Abstract

HomeCirculation ResearchVol. 99, No. 7Vascular Endothelial Growth Factor Signaling to Endothelial Nitric Oxide Synthase Free AccessEditorialPDF/EPUBAboutView PDFView EPUBSections ToolsAdd to favoritesDownload citationsTrack citationsPermissions ShareShare onFacebookTwitterLinked InMendeleyReddit Jump toFree AccessEditorialPDF/EPUBVascular Endothelial Growth Factor Signaling to Endothelial Nitric Oxide SynthaseMore than a FLeeTing Moment Michelle I. Lin and William C. Sessa Michelle I. LinMichelle I. Lin From the Yale School of Medicine (W.C.S., M.I.L.), Vascular Biology & Transplantation Program, Boyer Center for Molecular Medicine, New Haven, Conn. Search for more papers by this author and William C. SessaWilliam C. Sessa From the Yale School of Medicine (W.C.S., M.I.L.), Vascular Biology & Transplantation Program, Boyer Center for Molecular Medicine, New Haven, Conn. Search for more papers by this author Originally published29 Sep 2006https://doi.org/10.1161/01.RES.0000245430.24075.a4Circulation Research. 2006;99:666–668There is substantial evidence supporting the idea that the process of angiogenesis requires the synthesis of endothelium-derived nitric oxide (NO). NO, characterized as a major endothelial-derived relaxing factor, exerts paracrine and autocrine roles in maintaining cardiovascular homeostasis, vascular tone, and microvascular permeability. During the early steps of angiogenesis in which new blood vessels sprout from existing vascular beds, there is a persistence of vasodilation and increase in vascular permeability, suggesting that these hemodynamic changes in the existing vasculature are indispensable during an angiogenic process. A number of angiogenic factors can triggers the release of NO, synthesized by the endothelial isoform of NO synthase (eNOS). One such factor is the vascular endothelial growth factor (VEGF), which as its name implies, is a critical mitogenic, chemoattractant, and survival factor for endothelial cells1 in addition to being characterized as a potent vascular permeability factor.2 Early studies have demonstrated that VEGF can readily stimulate NO production in cultured cells and isolated blood vessels and that NO is essential in mediating the VEGF-induced endothelial cell proliferation and organization into tubes in 3D cultures.3,4 Subsequent in vivo studies have also demonstrated that eNOS knockout mice had significantly attenuated VEGF and ischemia induced angiogenesis and vascular permeability.5,6 These studies place eNOS derived NO as a key mediator of VEGF induced angiogenesis in postnatal mice.Since the discovery of VEGF, or VEGF-A, and subsequent members of the VEGF family (VEGF-B, -C, -D, -E, and placenta growth factor, PlGF), intense research has been performed to elucidate their modes of action. These growth factor ligands bind to 3 receptor tyrosine kinases (RTKs), namely VEGF receptor-1, -2, and -3 as well as to coreceptors such as heparan sulfate proteoglycans or neuropilins. Like many other RTKs, these receptors are able to form homodimers or heterodimers depending on the binding properties of the ligand. On dimerization of the receptors, activation of the tyrosine kinase follows leading to autophosphorylation of the receptor, thus creating docking sites for subsequent binding and activation of downstream signaling molecules.Much attention has focused on the signaling events elicited by VEGF-A (herein after referred to as VEGF), which can bind to the receptors, VEGFR-1 (also called Flt-1) and VEGFR-2 (also called Flk-1 in mouse or KDR in human). Gene ablation of either of these 2 receptors revealed important and distinct functional differences during vascular development: whereas the Flk-1 null mice die between embryonic days 8.5 to 9.5 as a result of decreased vasculogenesis because of early defect in hematopoietic and endothelial cells in blood islands,7 the Flt-1 null mice die between embryonic days 8.5 to 9, attributable to an obstruction of vessels by an overgrowth of endothelial cells.8 These studies suggest that whereas Flk-1 functions as a positive regulator for endothelial cell proliferation and differentiation, Flt-1 functions as the negative regulator. Interestingly, a subsequent study examining mice which were lacking the Flt-1 tyrosine-kinase domain revealed normal vascular development, which raised an interesting hypothesis that Flt-1 functions to sequester VEGF from Flk-1 during embryogenesis.9 This is supported by the evidence that Flt-1 binds to VEGF with much higher affinity than Flk-1 although exhibiting ≈10-fold less kinase activity, thus suggesting that Flt-1 may serve to “trap” VEGF, limiting Flk-1 signaling. Despite the fact that the tyrosine kinase domain of Flt-1 is dispensable during embryonic vasculogenesis/angiogenesis, other cells from these mice are impaired and experiments using chimeras of Flt-1 and the juxtamembrane region of Flk-1 (and vice versa) have revealed an uncoupling of signaling pathways between migratory and proliferative responses to the same VEGF stimulus.10Because of the complexity of VEGF signaling, it remains difficult to delineate the signaling mechanism that lies downstream of either Flt-1 or Flk-1 as both receptors can signal through common ligands. Furthermore, there is increasing evidence that suggest these 2 receptors may interact and possibly regulate each other (reviewed by Matsumoto and Claesson-Welsh11). Many studies have used overexpression of either receptor in porcine aortic endothelial cells (PAECs), which do not normally express these receptors12; antibodies directed against either receptor to abolish individual downstream signaling pathways13; or overexpression of a chimeric receptor fusing the intracellular and juxtamembrane domains of Flk-1 or Flt-1 to different extracellular domain such as colony-stimulating factor-1 receptor,14 or epidermal growth factor (EGF) receptor (EGFR).15 The latter system allows independent activation of either receptor (EGFR–Flt-1, called EGLT, or EGFR–Flk-1, called EGDR) using EGF as an agonist to stimulate the kinase activity of Flt or Flk without activating the endogenous VEGF receptors. This may be advantageous in bypassing issues in which endogenous receptors can heterodimerize and complicate the signaling pathways downstream from each receptor.In this present study in Circulation Research, Ahmed et al described that the activation of eNOS via VEGF signaling can occur via both Flt-1 and Flk-1. They demonstrated that eNOS phosphorylation on Ser1177 via a phosphatidylinositol 3′-kinase (PI3K) mechanism can occur through EGF activation of the chimeric EGLT receptor and this promotes in vitro angiogenesis. Several key tyrosine residues have been identified on both Flt-1 and Flk-1 whereby on receptor dimerization, autophosphorylation of these residues can lead to binding and activation of downstream signaling molecules. Of particular interest, the tyrosine residues at 794 and 1169 have been identified as major binding sites for phospholipase C gamma (PLCγ) whereby the amino acids spanning these residues contain consensus sequence for the binding of the N-terminal Src homology 2 (SH2) domain in PLCγ.16,17 However, very little is known about the downstream signaling events from these residues as well as other tyrosine residues identified to date.Ahmed et al demonstrated that the activation of eNOS via EGLT is dependent on the phosphorylation on the Tyr794 residue on Flt-1, as the truncated mutant, EGLT-793S, abolished the EGF stimulated eNOS phosphorylation on Ser1177. Furthermore, introduction of a nonphosphorylatable mutant Tyr794Phe into HUVECs significantly diminished NO release on EGF stimulation.15 An earlier report published by the same group demonstrated that VEGF mediated in vitro endothelial cell tube formation was dependent on Flt-1 signaling and that this was NO dependent as exogenous NO donor partially reversed the decrease in tube formation when an antibody against Flt-1 was added to neutralize the effect of VEGF.13 Using the same EGLT and EGDR chimeric receptor system, it was previously demonstrated that Tyr794 was critical in the inhibitory actions by which Flt-1 can exert on Flk-1 in stimulating endothelial cell proliferation and that this was because of the passive trapping of ligand by Flt-1.18 This inhibitory action of Flt-1 was also dependent on the p85 regulatory subunit of PI3K.18 Interestingly, the negative effect of Flt-1 on Flk-1 does not alter endothelial cell migration, suggesting the existence of a divergent regulatory mechanism by these receptors on different endothelial functions.18 These studies contrast a previous report in which the juxtamembrane domain (in which Tyr794 is located) of Flt-1 was swapped with that from Flk-1 and this domain negatively regulated Flk-1 downstream signaling events, such as p85 phosphorylation and endothelial cell migration, but not phosphorylation of mitogen activated protein kinase (MAPK).19 Despite the importance of the Tyr794 residue in mediating several endothelial functions, it remains to be elucidated whether or not the negative regulation of Flt-1 on Flk-1 also applies to the subsequent activation of eNOS.Ahmed et al report regulation of eNOS activation by via Tyr951 phosphorylation on Flk-1. Flk-1 was long been thought to be the major mediator of physiological and pathological effects of VEGF (reviewed by Cross et al20). Several tyrosine residues on Flk-1 have been mapped and examined in the context of signal transduction. In particular, Tyr1054 and Tyr1059 have been found to be required for maximal activation of the Flk-1 tyrosine kinase.21 Furthermore, Tyr951 has been shown to be important in VEGF mediated migration but not proliferation.22 Indeed, the phosphorylation of Tyr951 can also mediate the binding of the VEGF receptor associated protein (VRAP), which can serve as an adaptor protein for the binding of PLCγ and PI3K,23 or mediate the direct binding to the N-terminal or C-terminal SH2 domain in PLCγ.24 Thus, it is interesting that Ahmed et al observed that this residue is also important in the activation of eNOS through a PLCγ dependent pathway. PLCγ activation downstream of Flk-1 has been thought to be mainly attributable to Tyr1175 autophosphorylation,25 leading to subsequent activation of MAP kinase, resulting in increased endothelial cell proliferation.26 Moreover, mice which express the nonphosphorylatable phenylalanine mutant of this residue (Tyr1173Phe in mice) die in utero at embryonic day 8.5 to 9.5, similar to Flk-1 null mice, suggesting that phosphorylation of Tyr1175 and subsequent signaling downstream from this residue are critical during vascular development.27 This residue has also been thought to be essential for the activation of PI3K and protein kinase B (Akt) activities supported by the evidence that Tyr1175Phe mutant had decreased association of the Flk-1 receptor to the p85 regulatory subunit of PI3K.28 Thus, Tyr1175 is also likely to be involved in Flk-1 activation of eNOS through a PI3K/Akt pathway as, Ahmed et al demonstrated, and both receptors converge on the activation of Akt leading to eNOS phosphorylation. Introduction of a dominant negative form of Akt completely abolished EGF induced NO release and in vitro tube formation in HUVEC expressing either EGLT or EGDR.As a final thought, it remains enigmatic what the relative contributions of Flt-1 versus Flk-1 signaling are to VEGF biology, as VEGF binds with very high affinity to Flt-1 (Kd ≈1 to 20 pM) compared with that of Flk-1 (Kd ≈50 to 770 pM); yet, Flt-1 receptor phosphorylation in response to VEGF is quite low, and can only be readily observed in overexpression models. Although studies in which the VEGF receptors or chimeric receptors are overexpressed can offer insights into dissecting the signaling mechanisms through individual receptors without confounding factors, it remains difficult to validate how these models mimic the physiological situation. For instance, it has been estimated that the expression of Flt-1 is 3,000 copies per cell whereas Flk-1 is expressed at 40,000 copies per cell. The overexpression of these chimeric receptors may not resemble the natural frequency or distribution of endogenous Flt-1 versus Flk-1. Despite this caveat, the study by Ahmed et al showing that eNOS activation by VEGF can occur through Flt-1 adds another level of complexity to the VEGF receptor signaling story. Precisely how these 2 receptors respond to the same ligand to trigger different signaling events, including cross talk or feedback and eventually converge on the activation of eNOS through Akt still needs to be thoroughly investigated. Download figureDownload PowerPointFigure. eNOS activation by VEGF can occur either through the activation of PI3K subsequent to autophosphorylation of Tyr794 on Flt-1 or through a PLCγ dependent manner downstream of KDR/Flk-1 autophosphorylation of Tyr951. Once activated, both pathways converge on Akt phosphorylation of eNOS, releasing nitric oxide from endothelial cells. Other reported interaction between Tyr951 and VRAP (and subsequent recruitment of PI3K or PLCγ) or Tyr1175 and PLCγ may also contribute to KDR/Flk-1 activation of Akt-eNOS pathway.The opinions expressed in this editorial are not necessarily those of the editors or of the American Heart Association.Sources of FundingW.C.S. is supported by grants from the National Institute of Health (R01 HL64793, RO1 HL61371, R01 HL57665, PO1 HL70295, and contract No. N01-HV-28186 [NHLBI-Yale Proteomics Contract]).DisclosuresNone.FootnotesCorrespondence to William C. Sessa, Yale School of Medicine, Vascular Biology & Transplantation Program, Boyer Center for Molecular Medicine, 295 Congress Ave, New Haven, CT 06536. E-mail [email protected] References 1 Leung DW, Cachianes G, Kuang WJ, Goeddel DV, Ferrara N. Vascular endothelial growth factor is a secreted angiogenic mitogen. Science. 1989; 246: 1306–1309.CrossrefMedlineGoogle Scholar2 Senger DR, Galli SJ, Dvorak AM, Perruzzi CA, Harvey VS, Dvorak HF. Tumor cells secrete a vascular permeability factor that promotes accumulation of ascites fluid. Science. 1983; 219: 983–985.CrossrefMedlineGoogle Scholar3 Papapetropoulos A, Garcia-Cardena G, Madri JA, Sessa WC. Nitric oxide production contributes to the angiogenic properties of vascular endothelial growth factor in human endothelial cells. J Clin Invest. 1997; 100: 3131–3139.CrossrefMedlineGoogle Scholar4 van der Zee R, Murohara T, Luo Z, Zollmann F, Passeri J, Lekutat C, Isner JM. Vascular endothelial growth factor/vascular permeability factor augments nitric oxide release from quiescent rabbit and human vascular endothelium. Circulation. 1997; 95: 1030–1037.CrossrefMedlineGoogle Scholar5 Fukumura D, Gohongi T, Kadambi A, Izumi Y, Ang J, Yun CO, Buerk DG, Huang PL, Jain RK. Predominant role of endothelial nitric oxide synthase in vascular endothelial growth factor-induced angiogenesis and vascular permeability. Proc Natl Acad Sci U S A. 2001; 98: 2604–2609.CrossrefMedlineGoogle Scholar6 Murohara T, Asahara T, Silver M, Bauters C, Masuda H, Kalka C, Kearney M, Chen D, Symes JF, Fishman MC, Huang PL, Isner JM. Nitric oxide synthase modulates angiogenesis in response to tissue ischemia. J Clin Invest. 1998; 101: 2567–2578.CrossrefMedlineGoogle Scholar7 Shalaby F, Rossant J, Yamaguchi TP, Gertsenstein M, Wu XF, Breitman ML, Schuh AC. Failure of blood-island formation and vasculogenesis in Flk-1-deficient mice. Nature. 1995; 376: 62–66.CrossrefMedlineGoogle Scholar8 Fong GH, Rossant J, Gertsenstein M, Breitman ML. Role of the Flt-1 receptor tyrosine kinase in regulating the assembly of vascular endothelium. Nature. 1995; 376: 66–70.CrossrefMedlineGoogle Scholar9 Hiratsuka S, Minowa O, Kuno J, Noda T, Shibuya M. Flt-1 lacking the tyrosine kinase domain is sufficient for normal development and angiogenesis in mice. Proc Natl Acad Sci U S A. 1998; 95: 9349–9354.CrossrefMedlineGoogle Scholar10 Gille H, Kowalski J, Yu L, Chen H, Pisabarro MT, Davis-Smyth T, Ferrara N. A repressor sequence in the juxtamembrane domain of Flt-1 (VEGFR-1) constitutively inhibits vascular endothelial growth factor-dependent phosphatidylinositol 3′-kinase activation and endothelial cell migration. Embo J. 2000; 19: 4064–4073.CrossrefMedlineGoogle Scholar11 Matsumoto T, Claesson-Welsh L. VEGF receptor signal transduction. Science [serial online] 2001:RE21. Available from: Signal Transduction Knowledge Environment.Google Scholar12 Ito N, Huang K, Claesson-Welsh L. Signal transduction by VEGF receptor-1 wild type and mutant proteins. Cell Signal. 2001; 13: 849–854.CrossrefMedlineGoogle Scholar13 Bussolati B, Dunk C, Grohman M, Kontos CD, Mason J, Ahmed A. Vascular endothelial growth factor receptor-1 modulates vascular endothelial growth factor-mediated angiogenesis via nitric oxide. Am J Pathol. 2001; 159: 993–1008.CrossrefMedlineGoogle Scholar14 Rahimi N, Dayanir V, Lashkari K. Receptor chimeras indicate that the vascular endothelial growth factor receptor-1 (VEGFR-1) modulates mitogenic activity of VEGFR-2 in endothelial cells. J Biol Chem. 2000; 275: 16986–16992.CrossrefMedlineGoogle Scholar15 Ahmed S, Hewett PW, Wang P, Al-Ani B, Cudmore M, Fujisawa T, Haigh JJ, le Nobel F, Wang L, Mukhopadhyay D, Ahmed A. Direct evidence for endothelial VEGF receptor-1 funtion in nitric oxide mediated angiogenesis. Circ Res. 2006; 99: 715–722.LinkGoogle Scholar16 Cunningham SA, Arrate MP, Brock TA, Waxham MN. Interactions of FLT-1 and KDR with phospholipase C gamma: identification of the phosphotyrosine binding sites. Biochem Biophys Res Commun. 1997; 240: 635–639.CrossrefMedlineGoogle Scholar17 Sawano A, Takahashi T, Yamaguchi S, Shibuya M. The phosphorylated 1169-tyrosine containing region of flt-1 kinase (VEGFR-1) is a major binding site for PLCgamma. Biochem Biophys Res Commun. 1997; 238: 487–491.CrossrefMedlineGoogle Scholar18 Zeng H, Dvorak HF, Mukhopadhyay D. Vascular permeability factor (VPF)/vascular endothelial growth factor (VEGF) peceptor-1 down-modulates VPF/VEGF receptor-2-mediated endothelial cell proliferation, but not migration, through phosphatidylinositol 3-kinase-dependent pathways. J Biol Chem. 2001; 276: 26969–26979.CrossrefMedlineGoogle Scholar19 Gille H, Kowalski J, Li B, LeCouter J, Moffat B, Zioncheck TF, Pelletier N, Ferrara N. 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. J Biol Chem. 2001; 276: 3222–3230.CrossrefMedlineGoogle Scholar20 Cross MJ, Dixelius J, Matsumoto T, Claesson-Welsh L. VEGF-receptor signal transduction. Trends Biochem Sci. 2003; 28: 488–494.CrossrefMedlineGoogle Scholar21 Dougher M, Terman BI. Autophosphorylation of KDR in the kinase domain is required for maximal VEGF-stimulated kinase activity and receptor internalization. Oncogene. 1999; 18: 1619–1627.CrossrefMedlineGoogle Scholar22 Zeng H, Sanyal S, Mukhopadhyay D. Tyrosine residues 951 and 1059 of vascular endothelial growth factor receptor-2 (KDR) are essential for vascular permeability factor/vascular endothelial growth factor-induced endothelium migration and proliferation, respectively. J Biol Chem. 2001; 276: 32714–32719.CrossrefMedlineGoogle Scholar23 Wu LW, Mayo LD, Dunbar JD, Kessler KM, Ozes ON, Warren RS, Donner DB. VRAP is an adaptor protein that binds KDR, a receptor for vascular endothelial cell growth factor. J Biol Chem. 2000; 275: 6059–6062.CrossrefMedlineGoogle Scholar24 Wu LW, Mayo LD, Dunbar JD, Kessler KM, Baerwald MR, Jaffe EA, Wang D, Warren RS, Donner DB. 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. 2000; 275: 5096–5103.CrossrefMedlineGoogle Scholar25 Knight EL, Warner AJ, Maxwell A, Prigent SA. Chimeric VEGFRs are activated by a small-molecule dimerizer and mediate downstream signalling cascades in endothelial cells. Oncogene. 2000; 19: 5398–5405.CrossrefMedlineGoogle Scholar26 Takahashi T, Yamaguchi S, Chida K, Shibuya M. A single autophosphorylation site on KDR/Flk-1 is essential for VEGF-A-dependent activation of PLC-gamma and DNA synthesis in vascular endothelial cells. Embo J. 2001; 20: 2768–2778.CrossrefMedlineGoogle Scholar27 Sakurai Y, Ohgimoto K, Kataoka Y, Yoshida N, Shibuya M. Essential role of Flk-1 (VEGF receptor 2) tyrosine residue 1173 in vasculogenesis in mice. Proc Natl Acad Sci U S A. 2005; 102: 1076–1081.CrossrefMedlineGoogle Scholar28 Dayanir V, Meyer RD, Lashkari K, Rahimi N. Identification of tyrosine residues in vascular endothelial growth factor receptor-2/FLK-1 involved in activation of phosphatidylinositol 3-kinase and cell proliferation. J Biol Chem. 2001; 276: 17686–17692.CrossrefMedlineGoogle Scholar Previous Back to top Next FiguresReferencesRelatedDetailsCited By Shi Y, Huang X, Du Z and Tan J (2022) Analysis of single-cell RNA-sequencing data identifies a hypoxic tumor subpopulation associated with poor prognosis in triple-negative breast cancer, Mathematical Biosciences and Engineering, 10.3934/mbe.2022271, 19:6, (5793-5812), . Al Kawas H, Saaid I, Jank P, Westhoff C, Denkert C, Pross T, Weiler K and Karsten M (2022) How VEGF-A and its splice variants affect breast cancer development – clinical implications, Cellular Oncology, 10.1007/s13402-022-00665-w, 45:2, (227-239), Online publication date: 1-Apr-2022. Kato M, Ayaki I, Tanaka I, Kimura M, Arai K, Akimoto R, Nozaki T and Ishihara K (2020) Camellia japonica Seed Extract Stimulates Nitric Oxide Production via Activation of Phosphoinositide 3-Kinase/Akt/endothelial Nitric Oxide Synthase Pathway in Endothelial Cells, Food Science and Technology Research, 10.3136/fstr.26.875, 26:6, (875-881), . Sun Y, Zhao Z, Zhang H, Li J, Chen J, Luan X, Min W and He Y (2020) The interaction of lead exposure and CCM3 defect plays an important role in regulating angiogenesis through eNOS/NO pathway, Environmental Toxicology and Pharmacology, 10.1016/j.etap.2020.103407, 79, (103407), Online publication date: 1-Oct-2020. Mendes R, Nguyen D, Stephens D, Pamuk F, Fernandes D, Hasturk H, Van Dyke T and Kantarci A (2018) Hypoxia‐induced endothelial cell responses – possible roles during periodontal disease, Clinical and Experimental Dental Research, 10.1002/cre2.135, 4:6, (241-248), Online publication date: 1-Dec-2018. Grant M and Karsan A (2018) The Blood Vessel Wall Hematology, 10.1016/B978-0-323-35762-3.00123-2, (1843-1856.e6), . Bubb K, Birgisdottir A, Tang O, Hansen T and Figtree G (2017) Redox modification of caveolar proteins in the cardiovascular system- role in cellular signalling and disease, Free Radical Biology and Medicine, 10.1016/j.freeradbiomed.2017.02.012, 109, (61-74), Online publication date: 1-Aug-2017. Li Y, Zhang G, He J, Zhang D, Kong X, Yuan H and Chen F (2017)(2017) Ufm1 inhibits LPS-induced endothelial cell inflammatory responses through the NF-κB signaling pathway, International Journal of Molecular Medicine, 10.3892/ijmm.2017.2947, 39:5, (1119-1126), Online publication date: 1-May-2017. Sato Y (2016) Novel Molecular Basis for Vascular Health Regulated by Vasohibin-1, Journal of Lipid and Atherosclerosis, 10.12997/jla.2016.5.2.107, 5:2, (107), . Mendes R, Nguyen D, Stephens D, Pamuk F, Fernandes D, Van Dyke T, Kantarci A and McCormick B (2016) Endothelial Cell Response to Fusobacterium nucleatum, Infection and Immunity, 10.1128/IAI.01305-15, 84:7, (2141-2148), Online publication date: 1-Jul-2016. Sato Y (2015) Novel Link between Inhibition of Angiogenesis and Tolerance to Vascular Stress, Journal of Atherosclerosis and Thrombosis, 10.5551/jat.28902, 22:4, (327-334), . Pierro M and Thébaud B (2014) Understanding and treating pulmonary hypertension in congenital diaphragmatic hernia, Seminars in Fetal and Neonatal Medicine, 10.1016/j.siny.2014.09.008, 19:6, (357-363), Online publication date: 1-Dec-2014. Mittermayr R and Schaden W (2014) Extracorporeal Shock Wave Therapy for Acute and Chronic Soft Tissue Wounds Urgent Interventional Therapies, 10.1002/9781118504499.ch60, (549-558) Arboleda-Velasquez J and D’Amore P (2014) Vasculogenesis and Angiogenesis Cellular and Molecular Pathobiology of Cardiovascular Disease, 10.1016/B978-0-12-405206-2.00010-7, (181-196), . Gentile C, Muise-Helmericks R and Drake C (2013) VEGF-mediated phosphorylation of eNOS regulates angioblast and embryonic endothelial cell proliferation, Developmental Biology, 10.1016/j.ydbio.2012.10.020, 373:1, (163-175), Online publication date: 1-Jan-2013. Follin B, Tratwal J, Haack-Sørensen M, Elberg J, Kastrup J and Ekblond A (2013) Identical effects of VEGF and serum-deprivation on phenotype and function of adipose-derived stromal cells from healthy donors and patients with ischemic heart disease, Journal of Translational Medicine, 10.1186/1479-5876-11-219, 11:1, Online publication date: 1-Dec-2013. Olszewska-Pazdrak B and Carney D (2013) Systemic Administration of Thrombin Peptide TP508 Enhances VEGF-Stimulated Angiogenesis and Attenuates Effects of Chronic Hypoxia, Journal of Vascular Research, 10.1159/000348250, 50:3, (186-196), . Cudmore M, Hewett P, Ahmad S, Wang K, Cai M, Al-Ani B, Fujisawa T, Ma B, Sissaoui S, Ramma W, Miller M, Newby D, Gu Y, Barleon B, Weich H and Ahmed A (2012) The role of heterodimerization between VEGFR-1 and VEGFR-2 in the regulation of endothelial cell homeostasis, Nature Communications, 10.1038/ncomms1977, 3:1, Online publication date: 1-Jan-2012. Zandifar E, Sohrabi Beheshti S, Zandifar A and Haghjooy Javanmard S (2012) The Effect of Captopril on Impaired Wound Healing in Experimental Diabetes, International Journal of Endocrinology, 10.1155/2012/785247, 2012, (1-6), . Altorjay I, Veréb Z, Serfőző Z, Bacskai I, Bátori R, Erdődi F, Udvardy M, Sipka S, Lányi Á, Rajnavölgyi É and Palatka K (2011) Anti-TNF-α Antibody (Infliximab) Therapy Supports the Recovery of eNOS and VEGFR2 Protein Expression in Endothelial Cells, International Journal of Immunopathology and Pharmacology, 10.1177/039463201102400206, 24:2, (323-335), Online publication date: 1-Apr-2011. Kryzhanovskii S and Vititnova M (2011) Cardiovascular effects of nerve growth factor: An analytical review. Part II, Human Physiology, 10.1134/S0362119711030066, 37:3, (361-378), Online publication date: 1-May-2011. Lu Y, Xiong Y, Huo Y, Han J, Yang X, Zhang R, Zhu D, Klein-Heßling S, Li J, Zhang X, Han X, Li Y, Shen B, He Y, Shibuya M, Feng G and Luo J (2011) Grb-2–associated binder 1 (Gab1) regulates postnatal ischemic and VEGF-induced angiogenesis through the protein kinase A–endothelial NOS pathway, Proceedings of the National Academy of Sciences, 10.1073/pnas.1009395108, 108:7, (2957-2962), Online publication date: 15-Feb-2011. Shi Q, Cox L, Glenn J, Tejero M, Hondara V, VandeBerg J and Wang X (2010) Molecular pathways mediating differential responses to lipopolysaccharide between human and baboon arterial endothelial cells, Clinical and Experimental Pharmacology and Physiology, 10.1111/j.1440-1681.2009.05260.x, 37:2, (178-184), Online publication date: 1-Feb-2010. Franklin P, Banfor P, Tapang P, Segreti J, Widomski D, Larson K, Noonan W, Gintant G, Davidsen S, Albert D, Fryer R and Cox B (2009) Effect of the Multitargeted Receptor Tyrosine Kinase Inhibitor, ABT-869 [ N -(4-(3-Amino-1 H -indazol-4-yl)phenyl)- N ′-(2-fluoro-5-methylphenyl)urea], on Blood Pressure in Conscious Rats and Mice: Reversal with Antihypertensive Agents and Effect on Tumor Growth Inhibition , Journal of Pharmacology and Experimental Therapeutics, 10.1124/jpet.108.144816, 329:3, (928-937), Online publication date: 1-Jun-2009. Olszewska-Pazdrak B, Hein T, Olszewska P and Carney D (2009) Chronic hypoxia attenuates VEGF signaling and angiogenic responses by downregulation of KDR in human endothelial cells, American Journal of Physiology-Cell Physiology, 10.1152/ajpcell.00533.2008, 296:5, (C1162-C1170), Online publication date: 1-May-2009. Pournaras C, Rungger-Brändle E, Riva C, Hardarson S and Stefansson E (2008) Regulation of retinal blood flow in health and disease, Progress in Retinal and Eye Research, 10.1016/j.preteyeres.2008.02.002, 27:3, (284-330), Online publication date: 1-May-2008. Chen Z, Joshi S, Kauser K and Brooks A (2007) Knockdown of endothelial NOS by lentivirus-mediated short hairpin RNA in hemangioendothelioma cells increases proliferation and tumor formation, Nitric Oxide, 10.1016/j.niox.2007.03.001, 16:4, (403-412), Online publication date: 1-Jun-2007. Lorigo M, Oliveira N and Cairrao E (2021) PDE-Mediated Cyclic Nucleotide Compartmentation in Vascular Smooth Muscle Cells: From Basic to a Clinical Perspective, Journal of Cardiovascular Development and Disease, 10.3390/jcdd9010004, 9:1, (4) September 29, 2006Vol 99, Issue 7 Advertisement Article InformationMetrics https://doi.org/10.1161/01.RES.0000245430.24075.a4PMID: 17008595 Originally publishedSeptember 29, 2006 KeywordsreceptorsVEGFchimericangiogenesisNOPDF download Advertisement

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call