当前位置: 首页 > 期刊 > 《内分泌学杂志》 > 2006年第3期 > 正文
编号:11416490
Transactivation of Fetal Liver Kinase-1/Kinase-Insert Domain-Containing Receptor by Lysophosphatidylcholine Induces Vascular Endot
http://www.100md.com 《内分泌学杂志》
     Department of Pharmacology (Y.F., M.Y., Y.I., N.A., H.O., Y.K., K.I., T.T.), Graduate School of Medical Sciences

    Department of Clinical Pharmacology (K.T.), Graduate School of Pharmaceutical Sciences, The University of Tokushima Graduate School, Tokushima 770-8503, Japan

    Abstract

    Lysophosphatidylcholine (LPC), a major lipid component of oxidized low-density lipoprotein, is a bioactive lipid molecule involved in numerous biological processes including the progression of atherosclerosis. Recently orphan G protein-coupled receptors were identified as high-affinity receptors for LPC. Although several G protein-coupled receptor ligands transactivate receptor tyrosine kinases, LPC-stimulated transactivation of receptor tyrosine kinase has not yet been reported. Here we observed for the first time that LPC treatment of human umbilical vein endothelial cells (HUVECs) induces tyrosyl phosphorylation of vascular endothelial growth factor receptor 2 [fetal liver kinase-1/kinase-insert domain-containing receptor, Flk-1/KDR)]. Flk-1/KDR transactivation by LPC was inhibited by vascular endothelial growth factor receptor tyrosine kinase inhibitors, SU1498 and 4-[(4'-chrolo-2'-fluoro) phenylamino]6,7-dimethoxyquinazoline (VTKi) in immunoprecipitation. Furthermore, we examined the effects of the Src family kinases inhibitors, herbimycin A and 4-amino-5-(4-chlorophenyl)-7-(t-butyl) pyrazolo[3,4-d] pyrimidine (PP2), on LPC-induced Flk-1/KDR transactivation. Results from Western blots, c-Src is involved in LPC-induced Flk-1/KDR transactivation because herbimycin A and PP2 inhibited this transactivation. Kinase-inactive (KI) Src transfection also inhibited LPC-induced Flk-1/KDR transactivation. In addition, results from Western blots, ERK1/2 and Akt, which are downstream effectors of Flk-1/KDR, were also activated by LPC, and this was inhibited by SU1498, VTKi, herbimycin A, PP2, and KI Src transfection in HUVECs. LPC-induced stimulation of HUVEC proliferation was shown to be secondary to transactivation because it was suppressed by SU1498, VTKi, herbimycin A, PP2, and KI Src transfection in dimethylthiazoldiphenyltetra-zoliumbromide assay. These findings suggest that LPC-induced Flk-1/KDR transactivation via c-Src may have important implications for the progression of atherosclerosis.

    Introduction

    LYSOPHOSPHATIDYLCHOLINE (LPC) is known to induce a variety of vascular endothelial responses ranging from the up-regulation of adhesion molecules and growth factors to the secretion of chemokines and superoxide anion radicals (1, 2). As a component of oxidized low-density lipoprotein (oxLDL), LPC is locally generated and accumulates at the sites of wounds, inflammation, and atherosclerosis (3, 4). Atherosclerosis can be characterized as a chronic inflammatory disease in which both cell proliferation and apoptotic/necrotic cell death occur within the vascular wall. In atherosclerotic lesions, pathological examination reveals the presence of angiogenesis (5, 6). It has previously been shown that LPC induces growth factor gene expression in cultured human endothelial cells (7). It has also been shown that oxLDL and LPC induces endothelial proliferation (8).

    Recently orphan G protein-coupled receptors (GPCRs) were identified as high-affinity receptors for LPC. LPC binds to G2A and GPR4, Gi-protein-coupled receptors, specifically and regulates both cell growth and immunologic responses (9, 10). Gs-protein-coupled receptor GPR119 is reported to be a novel LPC receptor involved in insulin secretion (11). LPC, interacting with its receptor, induces an elevation of Ca2+ concentration activates serum-responsive transcription factors via the MAPK kinase pathway (10, 12). Despite the continued accumulation of evidence, however, the exact mechanisms by which LPC exerts biological function in endothelial cells (ECs) have not yet been elucidated.

    Transactivation of receptor tyrosine kinases (RTKs) by the binding of ligands to GPCRs has been shown to have important physiological consequences. GPCR-mediated RTK transactivation has been implicated to have a crucial role in diseases such as cardiac hypertrophy and cancer and may have an important role in vascular diseases as well (13, 14). The role of Ca2+, reactive oxygen species, cSrc, Pyk2, protein kinase C, and membrane-bound metalloproteases has been reported in GPCR-mediated transactivation of RTKs (15, 16, 17, 18). Among these intracellular and extracellular molecules, a role for c-Src tyrosine kinase in GPCR-mediated RTK transactivation has been the focus (16, 19). LPC has been reported as a regulator of tyrosine kinase activity (20, 21). In these investigations, c-Src is suggested to have an important role in LPC signaling. It has been reported that transactivation of RTK in response to stimulation of GPCRs induces MAPK and Akt activation (15, 19). Vascular endothelial growth factor (VEGF) receptor-2 [fetal liver kinase-1/kinase-insert domain-containing receptor (Flk-1/KDR)] is a major receptor tyrosine kinase transducing the effects of VEGF into ECs. The signaling of Flk-1/KDR is necessary for the execution of VEGF-stimulated proliferation as well as the survival of cultured ECs and has been shown to be involved in atherosclerosis. Therefore, we hypothesized that LPC may transactivate Flk-1/KDR and investigated the specific role of LPC-induced Flk-1/KDR activation in ECs.

    In the present study, we examined whether LPC transactivates Flk-1/KDR in human umbilical vein endothelial cells (HUVECs). Thereafter we investigated the involvement of c-Src tyrosine kinase in Flk-1/KDR transactivation by LPC. The findings of the present study strongly suggest that the transactivation of Flk-1/KDR by LPC is mediated by c-Src in HUVECs. The influence of Flk-1/KDR transactivation on the activation of ERK 1/2 and Akt, which are downstream effectors of Flk-1/KDR, were also examined. In addition, it was observed that LPC caused HUVEC proliferation, which may be related to the progression of atherosclerosis.

    Materials and Methods

    Chemicals

    LPC (C18:0) and wortmannin were purchased from Sigma (St. Louis, MO). Recombinant human VEGF was from PepRo Tech EC, Ltd. (London, UK). VEGF receptor tyrosine kinase inhibitors, VTKi [(4-[4'-chrolo-2'-fluoro]phenylamino)6,7-dimethoxyquinazoline] and SU1498 [(E)-3-(3,5-diisopropyl-4-hydroxyphenyl)-2-[(3-phenyl-n-propyl) amino-carbonyl] acrylonitrile], and Src family tyrosine kinase inhibitors 4-amino-5-(4-chlorophenyl)-7-(t-butyl) pyrazolo [3,4-d] pyrimidine (PP2) and herbimycin A were from Calbiochem (San Diego, CA). MAPK kinase (MEK) 1/2 inhibitors PD98059 was from Wako Pure Chemical Industries, Ltd. (Osaka, Japan), and U0126 was from Promega (Madison, WI). Anti-Flk-1/KDR polyclonal antibody, protein A/G PLUS-agarose beads, and anti-ERK1/2 antibody were from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA). Antiphosphotyrosine, clone 4G10, and anti-Src antibody were from Upstate Biotechnology Inc. (Lake Placid, NY). Anti-phospho-ERK1/2 (Thr 202/Tyr 204) antibody, anti-phospho-Akt (Ser 473) antibody, and anti-Akt antibody were from Cell Signaling Technology Inc. (Beverly, MA). Anti-Src phospho-specific antibody (Tyr 418), which recognizes the activated form of c-Src, was from Biosource (Camarillo, CA). All other chemicals were of reagent grade, obtained from commercial sources, and used without further purification.

    Cell culture and transient transfection

    HUVECs and endothelial cell basal medium (EBM)-2 were purchased from Clonetics (San Diego, CA). HUVECs were cultured in EBM-2 supplemented with 10% fetal bovine serum (FBS), gentamicin sulfate (50 μg/ml), and amphotericin-B (50 ng/ml) in addition to human fibroblast growth factor-B (10 ng/ml), epidermal growth factor human recombinant in a buffered BSA saline solution (20 ng/ml), VEGF human recombinant (1 ng/ml), IGF-I in aqueous solution cell culture tested (1 ng/ml), ascorbic acid (1 μg/ml), heparin (3 ng/ml), and hydrocortisone (0.4 μg/ml) at 37 C and 5% CO2. For transfection of wild-type (WT) or kinase-inactive (KI) Src, commercially available pUSE mammalian expression vectors encoding pp60c-Src or catalytically inactive Src (K297R) were used (Upstate Biotechnology). For the transient expression experiments, HUVECs were transfected with CytoPure-huv transfection reagent (polyplus-transfection, QBIOGENE Inc., Carlsbad, CA) according to the manufacture’s instructions. Transfection efficiency was determined with pcDNA3.1-GFP transfection as about 30% in HUVECs.

    Immunoblot analysis

    HUVECs in 0.2% FBS-containing EBM-2 medium were treated with or without inhibitors for various times and then incubated with LPC. After treatment with reagents, the cells were washed twice with cold PBS. Thereafter the cells were harvested using 0.5 ml of lysis buffer [20 mM Tris-HCl (pH 7.4), 150 mM NaCl, 1 mM Na2EDTA, 1 mM EGTA, 1% Triton X-100, 2.5 mM sodium pyrophosphate, 1 mM -glycerophophosphate, 1 mM sodium orthovanadate, 1 μg/ml leupeptin, 1 mM phenylmethylsulfonylfluoride] and incubated on ice. After thawing, cells were harvested and cell lysates were sonicated (Handy Sonic UR-20 P, Tomy Seiko Co. Ltd., Tokyo, Japan) on ice for 15 sec and then centrifuged at 25,000 x g for 20 min at 4 C to precipitate cell debris. The supernatant of lysates was analyzed for protein concentration by the Bradford methods (Bio-Rad Laboratories, Hercules, CA), and equal amounts of cellular proteins were separated by SDS-PAGE. After transfer to nitrocellulose membrane, the activation levels of ERK1/2, Akt, and Src were examined using antiphosphospecific antibodies, as described previously (22, 23).

    Immunoprecipitation

    Lysates containing equal amounts of protein were incubated with anti-Flk-1/KDR antibody overnight and then incubated with protein A/G PLUS-agarose beads for 2 h on a roller system at 4 C. After immunoprecipitation, samples were evaluated with immunoblot assay using antiphosphotyrosine antibody (clone 4G10). Based on its 230-kDa molecular mass and tyrosyl phosphorylation, it was considered that the bands represent activated state of Flk-1/KDR as described previously (17).

    Cell viability assay

    When HUVECs reached 40–50% confluence in 35-mm collagen-coated dishes (IWAKI, Osaka, Japan), growth was arrested using EBM-2 medium with 0.2% FBS for 24 h. SU1498, VTKi, PD98059, U0126, and herbimycin A were added and incubated for 30 min. PP2 was added and incubated for 2 h. After 24 h incubation with LPC (20 μM), 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H tetrazolium bromide (MTT) was added at a final concentration of 0.5 mg/ml, and after a further 2 h incubation, HUVECs were lysed with isopropanol containing 0.04 M HCl. MTT reduction was read at 550 nm by a spectrophotometer.

    Statistical analysis

    Values are presented as means ± SD for five separate experiments. One-way ANOVA was used to determine significance among groups, after which post hoc test with the Bonferroni correction were used for comparison between individual groups. A value at P < 0.05 was considered to be significant.

    Results

    LPC stimulates transactivation of Flk-1/KDR in HUVECs

    We first examined whether Flk-1/KDR is activated by LPC in HUVECs. HUVECs were treated at various times and with various concentrations of LPC. Activation of Flk-1/KDR in the cell lysates was determined by tyrosyl phosphorylation as described in Materials and Methods. As shown in Fig. 1A, LPC rapidly activated Flk-1/KDR (with a peak at 10 min) and then sustained activation for at least 60 min. Figure 1B shows the dose response for the activation of Flk-1/KDR by LPC in HUVECs. Flk-1/KDR activation was determined by a 10-min incubation period. Flk-1/KDR activity was increased in a dose-dependent manner by LPC (1–20 μM), and maximal activation occurred at 20 μM of LPC. To confirm that these are transactivation events, we examined the effects of VEGF receptor tyrosine kinase inhibitors, SU1498 (30 μM) and VTKi (5 μM) on LPC-induced Flk-1/KDR activation in HUVECs. The cells were pretreated with SU1498 and VTKi for 30 min before the addition of LPC (20 μM) and VEGF (10 ng/ml) for 10 min. As shown in Fig. 2A, both compounds significantly inhibited phosphorylation of Flk-1/KDR in response to 20 μM LPC. Figure 2B shows that both compounds also inhibited 10 ng/ml VEGF-induced phosphorylation of Flk-1/KDR. There were no differences in the total amounts of Flk-1/KDR observed on Western blot analysis with anti-Flk-1/KDR antibodies. These findings suggest that LPC significantly transactivates Flk-1/KDR in HUVECs.

    c-Src tyrosine kinase is involved in the LPC-induced Flk-1/KDR transactivation

    Many tyrosine kinases, including those of both the receptor and nonreceptor type, are important for the activation of survival and/or proliferative pathways in various cell types. Several c-Src-mediated signal transduction pathways from GPCRs to receptor tyrosine kinases have been reported (17, 18). We hypothesized that c-Src might be involved in the LPC-induced transactivation of Flk-1/KDR. To elucidate whether LPC activates c-Src in HUVECs, an examination of the effect of LPC on c-Src tyrosine kinase activity in HUVECs was undertaken. As shown in Fig. 3A, an application of 20 μM LPC caused rapid and significant phosphorylation of c-Src (peak at 5 min) at tyrosine 418, an autophosphorylation cite that leads to full catalytic activity of the kinase. c-Src activity was increased by LPC in a dose-dependent manner (1–20 μM), and maximal activation occurred at 20 μM of LPC (Fig. 3B). A tyrosine kinase inhibitor, herbimycin A (1 μM), and a Src kinase family inhibitor, PP2 (10 μM), both inhibited LPC-induced Src kinase activation (data not shown). In addition, herbimycin A (0.1–10 μM) and PP2 (1–100 μM) both inhibited LPC-induced Flk-1/KDR activation in a dose-dependent manner (Fig. 4, A and B). The cells were transfected with KI Src for 24 h and pretreated with PP2 for 2 h before the addition of LPC (20 μM) for 10 min. As shown in Fig. 4C, KI Src transfection and PP2 (100 μM) treatment did not affect VEGF-induced Flk-1/KDR activation. Furthermore, we examined the effect of KI Src transfection on LPC-induced Flk-1/KDR activation. As shown in Fig. 5A, transfection of KI Src almost abolished LPC-induced Flk-1/KDR activation, whereas WT Src transfection caused Flk-1/KDR activation with or without LPC treatment. Transfection of KI Src also inhibited LPC-induced c-Src kinase activation in HUVECs (Fig. 5B). There were no differences in the total amounts of Flk-1/KDR observed on Western blot analysis with anti-Flk-1/KDR antibody. In WT Src and KI Src cDNA-transfected samples, c-Src protein expression was different from vector-alone-transfected sample. These findings strongly suggest that c-Src is involved in LPC-induced Flk-1/KDR transactivation in HUVECs.

    LPC stimulates downstream of Flk-1/KDR and c-Src

    It was reported that ERK1/2 and Akt exist in the downstream region of Flk-1/KDR (24). It has also been reported that LPC stimulates ERK1/2 and Akt activation, which results in the activation of cell survival pathways (25). Thus, we examined whether LPC activates ERK1/2 and Akt through transactivation of Flk-1/KDR in HUVECs. Activation of ERK1/2 and Akt in the cell lysates were determined as described in Experimental procedures. Application of 20 μM LPC caused significant activation of ERK1/2, which peaked at 60 min, and also of Akt, which peaked at 90 min (data not shown). First, we investigated whether LPC-induced activation of ERK1/2 and Akt was inhibited by SU1498 (30 μM) or VTKi (5 μM) in HUVECs. The cells were pretreated with SU1498 and VTKi for 30 min before the addition of LPC (20 μM) to examine their effects on ERK1/2 or Akt activation, respectively. Both compounds significantly inhibited ERK1/2 and Akt activation in response to 20 μM LPC stimulation (Fig. 6). These findings suggest that LPC-induced transactivation of Flk-1/KDR is involved in the activation of ERK1/2 and Akt downstream. Furthermore, we examined the effects of herbimycin A, PP2, and KI Src transfection on ERK1/2 and Akt activation by LPC to investigate whether c-Src is involved in these phenomena. The cells were pretreated with herbimycin A (1 μM) for 30 min and PP2 (10 μM) for 2 h before the addition of LPC (20 μM) to examine their respective effects on ERK1/2 or Akt activation. Herbimycin A and PP2 both inhibited LPC-induced ERK1/2 and Akt phosphorylation (Fig. 7, A and B). Transfection of KI Src also inhibited LPC-induced ERK1/2 and Akt phosphorylation in HUVECs (Fig. 7, C and D). These findings suggest that ERK1/2 and Akt are downstream effector molecules of c-Src, which is suggested to be involved in the LPC induction of Flk-1/KDR transactivation.

    LPC-induced Flk-1/KDR transactivation stimulates HUVEC proliferation

    Flk-1/KDR activation is reported to induce cell growth and differentiation (24). Therefore, to investigate pathophysiological implications of LPC-induced Flk-1/KDR transactivation, we examined the effect of LPC on HUVEC proliferation. As shown in Fig. 8A, MTT assay revealed that stimulation with LPC increased HUVEC viability over a 24-h incubation period in a dose-dependent manner (1–20 μM). In addition, we examined whether LPC-induced HUVEC proliferation was mediated by Flk-1/KDR transactivation as well as activation of ERK1/2 and Akt. The cells were pretreated with SU1498 (30 μM), VTKi (5 μM), the MEK1/2 inhibitors PD98059 (10 μM) and U0126 (10 μM), and the phosphatidylinositol 3-phosphate kinase (PI3K) inhibitor LY294002 (10 μM) and wortmannin (10 nM) for 30 min before the addition of LPC (20 μM). SU1498, VTKi, PD98059, U0126, LY294002, and wortmannin inhibited LPC-induced HUVEC proliferation (Fig. 8B). These results suggest that ERK1/2 and Akt are involved in LPC-induced proliferation of HUVECs. To characterize the role of c-Src in LPC-induced HUVEC proliferation, we then examined the effect of herbimycin A and PP2 on LPC-induced HUVEC proliferation. The cells were pretreated with herbimycin A (1 μM) for 30 min and PP2 (10 μM) for 2 h before the addition of LPC (20 μM). Both herbimycin A and PP2 significantly inhibited the HUVEC proliferation induced by LPC (Fig. 8C). Transfection of KI Src also inhibited LPC-induced HUVEC proliferation, whereas WT Src transfection induced HUVEC proliferation in the absence of LPC stimulation (Fig. 8D). These findings strongly suggest that c-Src is involved in LPC-induced HUVEC proliferation, which is mediated through Flk-1/KDR transactivation.

    Discussion

    The major finding of the present study is that LPC transactivates VEGF receptor Flk-1/KDR in HUVECs via c-Src tyrosine kinase activation. SU1498 and VTKi, which have been shown to inhibit Flk-1/KDR, inhibited LPC-induced Flk-1/KDR activation. Herbimycin A and PP2, specific inhibitors for Src family kinases, both inhibited LPC-induced Flk-1/KDR activation in a concentration-dependent manner. Transfection of KI Src also inhibited LPC-induced Flk-1/KDR activation. From these findings, it was suggested that c-Src is involved in Flk-1/KDR transactivation in HUVECs. Moreover, LPC-induced ERK1/2 and Akt activation, which are both reported to be downstream of Flk-1/KDR (17), were also inhibited by SU1498, VTKi, herbimycin A, PP2, and transfection with KI Src. It was also found that LPC-induced Flk-1/KDR transactivation resulted in a stimulation of HUVEC proliferation. VEGF receptor inhibitors, MEK1/2 inhibitors, PI3K inhibitors, Src family inhibitors, and transfection with KI Src all inhibited LPC-induced HUVEC proliferation.

    The transactivation of RTK in response to the stimulation of a number of GPCRs, such as transactivation of the epithelial growth factor receptor by GPCR ligands such as thrombin, angiotensin II, lysophosphatidic acid, and endothelin-1 has been reported (18, 26). However, LPC-stimulated transactivation of RTK has not yet been reported or elucidated. Nevertheless, the role of LPC in the pathogenesis of atherosclerosis and systemic autoimmune diseases is well documented (27, 28), even though its specific cell surface receptor was not identified until quite recently. To date, a subfamily of GPCRs consisting of G2A and GPR4 have been identified as the receptors for LPC (9, 10). It is reported that G2A is expressed in atherosclerotic lesions (29). On the other hand, it is reported that G2A and GPR4 are proton-sensing GPCRs (30, 31). However, it is also reported that GPR119 is a newly identified receptor for LPC (11). Although it still remains to be clarified what GPCRs are responsible for LPC action, we found that LPC caused Flk-1/KDR transactivation in HUVECs. LPC has been implicated in the pathological states of vascular ECs that may be related to the progressive pathological events of atherosclerosis. It has been reported that the plasma concentration of LPC is elevated in patients with coronary artery diseases, compared with that of normal subjects (32). However, the precise intracellular mechanisms mediated by LPC stimulation in ECs have not yet been clarified. In the present study, we found for the first time that LPC induces Flk-1/KDR transactivation in HUVECs (Figs. 1 and 2). Because Flk-1/KDR activation is reported to be an important factor for atherogenesis (33, 34), LPC-induced Flk-1/KDR transactivation is taken to be one of the pathological developments of atherosclerosis.

    Several mechanisms for RTK transactivation have been proposed. For example, Pyk2, reactive oxygen species, intracellular-free Ca2+, and matrix metalloproteases have been identified as factors that activate RTK in many types of cells (17, 18). We and others previously reported that c-Src is involved in the downstream signaling events after stimulation of GPCR and RTK (15, 16, 19). Therefore, we examined whether c-Src mediates LPC-induced Flk-1/KDR transactivation in the present study. As shown in Fig. 3, c-Src was rapidly and significantly activated by LPC in HUVECs. Furthermore, herbimycin A, PP2, and transfection of KI Src significantly inhibited LPC-induced Flk-1/KDR transactivation (Figs. 4 and 5). From these findings, it is strongly suggested that c-Src is involved in LPC-induced Flk-1/KDR transactivation, and c-Src may be localized near the receptor and plasma membrane. However, because we did not determine whether c-Src directly regulates Flk-1/KDR activity in this study, further studies are required to define the precise role and the mechanisms of Src kinase in Flk-1/KDR transactivation.

    One previous report suggested that a PI3K-Akt- endothelial nitric oxide synthase pathway exists downstream of the Flk-1/KDR transactivation by sphingosine-1 phosphate because the sphingosine-1 phosphate-induced Akt and endothelial nitric oxide synthase activation were inhibited by SU1498 and VTKi (17). In the present study, we found similarly that LPC-induced ERK1/2 and Akt activation were inhibited by SU1498 and VTKi (Fig. 6). It is reported that ERK1/2 and Akt are activated by VEGF as early as 5–10 min after administration (35). In the present study, we found that LPC-induced ERK1/2 and Akt activation were observed. Both the ERK1/2 and Akt activation after transactivation are likely to have an important role in EC function.

    The proliferation of ECs is a prominent feature of atherosclerotic lesions because it is an important step toward angiogenesis as well as contributing to the intimal thickening that develops after endothelial injury (8, 36). LPC is closely related to atherosclerosis because LPC is a component of the oxLDL, which is widely considered to be a major risk factor for atherosclerosis (3, 4). Therefore, we further examined the effect of LPC on HUVEC proliferation. LPC-induced HUVEC proliferation in a concentration-dependent manner with the maximal effect of LPC at 20 μM (Fig. 8A). This result is consistent with the findings of Wolfram et al. (8), who reported that 20 μM LPC induces vascular endothelial proliferation. However, we also observed that LPC at higher concentrations than 25 μM brought about HUVEC death rather than proliferation (data not shown). At higher concentration than 25 μM, LPC has been shown to cause vascular smooth muscle cell apoptosis (37). It is also reported that LPC can induce GPCR-mediated apoptosis (38). However, in contrast with these findings, it is reported that LPC at 10–20 μM facilitates cell growth, differentiation, and proliferation (8, 39). In agreement with these later findings, our results show that 20 μM LPC significantly induced HUVEC proliferation (Fig. 8A). As shown by the results of pretreatment with SU1498, VTKi, PD98059, U0126, LY294002, wortmannin, herbimycin A, and PP2 and transfection with KI Src, LPC-induced HUVEC proliferation was inhibited (Fig. 8, B–D). On the other hand, we measured lysophospholipase D that produces physiologically active lysophosphatidic acid from LPC (40, 41). In this study, LPC did not activate lysophospholipase D in HUVEC (data not shown). From these findings, it is suggested that LPC-induced Flk-1/KDR transactivation via c-Src is implicated as a contributor to the development of atherosclerosis, especially those steps in which EC proliferation plays a pivotal role.

    In summary, we show for the first time that VEGF receptor Flk-1/KDR is transactivated by LPC in HUVECs. c-Src activation is suggested to be involved in LPC-induced Flk-1/KDR transactivation. In addition, LPC-induced Flk-1/KDR transactivation caused ERK1/2 and Akt activation in HUVECs. Although the specific mechanistic steps of the pathophysiological role of LPC remain to be elucidated, LPC-induced Flk-1/KDR transactivation and the resultant proliferation of vascular ECs very likely have a role in cardiovascular diseases such as atherosclerosis.

    Acknowledgments

    We thank Dr. A. Tokumura (Faculty of Pharmaceutical Sciences, Institute of Health Biosciences, The University of Tokushima Graduate School) for excellent advisement and technical support.

    Footnotes

    First Published Online December 1, 2005

    Abbreviations: EBM, Endothelial cell basal medium; EC, endothelial cell; FBS, fetal bovine serum; Flk-1, fetal liver kinase-1; GPCR, G protein-coupled receptor; HUVEC, human umbilical vein endothelial cell; KDR, kinase-insert domain-containing receptor; KI, kinase inactive; LPC, lysophosphatidylcholine; MEK, MAPK kinase; MTT, 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H tetrazolium bromide; oxLDL, oxidized low-density lipoprotein; PI3K, phosphatidylinositol 3-phosphate kinase; PP2, 4-amino-5-(4-chlorophenyl)-7-(t-butyl) pyrazolo [3,4-d] pyrimidine; RTK, receptor tyrosine kinase; SU1498, (E)-3-(3,5-diisopropyl-4-hydroxyphenyl)-2-[(3-phenyl-n-propyl) amino-carbonyl; VEGF, vascular endothelial growth factor; VTKi, (4-[4'-chrolo-2'-fluoro]phenylamino)6,7-dimethoxyquinazoline; WT, wild type.

    Accepted for publication November 21, 2005.

    References

    Kugiyama K, Sugiyama S, Ogata N, Oka H, Doi H, Ota Y, Yasue H 1999 Burst production of superoxide anion in human endothelial cells by lysophosphatidylcholine. Atherosclerosis 143:201–204

    Kume N, Cybulsky MI, Gimbrone M-J 1992 Lysophosphatidylcholine, a component of atherogenic lipoproteins, induces mononuclear leukocyte adhesion molecules in cultured human and rabbit arterial endothelial cells. J Clin Invest 90:1138–1144

    Kugiyama K, Kerns SA, Morrisett JD, Roberts R, Henry PD 1990 Impairment of endothelium-dependent arterial relaxation by lysolecithin in modified low-density lipoproteins. Nature 344:160–162

    Tokumura A, Nishioka Y, Yoshimoto O, Shinomiya J, Fukuzawa K 1999 Substrate specificity of lysophospholipase D which produces bioactive lysophosphatidic acids in rat plasma. Biochim Biophys Acta 1437:235–245

    Caplan BA, Schwartz CJ 1973 Increased endothelial cell turnover in areas of in vivo Evans Blue uptake in the pig aorta. Atherosclerosis 17:401–417

    Ross R 1999 Atherosclerosis—an inflammatory disease. N Engl J Med 340:115–126

    Kume N, Gimbrone Jr MA 1994 Lysophosphatidylcholine transcriptionally induces growth factor gene expression in cultured human endothelial cells. J Clin Invest 93:907–911

    Wolfram K-R, Wiebke LD, Schaefer CA, Kerstin MA, Backenkohler U, Neumann T, Tillmanns H, Erdogan A 2004 Lysophosphatidylcholine-induced modulation of Ca2+-activated K+ channels contributes to ROS-dependent proliferation of cultured human endothelial cells. J Mol Cell Cardiol 36:675–682

    Zhu K, Baudhuin LM, Hong G, Williams FS, Cristina KL, Kabarowski JH, Witte ON, Xu Y 2001 Sphingosylphosphorylcholine and lysophosphatidylcholine are ligands for the G protein-coupled receptor GPR4. J Biol Chem 276:41325–41335

    Kabarowski JH, Zhu K, Le LQ, Witte ON, Xu Y 2001 Lysophosphatidylcholine as a ligand for the immunoregulatory receptor G2A. Science 293:702–705

    Soga T, Ohishi T, Matsui T, Saito T, Matsumoto M, Takasaki J, Matsumoto S, Kamohara M, Hiyama H, Yoshida S, Momose K, Ueda Y, Matsushime H, Kobori M, Furuichi K 2005 Lysophosphatidylcholine enhances glucose-dependent insulin secretion via an orphan G-protein-coupled receptor. Biochem Biophys Res Commun 326:744–751

    Huang TY, Chen HI, Liu CY, Jen CJ 2002 Lysophosphatidylcholine alters vascular tone in rat aorta by suppressing endothelial [Ca2+]i signaling. J Biomed Sci 9:327–333

    Asakura M, Kitakaze M, Takashima S, Liao Y, Ishikura F, Yoshinaka T, Ohmoto H, Node K, Yoshino K, Ishiguro H, Asanuma H, Sanada S, Matsumura Y, Takeda H, Beppu S, Tada M, Hori M, Higashiyama S 2002 Cardiac hypertrophy is inhibited by antagonism of ADAM12 processing of HB-EGF: metalloproteinase inhibitors as a new therapy. Nat Med 8:35–40

    Gschwind A, Prenzel N, Ullrich A 2002 Lysophosphatidic acid-induced squamous cell carcinoma cell proliferation and motility involves epidermal growth factor receptor signal transactivation. Cancer Res 62:6329–6336

    Tanimoto T, Lungu AO, Berk BC 2004 Sphingosine 1-phosphate transactivates the platelet-derived growth factor receptor and epidermal growth factor receptor in vascular smooth muscle cells. Circ Res 94:1050–1058

    Shah BH, Farshori MP, Jambusaria A, Catt KJ 2003 Roles of Src and epidermal growth factor receptor transactivation in transient and sustained ERK1/2 responses to gonadotropin-releasing hormone receptor activation. J Biol Chem 278:19118–19126

    Tanimoto T, Jin ZG, Berk BC 2002 Transactivation of vascular endothelial growth factor (VEGF) receptor Flk-1/KDR is involved in sphingosine 1-phosphate-stimulated phosphorylation of Akt and endothelial nitric-oxide synthase (eNOS). J Biol Chem 277:42997–43001

    Ushio-Fukai M, Griendling KK, Becker PL, Hilenski L, Halleran S, Alexander RW 2001 Epidermal growth factor receptor transactivation by angiotensin II requires reactive oxygen species in vascular smooth muscle cells. Arterioscler Thromb Vasc Biol 21:489–495

    Yang CM, Lin MI, Hsieh HL, Sun CC, Ma YH, Hsiao LD 2005 Bradykinin-induced p42/p44 MAPK phosphorylation and cell proliferation via Src, EGF receptors, and PI3-K/Akt in vascular smooth muscle cells. J Cell Physiol 203:538–546

    Legradi A, Chitu V, Szukacsov V, Fajka-Boja R, Szekely SK, Monostori E 2004 Lysophosphatidylcholine is a regulator of tyrosine kinase activity and intracellular Ca2+ level in Jurkat T cell line. Immunol Lett 91:17–21

    Rikitake Y, Kawashima S, Takahashi T, Ueyama T, Ishido S, Inoue N, Hirata K, Yokoyama M 2001 Regulation of tyrosine phosphorylation of PYK2 in vascular endothelial cells by lysophosphatidylcholine. Am J Physiol Heart Circ Physiol 281:266–274

    Kyaw M, Yoshizumi M, Tsuchiya K, Kagami S, Izawa Y, Fujita Y, Ali N, Kanematsu Y, Toida K, Ishimura K, Tamaki T 2004 Src and Cas are essentially but differentially involved in angiotensin II-stimulated migration of vascular smooth muscle cells via extracellular signal-regulated kinase 1/2 and c-Jun NH2-terminal kinase activation. Mol Pharmacol 65:832–841

    Yoshizumi M, Tsuchiya K, Kirima K, Kyaw M, Suzaki Y, Tamaki T 2001 Quercetin inhibits Shc- and phosphatidylinositol 3-kinase-mediated c-Jun N-terminal kinase activation by angiotensin II in cultured rat aortic smooth muscle cells. Mol Pharmacol 60:656–665

    Suzuma K, Naruse K, Suzuma I, Takahara N, Ueki K, Aiello LP, King GL 2000 Vascular endothelial growth factor induces expression of connective tissue growth factor via KDR, Flt1, and phosphatidylinositol 3-kinase-akt-dependent pathways in retinal vascular cells. J Biol Chem 275:40725–40731

    Wu LW, Mayo LD, Dunbar JD, Kessler KM, Baerwald MR, Jaffe EA, Wang D, Warren RS, Donner DB 2000 Utilization of distinct signaling pathways by receptors for vascular endothelial cell growth factor and other mitogens in the induction of endothelial cell proliferation. J Biol Chem 275:5096–5103

    Prenzel N, Zwick E, Daub H, Leserer M, Abraham R, Wallasch C, Ullrich A 1999 EGF receptor transactivation by G-protein-coupled receptors requires metalloproteinase cleavage of proHB-EGF. Nature 402:884–888

    Koh JS, Wang Z, Levine JS 2000 Cytokine dysregulation induced by apoptotic cells is a shared characteristic of murine lupus. J Immunol 165:4190–4201

    Lusis AJ 2000 Atherosclerosis. Nature 407:233–241

    Rikitake Y, Hirata K, Yamashita T, Iwai K, Kobayashi S, Itoh H, Ozaki M, Ejiri J, Shiomi M, Inoue N, Kawashima S, Yokoyama M 2002 Expression of G2A, a receptor for lysophosphatidylcholine, by macrophages in murine, rabbit, and human atherosclerotic plaques. Arterioscler Thromb Vasc Biol 22:2049–2053

    Murakami N, Yokomizo T, Okuno T, Shimizu T 2004 G2A is a proton-sensing G-protein-coupled receptor antagonized by lysophosphatidylcholine. J Biol Chem 279:42484–42491

    Ludwig MG, Vanek M, Guerini D, Gasser JA, Jones CE, Junker U, Hofstetter H, Wolf RM, Seuwen K 2003 Proton-sensing G-protein-coupled receptors. Nature 425:93–98

    Wells IC, Peitzmeier G, Vincent JK 1986 Lecithin: cholesterol acyltransferase and lysolecithin in coronary atherosclerosis. Exp Mol Pathol 45:303–310

    Inoue M, Itoh H, Ueda M, Naruko T, Kojima A, Komatsu R, Doi K, Ogawa Y, Tamura N, Takaya K, Igaki T, Yamashita J, Chun TH, Masatsugu K, Becker AE, Nakao K 1998 Vascular endothelial growth factor (VEGF) expression in human coronary atherosclerotic lesions: possible pathophysiological significance of VEGF in progression of atherosclerosis. Circulation 98:2108–2116

    Parsons JT, Parsons SJ 1997 Src family protein tyrosine kinases: cooperating with growth factor and adhesion signaling pathways. Curr Opin Cell Biol 9:187–192

    Bernatchez PN, Allen BG, Gelinas DS, Guillemette G, Sirois MG 2001 Regulation of VEGF-induced endothelial cell PAF synthesis: role of p42/44 MAPK, p38 MAPK and PI3K pathways. Br J Pharmacol 134:1253–1262

    O’Brien ER, Garvin MR, Dev R, Stewart DK, Hinohara T, Simpson JB, Schwartz SM 1994 Angiogenesis in human coronary atherosclerotic plaques. Am J Pathol 145:883–894

    Kogure K, Nakashima S, Tsuchie A, Tokumura A, Fukuzawa K 2003 Temporary membrane distortion of vascular smooth muscle cells is responsible for their apoptosis induced by platelet-activating factor-like oxidized phospholipids and their degradation product, lysophosphatidylcholine. Chem Phys Lipids 126:29–38

    Lin P, Ye RD 2003 The lysophospholipid receptor G2A activates a specific combination of G proteins and promotes apoptosis. J Biol Chem 278:14379–14386

    Heinloth A, Heermeier K, Raff U, Wanner C, Galle J 2000 Stimulation of NADPH oxidase by oxidized low-density lipoprotein induces proliferation of human vascular endothelial cells. J Am Soc Nephrol 11:1819–1825

    Tokumura A, Majima E, Kariya Y, Tominaga K, Kogure K, Yasuda K, Fukuzawa K 2002 Identification of human plasma lysophospholipase D, a lysophosphatidic acid-producing enzyme, as a autotoxin, a multifunctional phosphodiesterase. Biol Chem 277:39436–39442

    Tokumura A, Harada K, Fukuzawa K, Tsukatani H 1986 Involvement of lysophospholipase D in the production of lysophosphatidic acid in rat plasma. Biochim Biophys Acta 875:31–38(Yoshiko Fujita, Masanori Yoshizumi, Yuki)