This application proposes experiments that will further explore the molecular mechanisms by which the cell surface associated mucin, MUC1, contributes to the progression of pancreatic adenocarcinoma. Previous work has established that MUC1 is overexpressed and differentially glycosylated by different adenocarcinomas, and that this overexpression is associated with aggressive (invasive and metastatic) forms of pancreatic and other cancers. MUC1 plays multiple and key roles at the cell surface of epithelial cells: it configures some aspects of cell surface adhesion properties, and it communicates information about the status of the cell surface to the nucleus through participation in different pathways of signal transduction. We now know that alterations in MUC1 structure or binding status are communicated to internal compartments of the cell through modifications of the cytoplasmic tail, which can be phosphorylated by different serine, threonine and tyrosine kinases. Our recent results have revealed that MUC1 can have dramatic modulatory effects on cellular functions in response to external growth factor/cytokine stimulation through interactions with receptor tyrosine kinases. MUC1 overexpression on pancreatic tumor cells significantly increases the invasive and metastatic properties of cells stimulated with PDGF-BB. The mechanism whereby this occurs involves phosphorylation of the MUC1 cytoplasmic tail by PDGFR2 and subsequent signaling by the MUC1 cytoplasmic tail, which is transported to and modifies the transcriptional activity of specific promoter sites in the nucleus in association with other signaling and transcription factors. In contrast to the results with PDGF-BB, stimulation of pancreatic tumor cells with hepatocyte growth factor (HGF) under conditions of MUC1 overexpression decreases cell invasive through a mechanism in which MUC1 cytoplasmic tail phosphorylated by cMet associates with p53 and modulates the ability of p53 to associate with AP-1 at promoter sites and activate expression of genes associated with invasion (e.g. MMP1). These results support previous findings that MUC1 can influence the invasive and metastatic properties of tumors in different ways (both enhancing and decreasing). Our results provide a molecular explanation for these different effects by showing that they are context dependent and affected by the cytokine and tissue environment of the cell on which MUC1 is expressed. These results provide new insight into the overall metastatic process, in which cells first take on activities of invasion to leave a primary tumor site, which can be shut down when metastatic tumor cells take up residence in a new organ site and no longer need to migrate. As we go forward we have chosen to focus this renewal application on the mechanisms by which MUC1 is involved in signal transduction and transcriptional regulation, and how this influences invasion and metastasis of pancreatic tumor cells.

Public Health Relevance

This application proposes experiments that will further explore the molecular mechanisms by which the cell surface associated mucin, MUC1, contributes to the progression of pancreatic adenocarcinoma. This renewal application will investigate the mechanisms by which MUC1 is involved in signal transduction and transcriptional regulation, and how this influences invasion and metastasis of pancreatic tumor cells.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA057362-15
Application #
7800427
Study Section
Tumor Progression and Metastasis Study Section (TPM)
Program Officer
Sathyamoorthy, Neeraja
Project Start
1992-09-01
Project End
2013-04-30
Budget Start
2010-05-01
Budget End
2011-04-30
Support Year
15
Fiscal Year
2010
Total Cost
$263,177
Indirect Cost
Name
University of Nebraska Medical Center
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
168559177
City
Omaha
State
NE
Country
United States
Zip Code
68198
Fink, Darci M; Steele, Maria M; Hollingsworth, Michael A (2016) The lymphatic system and pancreatic cancer. Cancer Lett 381:217-36
Hanson, Ryan L; Brown, Roger B; Steele, Maria M et al. (2016) Identification of FRA-1 as a novel player in pancreatic cancer in cooperation with a MUC1: ERK signaling axis. Oncotarget 7:39996-40011
Liu, X; Caffrey, T C; Steele, M M et al. (2014) MUC1 regulates cyclin D1 gene expression through p120 catenin and ?-catenin. Oncogenesis 3:e107
Liu, Xiang; Huang, Huocong; Remmers, Neeley et al. (2014) Loss of E-cadherin and epithelial to mesenchymal transition is not required for cell motility in tissues or for metastasis. Tissue Barriers 2:e969112
Thege, Fredrik I; Lannin, Timothy B; Saha, Trisha N et al. (2014) Microfluidic immunocapture of circulating pancreatic cells using parallel EpCAM and MUC1 capture: characterization, optimization and downstream analysis. Lab Chip 14:1775-84
Radhakrishnan, Prakash; Dabelsteen, Sally; Madsen, Frey Brus et al. (2014) Immature truncated O-glycophenotype of cancer directly induces oncogenic features. Proc Natl Acad Sci U S A 111:E4066-75
Liu, Xiang; Yi, Chunhui; Wen, Yunfei et al. (2014) Interactions between MUC1 and p120 catenin regulate dynamic features of cell adhesion, motility, and metastasis. Cancer Res 74:1609-20
Remmers, Neeley; Anderson, Judy M; Linde, Erin M et al. (2013) Aberrant expression of mucin core proteins and o-linked glycans associated with progression of pancreatic cancer. Clin Cancer Res 19:1981-93
Radhakrishnan, Prakash; Grandgenett, Paul M; Mohr, Ashley M et al. (2013) Expression of core 3 synthase in human pancreatic cancer cells suppresses tumor growth and metastasis. Int J Cancer 133:2824-33
Radhakrishnan, Prakash; Mohr, Ashley M; Grandgenett, Paul M et al. (2013) MicroRNA-200c modulates the expression of MUC4 and MUC16 by directly targeting their coding sequences in human pancreatic cancer. PLoS One 8:e73356

Showing the most recent 10 out of 59 publications