Multiple Endocrine Neoplasias are characterized by a series of oncogenic events. Patients with the most severe form, MEN2B, exhibit metastatic medullary thyroid carcinoma (MTC), pheochromocytomas, ganglioneuromas, and mucosal neuromas, in addition to earlier developmental defects. Most patients diagnosed with MEN2B contain a dominantly-acting point mutation in the intracellular tyrosine kinase catalytic domain of the Ret receptor tyrosine kinase, resulting in ligand-and dimerization-independent activation of the receptor. Similar effects have been observed with sporadic MTCs, which often have the same M918T mutation. The specific effect of this mutation is not known, but biochemical evidence suggests that the catalytic specificity of the receptor is altered. The identity of this putative new signaling pathway is not known. This proposal seeks to use the Drosophila retina as a model system to identify both the signaling pathways activated by the M918T mutation and to identify candidate drug targets. The Drosophila retina was chosen due to the availability of powerful molecular and genetic tools and the exceptional accessibility of the retina to genetic screens. The orthologous mutation was made in the Drosophila Ret ortholog dRet; the mutant construct was fused to a retinal-specific promoter and stable transgenic lines were established. Expression of dRetM1007T in the retina resulted in overproliferation and developmental defects that are strikingly reminiscent of the symptoms observed in MEN2B and sporadic MTC patients. A dRetM1007T transgenic line was used in a genetic screen to identify genetic modifiers of the dRetM1007T phenotype. Preliminary data are suggestive of activation of at least two separable signal transduction pathways, at least one of which fails to demonstrate genetic interaction with an activated form of the dEGFR. Of particular importance was the identification of genetic suppressors, loci which when their genetic dose was halved brought the severe dRetM1007T phenotype nearly back to wild type. The corresponding genes are of particular interest as potential drug targets, as partial reduction of their activity may have important therapeutic effects. This Proposal seeks to further characterize the role of these genetic modifiers both in the Drosophila retina model system and directly in human medullary thyroid carcinoma tissue.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
1R01CA084309-01
Application #
6039344
Study Section
Special Emphasis Panel (ZCA1-SRRB-X (O1))
Program Officer
Mietz, Judy
Project Start
2000-01-01
Project End
2004-12-31
Budget Start
2000-01-01
Budget End
2000-12-31
Support Year
1
Fiscal Year
2000
Total Cost
$225,082
Indirect Cost
Name
Washington University
Department
Anatomy/Cell Biology
Type
Schools of Medicine
DUNS #
062761671
City
Saint Louis
State
MO
Country
United States
Zip Code
63130
Bangi, Erdem; Murgia, Claudio; Teague, Alexander G S et al. (2016) Functional exploration of colorectal cancer genomes using Drosophila. Nat Commun 7:13615
Das, Tirtha K; Cagan, Ross L (2013) A Drosophila approach to thyroid cancer therapeutics. Drug Discov Today Technol 10:e65-71
Das, T K; Sangodkar, J; Negre, N et al. (2013) Sin3a acts through a multi-gene module to regulate invasion in Drosophila and human tumors. Oncogene 32:3184-97
Dar, Arvin C; Das, Tirtha K; Shokat, Kevan M et al. (2012) Chemical genetic discovery of targets and anti-targets for cancer polypharmacology. Nature 486:80-4
Cordero, Julia B; Macagno, Juan P; Stefanatos, Rhoda K et al. (2010) Oncogenic Ras diverts a host TNF tumor suppressor activity into tumor promoter. Dev Cell 18:999-1011
Vidal, Marcos; Salavaggione, Lorena; Ylagan, Lourdes et al. (2010) A role for the epithelial microenvironment at tumor boundaries: evidence from Drosophila and human squamous cell carcinomas. Am J Pathol 176:3007-14
Vidal, Marcos; Warner, Stephen; Read, Renee et al. (2007) Differing Src signaling levels have distinct outcomes in Drosophila. Cancer Res 67:10278-85
Vidal, Marcos; Cagan, Ross L (2006) Drosophila models for cancer research. Curr Opin Genet Dev 16:10-6
Vidal, Marcos; Larson, David E; Cagan, Ross L (2006) Csk-deficient boundary cells are eliminated from normal Drosophila epithelia by exclusion, migration, and apoptosis. Dev Cell 10:33-44
Vidal, Marcos; Wells, Samuel; Ryan, Anderson et al. (2005) ZD6474 suppresses oncogenic RET isoforms in a Drosophila model for type 2 multiple endocrine neoplasia syndromes and papillary thyroid carcinoma. Cancer Res 65:3538-41

Showing the most recent 10 out of 13 publications