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.

  • Letter
  • Published:

Nf1;Trp53 mutant mice develop glioblastoma with evidence of strain-specific effects

A Correction to this article was published on 01 January 2001

Abstract

Astrocytomas are the leading cause of brain cancer in humans. Because these tumours are highly infiltrative, current treatments that rely on targeting the tumour mass are often ineffective. A mouse model for astrocytoma would be a powerful tool for dissecting tumour progression and testing therapeutics. Mouse models of astrocytoma have been designed to express oncogenic proteins in astrocytes, but have had limited success due to low tumour penetrance or limited tumour progression1,2,3. We present here a mouse model of astrocytomas involving mutation of two tumour-suppressor genes, Nf1 and Trp53. Humans with mutations in NF1 develop neurofibromatosis type I (NF1) and have increased risk of optic gliomas, astrocytomas and glioblastomas4,5. The TP53 tumour suppressor is often mutated in a subset of astrocytomas that develop at a young age and progress slowly to glioblastoma (termed secondary glioblastomas, in contrast to primary glioblastomas that develop rapidly de novo6,7,8,9,10). This mouse model shows a range of astrocytoma stages, from low-grade astrocytoma to glioblastoma multiforme, and may accurately model human secondary glioblastoma involving TP53 loss. This is the first reported mouse model of astrocytoma initiated by loss of tumour suppressors, rather than overexpression of transgenic oncogenes.

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: Histology of astrocytomas in NPcis mice.
Figure 2: Glioblastoma multiforme in an NPcis mouse on a (H×B6)F1 background. This figure shows a tumour in the thalamus of an NPcis mouse with the criteria of a human glioblastoma multiforme.
Figure 3: Immunohistochemistry of astrocytomas in NPcis mice.
Figure 4: Astrocytomas show loss of the wild-type alleles of Nf1 and Trp53.
Figure 5: Tissue culture of astrocytomas.
Figure 6: Incidence of brain tumours in NPcis mice.

Similar content being viewed by others

References

  1. Danks, R. et al. Transformation of astrocytes in transgenic mice expressing SV40 T antigen under the transcriptional control of the glial fibrillary acidic protein promoter. Cancer Res. 55, 4302–4310 (1995).

    CAS  PubMed  Google Scholar 

  2. Holland, E., Hively, W., DePinho, R. & Varmus, H. A constitutively active epidermal growth factor receptor cooperates with disruption of G1 cell-cycle arrest pathways to induce glioma-like lesions in mice. Genes Dev. 12, 3675–3685 (1998).

    Article  CAS  Google Scholar 

  3. Weissenberger, J. et al. Development and malignant progression of astrocytomas in GFAP-v-src transgenic mice. Oncogene 14, 2005–2013 (1997).

    Article  CAS  Google Scholar 

  4. Huson, S. & Hughes, R. The Neurofibromatoses: A Pathogenetic and Clinical Overview (Chapman & Hall Medical, London, 1994).

    Google Scholar 

  5. Sorensen, S.A., Mulvihill, J.J. & Nielsen, A. Long-term follow-up of von Recklinghausen neurofibromatosis. Survival and malignant neoplasms. N. Engl. J. Med. 314, 1010–1015 (1986).

    Article  CAS  Google Scholar 

  6. Watanabe, K. et al. Incidence and timing of p53 mutations during astrocytoma progression in patients with multiple biopsies. Clin. Cancer Res. 3, 523–530 (1997).

    CAS  PubMed  Google Scholar 

  7. Watanabe, K. et al. Overexpression of the EGF receptor and p53 mutations are mutually exclusive in the evolution of primary and secondary glioblastomas. Brain Pathol. 6, 217–223 (1996).

    Article  CAS  Google Scholar 

  8. van Meyel, D.J. et al. p53 mutation, expression, and DNA ploidy in evolving gliomas: evidence for two pathways of progression. J. Natl Cancer Inst. 86, 1011–1017 (1994).

    Article  CAS  Google Scholar 

  9. Rasheed, B.K. et al. Alterations of the TP53 gene in human gliomas. Cancer Res. 54, 1324–1330 (1994).

    CAS  PubMed  Google Scholar 

  10. Lang, F.F., Miller, D.C., Koslow, M. & Newcomb, E.W. Pathways leading to glioblastoma multiforme: a molecular analysis of genetic alterations in 65 astrocytic tumors. J. Neurosurg. 81, 427–436 (1994).

    Article  CAS  Google Scholar 

  11. Cichowski, K. et al. Mouse models of tumor development in neurofibromatosis type I. Science 286, 2172–2176 (1999).

    Article  CAS  Google Scholar 

  12. Vogel, K. et al. Mouse tumor model for neurofibromatosis type 1. Science 286, 2176–2179 (1999).

    Article  CAS  Google Scholar 

  13. Rizvi, T. et al. Region-specific astrogliosis in brains of mice heterozygous for mutations in the neurofibromatosis type 1 (Nf1) tumor suppressor. Brain Res. 816, 111–123 (1999).

    Article  CAS  Google Scholar 

  14. Gutmann, D. et al. Haploinsufficiency for the neurofibromatosis 1 (NF1) tumor suppressor results in increased astrocyte proliferation. Oncogene 18, 4450–4459 (1999).

    Article  CAS  Google Scholar 

  15. Tischler, A., Shih, T., Williams, B. & Jacks, T. Characterization of pheochromocytomas in a mouse strain with a targeted disruptive mutation of the neurofibromatosis gene Nf1. Endocrine Pathol. 6, 323–335 (1995).

    Article  Google Scholar 

  16. Kleihues, P. & Cavenee, W. Pathology and Genetics of Tumours of the Nervous System (International Agency for Research on Cancer, Lyon, 1997).

    Google Scholar 

  17. Kleihues, P., Burger, P.C. & Scheithauer, B.W. The new WHO classification of brain tumours. Brain Pathol. 3, 255–268 (1993).

    Article  CAS  Google Scholar 

  18. Scherer, H. The forms of growth in gliomas and their practical significance. Brain 63, 1–35 (1940).

    Article  Google Scholar 

  19. Marsden, H.B., Kumar, S., Kahn, J. & Anderton, B.J. A study of glial fibrillary acidic protein (GFAP) in childhood brain tumours. Int. J. Cancer 31, 439–445 (1983).

    Article  CAS  Google Scholar 

  20. Gould, V.E. et al. Synaptophysin: a novel marker for neurons, certain neuroendocrine cells, and their neoplasms. Hum. Pathol. 17, 979–983 (1986).

    Article  CAS  Google Scholar 

  21. Bonner, R. et al. Laser capture microdissection: molecular analysis of tissue. Science 278, 1481–1483 (1997).

    Article  CAS  Google Scholar 

  22. Easton, D., Ponder, M., Huson, S. & Ponder, B. An analysis of variation in expression of neurofibromatosis (NF) type 1 (NF1): evidence for modifying genes. Am. J. Hum. Genet. 53, 305–313 (1993).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Jacks, T. Tumor predisposition in mice heterozygous for a targeted mutation in Nf1. Nature Genet. 7, 353–361 (1994).

    Article  CAS  Google Scholar 

  24. Menon, A. et al. Chromosome 17p deletions and p53 gene mutations associated with the formation of malignant neurofibrosarcomas in von Recklinghausen neurofibromatosis. Proc. Natl Acad. Sci. USA 87, 5435–5439 (1990).

    Article  CAS  Google Scholar 

  25. Jacks, T. Tumor spectrum analysis in p53-mutant mice. Curr. Biol. 4, 1–7 (1994).

    Article  CAS  Google Scholar 

  26. Lowe, S., Jacks, T., Housman, D. & Ruley, H. Abrogation of oncogene-associated apoptosis allows transformation of p53-deficient cells. Proc. Natl Acad. Sci. USA 91, 2026–2030 (1994).

    Article  CAS  Google Scholar 

  27. Brugarolas, J., Bronson, R. & Jacks, T. p21 is a critical CDK2 regulator essential for proliferation control in Rb-deficient cells. J. Cell Biol. 141, 503–514 (1998).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank D. Crowley, N.H. Fong, K. Mercer and B. Williams for technical assistance; A. Charest, J. Sage, K. Cichowski, K. Olive, K. Johnson, K. Tsai and D. Tuveson for discussions; C. Whittaker, C. Fry, M. Bryce and J. Reilly for assistance in preparation of the manuscript and figures; and S. Shih for assistance with inbreeding of Nf1+/− mice. K.M.R. is supported by a post-doctoral fellowship from the Leukemia & Lymphoma Society. T.J. is an Associate Investigator of the Howard Hughes Medical Institute. This work was supported in part by a grant from the Department of the Army and the Medallion Foundation.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Tyler Jacks.

Supplementary information

Table 1

Tumor phenotype of NPcis mice (XLS 40 kb)

The table summarizes for the mice in this study the sex; strain background (background); generation (gen); age at time of death (age); description of tumor histology as scored by atypical nuclei (AN), mitotic figures (M), presence of a focal tumor (FT), necrosis (N), excessive or aberrant blood vessels (V), multinucleated giant cells (GC), and spread to the leptomeninges (LM); grade of tumor based on the grading criteria described in figure 6 legend (grade); immunoreactivity for GFAP, synaptophysin (SNPN), and nestin; the number of days the animal was monitored from the time it was noticed to be sick to the time of sacrifice (days to sac); the observations at the time of sacrifice (reason for sacrifice); whether other tumor types were present (other tumors); and location of tumor (location). The age of the mice is given in months. Immunoreactivity for GFAP is indicated by "+++" for widespread staining of tumor cells, "++" for staining of sparse tumor cells within the tumor, "+" for cells staining within the tumor that could not be distinguished from reactive astrocytes. Tumors scored "+" for GFAP may contain positive staining tumor cells; however, the morphology of the stained cells did not allow us to determine whether the cells were tumor cells or normal cells. Immunoreactivity for synaptophysin is indicated by "-" for no positive cells within the tumor and "+/-" for tumors showing staining of normal neurons within the region of the tumor. These tumors were located in the olfactory bulb or pons and had invaded regions of dense neuronal processes. No tumor showed immunoreactivity for nestin. It should be noted that for the purposes of this study any mouse showing signs of neurological impairment was sacrificed immediately; therefore the days to sacrifice cannot be taken as a measure of brain tumor progression. The mice that were followed for long periods of time showed slow growing, externally visible soft tissue sarcomas. The mice listed under Laser Captured Samples and Additional Tumor Cell Line Samples were not from the same sibling set as the mice in the upper section of the table. They were not included in the calculation of penetrance of the phenotype or in the percentage of tumors immunoreative for GFAP. Cell lines were also generated from KR158 (B6) and KR130 (SXB6). Within each strain group the mice are listed in order from youngest at time of death to oldest. The B6, HXB6, and SXB6 all show a clear trend towards higher grade tumors in older mice and 90-100% penetrance of brain tumors after 6 months of age.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Reilly, K., Loisel, D., Bronson, R. et al. Nf1;Trp53 mutant mice develop glioblastoma with evidence of strain-specific effects. Nat Genet 26, 109–113 (2000). https://doi.org/10.1038/79075

Download citation

  • Received:

  • Accepted:

  • Issue Date:

  • DOI: https://doi.org/10.1038/79075

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