One of the most elegant and tightly regulated mechanisms for gene expression is alternative pre-mRNA splicing. Despite the undisputed importance of regulated splicing in several developmental processes and human disease, relatively little is known about the mechanisms by which specific alternative splice choices are regulated. Importantly, the identification of numerous alternatively spliced isoforms in cancer highlights the potential role of the spliceome in the tumor phenotype. Recently, we have shown that specific types of cell stress initiate coordinated alternative splicing of the p53 modulators mdm2 and mdm4 (mdmx). The predominant activity of the mdm2 alternative transcripts is to activate the p53 pathway by inhibiting MDM2 in a dominant negative fashion. These same alternative transcripts have been identified in numerous human tumors. We are interested in studying the regulation of mdm2 splicing to gain a better understanding of 1) regulation of the p53 pathway by alternative splicing in cancer 2) the relationship between the cancer spliceome and the cancer phenotype and 3) the role of the resultant spliced forms in the induction of cancer. Our data suggest that some transformed cells provide a constant signal to activate p53 through sustained expression of mdm2 alternatively spliced transcripts. The proposed research will elucidate the molecular connection between RNA splicing, the p53 response, and ultimately cancer. Unraveling this pathway will undoubtedly lead to the discovery of novel therapeutic intervention points and thus be a crucial stepping-stone for drug discovery.

Public Health Relevance

p53 is one of the most important tumor suppressor genes whose normal function is to prevent inappropriate cell growth and proliferation. The proposed research will decipher the role of alternative splicing in transformation and disruption of the p53 pathway. Since activation of the p53 tumor suppressor pathway is a central target for cancer therapy, understanding the way that splicing contributes to its silencing could lead to novel therapies to reactive the p53 tumor suppressor activity.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA133571-04
Application #
8447373
Study Section
Cancer Molecular Pathobiology Study Section (CAMP)
Program Officer
Strasburger, Jennifer
Project Start
2010-04-01
Project End
2015-01-31
Budget Start
2013-02-01
Budget End
2014-01-31
Support Year
4
Fiscal Year
2013
Total Cost
$245,202
Indirect Cost
$74,923
Name
Nationwide Children's Hospital
Department
Type
DUNS #
147212963
City
Columbus
State
OH
Country
United States
Zip Code
43205
Comiskey Jr, D F; Jacob, A G; Sanford, B L et al. (2018) A novel mouse model of rhabdomyosarcoma underscores the dichotomy of MDM2-ALT1 function in vivo. Oncogene 37:95-106
Jacob, Aishwarya G; Singh, Ravi K; Mohammad, Fuad et al. (2014) The splicing factor FUBP1 is required for the efficient splicing of oncogene MDM2 pre-mRNA. J Biol Chem 289:17350-64
Jacob, Aishwarya G; Singh, Ravi K; Comiskey Jr, Daniel F et al. (2014) Stress-induced alternative splice forms of MDM2 and MDMX modulate the p53-pathway in distinct ways. PLoS One 9:e104444
Jacob, Aishwarya G; O'Brien, Dennis; Singh, Ravi K et al. (2013) Stress-induced isoforms of MDM2 and MDM4 correlate with high-grade disease and an altered splicing network in pediatric rhabdomyosarcoma. Neoplasia 15:1049-63
O'Brien, D; Jacob, A G; Qualman, S J et al. (2012) Advances in pediatric rhabdomyosarcoma characterization and disease model development. Histol Histopathol 27:13-22