The goal of this application is to study the molecular mechanisms underlying the anti-tumor activity of inhibitors of deacetylase enzymes (HDACs). These enzymes, which catalyze the removal of an acetyl group from the lysine residues of proteins, are well recognized to play an important role in the regulation of gene expression, and have also been implicated in malignant transformation. In recent years, an increasing number of structurally diverse HDAC inhibitors have been identified that block proliferation and induce differentiation and/or apoptosis of tumor cells in culture and in animal models. Quite surprisingly, the effects of HDAC inhibitors seem to be somewhat selective for tumor cells and several of these compounds have now entered phase I clinical trials. From a molecular point of view, HDAC inhibition not only results in hyperacetylation of histones but also of key transcription factors such as p53, GATA-1 and estrogen receptoralpha. However thus far, the functional significance of acetylation of non-histone proteins in regulation of cell growth, and the precise mechanisms through which HDAC inhibitors induce tumor cell growth arrest remain poorly understood. We have now identified the p53 gene product as a major determinant of sensitivity to these agents, though in an unexpected way. We found that cells harboring mutations of the p53 gene, which generally confer resistance to treatment with canonical chemotherapeutic agents, are sensitive to the action of the HDAC inhibitor, TSA. We provide evidence that inhibition of HDACs, via TSA treatment, restores function from several types of p53 mutants at least in part due to p53 acetylation, and thus promotes apoptosis. By contrast, in the case of wild-type p53 acetylation of a particular residue, Lysine 320, confers chemo-protection. Based on these data we hypothesize that acetylation differentially influences the activity of wild-type and mutant forms of p53. To address this issue we will take advantage of mice genetically modified in components of the acetylation and of the p53 pathway, of unique cell types, and of a panel of p53 proteins harboring mutations at the known acetylation sites. Furthermore, we propose to identify the cellular deacetylase(s) that targets K320 and to exploit acetylation of this residue for therapeutic purposes. We expect that these studies will generate important information on the pathogenesis of cancer disease, and will provide new leads for the therapy of many types of cancer.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA102746-05
Application #
7359670
Study Section
Drug Discovery and Molecular Pharmacology Study Section (DMP)
Program Officer
Blair, Donald G
Project Start
2004-04-08
Project End
2010-02-28
Budget Start
2008-03-01
Budget End
2010-02-28
Support Year
5
Fiscal Year
2008
Total Cost
$271,505
Indirect Cost
Name
Georgetown University
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
049515844
City
Washington
State
DC
Country
United States
Zip Code
20057
Kolukula, Vamsi K; Sahu, Geetaram; Wellstein, Anton et al. (2014) SLC25A1, or CIC, is a novel transcriptional target of mutant p53 and a negative tumor prognostic marker. Oncotarget 5:1212-25
Cimini, Annamaria; Gentile, Roberta; D'Angelo, Barbara et al. (2013) Cocoa powder triggers neuroprotective and preventive effects in a human Alzheimer's disease model by modulating BDNF signaling pathway. J Cell Biochem 114:2209-20
Choudhury, Sujata; Kolukula, Vamsi K; Preet, Anju et al. (2013) Dissecting the pathways that destabilize mutant p53: the proteasome or autophagy? Cell Cycle 12:1022-9
Ullmann, Rebecca; Chien, Christopher D; Avantaggiati, Maria Laura et al. (2012) An acetylation switch regulates SUMO-dependent protein interaction networks. Mol Cell 46:759-70
Rodriguez, Olga Catalina; Choudhury, Sujatra; Kolukula, Vamsi et al. (2012) Dietary downregulation of mutant p53 levels via glucose restriction: mechanisms and implications for tumor therapy. Cell Cycle 11:4436-46
Kirilyuk, Alexander; Shimoji, Mika; Catania, Jason et al. (2012) An intrinsically disordered region of the acetyltransferase p300 with similarity to prion-like domains plays a role in aggregation. PLoS One 7:e48243
Vissapragada, Sarada; Ghosh, Anup; Ringer, Lymor et al. (2010) Dietary n-3 polyunsaturated fatty acids fail to reduce prostate tumorigenesis in the PB-ErbB-2 x Pten(+/-) preclinical mouse model. Cell Cycle 9:1824-9
Cheema, Amrita; Knights, Chad D; Rao, Mahadev et al. (2010) Functional mimicry of the acetylated C-terminal tail of p53 by a SUMO-1 acetylated domain, SAD. J Cell Physiol 225:371-84
Perez, Ricardo E; Knights, Chad D; Sahu, Geetaram et al. (2010) Restoration of DNA-binding and growth-suppressive activity of mutant forms of p53 via a PCAF-mediated acetylation pathway. J Cell Physiol 225:394-405
Di Giovanni, Simone; Knights, Chad D; Rao, Mahadev et al. (2006) The tumor suppressor protein p53 is required for neurite outgrowth and axon regeneration. EMBO J 25:4084-96

Showing the most recent 10 out of 11 publications