Abstract

Vasculogenesis, an embryological process, defines the formation of capillary plexi from endothelial precursor cells. A primitive vascular network is built via the assembly of angioblasts. Initial patterning of embryonic vascular network is independent of hemodynamic forces. The onset of blood circulation contributes to vasculature remodeling.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Subscribe and save

Springer+ Basic
EUR 32.99 /Month
  • Get 10 units per month
  • Download Article/Chapter or Ebook
  • 1 Unit = 1 Article or 1 Chapter
  • Cancel anytime
Subscribe now

Buy Now

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 169.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 219.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free ship** worldwide - see info
Hardcover Book
USD 219.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free ship** worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Notes

  1. 1.

    E.g., Hairy enhancer of Split [HES]-related transcriptional regulators HRT1 and HRT2 promotes arterial differentiation; nuclear receptor NR2f2 favors venous differentiation [886].

  2. 2.

    Haptotaxis corresponds to the adhesion gradient associated with the concentration of the constituents of the support medium, i.e., gradient in extracellular matrix density.

  3. 3.

    Tumors are hypoxic at some stage because of high oxygen consumption and inadequate blood supply. In response to hypoxia, tumor cells secrete angiogenic factors. Angiogenesis promotes tumor progression and metastasis.

  4. 4.

    In addition to mesenchymal stem cells, basophils, eosinophils, neutrophils, T lymphocytes, NK cells, and monocytes use CCL7 chemoattractant.

  5. 5.

    Vascular network development is regulated by local interactions between vascular cells and hemodynamic conditions.

  6. 6.

    Intussusception corresponds to pillar formation for capillary genesis. It is observed in the rapidly expanding postnatal lung capillary bed. The shape of these pillars is maintained by endothelial cells and pericytes. The pillars can grow in size and number. They can also merge to form folds and new vascular branches. There are 3 modes of intussusception [1176]: intussusceptive microvascular growth, arborization, and branching remodeling. Intussusceptive microvascular growth is characterized by insertion of transluminal pillars, which induces a rapid expansion of the capillary plexus. Vessels are generated from the capillary plexus by intussusceptive arborization by perpendicular pillar formation in rows, which delineate future vessels. Pillar resha** and fusions determine the new vascular entity. Pillars and folds are then formed in the capillary sheet, which separate the new vessels from the old capillary plexus. Intussusceptive branching remodeling adapts the branching angle and the bores of the branches of new supplying and draining vessels by insertion of transluminal pillars at branching points (abundant small holes are observed at branching sites). The combined cross-sectional area of the branches is greater than that of the stem: R\(_{t}^{p} =\) R\(_{b1}^{p}+\) R\(_{b2}^{p}\) (subscripts t and b for trunk and branch) with 2  < p  < 4, p depending both on the trunk bore and the vessel type (supplying or draining). The area ratio is indeed significantly different in arteries and veins [1176]. The distribution of blood flow at each bifurcation depends on the asymmetry ratio between the two branches. Strong asymmetry can lead to vascular pruning (non-perfusion and removal of branches at bifurcations). The time needed to complete pillar formation ranges from 40 to 120 mn.

  7. 7.

    The exocyst complex is composed of 8 proteins: Sec3, Sec5, Sec6, Sec8, Sec10, and Sec15, as well as Exo70 and Exo84.

  8. 8.

    Protein RabIn8 is homologous to Rab3a-interacting protein (RabIn3), hence its name RabIn3-like protein. Protein RabIn8 stimulates nucleotide exchange on Rab8, but not Rab3a and Rab5, hence being a Rab8-specific activator [1179].

  9. 9.

    Monomeric GTPase CDC42 is recruited to PIP2-rich sites of forming lumens by annexin-2, where it recruits atypical protein kinase-C.Annexin-2 resides transiently in Rab11a + vesicles during lumen formation [1178].

  10. 10.

    In nephrons, apical proteins, such as podocalyxin and Crumbs polarity complexes are delivered to Rab11a + recycling endosomes. Podocalyxin is a sialoglycoprotein of the CD34 family of transmembrane sialomucins. It is a constituent of the glycocalyx of podocytes that acts in podocyte morphogenesis. The apical Crumbs homolog complex keeps the atypical protein kinase-C apical during complex cellular shape changes. The apical-polarity determinant Crumbs complex is essential for lumen formation [1177]. The Crumbs complex associates with the Par complex directly or via the Crb complex protein Membrane protein, palmitoylated MPP5 of the P55-like MAGUK (membrane-associated guanylate kinase) subfamily (a.k.a. protein associated with Lin-7 PALS1). In addition, Crumbs homologs promote the dissociation of Par3 from Par6 and aPKC, hence allowing Par3 to localize to apicolateral borders during the transition from AMISs to preapical patches [1177].

  11. 11.

    Rab8 GTPase modulates polarized membrane transport via reorganization of actin and microtubules. Rab8 GTPase regulates membrane transport associated with TNFα-mediated stress responses and cell morphogenesis [1179].

  12. 12.

    TBC (Tre2 proto-oncogene [ubiquitin-specific peptidase USP6], BUB2 [budding uninhibited by benzimidazoles], and CDC16) motif-containing proteins pertains to GTPase activators of Rab and Rab-like GTPases.

  13. 13.

    The CDC42-specific GEF dynamin-binding protein (DnmBP; a.k.a. Tuba according to the tradition of naming large synaptic proteins after musical instruments) concentrates at the apical region of cell junctions in epithelia via its interaction with zonula occludens protein ZO1. Scaffold Tuba links dynamin, Rho GTPase, and the actin cytoskeleton. It connects dynamin to numerous actin regulators [1180].

  14. 14.

    A.k.a. chicken ovalbumin upstream promoter transcription factor COUPTF2).

  15. 15.

    A.k.a. arresten, canstatin, metastatin, and tumstatin.

  16. 16.

    A.k.a. endorepellin.

  17. 17.

    A.k.a. anastellin.

  18. 18.

    A.k.a. endostatin and neostatin.

  19. 19.

    A.k.a. vastatin.

  20. 20.

    A.k.a. restin.

  21. 21.

    A.k.a. basement-membrane protein BM40 and osteonectin.

  22. 22.

    The Sparc gene is generally detected in cells with high rates of matrix production and proliferation. It can be identified in osteoblasts, odontoblasts, and chondrocytes, as well as megacaryocytes, steroid-producing cells of adrenal glands and gonads, and cells of kidney glomeruli, bronchi, skin, and large vessels [1188].

  23. 23.

    Peptides derived from SPARC domain-4 binds to endothelial cells and impedes endothelial cell proliferation, like SPARC. This inhibition also results from the action of a peptide derived from the follistatin-like domain-2. On the other hand, peptides from SPARC domain-1 and -3 have no effect on VEGF-stimulated DNA synthesis in endothelial cells [1187]. The inhibition of proliferation of human microvascular endothelial cells is caused by, at least partly, VEGF–SPARC binding.

  24. 24.

    Lactadherin is also known as milk fat globule-EGF factor-8 (MFGE8).

  25. 25.

    Transmembrane glutamate carboxypeptidase-2 is also named prostate-specific membrane antigen.

  26. 26.

    Inhibition of P21-activated kinase-1 enhances the association of glutamate carboxypeptidase-2 with filamin-A.

  27. 27.

    The vascular network is locally composed of arteriolar branched and venular junctional networks interconnected by the capillary bed. These arteriolar and venular networks are afferent to and efferent from irrigated tissues, or efferent from and afferent to the heart pump, respectively. As the heart can be defined as the center of circulating blood circuitry, the latter meaning is preferable.

  28. 28.

    The ephrin-B2–EPHb4 signaling is involved in arteriovenous differentiation. Four Slit receptors exist, the Roundabouts (Robo). Receptor Robo4 and plexin-D1 are expressed by endothelium. Semaphorin-3 inhibits endothelial cell motility via plexin-D1. Neuropilins, in competition with Sema3a, associate with VEGF receptors and allow binding of heavy VEGF isoforms to endothelial cell receptors. Netrin-1 acts on both the endothelial cells and smooth myocytes of the vessel wall. It can have repulsive and attractive effects, according to the context. Neogenin is expressed in vascular smooth myocytes, whereas Unc5 is expressed in endothelial cells.

  29. 29.

    Slits are proteic chemorepellants in axon guidance and neuronal migration via the Roundabout receptor. Robo1 is expressed by vascular endothelial cells. Slit2 attracts endothelial cells.

  30. 30.

    Neuropilin-1 and -2 that bind semaphorin-3A (or collapsin-1) are involved in axon guidance. They are also receptors for heparin-binding VEGFa isoforms. Both Sema3a–Nrp1 and VEGF–Nrp1 signaling are required for heart development.

  31. 31.

    Netrin-1 is particularly expressed in the develo** lungs, pancreas, and mammary gland.

  32. 32.

    Presenilin-1 is a widespread transmembrane protein. It is cleaved into N-terminal (NPS1) and C-terminal (CPS1) fragments that associate to form a functional heterodimer. It promotes γ-secretase processing of substrates.

  33. 33.

    C-terminal Src kinase (CSK) binds Src kinase and prevents its activation by precluding its autophosphorylation (Tyr418).

  34. 34.

    A.k.a. translocation ETS leukemia protein (Tel or Tel1) and malignant proliferation, eosinophil (MPE).

  35. 35.

    Plasmin hydrolyzes many extracellular proteins. Urokinase and tissue-type plasminogen activators have high affinity for inactive plasminogen to produce plasmin. Conversely, plasminogen activator inhibitors PAI1 and PAI2 prevent the activation of plasminogen into plasmin.

  36. 36.

    Enzyme HDAC7 is transiently and strongly expressed in CD4 + , CD8 + thymocytes, as it represses a pro-apoptotic gene that encodes nuclear receptor NR4a1 (a.k.a. testicular receptor TR3, nerve growth factor NGF1b, and Nur77). Enzyme HDAC7 in the vascular endothelium downregulates MMP10 matrix metallopeptidase.

  37. 37.

    Two miR23–miR27–miR24 clusters exist: an intergenic miR23a–miR27a–miR24-2 and intronic miR23b–miR27b–miR24-1 cluster.

  38. 38.

    Mathematical reaction–diffusion discretized and continuous models of pattern formation represent either cells as points or as local physicochemical characteristics in a continuum. In the cell form of the model, mechanical aspects are related to cell positions, masses, velocities, and elastic properties, as well as intercellular forces, and chemical aspects to the chemical composition and diffusivity of each separate cell. In the continuous form of the model, mechanical aspects are described by stress, velocity, density, and elasticity of the matter, and chemical aspects by concentration and diffusivity in each point. Chemical reactions include production, conversion, and degradation of morphogens.

  39. 39.

    Delta-like ligand DLL4 is a Notch agonist via Notch-1 and Notch-4 receptors devoted to the specific development of the arterial endothelium.

  40. 40.

    Nuclear receptor NR2f2, or chicken ovalbumin upstream promoter transcription factor COUPTF2, inhibits the activity of many transcription factors, such as intracellular receptors retinoic acid (RAR or NR1b), retinoid X (RXR or NR2b), and thyroid (TR or NR1a) and steroid hormone (SF1 or NR5a1) receptors. Transcription factor NR2f2 is able to bind to DNA either as a homo- or heterodimer. Two NR2f types are encoded by the NR2F1 and NR2F2 genes. Factor NR2f2 is highly expressed in the venous endothelium, but not the arterial one. Transcription factor NR2f2 is also involved in the formation of atrioventricular septum and coronary vessels.

  41. 41.

    The homeobox gene product Prox1 is expressed in certain endothelial cells that yield buds from veins to give rise to lymphatics.

  42. 42.

    Tumor growth rate is not correlated with tumor vascular density. Tumor perfusion could be improved by vasculature pruning. Repression of DLL4 results in excessive angiogenesis. Normally, endothelial cell stimulation by multiple growth factors leads to cell proliferation with controlled degradation of the extracellular matrix and migration using various chemoattractants and adhesion molecules. Endothelial cells then form a central lumen and mature into a functional vessel. Neutralized DLL4 leads to defective cell fate and impairs formation of tubular structures by endothelial cells. Most tumor vessels are not functional; they are either not perfused or convey a reduced blood flow.

  43. 43.

    Protein CCM1 is produced in astrocytes and endothelial cells. It complexes with CCM2 and CCM3, as well as lipids and components of the cytoskeleton, cell junctions, and signal transduction pathways.

  44. 44.

    A.k.a. pre-adipocyte factor-1 (PreF1). It precludes differentiation into adipocytes, neuroendocrine cells, osteoblasts, myocytes, and hematopoietic cells.

  45. 45.

    In vitro, VEGF is implicated in the survival, proliferation, and migration of Schwann cells, astrocytes, and microglia [1258]. In vivo, VEGF is required for migration of certain motoneurons.

  46. 46.

    The PKD family includes PKD1 to PKD3. Kinases PKD1 and PKD3 are also called PKCμ and PKCν, respectively. PKD1 is a class-2a HDAC export kinase.

  47. 47.

    Receptor VEGFR1 is implicated in chemotaxis and vascular permeability.

  48. 48.

    Receptor VEGFR2 is involved via protein kinase-B and mitogen-activated protein kinase in endothelial cell proliferation and survival.

  49. 49.

    Growth factors VEGFb and PlGF are selective ligands of VEGFR1 (Vol. 3 – Chap. 8. Receptor Kinases).

  50. 50.

    A.k.a. calcipressin-2 [(Cps2).

  51. 51.

    Kinase LIMK1 is also phosphorylated by RoCK and PAK, downstream from Rho GTPases.

  52. 52.

    Cofilin phosphatases slingshot and chronophin dephosphorylate (activate) cofilin. Overexpression of LIMK1 suppresses cell motility [1268, 1269].

  53. 53.

    Filamin-A, the most abundant, is widespread, whereas filamin-B, also broadly expressed, is produced mainly in endothelial cells. Filamin-C is primarily expressed in skeletal myocutes.

  54. 54.

    A.k.a. metal-regulatory transcription factor-1 (MTF1).

  55. 55.

    The AP1 family is composed of dimeric protein complexes formed by products of Jun, Fos, and ATF gene families. Hypoxia upregulates AP1 expression that activates target genes for endothelin-1 and PDGFb in endothelial cells.

  56. 56.

    Hypoxia-responsive transcription factor JunB is activated by numerous stimuli, such as NFκ B and inflammatory cytokines. Other AP1 subunits Jun and Fos act as partners of VEGF response to hypoxia.

  57. 57.

    A cis-acting RNA element in the 3 UTR of the ceruloplasmin mRNA.

  58. 58.

    Interferon-γ activates a kinase cascade.Death-associated protein kinase DAPK1 activates DAPK3 that phosphorylates L13a [1276]. Both Dapk1 and Dapk3 mRNAs contain a 3UTR GAIT element. They are thus translationally repressed by the GAIT complex. This negativefeedback prevents complete suppression of translation, thereby allowing a low-level translation of target transcripts such as VegfA mRNA.

  59. 59.

    Platelet-derived growth factors form dimers that bind and activate their receptor tyrosine kinases PDGFRα and PDGFRβ that also build dimers. Both PDGFc and PDGFd are produced as latent growth factors that require proteolytic removal of their N-terminal CUB (complement subcomponents C1r and C1s, fibropellin-1, or uEGF, from Strongylocentrotus purpuratus (Purple sea urchin), and bone morphogenetic protein BMP1) domain to become active.

  60. 60.

    Platelet-derived growth factors PDGFaa, PDGFab, PDGFbb, and PDGFcc activate PDGFRαα homodimers, PDGFbb and PDGFdd stimulate PDGFRββ homodimers, and PDGFab, PDGFbb, PDGFcc, and PDGFdd activate PDGFRαβ heterodimers.

  61. 61.

    During tumoral angiogenesis, pericytes have morphological abnormalities.

  62. 62.

    The transient apoptotic effect on endothelial cells of TGFβ1 is followed by refractoriness of these cells to TGFβ1-induced apoptosis.

  63. 63.

    Prolactin operates in reproduction, growth, metabolism, immunity, osmoregulation, and behavior. Growth hormone not only stimulates growth, but also modulates metabolism, reproduction, and immunity, among other activities. Placental lactogen acts on mammary development, corpus luteum maintenance, and progesterone production.

  64. 64.

    The bone morphogenetic protein BMP1 family comprises structurally similar metallopeptidases: BMP1, mammalian Tolloid, and mammalian Tolloid-like Tll1 and Tll2. BMP1-like peptidases participate in the formation of the extracellular matrix by processing precursors: procollagens-1, -3, -5, -7, and -11 into functional ECM components; prolysyl oxidase zymogen into enzyme that catalyzes crosslinks in collagens and elastins; prolaminin-5; precursors of proteoglycans biglycan and osteoglycin; etc. BMP1-like peptidases activate certain TGFβ complexes, including BMP2, BMP4, GDF8, GDF11, and TGFβ1.

  65. 65.

    Interleukin-20 belongs to the IL10 family with IL10, IL19, IL22, IL24, and IL26. Many cell types produce IL20, such as monocytes and epithelial and endothelial cells.

  66. 66.

    The endothelial receptor protein Tyr kinase with Ig and EGF homology TIE2 is also termed Tyr-protein endothelial kinase TEK. Receptor TIE1 may build TIE1–TIE2 heterodimers to contribute to TIE2 signal transduction. Besides, Ang1–TIE2 signaling operates in the maintenance of hematopoietic stem cells.

  67. 67.

    Apelin localizes preferentially in blood vessels. An immature peptide, preproapelin, is secreted and cleaved by proteases to form apelin13, apelin17, and apelin36, among others. These isoforms have different receptor-binding affinities and activities. Apelin has a positive inotropic effect and can cause vasoconstriction and vasodilation (via NO) according to circumstances.

  68. 68.

    Receptor APJ is widespread in embryos and adults, especially in the lung and heart, as well as in vascular and endocardial endothelial and smooth muscle cells.

  69. 69.

    In endothelial cells, PI3Kc1β operates downstream from liganded G-protein-coupled receptors such as CXCR4 α-chemokine receptor. In these cells, PI3Kc1δ is expressed at low concentration. Catalytic subunits of class-1A PI3K isoforms PI3Kc1α and PI3Kc1β are ubiquitous, whereas PI3Kc1δ is only enriched in leukocytes.

  70. 70.

    This transient signaling is associated with normal microvascular development, acute PKB activation of myristoylated PKB (PKBM) being cardioprotective.

  71. 71.

    Thrombospondins are involved not only in endothelial cell proliferation and apoptosis, but also in collagen assembly and maintenance of vascular integrity. Thrombospondin-2 controls the level of matrix metallopeptidase MMP2 [1316].

  72. 72.

    Substrates of ADAMTS1 also include aggrecan, versican, and nidogen. It is associated with catalytic modifiers such as fibulin-1. Moreover, ADAMTS1 has a non-catalytic function, as it sequesters VEGF factor.

  73. 73.

    Girdin is also known as Akt (PKB) phosphorylation enhancer (APE), Gα-interacting vesicle-associated protein (GIV), and Hook-related protein-1 (HkRP1). Kinase PKB phosphorylates girdin to regulate its subcellular location and fibroblast migration.

  74. 74.

    After blood vessel injury, tissue factor with factor VIIa activates factor Xa, thrombin, and fibrin to build a hemostatic plug.

  75. 75.

    Hydrogen sulfide activates ERK1 and ERK2 in monocytes, smooth myocytes, and gastric epithelial cells [1327]. It stimulates P38MAPK in smooth muscle and β cells, but precludes P38MAPK phosphorylation in neutrophils and microglial cells.

  76. 76.

    Inhibition of endothelial NO synthase (NOS3) or PKG1 abolishes H2S-triggered angiogenesis and attenuates H2S-primed vasorelaxation. Conversely, inactivation of H2S synthase cystathionine γ-lyase supresses NO-stimulated cGMP accumulation and angiogenesis and reduces acetylcholine-stimulated vasorelaxation [1328].

  77. 77.

    These adducts localize to oxidized low-density lipoprotein and accumulate in atherosclerotic plaques as well as retina, where they promote choroidal neovascularization and age-related macular degeneration. These adducts, carboxyethylpyrrole (CEP) in particular, are transiently observed during wound healing. At injury sites, they reach a maximum concentration 3 days after damage and return to original levels when the healing is completed. High levels of CEP correlate with intense vascularization of damaged tissue.

  78. 78.

    Factor VEGF during the early stage of angiogenesis activates cell migration and sprouting. Angiopoietin-1 mediates the interactions between endothelial and smooth muscle cells. Angiopoietin-2 disrupts these interactions to promote independent migration. Both Ang1 and Ang2 are produced by vascular cells, bind to receptor TIE2, and act synergistically with VEGF to regulate angiogenesis. Homodimer PDGFbb released by endothelial cells at a late stage of angiogenesis recruits smooth muscle cells to stabilize the nascent sprouts.

  79. 79.

    NADPH oxidase also localizes to the endoplasmic reticulum. Mitochondria are another major ROS sources.

  80. 80.

    Soluble VEGFR1S also binds VEGFb and placenta growth factor. Agent VEGFR1S forms heterodimers with VEGFR2 receptor. Avascularity may be maintained by multiple redundant mechanisms associated with many anti-angiogenic molecules that exist in the cornea, but neutralization of VEGFR1S alone abolishes corneal avascularity. VEGFR1S also regulates the availability of VEGFa in cyclic vascularization and embryonic sprouting.

  81. 81.

    Thrombospondins constitute 2 subsets: subset A with Tsp1 and Tsp2 homotrimers and subset B with Tsp3 to Tsp5 homopentamers.

  82. 82.

    Regulator of G-protein signaling RGS5 is a marker for pericyte progenitor cells.

  83. 83.

    Activation of matrix metallopeptidase-9 leads to the release of soluble SCF, which promotes the proliferation and motility of circulating endothelial precursors.

  84. 84.

    Molecule CA4P binds to tubulin. It selectively targets endothelial cells, but not smooth myocytes.

  85. 85.

    A.k.a. RING finger protein RNF71 and tripartite motif-containing protein TRIM19.

  86. 86.

    Promyelocytic leukemia, which regulates tumor suppressor transcription factors, is lost in various types of human cancers.

  87. 87.

    Many pericytes survive after VEGF inhibitor administration. However, the pericyte phenotype reversibly changes during treatment, with a downregulation of the expression of α-smooth muscle actin. This change reverses when the treatment is stopped.

  88. 88.

    Manganese-containing enzyme SOD2 (MnSOD) resides in the mitochondrial matrix. Secreted, copper-containing SOD3 (CuSOD) lodges in the extracellular matrix.

  89. 89.

    A.k.a. liver receptor homolog-1.

  90. 90.

    Nineteen ETS transcription factors are expressed in blood vessel endothelial cells.

  91. 91.

    Receptor VEGFR3 exists in all vascular endothelia during embryo- and fetogenesis, whereas in adults, it is restricted to the lymphatic endothelium. However, VEGFR3 is observed in the microvasculature of tumors and wound healing. In adult humans, VEGFR3 is also synthesized in some fenestrated and discontinuous endothelia of blood vessels. Receptor VEGFR3 promotes the migration and proliferation of lymphatic endothelial cells.

  92. 92.

    Neuropilin-2 is a semaphorin receptor of the nervous system and lymphatic capillaries. Combined inhibition of VEGFR2 and VEGFR3 prevents angiogenesis and tumor growth [1366].

References

Chap. 10. Vasculature Growth

  1. Koivisto L, Larjava K, Hakkinen L, Uitto VJ, Heino J, Larjava H (1999) Different integrins mediate cell spreading, haptotaxis and lateral migration of HaCaT keratinocytes on fibronectin. Cell Adhesion and Communication 7:245–257

    Google Scholar 

  2. DuBois-Reymond E (1843) Vorlufiger Abriss einer Untersuchung ber den sogenannten Froschstrom und ber die elektromotorischen Fische [Preliminary outline of an investigation of the so-called frog current and of the electromotive fish]. Annalen der Physik und Chemie 58:1–30

    ADS  Google Scholar 

  3. Zhao M, Song B, Pu J, Wada T, Reid B, Tai G, Wang F, Guo A, Walczysko P, Gu Y, Sasaki T, Suzuki A, Forrester JV, Bourne HR, Devreotes PN, McCaig CD, Penninger JM (2006) Electrical signals control wound healing through phosphatidylinositol-3-OH kinase-big gamma and PTEN. Nature 442:457–460

    ADS  Google Scholar 

  4. Coultas L, Chawengsaksophak K, Rossant J (2005) Endothelial cells and VEGF in vascular development. Nature 438:937–945

    ADS  Google Scholar 

  5. Kokovay E, Li L, Cunningham LA (2006) Angiogenic recruitment of pericytes from bone marrow after stroke. Journal of Cerebral Blood Flow & Metabolism 26:545–555

    Google Scholar 

  6. Huu SN, Oster M, Uzan S, Chareyre F, Aractingi S, Khosrotehrani K (2007) Maternal neoangiogenesis during pregnancy partly derives from fetal endothelial progenitor cells. Proceedings of the National Academy of Sciences of the United States of America 104:1871–1876

    ADS  Google Scholar 

  7. Bousquenaud M, Schwartz C, Léonard F, Rolland-Turner M, Wagner D, Devaux Y (2012) Monocyte chemotactic protein 3 is a homing factor for circulating angiogenic cells. Cardiovascular Research 94:519–525

    Google Scholar 

  8. Gao D, Nolan DJ, Mellick AS, Bambino K, McDonnell K, Mittal V (2008) Endothelial progenitor cells control the angiogenic switch in mouse lung metastasis. Science 319:195–198

    ADS  Google Scholar 

  9. Bailey AS, Willenbring H, Jiang S, Anderson DA, Schroeder DA, Wong MH, Grompe M, Fleming WH (2006) Myeloid lineage progenitors give rise to vascular endothelium. Proceedings of the National Academy of Sciences of the United States of America 103: 13156-13161

    ADS  Google Scholar 

  10. Szczerba D, Szkely G (2005) A computational model of micro-vascular growth, In: Lecture Notes in Computer Science, Vol. 3516, 17–24, Springer, Berlin

    Google Scholar 

  11. De Palma M, Venneri MA, Galli R, Sergi LS, Politi LS, Sampaolesi M, Naldini L (2005) Tie2 identifies a hematopoietic lineage of proangiogenic monocytes required for tumor vessel formation and a mesenchymal population of pericyte progenitors. Cancer Cell 8:211–226

    Google Scholar 

  12. Jakobsson L, Franco CA, Bentley K, Collins RT, Ponsioen B, Aspalter IM, Rosewell I, Busse M, Thurston G, Medvinsky A, Schulte-Merker S, Gerhardt H (2010) Endothelial cells dynamically compete for the tip cell position during angiogenic sprouting. Nature – Cell Biology 12:943–953

    Google Scholar 

  13. Kurz H, Burri PH, Djonov VG (2003) Angiogenesis and vascular remodeling by intussusception: From form to function. News in Physiological Sciences 18:65–70

    Google Scholar 

  14. Apodaca G (2010) Opening ahead: early steps in lumen formation revealed. Nature – Cell Biology 12:1026–1028

    Google Scholar 

  15. Bryant DM, Datta A, Rodríguez-Fraticelli AE, Peränen J, Martn-Belmonte F, Mostov KE (2010) A molecular network for de novo generation of the apical surface and lumen. Nature – Cell Biology 12:1035–1045

    Google Scholar 

  16. Hattula K, Furuhjelm J, Arffman A, Pernen J (2002) A Rab8-specific GDP/GTP exchange factor is involved in actin remodeling and polarized membrane transport. Molecular Biology of the Cell 13:3268–3280

    Google Scholar 

  17. Salazar MA, Kwiatkowski AV, Pellegrini L, Cestra G, Butler MH, Rossman KL, Serna DM, Sondek J, Gertler FB, De Camilli P (2003) Tuba, a novel protein containing bin/amphiphysin/Rvs and Dbl homology domains, links dynamin to regulation of the actin cytoskeleton. Journal of Biological Chemistry 278:49031–49043.

    Google Scholar 

  18. van Royen N, Piek JJ, Buschmann I, Hoefer I, Voskuil M, Schaper W (2001) Stimulation of arteriogenesis: a new concept for the treatment of arterial occlusive disease. Cardiovascular Research 49:543–553

    Google Scholar 

  19. Murray CD (1926) The physiological principle of minimum work I: The vascular system and the cost of blood volume. Proceedings of the National Academy of Sciences of the United States of America 12:207–214

    ADS  Google Scholar 

  20. le Noble F, Moyon D, Pardanaud L, Yuan L, Djonov V, Matthijsen R, Breant C, Fleury V, Eichmann A (2004) Flow regulates arterial-venous differentiation in the chick embryo yolk sac. Development 131:361–375

    Google Scholar 

  21. Adams RH, Alitalo K (2007) Molecular regulation of angiogenesis and lymphangiogenesis. Nature Reviews – Molecular Cell Biology 8:464–478

    Google Scholar 

  22. Arroyo AG, Iruela-Arispe ML (2010) Extracellular matrix, inflammation, and the angiogenic response. Cardiovascular Research 86:226–235

    Google Scholar 

  23. Armulik A, Abramsson A, Betsholtz C (2005) Endothelial/pericyte interactions. Circulation Research 97:512–523

    Google Scholar 

  24. Kupprion C, Motamed K, Sage EH (1998) SPARC (BM-40, osteonectin) inhibits the mitogenic effect of vascular endothelial growth factor on microvascular endothelial cells. Journal of Biological Chemistry 273:29635–29640

    Google Scholar 

  25. Mundlos S, Schwahn B, Reichert T, Zabel B (1992) Distribution of osteonectin mRNA and protein during human embryonic and fetal development. Journal of Histochemistry and Cytochemistry 40:283–291

    Google Scholar 

  26. Chlenski A, Guerrero LJ, Peddinti R, Spitz JA, Leonhardt PT, Yang Q, Tian Y, Salwen HR, Cohn SL (2010) Anti-angiogenic SPARC peptides inhibit progression of neuroblastoma tumors. Molecular Cancer 9:138

    Google Scholar 

  27. Iozzo RV (2005) Basement membrane proteoglycans: from cellar to ceiling. Nature Reviews – Molecular Cell Biology 6:646–656

    Google Scholar 

  28. Sunshine SB, Dallabrida SM, Durand E, Ismail NS, Bazinet L, Birsner AE, Sohn R, Ikeda S, Pu WT, Kulke MH, Javaherian K, Zurakowski D, Folkman JM, Rupnick M (2012) Endostatin lowers blood pressure via nitric oxide and prevents hypertension associated with VEGF inhibition. Proceedings of the National Academy of Sciences of the United States of America 109:11306–11311

    ADS  Google Scholar 

  29. Kitazume S, Imamaki R, Ogawa K, Komi Y, Futakawa S, Kojima S, Hashimoto Y, Marth JD, Paulson JC, Taniguchi N (2010) α-2,6-sialic acid on platelet endothelial cell adhesion molecule (PECAM) regulates its homophilic interactions and downstream antiapoptotic signaling. Journal of Biological Chemistry 285:6515–6521

    Google Scholar 

  30. Francis SE, Goh KL, Hodivala-Dilke K, Bader BL, Stark M, Davidson D, Hynes RO (2002) Central roles of α5β1 integrin and fibronectin in vascular development in mouse embryos and embryoid bodies. Arteriosclerosis, Thrombosis, and Vascular Biology 22:927–933

    Google Scholar 

  31. Huang S, Ingber DE (1999) The structural and mechanical complexity of cell-growth control. Nature – Cell Biology 1:E131–E138

    Google Scholar 

  32. Silvestre J-S, Thry C, Hamard G, Boddaert J, Aguilar B, Delcayre A, Houbron C, Tamarat R, Blanc-Brude O, Heeneman S, Clergue M, Duriez M, Merval R, Lvy B, Tedgui A, Amigorena S, Mallat Z (2005) Lactadherin promotes VEGF-dependent neovascularization. Nature – Medicine 11:499–506

    Google Scholar 

  33. Lorger M, Krueger JS, O’Neal M, Staflin K, Felding-Habermann B (2009) Activation of tumor cell integrin αVβ3 controls angiogenesis and metastatic growth in the brain. Proceedings of the National Academy of Sciences of the United States of America 106:10666–10671

    ADS  Google Scholar 

  34. Jones FS, Jones PL (2000) The tenascin family of ECM glycoproteins: structure, function, and regulation during embryonic development and tissue remodeling. Developmental Dynamics 218:235–259.

    Google Scholar 

  35. Chung CY, Murphy-Ullrich JE, Erickson HP (1996) Mitogenesis, cell migration, and loss of focal adhesions induced by tenascin-C interacting with its cell surface receptor, annexin II. Molecular Biology of the Cell 7:883–892

    Google Scholar 

  36. Twal WO, Czirok A, Hegedus B, Knaak C, Chintalapudi MR, Okagawa H, Sugi Y, Argraves WS (2001) Fibulin-1 suppression of fibronectin-regulated cell adhesion and motility. Journal of Cell Science 114:4587–4598

    Google Scholar 

  37. Hunzelmann N, Nischt R, Brenneisen P, Eickert A, Krieg T (2001) Increased deposition of fibulin-2 in solar elastosis and its colocalization with elastic fibres. British Journal of Dermatology 145:217–222

    Google Scholar 

  38. Hopf M, Göhring W, Mann K, Timpl R (2001) Map** of binding sites for nidogens, fibulin-2, fibronectin and heparin to different IG modules of perlecan. Journal of Molecular Biology 311:529–541

    Google Scholar 

  39. Seeliger H, Camaj P, Ischenko I, Kleespies A, De Toni EN, Thieme SE, Blum H, Assmann G, Jauch KW, Bruns CJ (2009) EFEMP1 expression promotes in vivo tumor growth in human pancreatic adenocarcinoma. Molecular Cancer Research 7:189–198

    Google Scholar 

  40. Chen Q, Zhang T, Roshetsky JF, Ouyang Z, Essers J, Fan C, Wang Q, Hinek A, Plow EF, Dicorleto PE (2009) Fibulin-4 regulates expression of the tropoelastin gene and consequent elastic-fibre formation by human fibroblasts. Biochemical Journal 423:79–89

    Google Scholar 

  41. Nakamura T, Ruiz-Lozano P, Lindner V, Yabe D, Taniwaki M, Furukawa Y, Kobuke K, Tashiro K, Lu Z, Andon NL, Schaub R, Matsumori A, Sasayama S, Chien KR, Honjo T (1999) DANCE, a novel secreted RGD protein expressed in develo**, atherosclerotic, and balloon-injured arteries. Journal of Biological Chemistry 274:22476–22483

    Google Scholar 

  42. Conway RE, Petrovic N, Li Z, Heston W, Wu D, Shapiro LH (2006) Prostate-specific membrane antigen regulates angiogenesis by modulating integrin signal transduction. Molecular and Cellular Biology 26:5310–5324

    Google Scholar 

  43. Teodoro JG, Parker AE, Zhu X, Green MR (2006) p53-mediated inhibition of angiogenesis through up-regulation of a collagen prolyl hydroxylase. Science 313:968–971

    ADS  Google Scholar 

  44. Klagsbrun M, Eichmann A (2005) A role for axon guidance receptors and ligands in blood vessel development and tumor angiogenesis. Cytokine Growth Factor Reviews 16:535–548

    Google Scholar 

  45. Weinstein BM (2005) Vessels and nerves: marching to the same tune. Cell 120:299–302

    Google Scholar 

  46. Serini G, Valdembri D, Zanivan S, Morterra G, Burkhardt C, Caccavari F, Zammataro L, Primo L, Tamagnone L, Logan M, Tessier-Lavigne M, Taniguchi M, Puschel AW, Bussolino F (2003) Class 3 semaphorins control vascular morphogenesis by inhibiting integrin function. Nature 424:391–397

    ADS  Google Scholar 

  47. Kawasaki T, Kitsukawa T, Bekku Y, Matsuda Y, Sanbo M, Yagi T, Fujisawa H (1999) A requirement for neuropilin-1 in embryonic vessel formation. Development 126:4895–4902

    Google Scholar 

  48. Gu C, Rodriguez ER, Reimert DV, Shu T, Fritzsch B, Richards LJ, Kolodkin AL, Ginty DD (2003) Neuropilin-1 conveys semaphorin and VEGF signaling during neural and cardiovascular development. Developmental Cell 5:45–57

    MATH  Google Scholar 

  49. Gu C, Yoshida Y, Livet J, Reimert DV, Mann F, Merte J, Henderson CE, Jessell TM, Kolodkin AL, Ginty DD (2005) Semaphorin 3E and plexin-D1 control vascular pattern independently of neuropilins. Science 307:265–268

    ADS  Google Scholar 

  50. Toyofuku T, Yabuki M, Kamei J, Kamei M, Makino N, Kumanogoh A, Hori M (2007) Semaphorin-4A, an activator for T-cell-mediated immunity, suppresses angiogenesis via Plexin-D1. EMBO Journal 26:1373–1384

    Google Scholar 

  51. Conrotto P, Valdembri D, Corso S, Serini G, Tamagnone L, Comoglio PM, Bussolino F, Giordano S (2005) Sema4D induces angiogenesis through Met recruitment by Plexin B1. Blood 105:4321–4329

    Google Scholar 

  52. Cirulli V, Yebra M (2007) Netrins: beyond the brain. Nature Reviews – Molecular Cell Biology 8, 296-306

    Google Scholar 

  53. Pandey A, Shao H, Marks RM, Polverini PJ, Dixit VM (1995) Role of B61, the ligand for the Eck receptor tyrosine kinase, in TNF-alpha-induced angiogenesis. Science 268:567–569

    ADS  Google Scholar 

  54. Georgakopoulos A, Litterst C, Ghersi E, Baki L, Xu C, Serban G, Robakis NK (2006) Metalloproteinase/Presenilin1 processing of ephrinB regulates EphB-induced Src phosphorylation and signaling. EMBO Journal 25:1242–1252

    Google Scholar 

  55. Erber R, Eichelsbacher U, Powajbo V, Korn T, Djonov V, Lin J, Hammes HP, Grobholz R, Ullrich A, Vajkoczy P (2006) EphB4 controls blood vascular morphogenesis during postnatal angiogenesis. EMBO Journal 25:628–641

    Google Scholar 

  56. Park KW, Morrison CM, Sorensen LK, Jones CA, Rao Y, Chien CB, Wu JY, Urness LD, Li DY (2003) Robo4 is a vascular-specific receptor that inhibits endothelial migration. Developmental Biology 61:251–267

    Google Scholar 

  57. Fischer A, Schumacher N, Maier M, Sendtner M, Gessler M (2004) The Notch target genes Hey1 and Hey2 are required for embryonic vascular development. Genes and Development 18:901–911

    Google Scholar 

  58. Winnik S, Klinkert M, Kurz H, Zoeller C, Heinke J, Wu Y, Bode C, Patterson C, Moser M (2009) HoxB5 induces endothelial sprouting in vitro and modifies intussusceptive angiogenesis in vivo involving angiopoietin-2. Cardiovascular Research 83:558–565

    Google Scholar 

  59. Vasudevan A, Long JE, Crandall JE, Rubenstein JLR, Bhide PG (2008) Compartment-specific transcription factors orchestrate angiogenesis gradients in the embryonic brain. Nature – Neuroscience 11:429–439

    Google Scholar 

  60. Roukens MG, Alloul-Ramdhani M, Baan B, Kobayashi K, Peterson-Maduro J, van Dam H, Schulte-Merker S, Baker DA (2010) Control of endothelial sprouting by a Tel-CtBP complex. Nature – Cell Biology 12:933–942

    Google Scholar 

  61. Tabruyn SP, Griffioen AW (2007) A new role for NF-kappaB in angiogenesis inhibition. Cell Death and Differentiation 14:1393–1397

    Google Scholar 

  62. Scoditti E, Massaro M, Carluccio MA, Distante A, Storelli C, De Caterina R (2010) PPARγ agonists inhibit angiogenesis by suppressing PKCα- and CREB-mediated COX-2 expression in the human endothelium. Cardiovascular Research 86:302–310

    Google Scholar 

  63. Chinsomboon J, Ruas J, Gupta RK, Thom R, Shoag J, Rowe GC, Sawada N, Raghuram S, Arany Z (2009) The transcriptional coactivator PGC-1α mediates exercise-induced angiogenesis in skeletal muscle. Proceedings of the National Academy of Sciences of the United States of America 106:21401–21406

    ADS  Google Scholar 

  64. Ray LB (2009) Physiology: marathon mouse mechanism. Science Signaling 2:ec403

    Google Scholar 

  65. Boon RA, Urbich C, Fischer A, Fontijn RD, Seeger FH, Koyanagi M, Horrevoets AJG, Dimmeler S (2011) Krüppel-like factor 2 improves neovascularization capacity of aged proangiogenic cells. European Heart Journal 32:371–377

    Google Scholar 

  66. Rey S, Semenza GL (2010) Hypoxia-inducible factor-1-dependent mechanisms of vascularization and vascular remodelling. Cardiovascular Research 86:236–242

    Google Scholar 

  67. Martin M, Potente M, Janssens V, Vertommen D, Twizere JC, Rider MH, Goris J, Dimmeler S, Kettmann R, Dequiedt F (2008) Protein phosphatase 2A controls the activity of histone deacetylase 7 during T cell apoptosis and angiogenesis. Proceedings of the National Academy of Sciences of the United States of America 105:4727–4732

    ADS  Google Scholar 

  68. Economopoulou M, Langer HF, Celeste A, Orlova VV, Choi EY, Ma M, Vassilopoulos A, Callen E, Deng C, Bassing CH, Boehm M, Nussenzweig A, Chavakis T (2009) Histone H2AX is integral to hypoxia-driven neovascularization. Nature – Medicine 15:553–558

    Google Scholar 

  69. Kaikkonen MU, Lam MTY, Glass CK (2011) Non-coding RNAs as regulators of gene expression and epigenetics. Cardiovascular Research 90:430–440

    Google Scholar 

  70. Suarez Y, Fernandez-Hernando C, Yu J, Gerber SA, Harrison KD, Pober JS, Iruela-Arispe L, Merkenschlager M, Sessa WC (2008) Dicer-dependent endothelial microRNAs are necessary for postnatal angiogenesis. Proceedings of the National Academy of Sciences of the United States of America 105:14082-14087

    ADS  Google Scholar 

  71. Nicoli S, Standley C, Walker P, Hurlstone A, Fogarty KE, Lawson ND (2010) MicroRNA-mediated integration of haemodynamics and Vegf signalling during angiogenesis. Nature 464:1196–1200

    ADS  Google Scholar 

  72. Halberg N, Alarcón C, Tavazoie SF (2012) MicroRNA regulation of cancer–endothelial interactions: Vesicular microRNAs on the move... EMBO Journal 31:3509–3510

    Google Scholar 

  73. Anand S, Majeti BK, Acevedo LM, Murphy EA, Mukthavaram R, Scheppke L, Huang M, Shields DJ, Lindquist JN, Lapinski PE, King PD, Weis SM, Cheresh DA (2010) MicroRNA-132-mediated loss of p120RasGAP activates the endothelium to facilitate pathological angiogenesis. Nature – Medicine 16:909–914

    Google Scholar 

  74. Zhou Q, Gallagher R, Ufret-Vincenty R, Li X, Olson EN, Wang S (2011) Regulation of angiogenesis and choroidal neovascularization by members of microRNA-23 27 24 clusters. Proceedings of the National Academy of Sciences of the United States of America 108:8287–8292

    Google Scholar 

  75. Zhuang G, Wu X, Jiang Z, Kasman I, Yao J, Guan Y, Oeh J, Modrusan Z, Bais C, Sampath D, Ferrara N (2012) Tumour-secreted miR-9 promotes endothelial cell migration and angiogenesis by activating the JAK-STAT pathway. EMBO Journal 31:3513–3523

    Google Scholar 

  76. Turing AM (1952) The chemical basis of morphogenesis. Philosophicals Transactions of the Royal Society of London, Series B, Biological Sciences 237:37–72

    ADS  Google Scholar 

  77. Hellström M, Phng LK, Hofmann JJ, Wallgard E, Coultas L, Lindblom P, Alva J, Nilsson AK, Karlsson L, Gaiano N, Yoon K, Rossant J, Iruela-Arispe L, Kalén M, Gerhardt H, Betsholtz C (2007) Dll4 signalling through Notch1 regulates formation of tip cells during angiogenesis. Nature 445:776–780

    ADS  Google Scholar 

  78. Siekmann AF, Lawson ND (2007) Notch signalling limits angiogenic cell behaviour in develo** zebrafish arteries. Nature 445:781–784

    ADS  Google Scholar 

  79. Noguera-Troise I, Daly C, Papadopoulos NJ, Coetzee S, Boland P, Gale NW, Lin HC, Yancopoulos GD, Thurston G (2006) Blockade of Dll4 inhibits tumour growth by promoting non-productive angiogenesis. Nature 444:1032–1037

    ADS  Google Scholar 

  80. Ridgway J, Zhang G, Wu Y, Stawicki S, Liang WC, Chanthery Y, Kowalski J, Watts RJ, Callahan C, Kasman I, Singh M, Chien M, Tan C, Hongo JAS, de Sauvage F, Plowman G, Yan M (2006) Inhibition of Dll4 signalling inhibits tumour growth by deregulating angiogenesis. Nature 444:1083–1087

    ADS  Google Scholar 

  81. Benedito R, Roca C, Sörensen I, Adams S, Gossler A, Fruttiger M, Adams RH (2009) The notch ligands Dll4 and Jagged1 have opposing effects on angiogenesis. Cell 137:1124–1135

    Google Scholar 

  82. Wüstehube J, Bartol A, Liebler SS, Brütsch R, Zhu Y, Felbor U, Sure U, Augustin HG, Fischer A (2010) Cerebral cavernous malformation protein CCM1 inhibits sprouting angiogenesis by activating DELTA-NOTCH signaling. Proceedings of the National Academy of Sciences of the United States of America 107:12640–12645

    ADS  Google Scholar 

  83. Benedito R, Rocha SF, Woeste M, Zamykal M, Radtke F, Casanovas O, Duarte A, Pytowski B, Adams RH (2012) Notch-dependent VEGFR3 upregulation allows angiogenesis without VEGF–VEGFR2 signalling. Nature 484:110–114

    ADS  Google Scholar 

  84. Rodríguez P, Higueras MA, Gonzlez-Rajal A, Alfranca A, Fierro-Fernndez M, Garca-Fernndez RA, Ruiz-Hidalgo MJ, Monsalve M, Rodríguez-Pascual F, Redondo JM, de la Pompa JL, Laborda J, Lamas S (2012) The non-canonical NOTCH ligand DLK1 exhibits a novel vascular role as a strong inhibitor of angiogenesis. Cardiovascular Research 93:232–241

    Google Scholar 

  85. Zhao N, Liu H, Lilly B (2012) Reciprocal regulation of syndecan-2 and Notch signaling in vascular smooth muscle cells. Journal of Biological Chemistry 287:16111–16120

    Google Scholar 

  86. Ju R, Cirone P, Lin S, Griesbach H, Slusarski DC, Crews CM (2010) Activation of the planar cell polarity formin DAAM1 leads to inhibition of endothelial cell proliferation, migration, and angiogenesis. Proceedings of the National Academy of Sciences of the United States of America 107:6906–6911

    ADS  Google Scholar 

  87. Stefater JA, Lewkowich I, Rao S, Mariggi G, Carpenter AC, Burr AR, Fan J, Ajima R, Molkentin JD, Williams BO, Wills-Karp M, Pollard JW, Yamaguchi T, Ferrara N, Gerhardt H, Lang RA (2011) Regulation of angiogenesis by a non-canonical Wnt–Flt1 pathway in myeloid cells. Nature 474:511–515

    Google Scholar 

  88. Coultas L, Nieuwenhuis E, Anderson GA, Cabezas J, Nagy A, Henkelman RM, Hui CC, Rossant J (2010) Hedgehog regulates distinct vascular patterning events through VEGF-dependent and -independent mechanisms. Blood 116:653–660

    Google Scholar 

  89. Tedgui A, Lévy B (1994) Biologie de la paroi artrielle  [Biology of the arterial wall]. Masson, Paris

    Google Scholar 

  90. Wahlberg E (2005) Angiogenesis and arteriogenesis – the basics. svs.vascularweb.org/_CONTRIBUTION_ PAGES/Research/Basic_Articles/Angiogenesis_Wahlberg.html

    Google Scholar 

  91. Angiogenesis: an introduction. http://www.med.unibs.it/˜airc/

    Google Scholar 

  92. Ferrari G, Pintucci G, Seghezzi G, Hyman K, Galloway AC, Mignatti P (2006) VEGF, a prosurvival factor, acts in concert with TGF-beta1 to induce endothelial cell apoptosis. Proceedings of the National Academy of Sciences of the United States of America 103:17260–17265

    ADS  Google Scholar 

  93. Ferrara N, Davis-Smyth T (1997) The biology of vascular endothelial growth factor. Endocrine Reviews 18:4–25

    Google Scholar 

  94. www.sigmaaldrich.com/

    Google Scholar 

  95. Storkebaum E, Carmeliet P (2004) VEGF: a critical player in neurodegeneration. Journal of Clinical Investigation 113:14–18

    Google Scholar 

  96. Witmer AN, van Blijswijk BC, van Noorden CJ, Vrensen GF, Schlingemann RO (2004) In vivo angiogenic phenotype of endothelial cells and pericytes induced by vascular endothelial growth factor-A. Journal of Histochemistry and Cytochemistry 52:39–52

    Google Scholar 

  97. Dominguez MG, Hughes VC, Pan L, Simmons M, Daly C, Anderson K, Noguera-Troise I, Murphy AJ, Valenzuela DM, Davis S, Thurston G, Yancopoulos GD, Gale NW (2007) Vascular endothelial tyrosine phosphatase (VE-PTP)-null mice undergo vasculogenesis but die embryonically because of defects in angiogenesis. Proceedings of the National Academy of Sciences of the United States of America 104:3243–3248

    ADS  Google Scholar 

  98. Hiratsuka S, Nakamura K, Iwai S, Murakami M, Itoh T, Kijima H, Shipley JM, Senior RM, Shibuya M (2002) MMP9 induction by vascular endothelial growth factor receptor-1 is involved in lung-specific metastasis. Cancer Cell 2:289–300

    Google Scholar 

  99. LeCouter J, Moritz DR, Li B, Phillips GL, Liang XH, Gerber HP, Hillan KJ, Ferrara N (2003) Angiogenesis-independent endothelial protection of liver: role of VEGFR-1. Science 299:890–893

    ADS  Google Scholar 

  100. Wang S, Li X, Parra M, Verdin E, Bassel-Duby R, Olson EN (2008) Control of endothelial cell proliferation and migration by VEGF signaling to histone deacetylase 7. Proceedings of the National Academy of Sciences of the United States of America 105:7738–7743

    ADS  Google Scholar 

  101. Zeng H, Zhao D, Yang S, Datta K, Mukhopadhyay D (2003) Heterotrimeric Gα q /Gα11 proteins function upstream of vascular endothelial growth factor (VEGF) receptor-2 (KDR) phosphorylation in vascular permeability factor/VEGF signaling. The Journal of Biological Chemistry 278:20738–20745

    Google Scholar 

  102. Rousseau S, Houle F, Huot J (2000) Integrating the VEGF signals leading to actin-based motility in vascular endothelial cells. Trends in Cardiovascular Medicine 10:321–327

    Google Scholar 

  103. Landry J, Huot J (1999) Regulation of actin dynamics by stress-activated protein kinase 2 (SAPK2)-dependent phosphorylation of heat-shock protein of 27 kDa (Hsp27). Biochemical Society Symposium 64:79–89

    Google Scholar 

  104. Kobayashi M, Nishita M, Mishima T, Ohashi K, Mizuno K (2006) MAPKAPK-2-mediated LIM-kinase activation is critical for VEGF-induced actin remodeling and cell migration. EMBO Journal 25:713–726

    Google Scholar 

  105. Dawe HR, Minamide LS, Bamburg JR, Cramer LP (2003) ADF/cofilin controls cell polarity during fibroblast migration. Current Biology 13:252–257

    Google Scholar 

  106. Endo M, Ohashi K, Sasaki Y, Goshima Y, Niwa R, Uemura T, Mizuno K (2003) Control of growth cone motility and morphology by LIM kinase and Slingshot via phosphorylation and dephosphorylation of cofilin. Journal of Neuroscience 23:2527–2537

    Google Scholar 

  107. Del Valle-Pérez B, Martínez VG, Lacasa-Salavert C, Figueras A, Shapiro SS, Takafuta T, Casanovas O, Capellà G, Ventura F, Viñals F (2010) Filamin B plays a key role in vascular endothelial growth factor-induced endothelial cell motility through its interaction with Rac-1 and Vav-2. Journal of Biological Chemistry 285:10748–10760

    Google Scholar 

  108. Gavard J, Gutkind JS (2006) VEGF controls endothelial-cell permeability by promoting the bold beta-arrestin-dependent endocytosis of VE-cadherin. Nature – Cell Biology 8:1223–1234

    Google Scholar 

  109. Powis G, Kirkpatrick L (2004) Hypoxia inducible factor-1 as a cancer drug target. Molecular Cancer Therapeutics 3:647–654

    Google Scholar 

  110. Schmidt D, Textor B, Pein OT, Licht AH, Andrecht S, Sator-Schmitt M, Fusenig NE, Angel P, Schorpp-Kistner M (2007) Critical role for NF-κ B-induced JunB in VEGF regulation and tumor angiogenesis. EMBO Journal 26:710–719

    Google Scholar 

  111. Licht AH, Pein OT, Florin L, Hartenstein B, Reuter H, Arnold B, Lichter P, Angel P, Schorpp-Kistner MJ (2006) JunB is required for endothelial cell morphogenesis by regulating core-binding factor β. Journal of Cell Biology 175:981–991

    Google Scholar 

  112. Ray PS, Fox PL (2007) A post-transcriptional pathway represses monocyte VEGF-A expression and angiogenic activity. EMBO Journal 26:3360–3372

    Google Scholar 

  113. Fox PL, Mukhopadhyay R, Ray PS, Arif A (2008) The GAIT (interferon-γ-activated inhibitor of translation) system defines an auto-regulatory, negative-feedback circuit that controls inflammatory gene expression. FASEB Journal 22:601.4

    Google Scholar 

  114. Fischer C, Jonckx B, Mazzone M, Zacchigna S, Loges S, Pattarini L, Chorianopoulos E, Liesenborghs L, Koch M, De Mol M, Autiero M, Wyns S, Plaisance S, Moons L, van Rooijen N, Giacca M, Stassen JM, Dewerchin M, Collen D, Carmeliet P (2007) Anti-PlGF inhibits growth of VEGF(R)-inhibitor-resistant tumors without affecting healthy vessels. Cell 131:463–475

    Google Scholar 

  115. Greenberg JI, Shields DJ, Barillas SG, Acevedo LM, Murphy E, Huang J, Scheppke L, Stockmann C, Johnson RS, Angle N, Cheresh DA (2008) A role for VEGF as a negative regulator of pericyte function and vessel maturation. Nature 456:809–813

    ADS  Google Scholar 

  116. Dong J, Grunstein J, Tejada M, Peale F, Frantz G, Liang WC, Bai W, Yu L, Kowalski J, Liang X, Fuh G, Gerber HP, Ferrara N (2004) VEGF-null cells require PDGFR alpha signaling-mediated stromal fibroblast recruitment for tumorigenesis. EMBO Journal 23:2800–2810

    Google Scholar 

  117. Hou X, Kumar A, Lee C, Wang B, Arjunan P, Dong L, Maminishkis A, Tang Z, Li Y, Zhang F, Zhang SZ, Wardega P, Chakrabarty S, Liu B, Wu Z, Colosi P, Fariss RN, Lennartsson J, Nussenblatt R, Gutkind JS, Cao Y, Li X (2010) PDGF-CC blockade inhibits pathological angiogenesis by acting on multiple cellular and molecular targets. Proceedings of the National Academy of Sciences of the United States of America 107:12216–12221

    ADS  Google Scholar 

  118. Song S, Ewald AJ, Stallcup W, Werb Z, Bergers G (2005) PDGFRβ+ perivascular progenitor cells in tumours regulate pericyte differentiation and vascular survival. Nature – Cell Biology 7:870–879

    Google Scholar 

  119. Shojaei F, Wu X, Zhong C, Yu L, Liang XH, Yao J, Blanchard D, Bais C, Peale FV, van Bruggen N, Ho C, Ross J, Tan M, Carano RAD, Meng YG, Ferrara N (2007) Bv8 regulates myeloid-cell-dependent tumour angiogenesis. Nature 450:825–831

    ADS  Google Scholar 

  120. Weckbach LT, Groesser L, Borgolte J, Pagel JI, Pogoda F, Schymeinsky J, Mller-Hcker J, Shakibaei M, Muramatsu T, Deindl E, Walzog B (2012) Midkine acts as proangiogenic cytokine in hypoxia-induced angiogenesis. American Journal of Physiology – Heart and Circulatory Physiology 303:H429–H438

    Google Scholar 

  121. Ge G, Fernndez CA, Moses MA, Greenspan DS (2007) Bone morphogenetic protein 1 processes prolactin to a 17-kDa antiangiogenic factor. Proceedings of the National Academy of Sciences of the United States of America 104:10010–10015

    ADS  Google Scholar 

  122. Tritsaris K, Myren M, Ditlev SB, Hbschmann MV, van der Blom I, Hansen AJ, Olsen UB, Cao R, Zhang J, Jia T, Wahlberg E, Dissing S, Cao Y (2007) IL-20 is an arteriogenic cytokine that remodels collateral networks and improves functions of ischemic hind limbs. Proceedings of the National Academy of Sciences of the United States of America 104:15364–15369

    ADS  Google Scholar 

  123. Imhof BA, Aurrand-Lions M (2006) Angiogenesis and inflammation face off. Nature – Medicine 12:171–172

    Google Scholar 

  124. Fiedler U, Reiss Y, Scharpfenecker M, Grunow V, Koidl S, Thurston G, Gale NW, Witzenrath M, Rosseau S, Suttorp N, Sobke A, Herrmann M, Preissner KT, Vajkoczy P, Augustin HG (2006) Angiopoietin-2 sensitizes endothelial cells to TNF-alpha and has a crucial role in the induction of inflammation. Nature – Medicine 12:235–239

    Google Scholar 

  125. Suri C, Jones PF, Patan S, Bartunkova S, Maisonpierre PC, Davis S, Sato TN (1996) Yancopoulos G.D. Requisite role of angiopoietin-1, a ligand for the TIE2 receptor, during embryonic angiogenesis. Cell 87:1171–1180

    Google Scholar 

  126. Augustin HG, Koh GY, Thurston G, Alitalo K (2009) Control of vascular morphogenesis and homeostasis through the angiopoietin–Tie system. Nature Reviews – Molecular Cell Biology 10:165–177

    Google Scholar 

  127. Gale NW, Thurston G, Hackett SF, Renard R, Wang Q, McClain J, Martin C, Witte C, Witte MH, Jackson D, Suri C, Campochiaro PA, Wiegand SJ, Yancopoulos GD. Angiopoietin-2 is required for postnatal angiogenesis and lymphatic patterning, and only the latter role is rescued by Angiopoietin-1. Developmental Cell 3:411–423

    Google Scholar 

  128. Li Z, Huang H, Boland P, Dominguez MG, Burfeind P, Lai KM, Lin HC, Gale NW, Daly C, Auerbach W, Valenzuela D, Yancopoulos GD, Thurston G (2009) Embryonic stem cell tumor model reveals role of vascular endothelial receptor tyrosine phosphatase in regulating Tie2 pathway in tumor angiogenesis. Proceedings of the National Academy of Sciences of the United States of America 106:22399–22404

    ADS  Google Scholar 

  129. Kidoya H, Ueno M, Yamada Y, Mochizuki N, Nakata M, Yano T, Fujii R, Takakura N (2008) Spatial and temporal role of the apelin/APJ system in the caliber size regulation of blood vessels during angiogenesis. EMBO Journal 27:522–534

    Google Scholar 

  130. Kidoya H, Takakura N (2012) Biology of the apelin–APJ axis in vascular formation. Journal of Biochemistry 152:125–131

    Google Scholar 

  131. Ribatti D, Nico B, Spinazzi R, Vacca A, Nussdorfer GG (2005) The role of adrenomedullin in angiogenesis. Peptides 26:1670–1675

    Google Scholar 

  132. Smith RS Jr, Gao L, Bledsoe G, Chao L, Chao J (2009) Intermedin is a new angiogenic growth factor. American Journal of Physiology – Heart and Circulatory Physiology 297:H1040–H1047

    Google Scholar 

  133. English D, Welch Z, Kovala AT, Harvey K, Volpert OV, Brindley DN, Garcia JGN (2000) Sphingosine 1-phosphate released from platelets during clotting accounts for the potent endothelial cell chemotactic activity of blood serum and provides a novel link between hemostasis and angiogenesis. FASEB Journal 14:2255–2265

    Google Scholar 

  134. Watanabe K, Hasegawa Y, Yamashita H, Shimizu K, Ding Y, Abe M, Ohta H, Imagawa K, Hojo K, Maki H, Sonoda H, Sato Y (2004) Vasohibin as an endothelium-derived negative feedback regulator of angiogenesis. Journal of Clinical Investigation 114:898–907

    Google Scholar 

  135. Naito H, Kidoya H, Sato Y, Takakura N (2009) Induction and expression of anti-angiogenic vasohibins in the hematopoietic stem/progenitor cell population. Journal of Biochemistry 145:653–659

    Google Scholar 

  136. Shimizu K, Watanabe K, Yamashita H, Abe M, Yoshimatsu H, Ohta H, Sonoda H, Sato Y (2005) Gene regulation of a novel angiogenesis inhibitor, vasohibin, in endothelial cells. Biochemical and Biophysical Research Communications 327:700–706

    Google Scholar 

  137. Shibuya T, Watanabe K, Yamashita H, Shimizu K, Miyashita H, Abe M, Moriya T, Ohta H, Sonoda H, Shimosegawa T, Tabayashi K, Sato Y (2006) Isolation and characterization of vasohibin-2 as a homologue of VEGF-inducible endothelium-derived angiogenesis inhibitor vasohibin. Arteriosclerosis, Thrombosis, and Vascular Biology 26:1051–1057

    Google Scholar 

  138. Sonoda H, Ohta H, Watanabe K, Yamashita H, Kimura H, Sato Y (2006) Multiple processing forms and their biological activities of a novel angiogenesis inhibitor vasohibin. Biochemical and Biophysical Research Communications 342:640–646

    Google Scholar 

  139. Kimura H, Miyashita H, Suzuki Y, Kobayashi M, Watanabe K, Sonoda H, Ohta H, Fujiwara T, Shimosegawa T, Sato Y (2009) Distinctive localization and opposed roles of vasohibin-1 and vasohibin-2 in the regulation of angiogenesis. Blood 113:4810–4818

    Google Scholar 

  140. Sato Y (2010) The vasohibin family. Pharmaceuticals 3:433–440

    Google Scholar 

  141. Kuhnert F, Mancuso MR, Shamloo A, Wang HT, Choksi V, Florek M, Su H, Fruttiger M, Young WL, Heilshorn SC, Kuo CJ (2010) Essential regulation of CNS angiogenesis by the orphan G-protein-coupled receptor GPR124. Science 330:985–989

    ADS  Google Scholar 

  142. Mammoto A, Connor KM, Mammoto T, Yung CW, Huh D, Aderman CM, Mostoslavsky G, Smith LEH, Ingber DE (2009) A mechanosensitive transcriptional mechanism that controls angiogenesis. Nature 457:1103–1108

    ADS  Google Scholar 

  143. Wieland T, Mittmann C (2003) Regulators of G-protein signalling: multifunctional proteins with impact on signalling in the cardiovascular system. Pharmacology & Therapeutics 97:95–115

    Google Scholar 

  144. Albig AR, Schiemann WP (2005) Identification and characterization of regulator of G protein signaling 4 (RGS4) as a novel inhibitor of tubulogenesis: RGS4 inhibits mitogen-activated protein kinases and vascular endothelial growth factor signaling. Molecular Biology of the Cell 16:609–625

    Google Scholar 

  145. Hamzah J, Jugold M, Kiessling F, Rigby P, Manzur M, Marti HH, Rabie T, Kaden S, Grne HJ, Hmmerling GJ, Arnold B, Ganss R (2008) Vascular normalization in Rgs5-deficient tumours promotes immune destruction. Nature 453:410–414

    ADS  Google Scholar 

  146. Im E, Kazlauskas A (2006) Regulating angiogenesis at the level of PtdIns-4,5-P2. EMBO Journal 25:2075–2082

    Google Scholar 

  147. Graupera M, Guillermet-Guibert J, Foukas LC, Phng LK, Cain RJ, Salpekar A, Pearce W, Meek S, Millan J, Cutillas PR, Smith AJH, Ridley AJ, Ruhrberg C, Gerhardt H, Vanhaesebroeck B (2008) Angiogenesis selectively requires the p110alpha isoform of PI3K to control endothelial cell migration. Nature 453:662–666

    ADS  Google Scholar 

  148. Gambardella L, Hemberger M, Hughes B, Zudaire E, Andrews S, Vermeren S (2010) PI3K Signaling through the dual GTPase-activating protein ARAP3 is essential for developmental angiogenesis. Science Signaling 3:ra76

    Google Scholar 

  149. Chen J, Somanath PR, Razorenova O, Chen WS, Hay N, Bornstein P, Byzova TV (2005) Akt1 regulates pathological angiogenesis, vascular maturation and permeability in vivo. Nature – Medicine 11:1188–1196

    Google Scholar 

  150. Shiojima I, Sato K, Izumiya Y, Schiekofer S, Ito M, Liao R, Colucci WS, Walsh K (2005) Disruption of coordinated cardiac hypertrophy and angiogenesis contributes to the transition to heart failure. The Journal of Clinical Investigation 115:2059–2064

    Google Scholar 

  151. Sun JF, Phung T, Shiojima I, Felske T, Upalakalin JN, Feng D, Kornaga T, Dor T, Dvorak AM, Walsh K, Benjamin LE (2005) Microvascular patterning is controlled by fine-tuning the Akt signal. Proceedings of the National Academy of Sciences of the United States of America 102:128–133

    ADS  Google Scholar 

  152. Nagoshi T, Matsui T, Aoyama T, Leri A, Anversa P, Li L, Ogawa W, del Monte F, Gwathmey JK, Grazette L, Hemmings B, Kass DA, Champion HC, Rosenzweig A (2005) PI3K rescues the detrimental effects of chronic Akt activation in the heart during ischemia/reperfusion injury. The Journal of Clinical Investigation 115:2059–2064

    Google Scholar 

  153. Bornstein P, Agah A, Kyriakides TR (2004) The role of thrombospondins 1 and 2 in the regulation of cell-matrix interactions, collagen fibril formation, and the response to injury. The International Journal of Biochemistry & Cell Biology 36:1115–1125

    Google Scholar 

  154. Lee NV, Sato M, Annis DS, Loo JA, Wu L, Mosher DF, Iruela-Arispe ML (2006) ADAMTS1 mediates the release of antiangiogenic polypeptides from TSP1 and 2. EMBO Journal 25:5270–5283

    Google Scholar 

  155. Kitamura T, Asai N, Enomoto A, Maeda K, Kato T, Ishida M, Jiang P, Watanabe T, Usukura J, Kondo T, Costantini F, Murohara T, Takahashi M (2008) Regulation of VEGF-mediated angiogenesis by the Akt/PKB substrate Girdin. Nature – Cell Biology 10:329–337

    Google Scholar 

  156. Alfranca A, Lpez-Oliva JM, Gens L, Lpez-Maderuelo D, Mirones I, Salvado D, Quesada AJ, Arroyo AG, Redondo JM (2008) PGE2 induces angiogenesis via MT1-MMP-mediated activation of the TGFbeta/Alk5 signaling pathway. Blood 112:1120–1128

    Google Scholar 

  157. van den Berg YW, van den Hengel LG, Myers HR, Ayachi O, Jordanova E, Ruf W, Spek CA, Reitsma PH, Bogdanov VY, Versteeg HH (2009) Alternatively spliced tissue factor induces angiogenesis through integrin ligation. Proceedings of the National Academy of Sciences of the United States of America 106:19497–19502

    ADS  Google Scholar 

  158. Yao YY, Yin H, Shen B, Smith RS, Liu Y, Gao L, Chao L, Chao J (2008) Tissue kallikrein promotes neovascularization and improves cardiac function by the Akt-glycogen synthase kinase-3β pathway. Cardiovascular Research 80:354–364

    Google Scholar 

  159. Emanueli C, Salis MB, Van Linthout S, Meloni M, Desortes E, Silvestre JS, Clergue M, Figueroa CD, Gadau S, Condorelli G, Madeddu P (2004) Akt/protein kinase B and endothelial nitric oxide synthase mediate muscular neovascularization induced by tissue kallikrein gene transfer. Circulation 110:1638–1644

    Google Scholar 

  160. Coffman LG, Parsonage D, D’Agostino R Jr, Torti FM, Torti SV (2009) Regulatory effects of ferritin on angiogenesis. Proceedings of the National Academy of Sciences of the United States of America 106:570–575

    ADS  Google Scholar 

  161. Khan MM, Liu Y, Khan ME, Gilman ML, Khan ST, Bromberg ME, Colman RW (2010) Upregulation of tissue factor in monocytes by cleaved high molecular weight kininogen is dependent on TNFα and IL-1β. American Journal of Physiology – Heart and Circulatory Physiology 298:H652–H658

    Google Scholar 

  162. Wang R (2012) Shared signaling pathways among gasotransmitters. Proceedings of the National Academy of Sciences of the United States of America 109:8801–8802

    ADS  Google Scholar 

  163. Tang G, Wu L, Liang W, Wang R (2005) Direct stimulation of K ATP channels by exogenous and endogenous hydrogen sulfide in vascular smooth muscle cells. Molecular Pharmacology 68:1757–1764

    Google Scholar 

  164. Papapetropoulos A, Pyriochou A, Altaany Z, Yang G, Marazioti A, Zhou Z, Jeschke MG, Branski LK, Herndon DN, Wang R, Szabó C (2009) Hydrogen sulfide is an endogenous stimulator of angiogenesis. Proceedings of the National Academy of Sciences of the United States of America 106:21972–21977

    ADS  Google Scholar 

  165. Coletta C, Papapetropoulos A, Erdelyi K, Olah G, Módis K, Panopoulos P, Asimakopoulou A, Gerö D, Sharina I, Martin E, Szabo C (2012) Hydrogen sulfide and nitric oxide are mutually dependent in the regulation of angiogenesis and endothelium-dependent vasorelaxation. Proceedings of the National Academy of Sciences of the United States of America 109:9161–9166

    ADS  Google Scholar 

  166. West XZ, Malinin NL, Merkulova AA, Tischenko M, Kerr BA, Borden EC, Podrez EA, Salomon RG, Byzova TV (2010) Oxidative stress induces angiogenesis by activating TLR2 with novel endogenous ligands. Nature 467:972–976

    ADS  Google Scholar 

  167. Zhang M, Brewer AC, Schröder K, Santos CX, Grieve DJ, Wang M, Anilkumar N, Yu B, Dong X, Walker SJ, Brandes RP, Shah AM (2010) NADPH oxidase-4 mediates protection against chronic load-induced stress in mouse hearts by enhancing angiogenesis. Proceedings of the National Academy of Sciences of the United States of America 107:18121–18126

    ADS  Google Scholar 

  168. Kilarski WW, Samolov B, Petersson L, Kvanta A, Gerwins P (2009) Biomechanical regulation of blood vessel growth during tissue vascularization. Nature – Medicine 15:657–664

    Google Scholar 

  169. Yung YC, Chae J, Buehler MJ, Hunter CP, Mooney DJ (2009) Cyclic tensile strain triggers a sequence of autocrine and paracrine signaling to regulate angiogenic sprouting in human vascular cells. Proceedings of the National Academy of Sciences of the United States of America 106:15279–15284

    ADS  Google Scholar 

  170. Song JW, Munn LL (2011) Fluid forces control endothelial sprouting. Proceedings of the National Academy of Sciences of the United States of America 108:15342–15347

    ADS  Google Scholar 

  171. Rivron NC, Vrij EJ, Rouwkema J, Le Gac S, van den Berg A, Truckenmller RK, van Blitterswijk CA (2012) Tissue deformation spatially modulates VEGF signaling and angiogenesis. Proceedings of the National Academy of Sciences of the United States of America 109:6886–6891

    ADS  Google Scholar 

  172. Boerckel JD, Uhrig BA, Willett NJ, Huebsch N, Guldberg RE (2011) Mechanical regulation of vascular growth and tissue regeneration in vivo. Proceedings of the National Academy of Sciences of the United States of America 108:E674–E680

    ADS  Google Scholar 

  173. Charlesworth PJS, Harris AL (2006) Mechanisms of disease: angiogenesis in urologic malignancies. Nature – Clinical Practice – Urology 3:157–169

    Google Scholar 

  174. Murohara T, Ikeda H, Duan J, Shintani S, Sasaki KI, Eguchi H, Onitsuka I, Matsui K, Imaizumi T (2000) Transplanted cord blood-derived endothelial precursor cells augment postnatal neovascularization. Journal of Clinical Investigation 105:1527–1536

    Google Scholar 

  175. Madeddu P, Emanueli C, Pelosi E, Salis MB, Cerio AM, Bonanno G, Patti M, Stassi G, Condorelli G, Peschle C (2004) Transplantation of low dose CD34+KDR+ cells promotes vascular and muscular regeneration in ischemic limbs. FASEB Journal 18:1737–1739

    Google Scholar 

  176. Bir SC, **ong Y, Kevil CG, Luo J (2012) Emerging role of PKA/eNOS pathway in therapeutic angiogenesis for ischaemic tissue diseases. Cardiovascular Research 95:7–18

    Google Scholar 

  177. Ambati BK, Nozaki M, Singh N, Takeda A, Jani PD, Suthar T, Albuquerque RJC, Richter E, Sakurai E, Newcomb MT, Kleinman ME, Caldwell RB, Lin Q, Ogura Y, Orecchia A, Samuelson DA, Agnew DW, St Leger J, Green WR, Mahasreshti PJ, Curiel DT, Kwan D, Marsh H, Ikeda I, Leiper LJ, Collinson JM, Bogdanovich S, Khurana TS, Shibuya M, Baldwin ME, Ferrara N, Gerber HP, De Falco S, Witta J, Baffi JZ, Raisler BJ, Ambati J (2006) Corneal avascularity is due to soluble VEGF receptor-1. Nature 443:993–997

    ADS  Google Scholar 

  178. Karagiannis ED, Popel AS (2008) A systematic methodology for proteome-wide identification of peptides inhibiting the proliferation and migration of endothelial cells. Proceedings of the National Academy of Sciences of the United States of America 105:13775–13780

    ADS  Google Scholar 

  179. Singhn AJ, Meyer RD, Navruzbekov G, Shelke R, Duan L, Band H, Leeman SE, Rahimi N (2007) A critical role for the E3-ligase activity of c-Cbl in VEGFR-2-mediated PLCγ 1 activation and angiogenesis. Proceedings of the National Academy of Sciences of the United States of America 104:5413–5418

    ADS  Google Scholar 

  180. Streit M, Velasco P, Brown LF, Skobe M, Richard L, Riccardi L, Lawler J, Detmar M (1999) Overexpression of thrombospondin-1 decreases angiogenesis and inhibits the growth of human cutaneous squamous cell carcinomas. American Journal of Pathology 155:441–452

    Google Scholar 

  181. Hamzah J, Jugold M, Kiessling F, Rigby P, Manzur M, Marti HH, Rabie T, Kaden S, Grne HJ, Hmmerling GJ, Arnold B, Ganss R (2008) Vascular normalization in Rgs5-deficient tumours promotes immune destruction. Nature 453:410-414

    ADS  Google Scholar 

  182. Seaman S, Stevens J, Yang MY, Logsdon D, Graff-Cherry C, St. Croix B (2007) Genes that distinguish physiological and pathological angiogenesis. Cancer Cell 11:539–554

    Google Scholar 

  183. Ardi VC, Kupriyanova TA, Deryugina EI, Quigley JP (2007) Human neutrophils uniquely release TIMP-free MMP-9 to provide a potent catalytic stimulator of angiogenesis. Proceedings of the National Academy of Sciences of the United States of America 104:20262–20267

    ADS  Google Scholar 

  184. Lee DY, Deng Z, Wang CH, Yang BB (2007) MicroRNA-378 promotes cell survival, tumor growth, and angiogenesis by targeting SuFu and Fus-1 expression. Proceedings of the National Academy of Sciences of the United States of America 104:20350–20355

    ADS  Google Scholar 

  185. Gomez DE, Alonso DF, Yoshiji H, Thorgeirsson UP (1997) Tissue inhibitors of metalloproteinases: structure, regulation and biological functions. European Journal of Cell Biology 74:111–122

    Google Scholar 

  186. Stromblad S, Becker JC, Yebra M, Brooks PC, Cheresh DA (1996) Suppression of p53 activity and p21WAF1/CIP1 expression by vascular cell integrin alphaVbeta3 during angiogenesis. Journal of Clinical Investigation 98:426–433

    Google Scholar 

  187. Mizukami Y, Jo WS, Duerr EM, Gala M, Li J, Zhang X, Zimmer MA, Iliopoulos O, Zukerberg LR, Kohgo Y, Lynch MP, Rueda BR, Chung DC (2005) Induction of interleukin-8 preserves the angiogenic response in HIF-1-deficient colon cancer cells. Nature Medicine 11:992–997

    Google Scholar 

  188. Vincent L, Kermani P, Young LM, Cheng J, Zhang F, Shido K, Lam G, Bompais-Vincent H, Zhu Z, Hicklin DJ, Bohlen P, Chaplin DJ, May C, Rafii S (2005) Combretastatin A4 phosphate induces rapid regression of tumor neovessels and growth through interference with vascular endothelial-cadherin signaling. The Journal of Clinical Investigation 115:2992–3006

    Google Scholar 

  189. Thijssen VLJL, Postel R, Brandwijk RJMGE, Dings RPM, Nesmelova I, Satijn S, Verhofstad N, Nakabeppu Y, Baum LG, Bakkers J, Mayo KH, Poirier F, Griffioen AW (2006) Galectin-1 is essential in tumor angiogenesis and is a target for antiangiogenesis therapy. Proceedings of the National Academy of Sciences of the United States of America 103:15975–15980

    ADS  Google Scholar 

  190. Bernardi R, Guernah I, ** D, Grisendi S, Alimonti A, Teruya-Feldstein J, Cordon-Cardo C, Simon MC, Rafii S, Pandolfi PP (2006) PML inhibits HIF-1alpha translation and neoangiogenesis through repression of mTOR. Nature 442:779–785

    ADS  Google Scholar 

  191. Mancuso MR, Davis R, Norberg SM, O’Brien S, Sennino B, Nakahara T, Yao VJ, Inai T, Brooks P, Freimark B, Shalinsky DR, Hu-Lowe DD, McDonald DM (2006) Rapid vascular regrowth in tumors after reversal of VEGF inhibition. Journal of Clinical Investigation 116:2610–2621

    Google Scholar 

  192. Juarez JC, Manuia M, Burnett M, Betancourt O, Boivin B, Shaw DE, Tonks NK, Mazar AP, Doate F (2008) Superoxide dismutase 1 (SOD1) is essential for H2O2-mediated oxidation and inactivation of phosphatases in growth factor signaling. Proceedings of the National Academy of Sciences of the United States of America 105:7147–7152

    ADS  Google Scholar 

  193. Kleinman ME, Yamada K, Takeda A, Chandrasekaran V, Nozaki M, Baffi JZ, Albuquerque RJC, Yamasaki S, Itaya M, Pan Y, Appukuttan B, Gibbs D, Yang Z, Karik K, Ambati BK, Wilgus TA, DiPietro LA, Sakurai E, Zhang K, Smith JR, Taylor EW, Ambati J (2008) Sequence- and target-independent angiogenesis suppression by siRNA via TLR3. Nature 452:591–597

    ADS  Google Scholar 

  194. Goel S, Fukumura D, Jain RK (2012) Normalization of the tumor vasculature through oncogenic inhibition: An emerging paradigm in tumor biology. Proceedings of the National Academy of Sciences of the United States of America 109:E1214

    Google Scholar 

  195. Bottos A, Martini M, Di Nicolantonio F, Comunanza V, Maione F, Minassi A, Appendino G, Bussolino F, Bardelli A (2012) Targeting oncogenic serine/threonine-protein kinase BRAF in cancer cells inhibits angiogenesis and abrogates hypoxia. Proceedings of the National Academy of Sciences of the United States of America 109:E353–E359

    ADS  Google Scholar 

  196. Oliver G, Harvey N (2002) A stepwise model of the development of lymphatic vasculature. Annals of the New York Academy of Sciences 979:159–165

    ADS  Google Scholar 

  197. Gale NW, Prevo R, Espinosa J, Ferguson DJ, Dominguez MG, Yancopoulos GD, Thurston G, Jackson DG (2007) Normal lymphatic development and function in mice deficient for the lymphatic hyaluronan receptor LYVE-1. Molecular and Cellular Biology 27:595–604

    Google Scholar 

  198. Saharinen P, Petrova TV (2004) Molecular regulation of lymphangiogenesis. Annals of the New York Academy of Sciences 1014:76–87

    ADS  Google Scholar 

  199. Franois M, Caprini A, Hosking B, Orsenigo F, Wilhelm D, Browne C, Paavonen K, Karnezis T, Shayan R, Downes M, Davidson T, Tutt D, Cheah KSE, Stacker SA, Muscat GEO, Achen MG, Dejana E, Koopman P (2008) Sox18 induces development of the lymphatic vasculature in mice. Nature 456:643–647

    ADS  Google Scholar 

  200. Watabe T (2012) Roles of transcriptional network during the formation of lymphatic vessels. Journal of Biochemistry 152:213–220

    ADS  Google Scholar 

  201. Dagenais SL, Hartsough RL, Erickson RP, Witte MH, Butler MG, Glover TW (2004) Foxc2 is expressed in develo** lymphatic vessels and other tissues associated with lymphedema-distichiasis syndrome. Gene Expression Patterns 4:611–619

    Google Scholar 

  202. Petrova TV, Karpanen T, Norrmén C, Mellor R, Tamakoshi T, Finegold D, Ferrell R, Kerjaschki D, Mortimer P, Ylä-Herttuala S, Miura N, Alitalo K (2004) Defective valves and abnormal mural cell recruitment underlie lymphatic vascular failure in lymphedema distichiasis. Nature – Medicine 10:974–981

    Google Scholar 

  203. Tammela T, Zarkada G, Wallgard E, Murtomäk A, Suchting S, Wirzenius M, Waltari M, Hellström M, Schomber T, Peltonen R, Freitas C, Duarte A, Isoniemi H, Laakkonen P, Christofori G, Ylä-Herttuala S, Shibuya M, Pytowski B, Eichmann A, Betsholtz C, Alitalo K (2008) Blocking VEGFR3 suppresses angiogenic sprouting and vascular network formation. Nature 454:656–660

    ADS  Google Scholar 

  204. Alitalo K, Tammela T, Petrova TV (2005) Lymphangiogenesis in development and human disease. Nature 438:946–953

    ADS  Google Scholar 

  205. Böhmer R, Neuhaus B, Bühren S, Zhang D, Stehling M, Böck B, Kiefer F (2010) Regulation of developmental lymphangiogenesis by Syk + leukocytes. Developmental Cell 18:437–449

    Google Scholar 

  206. Tammela T, Petrova TV, Alitalo K (2005) Molecular lymphangiogenesis: new players. Trends in Cell Biology 15:434–441

    Google Scholar 

  207. Gale NW, Thurston G, Hackett SF, Renard R, Wang Q, McClain J, Martin C, Witte C, Witte MH, Jackson D, Suri C, Campochiaro PA, Wiegand SJ, Yancopoulos GD (2002) Angiopoietin-2 is required for postnatal angiogenesis and lymphatic patterning, and only the latter role is rescued by Angiopoietin-1. Developmental Cell 3:411–423

    Google Scholar 

  208. Mäkinen T, Adams RH, Bailey J, Lu Q, Ziemiecki A, Alitalo K, Klein R, Wilkinson GA (2005) PDZ interaction site in ephrinB2 is required for the remodeling of lymphatic vasculature. Genes and Development 19:397–410

    Google Scholar 

  209. Foo SS, Turner CJ, Adams S, Compagni A, Aubyn D, Kogata N, Lindblom P, Shani M, Zicha D, Adams RH (2006) Ephrin-B2 controls cell motility and adhesion during blood-vessel-wall assembly. Cell 124:161–173

    Google Scholar 

  210. Qutub AA, Gabhann FM, Karagiannis ED, Vempati P, Popel AS (2009) Multiscale models of angiogenesis. IEEE Engineering in Medicine and Biology Magazine 28:14–31

    Google Scholar 

  211. Schneider M, Reichold J, Weber B, Székely G, Hirsch S (2012) Tissue metabolism driven arterial tree generation. Medical Image Analysis (dx.doi.org/10.1016/j.media.2012.04.009)

    Google Scholar 

  212. d’Onofrio A, Gandolfi A (2009) A family of models of angiogenesis and anti-angiogenesis anti-cancer therapy. Mathematical Medicine and Biology 26:63–95

    Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Rights and permissions

Reprints and permissions

Copyright information

© 2013 Springer Science+Business Media New York

About this chapter

Cite this chapter

Thiriet, M. (2013). Vasculature Growth. In: Tissue Functioning and Remodeling in the Circulatory and Ventilatory Systems. Biomathematical and Biomechanical Modeling of the Circulatory and Ventilatory Systems, vol 5. Springer, New York, NY. https://doi.org/10.1007/978-1-4614-5966-8_10

Download citation

  • DOI: https://doi.org/10.1007/978-1-4614-5966-8_10

  • Published:

  • Publisher Name: Springer, New York, NY

  • Print ISBN: 978-1-4614-5965-1

  • Online ISBN: 978-1-4614-5966-8

  • eBook Packages: Physics and AstronomyPhysics and Astronomy (R0)

Publish with us

Policies and ethics

Navigation