Skip to main content
Top
Published in: Journal of Neuro-Oncology 3/2008

Open Access 01-02-2008 | Lab. Investigation - human/Animal Tissue

Medulloblastomas overexpress the p53-inactivating oncogene WIP1/PPM1D

Authors: Robert C. Castellino, Massimiliano De Bortoli, Xiongbin Lu, Sung-Hwan Moon, Thuy-Ai Nguyen, Mark A. Shepard, Pulivarthi H. Rao, Lawrence A. Donehower, John Y. H. Kim

Published in: Journal of Neuro-Oncology | Issue 3/2008

Login to get access

Abstract

Medulloblastoma is the most common malignant brain tumor of childhood. Despite numerous advances, clinical challenges range from recurrent and progressive disease to long-term toxicities in survivors. The lack of more effective, less toxic therapies results from our limited understanding of medulloblastoma growth. Although TP53 is the most commonly altered gene in cancers, it is rarely mutated in medulloblastoma. Accumulating evidence, however, indicates that TP53 pathways are disrupted in medulloblastoma. Wild-type p53-induced phosphatase 1 (WIP1 or PPM1D) encodes a negative regulator of p53. WIP1 amplification (17q22-q23) and its overexpression have been reported in diverse cancer types. We examined primary medulloblastoma specimens and cell lines, and detected WIP1 copy gain and amplification prevalent among but not exclusively in the tumors with 17q gain and isochromosome 17q (i17q), which are among the most common cytogenetic lesions in medulloblastoma. WIP1 RNA levels were significantly higher in the tumors with 17q gain or i17q. Immunoblots confirmed significant WIP1 protein in primary tumors, generally higher in those with 17q gain or i17q. Under basal growth conditions and in response to the chemotherapeutic agent, etoposide, WIP1 antagonized p53-mediated apoptosis in medulloblastoma cell lines. These results indicate that medulloblastoma express significant levels of WIP1 that modulate genotoxic responsiveness by negatively regulating p53.
Literature
1.
go back to reference Giangaspero F, Bigner SH, Giordana MT, Kleihues P, Trojanowski JQ (2000) Medulloblastoma. In: Kleihues P, Cavenee WK (eds) Pathology and genetics: tumours of the nervous system. World Health Organization Classification of Tumours. International Agency for Research of Cancer, Lyon, France, pp 96–103 Giangaspero F, Bigner SH, Giordana MT, Kleihues P, Trojanowski JQ (2000) Medulloblastoma. In: Kleihues P, Cavenee WK (eds) Pathology and genetics: tumours of the nervous system. World Health Organization Classification of Tumours. International Agency for Research of Cancer, Lyon, France, pp 96–103
2.
go back to reference McNeil DE, Cote TR, Clegg L, Rorke LB (2002) Incidence and trends in pediatric malignancies medulloblastoma/primitive neuroectodermal tumor: a SEER update. Surveillance epidemiology and end results Med Pediatr Oncol 39:190–194PubMedCrossRef McNeil DE, Cote TR, Clegg L, Rorke LB (2002) Incidence and trends in pediatric malignancies medulloblastoma/primitive neuroectodermal tumor: a SEER update. Surveillance epidemiology and end results Med Pediatr Oncol 39:190–194PubMedCrossRef
3.
go back to reference Rood BR, Macdonald TJ, Packer RJ (2004) Current treatment of medulloblastoma: recent advances and future challenges. Semin Oncol 31:666–675PubMedCrossRef Rood BR, Macdonald TJ, Packer RJ (2004) Current treatment of medulloblastoma: recent advances and future challenges. Semin Oncol 31:666–675PubMedCrossRef
4.
go back to reference Biegel JA (1999) Cytogenetics and molecular genetics of childhood brain tumors. Neuro Oncol 1:139–151PubMedCrossRef Biegel JA (1999) Cytogenetics and molecular genetics of childhood brain tumors. Neuro Oncol 1:139–151PubMedCrossRef
5.
go back to reference Ellison D (2002) Classifying the medulloblastoma: insights from morphology and molecular genetics. Neuropathol Appl Neurobiol 28:257–282PubMedCrossRef Ellison D (2002) Classifying the medulloblastoma: insights from morphology and molecular genetics. Neuropathol Appl Neurobiol 28:257–282PubMedCrossRef
6.
7.
go back to reference Benard J, Douc-Rasy S, Ahomadegbe JC (2003) TP53 family members and human cancers. Hum Mutat 21:182–191PubMedCrossRef Benard J, Douc-Rasy S, Ahomadegbe JC (2003) TP53 family members and human cancers. Hum Mutat 21:182–191PubMedCrossRef
8.
go back to reference Ohgaki H, Eibl RH, Wiestler OD, Yasargil MG, Newcomb EW, Kleihues P (1991) p53 mutations in nonastrocytic human brain tumors. Cancer Res 51:6202–6205PubMed Ohgaki H, Eibl RH, Wiestler OD, Yasargil MG, Newcomb EW, Kleihues P (1991) p53 mutations in nonastrocytic human brain tumors. Cancer Res 51:6202–6205PubMed
9.
go back to reference Saylors RL III, Sidransky D, Friedman HS, et al (1991) Infrequent p53 gene mutations in medulloblastomas. Cancer Res 51:4721–4723PubMed Saylors RL III, Sidransky D, Friedman HS, et al (1991) Infrequent p53 gene mutations in medulloblastomas. Cancer Res 51:4721–4723PubMed
10.
go back to reference Frank AJ, Hernan R, Hollander A, et al (2004) The TP53-ARF tumor suppressor pathway is frequently disrupted in large/cell anaplastic medulloblastoma. Brain Res Mol Brain Res 121:137–140PubMedCrossRef Frank AJ, Hernan R, Hollander A, et al (2004) The TP53-ARF tumor suppressor pathway is frequently disrupted in large/cell anaplastic medulloblastoma. Brain Res Mol Brain Res 121:137–140PubMedCrossRef
11.
go back to reference Eberhart CG, Chaudhry A, Daniel RW, Khaki L, Shah KV, Gravitt PE (2005) Increased p53 immunopositivity in anaplastic medulloblastoma and supratentorial PNET is not caused by JC virus. BMC Cancer 5:19PubMedCrossRef Eberhart CG, Chaudhry A, Daniel RW, Khaki L, Shah KV, Gravitt PE (2005) Increased p53 immunopositivity in anaplastic medulloblastoma and supratentorial PNET is not caused by JC virus. BMC Cancer 5:19PubMedCrossRef
12.
go back to reference Wetmore C, Eberhart DE, Curran T (2001) Loss of p53 but not ARF accelerates medulloblastoma in mice heterozygous for patched. Cancer Res 61:513–516PubMed Wetmore C, Eberhart DE, Curran T (2001) Loss of p53 but not ARF accelerates medulloblastoma in mice heterozygous for patched. Cancer Res 61:513–516PubMed
13.
go back to reference Toledo F, Wahl GM (2006) Regulating the p53 pathway: in vitro hypotheses, in vivo veritas. Nat Rev Cancer 6:909–923PubMedCrossRef Toledo F, Wahl GM (2006) Regulating the p53 pathway: in vitro hypotheses, in vivo veritas. Nat Rev Cancer 6:909–923PubMedCrossRef
14.
go back to reference Adesina AM, Nalbantoglu J, Cavenee WK (1994) p53 gene mutation and mdm2 gene amplification are uncommon in medulloblastoma. Cancer Res 54:5649–5651PubMed Adesina AM, Nalbantoglu J, Cavenee WK (1994) p53 gene mutation and mdm2 gene amplification are uncommon in medulloblastoma. Cancer Res 54:5649–5651PubMed
15.
go back to reference Batra SK, McLendon RE, Koo JS, et al (1995) Prognostic implications of chromosome 17p deletions in human medulloblastomas. J Neurooncol 24:39–45PubMedCrossRef Batra SK, McLendon RE, Koo JS, et al (1995) Prognostic implications of chromosome 17p deletions in human medulloblastomas. J Neurooncol 24:39–45PubMedCrossRef
16.
go back to reference Giordana MT, Duo D, Gasverde S, et al (2002) MDM2 overexpression is associated with short survival in adults with medulloblastoma. Neuro Oncol 4:115–122PubMedCrossRef Giordana MT, Duo D, Gasverde S, et al (2002) MDM2 overexpression is associated with short survival in adults with medulloblastoma. Neuro Oncol 4:115–122PubMedCrossRef
17.
go back to reference Bulavin DV, Demidov ON, Saito S, et al (2002) Amplification of PPM1D in human tumors abrogates p53 tumor-suppressor activity. Nat Genet 31:210–215PubMedCrossRef Bulavin DV, Demidov ON, Saito S, et al (2002) Amplification of PPM1D in human tumors abrogates p53 tumor-suppressor activity. Nat Genet 31:210–215PubMedCrossRef
18.
go back to reference Li J, Yang Y, Peng Y, et al (2002) Oncogenic properties of PPM1D located within a breast cancer amplification epicenter at 17q23. Nat Genet 31:133–134PubMedCrossRef Li J, Yang Y, Peng Y, et al (2002) Oncogenic properties of PPM1D located within a breast cancer amplification epicenter at 17q23. Nat Genet 31:133–134PubMedCrossRef
19.
go back to reference Saito-Ohara F, Imoto I, Inoue J, et al (2003) PPM1D is a potential target for 17q gain in neuroblastoma. Cancer Res 63:1876–1883PubMed Saito-Ohara F, Imoto I, Inoue J, et al (2003) PPM1D is a potential target for 17q gain in neuroblastoma. Cancer Res 63:1876–1883PubMed
20.
go back to reference Bulavin DV, Phillips C, Nannenga B, et al (2004) Inactivation of the Wip1 phosphatase inhibits mammary tumorigenesis through p38 MAPK-mediated activation of the p16(Ink4a)-p19(Arf) pathway. Nat Genet 36:343–350PubMedCrossRef Bulavin DV, Phillips C, Nannenga B, et al (2004) Inactivation of the Wip1 phosphatase inhibits mammary tumorigenesis through p38 MAPK-mediated activation of the p16(Ink4a)-p19(Arf) pathway. Nat Genet 36:343–350PubMedCrossRef
21.
go back to reference Lu X, Nannenga B, Donehower LA (2005) PPM1D dephosphorylates Chk1 and p53 and abrogates cell cycle checkpoints. Genes Dev 19:1162–1174PubMedCrossRef Lu X, Nannenga B, Donehower LA (2005) PPM1D dephosphorylates Chk1 and p53 and abrogates cell cycle checkpoints. Genes Dev 19:1162–1174PubMedCrossRef
22.
go back to reference Mendrzyk F, Radlwimmer B, Joos S, et al (2005) Genomic and protein expression profiling identifies CDK6 as novel independent prognostic marker in medulloblastoma. J Clin Oncol 23:8853–8862PubMedCrossRef Mendrzyk F, Radlwimmer B, Joos S, et al (2005) Genomic and protein expression profiling identifies CDK6 as novel independent prognostic marker in medulloblastoma. J Clin Oncol 23:8853–8862PubMedCrossRef
23.
go back to reference Ehrbrecht A, Muller U, Wolter M, et al (2006) Comprehensive genomic analysis of desmoplastic medulloblastomas: identification of novel amplified genes and separate evaluation of the different histological components. J Pathol 208:554–563PubMedCrossRef Ehrbrecht A, Muller U, Wolter M, et al (2006) Comprehensive genomic analysis of desmoplastic medulloblastomas: identification of novel amplified genes and separate evaluation of the different histological components. J Pathol 208:554–563PubMedCrossRef
24.
go back to reference De Bortoli M, Castellino RC, Lu XY, et al (2006) Medulloblastoma outcome is adversely associated with overexpression of EEF1D, RPL30, and RPS20 on the long arm of chromosome 8. BMC Cancer 6:223PubMedCrossRef De Bortoli M, Castellino RC, Lu XY, et al (2006) Medulloblastoma outcome is adversely associated with overexpression of EEF1D, RPL30, and RPS20 on the long arm of chromosome 8. BMC Cancer 6:223PubMedCrossRef
25.
go back to reference Verma RS, Babu A (eds) (1995) Human chromosomes: principles and techniques. McGraw-Hill, New York, NY Verma RS, Babu A (eds) (1995) Human chromosomes: principles and techniques. McGraw-Hill, New York, NY
26.
go back to reference Rauta J, Alarmo EL, Kauraniemi P, Karhu R, Kuukasjarvi T, Kallioniemi A (2006) The serine-threonine protein phosphatase PPM1D is frequently activated through amplification in aggressive primary breast tumours. Breast Cancer Res Treat 95:257–263PubMedCrossRef Rauta J, Alarmo EL, Kauraniemi P, Karhu R, Kuukasjarvi T, Kallioniemi A (2006) The serine-threonine protein phosphatase PPM1D is frequently activated through amplification in aggressive primary breast tumours. Breast Cancer Res Treat 95:257–263PubMedCrossRef
27.
go back to reference Rao PH, Murty VV, Gaidano G, Hauptschein R, Dalla-Favera R, Chaganti RS (1994) Subregional mapping of 8 single copy loci to chromosome 6 by fluorescence in situ hybridization. Cytogenet Cell Genet 66(4):272–273PubMedCrossRef Rao PH, Murty VV, Gaidano G, Hauptschein R, Dalla-Favera R, Chaganti RS (1994) Subregional mapping of 8 single copy loci to chromosome 6 by fluorescence in situ hybridization. Cytogenet Cell Genet 66(4):272–273PubMedCrossRef
28.
go back to reference el-Deiry WS, Tokino T, Velculescu VE, et al (1993) WAF1, a potential mediator of p53 tumor suppression. Cell 75:817–825PubMedCrossRef el-Deiry WS, Tokino T, Velculescu VE, et al (1993) WAF1, a potential mediator of p53 tumor suppression. Cell 75:817–825PubMedCrossRef
29.
go back to reference Tischer E, Mitchell R, Hartman T, et al (1991) The human gene for vascular endothelial growth factor. Multiple protein forms are encoded through alternative exon splicing. J Biol Chem 266:11947–11954PubMed Tischer E, Mitchell R, Hartman T, et al (1991) The human gene for vascular endothelial growth factor. Multiple protein forms are encoded through alternative exon splicing. J Biol Chem 266:11947–11954PubMed
30.
go back to reference Takekawa M, Adachi M, Nakahata A, et al (2000) p53-inducible wip1 phosphatase mediates a negative feedback regulation of p38 MAPK-p53 signaling in response to UV radiation. EMBO J 19:6517–6526PubMedCrossRef Takekawa M, Adachi M, Nakahata A, et al (2000) p53-inducible wip1 phosphatase mediates a negative feedback regulation of p38 MAPK-p53 signaling in response to UV radiation. EMBO J 19:6517–6526PubMedCrossRef
31.
go back to reference Fiscella M, Zhang H, Fan S, et al (1997) Wip1, a novel human protein phosphatase that is induced in response to ionizing radiation in a p53-dependent manner. Proc Natl Acad Sci USA 94:6048–6053PubMedCrossRef Fiscella M, Zhang H, Fan S, et al (1997) Wip1, a novel human protein phosphatase that is induced in response to ionizing radiation in a p53-dependent manner. Proc Natl Acad Sci USA 94:6048–6053PubMedCrossRef
32.
go back to reference Demidov ON, Kek C, Shreeram S, et al (2006) The role of the MKK6/p38 MAPK pathway in Wip1-dependent regulation of ErbB2-driven mammary gland tumorigenesis. Oncogene 26:2502–2506PubMedCrossRef Demidov ON, Kek C, Shreeram S, et al (2006) The role of the MKK6/p38 MAPK pathway in Wip1-dependent regulation of ErbB2-driven mammary gland tumorigenesis. Oncogene 26:2502–2506PubMedCrossRef
33.
go back to reference Zhang X, Kim J, Ruthazer R, et al (2006) The HBP1 transcriptional repressor participates in RAS-induced premature senescence. Mol Cell Biol 26:8252–8266PubMedCrossRef Zhang X, Kim J, Ruthazer R, et al (2006) The HBP1 transcriptional repressor participates in RAS-induced premature senescence. Mol Cell Biol 26:8252–8266PubMedCrossRef
34.
go back to reference Hirasawa A, Saito-Ohara F, Inoue J, et al (2003) Association of 17q21-q24 gain in ovarian clear cell adenocarcinomas with poor prognosis and identification of PPM1D and APPBP2 as likely amplification targets. Clin Cancer Res 9:1995–2004PubMed Hirasawa A, Saito-Ohara F, Inoue J, et al (2003) Association of 17q21-q24 gain in ovarian clear cell adenocarcinomas with poor prognosis and identification of PPM1D and APPBP2 as likely amplification targets. Clin Cancer Res 9:1995–2004PubMed
35.
go back to reference Attardi LD, de VA, Jacks T (2004) Activation of the p53-dependent G1 checkpoint response in mouse embryo fibroblasts depends on the specific DNA damage inducer. Oncogene 23:973–980PubMedCrossRef Attardi LD, de VA, Jacks T (2004) Activation of the p53-dependent G1 checkpoint response in mouse embryo fibroblasts depends on the specific DNA damage inducer. Oncogene 23:973–980PubMedCrossRef
36.
go back to reference Clifford B, Beljin M, Stark GR, Taylor WR (2003) G2 arrest in response to topoisomerase II inhibitors: the role of p53. Cancer Res 63:4074–4081PubMed Clifford B, Beljin M, Stark GR, Taylor WR (2003) G2 arrest in response to topoisomerase II inhibitors: the role of p53. Cancer Res 63:4074–4081PubMed
37.
go back to reference Nam C, Yamauchi H, Nakayama H, Doi K (2006) Etoposide induces apoptosis and cell cycle arrest of neuroepithelial cells in a p53-related manner. Neurotoxicol Teratol 28:664–672PubMedCrossRef Nam C, Yamauchi H, Nakayama H, Doi K (2006) Etoposide induces apoptosis and cell cycle arrest of neuroepithelial cells in a p53-related manner. Neurotoxicol Teratol 28:664–672PubMedCrossRef
38.
go back to reference Belova GI, Demidov ON, Fornace AJ Jr, Bulavin DV (2005) Chemical inhibition of Wip1 phosphatase contributes to suppression of tumorigenesis. Cancer Biol Ther 4:1154–1158PubMedCrossRef Belova GI, Demidov ON, Fornace AJ Jr, Bulavin DV (2005) Chemical inhibition of Wip1 phosphatase contributes to suppression of tumorigenesis. Cancer Biol Ther 4:1154–1158PubMedCrossRef
39.
go back to reference Yamaguchi H, Durell SR, Feng H, Bai Y, Anderson CW, Appella E (2006) Development of a substrate-based cyclic phosphopeptide inhibitor of protein phosphatase 2Cdelta, Wip1. Biochemistry 45:13193–13202PubMedCrossRef Yamaguchi H, Durell SR, Feng H, Bai Y, Anderson CW, Appella E (2006) Development of a substrate-based cyclic phosphopeptide inhibitor of protein phosphatase 2Cdelta, Wip1. Biochemistry 45:13193–13202PubMedCrossRef
Metadata
Title
Medulloblastomas overexpress the p53-inactivating oncogene WIP1/PPM1D
Authors
Robert C. Castellino
Massimiliano De Bortoli
Xiongbin Lu
Sung-Hwan Moon
Thuy-Ai Nguyen
Mark A. Shepard
Pulivarthi H. Rao
Lawrence A. Donehower
John Y. H. Kim
Publication date
01-02-2008
Publisher
Springer US
Published in
Journal of Neuro-Oncology / Issue 3/2008
Print ISSN: 0167-594X
Electronic ISSN: 1573-7373
DOI
https://doi.org/10.1007/s11060-007-9470-8

Other articles of this Issue 3/2008

Journal of Neuro-Oncology 3/2008 Go to the issue