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.

  • Short Communication
  • Published:

DNA damage-induced sustained p53 activation contributes to inflammation-associated hepatocarcinogenesis in rats

Abstract

The tumor suppressor p53 has an important role in inducing cell-intrinsic responses to DNA damage, including cellular senescence or apoptosis, which act to thwart tumor development. It has been shown, however, that senescent or dying cells are capable of eliciting inflammatory responses, which can have pro-tumorigenic effects. Whether DNA damage-induced p53 activity can contribute to senescence- or apoptosis-associated pro-tumorigenic inflammation is unknown. Recently, we generated a p53 knock-out rat via homologous recombination in rat embryonic stem cells. Here we show that in a rat model of inflammation-associated hepatocarcinogenesis, heterozygous deficiency of p53 resulted in attenuated inflammatory responses and ameliorated hepatic cirrhosis and tumorigenesis. Chronic administration of hepatocarcinogenic compound, diethylnitrosamine, led to persistent DNA damage and sustained induction of p53 protein in the wild-type livers, and much less induction in p53 heterozygous livers. Sustained p53 activation subsequent to DNA damage was accompanied by apoptotic rather than senescent hepatic injury, which gave rise to the hepatic inflammatory responses. In contrast, the non-hepatocarcinogenic agent, carbon tetrachloride, failed to induce p53, and caused a similar degree of chronic hepatic inflammation and cirrhosis in wild type and p53 heterozygous rats. These results suggest that although p53 is usually regarded as a tumor suppressor, its constant activation can promote pro-tumorigenic inflammation, especially in livers exposed to agents that inflict lasting mutagenic DNA damage.

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

Similar content being viewed by others

References

  1. Hanahan D, Weinberg RA . Hallmarks of cancer: the next generation. Cell 2011; 144: 646–674.

    Article  CAS  PubMed  Google Scholar 

  2. Thorgeirsson SS, Grisham JW . Molecular pathogenesis of human hepatocellular carcinoma. Nat Genet 2002; 31: 339–346.

    Article  CAS  PubMed  Google Scholar 

  3. Kang TW, Yevsa T, Woller N, Hoenicke L, Wuestefeld T, Dauch D et al. Senescence surveillance of pre-malignant hepatocytes limits liver cancer development. Nature 2011; 479: 547–551.

    Article  CAS  PubMed  Google Scholar 

  4. Fausto N . Mouse liver tumorigenesis: models, mechanisms, and relevance to human disease. Semin Liver Dis 1999; 19: 243–252.

    Article  CAS  PubMed  Google Scholar 

  5. Levine AJ, Oren M . The first 30 years of p53: growing ever more complex. Nat Rev Cancer 2009; 9: 749–758.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Livni N, Eid A, Ilan Y, Rivkind A, Rosenmann E, Blendis LM et al. p53 expression in patients with cirrhosis with and without hepatocellular carcinoma. Cancer 1995; 75: 2420–2426.

    Article  CAS  PubMed  Google Scholar 

  7. Sarfraz S, Hamid S, Siddiqui A, Hussain S, Pervez S, Alexander G . Altered expression of cell cycle and apoptotic proteins in chronic hepatitis C virus infection. BMC Microbiol 2008; 8: 133.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Greenblatt MS, Feitelson MA, Zhu M, Bennett WP, Welsh JA, Jones R et al. Integrity of p53 in hepatitis B x antigen-positive and -negative hepatocellular carcinomas. Cancer Res 1997; 57: 426–432.

    CAS  PubMed  Google Scholar 

  9. Kodama T, Takehara T, Hikita H, Shimizu S, Shigekawa M, Tsunematsu H et al. Increases in p53 expression induce CTGF synthesis by mouse and human hepatocytes and result in liver fibrosis in mice. J Clin Invest 2011; 121: 3343–3356.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Krizhanovsky V, Yon M, Dickins RA, Hearn S, Simon J, Miething C et al. Senescence of activated stellate cells limits liver fibrosis. Cell 2008; 134: 657–667.

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Abbott A . Laboratory animals: the Renaissance rat. Nature 2004; 428: 464–466.

    Article  CAS  PubMed  Google Scholar 

  12. Tong C, Li P, Wu NL, Yan Y, Ying QL . Production of p53 gene knockout rats by homologous recombination in embryonic stem cells. Nature 2010; 467: 211–213.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Rajewsky MF, Dauber W, Frankenberg H . Liver carcinogenesis by diethylnitrosamine in the rat. Science 1966; 152: 83–85.

    Article  CAS  PubMed  Google Scholar 

  14. Verna L, Whysner J, Williams GM . N-nitrosodiethylamine mechanistic data and risk assessment: bioactivation, DNA-adduct formation, mutagenicity, and tumor initiation. Pharmacol Ther 1996; 71: 57–81.

    Article  CAS  PubMed  Google Scholar 

  15. Polyak K, Xia Y, Zweier JL, Kinzler KW, Vogelstein B . A model for p53-induced apoptosis. Nature 1997; 389: 300–305.

    Article  CAS  PubMed  Google Scholar 

  16. Farazi PA, DePinho RA . Hepatocellular carcinoma pathogenesis: from genes to environment. Nat Rev Cancer 2006; 6: 674–687.

    Article  CAS  PubMed  Google Scholar 

  17. Newell P, Villanueva A, Friedman SL, Koike K, Llovet JM . Experimental models of hepatocellular carcinoma. J Hepatol 2008; 48: 858–879.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Kemp CJ . Hepatocarcinogenesis in p53-deficient mice. Mol Carcinog 1995; 12: 132–136.

    Article  CAS  PubMed  Google Scholar 

  19. Kuraishy A, Karin M, Grivennikov SI . Tumor promotion via injury- and death-induced inflammation. Immunity 2011; 35: 467–477.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Kemp CJ, Donehower LA, Bradley A, Balmain A . Reduction of p53 gene dosage does not increase initiation or promotion but enhances malignant progression of chemically induced skin tumors. Cell 1993; 74: 813–822.

    Article  CAS  PubMed  Google Scholar 

  21. van Gijssel HE, Maassen CB, Mulder GJ, Meerman JH . p53 protein expression by hepatocarcinogens in the rat liver and its potential role in mitoinhibition of normal hepatocytes as a mechanism of hepatic tumour promotion. Carcinogenesis 1997; 18: 1027–1033.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank Jiaohong Wang, Shoudong Ye and Zhengmao Lu for technical assistance. This work was supported by a NIH grant to QLY (R01OD010926).

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to H-X Yan or Q-L Ying.

Ethics declarations

Competing interests

The authors declare no conflict of interest.

Additional information

Supplementary Information accompanies the paper on the Oncogene website

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Cite this article

Yan, HX., Wu, HP., Zhang, HL. et al. DNA damage-induced sustained p53 activation contributes to inflammation-associated hepatocarcinogenesis in rats. Oncogene 32, 4565–4571 (2013). https://doi.org/10.1038/onc.2012.451

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/onc.2012.451

Keywords

This article is cited by

Search

Quick links