Chromosome 18q is affected by loss of heterozygosity (LOH) in greater than 70 percent of colorectal cancers, and patients whose tumors have sustained 18q LOH have a markedly worse prognosis than those whose tumors have not. The DCC (deleted in colorectal cancer) gene was identified and cloned on the basis of localized genetic alterations affecting the common region of LOH on 18q, and several lines of evidence suggest DCC is a promising candidate colorectal tumor suppressor gene. Inactivation of DCC may also occur in some cancers of the breast, pancreas, stomach, uterus, prostate, brain, and cervix, as well as in some leukemias. The sequence of DCC predicts a 1447 amino acid transmembrane protein whose four immunoglobulin-like and six fibronectin type III-like extracellular domains show similarity to the neural cell adhesion molecule (N- CAM) family of cell surface proteins. The DCC cytoplasmic domain, however, shares little similarity with any other proteins. The structural similarity of DCC with the N-CAMs suggests it may function through specific cell-cell or cell-extracellular matrix interactions. Nevertheless, at present, many questions remain regarding the role of 18q and DCC alterations in tumorigenesis and the expression pattern and function of DCC in normal and neoplastic cells. For example, specific somatic genetic alterations in the DCC gene in tumors have not been well characterized. In addition, while DCC transcripts are present in most adult tissues at very low levels, with highest levels in brain tissues, DCC expression has not been fully characterized in normal and neoplastic tissues. Furthermore, the means by which DCC functions to regulate cell growth and differentiation is unknown. To address these and other questions four specific aims are proposed: 1) To identify somatic genetic alterations on chromosome 18q, and particularly those present in the DCC gene that inactivate it and/or account for the marked decrease in its expression in colon and other cancers. 2) To obtain detailed information on the structure and expression pattern of DCC transcripts and portein(s) in normal and neoplastic tissues, particularly those of the gastrointestinal tract, using RT-PCR to characterize DCC transcripts and immunoblotting, immunoprecipitation and immunohistochemical techniques to characterize the DCC protein(s). 3) To identify proteins which bind specifically to the cytoplastic domain of DCC. 4) To determine if DCC functions as a tumor suppressor gene in colorectal and other gastrointestinal cancer cell lines. The proposed studies will determine if DCC is the gene targeted for inactivation by 18q allelic losses in colon and other tumor types. In addition, the studies will begin to address the means by which DCC functions to regulate normal cell growth and suppress tumorigenesis. Finally, in addition to furthering the understanding of the pathogenesis of cancer, the studies in this application may provide novel insights and reagents for improving the diagnosis and management of patients with cancer.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA070097-04
Application #
2712776
Study Section
Pathology B Study Section (PTHB)
Program Officer
Gallahan, Daniel L
Project Start
1995-08-28
Project End
2000-05-31
Budget Start
1998-06-01
Budget End
1999-05-31
Support Year
4
Fiscal Year
1998
Total Cost
Indirect Cost
Name
University of Michigan Ann Arbor
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
791277940
City
Ann Arbor
State
MI
Country
United States
Zip Code
48109
Hilgers, W; Song, J J; Haye, M et al. (2000) Homozygous deletions inactivate DCC, but not MADH4/DPC4/SMAD4, in a subset of pancreatic and biliary cancers. Genes Chromosomes Cancer 27:353-7
Kolligs, F T; Kolligs, B; Hajra, K M et al. (2000) gamma-catenin is regulated by the APC tumor suppressor and its oncogenic activity is distinct from that of beta-catenin. Genes Dev 14:1319-31
Hu, G; Fearon, E R (1999) Siah-1 N-terminal RING domain is required for proteolysis function, and C-terminal sequences regulate oligomerization and binding to target proteins. Mol Cell Biol 19:724-32
Meyerhardt, J A; Caca, K; Eckstrand, B C et al. (1999) Netrin-1: interaction with deleted in colorectal cancer (DCC) and alterations in brain tumors and neuroblastomas. Cell Growth Differ 10:35-42
Rimm, D L; Caca, K; Hu, G et al. (1999) Frequent nuclear/cytoplasmic localization of beta-catenin without exon 3 mutations in malignant melanoma. Am J Pathol 154:325-9
Caca, K; Kolligs, F T; Ji, X et al. (1999) Beta- and gamma-catenin mutations, but not E-cadherin inactivation, underlie T-cell factor/lymphoid enhancer factor transcriptional deregulation in gastric and pancreatic cancer. Cell Growth Differ 10:369-76
Hajra, K M; Ji, X; Fearon, E R (1999) Extinction of E-cadherin expression in breast cancer via a dominant repression pathway acting on proximal promoter elements. Oncogene 18:7274-9
Kolligs, F T; Hu, G; Dang, C V et al. (1999) Neoplastic transformation of RK3E by mutant beta-catenin requires deregulation of Tcf/Lef transcription but not activation of c-myc expression. Mol Cell Biol 19:5696-706
Meyerhardt, J A; Look, A T; Bigner, S H et al. (1997) Identification and characterization of neogenin, a DCC-related gene. Oncogene 14:1129-36
Reyes-Mugica, M; Rieger-Christ, K; Ohgaki, H et al. (1997) Loss of DCC expression and glioma progression. Cancer Res 57:382-6

Showing the most recent 10 out of 13 publications