The Wnt signaling pathway provides a paradigm for how misregulation of cell signaling contributes to oncogenesis. Loss-of-function mutations in the tumor suppressor protein, Adenomatous polyposis coli (APC), inappropriately activate Wnt signaling and frequently initiate cancer progression. In fact, mutations in APC are responsible for over 80% of all colon cancer cases, thus it is clear that understanding APC biology could provide important insight to develop novel therapeutic strategies. In the current model of Wnt signaling, APC participates in a multi-protein ?destruction complex? that targets the key effector protein ?-catenin for ubiquitin-mediated proteolysis. In the absence of APC, the destruction complex is inactivated, ?-catenin hyper-accumulates, and it translocates to the nucleus where pro-proliferative genes such as c-myc and cyclinD1 are transactivated. A long-term goal of my lab is to understand how ?-catenin signaling is regulated, and we are currently focused on understanding how the destruction complex works as a molecular machine. While it is clear that APC participates in the complex, its mechanistic role remains unknown. In the past decade, several models have been proposed to explain APC's mechanistic function in the destruction complex. While these models are based on convincing biochemical data, they remain to be rigorously tested using functional studies. Drosophila APC2 is an ideal model for such functional studies, as Wnt pathway components are highly conserved across metazoans, and I have previously shown that Drosophila APC2 works as effectively as human APC in human colon cancer cell lines. My lab therefore uses a multi-disciplinary approach that combines state-of-the-art cell biological and biochemical tools in human cell culture, with the powerful genetic in vivo system of Drosophila development to investigate questions important to the progression and treatment of human colon cancer. During our prior funding cycle, my lab utilized a structure/function approach to investigate the importance of different ?-catenin binding sites on APC for proper Wnt regulation. Our findings are inconsistent with existing models, and instead suggest an alternative model of the inner workings of the destruction complex. Here, I propose experiments to directly evaluate this model, and to pursue studies investigating how APC can modulate Wnt signaling outputs independent of ?- catenin destruction.

Public Health Relevance

Colon cancer is the third leading cause of cancer-related deaths in the US with an estimated 140,000 new diagnoses and 50,000 deaths per year. Over 80% of all colon cancer cases are initiated by inactivating mutations in the tumor suppressor protein, Adenomatous Polyposis Coli (APC), which is a negative regulator of the Wnt signaling pathway. Despite intense research on APC, its mechanistic function in Wnt signaling has remained elusive; therefore, in this proposal I outline functional studies to investigate APC's role. The results of these studies will provide insight into how APC mutations stimulate the oncogenic phenotype, and may ultimately help illuminate novel treatment options for colon cancer.

Agency
National Institute of Health (NIH)
Institute
National Institute of General Medical Sciences (NIGMS)
Type
Academic Research Enhancement Awards (AREA) (R15)
Project #
2R15GM107796-02
Application #
9305192
Study Section
Development - 2 Study Section (DEV2)
Program Officer
Melillo, Amanda A
Project Start
2013-09-01
Project End
2020-05-31
Budget Start
2017-06-01
Budget End
2020-05-31
Support Year
2
Fiscal Year
2017
Total Cost
$385,329
Indirect Cost
$85,371
Name
Franklin and Marshall College
Department
Biology
Type
Schools of Arts and Sciences
DUNS #
069773646
City
Lancaster
State
PA
Country
United States
Zip Code
17604
Schaefer, Kristina N; Bonello, Teresa T; Zhang, Shiping et al. (2018) Supramolecular assembly of the beta-catenin destruction complex and the effect of Wnt signaling on its localization, molecular size, and activity in vivo. PLoS Genet 14:e1007339
Croy, Heather E; Fuller, Caitlyn N; Giannotti, Jemma et al. (2016) The Poly(ADP-ribose) Polymerase Enzyme Tankyrase Antagonizes Activity of the ?-Catenin Destruction Complex through ADP-ribosylation of Axin and APC2. J Biol Chem 291:12747-60
Kunttas-Tatli, Ezgi; Von Kleeck, Ryan A; Greaves, Bradford D et al. (2015) The two SAMP repeats and their phosphorylation state in Drosophila Adenomatous polyposis coli-2 play mechanistically distinct roles in negatively regulating Wnt signaling. Mol Biol Cell 26:4503-18
Hales, Karen G; Korey, Christopher A; Larracuente, Amanda M et al. (2015) Genetics on the Fly: A Primer on the Drosophila Model System. Genetics 201:815-42
Yamulla, Robert J; Kane, Eric G; Moody, Alexandra E et al. (2014) Testing models of the APC tumor suppressor/?-catenin interaction reshapes our view of the destruction complex in Wnt signaling. Genetics 197:1285-302
Kunttas-Tatli, Ezgi; Roberts, David M; McCartney, Brooke M (2014) Self-association of the APC tumor suppressor is required for the assembly, stability, and activity of the Wnt signaling destruction complex. Mol Biol Cell 25:3424-36
Poulton, John S; Mu, Frank W; Roberts, David M et al. (2013) APC2 and Axin promote mitotic fidelity by facilitating centrosome separation and cytoskeletal regulation. Development 140:4226-36