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.

  • Original Article
  • Published:

Cellular senescence checkpoint function determines differential Notch1-dependent oncogenic and tumor-suppressor activities

Abstract

Notch activity regulates tumor biology in a context-dependent and complex manner. Notch may act as an oncogene or a tumor-suppressor gene even within the same tumor type. Recently, Notch signaling has been implicated in cellular senescence. Yet, it remains unclear as to how cellular senescence checkpoint functions may interact with Notch-mediated oncogenic and tumor-suppressor activities. Herein, we used genetically engineered human esophageal keratinocytes and esophageal squamous cell carcinoma cells to delineate the functional consequences of Notch activation and inhibition along with pharmacological intervention and RNA interference experiments. When expressed in a tetracycline-inducible manner, the ectopically expressed activated form of Notch1 (ICN1) displayed oncogene-like characteristics inducing cellular senescence corroborated by the induction of G0/G1 cell-cycle arrest, Rb dephosphorylation, flat and enlarged cell morphology and senescence-associated β-galactosidase activity. Notch-induced senescence involves canonical CSL/RBPJ-dependent transcriptional activity and the p16INK4A-Rb pathway. Loss of p16INK4A or the presence of human papilloma virus (HPV) E6/E7 oncogene products not only prevented ICN1 from inducing senescence but permitted ICN1 to facilitate anchorage-independent colony formation and xenograft tumor growth with increased cell proliferation and reduced squamous-cell differentiation. Moreover, Notch1 appears to mediate replicative senescence as well as transforming growth factor-β-induced cellular senescence in non-transformed cells and that HPV E6/E7 targets Notch1 for inactivation to prevent senescence, revealing a tumor-suppressor attribute of endogenous Notch1. In aggregate, cellular senescence checkpoint functions may influence dichotomous Notch activities in the neoplastic context.

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
Figure 2
Figure 3
Figure 4
Figure 5
Figure 6
Figure 7
Figure 8

Similar content being viewed by others

References

  1. Enzinger PC, Mayer RJ . Esophageal cancer. New Engl J Med 2003; 349: 2241–2252.

    Article  CAS  Google Scholar 

  2. Nakagawa H, Katzka D, Rustgi AK . Biology of esophageal cancer. In: Rustgi AK (ed). Gastrointestinal Cancers. Elsevier: London, UK, 2003, pp 241–251.

    Google Scholar 

  3. Harada H, Nakagawa H, Oyama K, Takaoka M, Andl CD, Jacobmeier B et al. Telomerase induces immortalization of human esophageal keratinocytes without p16INK4a inactivation. Mol Cancer Res 2003; 1: 729–738.

    CAS  Google Scholar 

  4. Sashiyama H, Shino Y, Kawamata Y, Tomita Y, Ogawa N, Shimada H et al. Immortalization of human esophageal keratinocytes by E6 and E7 of human papillomavirus type 16. Int J Oncol 2001; 19: 97–103.

    CAS  Google Scholar 

  5. Takaoka M, Harada H, Deramaudt TB, Oyama K, Andl CD, Johnstone CN et al. Ha-Ras(G12V) induces senescence in primary and immortalized human esophageal keratinocytes with p53 dysfunction. Oncogene 2004; 23: 6760–6768.

    Article  CAS  Google Scholar 

  6. Oyama K, Okawa T, Nakagawa H, Takaoka M, Andl CD, Kim SH et al. AKT induces senescence in primary esophageal epithelial cells but is permissive for differentiation as revealed in organotypic culture. Oncogene 2007; 26: 2353–2364.

    Article  CAS  Google Scholar 

  7. Ohashi S, Natsuizaka M, Wong GS, Michaylira CZ, Grugan KD, Stairs DB et al. Epidermal growth factor receptor and mutant p53 expand an esophageal cellular subpopulation capable of epithelial-to-mesenchymal transition through ZEB transcription factors. Cancer Res 2010; 70: 4174–4184.

    Article  CAS  Google Scholar 

  8. Okawa T, Michaylira CZ, Kalabis J, Stairs DB, Nakagawa H, Andl CD et al. The functional interplay between EGFR overexpression, hTERT activation, and p53 mutation in esophageal epithelial cells with activation of stromal fibroblasts induces tumor development, invasion, and differentiation. Genes Dev 2007; 21: 2788–2803.

    Article  CAS  Google Scholar 

  9. Kim SH, Nakagawa H, Navaraj A, Naomoto Y, Klein-Szanto AJ, Rustgi AK et al. Tumorigenic conversion of primary human esophageal epithelial cells using oncogene combinations in the absence of exogenous Ras. Cancer Res 2006; 66: 10415–10424.

    Article  CAS  Google Scholar 

  10. McElhinny AS, Li JL, Wu L . Mastermind-like transcriptional co-activators: emerging roles in regulating cross talk among multiple signaling pathways. Oncogene 2008; 27: 5138–5147.

    Article  CAS  Google Scholar 

  11. Blanpain C, Lowry WE, Pasolli HA, Fuchs E . Canonical notch signaling functions as a commitment switch in the epidermal lineage. Genes Dev 2006; 20: 3022–3035.

    Article  CAS  Google Scholar 

  12. Rangarajan A, Talora C, Okuyama R, Nicolas M, Mammucari C, Oh H et al. Notch signaling is a direct determinant of keratinocyte growth arrest and entry into differentiation. EMBO J 2001; 20: 3427–3436.

    Article  CAS  Google Scholar 

  13. Ohashi S, Natsuizaka M, Yashiro-Ohtani Y, Kalman RA, Nakagawa M, Wu L et al. NOTCH1 and NOTCH3 coordinate esophageal squamous differentiation through a CSL-dependent transcriptional network. Gastroenterology 2010; 139: 2113–2123.

    Article  CAS  Google Scholar 

  14. Ranganathan P, Weaver KL, Capobianco AJ . Notch signalling in solid tumours: a little bit of everything but not all the time. Nat Rev Cancer 2011; 11: 338–351.

    Article  CAS  Google Scholar 

  15. Hijioka H, Setoguchi T, Miyawaki A, Gao H, Ishida T, Komiya S et al. Upregulation of Notch pathway molecules in oral squamous cell carcinoma. Int J Oncol 2010; 36: 817–822.

    CAS  Google Scholar 

  16. Zagouras P, Stifani S, Blaumueller CM, Carcangiu ML, Artavanis-Tsakonas S . Alterations in Notch signaling in neoplastic lesions of the human cervix. Proc Natl Acad Sci USA 1995; 92: 6414–6418.

    Article  CAS  Google Scholar 

  17. Rangarajan A, Syal R, Selvarajah S, Chakrabarti O, Sarin A, Krishna S . Activated Notch1 signaling cooperates with papillomavirus oncogenes in transformation and generates resistance to apoptosis on matrix withdrawal through PKB/Akt. Virology 2001; 286: 23–30.

    Article  CAS  Google Scholar 

  18. Weijzen S, Zlobin A, Braid M, Miele L, Kast WM . HPV16 E6 and E7 oncoproteins regulate Notch-1 expression and cooperate to induce transformation. J Cell Physiol 2003; 194: 356–362.

    Article  CAS  Google Scholar 

  19. Talora C, Sgroi DC, Crum CP, Dotto GP . Specific down-modulation of Notch1 signaling in cervical cancer cells is required for sustained HPV-E6/E7 expression and late steps of malignant transformation. Genes Dev 2002; 16: 2252–2263.

    Article  CAS  Google Scholar 

  20. Agrawal N, Frederick MJ, Pickering CR, Bettegowda C, Chang K, Li RJ et al. Exome sequencing of head and neck squamous cell carcinoma reveals inactivating mutations in NOTCH1. Science 2011; 333: 1154–1157.

    Article  CAS  Google Scholar 

  21. Stransky N, Egloff AM, Tward AD, Kostic AD, Cibulskis K, Sivachenko A et al. The mutational landscape of head and neck squamous cell carcinoma. Science 2011; 333: 1157–1160.

    Article  CAS  Google Scholar 

  22. Wang NJ, Sanborn Z, Arnett KL, Bayston LJ, Liao W, Proby CM et al. Loss-of-function mutations in Notch receptors in cutaneous and lung squamous cell carcinoma. Proc Natl Acad Sci USA 2011; 108: 17761–17766.

    Article  CAS  Google Scholar 

  23. Agrawal N, Jiao Y, Bettegowda C, Hutfless SM, Wang Y, David S et al. Comparative genomic analysis of esophageal adenocarcinoma and squamous cell carcinoma. Cancer Discov 2012; 2: 899–905.

    Article  CAS  Google Scholar 

  24. Lefort K, Mandinova A, Ostano P, Kolev V, Calpini V, Kolfschoten I et al. Notch1 is a p53 target gene involved in human keratinocyte tumor suppression through negative regulation of ROCK1/2 and MRCKalpha kinases. Genes Dev 2007; 21: 562–577.

    Article  CAS  Google Scholar 

  25. Li T, Wen H, Brayton C, Laird FM, Ma G, Peng S et al. Moderate reduction of gamma-secretase attenuates amyloid burden and limits mechanism-based liabilities. J Neurosci 2007; 27: 10849–10859.

    Article  CAS  Google Scholar 

  26. Nicolas M, Wolfer A, Raj K, Kummer JA, Mill P, van Noort M et al. Notch1 functions as a tumor suppressor in mouse skin. Nat Genet 2003; 33: 416–421.

    Article  CAS  Google Scholar 

  27. Pan Y, Lin MH, Tian X, Cheng HT, Gridley T, Shen J et al. gamma-secretase functions through Notch signaling to maintain skin appendages but is not required for their patterning or initial morphogenesis. Dev Cell 2004; 7: 731–743.

    Article  CAS  Google Scholar 

  28. Proweller A, Tu L, Lepore JJ, Cheng L, Lu MM, Seykora J et al. Impaired notch signaling promotes de novo squamous cell carcinoma formation. Cancer Res 2006; 66: 7438–7444.

    Article  CAS  Google Scholar 

  29. Zhang YW, Wang R, Liu Q, Zhang H, Liao FF, Xu H . Presenilin/gamma-secretase-dependent processing of beta-amyloid precursor protein regulates EGF receptor expression. Proc Natl Acad Sci USA 2007; 104: 10613–10618.

    Article  CAS  Google Scholar 

  30. Demehri S, Turkoz A, Kopan R . Epidermal Notch1 loss promotes skin tumorigenesis by impacting the stromal microenvironment. Cancer Cell 2009; 16: 55–66.

    Article  CAS  Google Scholar 

  31. Venkatesh D, Fredette N, Rostama B, Tang Y, Vary CP, Liaw L et al. RhoA-mediated signaling in Notch-induced senescence-like growth arrest and endothelial barrier dysfunction. Arterioscler Thromb Vasc Biol 2011; 31: 876–882.

    Article  CAS  Google Scholar 

  32. Liu ZJ, Tan Y, Beecham GW, Seo DM, Tian R, Li Y et al. Notch activation induces endothelial cell senescence and pro-inflammatory response: implication of Notch signaling in atherosclerosis. Atherosclerosis 2012; 225: 296–303.

    Article  CAS  Google Scholar 

  33. Lowell S, Jones P, Le Roux I, Dunne J, Watt FM . Stimulation of human epidermal differentiation by delta-notch signalling at the boundaries of stem-cell clusters. Curr Biol 2000; 10: 491–500.

    Article  CAS  Google Scholar 

  34. Dickson MA, Hahn WC, Ino Y, Ronfard V, Wu JY, Weinberg RA et al. Human keratinocytes that express hTERT and also bypass a p16(INK4a)-enforced mechanism that limits life span become immortal yet retain normal growth and differentiation characteristics. Mol Cell Biol 2000; 20: 1436–1447.

    Article  CAS  Google Scholar 

  35. Ohashi S, Natsuizaka M, Naganuma S, Kagawa S, Kimura S, Itoh H et al. A NOTCH3-mediated squamous cell differentiation program limits expansion of EMT-competent cells that express the ZEB transcription factors. Cancer Res 2011; 71: 6836–6847.

    Article  CAS  Google Scholar 

  36. Jia LQ, Osada M, Ishioka C, Gamo M, Ikawa S, Suzuki T et al. Screening the p53 status of human cell lines using a yeast functional assay. Mol Carcinog 1997; 19: 243–253.

    Article  CAS  Google Scholar 

  37. Liu Q, Yan YX, McClure M, Nakagawa H, Fujimura F, Rustgi AK . MTS-1 (CDKN2) tumor suppressor gene deletions are a frequent event in esophagus squamous cancer and pancreatic adenocarcinoma cell lines. Oncogene 1995; 10: 619–622.

    CAS  Google Scholar 

  38. Dotto GP . Crosstalk of Notch with p53 and p63 in cancer growth control. Nat Rev Cancer 2009; 9: 587–595.

    Article  CAS  Google Scholar 

  39. Bertwistle D, Sugimoto M, Sherr CJ . Physical and functional interactions of the Arf tumor suppressor protein with nucleophosmin/B23. Mol Cell Biol 2004; 24: 985–996.

    Article  CAS  Google Scholar 

  40. Ovcharenko I, Nobrega MA, Loots GG, Stubbs L . ECR Browser: a tool for visualizing and accessing data from comparisons of multiple vertebrate genomes. Nucleic Acids Res 2004; 32: W280–W286.

    Article  CAS  Google Scholar 

  41. Lee DK, Kim BC, Kim IY, Cho EA, Satterwhite DJ, Kim SJ . The human papilloma virus E7 oncoprotein inhibits transforming growth factor-beta signaling by blocking binding of the Smad complex to its target sequence. J Biol Chem 2002; 277: 38557–38564.

    Article  CAS  Google Scholar 

  42. Zavadil J, Cermak L, Soto-Nieves N, Bottinger EP . Integration of TGF-beta/Smad and Jagged1/Notch signalling in epithelial-to-mesenchymal transition. EMBO J 2004; 23: 1155–1165.

    Article  CAS  Google Scholar 

  43. Bierie B, Moses HL . Tumour microenvironment: TGFbeta: the molecular Jekyll and Hyde of cancer. Nat Rev Cancer 2006; 6: 506–520.

    Article  CAS  Google Scholar 

  44. Natsugoe S, Xiangming C, Matsumoto M, Okumura H, Nakashima S, Sakita H et al. Smad4 and transforming growth factor beta1 expression in patients with squamous cell carcinoma of the esophagus. Clin Cancer Res 2002; 8: 1838–1842.

    CAS  Google Scholar 

  45. Natsuizaka M, Ohashi S, Wong GS, Ahmadi A, Kalman RA, Budo D et al. Insulin-like growth factor-binding protein-3 promotes transforming growth factor-{beta}1-mediated epithelial-to-mesenchymal transition and motility in transformed human esophageal cells. Carcinogenesis 2010; 31: 1344–1353.

    Article  CAS  Google Scholar 

  46. Natsuizaka M, Naganuma S, Kagawa S, Ohashi S, Ahmadi A, Subramanian H et al. Hypoxia induces IGFBP3 in esophageal squamous cancer cells through HIF-1alpha-mediated mRNA transcription and continuous protein synthesis. FASEB J 2012; 26: 2620–2630.

    Article  CAS  Google Scholar 

  47. Gustafsson MV, Zheng X, Pereira T, Gradin K, Jin S, Lundkvist J et al. Hypoxia requires notch signaling to maintain the undifferentiated cell state. Dev Cell 2005; 9: 617–628.

    Article  CAS  Google Scholar 

  48. Cui H, Kong Y, Xu M, Zhang H . Notch3 functions as a tumor suppressor by controlling cellular senescence. Cancer Res 2013; 73: 3451–3459.

    Article  CAS  Google Scholar 

  49. Yugawa T, Handa K, Narisawa-Saito M, Ohno S, Fujita M, Kiyono T . Regulation of Notch1 gene expression by p53 in epithelial cells. Mol Cell Biol 2007; 27: 3732–3742.

    Article  CAS  Google Scholar 

  50. Pascal T, Debacq-Chainiaux F, Chretien A, Bastin C, Dabee AF, Bertholet V et al. Comparison of replicative senescence and stress-induced premature senescence combining differential display and low-density DNA arrays. FEBS Lett 2005; 579: 3651–3659.

    Article  CAS  Google Scholar 

  51. Tremain R, Marko M, Kinnimulki V, Ueno H, Bottinger E, Glick A . Defects in TGF-beta signaling overcome senescence of mouse keratinocytes expressing v-Ha-ras. Oncogene 2000; 19: 1698–1709.

    Article  CAS  Google Scholar 

  52. Lin S, Yang J, Elkahloun AG, Bandyopadhyay A, Wang L, Cornell JE et al. Attenuation of TGF-beta signaling suppresses premature senescence in a p21-dependent manner and promotes oncogenic Ras-mediated metastatic transformation in human mammary epithelial cells. Mol Biol Cell 2012; 23: 1569–1581.

    Article  CAS  Google Scholar 

  53. Kolev V, Mandinova A, Guinea-Viniegra J, Hu B, Lefort K, Lambertini C et al. EGFR signalling as a negative regulator of Notch1 gene transcription and function in proliferating keratinocytes and cancer. Nat Cell Biol 2008; 10: 902–911.

    Article  CAS  Google Scholar 

  54. Kuilman T, Michaloglou C, Mooi WJ, Peeper DS . The essence of senescence. Genes Dev 2010; 24: 2463–2479.

    Article  CAS  Google Scholar 

  55. Thiery JP, Acloque H, Huang RY, Nieto MA . Epithelial-mesenchymal transitions in development and disease. Cell 2009; 139: 871–890.

    Article  CAS  Google Scholar 

  56. Ansieau S, Bastid J, Doreau A, Morel AP, Bouchet BP, Thomas C et al. Induction of EMT by twist proteins as a collateral effect of tumor-promoting inactivation of premature senescence. Cancer Cell 2008; 14: 79–89.

    Article  CAS  Google Scholar 

  57. Pickering CR, Zhang J, Yoo SY, Bengtsson L, Moorthy S, Neskey DM et al. Integrative genomic characterization of oral squamous cell carcinoma identifies frequent somatic drivers. Cancer Discov 2013; 3: 770–781.

    Article  CAS  Google Scholar 

  58. Ling H, Sylvestre JR, Jolicoeur P . Cyclin D1-dependent induction of luminal inflammatory breast tumors by activated Notch3. Cancer Res 2013; 73: 5963–5973.

    Article  CAS  Google Scholar 

  59. Aster JC, Xu L, Karnell FG, Patriub V, Pui JC, Pear WS . Essential roles for ankyrin repeat and transactivation domains in induction of T-cell leukemia by notch1. Mol Cell Biol 2000; 20: 7505–7515.

    Article  CAS  Google Scholar 

  60. Jeffries S, Capobianco AJ . Neoplastic transformation by Notch requires nuclear localization. Mol Cell Biol 2000; 20: 3928–3941.

    Article  CAS  Google Scholar 

  61. Wrana JL, Attisano L, Carcamo J, Zentella A, Doody J, Laiho M et al. TGF beta signals through a heteromeric protein kinase receptor complex. Cell 1992; 71: 1003–1014.

    Article  CAS  Google Scholar 

  62. Mroz EA, Baird AH, Michaud WA, Rocco JW . COOH-terminal binding protein regulates expression of the p16INK4A tumor suppressor and senescence in primary human cells. Cancer Res 2008; 68: 6049–6053.

    Article  CAS  Google Scholar 

  63. Maruo S, Zhao B, Johannsen E, Kieff E, Zou J, Takada K . Epstein-Barr virus nuclear antigens 3C and 3A maintain lymphoblastoid cell growth by repressing p16INK4A and p14ARF expression. Proc Natl Acad Sci USA 2011; 108: 1919–1924.

    Article  CAS  Google Scholar 

  64. Cordenonsi M, Dupont S, Maretto S, Insinga A, Imbriano C, Piccolo S . Links between tumor suppressors: p53 is required for TGF-beta gene responses by cooperating with Smads. Cell 2003; 113: 301–314.

    Article  CAS  Google Scholar 

  65. Young AR, Narita M, Ferreira M, Kirschner K, Sadaie M, Darot JF et al. Autophagy mediates the mitotic senescence transition. Genes Dev 2009; 23: 798–803.

    Article  CAS  Google Scholar 

  66. Lee BY, Han JA, Im JS, Morrone A, Johung K, Goodwin EC et al. Senescence-associated beta-galactosidase is lysosomal beta-galactosidase. Aging Cell 2006; 5: 187–195.

    Article  CAS  Google Scholar 

  67. Kurz DJ, Decary S, Hong Y, Erusalimsky JD . Senescence-associated (beta)-galactosidase reflects an increase in lysosomal mass during replicative ageing of human endothelial cells. J Cell Sci 2000; 113 (Pt 20): 3613–3622.

    CAS  Google Scholar 

Download references

Acknowledgements

We thank Dr Hiroshi Shirasawa (Chiba University, Chiba, Japan) for the gift of EN60 cells. We are grateful to the Molecular Pathology & Imaging, Molecular Biology/Gene Expression and Cell Culture Core Facilities of the NIH/NIDDK Center for Molecular Studies in Digestive and Liver Diseases (P30-DK050306) and of the NIH P01CA098101. This study was supported in part by NIH Grants P01CA098101 (to SK, MN, KAW, DB, HK, SN, SO, PAG, AJK-S, AB, K-KW, JAD, HN and AKR), U01CA143056 (to AKR), R01DK077005 (to HN), K26 RR032714 (to HN), Pennsylvania CURE Program Grant (to HN), F32-CA174176 (to KAW), F32-DE024685 to (NF), K08DE022842, Trio/ACS Career Award and VA CPPF Grant (to DB), K07CA137140 (to AME), University of Pennsylvania University Research Foundation Award (to HN), University of Pennsylvania, Abramson Cancer Center Pilot Project Grant (to HN), and the American Cancer Society RP-10-033-01-CCE (to AKR).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to H Nakagawa.

Ethics declarations

Competing interests

The authors declare no conflict of interest.

Additional information

Supplementary Information accompanies this paper on the Oncogene website

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kagawa, S., Natsuizaka, M., Whelan, K. et al. Cellular senescence checkpoint function determines differential Notch1-dependent oncogenic and tumor-suppressor activities. Oncogene 34, 2347–2359 (2015). https://doi.org/10.1038/onc.2014.169

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/onc.2014.169

This article is cited by

Search

Quick links