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:

Functional interplay between the DNA-damage-response kinase ATM and ARF tumour suppressor protein in human cancer

A Corrigendum to this article was published on 01 October 2013

This article has been updated

Abstract

The DNA damage response (DDR) pathway and ARF function as barriers to cancer development. Although commonly regarded as operating independently of each other, some studies proposed that ARF is positively regulated by the DDR. Contrary to either scenario, we found that in human oncogene-transformed and cancer cells, ATM suppressed ARF protein levels and activity in a transcription-independent manner. Mechanistically, ATM activated protein phosphatase 1, which antagonized Nek2-dependent phosphorylation of nucleophosmin (NPM), thereby liberating ARF from NPM and rendering it susceptible to degradation by the ULF E3-ubiquitin ligase. In human clinical samples, loss of ATM expression correlated with increased ARF levels and in xenograft and tissue culture models, inhibition of ATM stimulated the tumour-suppressive effects of ARF. These results provide insights into the functional interplay between the DDR and ARF anti-cancer barriers, with implications for tumorigenesis and treatment of advanced tumours.

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

Access options

Buy this article

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

Figure 1: ATM regulates p14ARF protein levels in vitro.
Figure 2: ATM regulates p14ARF protein stability.
Figure 3: p14ARF protein stability is regulated by the ATM–PP1–Nek2-mediated pathway.
Figure 4: Phosphorylation of NPM/B23 at Ser 70 and Ser 88 enhances p14ARF protein stability.
Figure 5: ATM-dependent p14ARF regulation affects ribosomal biogenesis.
Figure 6: ATM-dependent p14ARF regulation affects cell growth.
Figure 7: ATM abrogation is inversely related to ARF expression in lung cancer.
Figure 8: ATM abrogation stalls growth in p53-null xenografts in an ARF-dependent manner.

Similar content being viewed by others

Change history

  • 28 August 2013

    In the version of this Article originally published, there was an error in Fig. 8e. The arrow beside ARF in green should have been pointing up and the arrow beside ARF in red should have been pointing down. This has been corrected in the HTML and PDF versions of the Article.

References

  1. Sherr, C. J. Divorcing ARF and p53: an unsettled case. Nat. Rev. Cancer 6, 663–673 (2006).

    Article  CAS  PubMed  Google Scholar 

  2. Kim, W. Y. & Sharpless, N. E. The regulation of INK4/ARF in cancer and aging. Cell 127, 265–275 (2006).

    Article  CAS  PubMed  Google Scholar 

  3. Bartek, J., Bartkova, J. & Lukas, J. DNA damage signalling guards against activated oncogenes and tumour progression. Oncogene 26, 7773–7779 (2007).

    Article  CAS  PubMed  Google Scholar 

  4. Halazonetis, T. D., Gorgoulis, V. G. & Bartek, J. An oncogene-induced DNA damage model for cancer development. Science 319, 1352–1355 (2008).

    Article  CAS  PubMed  Google Scholar 

  5. Jackson, S. P. & Bartek, J. The DNA-damage response in human biology and disease. Nature 461, 1071–1078 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Negrini, S., Gorgoulis, V. G. & Halazonetis, T. D. Genomic instability–an evolving hallmark of cancer. Nat. Rev. Mol. Cell Biol. 11, 220–228 (2010).

    Article  CAS  PubMed  Google Scholar 

  7. Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011).

    Article  CAS  PubMed  Google Scholar 

  8. Kastan, M. B. & Bartek, J. Cell-cycle checkpoints and cancer. Nature 432, 316–323 (2004).

    Article  CAS  PubMed  Google Scholar 

  9. Kamijo, T. et al. Tumor suppression at the mouse INK4a locus mediated by the alternative reading frame product p19ARF. Cell 91, 649–659 (1997).

    Article  CAS  PubMed  Google Scholar 

  10. Stott, F. J. et al. The alternative product from the human CDKN2A locus, p14ARF, participates in a regulatory feedback loop with p53 and MDM2. EMBO J. 17, 5001–5014 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Kamijo, T. et al. Loss of the ARF tumor suppressor reverses premature replicative arrest but not radiation hypersensitivity arising from disabled atm function. Cancer Res. 59, 2464–2469 (1999).

    CAS  PubMed  Google Scholar 

  12. Shiloh, Y. ATM and related protein kinases: safeguarding genome integrity. Nat. Rev. Cancer 3, 155–168 (2003).

    Article  CAS  PubMed  Google Scholar 

  13. Efeyan, A. et al. Limited role of murine ATM in oncogene-induced senescence and p53-dependent tumor suppression. PLoS One 4, e5475 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Hickson, I. et al. Identification and characterization of a novel and specific inhibitor of the ataxia-telangiectasia mutated kinase ATM. Cancer Res. 64, 9152–9159 (2004).

    Article  CAS  PubMed  Google Scholar 

  15. DiTullio, R. A. Jr et al. 53BP1 functions in an ATM-dependent checkpoint pathway that is constitutively activated in human cancer. Nat. Cell Biol. 12, 998–1002 (2002).

    Article  CAS  Google Scholar 

  16. Tsantoulis, P. K. & Gorgoulis, V. G. Involvement of E2F transcription factor family in cancer. Eur. J. Cancer 41, 2403–2414 (2005).

    Article  CAS  PubMed  Google Scholar 

  17. Liontos, M. et al. Modulation of the E2F1-driven cancer cell fate by the DNA damage response machinery and potential novel E2F1 targets in osteosarcomas. Am. J. Pathol. 175, 376–391 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Ramirez, R. D. et al. Immortalization of human bronchial epithelial cells in the absence of viral oncoproteins. Cancer Res. 64, 9027–9034 (2004).

    Article  CAS  PubMed  Google Scholar 

  19. Sato, M. et al. Multiple oncogenic changes (K-RAS(V12), p53 knockdown, mutant EGFRs, p16 bypass, telomerase) are not sufficient to confer a full malignant phenotype on human bronchial epithelial cells. Cancer Res. 66, 2116–2128 (2006).

    Article  CAS  PubMed  Google Scholar 

  20. Inoue, R., Asker, C., Klangby, U., Pisa, P. & Wiman, K. G. Induction of the human ARF protein by serum starvation. Anticancer Res. 19, 2939–2943 (1999).

    CAS  PubMed  Google Scholar 

  21. Chen, D., Shan, J., Zhu, W. G., Qin, J. & Gu, W. Transcription-independent ARF regulation in oncogenic stress-mediated p53 responses. Nature 464, 624–627 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Enomoto, T., Lindstrom, M. S., Jin, A., Ke, H. & Zhang, Y. Essential role of the B23/NPM core domain in regulating ARF binding and B23 stability. J. Biol. Chem. 281, 18463–18472 (2006).

    Article  CAS  PubMed  Google Scholar 

  23. Colombo, E. et al. Nucleophosmin is required for DNA integrity and p19Arf protein stability. Mol. Cell. Biol. 25, 8874–8886 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Kuo, M. L., den Besten, W., Bertwistle, D., Roussel, M. F. & Sherr, C. J. N-terminal polyubiquitination and degradation of the Arf tumor suppressor. Genes Dev. 18, 1862–1874 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Huang, M., Itahana, K., Zhang, Y. & Mitchell, B. S. Depletion of guanine nucleotides leads to the Mdm2-dependent proteasomal degradation of nucleostemin. Cancer Res. 69, 3004–3012 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Lee, C., Smith, B. A., Bandyopadhyay, K. & Gjerset, R. A. DNA damage disrupts the p14ARF-B23 (nucleophosmin) interaction and triggers a transient subnuclear redistribution of p14ARF. Cancer Res. 65, 9834–9842 (2005).

    Article  CAS  PubMed  Google Scholar 

  27. Matsuoka, S. et al. ATM and ATR substrate analysis reveals extensive protein networks responsive to DNA damage. Science 316, 1160–1166 (2007).

    Article  CAS  PubMed  Google Scholar 

  28. Lin, C. Y. et al. Dephosphorylation of nucleophosmin by PP1 β facilitates pRB binding and consequent E2F1-dependent DNA repair. Mol. Biol. Cell 21, 4409–4417 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Tang, X. et al. A novel ATM-dependent pathway regulates protein phosphatase 1 in response to DNA damage. Mol. Cell. Biol. 28, 2559–2566 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Scheer, U. & Benavente, R. Functional and dynamic aspects of the mammalian nucleolus. Bioessays 12, 14–21 (1990).

    Article  CAS  PubMed  Google Scholar 

  31. Trinkle-Mulcahy, L., Sleeman, J. E. & Lamond, A. I. Dynamic targeting of protein phosphatase 1 within the nuclei of living mammalian cells. J. Cell Sci. 114, 4219–4228 (2001).

    CAS  PubMed  Google Scholar 

  32. Hayward, D. G. & Fry, A. M. Nek2 kinase in chromosome instability and cancer. Cancer Lett. 237, 155–166 (2006).

    Article  CAS  PubMed  Google Scholar 

  33. Noguchi, K., Fukazawa, H., Murakami, Y. & Uehara, Y. Nucleolar Nek11 is a novel target of Nek2A in G1/S-arrested cells. J. Biol. Chem. 279, 32716–32727 (2004).

    Article  CAS  PubMed  Google Scholar 

  34. Mitsuhashi, S. et al. Tautomycetin is a novel and specific inhibitor of serine/threonine protein phosphatase type 1, PP1. Biochem. Biophys. Res. Commun. 287, 328–331 (2001).

    Article  CAS  PubMed  Google Scholar 

  35. Colombo, E., Alcalay, M. & Pelicci, P. G. Nucleophosmin and its complex network: a possible therapeutic target in hematological diseases. Oncogene 30, 2595–2609 (2011).

    Article  CAS  PubMed  Google Scholar 

  36. Sugimoto, M., Kuo, M. L., Roussel, M. F. & Sherr, C. J. Nucleolar Arf tumor suppressor inhibits ribosomal RNA processing. Mol. Cell 11, 415–424 (2003).

    Article  CAS  PubMed  Google Scholar 

  37. Itahana, K. et al. Tumor suppressor ARF degrades B23, a nucleolar protein involved in ribosome biogenesis and cell proliferation. Mol. Cell 12, 1151–1164 (2003).

    Article  CAS  PubMed  Google Scholar 

  38. Grandori, C. et al. c-Myc binds to human ribosomal DNA and stimulates transcription of rRNA genes by RNA polymerase I. Nat. Cell Biol. 7, 311–318 (2005).

    Article  CAS  PubMed  Google Scholar 

  39. Lessard, F. et al. The ARF tumor suppressor controls ribosome biogenesis by regulating the RNA polymerase I transcription factor TTF-I. Mol. Cell 38, 539–550 (2010).

    Article  CAS  PubMed  Google Scholar 

  40. Huang, M., Ji, Y., Itahana, K., Zhang, Y. & Mitchell, B. Guanine nucleotide depletion inhibits pre-ribosomal RNA synthesis and causes nucleolar disruption. Leuk. Res. 32, 131–141 (2008).

    Article  CAS  PubMed  Google Scholar 

  41. Cox, J. & Mann, M. Quantitative, high-resolution proteomics for data-driven systems biology. Annu. Rev. Biochem. 80, 273–299 (2011).

    Article  CAS  PubMed  Google Scholar 

  42. Li, Z. et al. Systematic comparison of label-free, metabolic labeling, and isobaric chemical labeling for quantitative proteomics on LTQ Orbitrap Velos. J. Proteome. Res. 11, 1582–1590 (2012).

    Article  CAS  PubMed  Google Scholar 

  43. Falck, J., Mailand, N., Syljuasen, R. G., Bartek, J. & Lukas, J. The ATMChk2—Cdc25A checkpoint pathway guards against radioresistant DNA synthesis. Nature 410, 842–847 (2001).

    Article  CAS  PubMed  Google Scholar 

  44. Choi, J. et al. Selective requirement of H2B N-Terminal tail for p14ARF induced chromatin silencing. Nucleic. Acids Res. 39, 9167–9180 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Sarkaria, J. N. et al. Inhibition of ATM and ATR kinase activities by the radiosensitizing agent, caffeine. Cancer Res. 59, 4375–4382 (1999).

    CAS  PubMed  Google Scholar 

  46. Gorgoulis, V. G. et al. Alterations of the p16-pRb pathway and the chromosome locus 9p21-22 in non-small-cell lung carcinomas: relationship with p53 and MDM2 protein expression. Am. J. Pathol. 153, 1749–1765 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Gorgoulis, V. G. et al. Activation of the DNA damage checkpoint and genomic instability in human precancerous lesions. Nature 434, 907–913 (2005).

    Article  CAS  PubMed  Google Scholar 

  48. Zeng, Y., Kotake, Y., Pei, X. H., Smith, M. D. & Xiong, Y. p53 binds to and is required for the repression of Arf tumor suppressor by HDAC and polycomb. Cancer Res. 71, 2781–2792 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Jiang, H. et al. The combined status of ATM and p53 link tumor development with therapeutic response. Genes Dev. 23, 1895–1909 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Gorgoulis, V. G. & Halazonetis, T. D. Oncogene-induced senescence: the bright and dark side of the response. Curr. Opin. Cell Biol. 22, 816–827 (2010).

    Article  CAS  PubMed  Google Scholar 

  51. Di Micco, R. et al. DNA damage response activation in mouse embryonic fibroblasts undergoing replicative senescence and following spontaneous immortalization. Cell Cycle 7, 3601–3606 (2008).

    Article  CAS  PubMed  Google Scholar 

  52. Churchman, M. L., Roig, I., Jasin, M., Keeney, S. & Sherr, C. J. Expression of ARF tumor suppressor in spermatogonia facilitates meiotic progression in male germ cells. Plos Genetics 7, e1002157 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Takubo, K. et al. Stem cell defects in ATM-deficient undifferentiated spermatogonia through DNA-induced cell-cycle arrest. Cell Stem. Cell 2, 170–182 (2008).

    Article  CAS  PubMed  Google Scholar 

  54. Li, Y. et al. ATM activity contributes to the tumor-suppressing functions of p14ARF. Oncogene 23, 7355–7365 (2004).

    Article  CAS  PubMed  Google Scholar 

  55. Eymin, B. et al. p14ARF activates a Tip60-dependent and p53-independent ATM/ATR/CHK pathway in response to genotoxic stress. Mol. Cell Biol. 26, 4339–4350 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Khan, S. H., Moritsugu, J. & Wahl, G. M. Differential requirement for p19ARF in the p53-dependent arrest induced by DNA damage, microtubule disruption, and ribonucleotide depletion. Proc. Natl Acad. Sci. USA 97, 3266–3271 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Gorgoulis, V. G. et al. p53 activates ICAM-1 (CD54) expression in an NF-kB-independent manner. EMBO J. 22, 1567–1578 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Liontos, M. et al. Deregulated overexpression of hCdt1 and hCdc6 promotes malignant behavior. Cancer Res. 67, 10899–10909 (2007).

    Article  CAS  PubMed  Google Scholar 

  59. Sideridou, M. et al. Cdc6 expression represses E-cadherin transcription and activates adjacent replication origins. J Cell Biol. 195, 1123–1140 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Bartkova, J. et al. Oncogene-induced senescence is part of the tumorigenesis barrier imposed by DNA damage checkpoints. Nature 444, 633–637 (2006).

    Article  CAS  PubMed  Google Scholar 

  61. Hershko, T. & Ginsberg, D. Up-regulation of Bcl-2 homology 3 (BH3)-only proteins by E2F1 mediates apoptosis. J. Biol. Chem. 279, 8627–8634 (2004).

    Article  CAS  PubMed  Google Scholar 

  62. Cescutti, R., Negrini, S., Kohzaki, M. & Halazonetis, T. D. TopBP1 functions with 53BP1 in the G1 DNA damage checkpoint. EMBO J. 29, 3723–3732 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Andersen, J. S. et al. Directed proteomic analysis of the human nucleolus. Curr. Biol 12, 1–11 (2002).

    Article  PubMed  Google Scholar 

  64. Karakaidos, P. et al. Overexpression of the replication licensing regulators hCdt1 and hCdc6 characterizes a subset of non-small-cell lung carcinomas: synergistic effect with mutant p53 on tumor growth and chromosomal instability–evidence of E2F-1 transcriptional control over hCdt1. Am. J. Pathol. 165, 1351–1365 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Zacharatos, P. et al. Distinct expression patterns of the transcription factor E2F-1 in relation to tumour growth parameters in common human carcinomas. J. Pathol. 203, 744–753 (2004).

    Article  CAS  PubMed  Google Scholar 

  66. Honrado, E. et al. Immunohistochemical expression of DNA repair proteins in familial breast cancer differentiate BRCA2-associated tumors. J. Clin. Oncol. 23, 7503–7511 (2005).

    Article  CAS  PubMed  Google Scholar 

  67. Kilpivaara, O. et al. Correlation of CHEK2 protein expression and c.1100delC mutation status with tumor characteristics among unselected breast cancer patients. Int. J. Cancer 113, 575–580 (2005).

    Article  CAS  PubMed  Google Scholar 

  68. Geradts, J., Kratzke, R. A., Niehans, G. A. & Lincoln, C. E. Immunohistochemical detection of the cyclin-dependent kinase inhibitor 2/multiple tumor suppressor gene 1 (CDKN2/MTS1) product p16INK4A in archival human solid tumors: correlation with retinoblastoma protein expression. Cancer Res. 55, 6006–6011 (1995).

    CAS  PubMed  Google Scholar 

  69. Vestey, S. B. et al. p14ARF expression in invasive breast cancers and ductal carcinoma in situ–relationships to p53 and Hdm2. Breast Cancer Res 6, R571–R585 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Shilov, I. V. et al. The Paragon Algorithm, a next generation search engine that uses sequence temperature values and feature probabilities to identify peptides from tandem mass spectra. Mol. Cell Proteom. 6, 1638–1655 (2007).

    Article  CAS  Google Scholar 

  71. Tang, W. H., Shilov, I. V. & Seymour, S. L. Nonlinear fitting method for determining local false discovery rates from decoy database searches. J. Proteome Res. 7, 3661–3667 (2008).

    Article  CAS  PubMed  Google Scholar 

  72. R Development Core Team. R: a Language and Environment for Statistical Computing. R Foundation for Statistical Computing, Vienna, Austria. 409 (2012).

  73. Subramanian, A. et al. Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Mootha, V. K. et al. PGC-1alpha-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat. Genet. 34, 267–273 (2003).

    Article  CAS  PubMed  Google Scholar 

  75. Warnes, G. R. Includes R source code and/or documentation contributed by (in alphabetical order): Bolker, B. et al. gplots: various R programming tools for plotting data. R package version 2.10.1 (2011).

  76. Brooks, D. G., James, R. M, Patek, C. E., Williamson, J. & Arends, M. J. Mutant K-ras enhances apoptosis in embryonic stem cells in combination with DNA damage and is associated with increased levels of p19(ARF). Oncogene 20, 2144–2152 (2001).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We would like to thank G. Peters (Cancer Research UK London Research Institute, UK), D. Ginsberg (Bar Ilan University, Israel), P. G. Pelicci (European Institute of Oncology, Italy; University of Milan, Italy), M. Serrano (Spanish National Cancer Research Centre, Spain), Y. Shiloh (Tel Aviv University, Israel) and K. Vousden (Cancer Research-UK, Beatson Institute, UK) for providing reagents for this work. Special thanks for expert technical support from A. Damalas on the BJ cell treatments with serum starvation and T. Liloglou for manipulations with the HBECs and their derivatives. Financial support was from the European Commission FP7 (projects GENICA, INFLA-CARE, BioMedReg, DDResponse and INsPiRE), the Danish Cancer Society, and the Danish National Research Foundation. G.V. is a recipient of a Hellenic Association for Molecular Cancer Research scholarship. Dr A. Kotsinas is a recipient of an Empeirikeion Foundation fellowship. This work is dedicated to the memory of G. V. Gorgoulis.

Author information

Authors and Affiliations

Authors

Contributions

G.V. and M.L.: cell culture and manipulations, siRNA/plasmid/viral transfections/transductions/infections, ubiquitination assay, western blots, cell growth analyses and soft agar assays.

K.V., T.R. and V.G.G.: proteomic analysis design and experimentation.

E.K., J. Bartkova, I.S.P. and J. Bartek: human and mouse pathological evaluations, immunohistochemical analyses and evaluations, and tumour immunofluorescence analyses.

A.K.: real-time (RT-)PCR analyses.

M.S.: ChIP assay.

M.S. and G.V.: rRNA pulse-chase analysis, immunoprecipitation analyses, plasmid and lentiviral production.

A.D-O., M.K. and T.D.H.: I-2 mutagenesis analyses and corresponding immunofluorescence and western blot analyses.

I.O., S.B., M.C-B. and S.V.: nuclear fractionation, nucleolar immunofluorescence and western blot analyses, NPM site-directed mutagenesis and 47S pre-rRNA real-time RT–PCR analysis.

V.Z., D.K., G.P., A. Klinakis and V.G.G.: xenografts and mice manipulations.

K.V., M.L. and A. Kotsinas: Bioinformatic and statistical analyses.

W.G.: data analysis and production of reagents.

J. Bartek and T.D.H.: data analysis and interpretation, and assistance in manuscript preparation.

VG.: experimental design, guidance, manuscript preparation and writing.

Corresponding authors

Correspondence to Jiri Bartek or Vassilis G. Gorgoulis.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 HeLa and H1299 cells exhibit signs of DDR activation and detectable levels of ARF expression, but Chk2 silencing does not affect p14ARF protein levels.

a. Immunoblot (IB) analysis shows decreased phospho-Chk2 levels upon ATM inhibition (ATMi) in H1299 and HeLa cells, demonstrating the effectiveness of the inhibitor. b. IBs depicting signs of DDR activity, assessed by gH2AX (see also Fig. 1a), and detectable levels of p14ARF expression in HeLa and H1299 cells. c. IBs demonstrating that Chk2 silencing in HeLa and H1299 cells has no effect on p14ARF levels. p14ARF mRNA levels are induced by oncogenic stimuli (Cdk4, Ras and E2F1), but are unaltered after inhibition or silencing of ATM. Bars represent quantification of p14ARF mRNA levels as assessed by semi-quantitative real time RT–PCR in: (d) Human Bronchial Epithelial Cells (HBECS) (KT: hTERT, Cdk4 and KT transfected with activated K-RasV12), NARF cells (with or without IPTG induction) and Saos2-E2F1-ER (non-induced, TAM-induced and TAM-induced + ATMi), and in: (e) H1299, HeLa, HBECs cells transfected with control siRNA, siATM, DMSO (control) and Ku55933 (ATMi), induced NARF2 treated with Doxorubicin and Doxorubicin +Ku55933, respectively. (p<0.005, t-test, error bars indicate SDs, n = 4 real time RT–PCR runs). Silencing of ATM enhances oncogene-induced p14ARF expression in BJ cells and HBECs. f. IB analysis complementing Fig. 1c results showing the status of DDR activation and p14ARF levels in immortalized HBECs (hTERT, Cdk4) and immortalized HBECs with K-RasV12. Genetically manipulated HBECs expressing various oncogenes16,17,76 were employed as normal cells do not demonstrate signs of DDR activation47 and ARF levels are negligible1. g. IB analysis results showing that β-catenin transfected diploid BJ human fibroblasts exhibit DDR activation, while upregulation of p14ARF levels require additionally low serum conditions, as previously reported20. h. IB analyses, complementing panel g, demonstrating that the increased p14ARF protein levels in BJ primary human fibroblasts due to β-catenin transfection and serum depletion was further enhanced after silencing of ATM. ATM regulates p14ARF protein stability. i Silencing of ATM (shATM H1299 cells) protects ARF from DNA damage mediated downregulation. Oncogenic stimuli compete with active ATM in regulating ARF expression. j. IB analysis of p14ARF in H1299 cells infected with pBabe (control) or pBabe-Ha-RasV12 and in the presence or absence of ATM inhibitor Ku55933, showing that the oncogenic challenge of H1299 cell (that already harbour mutant and activated K-Ras) decreases the endogenous levels of ARF that were re-established when ATM was inhibited. Apparently, the oncogene-ARF pathway, in this setting, has reached an activation plateau and any additional oncogenic stimulus activates ATM leading to ARF suppression. b. Two signalling routes lead to ARF induction, oncogenic challenge1 and ATM suppression. Given that oncogenes activate ATM, as well4,7,47,50,60, oncogenic insults trigger two pathways with opposing effects on ARF expression. The outcome of this antagonism will depend on whether the rate of ARF production by oncogenes exceeds or not the rate of ARF destruction by the oncogene induced ATM pathway. Actin serves as loading control. ATMi = Ku55933 addition, PBGD = Porphobilinogen deaminase (house-keeping gene), Dox = Doxorubicin, ATMi = Ku55933 addition, ctrsi = control siRNA, TAM = 4-OH-Tamoxifen.

Supplementary Figure 2 Activation of ATM promotes the degradation of ARF by disrupting the ARF-NPM/B23 complex.

a. TRIP12/ULF protein levels remain unchanged after ATMi or Doxorubicin treatment in H1299 cells. Actin serves as loading control. ATMi = Ku55933 addition. b. IB analysis in siNPM/B23 H1299 cells showing downregulation of p14ARF denoting the significance of NPM/B23 in p14ARF stabilization. c. Nucleolar changes after exposure to ATM inhibition and Doxorubicine. H1299 cells were treated with doxorubicine (2uM) and the ATM inhibitor Ku55933 (10uM) for 24h. Untreated and treated cells were fixed and subjected to immunofluorescence staining with antibodies against the indicated nucleolar markers. Fibrillarin and UBF nucleolar cap structures are indicated by arrowheads. Fluorescence signals were analyzed by CLSM. Dox = Doxorubicin, ATMi = Ku55933 addition.

Supplementary Figure 3 ATM regulates PP1 activity through I-2.

a. IB analysis of I-2 in H1299 cells treated with DMSO, Ku55933 or Doxorubicin. pI-2 designates the slower migrating phosphorylated form of I-2. b. The phosphomimetic S43D mutation of I-2 abrogates its inhibitory effect over PP1. Immunoblot analysis for H3Ser10 in H1299 cells transfected with empty vector, wtI-2 or I-2S43D, which served as a positive control of increased PP1 activity. Histogram depicting quantification of H3Ser10 protein levels in H1299 cells transfected with the phosphomimetic S43D (p = NS and p<0.001 (S43D) respectively, t-test, error bars indicate SDs, n = 3 blots). c. Immunofluorescence analysis demonstrating absence of p14ARF staining in nuclei transfected with the phosphomimetic I-2S44D-GFP compared to I-2-GFP and I-2S44A-GFP mutant form in H1299 cells. Overexpression of wtI-2 or I-2S43A and I-2S43D mutants does not affect nucleolar integrity or NPM/B23 localization, as shown by immunofluorescence analysis for the nucleolar marker fibrillarin (d) and NPM/B23. (e) The efficiency of the transfection was examined with GFP. Nuclei were stained with DAPI. The efficiency of the transfection was examined with GFP. Dotted lines define nuclei. Actin and H3 serve as loading control. NS = non-significant, Dox = Doxorubicin, ATMi = Ku55933 addition, A.U. = arbitrary optical density units.

Supplementary Figure 4 Representative immunoblot showing the specificity of the sip14ARF as it does not affect p16INK4A expression.

Supplementary Figure 5 ATM-dependent p14ARF regulation affects ribosomal biogenesis.

a. Schematic presentation of the findings described in panels b and c. b. IBs depicting the localization over time of NPM/B23 and p14ARF in ATM compromised H1299 cells in total lysates, nucleoplasmic and nucleolar fractions. c. Modulation of p14ARF levels by ATMi affects TTF-I nucleoplasmic/nucleolar localization. IBs depicting the status of TTF-I and p14ARF in H1299 total cell lysates, nucleoplasmic and nucleolar fractions, respectively upon DMSO, ATMi and ATMi/siARF treatments.

Supplementary Figure 6 Gene Set enrichment analyses revealed the depicted ‘translation pathway’ to be among the most significantly affected ones (p<10−4) by ATM inhibition (ATMi). ATMi = Ku55933 addition.

Supplementary Figure 7 ATM abrogation is inversely related with ARF expression in human cancer.

a. Cumulative data (histograms) depicting the distribution of the lung cancer cases examined in the current work, according to the status of ATM, p-ATM, Chk2, p14ARF and p16INK4A66,67,68,69. b. Meta-analysis regarding alterations in the ATM gene and its protein product levels. Histograms represent the percentage of ATM alterations in the corresponding neoplasias. The red bar is the result of the current study. d. Phosphorylated I-2 PP1 regulatory subunit expression corroborates with ATM status in human lung carcinomas. Cases with normal ATM demonstrate by IB analysis increased expression of the phosphorylated I-2 PP1 regulatory subunit (pI-2). The reduced intensity of the slower migrating pI-2 band upon calf intestinal phosphatase (CIP) treatment of the blots denotes the specificity of the antibody used for the detection of the phosphorylated form of the I-2 subunit. In contrast, cases with low ATM levels do not show any levels of the pI-2 subunit14,30,33. Actin serves as loading control.

Supplementary Figure 8 ATM abrogation is inversely related with ARF expression in tumours generated from grafted H1299 cells in immuno-compromised mice.

a. Validation of lenti-shATM silencing sequences (see Suppl. Tables S4) in H1299 cells showing down-regulation of ATM followed by increased expression of ARF. Actin serves as loading control. b Successful transduction of H1299 cells with lenti-GFP. Scale bar: 25 μm. c. Successful delivery and expression of GFP after injecting H1299-mock xenografts with lenti-GFP. Nuclei were stained with DAPI. Scale bar: 50 μm. d. Timetable of injections containing either pLKO.1 lenti-shATM or pLKO.1 lenti-non-target shRNA to H1299-mock and H1299-shARF xenografts. e. Tumours generated from grafted H1299 cells in immuno-compromised mice exhibited significantly reduced size after suppressing ATM expression for a period of 4 weeks (see also panels f-h) by injecting the tumours with lenti-shATM. The tumours remained unaffected when shATM was administrated in H1299-shARF xenografts. f. Tumours generated from grafted H1299 cells in immuno-compromised mice exhibited significantly reduced size after inhibition of ATM activity (ATMi), but remained unaffected in size when ATM inhibitor (ATMi) was administrated in H1299-shARF xenografts. Hematoxylin-eosin sections from the developed tumours (yellow arrow depicts invasion of abdominal wall muscle layer). Scale bar: 200 μm. IB analysis depicting the p14ARF protein levels in the control (EV), ATMi/EV and ATMi/shARF tumours. g. There was no significant difference in mass between tumours developed from H1299-shARF injected cells, with or without ATMi administration. h. Histogram presents the average mass between the groups of generated tumours (p = 0.007 and p = NS respectively, t-test, error bars indicate SDs, from 3 groups of 4 animals/group). Dependence of ARF expression upon ATM status in MEFs and mouse testis. i IBs showing that ATM null mouse embryo fibroblasts (MEFs) demonstrate higher levels of ARF compared to ATM+/+ MEFs, which remain unaffected upon irradiation, while in irradiated ATM+/+ MEFs ARF expression is lost. j. IHC staining demonstrating loss of ARF expression in mouse seminiferous tubules after irradiation (Scale bar: 50 μm). Histogram depicts p19ARF mRNA levels in mouse testis before and after irradiation (p = NS, t-test, error bars indicate SDs, from 3 animals/group). EV = empty vector, ATMi = caffeine addition, IR = irradiation, NS = non-significant.

Supplementary Figure 9 Full scans of blots.

Supplementary Table 1 Table 3: Studies examining genetic and epigenetic alterations in ATM and its mRNA and protein expression status in several sporadic malignancies.
Supplementary Table 2 Table 4: Lentiviral vectors (MISSION) and inserts used from Sigma-Aldrich.
Supplementary Table 3 Table 5: List of antibodies employed in immunohistochemistry, immunofluorescence, immunoblotting, immunoprecipitation and chromatin immunoprecipitation analyses.
Supplementary Table 4 Table 6: Primers for mutagenesis and sequencing.
Supplementary Table 5 Table 7: Datasets accession numbers for proteomic results deposited in PRIDE.

Supplementary information

Supplementary Information

Supplementary Information (PDF 2154 kb)

Supplementary Table 1

Analytical presentation of all peptides and their modification recognized by the LC-MS/MS analysis from NPM-immunoprecipitated H1299. (see full table organized in accompanying excel file). (XLSX 235 kb)

Supplementary Table 2

Peptides (a), proteins (b) and filtered proteins (c) presenting high degree of reproducibility. Section (c) also contains p-values indicative of differential expression with confidence being set to 95%. (see full table organized in accompanying excel file). (XLSX 4173 kb)

Supplementary Table 3

Studies examining genetic and epigenetic alterations in ATM and its mRNA and protein expression status in several sporadic malignancies. (XLSX 16 kb)

Supplementary Table 4

Lentiviral vectors (MISSION®) and inserts used from Sigma-Aldrich. (XLSX 11 kb)

Supplementary Table 5

List of antibodies employed in immunohistochemistry, immunofluorescence, immunoblotting, immunoprecipitation and chromatin immunoprecipitation analyses. (XLSX 14 kb)

Supplementary Table 6

Primers for mutagenesis and sequencing. (XLSX 11 kb)

Supplementary Table 7

Datasets accession numbers for proteomic results deposited in PRIDE. (XLSX 11 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Velimezi, G., Liontos, M., Vougas, K. et al. Functional interplay between the DNA-damage-response kinase ATM and ARF tumour suppressor protein in human cancer. Nat Cell Biol 15, 967–977 (2013). https://doi.org/10.1038/ncb2795

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb2795

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