The goal of cancer biology is to understand the progression of genetic and epigenetic changes occurring in cells as they become tumorigenic. Many studies have demonstrated a critical role for CXCR4 in guiding metastasizing breast cancer cells to sites with high levels of its ligand SDF1 (CXCL12). In contrast, relatively few studies have explored the consequences of having this pathway deregulated early in lesion development, even though a large fraction (>90%) of human breast tumors display inappropriate CXCR4 expression at very early stages of transformation. The observation that CXCR4 is upregulated early in the disease process, combined with recent studies demonstrating that carcinoma associated fibroblasts serve as a local source of its ligand SDF1, have fueled speculation that CXCR4 may also play a pivotal role in primary tumor growth. We have identified such a disease signature in hyperplastic lesions of mammary glands harboring loss-of-function in Slit2 and Slit3 or its Robo1 receptor, giving us the unique opportunity to understand the function of CXCR4 and SDF1 in the early stages of breast transformation within the integrated physiology of an animal model. Our preliminary data demonstrate that loss-of-function mutations in Slits or their Robo1 receptor lead to loss of tissue organization, elevated proliferation and a host of changes in the microenvironment, including increased angiogenesis. Based on our preliminary data, the overall hypothesis of the application is that loss of SLIT/ROBO1 signaling in breast leads to upregulated SDF1/CXCR4, which, in turn, contributes to epithelial transformation and generation of the tumor microenvironment. To address this hypothesis, the Specific Aims of this proposal are three-fold.
In Aim I, we propose to investigate the role SDF1/CXCR4 signaling plays in early lesion development by determining the signaling status of CXCR4 in Slit2-/-;Slit3-/- and Robo1-/- tissue using both gain- and loss-of-function approaches. We will also evaluate whether similar expression changes occur between Slits, Robo1 and Cxcr4 in human breast tumors.
In Aim II, we propose to define the cross talk that occurs between mammary epithelia and stroma by generating glands in which SLIT/ROBO1 signaling is selectively eliminated in the epithelia or stroma.
In Aim III, we investigate the pro-angiogenic environment that arises in the absence of SLIT/ROBO1 signaling. We propose to elucidate the role of SDF1, alone, in promoting neoangiogenesis by eliminating VEGF activation in the Robo1-null background. We also propose to explore the role of Slit2 and Slit3 as tumor suppressors by re-expressing the genes in breast cancer cells and evaluating the effects on tumor growth and tumor angiogenesis. In summary, we have identified SDF1/CXCR4 as key, downstream regulatory targets of SLIT/ROBO1 signaling in vivo. We propose to elucidate how misregulation of this chemokine axis orchestrates inappropriate interactions between cells and their environment, leading to transformation of the tissue and surrounding microenvironment.

Public Health Relevance

The genesis of breast cancer has remained elusive. Even though it is considered a heritable disease, it is estimated that only 5-10% of all human breast cancers are causally linked to known genetic mutations. Identifying genes whose mutations allow a lesion to progress and become malignant is crucial for identifying potential therapeutic targets. For a number of cancers, prominently breast, a candidate target with an established role in metastasis is the G-protein coupled receptor CXCR4. The goal of this application is to understand how CXCR4 contributes to tumor progression using breast as a model system.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
3R01CA128902-02S1
Application #
7914909
Study Section
Tumor Microenvironment Study Section (TME)
Program Officer
Ogunbiyi, Peter
Project Start
2008-07-01
Project End
2013-04-30
Budget Start
2009-05-01
Budget End
2010-04-30
Support Year
2
Fiscal Year
2009
Total Cost
$71,614
Indirect Cost
Name
University of California Santa Cruz
Department
Biochemistry
Type
Schools of Arts and Sciences
DUNS #
125084723
City
Santa Cruz
State
CA
Country
United States
Zip Code
95064
Macias, Hector; Hinck, Lindsay (2012) Mammary gland development. Wiley Interdiscip Rev Dev Biol 1:533-57
Hinck, Lindsay (2011) Tumor suppressors: heroes and villains? J Mammary Gland Biol Neoplasia 16:169-71
Harburg, Gwyndolen C; Hinck, Lindsay (2011) Navigating breast cancer: axon guidance molecules as breast cancer tumor suppressors and oncogenes. J Mammary Gland Biol Neoplasia 16:257-70
Macias, Hector; Moran, Angel; Samara, Yazeed et al. (2011) SLIT/ROBO1 signaling suppresses mammary branching morphogenesis by limiting basal cell number. Dev Cell 20:827-40
Smith-Berdan, Stephanie; Nguyen, Andrew; Hassanein, Deena et al. (2011) Robo4 cooperates with CXCR4 to specify hematopoietic stem cell localization to bone marrow niches. Cell Stem Cell 8:72-83
Marlow, Rebecca; Binnewies, Mikhail; Sorensen, Lise K et al. (2010) Vascular Robo4 restricts proangiogenic VEGF signaling in breast. Proc Natl Acad Sci U S A 107:10520-5
Marlow, Rebecca; Strickland, Phyllis; Lee, Ji Shin et al. (2008) SLITs suppress tumor growth in vivo by silencing Sdf1/Cxcr4 within breast epithelium. Cancer Res 68:7819-27