Skip to main content

Molecular Mechanisms of Hypoxia-Regulated Angiogenesis

  • Chapter
  • First Online:
Angiogenesis and Vascularisation

Abstract

Hypoxia is an important regulator of angiogenesis and it stimulates neovascularization from existing blood vessels. At the molecular level, it occurs mostly through transcriptional regulation of genes which contain a core consensus sequence called hypoxia response element (HRE) via hypoxia inducible factors (HIFs) action. The discovery of HIFs hydroxylases as a family of dioxygenases that regulate HIFs dependently on the oxygen availability have significantly improved our understanding of the mechanisms of hypoxia signaling. Moreover, a broad number of factors have been shown to influence HIF stability and their effects could be mediated via several possible mechanisms including nitrosylation, deacetylation, or oxidation. The induction of HIF leads to the complex regulation of the pro- and antiangiogenic factors and extensive research will be essential for thorough understanding of the role of hypoxia in disease development and will help to identify new therapeutic targets for treatment of such hypoxia-dependent disorders.

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

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 129.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 169.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 169.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Semenza GL, Nejfelt MK, Chi SM, Antonarakis SE (1991) Hypoxia-inducible nuclear factors bind to an enhancer element located 3′ to the human erythropoietin gene. Proc Natl Acad Sci USA 88(13):5680–5684

    CAS  PubMed  Google Scholar 

  2. Pugh CW, Tan CC, Jones RW, Ratcliffe PJ (1991) Functional analysis of an oxygen-regulated transcriptional enhancer lying 3′ to the mouse erythropoietin gene. Proc Natl Acad Sci USA 88(23):10553–10557

    CAS  PubMed  Google Scholar 

  3. Semenza GL, Jiang BH, Leung SW, Passantino R, Concordet JP, Maire P, Giallongo A (1996) Hypoxia response elements in the aldolase A, enolase 1, and lactate dehydrogenase A gene promoters contain essential binding sites for hypoxia-inducible factor 1. J Biol Chem 271(51):32529–32537

    CAS  PubMed  Google Scholar 

  4. Kimura H, Weisz A, Ogura T, Hitomi Y, Kurashima Y, Hashimoto K, D’Acquisto F, Makuuchi M, Esumi H (2001) Identification of hypoxia-inducible factor 1 ancillary sequence and its function in vascular endothelial growth factor gene induction by hypoxia and nitric oxide. J Biol Chem 276(3):2292–2298

    CAS  PubMed  Google Scholar 

  5. Wang GL, Jiang BH, Rue EA, Semenza GL (1995) Hypoxia-inducible factor 1 is a basic-helix-loop-helix-PAS heterodimer regulated by cellular O2 tension. Proc Natl Acad Sci USA 92(12):5510–5514

    CAS  PubMed  Google Scholar 

  6. Makino Y, Kanopka A, Wilson WJ, Tanaka H, Poellinger L (2002) Inhibitory PAS domain protein (IPAS) is a hypoxia-inducible splicing variant of the hypoxia-inducible factor-3alpha locus. J Biol Chem 277(36):32405–32408

    CAS  PubMed  Google Scholar 

  7. Maynard MA, Qi H, Chung J, Lee EH, Kondo Y, Hara S, Conaway RC, Conaway JW, Ohh M (2003) Multiple splice variants of the human HIF-3 alpha locus are targets of the von Hippel-Lindau E3 ubiquitin ligase complex. J Biol Chem 278(13):11032–11040

    CAS  PubMed  Google Scholar 

  8. Makino Y, Cao R, Svensson K, Bertilsson G, Asman M, Tanaka H, Cao Y, Berkenstam A, Poellinger L (2001) Inhibitory PAS domain protein is a negative regulator of hypoxia-inducible gene expression. Nature 414(6863):550–554

    CAS  PubMed  Google Scholar 

  9. Jaakkola P, Mole DR, Tian YM, Wilson MI, Gielbert J, Gaskell SJ, von Kriegsheim A, Hebestreit HF, Mukherji M, Schofield CJ, Maxwell PH, Pugh CW, Ratcliffe PJ (2001) Targeting of HIF-alpha to the von Hippel-Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation. Science 292(5516):468–472

    CAS  PubMed  Google Scholar 

  10. Ivan M, Kondo K, Yang H, Kim W, Valiando J, Ohh M, Salic A, Asara JM, Lane WS, Kaelin WG Jr (2001) HIFalpha targeted for VHL-mediated destruction by proline hydroxylation: implications for O2 sensing. Science 292(5516):464–468

    CAS  PubMed  Google Scholar 

  11. Epstein AC, Gleadle JM, McNeill LA, Hewitson KS, O’Rourke J, Mole DR, Mukherji M, Metzen E, Wilson MI, Dhanda A, Tian YM, Masson N, Hamilton DL, Jaakkola P, Barstead R, Hodgkin J, Maxwell PH, Pugh CW, Schofield CJ, Ratcliffe PJ (2001) C. elegans EGL-9 and mammalian homologs define a family of dioxygenases that regulate HIF by prolyl hydroxylation. Cell 107(1):43–54

    CAS  PubMed  Google Scholar 

  12. Bruick RK, McKnight SL (2001) A conserved family of prolyl-4-hydroxylases that modify HIF. Science 294(5545):1337–1340

    CAS  PubMed  Google Scholar 

  13. Berra E, Benizri E, Ginouves A, Volmat V, Roux D, Pouyssegur J (2003) HIF prolyl-hydroxylase 2 is the key oxygen sensor setting low steady-state levels of HIF-1alpha in normoxia. EMBO J 22(16):4082–4090

    CAS  PubMed  Google Scholar 

  14. Appelhoff RJ, Tian YM, Raval RR, Turley H, Harris AL, Pugh CW, Ratcliffe PJ, Gleadle JM (2004) Differential function of the prolyl hydroxylases PHD1, PHD2, and PHD3 in the regulation of hypoxia-inducible factor. J Biol Chem 279(37):38458–38465

    CAS  PubMed  Google Scholar 

  15. Maxwell PH, Wiesener MS, Chang GW, Clifford SC, Vaux EC, Cockman ME, Wykoff CC, Pugh CW, Maher ER, Ratcliffe PJ (1999) The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature 399(6733):271–275

    CAS  PubMed  Google Scholar 

  16. Ohh M, Park CW, Ivan M, Hoffman MA, Kim TY, Huang LE, Pavletich N, Chau V, Kaelin WG (2000) Ubiquitination of hypoxia-inducible factor requires direct binding to the beta-domain of the von Hippel-Lindau protein. Nat Cell Biol 2(7):423–427

    CAS  PubMed  Google Scholar 

  17. Cockman ME, Masson N, Mole DR, Jaakkola P, Chang GW, Clifford SC, Maher ER, Pugh CW, Ratcliffe PJ, Maxwell PH (2000) Hypoxia inducible factor-alpha binding and ubiquitylation by the von Hippel-Lindau tumor suppressor protein. J Biol Chem 275(33):25733–25741

    CAS  PubMed  Google Scholar 

  18. Chilov D, Camenisch G, Kvietikova I, Ziegler U, Gassmann M, Wenger RH (1999) Induction and nuclear translocation of hypoxia-inducible factor-1 (HIF-1): heterodimerization with ARNT is not necessary for nuclear accumulation of HIF-1alpha. J Cell Sci 112(Pt 8):1203–1212

    CAS  PubMed  Google Scholar 

  19. Lando D, Peet DJ, Gorman JJ, Whelan DA, Whitelaw ML, Bruick RK (2002) FIH-1 is an asparaginyl hydroxylase enzyme that regulates the transcriptional activity of hypoxia-inducible factor. Genes Dev 16(12):1466–1471

    CAS  PubMed  Google Scholar 

  20. Brunelle JK, Bell EL, Quesada NM, Vercauteren K, Tiranti V, Zeviani M, Scarpulla RC, Chandel NS (2005) Oxygen sensing requires mitochondrial ROS but not oxidative phosphorylation. Cell Metab 1(6):409–414

    CAS  PubMed  Google Scholar 

  21. Guzy RD, Hoyos B, Robin E, Chen H, Liu L, Mansfield KD, Simon MC, Hammerling U, Schumacker PT (2005) Mitochondrial complex III is required for hypoxia-induced ROS production and cellular oxygen sensing. Cell Metab 1(6):401–408

    CAS  PubMed  Google Scholar 

  22. Mansfield KD, Guzy RD, Pan Y, Young RM, Cash TP, Schumacker PT, Simon MC (2005) Mitochondrial dysfunction resulting from loss of cytochrome c impairs cellular oxygen sensing and hypoxic HIF-alpha activation. Cell Metab 1(6):393–399

    CAS  PubMed Central  PubMed  Google Scholar 

  23. Masson N, Singleton RS, Sekirnik R, Trudgian DC, Ambrose LJ, Miranda MX, Tian YM, Kessler BM, Schofield CJ, Ratcliffe PJ (2012) The FIH hydroxylase is a cellular peroxide sensor that modulates HIF transcriptional activity. EMBO Rep 13(3):251–257

    CAS  PubMed Central  PubMed  Google Scholar 

  24. Finkel T, Deng CX, Mostoslavsky R (2009) Recent progress in the biology and physiology of sirtuins. Nature 460(7255):587–591

    CAS  PubMed Central  PubMed  Google Scholar 

  25. Finley LW, Carracedo A, Lee J, Souza A, Egia A, Zhang J, Teruya-Feldstein J, Moreira PI, Cardoso SM, Clish CB, Pandolfi PP, Haigis MC (2011) SIRT3 opposes reprogramming of cancer cell metabolism through HIF1alpha destabilization. Cancer Cell 19(3):416–428

    CAS  PubMed Central  PubMed  Google Scholar 

  26. Bell EL, Emerling BM, Ricoult SJ, Guarente L (2011) SirT3 suppresses hypoxia inducible factor 1alpha and tumor growth by inhibiting mitochondrial ROS production. Oncogene 30(26):2986–2996

    CAS  PubMed Central  PubMed  Google Scholar 

  27. Yu W, Dittenhafer-Reed KE, Denu JM (2012) SIRT3 protein deacetylates isocitrate dehydrogenase 2 (IDH2) and regulates mitochondrial redox status. J Biol Chem 287(17):14078–14086

    CAS  PubMed  Google Scholar 

  28. Tao R, Coleman MC, Pennington JD, Ozden O, Park SH, Jiang H, Kim HS, Flynn CR, Hill S, Hayes McDonald W, Olivier AK, Spitz DR, Gius D (2010) Sirt3-mediated deacetylation of evolutionarily conserved lysine 122 regulates MnSOD activity in response to stress. Mol Cell 40(6):893–904

    CAS  PubMed Central  PubMed  Google Scholar 

  29. Lim JH, Lee YM, Chun YS, Chen J, Kim JE, Park JW (2010) Sirtuin 1 modulates cellular responses to hypoxia by deacetylating hypoxia-inducible factor 1alpha. Mol Cell 38(6):864–878

    CAS  PubMed  Google Scholar 

  30. Dioum EM, Chen R, Alexander MS, Zhang Q, Hogg RT, Gerard RD, Garcia JA (2009) Regulation of hypoxia-inducible factor 2alpha signaling by the stress-responsive deacetylase sirtuin 1. Science 324(5932):1289–1293

    CAS  PubMed  Google Scholar 

  31. Rane S, He M, Sayed D, Vashistha H, Malhotra A, Sadoshima J, Vatner DE, Vatner SF, Abdellatif M (2009) Downregulation of miR-199a derepresses hypoxia-inducible factor-1alpha and Sirtuin 1 and recapitulates hypoxia preconditioning in cardiac myocytes. Circ Res 104(7):879–886

    CAS  PubMed Central  PubMed  Google Scholar 

  32. Chen R, Dioum EM, Hogg RT, Gerard RD, Garcia JA (2011) Hypoxia increases sirtuin 1 expression in a hypoxia-inducible factor-dependent manner. J Biol Chem 286(16):13869–13878

    CAS  PubMed  Google Scholar 

  33. Zhong L, D’Urso A, Toiber D, Sebastian C, Henry RE, Vadysirisack DD, Guimaraes A, Marinelli B, Wikstrom JD, Nir T, Clish CB, Vaitheesvaran B, Iliopoulos O, Kurland I, Dor Y, Weissleder R, Shirihai OS, Ellisen LW, Espinosa JM, Mostoslavsky R (2010) The histone deacetylase Sirt6 regulates glucose homeostasis via Hif1alpha. Cell 140(2):280–293

    CAS  PubMed Central  PubMed  Google Scholar 

  34. Potente M, Ghaeni L, Baldessari D, Mostoslavsky R, Rossig L, Dequiedt F, Haendeler J, Mione M, Dejana E, Alt FW, Zeiher AM, Dimmeler S (2007) SIRT1 controls endothelial angiogenic functions during vascular growth. Genes Dev 21(20):2644–2658

    CAS  PubMed  Google Scholar 

  35. Geng H, Harvey CT, Pittsenbarger J, Liu Q, Beer TM, Xue C, Qian DZ (2011) HDAC4 protein regulates HIF1alpha protein lysine acetylation and cancer cell response to hypoxia. J Biol Chem 286(44):38095–38102

    CAS  PubMed  Google Scholar 

  36. Chen S, Sang N (2011) Histone deacetylase inhibitors: the epigenetic therapeutics that repress hypoxia-inducible factors. J Biomed Biotechnol 2011:197946

    PubMed Central  PubMed  Google Scholar 

  37. Karhausen J, Haase VH, Colgan SP (2005) Inflammatory hypoxia: role of hypoxia-inducible factor. Cell Cycle 4(2):256–258

    CAS  PubMed  Google Scholar 

  38. Frede S, Stockmann C, Freitag P, Fandrey J (2006) Bacterial lipopolysaccharide induces HIF-1 activation in human monocytes via p44/42 MAPK and NF-kappaB. Biochem J 396(3):517–527

    CAS  PubMed  Google Scholar 

  39. Jung YJ, Isaacs JS, Lee S, Trepel J, Neckers L (2003) IL-1beta-mediated up-regulation of HIF-1alpha via an NFkappaB/COX-2 pathway identifies HIF-1 as a critical link between inflammation and oncogenesis. FASEB J 17(14):2115–2117

    CAS  PubMed  Google Scholar 

  40. Frede S, Freitag P, Otto T, Heilmaier C, Fandrey J (2005) The proinflammatory cytokine interleukin 1beta and hypoxia cooperatively induce the expression of adrenomedullin in ovarian carcinoma cells through hypoxia inducible factor 1 activation. Cancer Res 65(11):4690–4697

    CAS  PubMed  Google Scholar 

  41. Jung Y, Isaacs JS, Lee S, Trepel J, Liu ZG, Neckers L (2003) Hypoxia-inducible factor induction by tumour necrosis factor in normoxic cells requires receptor-interacting protein-dependent nuclear factor kappa B activation. Biochem J 370(Pt 3):1011–1017

    CAS  PubMed  Google Scholar 

  42. Balkwill F, Mantovani A (2001) Inflammation and cancer: back to Virchow? Lancet 357(9255):539–545

    CAS  PubMed  Google Scholar 

  43. Hanahan D, Weinberg RA (2011) Hallmarks of cancer: the next generation. Cell 144(5):646–674

    CAS  PubMed  Google Scholar 

  44. Hollander AP, Corke KP, Freemont AJ, Lewis CE (2001) 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(7):1540–1544

    CAS  PubMed  Google Scholar 

  45. Huerta-Yepez S, Baay-Guzman GJ, Bebenek IG, Hernandez-Pando R, Vega MI, Chi L, Riedl M, Diaz-Sanchez D, Kleerup E, Tashkin DP, Gonzalez FJ, Bonavida B, Zeidler M, Hankinson O (2011) Hypoxia inducible factor promotes murine allergic airway inflammation and is increased in asthma and rhinitis. Allergy 66(7):909–918

    CAS  PubMed Central  PubMed  Google Scholar 

  46. Parathath S, Mick SL, Feig JE, Joaquin V, Grauer L, Habiel DM, Gassmann M, Gardner LB, Fisher EA (2011) Hypoxia is present in murine atherosclerotic plaques and has multiple adverse effects on macrophage lipid metabolism. Circ Res 109(10):1141–1152

    CAS  PubMed Central  PubMed  Google Scholar 

  47. Belaiba RS, Bonello S, Zahringer C, Schmidt S, Hess J, Kietzmann T, Gorlach A (2007) Hypoxia up-regulates hypoxia-inducible factor-1alpha transcription by involving phosphatidylinositol 3-kinase and nuclear factor kappaB in pulmonary artery smooth muscle cells. Mol Biol Cell 18(12):4691–4697

    CAS  PubMed Central  PubMed  Google Scholar 

  48. van Uden P, Kenneth NS, Rocha S (2008) Regulation of hypoxia-inducible factor-1alpha by NF-kappaB. Biochem J 412(3):477–484

    PubMed Central  PubMed  Google Scholar 

  49. van Uden P, Kenneth NS, Webster R, Muller HA, Mudie S, Rocha S (2011) Evolutionary conserved regulation of HIF-1beta by NF-kappaB. PLoS Genet 7(1):e1001285

    PubMed Central  PubMed  Google Scholar 

  50. Tafani M, Pucci B, Russo A, Schito L, Pellegrini L, Perrone GA, Villanova L, Salvatori L, Ravenna L, Petrangeli E, Russo MA (2013) Modulators of HIF1alpha and NFkB in cancer treatment: is it a rational approach for controlling malignant progression? Front Pharmacol 4:13

    PubMed Central  PubMed  Google Scholar 

  51. Sandau KB, Faus HG, Brune B (2000) Induction of hypoxia-inducible-factor 1 by nitric oxide is mediated via the PI 3K pathway. Biochem Biophys Res Commun 278(1):263–267

    CAS  PubMed  Google Scholar 

  52. Kimura H, Weisz A, Kurashima Y, Hashimoto K, Ogura T, D’Acquisto F, Addeo R, Makuuchi M, Esumi H (2000) Hypoxia response element of the human vascular endothelial growth factor gene mediates transcriptional regulation by nitric oxide: control of hypoxia-inducible factor-1 activity by nitric oxide. Blood 95(1):189–197

    CAS  PubMed  Google Scholar 

  53. Sogawa K, Numayama-Tsuruta K, Ema M, Abe M, Abe H, Fujii-Kuriyama Y (1998) Inhibition of hypoxia-inducible factor 1 activity by nitric oxide donors in hypoxia. Proc Natl Acad Sci USA 95(13):7368–7373

    CAS  PubMed  Google Scholar 

  54. Yin JH, Yang DI, Ku G, Hsu CY (2000) iNOS expression inhibits hypoxia-inducible factor-1 activity. Biochem Biophys Res Commun 279(1):30–34

    CAS  PubMed  Google Scholar 

  55. Mateo J, Garcia-Lecea M, Cadenas S, Hernandez C, Moncada S (2003) Regulation of hypoxia-inducible factor-1alpha by nitric oxide through mitochondria-dependent and -independent pathways. Biochem J 376(Pt 2):537–544

    CAS  PubMed  Google Scholar 

  56. Kimura H, Ogura T, Kurashima Y, Weisz A, Esumi H (2002) Effects of nitric oxide donors on vascular endothelial growth factor gene induction. Biochem Biophys Res Commun 296(4):976–982

    CAS  PubMed  Google Scholar 

  57. Wang F, Zhang R, Xia T, Hsu E, Cai Y, Gu Z, Hankinson O (2007) Inhibitory effects of nitric oxide on invasion of human cancer cells. Cancer Lett 257(2):274–282

    CAS  PubMed Central  PubMed  Google Scholar 

  58. Quintero M, Brennan PA, Thomas GJ, Moncada S (2006) Nitric oxide is a factor in the stabilization of hypoxia-inducible factor-1alpha in cancer: role of free radical formation. Cancer Res 66(2):770–774

    CAS  PubMed  Google Scholar 

  59. Nanni S, Benvenuti V, Grasselli A, Priolo C, Aiello A, Mattiussi S, Colussi C, Lirangi V, Illi B, D’Eletto M, Cianciulli AM, Gallucci M, De Carli P, Sentinelli S, Mottolese M, Carlini P, Strigari L, Finn S, Mueller E, Arcangeli G, Gaetano C, Capogrossi MC, Donnorso RP, Bacchetti S, Sacchi A, Pontecorvi A, Loda M, Farsetti A (2009) Endothelial NOS, estrogen receptor beta, and HIFs cooperate in the activation of a prognostic transcriptional pattern in aggressive human prostate cancer. J Clin Invest 119(5):1093–1108

    CAS  PubMed Central  PubMed  Google Scholar 

  60. Dulak J, Jozkowicz A, Dembinska-Kiec A, Guevara I, Zdzienicka A, Zmudzinska-Grochot D, Florek I, Wojtowicz A, Szuba A, Cooke JP (2000) Nitric oxide induces the synthesis of vascular endothelial growth factor by rat vascular smooth muscle cells. Arterioscler Thromb Vasc Biol 20(3):659–666

    CAS  PubMed  Google Scholar 

  61. Jozkowicz A, Cooke JP, Guevara I, Huk I, Funovics P, Pachinger O, Weidinger F, Dulak J (2001) Genetic augmentation of nitric oxide synthase increases the vascular generation of VEGF. Cardiovasc Res 51(4):773–783

    CAS  PubMed  Google Scholar 

  62. Namba T, Koike H, Murakami K, Aoki M, Makino H, Hashiya N, Ogihara T, Kaneda Y, Kohno M, Morishita R (2003) Angiogenesis induced by endothelial nitric oxide synthase gene through vascular endothelial growth factor expression in a rat hindlimb ischemia model. Circulation 108(18):2250–2257

    CAS  PubMed  Google Scholar 

  63. Ghiso N, Rohan RM, Amano S, Garland R, Adamis AP (1999) Suppression of hypoxia-associated vascular endothelial growth factor gene expression by nitric oxide via cGMP. Invest Ophthalmol Vis Sci 40(6):1033–1039

    CAS  PubMed  Google Scholar 

  64. Dulak J, Jozkowicz A (2003) Regulation of vascular endothelial growth factor synthesis by nitric oxide: facts and controversies. Antioxid Redox Signal 5(1):123–132

    CAS  PubMed  Google Scholar 

  65. Metzen E, Zhou J, Jelkmann W, Fandrey J, Brune B (2003) Nitric oxide impairs normoxic degradation of HIF-1alpha by inhibition of prolyl hydroxylases. Mol Biol Cell 14(8):3470–3481

    CAS  PubMed Central  PubMed  Google Scholar 

  66. Park YK, Ahn DR, Oh M, Lee T, Yang EG, Son M, Park H (2008) Nitric oxide donor, (+/−)-S-nitroso-N-acetylpenicillamine, stabilizes transactive hypoxia-inducible factor-1alpha by inhibiting von Hippel-Lindau recruitment and asparagine hydroxylation. Mol Pharmacol 74(1):236–245

    CAS  PubMed  Google Scholar 

  67. Ball KA, Nelson AW, Foster DG, Poyton RO (2012) Nitric oxide produced by cytochrome c oxidase helps stabilize HIF-1alpha in hypoxic mammalian cells. Biochem Biophys Res Commun 420(4):727–732

    CAS  PubMed Central  PubMed  Google Scholar 

  68. Castello PR, David PS, McClure T, Crook Z, Poyton RO (2006) Mitochondrial cytochrome oxidase produces nitric oxide under hypoxic conditions: implications for oxygen sensing and hypoxic signaling in eukaryotes. Cell Metab 3(4):277–287

    CAS  PubMed  Google Scholar 

  69. Lee RC, Feinbaum RL, Ambros V (1993) The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 75(5):843–854

    CAS  PubMed  Google Scholar 

  70. Chan SY, Loscalzo J (2010) MicroRNA-210: a unique and pleiotropic hypoxamir. Cell Cycle 9(6):1072–1083

    CAS  PubMed Central  PubMed  Google Scholar 

  71. Lei Z, Li B, Yang Z, Fang H, Zhang GM, Feng ZH, Huang B (2009) Regulation of HIF-1alpha and VEGF by miR-20b tunes tumor cells to adapt to the alteration of oxygen concentration. PLoS One 4(10):e7629

    PubMed Central  PubMed  Google Scholar 

  72. Taguchi A, Yanagisawa K, Tanaka M, Cao K, Matsuyama Y, Goto H, Takahashi T (2008) Identification of hypoxia-inducible factor-1 alpha as a novel target for miR-17-92 microRNA cluster. Cancer Res 68(14):5540–5545

    CAS  PubMed  Google Scholar 

  73. Zhang Z, Sun H, Dai H, Walsh RM, Imakura M, Schelter J, Burchard J, Dai X, Chang AN, Diaz RL, Marszalek JR, Bartz SR, Carleton M, Cleary MA, Linsley PS, Grandori C (2009) MicroRNA miR-210 modulates cellular response to hypoxia through the MYC antagonist MNT. Cell Cycle 8(17):2756–2768

    CAS  PubMed  Google Scholar 

  74. Liu F, Lou YL, Wu J, Ruan QF, Xie A, Guo F, Cui SP, Deng ZF, Wang Y (2012) Upregulation of microRNA-210 regulates renal angiogenesis mediated by activation of VEGF signaling pathway under ischemia/perfusion injury in vivo and in vitro. Kidney Blood Press Res 35(3):182–191

    PubMed  Google Scholar 

  75. Fasanaro P, D’Alessandra Y, Di Stefano V, Melchionna R, Romani S, Pompilio G, Capogrossi MC, Martelli F (2008) MicroRNA-210 modulates endothelial cell response to hypoxia and inhibits the receptor tyrosine kinase ligand Ephrin-A3. J Biol Chem 283(23):15878–15883

    CAS  PubMed  Google Scholar 

  76. Lou YL, Guo F, Liu F, Gao FL, Zhang PQ, Niu X, Guo SC, Yin JH, Wang Y, Deng ZF (2012) miR-210 activates notch signaling pathway in angiogenesis induced by cerebral ischemia. Mol Cell Biochem 370(1–2):45–51

    CAS  PubMed  Google Scholar 

  77. Levy AP, Levy NS, Wegner S, Goldberg MA (1995) Transcriptional regulation of the rat vascular endothelial growth factor gene by hypoxia. J Biol Chem 270(22):13333–13340

    CAS  PubMed  Google Scholar 

  78. Shima DT, Deutsch U, D’Amore PA (1995) Hypoxic induction of vascular endothelial growth factor (VEGF) in human epithelial cells is mediated by increases in mRNA stability. FEBS Lett 370(3):203–208

    CAS  PubMed  Google Scholar 

  79. Levy AP, Levy NS, Goldberg MA (1996) Post-transcriptional regulation of vascular endothelial growth factor by hypoxia. J Biol Chem 271(5):2746–2753

    CAS  PubMed  Google Scholar 

  80. Ray PS, Jia J, Yao P, Majumder M, Hatzoglou M, Fox PL (2009) A stress-responsive RNA switch regulates VEGFA expression. Nature 457(7231):915–919

    CAS  PubMed Central  PubMed  Google Scholar 

  81. Essafi-Benkhadir K, Onesto C, Stebe E, Moroni C, Pages G (2007) Tristetraprolin inhibits Ras-dependent tumor vascularization by inducing vascular endothelial growth factor mRNA degradation. Mol Biol Cell 18(11):4648–4658

    CAS  PubMed Central  PubMed  Google Scholar 

  82. Fellows A, Griffin ME, Petrella BL, Zhong L, Parvin-Nejad FP, Fava R, Morganelli P, Robey RB, Nichols RC (2012) AUF1/hnRNP D represses expression of VEGF in macrophages. Mol Biol Cell 23(8):1414–1422

    CAS  PubMed Central  PubMed  Google Scholar 

  83. Levy NS, Chung S, Furneaux H, Levy AP (1998) Hypoxic stabilization of vascular endothelial growth factor mRNA by the RNA-binding protein HuR. J Biol Chem 273(11):6417–6423

    CAS  PubMed  Google Scholar 

  84. de Vries S, Naarmann de Vries IS, Urlaub H, Lue H, Bernhagen J, Ostareck DH, Ostareck-Lederer A (2013) Identification of DEAD-box RNA helicase 6 (DDX6) as a cellular modulator of vascular endothelial growth factor expression under hypoxia. J Biol Chem 288(8):5815–5827

    PubMed  Google Scholar 

  85. Goldberg-Cohen I, Furneauxb H, Levy AP (2002) A 40-bp RNA element that mediates stabilization of vascular endothelial growth factor mRNA by HuR. J Biol Chem 277(16):13635–13640

    CAS  PubMed  Google Scholar 

  86. Dang LT, Lawson ND, Fish JE (2013) MicroRNA control of vascular endothelial growth factor signaling output during vascular development. Arterioscler Thromb Vasc Biol 33(2):193–200

    CAS  PubMed Central  PubMed  Google Scholar 

  87. Chang SH, Lu YC, Li X, Hsieh WY, Xiong Y, Ghosh M, Evans T, Elemento O, Hla T (2013) Antagonistic function of the RNA-binding protein HuR and miR-200b in post-transcriptional regulation of VEGF-A expression and angiogenesis. J Biol Chem 288:4908–4921

    CAS  PubMed  Google Scholar 

  88. Weijts BG, Bakker WJ, Cornelissen PW, Liang KH, Schaftenaar FH, Westendorp B, de Wolf CA, Paciejewska M, Scheele CL, Kent L, Leone G, Schulte-Merker S, de Bruin A (2012) E2F7 and E2F8 promote angiogenesis through transcriptional activation of VEGFA in cooperation with HIF1. EMBO J 31(19):3871–3884

    CAS  PubMed  Google Scholar 

  89. Li J, Ran C, Li E, Gordon F, Comstock G, Siddiqui H, Cleghorn W, Chen HZ, Kornacker K, Liu CG, Pandit SK, Khanizadeh M, Weinstein M, Leone G, de Bruin A (2008) Synergistic function of E2F7 and E2F8 is essential for cell survival and embryonic development. Dev Cell 14(1):62–75

    CAS  PubMed Central  PubMed  Google Scholar 

  90. Qin G, Kishore R, Dolan CM, Silver M, Wecker A, Luedemann CN, Thorne T, Hanley A, Curry C, Heyd L, Dinesh D, Kearney M, Martelli F, Murayama T, Goukassian DA, Zhu Y, Losordo DW (2006) Cell cycle regulator E2F1 modulates angiogenesis via p53-dependent transcriptional control of VEGF. Proc Natl Acad Sci USA 103(29):11015–11020

    CAS  PubMed  Google Scholar 

  91. Shie JL, Wu G, Wu J, Liu FF, Laham RJ, Oettgen P, Li J (2004) RTEF-1, a novel transcriptional stimulator of vascular endothelial growth factor in hypoxic endothelial cells. J Biol Chem 279(24):25010–25016

    CAS  PubMed  Google Scholar 

  92. Yan SF, Fujita T, Lu J, Okada K, Shan Zou Y, Mackman N, Pinsky DJ, Stern DM (2000) Egr-1, a master switch coordinating upregulation of divergent gene families underlying ischemic stress. Nat Med 6(12):1355–1361

    CAS  PubMed  Google Scholar 

  93. Jin Y, Wu J, Song X, Song Q, Cully BL, Messmer-Blust A, Xu M, Foo SY, Rosenzweig A, Li J (2011) RTEF-1, an upstream gene of hypoxia-inducible factor-1alpha, accelerates recovery from ischemia. J Biol Chem 286(25):22699–22705

    CAS  PubMed  Google Scholar 

  94. Messmer-Blust AF, Zhang C, Shie JL, Song Q, He P, Lubenec I, Liu Y, Sellke F, Li J (2012) Related transcriptional enhancer factor 1 increases endothelial-dependent microvascular relaxation and proliferation. J Vasc Res 49(3):249–259

    CAS  PubMed  Google Scholar 

  95. Xu M, Jin Y, Song Q, Wu J, Philbrick MJ, Cully BL, An X, Guo L, Gao F, Li J (2011) The endothelium-dependent effect of RTEF-1 in pressure overload cardiac hypertrophy: role of VEGF-B. Cardiovasc Res 90(2):325–334

    CAS  PubMed  Google Scholar 

  96. Stein I, Itin A, Einat P, Skaliter R, Grossman Z, Keshet E (1998) Translation of vascular endothelial growth factor mRNA by internal ribosome entry: implications for translation under hypoxia. Mol Cell Biol 18(6):3112–3119

    CAS  PubMed Central  PubMed  Google Scholar 

  97. Brahimi-Horn MC, Pouyssegur J (2005) The hypoxia-inducible factor and tumor progression along the angiogenic pathway. Int Rev Cytol 242:157–213

    CAS  PubMed  Google Scholar 

  98. Loboda A, Jazwa A, Jozkowicz A, Molema G, Dulak J (2006) Angiogenic transcriptome of human microvascular endothelial cells: Effect of hypoxia, modulation by atorvastatin. Vascul Pharmacol 44(4):206–214

    CAS  PubMed Central  PubMed  Google Scholar 

  99. Loboda A, Stachurska A, Florczyk U, Rudnicka D, Jazwa A, Wegrzyn J, Kozakowska M, Stalinska K, Poellinger L, Levonen AL, Yla-Herttuala S, Jozkowicz A, Dulak J (2009) HIF-1 induction attenuates Nrf2-dependent IL-8 expression in human endothelial cells. Antioxid Redox Signal 11(7):1501–1517

    CAS  PubMed  Google Scholar 

  100. Florczyk U, Czauderna S, Stachurska A, Tertil M, Nowak W, Kozakowska M, Poellinger L, Jozkowicz A, Loboda A, Dulak J (2011) Opposite effects of HIF-1alpha and HIF-2alpha on the regulation of IL-8 expression in endothelial cells. Free Radic Biol Med 51(10):1882–1892

    CAS  PubMed Central  PubMed  Google Scholar 

  101. 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-1alpha-deficient colon cancer cells. Nat Med 11(9):992–997

    CAS  PubMed  Google Scholar 

  102. Yu L, Hales CA (2011) Long-term exposure to hypoxia inhibits tumor progression of lung cancer in rats and mice. BMC Cancer 11:331

    CAS  PubMed Central  PubMed  Google Scholar 

  103. Kalliomaki TM, McCallum G, Lunt SJ, Wells PG, Hill RP (2008) Analysis of the effects of exposure to acute hypoxia on oxidative lesions and tumour progression in a transgenic mouse breast cancer model. BMC Cancer 8:151

    PubMed Central  PubMed  Google Scholar 

  104. Volm M, Koomagi R (2000) Hypoxia-inducible factor (HIF-1) and its relationship to apoptosis and proliferation in lung cancer. Anticancer Res 20(3A):1527–1533

    CAS  PubMed  Google Scholar 

  105. Volm M, Koomagi R, Mattern J, Efferth T (2002) Expression profile of genes in non-small cell lung carcinomas from long-term surviving patients. Clin Cancer Res 8(6):1843–1848

    CAS  PubMed  Google Scholar 

  106. Leite de Oliveira R, Deschoemaeker S, Henze AT, Debackere K, Finisguerra V, Takeda Y, Roncal C, Dettori D, Tack E, Jonsson Y, Veschini L, Peeters A, Anisimov A, Hofmann M, Alitalo K, Baes M, D’Hooge J, Carmeliet P, Mazzone M (2012) Gene-targeting of Phd2 improves tumor response to chemotherapy and prevents side-toxicity. Cancer Cell 22(2):263–277

    CAS  PubMed  Google Scholar 

  107. Fleitas T, Martinez-Sales V, Vila V, Reganon E, Mesado D, Martin M, Gomez-Codina J, Montalar J, Reynes G (2013) VEGF and TSP1 levels correlate with prognosis in advanced non-small cell lung cancer. Clin Transl Oncol 15(11):897–902

    CAS  PubMed  Google Scholar 

  108. Tenan M, Fulci G, Albertoni M, Diserens AC, Hamou MF, El Atifi-Borel M, Feige JJ, Pepper MS, Van Meir EG (2000) Thrombospondin-1 is downregulated by anoxia and suppresses tumorigenicity of human glioblastoma cells. J Exp Med 191(10):1789–1798

    CAS  PubMed Central  PubMed  Google Scholar 

  109. Wu P, Yonekura H, Li H, Nozaki I, Tomono Y, Naito I, Ninomiya Y, Yamamoto H (2001) Hypoxia down-regulates endostatin production by human microvascular endothelial cells and pericytes. Biochem Biophys Res Commun 288(5):1149–1154

    CAS  PubMed  Google Scholar 

  110. Emara M, Obaid L, Johnson S, Bigam DL, Cheung PY (2007) The effect of hypoxia on plasma angiostatin and related factors in newborn pigs. Proc West Pharmacol Soc 50:47–52

    CAS  PubMed  Google Scholar 

  111. Jin Y, An X, Ye Z, Cully B, Wu J, Li J (2009) RGS5, a hypoxia-inducible apoptotic stimulator in endothelial cells. J Biol Chem 284(35):23436–23443

    CAS  PubMed  Google Scholar 

  112. An X, Jin Y, Guo H, Foo SY, Cully BL, Wu J, Zeng H, Rosenzweig A, Li J (2009) Response gene to complement 32, a novel hypoxia-regulated angiogenic inhibitor. Circulation 120(7):617–627

    CAS  PubMed Central  PubMed  Google Scholar 

Download references

Acknowledgments

The authors apologize for the inability to cite all of the important studies that have been performed in the field due to space limitations.

AL is supported by the Foundation for Polish Science—PARENT-BRIDGE Programme co-financed by the European Union within European Regional Development Fund (POMOST/2010-2/8) and she is the recipient of L'Oreal Poland for Women in Science Scholarship. The Faculty of Biochemistry, Biophysics and Biotechnology of the Jagiellonian University is a beneficiary of the structural funds from the European Union and the Polish Ministry of Science and Higher Education (grants No: POIG.02.01.00-12 064/08, POIG 01.01.02-00-109/09, POIG.02.02.00-014/08 and 01.01.02-00-069/09).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Agnieszka Łoboda .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2013 Springer-Verlag Wien

About this chapter

Cite this chapter

Łoboda, A., Józkowicz, A., Dulak, J. (2013). Molecular Mechanisms of Hypoxia-Regulated Angiogenesis. In: Dulak, J., Józkowicz, A., Łoboda, A. (eds) Angiogenesis and Vascularisation. Springer, Vienna. https://doi.org/10.1007/978-3-7091-1428-5_8

Download citation

Publish with us

Policies and ethics