Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

VEGFR-3 controls tip to stalk conversion at vessel fusion sites by reinforcing Notch signalling

Abstract

Angiogenesis, the growth of new blood vessels, involves specification of endothelial cells to tip cells and stalk cells, which is controlled by Notch signalling, whereas vascular endothelial growth factor receptor (VEGFR)-2 and VEGFR-3 have been implicated in angiogenic sprouting. Surprisingly, we found that endothelial deletion of Vegfr3, but not VEGFR-3-blocking antibodies, postnatally led to excessive angiogenic sprouting and branching, and decreased the level of Notch signalling, indicating that VEGFR-3 possesses passive and active signalling modalities. Furthermore, macrophages expressing the VEGFR-3 and VEGFR-2 ligand VEGF-C localized to vessel branch points, and Vegfc heterozygous mice exhibited inefficient angiogenesis characterized by decreased vascular branching. FoxC2 is a known regulator of Notch ligand and target gene expression, and Foxc2+/−;Vegfr3+/− compound heterozygosity recapitulated homozygous loss of Vegfr3. These results indicate that macrophage-derived VEGF-C activates VEGFR-3 in tip cells to reinforce Notch signalling, which contributes to the phenotypic conversion of endothelial cells at fusion points of vessel sprouts.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Blood vascular hyperplasia and excessive filopodia projection in mice with a targeted deletion of Vegfr3 in the endothelium.
Figure 2: Role of VEGFR-3 tyrosine kinase activity in angiogenesis.
Figure 3: An increased level of VEGFR-2 signalling contributes to vascular hyperplasia in Vegfr3iΔEC retinas.
Figure 4: A decreased level of Notch signalling underlies excessive angiogenesis in Vegfr3iΔEC retinas.
Figure 5: Vegfc haploinsufficiency leads to instability of sprout fusion points and inefficient angiogenesis.
Figure 6: VEGF-C promotes Notch signalling in endothelial cells through VEGFR-3 and PI(3)K.
Figure 7: VEGFR-3 interacts with the transcription factor FoxC2 to control angiogenesis.

Similar content being viewed by others

Zixuan Zhao, Xinyi Chen, … Hanry Yu

References

  1. Ferrara, N. et al. Heterozygous embryonic lethality induced by targeted inactivation of the VEGF gene. Nature 380, 438–442 (1996).

    Article  Google Scholar 

  2. Carmeliet, P. et al. Abnormal blood vessel development and lethality in embryos lacking a single VEGF allele. Nature 380, 435–439 (1996).

    Article  CAS  Google Scholar 

  3. Shalaby, F. et al. Failure of blood island formation and vasculogenesis in Flk-1-deficient mice. Nature 376, 62–66 (1995).

    Article  CAS  Google Scholar 

  4. Gille, H. et al. 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. 276, 3222–3230 (2001).

    Article  CAS  Google Scholar 

  5. Tammela, T. & Alitalo, K. Lymphangiogenesis: molecular mechanisms and future promise. Cell 140, 460–476 (2010).

    Article  CAS  Google Scholar 

  6. Dixelius, J. et al. Ligand-induced vascular endothelial growth factor receptor-3 (VEGFR-3) heterodimerization with VEGFR-2 in primary lymphatic endothelial cells regulates tyrosine phosphorylation sites. J. Biol. Chem. 278, 40973–40979 (2003).

    Article  CAS  Google Scholar 

  7. Nilsson, I. et al. VEGF receptor 2/-3 heterodimers detected in situ by proximity ligation on angiogenic sprouts. EMBO J. 29, 1377–1388 (2010).

    Article  CAS  Google Scholar 

  8. Dumont, D. J. et al. Cardiovascular failure in mouse embryos deficient in VEGF receptor-3. Science 282, 946–949 (1998).

    Article  CAS  Google Scholar 

  9. Covassin, L. D., Villefranc, J. A., Kacergis, M. C., Weinstein, B. M. & Lawson, N. D. Distinct genetic interactions between multiple Vegf receptors are required for development of different blood vessel types in zebrafish. Proc. Natl Acad. Sci. USA 103, 6554–6559 (2006).

    Article  CAS  Google Scholar 

  10. Kaipainen, A. et al. Expression of the fms-like tyrosine kinase FLT4 gene becomes restricted to lymphatic endothelium during development. Proc. Natl Acad. Sci. USA 92, 3566–3570 (1995).

    Article  CAS  Google Scholar 

  11. Lohela, M., Helotera, H., Haiko, P., Dumont, D. J. & Alitalo, K. Transgenic induction of vascular endothelial growth factor-C is strongly angiogenic in mouse embryos but leads to persistent lymphatic hyperplasia in adult tissues. Am. J. Pathol. 173, 1891–1901 (2008).

    Article  CAS  Google Scholar 

  12. Paavonen, K., Puolakkainen, P., Jussila, L., Jahkola, T. & Alitalo, K. Vascular endothelial growth factor receptor-3 in lymphangiogenesis in wound healing. Am. J. Pathol. 156, 1499–1504 (2000).

    Article  CAS  Google Scholar 

  13. Valtola, R. et al. VEGFR-3 and its ligand VEGF-C are associated with angiogenesis in breast cancer. Am. J. Pathol. 154, 1381–1390 (1999).

    Article  CAS  Google Scholar 

  14. Tammela, T. et al. Blocking VEGFR-3 suppresses angiogenic sprouting and vascular network formation. Nature 454, 656–660 (2008).

    Article  CAS  Google Scholar 

  15. Karkkainen, M. J. et al. Vascular endothelial growth factor C is required for sprouting of the first lymphatic vessels from embryonic veins. Nat. Immunol. 5, 74–80 (2004).

    Article  CAS  Google Scholar 

  16. Baldwin, M. E. et al. Vascular endothelial growth factor D is dispensable for development of the lymphatic system. Mol. Cell Biol. 25, 2441–2449 (2005).

    Article  CAS  Google Scholar 

  17. Haiko, P. et al. Deletion of vascular endothelial growth factor C (VEGF-C) and VEGF-D is not equivalent to VEGF receptor 3 deletion in mouse embryos. Mol. Cell Biol. 28, 4843–4850 (2008).

    Article  CAS  Google Scholar 

  18. Gerhardt, H. et al. VEGF guides angiogenic sprouting utilizing endothelial tip cell filopodia. J. Cell Biol. 161, 1163–1177 (2003).

    Article  CAS  Google Scholar 

  19. Hellstrom, M. et al. Dll4 signalling through Notch1 regulates formation of tip cells during angiogenesis. Nature 445, 776–780 (2007).

    Article  CAS  Google Scholar 

  20. Suchting, S. et al. The Notch ligand Delta-like 4 negatively regulates endothelial tip cell formation and vessel branching. Proc. Natl Acad. Sci. USA 104, 3225–3230 (2007).

    Article  CAS  Google Scholar 

  21. Roca, C. & Adams, R. H. Regulation of vascular morphogenesis by Notch signaling. Genes Dev. 21, 2511–2524 (2007).

    Article  CAS  Google Scholar 

  22. Kamei, M. et al. Endothelial tubes assemble from intracellular vacuoles in vivo. Nature 442, 453–456 (2006).

    Article  CAS  Google Scholar 

  23. Strilic, B. et al. The molecular basis of vascular lumen formation in the developing mouse aorta. Dev. Cell 17, 505–515 (2009).

    Article  CAS  Google Scholar 

  24. Fantin, A. et al. Tissue macrophages act as cellular chaperones for vascular anastomosis downstream of VEGF-mediated endothelial tip cell induction. Blood 116, 829–840 (2010).

    Article  CAS  Google Scholar 

  25. Kubota, Y. et al. M-CSF inhibition selectively targets pathological angiogenesis and lymphangiogenesis. J. Exp. Med. 206, 1089–1102 (2009).

    Article  CAS  Google Scholar 

  26. Qian, B. Z. & Pollard, J. W. Macrophage diversity enhances tumor progression and metastasis. Cell 141, 39–51 (2010).

    Article  CAS  Google Scholar 

  27. Siekmann, A. F. & Lawson, N. D. Notch signalling limits angiogenic cell behaviour in developing zebrafish arteries. Nature 445, 781–784 (2007).

    Article  CAS  Google Scholar 

  28. Benedito, R. et al. The notch ligands Dll4 and Jagged1 have opposing effects on angiogenesis. Cell 137, 1124–1135 (2009).

    Article  CAS  Google Scholar 

  29. Claxton, S. et al. Efficient, inducible Cre-recombinase activation in vascular endothelium. Genesis 46, 74–80 (2008).

    Article  CAS  Google Scholar 

  30. Le Bras, B. et al. VEGF-C is a trophic factor for neural progenitors in the vertebrate embryonic brain. Nat. Neurosci. 9, 340–348 (2006).

    Article  CAS  Google Scholar 

  31. Skobe, M. et al. Concurrent induction of lymphangiogenesis, angiogenesis, and macrophage recruitment by vascular endothelial growth factor-C in melanoma. Am. J. Pathol. 159, 893–903 (2001).

    Article  CAS  Google Scholar 

  32. Karkkainen, M. J. et al. A model for gene therapy of human hereditary lymphedema. Proc. Natl Acad. Sci. USA 98, 12677–12682 (2001).

    Article  CAS  Google Scholar 

  33. Galvagni, F. et al. Endothelial cell adhesion to the extracellular matrix induces c-Src-dependent VEGFR-3 phosphorylation without the activation of the receptor intrinsic kinase activity. Circ. Res. 106, 1839–1848 (2010).

    Article  CAS  Google Scholar 

  34. Jakobsson, L. et al. Endothelial cells dynamically compete for the tip cell position during angiogenic sprouting. Nat. Cell Biol. 12, 943–953 (2010).

    Article  CAS  Google Scholar 

  35. Wiktor-Jedrzejczak, W. W., Ahmed, A., Szczylik, C. & Skelly, R. R. Hematological characterization of congenital osteopetrosis in op/op mouse. Possible mechanism for abnormal macrophage differentiation. J. Exp. Med. 156, 1516–1527 (1982).

    Article  CAS  Google Scholar 

  36. Takeshita, K. et al. Critical role of endothelial Notch1 signaling in postnatal angiogenesis. Circ. Res. 100, 70–78 (2007).

    Article  CAS  Google Scholar 

  37. Yamamizu, K. et al. Convergence of Notch and β-catenin signaling induces arterial fate in vascular progenitors. J. Cell Biol. 189, 325–338 (2010).

    Article  CAS  Google Scholar 

  38. Hayashi, H. & Kume, T. Foxc transcription factors directly regulate Dll4 and Hey2 expression by interacting with the VEGF-Notch signaling pathways in endothelial cells. PLoS One 3, e2401 (2008).

    Article  CAS  Google Scholar 

  39. Burgering, B. M. A brief introduction to FOXOlogy. Oncogene 27, 2258–2262 (2008).

    Article  CAS  Google Scholar 

  40. Petrova, T. V. et al. Defective valves and abnormal mural cell recruitment underlie lymphatic vascular failure in lymphedema distichiasis. Nat. Med. 10, 974–981 (2004).

    Article  CAS  Google Scholar 

  41. Norrmen, C. et al. FOXC2 controls formation and maturation of lymphatic collecting vessels through cooperation with NFATc1. J. Cell Biol. 185, 439–457 (2009).

    Article  CAS  Google Scholar 

  42. Laakkonen, P. et al. Vascular endothelial growth factor receptor 3 is involved in tumor angiogenesis and growth. Cancer Res. 67, 593–599 (2007).

    Article  CAS  Google Scholar 

  43. Davis, G. E. & Senger, D. R. Endothelial extracellular matrix: biosynthesis, remodeling, and functions during vascular morphogenesis and neovessel stabilization. Circ. Res. 97, 1093–1107 (2005).

    Article  CAS  Google Scholar 

  44. Zhang, L. et al. VEGFR-3 ligand-binding and kinase activity are required for lymphangiogenesis but not for angiogenesis. Cell Res. 20, 1319–1331 (2010).

    Article  CAS  Google Scholar 

  45. Nilsson, I. et al. VEGF receptor 2/-3 heterodimers detected in situ by proximity ligation on angiogenic sprouts. EMBO J. 29, 1377–1388 (2010).

    Article  CAS  Google Scholar 

  46. Wang, Y. et al. Ephrin-B2 controls VEGF-induced angiogenesis and lymphangiogenesis. Nature 465, 483–486 (2010).

    Article  CAS  Google Scholar 

  47. Sawamiphak, S. et al. Ephrin-B2 regulates VEGFR2 function in developmental and tumour angiogenesis. Nature 465, 487–491 (2010).

    Article  CAS  Google Scholar 

  48. Saharinen, P. et al. Claudin-like protein 24 interacts with the VEGFR-2 and VEGFR-3 pathways and regulates lymphatic vessel development. Genes Dev. 24, 875–880 (2010).

    Article  CAS  Google Scholar 

  49. Whitaker, G. B., Limberg, B. J. & Rosenbaum, J. S. Vascular endothelial growth factor receptor-2 and neuropilin-1 form a receptor complex that is responsible for the differential signaling potency of VEGF(165) and VEGF(121). J. Biol. Chem. 276, 25520–25531 (2001).

    Article  CAS  Google Scholar 

  50. Carmeliet, P. et al. Targeted deficiency or cytosolic truncation of the VE-cadherin gene in mice impairs VEGF-mediated endothelial survival and angiogenesis. Cell 98, 147–157 (1999).

    Article  CAS  Google Scholar 

  51. Shawber, C. J. et al. Notch alters VEGF responsiveness in human and murine endothelial cells by direct regulation of VEGFR-3 expression. J. Clin. Invest. 117, 3369–3382 (2007).

    Article  CAS  Google Scholar 

  52. Ober, E. A. et al. Vegfc is required for vascular development and endoderm morphogenesis in zebrafish. EMBO Rep. 5, 78–84 (2004).

    Article  CAS  Google Scholar 

  53. De Palma, M., Venneri, M. A., Roca, C. & Naldini, L. Targeting exogenous genes to tumor angiogenesis by transplantation of genetically modified hematopoietic stem cells. Nat. Med. 9, 789–795 (2003).

    Article  CAS  Google Scholar 

  54. Mäkinen, T. et al. Isolated lymphatic endothelial cells transduce growth, survival and migratory signals via the VEGF-C receptor VEGFR-3. EMBO J. 20, 4762–4773 (2001).

    Article  Google Scholar 

  55. Soriano, P. Generalized lacZ expression with the ROSA26 Cre reporter strain. Nat. Genet. 21, 70–71 (1999).

    Article  CAS  Google Scholar 

  56. Iida, K. et al. Essential roles of the winged helix transcription factor MFH-1 in aortic arch patterning and skeletogenesis. Development 124, 4627–4638 (1997).

    PubMed  CAS  Google Scholar 

  57. Pytowski, B. et al. Complete and specific inhibition of adult lymphatic regeneration by a novel VEGFR-3 neutralizing antibody. J. Natl Cancer Inst. 97, 14–21 (2005).

    Article  CAS  Google Scholar 

  58. Prewett, M. et al. Antivascular endothelial growth factor receptor (fetal liver kinase 1) monoclonal antibody inhibits tumor angiogenesis and growth of several mouse and human tumors. Cancer Res. 59, 5209–5218 (1999).

    PubMed  CAS  Google Scholar 

  59. Weijzen, S. et al. The Notch ligand Jagged-1 is able to induce maturation of monocyte-derived human dendritic cells. J. Immunol. 169, 4273–4278 (2002).

    Article  CAS  Google Scholar 

  60. Tammela, T. et al. Angiopoietin-1 promotes lymphatic sprouting and hyperplasia. Blood 105, 4642–4648 (2005).

    Article  CAS  Google Scholar 

  61. Baluk, P. et al. Pathogenesis of persistent lymphatic vessel hyperplasia in chronic airway inflammation. J. Clin. Invest. 115, 247–257 (2005).

    Article  CAS  Google Scholar 

  62. Ruhrberg, C. et al. Spatially restricted patterning cues provided by heparin-binding VEGF-A control blood vessel branching morphogenesis. Genes Dev. 16, 2684–2698 (2002).

    Article  CAS  Google Scholar 

  63. Karpanen, T. et al. Lymphangiogenic growth factor responsiveness is modulated by postnatal lymphatic vessel maturation. Am. J. Pathol. 169, 708–718 (2006).

    Article  CAS  Google Scholar 

  64. Zheng, W. et al. Notch restricts lymphatic vessel sprouting induced by vascular endothelial growth factor. Blood 118, 1154–1162 (2011).

    Article  CAS  Google Scholar 

  65. Tvorogov, D. et al. Effective suppression of vascular network formation by combination of antibodies blocking VEGFR ligand binding and receptor dimerization. Cancer Cell 18, 630–640 (2010).

    Article  CAS  Google Scholar 

  66. Jussila, L. et al. Lymphatic endothelium and Kaposi’s sarcoma spindle cells detected by antibodies against the vascular endothelial growth factor receptor-3. Cancer Res. 58, 1599–1604 (1998).

    PubMed  CAS  Google Scholar 

  67. Ghalamkarpour, A. et al. Recessive primary congenital lymphoedema caused by a VEGFR3 mutation. J. Med. Genet. 46, 399–404 (2009).

    Article  CAS  Google Scholar 

  68. Persaud, K. et al. Involvement of the VEGF receptor 3 in tubular morphogenesis demonstrated with a human anti-human VEGFR-3 monoclonal antibody that antagonizes receptor activation by VEGF-C. J. Cell Sci. 117, 2745–2756 (2004).

    Article  CAS  Google Scholar 

  69. Karpanen, T. et al. Functional interaction of VEGF-C and VEGF-D with neuropilin receptors. FASEB J. 20, 1462–1472 (2006).

    Article  CAS  Google Scholar 

  70. Jakobsson, L. et al. Heparan sulfate in trans potentiates VEGFR-mediated angiogenesis. Dev. Cell 10, 625–634 (2006).

    Article  CAS  Google Scholar 

  71. Lobov, I. B. et al. Delta-like ligand 4 (Dll4) is induced by VEGF as a negative regulator of angiogenic sprouting. Proc. Natl Acad. Sci. USA 104, 3219–3224 (2007).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We would like to thank T. Petrova (CePO, CHUV and University of Lausanne, Switzerland) for the Foxc2+/− mice, M. Achen and S. Stacker (Peter MacCallum Cancer Centre, Melbourne, Australia) for the Vegfd−/− mice, B. Pytowski at Eli Lilly for VEGFR-2- and VEGFR-3-blocking antibodies, M. Jeltsch (Molecular/Cancer Biology Laboratory, University of Helsinki, Finland) for generating VEGF-C antibodies, S. Kaijalainen (Molecular/Cancer Biology Laboratory, University of Helsinki, Finland) for generating mDll4-Fc and mDll4–ECTM–eGFP expression vectors, A. Alitalo (Institute of Pharmaceutical Sciences, ETH Zurich, Switzerland) for valuable help with experiments and K. Helenius for critical comments on the manuscript. The Biomedicum Molecular Imaging Unit is acknowledged for microscopy services, and N. Ihalainen, T. Laakkonen, K. Salo and T. Tainola for excellent technical assistance, as well as personnel of the Meilahti Experimental Animal Center (University of Helsinki) for expert animal husbandry. We also thank I. Rosewell (London Research Institute, UK) for generation of chimaeric mice. This work was supported by grants from the Academy of Finland, the Association for International Cancer Research, the Finnish Cancer Organizations, the Helsinki University Research Fund, the Sigrid Juselius Foundation, the Louis-Jeantet Foundation and the European Research Council (ERC-2010-AdG-268804-TX-FACTORS). T.T. was supported by personal grants from the Emil Aaltonen Foundation, the K. Albin Johansson Foundation, the Finnish Medical Foundation, the Maud Kuistila Foundation, the Orion-Farmos Research Foundation and the Paulo Foundation. G.Z. was supported by personal grants from the K. Albin Johansson Foundation, the Finnish Medical Foundation, The Paulo Foundation, the Ida Montin Foundation and the Orion-Farmos Research Foundation. H.G. was supported by Cancer Research UK, the Lister Institute of Preventive Medicine, the European Molecular Biology Organization (EMBO) Young Investigator Programme and the Leducq Transatlantic Network ARTEMIS. L.J. was supported by an EMBO long-term postdoctoral fellowship. C.A.F. was supported by a Marie Curie FP7 postdoctoral fellowship.

Author information

Authors and Affiliations

Authors

Contributions

T.T. and G.Z. designed, directed and carried out experiments and data analysis, as well as interpreted results, and wrote the paper; H.N. designed and carried out cell culture and biochemistry experiments, and analysed data; L.J. carried out three-dimensional embryoid body sprouting experiments and analysed data; K.H. carried out cell culture, morphometry of retinal vessels and qRT-PCR, and analysed data; D.T. carried out biochemistry experiments and analysed data; W.Z. produced and validated Notch ligand and inhibitor proteins; C.A.F. carried out three-dimensional embryoid body sprouting experiments and analysed data; A.M. carried out retina experiments and analysed data; E.A. provided op/op retinas and carried out genotyping; N.M. generated FoxC2 antibodies; S.Y-H. generated adenoviral vectors; M.F. generated PdgfbCreERT2 mice; T.M. generated Vegfr3flox/floxmice; A.E. analysed retinas of Vegfr3+/LacZ mice; J.W.P. provided op/op retinas; H.G. directed experiments, interpreted results and helped write the paper; K.A. designed and directed experiments, interpreted results and wrote the paper.

Corresponding author

Correspondence to Kari Alitalo.

Ethics declarations

Competing interests

K.A. is the chairman of the Scientific Advisory Board of Circadian.

Supplementary information

Supplementary Information

Supplementary Information (PDF 2609 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Tammela, T., Zarkada, G., Nurmi, H. et al. VEGFR-3 controls tip to stalk conversion at vessel fusion sites by reinforcing Notch signalling. Nat Cell Biol 13, 1202–1213 (2011). https://doi.org/10.1038/ncb2331

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb2331

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing