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.

Regulation of angiogenesis by hypoxia: role of the HIF system

Abstract

The regulation of angiogenesis by hypoxia is an important component of homeostatic mechanisms that link vascular oxygen supply to metabolic demand. Molecular characterization of angiogenic pathways, identification of hypoxia-inducible factor (HIF) as a key transcriptional regulator of these molecules, and the definition of the HIF hydoxylases as a family of dioxygenases that regulate HIF in accordance with oxygen availability have provided new insights into this process. Here we review these findings, and the role of HIF in developmental, adaptive and neoplastic angiogenesis. We also discuss the implications of oncogenic activation of extensive, physiologically interconnected hypoxia pathways for the tumor phenotype.

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: Dual regulation of HIF-α subunits by prolyl and asparaginyl hydroxylation.
Figure 2: Pathways linking angiogenesis to oxygen availability through the regulation of HIF.
Figure 3: Multiple interfaces of hypoxia pathways with the angiogenic growth factor VEGF.
Figure 4
Figure 5: Schematic illustrating the coselection of pathways in cancer.

References

  1. Krogh, A. The number and distribution of capillaries in muscles with calculations of the oxygen pressure head necessary for supplying the tissue. J. Physiol. (London) 52, 409–415 (1919).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Hudlicka, O., Dodd, L., Renkin, E.M. & Gray, S.D. Early changes in fiber profile and capillary density in long-term stimulated muscles. Am. J. Physiol. 243, H528–H535 (1982).

    CAS  PubMed  Google Scholar 

  3. Jozsa, L., Balint, J., Reffy, A., Jarvinen, M. & Kvist, M. Capillary density of tenotomized skeletal muscles. II. Observations on human muscles after spontaneous rupture of tendon. Eur. J. Appl. Physiol. Occup. Physiol. 44, 183–188 (1980).

    CAS  PubMed  Google Scholar 

  4. Ashton, N., Ward, B. & Serpell, G. Effect of oxygen on developing retinal vessels with particular reference to the problem of retrolental fibroplasia. Br. J. Ophthalmol. 38, 397–432 (1954).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Thomlinson, R.H. & Gray, L.H. The histological structure of some human lung cancers and the possible implications for radio-therapy. Br. J. Cancer 9, 539–549 (1955).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Folkman, J., Merler, E., Abernathy, C. & Williams, G. Isolation of a tumor factor responsible for angiogenesis. J. Exp. Med. 133, 275–288 (1971).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Knighton, D.R., Silver, I.A. & Hunt, T.K. Regulation of wound-healing angiogenesis - effect of oxygen gradients and inspired oxygen concentration. Surgery 90, 262–270 (1981).

    CAS  PubMed  Google Scholar 

  8. Knighton, D.R. et al. Oxygen tension regulates the expression of angiogenesis factor by macrophages. Science 221, 1283–1285 (1983).

    CAS  PubMed  Google Scholar 

  9. Kourembanas, S., Hannan, R.L. & Faller, D.V. Oxygen tension regulates the expression of the platelet-derived growth factor-β chain gene in human endothelial cells. J. Clin. Invest. 86, 670–674 (1990).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Shweiki, D., Itin, A., Soffer, D. & Keshet, E. Vascular endothelial growth factor induced by hypoxia may mediate hypoxia-initiated angiogenesis. Nature 359, 843–845 (1992).

    Article  CAS  PubMed  Google Scholar 

  11. Plate, K.H., Breier, G., Weich, H.A. & Risau, W. Vascular endothelial growth factor is a potential tumour angiogenesis factor in human gliomas in vivo. Nature 359, 845–848 (1992).

    CAS  PubMed  Google Scholar 

  12. Adair, T.H., Gay, W.J. & Montani, J.-P. Growth regulation of the vascular system: evidence for a metabolic hypothesis. Am. J. Physiol. 259, R393–R404 (1990).

    CAS  PubMed  Google Scholar 

  13. Jelkmann, W. Erythropoietin: structure, control of production, and function. Physiol. Rev. 72, 449–489 (1992).

    CAS  PubMed  Google Scholar 

  14. Necas, E. & Thorling, E.B. Unresponsiveness of erythropoietin-producing cells to cyanide. Am. J. Physiol. 222, 1187–1190 (1972).

    CAS  PubMed  Google Scholar 

  15. Goldwasser, E., Jacobson, L.O., Fried, W. & Plazk, L.F. Studies on erythropoiesis V: the effect of cobalt on the production of erythropoietin. Blood 13, 55–60 (1958).

    CAS  PubMed  Google Scholar 

  16. Goldberg, M.A. & Schneider, T.J. Similarities between the oxygen-sensing mechanisms regulating the expression of vascular endothelial growth factor and erythropoietin. J. Biol. Chem. 269, 4355–4359 (1994).

    CAS  PubMed  Google Scholar 

  17. Gleadle, J.M., Ebert, B.L., Firth, J.D. & Ratcliffe, P.J. Regulation of angiogenic growth factor expression by hypoxia, transition metals, and chelating agents. Am. J. Physiol. 268, C1362–C1368 (1995).

    CAS  PubMed  Google Scholar 

  18. Liu, Y., Cox, S.R., Morita, T. & Kourembanas, S. Hypoxia regulates vascular endothelial growth factor gene expression in endothelial cells. Circ. Res. 77, 638–643 (1995).

    CAS  PubMed  Google Scholar 

  19. Forsythe, J.A. et al. Activation of vascular endothelial growth factor gene transcription by hypoxia-inducible factor 1. Mol. Cell. Biol. 16, 4604–4613 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Semenza, G.L. & Wang, G.L. A nuclear factor induced by hypoxia via de novo protein synthesis binds to the human erythropoietin gene enhancer at a site required for transcriptional activation. Mol. Cell. Biol. 12, 5447–5454 (1992).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Wang, G.L., Jiang, B.-H., Rue, E.A. & Semenza, G.L. Hypoxia-inducible factor 1 is a basic-helix-loop-helix-PAS heterodimer regulated by cellular O2 tension. Proc. Natl. Acad. Sci. USA 92, 5510–5514 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Maxwell, P.H., Pugh, C.W. & Ratcliffe, P.J. Inducible operation of the erythropoietin 3′ enhancer in multiple cell lines: evidence for a widespread oxygen sensing mechanism. Proc. Natl. Acad. Sci. USA 90, 2423–2427 (1993).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Semenza, G.L. HIF-1 and human disease: one highly involved factor. Genes Dev. 14, 1983–1991 (2000).

    CAS  PubMed  Google Scholar 

  24. Wenger, R.H. Cellular adaptation to hypoxia: O2-sensing protein hydroxylases, hypoxia-inducible transcription factors, and O2-regulated gene expression. FASEB J. 16, 1151–1162 (2002).

    CAS  PubMed  Google Scholar 

  25. Tian, H., McKnight, S.L. & Russell, D.W. Endothelial PAS domain protein 1 (EPAS1), a transcription factor selectively expressed in endothelial cells. Genes Dev. 11, 72–82 (1997).

    CAS  PubMed  Google Scholar 

  26. Wiesener, M.S. et al. Induction of endothelial PAS domain protein-1 by hypoxia: characterization and comparison with hypoxia-inducible factor-1α. Blood 92, 2260–2268 (1998).

    CAS  PubMed  Google Scholar 

  27. Makino, Y. et al. Inhibitory PAS domain protein is a negative regulator of hypoxia-inducible gene expression. Nature 414, 550–554 (2001).

    CAS  PubMed  Google Scholar 

  28. Ivan, M. et al. HIFα targeted for VHL-mediated destruction by proline hydroxylation: implications for O2 sensing. Science 292, 464–468 (2001).

    CAS  PubMed  Google Scholar 

  29. Jaakkola, P. et al. Targeting of HIF-α to the von Hippel-Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation. Science 292, 468–472 (2001).

    CAS  PubMed  Google Scholar 

  30. Yu, F., White, S.B., Zhao, Q. & Lee, F.S. HIF-1α binding to VHL is regulated by stimulus-sensitive proline hydroxylation. Proc. Natl. Acad. Sci. USA 98, 9630–9635 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Epstein, A.C.R. et al. C. elegans EGL-9 and mammalian homologues define a family of dioxygenases that regulate HIF by prolyl hydroxylation. Cell 107, 43–54 (2001).

    CAS  PubMed  Google Scholar 

  32. Bruick, R.K. & McKnight, S.L. A conserved family of prolyl-4-hydroxylases that modify HIF. Science 294, 1337–1340 (2001).

    CAS  PubMed  Google Scholar 

  33. Hewitson, K.S. et al. Hypoxia inducible factor (HIF) asparagine hydroxylase is identical to Factor Inhibiting HIF (FIH) and is related to the cupin structural family. J. Biol. Chem. 277, 26351–26355 (2002).

    CAS  PubMed  Google Scholar 

  34. Masson, N., Willam, C., Maxwell, P.H., Pugh, C.W. & Ratcliffe, P.J. Independent function of two destruction domains in hypoxia-inducible factor-α chains activated by prolyl hydroxylation. EMBO J. 20, 5197–5206 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Lando, D., Peet, D.J., Whelan, D.A., Gorman, J.J. & Whitelaw, M.L. Asparagine hydroxylation of the HIF transactivation domain: a hypoxic switch. Science 295, 858–861 (2002).

    CAS  PubMed  Google Scholar 

  36. Lando, D. et al. FIH-1 is an asparaginyl hydroxylase enzyme that regulates the transcriptional activity of hypoxia-inducible factor. Genes Dev. 16, 1466–1471 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Mahon, P.C., Hirota, K. & Semenza, G.L. FIH-1: a novel protein that interacts with HIF-1α and VHL to mediate repression of HIF-1 transcriptional activity. Genes Dev. 15, 2675–2686 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Schofield, C.J. & Zhang, Z. Structural and mechanistic studies on 2-oxoglutarate-dependent oxygenases and related enzymes. Curr. Opin. Struct. Biol. 9, 722–731 (1999).

    CAS  PubMed  Google Scholar 

  39. Webster, K.A., Discher, D.J. & Bishopric, N.H. Regulation of fos and jun immediate-early genes by redox or metabolic stress in cardiac myocytes. Circ. Res. 74, 679–686 (1994).

    CAS  PubMed  Google Scholar 

  40. Koong, A.C., Chen, E.Y. & Giaccia, A.J. Hypoxia causes the activation of nuclear factor κB through the phosphorylation of IκBα on tyrosine residues. Cancer Res. 54, 1425–1430 (1994).

    CAS  PubMed  Google Scholar 

  41. Graeber, T.G. et al. Hypoxia induces accumulation of p53 protein, but activation of a G1-phase checkpoint by low-oxygen conditions is independent of p53 status. Mol. Cell. Biol. 14, 6264–6277 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Alarcon, R., Koumenis, C., Geyer, R.K., Maki, C.G. & Giaccia, A.J. Hypoxia induces p53 accumulation through MDM2 down-regulation and inhibition of E6-mediated degradation. Cancer Res. 59, 6046–6051 (1999).

    CAS  PubMed  Google Scholar 

  43. Kuznetsova, A.V. et al. von Hippel-Lindau protein binds hyperphosphorylated large subunit of RNA polymerase II through a proline hydroxylation motif and targets it for ubiquitination. Proc. Natl. Acad. Sci. USA 100, 2706–2711 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Conway, E.M., Collen, D. & Carmeliet, P. Molecular mechanisms of blood vessel growth. Cardiovasc. Res. 49, 507–521 (2001).

    CAS  PubMed  Google Scholar 

  45. Melillo, G. et al. A hypoxia-responsive element mediates a novel pathway of activation of the inducible nitric oxide synthase promoter. J. Exp. Med. 182, 1683–1693 (1995).

    CAS  PubMed  Google Scholar 

  46. Enholm, B. et al. Comparison of VEGF, VEGF-B, VEGF-C and Ang-1 mRNA regulation by serum, growth factors, oncoproteins and hypoxia. Oncogene 14, 2475–2483 (1997).

    CAS  PubMed  Google Scholar 

  47. Currie, M.J. et al. Expression of the angiopoietins and their receptor Tie2 in human renal clear cell carcinomas; regulation by the von Hippel-Lindau gene and hypoxia. J. Pathol. 198, 502–510 (2002).

    CAS  PubMed  Google Scholar 

  48. Tuder, R.M., Flook, B.E. & Voelkel, N.F. Increased gene expression for VEGF and the VEGF receptors KDR/Flk and Flt in lungs exposed to acute or to chronic hypoxia. J. Clin. Invest. 95, 1798–1807 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Gerber, H.-P., Condorelli, F., Park, J. & Ferrara, N. Differential transcriptional regulation of the two vascular endothelial growth factor receptor genes. J. Biol. Chem. 272, 23659–23667 (1997).

    CAS  PubMed  Google Scholar 

  50. Waltenberger, J., Mayr, U., Pentz, S. & Hombach, V. Functional upregulation of the vascular endothelial growth factor receptor KDR by hypoxia. Circulation 94, 1647–1654 (1996).

    CAS  PubMed  Google Scholar 

  51. Oh, H. et al. Hypoxia and vascular endothelial growth factor selectively up-regulate angiopoietin-2 in bovine microvascular endothelial cells. J. Biol. Chem. 274, 15732–15739 (1999).

    CAS  PubMed  Google Scholar 

  52. Mandriota, S. et al. Hypoxia-inducible angiopoietin-2 expression is mimicked by iodonium compounds and occurs in the rat brain and skin in response to systemic hypoxia and tissue ischaemia. Am. J. Pathol. 156, 1–13 (2000).

    Google Scholar 

  53. Ben-Yosef, Y. et al. Regulation of endothelial matrix metalloproteinase-2 by hypoxia/reoxygenation. Circ. Res. 90, 784–791 (2002).

    CAS  PubMed  Google Scholar 

  54. Norman, J.T., Clark, I.M. & Garcia, P.L. Hypoxia promotes fibrogenesis in human renal fibroblasts. Kidney Int. 58, 2351–2366 (2000).

    CAS  PubMed  Google Scholar 

  55. Takahashi, Y., Takahashi, S., Shiga, Y., Yoshimi, T. & Miura, T. Hypoxic induction of prolyl 4-hydroxylase α(I) in cultured cells. J. Biol. Chem. 275, 14139–14146 (2000).

    CAS  PubMed  Google Scholar 

  56. Kietzmann, T., Roth, U. & Jungermann, K. Induction of the plasminogen activator inhibitor-1 gene expression by mild hypoxia via a hypoxia response element binding the hypoxia-inducible factor-1 in rat hepatocytes. Blood 94, 4177–4185 (1999).

    CAS  PubMed  Google Scholar 

  57. Graham, C.H., Fitzpatrick, T.E. & McCrae, K.R. Hypoxia stimulates urokinase receptor expression through a heme protein-dependent pathway. Blood 91, 3300–3307 (1998).

    CAS  PubMed  Google Scholar 

  58. Phelan, M.W., Forman, L.W., Perrine, S.P. & Faller, D.V. Hypoxia increases thrombospondin-1 transcript and protein in cultured endothelial cells. J. Lab. Clin. Med. 132, 519–529 (1998).

    CAS  PubMed  Google Scholar 

  59. Kuwubara, K. et al. Hypoxia-mediated induction of acidic/basic fibroblast growth factor and platelet-derived growth factor in mononuclear phagocytes stimulates growth of hypoxic endothelial cells. Proc. Natl. Acad. Sci. USA 92, 4606–4610 (1995).

    Google Scholar 

  60. Negus, R.P., Turner, L., Burke, F. & Balkwill, F.R. Hypoxia down-regulates MCP-1 expression: implications for macrophage distribution in tumors. J. Leukoc. Biol. 63, 758–765 (1998).

    CAS  PubMed  Google Scholar 

  61. Wykoff, C.C., Pugh, C.W., Maxwell, P.H., Harris, A.L. & Ratcliffe, P.J. Identification of novel hypoxia-dependent and independent target genes of the von Hippel-Lindau (VHL) tumor suppressor by mRNA differential expression profiling. Oncogene 19, 6297–6305 (2000).

    CAS  PubMed  Google Scholar 

  62. Sakuda, H., Nakashima, Y., Kuriyama, S. & Sueishi, K. Media conditioned by smooth muscle cells cultured in a variety of hypoxic environments stimulates in vitro angiogenesis. A relationship to transforming growth factor-β1. Am. J. Pathol. 141, 1507–1516 (1992).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Phillips, P.G., Birnby, L.M. & Narendran, A. Hypoxia induces capillary network formation in cultured bovine pulmonary microvessel endothelial cells. Am. J. Physiol. 268, L789–L800 (1995).

    CAS  PubMed  Google Scholar 

  64. Krishnamachary, B. et al. Regulation of colon carcinoma cell invasion by hypoxia-inducible factor 1. Cancer Res. 63, 1138–1143 (2003).

    CAS  PubMed  Google Scholar 

  65. Meininger, C.J., Schelling, M.E. & Granger, H.J. Adenosine and hypoxia stimulate proliferation and migration of endothelial cells. Am. J. Physiol. 255, H554–H562 (1988).

    CAS  PubMed  Google Scholar 

  66. Shreeniwas, R. et al. Macrovascular and microvascular endothelium during long-term hypoxia: alterations in cell growth, monolayer permeability, and cell surface coagulant properties. J. Cell. Physiol. 146, 8–17 (1991).

    CAS  PubMed  Google Scholar 

  67. Tucci, M. et al. Distinct effect of hypoxia on endothelial cell proliferation and cycling. Am. J. Physiol. 272, C1700–C1708 (1997).

    CAS  PubMed  Google Scholar 

  68. Iyer, N.V. et al. Cellular and developmental control of O2 homeostasis by hypoxia-inducible factor 1α. Genes Dev. 12, 149–162 (1997).

    Google Scholar 

  69. Carmeliet, P. et al. Role of HIF-1α in hypoxia-mediated apoptosis, cell proliferation and tumour angiogenesis. Nature 394, 485–490 (1998).

    CAS  PubMed  Google Scholar 

  70. Ryan, H.E., Lo, J. & Johnson, R.S. HIF-1α is required for solid tumor formation and embryonic vascularization. EMBO J. 17, 3005–3015 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Levy, N.S., Chung, S., Furneaux, H. & Levy, A.P. Hypoxic stabilization of vascular endothelial growth factor mRNA by the RNA-binding protein HuR. J. Biol. Chem. 273, 6417–6423 (1998).

    CAS  PubMed  Google Scholar 

  72. Stein, I. et al. Translation of vascular endothelial growth factor mRNA by internal ribosome entry: implications for translation under hypoxia. Mol. Cell. Biol. 18, 3112–3119 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Barleon, B. et al. Vascular endothelial growth factor up-regulates its receptor fms-like tyrosine kinase 1 (FLT-1) and a soluble variant of FLT-1 in human vascular endothelial cells. Cancer Res. 57, 5421–5425 (1997).

    CAS  PubMed  Google Scholar 

  74. Kotch, L.E., Iyer, N.V., Laughner, E. & Semenza, G.L. Defective vascularization of HIF-1α-null embryos is not associated with VEGF deficiency but with mesenchymal cell death. Dev. Biol. 209, 254–267 (1999).

    CAS  PubMed  Google Scholar 

  75. Maltepe, E., Schmidt, J.V., Baunoch, D., Bradfield, C.A. & Simon, M.C. Abnormal angiogenesis and responses to glucose and oxygen deprivation in mice lacking the protein ARNT. Nature 386, 403–407 (1997).

    CAS  PubMed  Google Scholar 

  76. Peng, J., Zhang, L., Drysdale, L. & Fong, G.H. The transcription factor EPAS-1/hypoxia-inducible factor 2α plays an important role in vascular remodeling. Proc. Natl. Acad. Sci. USA 97, 8386–8391 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Tian, H., Hammer, R.E., Matsumoto, A.M., Russell, D.W. & McKnight, S.L. The hypoxia responsive transcription factor EPAS1 is essential for catecholamine homeostasis and protection against heart failure during embryonic development. Genes Dev. 12, 3320–3324 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Compernolle, V. et al. Loss of HIF-2α and inhibition of VEGF impair fetal lung maturation, whereas treatment with VEGF prevents fatal respiratory distress in premature mice. Nat. Med. 8, 702–710 (2002).

    CAS  PubMed  Google Scholar 

  79. Lee, Y.M. et al. Determination of hypoxic region by hypoxia marker in developing mouse embryos in vivo: a possible signal for vessel development. Dev. Dyn. 220, 175–186 (2001).

    CAS  PubMed  Google Scholar 

  80. Morita, M. et al. HLF/HIF-2α is a key factor in retinopathy of prematurity in association with erythropoietin. EMBO J. 22, 1134–1146 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Grimm, C. et al. HIF-1-induced erythropoietin in the hypoxic retina protects against light-induced retinal degeneration. Nat. Med. 8, 718–724 (2002).

    CAS  PubMed  Google Scholar 

  82. Rosenberger, C. et al. Expression of hypoxia-inducible factor-1α and -2α in hypoxic and ischemic rat kidneys. J. Am. Soc. Nephrol. 13, 1721–1732 (2002).

    CAS  PubMed  Google Scholar 

  83. Lee, S.H. et al. Early expression of angiogenesis factors in acute myocardial ischemia and infarction. N. Engl. J. Med. 342, 626–633 (2000).

    CAS  PubMed  Google Scholar 

  84. Stroka, D.M. et al. HIF-1 is expressed in normoxic tissue and displays an organ-specific regulation under systemic hypoxia. FASEB J. 15, 2445–2453 (2001).

    CAS  PubMed  Google Scholar 

  85. Wiesener, M.S. et al. Widespread, hypoxia-inducible expression of HIF-2α in distinct cell populations of different organs. FASEB J. 17, 271–273 (2002).

    PubMed  Google Scholar 

  86. Rajakumar, A., Doty, K., Daftary, A., Harger, G. & Conrad, K.P. Impaired oxygen-dependent reduction of HIF-1α and -2α proteins in pre-eclamptic placentae. Placenta 24, 199–208 (2003).

    CAS  PubMed  Google Scholar 

  87. Hollander, A.P., Corke, K.P., Freemont, A.J. & Lewis, C.E. Expression of hypoxia-inducible factor 1alpha by macrophages in the rheumatoid synovium: implications for targeting of therapeutic genes to the inflamed joint. Arthritis Rheum. 44, 1540–1544 (2001).

    CAS  PubMed  Google Scholar 

  88. Ozaki, H. et al. Hypoxia inducible factor-1α is increased in ischemic retina: temporal and spatial correlation with VEGF expression. Invest. Ophthalmol. Vis. Sci. 40, 182–189 (1999).

    CAS  PubMed  Google Scholar 

  89. Elson, D.A., Ryan, H.E., Snow, J.W., Johnson, R. & Arbeit, J.M. Coordinate up-regulation of hypoxia inducible factor (HIF)-1α and HIF-1 target genes during multi-stage epidermal carcinogenesis and wound healing. Cancer Res. 60, 6189–6195 (2000).

    CAS  PubMed  Google Scholar 

  90. Elson, D.A. et al. Induction of hypervascularity without leakage or inflammation in transgenic mice overexpressing hypoxia-inducible factor-1α. Genes Dev. 15, 2520–2532 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Vincent, K.A. et al. Angiogenesis is induced in a rabbit model of hindlimb ischemia by naked DNA encoding an HIF-1α/VP16 hybrid transcription factor. Circulation 102, 2255–2261 (2000).

    CAS  PubMed  Google Scholar 

  92. Shyu, K.G. et al. Intramyocardial injection of naked DNA encoding HIF-1α/VP16 hybrid to enhance angiogenesis in an acute myocardial infarction model in the rat. Cardiovasc. Res. 54, 576–583 (2002).

    CAS  PubMed  Google Scholar 

  93. Li, J. et al. PR39, a peptide regulator of angiogenesis. Nat. Med. 6, 49–55 (2000).

    CAS  PubMed  Google Scholar 

  94. Willam, C. et al. Peptide blockade of HIFα degradation modulates cellular metabolism and angiogenesis. Proc. Natl. Acad. Sci. USA 99, 10423–10428 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Nwogu, N.I. et al. Inhibition of collagen synthesis with prolyl 4-hydroxylase inhibitor improves left ventricular function and alters the pattern of left ventricular dilatation after myocardial infarction. Circulation 104, 2216–2221 (2001).

    CAS  PubMed  Google Scholar 

  96. Ivan, M. et al. Biochemical purification and pharmacological inhibition of a mammalian prolyl hydroxylase acting on hypoxia-inducible factor. Proc. Natl. Acad. Sci. USA 99, 13459–13464 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Aravind, L. & Koonin, E.V. The DNA-repair protein AlkB, EGL-9, and leprecan define new families of 2-oxoglutarate- and iron-dependent dioxygenases. Genome Biol. 2, 0007.1–0007.8 (2001).

    Google Scholar 

  98. Elkins, J.M. et al. Structure of factor-inhibiting hypoxia-inducible factor (HIF) reveals mechanism of oxidative modification of HIF-1α. J. Biol. Chem. 278, 1802–1806 (2003).

    CAS  PubMed  Google Scholar 

  99. Semenza, G.L. Hypoxia, clonal selection, and the role of HIF-1 in tumor progression. Crit. Rev. Biochem. Mol. Biol. 35, 71–103 (2000).

    CAS  PubMed  Google Scholar 

  100. Maxwell, P.H., Pugh, C.W. & Ratcliffe, P.J. Activation of the HIF pathway in cancer. Curr. Opin. Genet. Dev. 11, 293–299 (2001).

    CAS  PubMed  Google Scholar 

  101. Richard, D.E., Berra, E., Gothie, E., Roux, D. & Pouysségur, J. p42/p44 mitogen-activated protein kinases phosphorylate hypoxia-inducible factor 1α (HIF-1α) and enhance the transcriptional activity of HIF-1. J. Biol. Chem. 274, 32631–32637 (1999).

    CAS  PubMed  Google Scholar 

  102. Zundel, W. et al. Loss of PTEN facilitates HIF-1-mediated gene expression. Genes Dev. 14, 391–396 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Chan, D.A., Sutphin, P.D., Denko, N.C. & Giaccia, A.J. Role of prolyl hydroxylation in oncogenically stabilized hypoxia-inducible factor-1α. J. Biol. Chem. 277, 40112–40117 (2002).

    CAS  PubMed  Google Scholar 

  104. Knowles, H.J., Raval, R.R., Harris, A.L. & Ratcliffe, P.J. Effect of ascorbate on the activity of hypoxia inducible factor (HIF) in cancer cells. Cancer Res. 63, 1764–1768 (2003).

    CAS  PubMed  Google Scholar 

  105. Le, N.T.V. & Richardson, D.R. The role of iron in cell cycle progression and the proliferation of neoplastic cells. Biochim. Biophys. Acta 1603, 31–46 (2002).

    CAS  PubMed  Google Scholar 

  106. Maxwell, P.H. et al. Hypoxia inducible factor-1 modulates gene expression in solid tumors and influences both angiogenesis and tumor growth. Proc. Natl. Acad. Sci. USA 94, 8104–8109 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Kung, A.L., Wang, S., Klco, J.M., Kaelin, W.G. & Livingston, D.M. Suppression of tumor growth through disruption of hypoxia-inducible transcription. Nat. Med. 6, 1335–1340 (2000).

    CAS  PubMed  Google Scholar 

  108. Hopfl, G. et al. Rescue of hypoxia-inducible factor-1α-deficient tumor growth by wild-type cells is independent of vascular endothelial growth factor. Cancer Res. 62, 2962–2970 (2002).

    CAS  PubMed  Google Scholar 

  109. Ryan, H.E. et al. Hypoxia-inducible factor-lα is a positive factor in solid tumor growth. Cancer Res. 60, 4010–4015 (2000).

    CAS  PubMed  Google Scholar 

  110. Kaelin, W.G. Molecular basis of the VHL hereditary cancer syndrome. Nat. Rev. Cancer 2, 673–682 (2002).

    CAS  PubMed  Google Scholar 

  111. Maxwell, P.H. et al. The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature 399, 271–275 (1999).

    CAS  PubMed  Google Scholar 

  112. Iliopoulos, O., Kibel, A., Gray, S. & Kaelin, W.G., Jr. Tumour supression by the human von Hippel-Lindau gene product. Nat. Med. 1, 822–826 (1995).

    CAS  PubMed  Google Scholar 

  113. Kondo, K., Kico, J., Nakamura, E., Lechpammer, M. & Kaelin, W.G.J. Inhibition of HIF is necessary for tumor suppression by the von Hippel-Lindau protein. Cancer Cell 1, 237–246 (2002).

    CAS  PubMed  Google Scholar 

  114. Maranchie, J.K. et al. The contribution of VHL substrate binding and HIF1-α to the phenotype of VHL loss in renal cell carcinoma. Cancer Cell 1, 247–255 (2002).

    CAS  PubMed  Google Scholar 

  115. Vaux, E.C. et al. Selection of mutant CHO cells with constitutive activation of the HIF system and inactivation of the von Hippel-Lindau tumor suppressor. J. Biol. Chem. 276, 44323–44330 (2001).

    CAS  PubMed  Google Scholar 

  116. Mack, F.A. et al. Loss of pVHL is sufficient to cause HIF dysregulation in primary cells but does not promote tumor growth. Cancer Cell 3, 75–88 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Knudson, A.G. Chasing the cancer demon. Annu. Rev. Genet. 34, 1–19 (2000).

    CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Peter J Ratcliffe.

Ethics declarations

Competing interests

C.W.P. and P.J.R. are scientific cofounders of ReOx, Ltd.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Pugh, C., Ratcliffe, P. Regulation of angiogenesis by hypoxia: role of the HIF system. Nat Med 9, 677–684 (2003). https://doi.org/10.1038/nm0603-677

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm0603-677

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