicated in cell migration. KDR stimulationcaused tyrosine phosphorylation of both phosphatidyl-inositol 3-kinase and phospholipase Cg in primary en-dothelial cells and stimulated cell migration. KDR-selec-tive VEGF was also able to induce angiogenesis in therat cornea to an extent indistinguishable from wild typeVEGF. We also demonstrate that KDR, but not Flt-1,stimulation is responsible for the induction of vascularpermeability by VEGF.

Angiogenesis is an important developmental process and isalso critically involved in several pathological conditions suchas rheumatoid arthritis, diabetic retinopathy, and growth ofsolid tumors (1, 2). VEGF1 is a major mediator of normal andabnormal angiogenesis (1). VEGF is secreted by tumor cellsand their surrounding stroma and is causally involved in theprogression of the majority of solid tumors (1). Inhibition ofVEGF activity using various inhibitors results in suppressionof tumor growth in vivo (3–6). VEGF is also up-regulated inischemic tissues and has been implicated as a potential therapyfor peripheral and myocardial ischemia (7).

VEGF is expressed as at least four different splice isoforms(1). The most abundantly expressed variant is VEGF165, which

contains a heparin-binding domain. Two VEGF receptor tyro-sine kinases, Flt-1 (VEGFR-1) and KDR (VEGFR-2), are ex-pressed on proliferating and quiescent endothelial cells (8).Although Flt-1 exhibits higher affinity for VEGF165, its func-tion in the adult vasculature is still poorly understood. Micelacking the complete Flt-1 gene display an increased number ofendothelial progenitors and vascular disorganization and die inutero at embryonic day 9 (9, 10). However, animals homozygousfor a deletion of the cytoplasmic domain are fertile and do notdisplay any obvious defects (11). Mice in which the flk-1/KDRgene has been inactivated also die at embryonic day 9. They aredeficient in vasculogenesis and also lack blood island formation(12).

Placenta growth factor (PlGF) shares 53% identity with theplatelet-derived growth factor-like domain of VEGF (13). Itbinds Flt-1 with high affinity but is unable to interact withKDR (14). PlGF exerts minimal effects on cell growth andmigration, suggesting that binding to Flt-1 alone is not suffi-cient to mediate these activities. In some instances, however,effects of PlGF on mitogenicity and MAP kinase activationhave been reported (15). In recent studies, the effects of PlGFon endothelial cells have been compared with those of anotherVEGF-like molecule, VEGForf. Members of the VEGForf genefamily have been isolated from the Orf parapoxyvirus, and theencoded proteins bind to KDR but not Flt-1 (16, 17). VEGForf

mediates migration of KDR-expressing PAE cells and cornealangiogenesis to an extent that is comparable with VEGF (17,18). However, the residues mediating VEGForf binding to KDRare unknown because the three basic amino acids comprisingthe major determinants of VEGF binding to the KDR receptorare absent in VEGForf. In addition, VEGForf does not possessthe heparin-binding domain found on VEGF. These differencesmay complicate conclusions about VEGF function, drawn fromstudies using VEGForf proteins. VEGF binding to KDR leads toreceptor phosphorylation and activation of MAP kinases aswell as tyrosine phosphorylation of PI3K and PLCg. AlthoughFlt-1 becomes phosphorylated and can activate several signal-ing molecules when expressed in heterologous cells (19), it isunclear whether these signaling events also occur in endothe-lial cells in response to Flt-1 engagement.

Recent studies have shown that KDR signaling results inendothelial nitric-oxide synthase up-regulation and activation(20). Nitric oxide plays a critical role in the VEGF-inducedendothelial cell proliferation, migration, and tube formation, aswell as increased vascular permeability, hypotension, and an-giogenesis in vivo (21–24). VEGF stimulates the formation ofvasodilator prostaglandins, which have been implicated as me-

* The costs of publication of this article were defrayed in part by thepayment of page charges. This article must therefore be hereby marked“advertisement” in accordance with 18 U.S.C. Section 1734 solely toindicate this fact.

** To whom correspondence should addressed: Dept. of MolecularOncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA94080. Tel.: 650-225-2968; Fax: 650-225-6327; E-mail: [email protected].

1 The abbreviations used are: VEGF, vascular endothelial growthfactor; PlGF, placenta growth factor; PLCg, phospholipase Cg; PI3K,phosphatidylinositol 3-kinase; HUVEC, human umbilical vein endothe-lial cell(s); PAE, porcine aortic endothelial cells; ERK, extracellularsignal-regulated kinase; MAP, mitogen-activated protein.

THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. 276, No. 5, Issue of February 2, pp. 3222–3230, 2001© 2001 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in U.S.A.

This paper is available on line at http://www.jbc.org3222

by guest on April 3, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 2: HE Printed in U.S.A. Analysis of Biological Effects and ... · Analysis of Biological Effects and Signaling Properties of Flt-1 (VEGFR-1) and KDR (VEGFR-2) A REASSESSMENT USING NOVEL

diators of VEGF-induced vascular permeability but not newblood vessel formation (21, 25). Recently, it was shown that aVEGF mutant chimeric protein that is unable to trigger KDRactivation can cause vascular permeability to an extent that is

similar to that of wild type VEGF (26). This led the authors toconclude that either Flt-1 or another as yet unknown receptoris responsible for the increase in vascular permeability in re-sponse to VEGF administration.

FIG. 1. Specificity and activity of KDR- and Flt-selective VEGF mutants. Competition of VEGF wild type and mutant protein binding with125I-VEGF to KDR. Binding of 125I-labeled VEGF was performed in the presence of the indicated concentrations of cold ligand. A, KDR bindingactivities. B, Flt-1 binding activities. C, KDR phosphorylation. The ability of mutant and wild type VEGF proteins to induce KDR phosphorylationwas analyzed over the indicated concentration rage. D, Flt-sel VEGF is biologically active. This mutant protein displays the same potency asVEGF165 in a competitive displacement of 125I-VEGF from transfected PAE cells expressing Flt(ANGG) (top panel). Flt-sel VEGF also promoteschemotaxis of PAE-Flt(ANGG) cells as efficiently as wild type VEGF, while KDR-sel VEGF is ineffective (bottom panel). Growth factors were addedat the indicated concentrations as described under “Experimental Procedures.” Cells were allowed to migrate for 14–16 h. Flt(ANGG) is a mutantFlt-1 receptor that, unlike wild type Flt-1, is able to mediate a motility response to VEGF (30). Data are expressed as stimulation index ofquadruplicate wells relative to basal, factor-independent migration. Bars represent the means 6 S.E.

KDR Mediates Angiogenesis and Vascular Permeability 3223

by guest on April 3, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 3: HE Printed in U.S.A. Analysis of Biological Effects and ... · Analysis of Biological Effects and Signaling Properties of Flt-1 (VEGFR-1) and KDR (VEGFR-2) A REASSESSMENT USING NOVEL

We sought to address these issues using novel, highly selec-tive, VEGF mutants generated by phage-display technology(27). Structure-function studies have demonstrated that VEGFinteracts differently with its cognate receptors KDR and Flt-1.Hence, it has been possible to generate VEGF mutants thatshow a strong preference for binding either one or the otherreceptor. Compared with an earlier generation of receptor spe-cific mutants (28), the novel mutants display a substantiallyincreased selectivity and thus are more likely to yield relevantresults pertaining to the function of each individual VEGFreceptor. By selectively activating Flt-1 or KDR in primaryendothelial cells, we have studied the involvement of eachindividual receptor in mediating VEGF signaling and cell mi-gration. KDR activation alone is sufficient for the activation ofsignal transducers involved in mitogenesis and cell migration.Selective KDR engagement also induces in vivo angiogenesisand vascular permeability.

EXPERIMENTAL PROCEDURES

Reagents—HUVEC were purchased from Cell Systems (Kirkland,WA). LY 294002 was purchased from Biomol. The construction andcharacterization of Flt-1- and KDR-selective VEGF are described by Liet al. (27). The mutations are as follows: KDR-selective VEGF D63S/G65M/L66R and Flt-selective VEGF I43A/I46A/Q79A/I83A. The muta-tions were introduced into VEGF165 and wild type VEGF, and thereceptor-selective mutants were expressed in Escherichia coli and pu-rified as described (28). The endotoxin content of the purified materialdid not exceed 0.2 Eu/mg.

Binding Assays—The determination of binding affinities of the re-ceptor-selective VEGF mutants was carried in solution as described (29)utilizing soluble IgG-Fc fusions of KDR and Flt-1 (14). To furthercharacterize the activities of the Flt-sel mutant, 125I-VEGF bindingassays were performed in PAE cells transfected with the mutant Flt-1receptor Flt(ANGG), as described previously (30). The PAE cell linelacks endogenous VEGF receptors (31).

Kinase Receptor Activation Assay—KDR phosphorylation was ana-lyzed in CHO cells stably expressing KDR with a C-terminal epitopetag. Kinase receptor activation assay assays were performed as de-scribed recently (32).

Cell Culture—Passage 4–7 HUVEC were maintained in CS-C me-dium (Cell Systems, Kirkland, WA) containing 10% fetal bovine serumand growth factors on gelatin-coated dishes and made quiescent by 14 hof starvation in 0.2% fetal bovine serum. Cells were treated as indicatedand washed once in ice-cold phosphate-buffered saline containing 0.1mM sodium orthovanadate. Cells were lysed in 0.5–1 ml of RIPA buffer(150 mM NaCl, 1% Nonidet P-40, 0.5% sodium deoxycholate, 50 mM

Tris, pH 8.0) containing 0.1 mM sodium orthovanadate, 5 mM para-nitrophenylphosphate, 10 mM sodium fluoride, 0.5 mM okadaic acid, anda protease inhibitor mixture (Roche MB 1836145). Culture of PAE cellswas performed as described previously (30, 31).

Immunoprecipitation—Protein A/G beads (Pierce) were blocked fornonspecific protein binding in 50 mM HEPES, pH 7.2, 0.1% TritonX-100, 150 mM NaCl, and 1 mg/ml ovalbumin for 30 min. Antibodieswere precoupled in the same buffer for 1 h at 4 °C with head-over-endrotation, and beads were washed three times in lysis buffer. Beads wereadded to the lysates and rotated overnight. Beads were washed sequen-tially in 50 mM Tris, pH 7.6, 150 mM NaCl, 1% Triton X-100, 1 mM

CaCl2; 50 mM Tris, pH 7.6, 500 mM NaCl, 0.1% Triton X-100, 1 mM

CaCl2; and 50 mM Tris, pH 7.6, 150 mM NaCl, 0.05% Triton X-100, 1 mM

CaCl2. Beads were resuspended in 23 sample buffer and boiled. Super-natants were applied directly to 4–12% Tris-glycine gradient gels(Novex).

Antisera—Anti-phospho-ERK antiserum was purchased from Pro-mega. Anti-phospho-p38 antibodies were from New England Biolabs.p85 was immunoprecipitated with antibodies from Transduction Labs(P13020) and Neomarkers (MS 424-P). A monoclonal antibody fromUpstate Biotechnologies, Inc. was used for the immunoprecipitation ofPLCg. The phosphotyrosine antibodies PY20 or E120H from Transduc-tion Labs were used.

HUVEC and PAE Cell Migration Assays—Migration assays wereperformed essentially as described previously (30). Falcon 8.0-mm filterinserts (Falcon 3097) were coated with type 1 collagen (VITROGEN,COHESION). PAE clones were grown in Ham’s F-12 medium, 10% fetalcalf serum, 0.25 mg/ml G418. HUVEC (obtained from Cell Systems,,p8) were grown in Cell Systems complete medium (4ZO-500) with 10%

fetal calf serum. Cells were trypsinized and transferred to endothelialbasal medium (Clonetics) with 0.1% bovine serum albumin for theassay. Cells were plated at 5 3 104/upper chamber. Growth factors wereplaced in the lower chamber, and inhibitors were in the upper chamber.Cells were allowed to migrate at 37 °C for 14–16 h. For the LY294002inhibitor experiments, cells were allowed to adhere for 30 min prior toaddition of the inhibitor. 20 min after inhibitor addition, VEGF wasadded to the bottom well, and the assay was allowed to proceed for only4 h to avoid the occurrence of apoptosis associated with the treatmentof these primary cells with LY294002.

Cells were removed from the upper side of the membrane by scrapingwith a polyurethane swab, and then the remaining cells on the bottomside of the membrane were fixed with methanol. Cells were stained withYo-Pro Iodide nuclear stain (Molecular Probes) and counted under lowpower fluorescence using an Image-Pro cell recognition program.

Corneal Pocket Angiogenesis Assay—Assays were performed as de-scribed previously (33), with the following modifications. Harlan Spra-gue-Dawley rats are anesthetized using a gas (isoflurane)/injectableketamine (80 mg/kg) xylazine (15 mg/kg) combination. The eyes aregently proptosed and secured in place using nontraumatic forceps. Witha number 15 blade, a 1.5-mm incision was made slightly below thecenter of the cornea. Using a microspatula (ST80017, ASSI), the inci-sion was carefully blunt-dissected through the stroma toward the outercanthus of the eye. A hydron-coated pellet (2 3 2 mm) containinggrowth factor (200 ng), methylcellulose, and aluminum sucralfate (100mg) was inserted into the base of the pocket. After surgery the eyes werecoated with gentamicin ointment. At day 6 the animals were injectedwith high molecular weight fluorescein isothiocyanate-dextran and eu-thanized to allow for visualization of the vasculature. Corneal wholemounts were made of the enucleated eyes and measurements ofneovascular area completed using computer-assisted image analysis(Image-Pro Plus).

Vascular Permeability Assay—Vascular permeability was assessedusing a modified Miles assay described previously (21, 25, 34–36).Hairless guinea pigs (600–800 g) were anesthetized by intramuscularadministration of xylazine (5 mg/kg) and ketamine (75 mg/kg). Theanimals then received an intracardiac injection of 1 ml of 1% Evans bluedye. Sixty minutes later, intradermal injections (0.1 ml) of VEGF or theappropriate VEGF mutant were made in a grid, which allowed us toconstruct a dose-response curve for each compound (n 5 4/molecule).VEGF165 was diluted in phosphate-buffered saline/bovine serum albu-min (1 mg/ml) and used at doses of 0, 0.3, 1, 3, 10, 30, and 100 ng/site.The dose of all other proteins was adjusted to inject equimolar amountsof protein. Sixty minutes after injections were made, animals wereeuthanized with pentobarbital (160 mg/kg IV), and their skin wasremoved, cleaned from connective tissues, and photographed. Quanti-fication of the dye extravasation area was carried out on the picturesusing an image analysis software (NIH Image®). For each dose, meas-urements were made in triplicate and averaged to minimize measure-ment errors.

RESULTS

Characterization of Receptor-selective Mutants—We set outto determine the different contributions of KDR and Flt-1 toVEGF signaling in primary endothelial cells. Because endothe-lial cells express both VEGF receptors, we made use of twonovel VEGF mutants engineered by the use of a phage displayselection process to bind selectively to either KDR or Flt-1 (27).These mutations were introduced into the VEGF165 cDNA andrecombinant proteins were produced in E. coli. The full-lengthVEGF mutant proteins were analyzed for their ability to bindto either Flt-1 or KDR in solution. Fig. 1A demonstrates therelative inability of the Flt-1-selective VEGF mutant (Flt-sel)to compete efficiently with wild type 125I-VEGF for binding toKDR. The reduction of binding of the KDR-selective mutant(KDR-sel2) to Flt-1 is shown in Fig. 1B. Table I summarizes theKd values and relative affinities of the individual mutants toFlt-1 and KDR. Binding of the KDR-selective variant to Flt-1 isreduced by a factor of 2000-fold, whereas Flt-selective VEGFbinds KDR 128-fold less well. To further characterize the spec-ificity of the mutants, we tested them with respect to theirability to induce KDR phosphorylation in intact cells (Fig. 1C).KDR-selective VEGF was essentially as potent and effective aswild type VEGF. Surprisingly, the ability of Flt-sel VEGF to

KDR Mediates Angiogenesis and Vascular Permeability3224

by guest on April 3, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 4: HE Printed in U.S.A. Analysis of Biological Effects and ... · Analysis of Biological Effects and Signaling Properties of Flt-1 (VEGFR-1) and KDR (VEGFR-2) A REASSESSMENT USING NOVEL

induce KDR phosphorylation was reduced by at least 4 ordersof magnitude. However, as illustrated in Fig. 1D, Flt-sel VEGFwas fully capable of competing for 125I-VEGF binding to theFlt-1 receptor even in the context of intact cells and was aspotent and effective as VEGF165 in promoting motility of PAEcells expressing Flt(ANGG). Unlike wild type Flt-1, this recep-tor mutant is able to mediate a KDR-like motility response toVEGF because of the removal of a repressor motif in the jux-tamembrane region (30). These findings argue against the pos-sibility that the inability of Flt-sel VEGF to induce KDR phos-phorylation may be due to inherent instability and/or lack ofactivity of the mutant protein.

Activation of MAP Kinases—Earlier work by this and otherlaboratories has generally shown that PlGF binding to Flt-1 isnot able to cause marked mitogenesis in endothelial cells (14,19), although evidence to the contrary has also been reported intransfected cell lines (15). We therefore tested whether KDR-selective VEGF was capable of mediating mitogenic signaling.As expected, activation by KDR-selective VEGF triggered phos-phorylation of ERK1 and ERK2 in HUVEC (Fig. 2A). Theextent of phosphorylation was indistinguishable from that ob-tained using wild type VEGF. Flt-1-selective VEGF at thehighest concentration used resulted in minimal phosphoryla-tion of ERK2. The homodimeric VEGF mutants utilized in thisstudy are not expected to promote receptor heterodimer forma-tion. Hence Flt-1 does not contribute to MAP kinase activation.

VEGF has been reported to stimulate the stress-activatedp38 MAP kinase (37, 38). To determine which VEGF receptor isinvolved, the phosphorylation status of p38 was analyzed afterstimulation with wild type Flt- and KDR-selective VEGF. Fig.2B demonstrates that only KDR-selective VEGF was able tostimulate p38 phosphorylation. However, as above indicated,Flt-selective VEGF was as effective as wild type VEGF165 ininducing migration of PAE cells expressing Flt(ANGG), dem-onstrating that this mutant is biologically active. Furthermore,in agreement with a previous report that implicates Flt-1 ac-

tivation in metalloproteinase release (39), Flt-selective VEGFstimulated the release of increased amounts of matrix metal-loprotease-9 proteolytic activity from human vascular smoothmuscle cells (27).

PI3K and PLCg Phosphorylation—PLCg phosphorylationand activation has been implicated in VEGF signaling. PLCgbinding to both KDR (40, 41) and Flt-1 (19, 42, 43) has beenreported. To determine which VEGF receptor(s) are involved inPLCg activation in primary endothelial cells, HUVEC weretreated with VEGF or VEGF receptor-selective mutants, andPLCg phosphorylation was assessed after immunoprecipita-tion (Fig. 3A). Both wild type and KDR-selective VEGF wereable to stimulate PLCg phosphorylation to a similar extent.Flt-selective VEGF did not increase PLCg phosphorylationover background levels, arguing against a role for Flt-1 in PLCgactivation in HUVEC.

PI3K has been demonstrated to transmit survival signalsthrough the activation of Akt in several cell types (44). VEGFalso acts as a survival factor for endothelial cells, and thissignal requires PI3K and Akt kinase activity (45). In a varietyof cell types, PI3K activity has been demonstrated to be in-volved in cytoskeletal changes following growth factor stimu-

FIG. 2. Activation of MAP kinases. Quiescent HUVEC were eitherleft untreated or stimulated with VEGF or VEGF mutant proteins for 5min. A, Western blots were probed for phosphorylated ERK1 and ERK2.B, the phosphorylation state of p38 stress-activated MAP kinase wasassessed with a phospho-specific antiserum. wt, wild type.

FIG. 3. KDR stimulates PLCg andPI3K phosphorylation. Quiescent HU-VEC were stimulated with 20 ng/ml of theindicated growth factors. A, PLCg was im-munoprecipitated from whole cell lysatesand analyzed for tyrosine phosphoryla-tion. B, lysates were immunoprecipitated(IP) with monoclonals antibodies againstp85 PI3K and tested for phosphotyrosine.wt, wild type.

TABLE IThe Kd values and relative binding affinities of wild type VEGF and

receptor-selective VEGF mutants

MutantsKDR binding FLT-1 binding

KdRelative toVEGF165

KdRelative toVEGF165

pM pM

V165 97 1 37 1KDR-sel2 100 1 87,000 2,000Flt-sel 12,416 128 98 2.6

KDR Mediates Angiogenesis and Vascular Permeability 3225

by guest on April 3, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 5: HE Printed in U.S.A. Analysis of Biological Effects and ... · Analysis of Biological Effects and Signaling Properties of Flt-1 (VEGFR-1) and KDR (VEGFR-2) A REASSESSMENT USING NOVEL

lation as well as cell migration (46, 47). Therefore, the ability ofthe VEGF proteins to cause phosphorylation of the p85 regu-latory subunit of PI3K was assessed after immunoprecipita-tion. Only wild type and KDR-selective VEGF were capable ofcausing phosphorylation the PI3K regulatory subunit (Fig. 3B).

Effect of Receptor-selective VEGF Mutants on EndothelialCell Migration—One of the central aspects of VEGF action onendothelial cells is its ability to act as a chemoattractant andstimulate the migration of endothelial cells. Fig. 4A shows theeffect of receptor-selective VEGF on HUVEC in a modifiedBoyden chamber assay. In several independent experiments,VEGF caused a 4–5-fold increase in HUVEC migration. KDR-selective VEGF is as effective as wild type VEGF in the pro-motion of HUVEC migration. Flt-selective VEGF is unable toincrease cell migration over background levels.

To determine the contribution of PI3K to endothelial cellmigration, different concentrations of the inhibitor LY 294002were added to the assay after the cells had been allowed toattach to the membrane. Because of the deleterious effects ofPI3K inhibition on endothelial cell survival a short-term assay

was performed (see “Experimental Procedures”). Fig. 4B showsthat at its highest concentration, and LY 294002 caused a 56%inhibition of HUVEC migration. Thus, PI3K activity contrib-utes significantly to endothelial cell migration.

KDR, but Not Flt-1, Signaling Causes in Vivo Angiogenesis—Because endothelial cells in adult organisms express both KDRand Flt-1 and previous approaches that interfere with in vivoangiogenesis have been targeted at interfering with VEGFbinding to both receptors, we sought to identify the receptor(s)responsible for in vivo angiogenesis. Hydron pellets containing200 ng of growth factors were implanted into rat corneas, andthe angiogenic areas were evaluated after 1 week (Fig. 5A).KDR-selective VEGF was as efficient as wild type VEGF ininducing corneal angiogenesis. Although Flt-selective VEGFoccasionally induced marginal angiogenesis (Fig. 5A), analysisof the angiogenic surface areas in several animals showed thatFlt-selective VEGF was unable to stimulate angiogenesis overcontrol levels. PlGF gave only a marginal response (Fig. 5B).Therefore, only KDR and not Flt-1 is capable of promotingangiogenesis in vivo.

FIG. 4. Analysis of HUVEC migra-tion. HUVEC migration was analyzed ina modified Boyden chamber assay. Exper-iments were performed in triplicate. Er-ror bars represent the standard error. A,HUVEC migration in response to the in-dicated concentrations of wild type, Flt-selective, and KDR-selective VEGF. B,PI3K inhibition impairs HUVEC migra-tion in response to wild type VEGF. wt,wild type.

KDR Mediates Angiogenesis and Vascular Permeability3226

by guest on April 3, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 6: HE Printed in U.S.A. Analysis of Biological Effects and ... · Analysis of Biological Effects and Signaling Properties of Flt-1 (VEGFR-1) and KDR (VEGFR-2) A REASSESSMENT USING NOVEL

Increased Vascular Permeability Is Mediated through KDRbut Not Flt-1 Activation—Similarly, we sought to determinethe relative importance of KDR and Flt-1 receptors for theVEGF-induced vascular permeability. Vascular permeabilitywas assessed by the Miles assay, and responses were evaluated60 min following the intradermal administration of VEGF andVEGF receptor-selective mutants. KDR-selective VEGF in-duced vascular permeability to a comparable extent as wildtype VEGF, whereas Flt-1 receptor-selective VEGF caused es-sentially no leakage (Fig. 6A). The results obtained in fouranimals are summarized in Fig. 6B. In additional experiments,we tested the Flt-1-selective mutant at much higher concentra-tions (up to 30 mg/site), and still no extravasation was observed(data not shown). Therefore, VEGF-induced vascular perme-ability is mediated by KDR receptor binding. To further con-firm these results, we compared the vascular permeability ofVEGF165 and the Flt-1-selective ligand PlGF. To rule out thepossibility that differences in Flt-1 receptor binding affinitybetween VEGF and PlGF could explain the lack of vascularpermeability induced by PlGF, we used PlGF at equimolardoses and at doses 20-fold higher relative to those of VEGF.Whereas VEGF induced a dose-dependent dye extravasation,PlGF did not induce any vascular permeability regardless ofwhether we used equimolar or 20-fold higher doses relative to

those of VEGF consistent with previous reports (Refs. 14 and48 and Fig. 6C). These results demonstrate the critical role ofKDR receptor activation in the VEGF-induced vascular perme-ability response.

DISCUSSION

The expression of at least two VEGF receptor tyrosine ki-nases on endothelial cells has made it difficult to elucidate theindividual contributions of each receptor to VEGF signaling.Although PlGF binds preferentially to Flt-1, its use has some-times led to conflicting results especially when heterologouscell types such as NIH3T3 fibroblasts were used (15, 19).

This study explores whether the activities of both Flt-1 andKDR are required to mediate VEGF functions in primary en-dothelial cells and in vivo. To this end, we employed ho-modimers of receptor-selective VEGF mutants. Flt-selectiveVEGF was unable to generate a mitogenic signal, as evidencedalso by its inability to stimulate ERK activity. This result is inagreement with previous work that examined the abilities ofearlier receptor-selective mutants to promote endothelial cellproliferation (28). Interestingly, although the novel Flt-1-selec-tive mutant demonstrated a 128-fold selectivity using solubleIgG chimeric receptors, in a kinase receptor activation assay

FIG. 5. Angiogenic effects of VEGFmutants in the rat cornea. A, repre-sentative examples of the extent of cor-neal angiogenesis in response to wildtype, Flt-selective, and KDR-selectiveVEGF. B, quantitative analysis of thesurface areas of corneal angiogenesiscaused by wild type and mutant VEGFproteins as well as PlGF.

KDR Mediates Angiogenesis and Vascular Permeability 3227

by guest on April 3, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 7: HE Printed in U.S.A. Analysis of Biological Effects and ... · Analysis of Biological Effects and Signaling Properties of Flt-1 (VEGFR-1) and KDR (VEGFR-2) A REASSESSMENT USING NOVEL

format it was virtually devoid of the ability to induce KDRphosphorylation, even at concentrations exceeding 10,000 ng/ml. Therefore, the receptor binding assays largely underesti-mated the extent of specificity of this mutant that may beobtained in a biological context. There are various examples oflack of good correlation between binding affinity and biologicalactivity of mutant proteins (49). In the case of our mutant, oneof the possible explanations for the discrepancy is that thereduced affinity leads to only a transient existence of ligand-receptor complexes, below a critical threshold for effective re-

ceptor phosphorylation and dimerization. Interestingly, a“minimum residency time threshold” for effective phosphoryl-ation has been recently described for the prolactin receptor(50). However, it was important to verify that Flt-sel VEGF iscapable of exerting the appropriate biological activity. Thismutant is able to compete for 125I-VEGF binding in PAE cellsexpressing Flt(ANGG) with a dose response nearly identical tothat of VEGF165, demonstrating its ability to bind Flt-1 notonly in soluble receptor assays but also in the context of intactcells. Furthermore, this binding results in stable receptor acti-

FIG. 6. VEGF-induced vascular permeability is mediated by binding to KDR receptors. A, representative pictures of skin of guinea pigsused in the Miles assay showing the vascular permeability in response to wild type VEGF165, and KDR receptor-selective VEGF mutant, and thelack of effect of the Flt receptor-selective VEGF mutant. B, dose-response curves for VEGF and receptor-selective VEGF mutants. Error barsrepresent S.E. C, comparison of the vascular permeabilty response obtained with equimolar doses of VEGF and PlGF and a 20-fold higher dose ofPlGF over VEGF.

KDR Mediates Angiogenesis and Vascular Permeability3228

by guest on April 3, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 8: HE Printed in U.S.A. Analysis of Biological Effects and ... · Analysis of Biological Effects and Signaling Properties of Flt-1 (VEGFR-1) and KDR (VEGFR-2) A REASSESSMENT USING NOVEL

vation, as assessed by the ability of Flt-sel VEGF to effectivelypromote migration of PAE cells expressing Flt(ANGG) in anovernight assay. Although wild type Flt-1 fails to transmit amotility signal (30, 31), the juxtamembrane mutant Flt(ANGG)is able to mediate VEGF-dependent migration in transfectedPAE cells, similar to that mediated by KDR (30). Additionally,as previously mentioned, Flt-selective VEGF has been shown tostimulate the release of matrix metalloprotease-9 proteolyticactivity from cultured human vascular smooth muscle cells(27).

Activation of the KDR receptor was also sufficient to causecell migration in HUVEC. Therefore, we also examined theactivation of proteins previously reported to be involved in themigratory response of normal endothelial cells. Both PI3K andPLCg phosphorylation were stimulated to the same extent bywild type VEGF and KDR-selective VEGF. HUVEC migrationwas inhibited by treatment with the PI3K inhibitor LY294002,demonstrating a prominent role for PI3K in endothelial cellmigration. This is consistent with results obtained for other celltypes, such as epithelial cells and fibroblasts, whereas PI3Kactivity is required for cytoskeletal changes and cell migration(46, 47).

Recently, the properties of novel VEGF family memberspresent in Orf parapoxyviruses have been analyzed (VEGForf).VEGForf is a distant relative of VEGF-A. It lacks a heparin-binding domain and binds preferentially to KDR. Additionally,VEGForf features a threonine- and proline-rich C-terminal do-main of unknown function that is not present in any othermammalian VEGF. VEGForf mediates endothelial cell mitogen-esis and migration of KDR-expressing PAE cells as well ascorneal angiogenesis to an extent that is comparable withVEGF165 (18, 43). Our rationally designed KDR-selectiveVEGF should be more representative for the functions of mam-malian VEGFs, because it is largely unchanged from wild typeVEGF, and only its ability to interact with Flt-1 has beenremoved.

Because our Flt-1-selective VEGF mutant shows substan-tially reduced binding and little or no ability to activate KDR,we tested whether this highly selective protein could inducedye extravasation. The absence of extravasation following ad-ministration of the Flt-1 receptor-selective mutant and thecomparable extent of vascular permeability caused by wild typeVEGF and the KDR receptor-selective mutant indicates thatVEGF-induced vascular permeability is solely mediatedthrough binding of VEGF to the KDR receptor.

However, a chimeric mutant of mouse VEGF, which showspreferential binding to Flt-1, has recently been reported tostimulate vascular permeability, leading the authors (26) toconclude that KDR activation is not required to mediate thisbiological activity of VEGF. These conflicting conclusions maybe explained, at least in part, by the fact that the chimericmurine VEGF mutant was obtained by site-directed mutagen-esis in which amino acids 83–89 were substituted by analogousregions of PlGF (26). The differences in structure and/or signaltransduction between VEGF and PlGF may be responsible inpart for this discrepancy. Furthermore, it is possible that thehigh local concentrations of the injected protein employed inthe vascular permeability assay (up to 1 mg/ml) require agreater degree of specifity of the mutant VEGF protein thanthat afforded by such chimeric mutant, to achieve meaningfulresults. In support of this possibility, our earlier generationFlt-1-selective mutants (28), which display a lower degree ofreceptor selectivity than the mutant used in the present study,are able to cause as much as 50% of the vascular permeabilityresponse as wild type VEGF (data not shown). Furthermore,the finding that our KDR-selective mutant, which exhibits

2000-fold selectivity for KDR versus Flt-1, is fully competent tocause vascular permeability provides strong evidence in favorof the involvement of KDR. Interestingly, a KDR-selectiveVEGF mutant in the background of the receptor-binding do-main (27) was also fully competent in promoting vascular per-meability (data not shown), indicating that the heparin-bind-ing domain has little or no contribution to this activity. The roleof the KDR receptor in mediating vascular permeability issupported by other studies that describe a mutant VEGF-C,which has lost both KDR receptor binding and vascular perme-ability activities (34) and a VEGF molecule produced by Orfviruses, which binds KDR but not Flt-1 receptors and inducesvascular permeability (16, 18). Additionally, PlGF fails to in-duce vascular permeability (14, 48). Therefore, we concludethat binding of VEGF to KDR is necessary and sufficient toinduce vascular permeability. Similar to the results obtained inthe in vivo angiogenesis experiments, a contribution from Flt-1is not required to elicit this biological effect.

Our findings ascertain that KDR can be the sole mediator ofVEGF-induced migration, angiogenesis and permeability inendothelial cells. Furthermore, these VEGF mutants shouldprovide especially valuable tools to further dissect Flt-1 andKDR function in vivo through the use in transgenic models,where the protein is overexpressed. Their high selectivity overa broad concentration range may be critical to correctly definethe VEGF receptor biology in such circumstances.

Acknowledgments—We thank Abraham M. de Vos for helpful discus-sions and Gloria Meng, Jane Lee, and Michael Sadick for assay support.

REFERENCES

1. Ferrara, N. (1999) Kidney Int. 56, 794–8142. Risau, W. (1997) Nature 386, 671–6743. Kim, K. J., Li, B., Winer, J., Armanini, M., Gillett, N., Phillips, H. S., and

Ferrara, N. (1993) Nature 362, 841–8444. Millauer, B., Shawver, L. K., Plate, K. H., Risau, W., and Ullrich, A. (1994)

Nature 367, 576–5795. Millauer, B., Longhi, M. P., Plate, K. H., Shawver, L. K., Risau, W., Ullrich, A.,

and Strawn, L. M. (1996) Cancer Res. 56, 1615–16206. Borgstrom, P., Hillan, K. J., Sriramarao, P., and Ferrara, N. (1996) Cancer

Res. 56, 4032–40397. Ferrara, N., and Alitalo, K. (1999) Nat. Med. 5, 1359–13648. Ferrara, N., and Davis-Smyth, T. (1997) Endocr. Rev. 18, 4–259. Fong, G. H., Zhang, L., Bryce, D. M., and Peng, J. (1999) Development 126,

3015–302510. Fong, G. H., Rossant, J., Gertsenstein, M., and Breitman, M. L. (1995) Nature

376, 66–7011. Hiratsuka, S., Minowa, O., Kuno, J., Noda, T., and Shibuya, M. (1998) Proc.

Natl. Acad. Sci. U. S. A. 4, 9349–935412. Shalaby, F., Rossant, J., Yamaguchi, T. P., Gertsenstein, M., Wu, X. F.,

Breitman, M. L., and Schuh, A. C. (1995) Nature 376, 62–6613. Maglione, D., Guerriero, V., Viglietto, G., Delli-Bovi, P., and Persico, M. G.

(1991) Proc. Natl. Acad. Sci. U. S. A. 88, 9267–927114. Park, J. E., Chen, H. H., Winer, J., Houck, K. A., and Ferrara, N. (1994) J. Biol.

Chem. 269, 25646–2565415. Landgren, E., Schiller, P., Cao, Y., and Claesson-Welsh, L. (1998) Oncogene 16,

359–36716. Wise, L. M., Veikkola, T., Mercer, A. A., Savory, L. J., Fleming, S. B., Caesar,

C., Vitali, A., Makinen, T., Alitalo, K., and Stacker, S. A. (1999) Proc. Natl.Acad. Sci. U. S. A. 96, 3071–3076

17. Meyer, M., Clauss, M., Lepple-Wienhues, A., Waltenberger, J., Augustin,H. G., Ziche, M., Lanz, C., Buttner, M., Rziha, H. J., and Dehio, C. (1999)EMBO J. 18, 363–374

18. Ogawa, S., Oku, A., Sawano, A., Yamaguchi, S., Yazaki, Y., and Shibuya, M.(1998) J. Biol. Chem. 273, 31273–31282

19. Seetharam, L., Gotoh, N., Maru, Y., Neufeld, G., Yamaguchi, S., and Shibuya,M. (1995) Oncogene 10, 135–147

20. Shen, B. Q., Lee, D. Y., and Zioncheck, T. F. (1999) J. Biol. Chem. 274,33057–33063

21. Fujii, E., Irie, K., Ohba, K., Ogawa, A., Yoshioka, T., Yamakawa, M., andMuraki, T. (1997) Naunyn-Schmiedebergs Arch. Pharmacol. 356, 475–480

22. Papapetropoulos, A., Garcia-Cardena, G., Madri, J. A., and Sessa, W. C. (1997)J. Clin. Invest. 100, 3131–3139

23. Hood, J. D., Meininger, C. J., Ziche, M., and Granger, H. J. (1998) Am. J.Physiol. 274, H1054–H1058

24. Ziche, M., Morbidelli, L., Choudhuri, R., Zhang, H. T., Donnini, S., Granger,H. J., and Bicknell, R. (1997) J. Clin. Invest. 99, 2625–2634

25. Murohara, T., Asahara, T., Silver, M., Bauters, C., Masuda, H., Kalka, C.,Kearney, M., Chen, D., Symes, J. F., Fishman, M. C., Huang, P. L., andIsner, J. M. (1998) J. Clin. Invest. 101, 2567–2578

26. Stacker, S. A., Vitali, A., Caesar, C., Domagala, T., Groenen, L. C., Nice, E.,Achen, M. G., and Wilks, A. F. (1999) J. Biol. Chem. 274, 34884–34892

KDR Mediates Angiogenesis and Vascular Permeability 3229

by guest on April 3, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 9: HE Printed in U.S.A. Analysis of Biological Effects and ... · Analysis of Biological Effects and Signaling Properties of Flt-1 (VEGFR-1) and KDR (VEGFR-2) A REASSESSMENT USING NOVEL

27. Li, B., Fuh, G., Meng, G., Xin, X., Gerritsen, M. E., Cunningham, B., andde Vos, A. M. (2000) J. Biol. Chem. 275, 29823–29828

28. Keyt, B. A., Nguyen, H. V., Berleau, L. T., Duarte, C. M., Park, J., Chen, H.,and Ferrara, N. (1996) J. Biol. Chem. 271, 5638–5646

29. Cunningham, B. C., Lowe, D. G., Li, B., Bennett, B. D., and Wells, J. A. (1994)EMBO J. 13, 2508–2515

30. Gille, H., Kowalski, J., Yu, L., Chen, H., Pisabarro, M. T., Davis-Smyth, T.,Ferrara, N.(2000) EMBO J. 15, 4064–4073

31. Waltenberger, J., Claesson-Welsh, L., Siegbahn, A., Shibuya, M., Heldin,C.-H.(1994) J. Biol. Chem. 269, 26988–26995

32. Sadick, M. D., Intintoli, A., Quarmby, V., McCoy, A., Canova-Davis, E., andLing, V. (1999) J. Pharm. Biomed. Anal. 19, 883–891

33. Polverini, P. J., Bouck, N. P., and Rastinejad, F. (1991) Methods Enzymol. 198,440–450

34. Senger, D. R., Galli, S. J., Dvorak, A. M., Perruzzi, C. A., Harvey, V. S., andDvorak, H. F. (1983) Science 219, 983–985

35. Park, J. E., Keller, H.-A., and Ferrara, N. (1993) Mol. Biol. Cell 4, 1317–132636. Joukov, V., Kumar, V., Sorsa, T., Arighi, E., Weich, H., Saksela, O., and

Alitalo, K. (1998) J. Biol. Chem. 273, 6599–660237. Rousseau, S., Houle, F., Landry, J., and Huot, J. (1997) Oncogene 15,

2169–217738. Yu, Y., and Sato, J. D. (1999) J. Cell. Physiol. 178, 235–246

39. Wang, H., and Keiser, J. A. (1998) Circulation Res. 83, 832–84040. Dougher, M., and Terman, B. I. (1999) Oncogene 18, 1619–162741. Cunningham, S. A., Arrate, M. P., Brock, T. A., and Waxham, M. N. (1997)

Biochem. Biophys. Res. Commun. 240, 635–63942. Sawano, A., Takahashi, T., Yamaguchi, S., and Shibuya, M. (1997) Biochem.

Biophys. Res. Commun. 238, 487–49143. Ito, N., Wernstedt, C., Engstrom, U., and Claesson-Welsh, L. (1998) J. Biol.

Chem. 273, 23410–2341844. Marte, B. M., and Downward, J. (1997) Trends Biochem. Sci. 22, 355–35845. Gerber, H. P., McMurtrey, A., Kowalski, J., Yan, M., Keyt, B. A., Dixit, V., and

Ferrara, N. (1998) J. Biol. Chem. 273, 30366–3034346. Wennstrom, S., Hawkins, P., Cooke, F., Hara, K., Yonezawa, K., Kasuga, M.,

Jackson, T., Claesson-Welsh, L., and Stephens, L. (1994) Curr. Biol. 4,385–393

47. Toker, A., and Cantley, L. C. (1997) Nature 387, 673–67648. Murohara, T., Horowitz, J. R., Silver, M., Tsurumi, Y., Chen, D., Sullivan, A.,

and Isner, J. M. (1998) Circulation 97, 99–10749. Lokker, N. A., Mark, M. R., Luis, E. A., Bennett, G. L., Robbins, K. A., Baker,

J. B., and Godowski, P. J. (1992) EMBO J. 11, 2503–251050. Elkins, P. A., Christinger, H. W., Gertler, A., Sandowski, Y., Sakal, E., de Vos,

A. M., and Kossiakoff, A. A. (2000) Nat. Struct. Biol., 7, 808–815

KDR Mediates Angiogenesis and Vascular Permeability3230

by guest on April 3, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 10: HE Printed in U.S.A. Analysis of Biological Effects and ... · Analysis of Biological Effects and Signaling Properties of Flt-1 (VEGFR-1) and KDR (VEGFR-2) A REASSESSMENT USING NOVEL

Zioncheck, Nicolas Pelletier and Napoleone FerraraHendrik Gille, Joe Kowalski, Bing Li, Jennifer LeCouter, Barbara Moffat, Thomas F.

VASCULAR ENDOTHELIAL GROWTH FACTOR MUTANTS(VEGFR-2): A REASSESSMENT USING NOVEL RECEPTOR-SPECIFIC

Analysis of Biological Effects and Signaling Properties of Flt-1 (VEGFR-1) and KDR

doi: 10.1074/jbc.M002016200 originally published online October 31, 20002001, 276:3222-3230.J. Biol. Chem. 

  10.1074/jbc.M002016200Access the most updated version of this article at doi:

 Alerts:

  When a correction for this article is posted• 

When this article is cited• 

to choose from all of JBC's e-mail alertsClick here

  http://www.jbc.org/content/276/5/3222.full.html#ref-list-1

This article cites 50 references, 20 of which can be accessed free at

by guest on April 3, 2020

http://ww

w.jbc.org/

Dow

nloaded from