Cancer predisposition syndromes are autosomal dominantly inherited conditions in which the affected individuals have a high relative risk for developing cancer. PTEN and neurofibromatosis-2 (NF2) are tumor suppressors that are inactivated in both somatic cancers and in cancer predisposition syndromes that belong to a clinically defined group (phakomatoses). The mechanism for tumor suppression is known only for PTEN and relies on its ability to dephosphorylate phosphoinositides and antagonize the growth promoting effects of the phosphatidylinositol-3 OH (PI-3) kinase. The regulation of both PTEN and NF2 (merlin) is not yet understood, but it appears to depend on the recruitment of both proteins to the plasma membrane and on their phosphorylation. In spite of the similarities between these tumor suppressors, no previous study attempted to link them in a common pathway. The broad objective of this project is to characterize a molecular connection between PTEN and NF2 tumor suppressors. The new finding that PTEN associates with an adaptor molecule that was previously shown to bind to NF2 will constitute the major focus of this study. The detailed project attempts to cover the following topics: (1) the role of the adaptor molecule in cell growth and migration of cultured gene-deficient cells, and in tumor formation and metastasis in the gene-deficient animals already generated in the laboratory;(2) the analysis of the complex between the three molecules in terms of specific binding affinities, size of the complex by gel filtration and membrane co-localization by density gradient fractionation and immunofluorescence;and (3) interrelations in regulation and signaling between PTEN and NF2 in terms of phosphorylation, stability, Akt/PKB kinase and Rac GTP-ase activation. Finding a common molecular link between PTEN and NF2 tumor suppressors that determine similar cancer predisposition syndromes and are frequently inactivated in brain cancer is fundamental to the understanding of the pathogenesis of cancer and constitutes the basis of a targeted cancer therapy.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA107201-05
Application #
7650264
Study Section
Cancer Molecular Pathobiology Study Section (CAMP)
Program Officer
Jhappan, Chamelli
Project Start
2005-09-12
Project End
2011-09-07
Budget Start
2009-07-01
Budget End
2011-09-07
Support Year
5
Fiscal Year
2009
Total Cost
$254,494
Indirect Cost
Name
University of Texas MD Anderson Cancer Center
Department
Neurology
Type
Other Domestic Higher Education
DUNS #
800772139
City
Houston
State
TX
Country
United States
Zip Code
77030
Georgescu, Maria-Magdalena; Mobley, Bret C; Orr, Brent A et al. (2016) NHERF1/EBP50 and NF2 as diagnostic markers for choroid plexus tumors. Acta Neuropathol Commun 4:55
Georgescu, Maria-Magdalena; Gagea, Mihai; Cote, Gilbert (2016) NHERF1/EBP50 Suppresses Wnt-?-Catenin Pathway-Driven Intestinal Neoplasia. Neoplasia 18:512-23
Georgescu, Maria-Magdalena; Yell, Paul; Mobley, Bret C et al. (2015) NHERF1/EBP50 is an organizer of polarity structures and a diagnostic marker in ependymoma. Acta Neuropathol Commun 3:11
Georgescu, Maria-Magdalena; Cote, Gilbert; Agarwal, Nitin Kumar et al. (2014) NHERF1/EBP50 controls morphogenesis of 3D colonic glands by stabilizing PTEN and ezrin-radixin-moesin proteins at the apical membrane. Neoplasia 16:365-74.e1-2
Agarwal, Nitin Kumar; Zhu, Xiaoping; Gagea, Mihai et al. (2014) PHLPP2 suppresses the NF-?B pathway by inactivating IKK? kinase. Oncotarget 5:815-23
Zhu, Xiaoping; Morales, Fabiana C; Agarwal, Nitin Kumar et al. (2013) Moesin is a glioma progression marker that induces proliferation and Wnt/?-catenin pathway activation via interaction with CD44. Cancer Res 73:1142-55
Molina, J R; Agarwal, N K; Morales, F C et al. (2012) PTEN, NHERF1 and PHLPP form a tumor suppressor network that is disabled in glioblastoma. Oncogene 31:1264-74
Morales, F C; Hayashi, Y; van Pelt, C S et al. (2012) NHERF1/EBP50 controls lactation by establishing basal membrane polarity complexes with prolactin receptor. Cell Death Dis 3:e391
Molina, Jennifer R; Morales, Fabiana C; Hayashi, Yuho et al. (2010) Loss of PTEN binding adapter protein NHERF1 from plasma membrane in glioblastoma contributes to PTEN inactivation. Cancer Res 70:6697-703
Molina, Jennifer R; Hayashi, Yuho; Stephens, Clifton et al. (2010) Invasive glioblastoma cells acquire stemness and increased Akt activation. Neoplasia 12:453-63

Showing the most recent 10 out of 16 publications