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:

Hypoxia-inducible TAp73 supports tumorigenesis by regulating the angiogenic transcriptome

Abstract

The functional significance of the overexpression of unmutated TAp73, a homologue of the tumour suppressor p53, in multiple human cancers is unclear, but raises the possibility of unidentified roles in promoting tumorigenesis. We show here that TAp73 is stabilized by hypoxia, a condition highly prevalent in tumours, through HIF-1α-mediated repression of the ubiquitin ligase Siah1, which targets TAp73 for degradation. Consequently, TAp73-deficient tumours are less vascular and reduced in size, and conversely, TAp73 overexpression leads to increased vasculature. Moreover, we show that TAp73 is a critical regulator of the angiogenic transcriptome and is sufficient to directly activate the expression of several angiogenic genes. Finally, expression of TAp73 positively correlates with these angiogenic genes in several human tumours, and the angiogenic gene signature is sufficient to segregate the TAp73Hi- from TAp73Low-expressing tumours. These data demonstrate a pro-angiogenic role for TAp73 in supporting tumorigenesis, providing a rationale for its overexpression in cancers.

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: Hypoxia induces the stabilization of TAp73.
Figure 2: HIF-1α is required for TAp73 stabilization in response to hypoxia.
Figure 3: HIF-1α suppresses siah1 to regulate TAp73 on hypoxia.
Figure 4: p73 is required for efficient vasculature in tumours.
Figure 5: The angiogenic transcriptome is regulated by TAp73.
Figure 6: TAp73 regulates angiogenic genes independent of HIF-1α and its apoptotic potential.
Figure 7: Positive correlation between TAp73, Vegf-A and HIF-1α protein expression in human tumours.
Figure 8: Correlation of the expression of TAp73, SIAH1 and several angiogenic genes in clinical gastric tumour samples.

Similar content being viewed by others

References

  1. Melino, G., De Laurenzi, V. & Vousden, K. H. p73: friend or foe in tumorigenesis. Nat. Rev. Cancer 2, 605–615 (2002).

    CAS  PubMed  Google Scholar 

  2. Stiewe, T. & Pützer, B. M. Role of p73 in malignancy: tumor suppressor or oncogene? Cell Death Differ. 9, 237–245 (2002).

    CAS  PubMed  Google Scholar 

  3. Bisso, A., Collavin, L. & Del Sal, G. p73 as a pharmaceutical target for cancer therapy. Curr. Pharm. Des. 17, 578–590 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Lin, K. W., Nam, S. Y., Toh, W. H., Dulloo, I. & Sabapathy, K. Multiple stress signals induce p73beta accumulation. Neoplasia 6, 546–557 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Tomasini, R. et al. TAp73 knockout shows genomic instability with infertility and tumor suppressor functions. Genes Dev. 22, 2677–2691 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Levrero, M. et al. The p53/p63/p73 family of transcription factors: overlapping and distinct functions. J. Cell Sci. 113, 1661–1670 (2000).

    CAS  PubMed  Google Scholar 

  7. Buhlmann, S. & Pützer, B. M. DNp73 a matter of cancer: mechanisms and clinical implications. Biochim. Biophys. Acta 1785, 207–216 (2008).

    CAS  PubMed  Google Scholar 

  8. Gong, J. G. et al. The tyrosine kinase c-Abl regulates p73 in apoptotic response to cisplatin-induced DNA damage. Nature 399, 806–809 (1999).

    CAS  PubMed  Google Scholar 

  9. Agami, R., Blandino, G., Oren, M. & Shaul, Y. Interaction of c-Abl and p73α and their collaboration to induce apoptosis. Nature 399, 809–813 (1999).

    CAS  PubMed  Google Scholar 

  10. Yuan, Z. M. et al. p73 is regulated by tyrosine kinase c-Abl in the apoptotic response to DNA damage. Nature 399, 814–817 (1999).

    CAS  PubMed  Google Scholar 

  11. Donehower, L. A. et al. Mice deficient for p53 are developmentally normal but susceptible to spontaneous tumours. Nature 356, 215–221 (1992).

    CAS  PubMed  Google Scholar 

  12. Zaika, A. I., Kovalev, S., Marchenko, N. D. & Moll, U. M. Overexpression of the wild type p73 gene in breast cancer tissues and cell lines. Cancer Res. 59, 3257–3263 (1999).

    CAS  PubMed  Google Scholar 

  13. Kang, M. J. et al. Loss of imprinting and elevated expression of wild-type p73 in human gastric adenocarcinoma. Clin. Cancer Res. 6, 1767–1771 (2000).

    CAS  PubMed  Google Scholar 

  14. Vikhanskaya, F. et al. p73 supports cellular growth through c-Jun-dependent AP-1 transactivation. Nat. Cell Biol. 9, 698–705 (2007).

    CAS  PubMed  Google Scholar 

  15. Toh, W. H., Logette, E., Corcos, L. & Sabapathy, K. TAp73β and DNp73β activate the expression of the pro-survival caspase-2S. Nucleic Acids Res. 36, 4498–5509 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Du, W. et al. TAp73 enhances the pentose phosphate pathway and supports cell proliferation. Nat. Cell. Biol. 15, 991–1000 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Lefkimmiatis, K. et al. p73 and p63 sustain cellular growth by transcriptional activation of cell cycle progression genes. Cancer Res. 69, 8563–8571 (2009).

    CAS  PubMed  Google Scholar 

  18. Bertout, J. A., Patel, S. A. & Simon, M. C. The impact of O2 availability on human cancer. Nat. Rev. Cancer 8, 967–975 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Semenza, G. L. Defining the role of hypoxia-inducible factor 1 in cancer biology and therapeutics. Oncogene 29, 625–634 (2010).

    CAS  PubMed  Google Scholar 

  20. Semenza, G. L. Targeting HIF-1 for cancer therapy. Nat. Rev. Cancer 3, 721–732 (2003).

    CAS  Google Scholar 

  21. Yan, Y. et al. Inhibitors of ubiquitin-activating enzyme (E1), a new class of potential cancer therapeutics. Cancer Res. 67, 9472–9481 (2007).

    Google Scholar 

  22. Pagano, M. et al. Role of the ubiquitin-proteasome pathway in regulating abundance of the cyclin-dependent kinase inhibitor p27. Science 269, 682–685 (1995).

    CAS  PubMed  Google Scholar 

  23. Kaelin, W. G. Jr The von Hippel–Lindau tumour suppressor protein: O2 sensing and cancer. Nat. Rev. Cancer 8, 865–873 (2008).

    CAS  PubMed  Google Scholar 

  24. Chew, E. H., Poobalasingam, T., Hawkey, C. J. & Hagen, T. Characterization of cullin-based E3 ubiquitin ligases in intact mammalian cells–evidence for cullin dimerization. Cell. Signal. 19, 1071–1080 (2007).

    CAS  PubMed  Google Scholar 

  25. Narita, T. et al. Identification of a novel small molecule HIF-1α translation inhibitor. Clin. Cancer Res. 15, 6128–6136 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Kung, A. L. et al. Small molecule blockade of transcriptional coactivation of the hypoxia-inducible factor pathway. Cancer Cell 6, 33–43 (2004).

    CAS  PubMed  Google Scholar 

  27. Hu, G. & Fearon, E. R. Siah-1 N-terminal RING domain is required for proteolysis function, and C-terminal sequences regulate oligomerization and binding to target proteins. Mol. Cell. Biol. 19, 724–732 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Pugh, C. W. & Ratcliffe, P. J. Regulation of angiogenesis by hypoxia: role of the HIF system. Nat. Med. 9, 677–684 (2003).

    CAS  Google Scholar 

  29. Liao, D. & Johnson, R. S. Hypoxia: a key regulator of angiogenesis in cancer. Cancer Metastasis Rev. 26, 281–290 (2007).

    CAS  PubMed  Google Scholar 

  30. Wernig, M. et al. In vitro reprogramming of fibroblasts into a pluripotent ES-cell-like state. Nature 448, 318–324 (2007).

    CAS  PubMed  Google Scholar 

  31. Battegay, E. J., Rupp, J., Iruela-Arispe, L., Sage, E. H. & Pech, M. PDGF-BB modulates endothelial proliferation and angiogenesis in vitro via PDGF beta-receptors. J. Cell Biol. 125, 917–928 (1994).

    CAS  PubMed  Google Scholar 

  32. Ten Dijke, P., Goumans, M. J. & Pardali, E. Endoglin in angiogenesis and vascular diseases. Angiogenesis 11, 79–89 (2008).

    PubMed  Google Scholar 

  33. Weckbach, L. T. et al. Midkine acts as proangiogenic cytokine in hypoxia-induced angiogenesis. Am. J. Physiol. Heart Circ. Physiol. 303, H429–H438 (2012).

    CAS  PubMed  Google Scholar 

  34. Rosenbluth, J. M., Mays, D. J., Pino, M. F., Tang, L. J. & Pietenpol, J. A. A gene signature-based approach identifies mTOR as a regulator of p73. Mol. Cell. Biol. 28, 5951–5964 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Koeppel, M. et al. Crosstalk between c-Jun and TAp73α/β contributes to the apoptosis-survival balance. Nucleic Acids Res. 39, 6069–6085 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Vikhanskaya, F. et al. p73 overexpression increases VEGF and reduces thrombospondin-1 production: implications for tumor angiogenesis. Oncogene 20, 7293–7300 (2001).

    CAS  PubMed  Google Scholar 

  37. Zhang, L. et al. Wild-type p53 suppresses angiogenesis in human leiomyosarcoma and synovial sarcoma by transcriptional suppression of vascular endothelial growth factor expression. Cancer Res. 60, 3655–3661 (2000).

    CAS  PubMed  Google Scholar 

  38. Chen, C., Pore, N., Behrooz, A., Ismail-Beigi, F. & Maity, A. Regulation of glut1 mRNA by hypoxia-inducible factor-1. Interaction between H-ras and hypoxia. J. Biol. Chem. 276, 9519–9525 (2001).

    CAS  PubMed  Google Scholar 

  39. Potter, C. & Harris, A. L. Hypoxia inducible carbonic anhydrase IX, marker of tumour hypoxia, survival pathway and therapy target. Cell Cycle 3, 164–167 (2004).

    CAS  PubMed  Google Scholar 

  40. Li, C. W. et al. Role of p63/p73 in epithelial remodeling and their response to steroid treatment in nasal polyposis. J. Allergy Clin. Immunol. 127, 765–772 (2011).

    CAS  PubMed  Google Scholar 

  41. Hsu, Y. C. et al. Increased expression of hypoxia-inducible factor 1α in the nasal polyps. Am. J. Otolaryngol. 28, 379–383 (2007).

    CAS  PubMed  Google Scholar 

  42. Ooi, C. H. et al. Oncogenic pathway combinations predict clinical prognosis in gastric cancer. PLoS Genet. 5, e1000676 (2009).

    PubMed  PubMed Central  Google Scholar 

  43. Kitadai, Y. Angiogenesis and lymphangiogenesis of gastric cancer. J. Oncol. 2010, 468725 (2010).

    PubMed  PubMed Central  Google Scholar 

  44. DiComo, C. J., Gaiddon, C. & Prives, C. p73 function is inhibited by tumor-derived p53 mutants in mammalian cells. Mol. Cell. Biol. 19, 1438–1449 (1999).

    CAS  Google Scholar 

  45. Pozniak, C. D. et al. An anti-apoptotic role for the p53 family member, p73, during developmental neuron death. Science 289, 304–306 (2000).

    CAS  PubMed  Google Scholar 

  46. Weiss, R. H. & Howard, L. L. p73 is a growth-regulated protein in vascular smooth muscle cells and is present at high levels in human atherosclerotic plaque. Cell Signal. 13, 727–733 (2001).

    CAS  PubMed  Google Scholar 

  47. Fernandez-Alonso, R. et al. p73 is required for endothelial cell differentiation, migration and the formation of vascular networks regulating VEGF and TGFβ signaling. Cell Death Differ. (2015)10.1038/cdd.2014.214

  48. Amelio, I. et al. TAp73 opposes tumor angiogenesis by promoting hypoxia-inducible factor 1α degradation.. Proc. Natl Acad. Sci. USA 112, 226–231 (2015).

    CAS  PubMed  Google Scholar 

  49. Stantic, M. et al. TAp73 suppresses tumor angiogenesis through repression of proangiogenic cytokines and HIF-1α activity. Proc. Natl Acad. Sci. USA 112, 220–2225 (2015).

    CAS  PubMed  Google Scholar 

  50. Nakayama, K. et al. Siah2 regulates stability of prolyl-hydroxylases, controls HIF1α abundance, and modulates physiological responses to hypoxia. Cell 117, 941–952 (2004).

    CAS  PubMed  Google Scholar 

  51. Senoo, M., Matsumura, Y. & Habu, S. TAp63γ (p51A) and dNp63α (p73L), two major isoforms of the p63 gene, exert opposite effects on the vascular endothelial growth factor (VEGF) gene expression. Oncogene 21, 2455–2465 (2002).

    CAS  PubMed  Google Scholar 

  52. Farhang, G. M., Goossens, S. & Haigh, J. J. The p53 family and VEGF regulation: ”It’s complicated”. Cell Cycle 12, 1331–1332 (2013).

    Google Scholar 

  53. Salimath, B., Marmé, D. & Finkenzeller, G. Expression of the vascular endothelial growth factor gene is inhibited by p73. Oncogene 20, 7293–7300 (2001).

    Google Scholar 

  54. Balint, E., Phillips, A. C., Kozlov, S., Stewart, C. L. & Vousden, K. H. Induction of p57(KIP2) expression by p73beta. Proc. Natl Acad. Sci. USA 99, 3529–3534 (2002).

    CAS  PubMed  Google Scholar 

  55. Woltjen, K. et al. piggyBac transposition reprograms fibroblasts to induced pluripotent stem cells. Nature 458, 766–770 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Hsiao, T. H. et al. Utilizing signature-score to identify oncogenic pathways of cholangiocarcinoma. Trans. Cancer Res. 2, 6–17 (2013).

    CAS  Google Scholar 

Download references

Acknowledgements

We thank Y. Li (NUS), E. Mationg-Kalaw (SGH Path-NCCS Translational Laboratory) and M. Koh (Singhealth Tissue Repository) for technical assistance, and C. Lim and X. Min for help with statistical analysis. We are grateful to F. Mechta-Grigoriou, T. Hagen, S-I. Matsuzawa and T. W. Mak for the vegf-A promoter–luciferase, dnHIF-1α, siah1 WT/ΔRING mutant plasmids, and the TAp73−/− and TAp73+/+ MEFs, respectively. We also acknowledge the Advanced Molecular Pathology Laboratory (AMPL) for the histological services provided. This study was supported by grants from the National Medical Research Council to K.S., and by grant SAF2012-36143 from the Spanish Ministerio de Economía y Competitividad, co-financed by FEDER funds to M.C.M.

Author information

Authors and Affiliations

Authors

Contributions

I.D. designed the experiments, performed the research work, analysed the data and helped with writing of the manuscript. B.H.P. and R.O. performed the research work and analysed the data. S.Y.T., C.W.L. and D.Y.W. performed the immunohistochemistry work on human samples. A.V. and L.K.G. performed the gastric cancer data set analysis. M.M-L., M.M.M. and M.C.M. performed the experiments related to iPSC generation and the associated teratoma studies. W.X. and F.M. provided the TAp73 antibody and advised on its use. K.S. designed the experiments, analysed the data, supervised the project and wrote the manuscript. All authors reviewed the manuscript.

Corresponding author

Correspondence to Kanaga Sabapathy.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 8 VHL is dispensable for oxygen-mediated expression of TAp73.

A Levels of transiently transfected TAp73α and TAp73β in H1299 cells were assessed by immunoblotting (IB) upon exposure to 1% O2 for 24 h (right panel). Saos-2 TAp73α-inducible cells were induced with 2 μg ml-1 of doxycycline for 24 h in 1% O2 and TAp73 levels were determined by IB (left panel). B,C Effects of 1% O2 and reoxygenation (Reox.) with 21% O2 on transfected TAp73β in H1299 in which vhl expression was silenced by siRNA-mediated silencing (using 2 independent siRNAs) (B) was assessed by IB. Similarly, HEK293 cells inducibly expressing the dominant negative form of Ubc12 which inhibits all cullin-based E3 ligases (left panel), or H1299 cells transiently transfected with HA-Ubc12 (right panel) (C) were used to analyze effect on TAp73β. Arrowhead represents Vhl and represents non-specific band in H1299 blot (B). Blots are representative of 3 independent experiments. Uncropped images of blots/gels are shown in Supplementary Fig. 6.

Supplementary Figure 9 Requirement of HIF-1α for suppression of Siah1 upon hypoxia.

A Evaluation of the effect of 1% O2 on expression of selected candidate E3 ligases in H1299 cells after 24 h by real-time qPCR. B Effects of silencing hif-1α and/or hif-1β in H1299 cells on siah1 and glut-1 were determined by real-time qPCR upon hypoxia. C,D Effects of treatment with chetomin (CHT) (C) or silencing hif-1α by siRNA (D) on other E3 ligases were determined by real-time qPCR. E Levels of TAp73β upon hypoxia and reoxygenation were determined after silencing the expression of siah1 using two different siRNAs, and levels of siah1 were determined by real-time qPCR. Data from one experiment is shown as average of two biological replicates from two separate cellular extracts, which yielded similar results (for Supplementary Fig. 2a–e) . All data are representative of 3 independent experiments. Uncropped images of blots/gels are shown in Supplementary Fig. 6.

Supplementary Figure 10 TAp73 regulates angiogenesis.

A Semi-quantitative RT-PCR shows the expression of TAp73 and DNp73 in wild-type, TAp73−/− and DNp73−/− MEFs. Arrowheads show TAp73 and DNp73 transcripts. B,C TAp73 and DNp73 transcripts in WT and p73-deficient (KO) induced pluripotent stem (iPS) cells were determined by real-time qPCR analyses (WT iPSC n = 8 clones, p73KO iPSC n = 5 clones) (B, left panel). Morphology of the iPS cells is shown on the right. Scale bars: 250 μm. (B). Kinetics of proliferation of these cells were determined by cellular counting over several passages (C). Bars represent mean values ± s.d.; experiments were repeated three times with two replicas each sample. (P < 0.001 by Student’s t-test). D Tumors generated by injection with H1299-TAp73β-inducible cells into SCID mice were stained for TUNEL and Ki67, and representative pictures are shown. Scale bars: 100 μm. Right most panels show quantification using Angiosight (n = 3 tumors (− Dox),n = 5 tumors (+ Dox), p = n.s by Student’s t-test). Data are presented as means ± s.d. E Saos2-TAp73β-inducible cells were subcutaneously injected into SCID mice and once tumors grew to a size of about 100 mm-3, mice were then divided into two groups: one control group (n = 3 mice) and one group where mice were gavaged with 2 mg ml-1 of doxycycline daily for 6 days (n = 3 mice) before tumors were harvested for immunohistochemical (IHC) analysis. Tumors were stained for H&E, TAp73 and CD34. Scale bars: 100 μm. Uncropped images of blots/gels are shown in Supplementary Fig. 6.

Supplementary Figure 11 TAp73 is required and sufficient to regulate expression of angiogenic genes.

A,B Real-time qPCR analysis was carried out for several pro-angiogenic genes using TAp73+/+ and TAp73−/− MEFs (n = 4 biological replicates from four separate cellular extracts) (A) or upon silencing of p73 by siRNA in H1299 cells (B). (P < 0.05 by unpaired Mann-Whitney test in A). Data are presented as means ± s.d. for A. C Expression of selected angiogenic genes upon siah1 silencing in TAp73+/+ and TAp73−/− MEFs was determined by real-time qPCR. Expression levels are shown as fold-change based on control scrambled siRNA levels. (Data from one of n = 3 independent experiments is shown). D,E Expression of several pro-angiogenic genes was assessed using Saos-2-TAp73β inducible cells induced for 6 h or 10 h (D), or in Saos2 cells expressing TAp73α, TAp73β or p53, after 8 h and 24 h (E), by real-time qPCR. Lower panel shows expression of TAp73α, TAp73β or p53 by IB (E). F Table showing details of pro-angiogenic genes found to have potential TAp73 binding sites in their genomic regulatory elements after analysis of chromatin immunoprecipitation (ChIP)-Seq data from Koeppel et al., 201135. Data from one experiment is shown as average of two biological replicates from two separate cellular extracts, which yielded similar results (for Supplementary Fig. 4b, d, e). All data are representative of 3 independent experiments. Uncropped images of blots/gels are shown in Supplementary Fig. 6.

Supplementary Figure 12 p73 expression correlates with angiogenic genes in human clinical samples.

A Effect of TAp73-specific blocking peptide was analysed by immunohistochemical staining with non-related antibodies against MNF116 or Ki67, in both normal and colonic adenocarcinoma tissues in the presence or absence of the peptide. Scale bars: 0.9 μm. B Expression of TAp73, Vegf-A, CAIX (another hypoxic marker) and HIF-1α in nasal poly samples from patients with nasal polyposis (n = 4 samples). Scale bars: 50 μm. Representative pictures are shown.

Supplementary Figure 13 Uncropped gel pictures for all gels in the article.

Supplementary Table 1 Differential regulation of several physiological processes by TAp73 in MEFs.
Supplementary Table 2 49-angiogenic genes signature from TAp73 differentially regulated genes.
Supplementary Table 3 Molecular networks associated with the 49-angiogenic genes signature.
Supplementary Table 4 Molecular networks associated with the 28-pro-angiogenic genes signature.
Supplementary Table 5 List of 28 pro-angiogenic genes used for further validation.
Supplementary Table 6 Selected genes with most significant correlation in gastric cancer dataset.
Supplementary Table 7 List of primers (mouse and human).

Supplementary information

Supplementary Information

Supplementary Information (PDF 2363 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dulloo, I., Phang, B., Othman, R. et al. Hypoxia-inducible TAp73 supports tumorigenesis by regulating the angiogenic transcriptome. Nat Cell Biol 17, 511–523 (2015). https://doi.org/10.1038/ncb3130

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3130

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer