Regulation of cell adhesion and actin cytoskeletal structures play important roles in normal and tumor cell movement. Rho GTPases, such as RhoA, are molecular switches that act to control cell adhesion, motility and have been implicated in tumor progression. The activation of Rho proteins is mediated by specific guanine- nucleotide exchange factors or RhoGEFs, which catalyze the exchange of GDP for GTP. As the number of RhoGEFs outnumber their target GTPases by a factor of 3, it is hypothesized that the spatial and temporal regulation of RhoGEF activity is a key factor in controlling RhoA function within cells. Rgnef is a 190 kDa RhoA-specific GEF (also termed p190RhoGEF) involved in the regulation of integrin-initiated RhoA activation, focal adhesion (FA) formation, and fibroblast cell motility. A novel feature of Rgnef is that it contains a binding site for the integrin-activated focal adhesion kinase (FAK). What remains unknown is how Rgnef is regulated within cells. This proposal will test the hypothesis that Rgnef (via FAK association and tyrosine phosphorylation) regulates RhoA activity, cell adhesion and motility, ultimately influencing ovarian carcinoma tumor progression. Experiments will use gain-of-function assays via reconstitution of mouse embryo fibroblasts (MEFs) obtained from Rgnef-null mice or rescue of knockdown phenotypes in human ovarian carcinoma cells.
Three aims are proposed:
Aim 1 will determine the necessity of Rgnef in FA formation and cell motility. Experiments will include RhoA activity assays, fluorescence microscopy monitoring of FA formation, and real-time imaging to evaluate Rgnef localization and the enhancement of cell motility.
Aim 2 will test the role of FAK binding and tyrosine phosphorylation in Rgnef function. Two sites within Rgnef have been identified by mass spectrometry as phosphorylated by FAK. These sites will be mutated and re-expression of these mutants in Rgnef-null MEFs will be evaluated for effects on intrinsic Rgnef activity, integrin-associated RhoA activation, FA formation, and cell motility.
Aim 3 will evaluate the role of Rgnef in human ovarian carcinoma tumor progression. Recent studies indicate that Rgnef expression is elevated as function of colon carcinoma tumor progression and cellular studies show that Rgnef-FAK interactions contribute to an invasive cell phenotype. Rgnef-specific antibody tissue array staining reveals enhanced expression in advanced stage ovarian carcinoma and preliminary data indicate that Rgnef knockdown inhibits ovarian carcinoma progression. Lentiviral-mediated Rgnef knockdown and re-expression of Rgnef mutants in two human ovarian carcinoma cell lines will be evaluated in cell culture and upon orthotopic tumor growth in nude mice for effects on FA formation-invasion and tumor progression, respectively. Together, these experiments will provide advanced training in cellular signaling, molecular mechanisms of cell motility, and cancer biology model systems. The results will enhance our current understanding of intracellular signaling pathways promoting ovarian carcinoma tumor progression.

Public Health Relevance

Ovarian cancer is a leading cause of mortality in gynecological cancer and approximately 1 in 60 women are diagnosed in the United States annually. The research proposed in this application will analyze the role of Rgnef, a protein implicated in cellular adhesion and motility, both critical factors in tumor cell progression and metastasis. These projects ultimately aim to add to the current understanding of intracellular signaling pathways that contribute to ovarian carcinoma tumor progression.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Postdoctoral Individual National Research Service Award (F32)
Project #
1F32CA159558-01
Application #
8124019
Study Section
Special Emphasis Panel (ZRG1-F09-E (20))
Program Officer
Jakowlew, Sonia B
Project Start
2011-07-01
Project End
2014-06-30
Budget Start
2011-07-01
Budget End
2012-06-30
Support Year
1
Fiscal Year
2011
Total Cost
$53,042
Indirect Cost
Name
University of California San Diego
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
804355790
City
La Jolla
State
CA
Country
United States
Zip Code
92093
Tancioni, Isabelle; Miller, Nichol L G; Uryu, Sean et al. (2015) FAK activity protects nucleostemin in facilitating breast cancer spheroid and tumor growth. Breast Cancer Res 17:47
Tancioni, Isabelle; Uryu, Sean; Sulzmaier, Florian J et al. (2014) FAK Inhibition disrupts a ?5 integrin signaling axis controlling anchorage-independent ovarian carcinoma growth. Mol Cancer Ther 13:2050-61
Jean, Christine; Chen, Xiao Lei; Nam, Ju-Ock et al. (2014) Inhibition of endothelial FAK activity prevents tumor metastasis by enhancing barrier function. J Cell Biol 204:247-63
Miller, N L G; Kleinschmidt, E G; Schlaepfer, D D (2014) RhoGEFs in cell motility: novel links between Rgnef and focal adhesion kinase. Curr Mol Med 14:221-34
Ward, Kristy K; Tancioni, Isabelle; Lawson, Christine et al. (2013) Inhibition of focal adhesion kinase (FAK) activity prevents anchorage-independent ovarian carcinoma cell growth and tumor progression. Clin Exp Metastasis 30:579-94
Miller, Nichol L G; Lawson, Christine; Kleinschmidt, Elizabeth G et al. (2013) A non-canonical role for Rgnef in promoting integrin-stimulated focal adhesion kinase activation. J Cell Sci 126:5074-85
Lim, Ssang-Taek; Miller, Nichol L G; Chen, Xiao Lei et al. (2012) Nuclear-localized focal adhesion kinase regulates inflammatory VCAM-1 expression. J Cell Biol 197:907-19
Miller, Nichol L G; Lawson, Christine; Chen, Xiao Lei et al. (2012) Rgnef (p190RhoGEF) knockout inhibits RhoA activity, focal adhesion establishment, and cell motility downstream of integrins. PLoS One 7:e37830