Two Tumor Suppressors, p27Kip1 and Patched-1, Collaborate to Prevent Medulloblastoma

Two cyclin-dependent kinase inhibitors, p18Ink4c and p27Kip1, are required for proper cerebellar development. Loss of either of these proteins conferred a proliferative advantage to granule neuron progenitors, although inactivation of Kip1 exerted a greater effect. Mice heterozygous for Patched-1 (Ptc1+/−) that are either heterozygous or nullizygous for Kip1 developed medulloblastoma rapidly and with high penetrance. All tumors from Ptc1+/−;Kip1+/− or Ptc1+/−;Kip1−/− mice failed to express the wild-type Ptc1 allele, consistent with its role as a canonical “two-hit” tumor suppressor. In contrast, expression of the wild-type p27Kip1 protein was invariably maintained in medulloblastomas arising in Ptc1+/−;Kip1+/− mice, indicating that Kip1 is haploinsufficient for tumor suppression. Although medulloblastomas occurring in Ptc1+/− mice were histopathologically heterogeneous and contained intermixed regions of both rapidly proliferating and nondividing more differentiated cells, tumors that also lacked Kip1 were uniformly less differentiated, more highly proliferative, and invasive. Molecular analysis showed that the latter medulloblastomas exhibited constitutive activation of the Sonic hedgehog signaling pathway without loss of functional p53. Apart from gains or losses of single chromosomes, with gain of chromosome 6 being the most frequent, no other chromosomal anomalies were identified by spectral karyotyping, and half of the medulloblastomas so examined retained a normal karyotype. In this respect, this mouse medulloblastoma model recapitulates the vast majority of human medulloblastomas that do not sustain TP53 mutations and are not aneuploid. (Mol Cancer Res 2009;7(1):33–40)

[1]  Ludger Hengst,et al.  The Cdk inhibitor p27 in human cancer: prognostic potential and relevance to anticancer therapy , 2008, Nature Reviews Cancer.

[2]  F. Zindy,et al.  Post-transcriptional down-regulation of Atoh1/Math1 by bone morphogenic proteins suppresses medulloblastoma development. , 2008, Genes & development.

[3]  S. Baylin,et al.  Cooperation between the Hic1 and Ptch1 tumor suppressors in medulloblastoma. , 2008, Genes & development.

[4]  Joel I. Pritchard,et al.  The Smo/Smo model: hedgehog-induced medulloblastoma with 90% incidence and leptomeningeal spread. , 2008, Cancer research.

[5]  D. Ellison,et al.  The origins of medulloblastoma subtypes. , 2008, Annual review of pathology.

[6]  Roger E. Taylor,et al.  Nodule Formation and Desmoplasia in Medulloblastomas—Defining the Nodular/Desmoplastic Variant and Its Biological Behavior , 2007, Brain pathology.

[7]  F. Zindy,et al.  Genetic alterations in mouse medulloblastomas and generation of tumors de novo from primary cerebellar granule neuron precursors. , 2007, Cancer research.

[8]  F. Zindy,et al.  N-Myc and the cyclin-dependent kinase inhibitors p18Ink4c and p27Kip1 coordinately regulate cerebellar development. , 2006, Proceedings of the National Academy of Sciences of the United States of America.

[9]  Michael M. Murphy,et al.  XRCC4 suppresses medulloblastomas with recurrent translocations in p53-deficient mice. , 2006, Proceedings of the National Academy of Sciences of the United States of America.

[10]  S. Pazzaglia Ptc1 heterozygous knockout mice as a model of multi-organ tumorigenesis. , 2006, Cancer letters.

[11]  Y. Ishizaki Control of proliferation and differentiation of neural precursor cells: focusing on the developing cerebellum. , 2006, Journal of pharmacological sciences.

[12]  M. Hatten,et al.  The tumor suppressors Ink4c and p53 collaborate independently with Patched to suppress medulloblastoma formation. , 2005, Genes & development.

[13]  J. Kessler,et al.  Loss of patched and disruption of granule cell development in a pre-neoplastic stage of medulloblastoma , 2005, Development.

[14]  S. Marino Medulloblastoma: developmental mechanisms out of control. , 2005, Trends in molecular medicine.

[15]  S. Pazzaglia,et al.  Modulation of Patched-Associated Susceptibility to Radiation Induced Tumorigenesis by Genetic Background , 2004, Cancer Research.

[16]  Deanna Naeve,et al.  Arf induces p53-dependent and -independent antiproliferative genes. , 2003, Cancer research.

[17]  V. Godfrey,et al.  Haploinsufficiency of p18INK4c Sensitizes Mice to Carcinogen-Induced Tumorigenesis , 2003, Molecular and Cellular Biology.

[18]  P. Pandolfi,et al.  p27 deficiency desensitizes Rb−/− cells to signals that trigger apoptosis during pituitary tumor development , 2003, Oncogene.

[19]  D. Rowitch,et al.  Development of mice expressing a single D-type cyclin. , 2002, Genes & development.

[20]  S. Pazzaglia,et al.  High incidence of medulloblastoma following X-ray-irradiation of newborn Ptc1 heterozygous mice , 2002, Oncogene.

[21]  James M. Olson,et al.  Medulloblastoma Growth Inhibition by Hedgehog Pathway Blockade , 2002, Science.

[22]  D. Ellison,et al.  Classifying the medulloblastoma: insights from morphology and molecular genetics , 2002, Neuropathology and applied neurobiology.

[23]  Huda Y. Zoghbi,et al.  Genetic regulation of cerebellar development , 2001, Nature Reviews Neuroscience.

[24]  R. Bronson,et al.  Deletion of the p27Kip1 gene restores normal development in cyclin D1-deficient mice. , 2001, Proceedings of the National Academy of Sciences of the United States of America.

[25]  T. Curran,et al.  Loss of p53 but not ARF accelerates medulloblastoma in mice heterozygous for patched. , 2001, Cancer research.

[26]  V. Godfrey,et al.  Functional Collaboration between Different Cyclin-Dependent Kinase Inhibitors Suppresses Tumor Growth with Distinct Tissue Specificity , 2000, Molecular and Cellular Biology.

[27]  T. Himi,et al.  A Role for p27/Kip1 in the Control of Cerebellar Granule Cell Precursor Proliferation , 2000, The Journal of Neuroscience.

[28]  M. Barbacid,et al.  Limited overlapping roles of P15INK4b and P18INK4c cell cycle inhibitors in proliferation and tumorigenesis , 2000, The EMBO journal.

[29]  M. Pagano,et al.  Regulation of the Cdk inhibitor p27 and its deregulation in cancer , 2000, Journal of cellular physiology.

[30]  R. Bronson,et al.  Deletion of the p 27 Kip 1 gene restores normal development in cyclin D 1-deficient mice , 2000 .

[31]  James M. Roberts,et al.  CDK inhibitors: positive and negative regulators of G1-phase progression. , 1999, Genes & development.

[32]  James M. Roberts,et al.  The p21Cip1 and p27Kip1 CDK ‘inhibitors’ are essential activators of cyclin D‐dependent kinases in murine fibroblasts , 1999, The EMBO journal.

[33]  M. Scott,et al.  Hedgehog and Patched in Neural Development and Disease , 1998, Neuron.

[34]  James M. Roberts,et al.  The murine gene p27Kip1 is haplo-insufficient for tumour suppression , 1998, Nature.

[35]  V. Godfrey,et al.  CDK inhibitors p18(INK4c) and p27(Kip1) mediate two separate pathways to collaboratively suppress pituitary tumorigenesis. , 1998, Genes & development.

[36]  V. Godfrey,et al.  CDK inhibitors p18 INK4c and p27 Kip1 mediate two separate pathways to collaboratively suppress pituitary tumorigenesis , 1998 .

[37]  H. Zoghbi,et al.  Math1 is essential for genesis of cerebellar granule neurons , 1997, Nature.

[38]  M. Scott,et al.  Altered neural cell fates and medulloblastoma in mouse patched mutants. , 1997, Science.

[39]  Nobuyuki Shishido,et al.  Mice Lacking p27 Kip1 Display Increased Body Size, Multiple Organ Hyperplasia, Retinal Dysplasia, and Pituitary Tumors , 1996, Cell.

[40]  James M. Roberts,et al.  A Syndrome of Multiorgan Hyperplasia with Features of Gigantism, Tumorigenesis, and Female Sterility in p27 Kip1 -Deficient Mice , 1996, Cell.

[41]  K. Manova-Todorova,et al.  Enhanced Growth of Mice Lacking the Cyclin-Dependent Kinase Inhibitor Function of p27 Kip1 , 1996, Cell.