Many human tumors, including breast cancer, display a hierarchical organization in which a subset of tumor cells with stem cell properties drives tumor growth and metastasis. Recent studies have shown that cancer stem cells (CSCs) are regulated by intrinsic self-renewal pathways in addition to extrinsic inflammatory pathways. Overexpression of HER2 has been shown to increase the CSC population but the underlying molecular mechanism is still largely unknown. We have systematically dissected the HER2-downstream signaling pathways and found that the NFkB and STAT3 pathways are the most important for self-renewal of CSCs. Interestingly, activation of NFkB and STAT3 is not directly mediated by signaling molecules immediately downstream of HER2, such as MAPK or PI3K/AKT kinases, but instead is mediated by HER2-induced interleukin-1 (IL-1) and interleukin-6 (IL-6) cytokines. IL-1 is a well-known proinflammatory cytokine that can induce the expression of a cascade of cytokines including IL-6 to propagate and sustain inflammation. Therefore, we hypothesize that HER2 induction of IL-1 serves as an important link between inflammation and cancer stem cells that switches on a cascade of cytokines in both tumor and stromal cells to regulate the CSC population in vivo. To test this hypothesis, we will pursue the following specific aims.
In Aim 1, we will elucidate the molecular mechanism by which HER-2 regulates the expression of IL-1 and IL-6 in breast cancer cells. Using a series of biochemical approaches, we will first identify the key HER2-downstream signaling pathways responsible for induction of IL-1 expression in breast cancer cells and address whether IL1 in turn activates IL- 6 expression.
In aim 2, we will study the function and mechanism of HER2-induced IL-1 and IL-6 signaling in the maintenance and regulation of cancer stem cells. We will use recombinant proteins and knockdown cell models to study how the interplay between IL-1-induced NFkB signaling and IL-6-induced STAT3 signaling regulates the CSC population.
In aim 3, we will investigate the sources and functions of IL-1 and IL-6 in promotion of HER2-induced mammary tumorigenesis. We will cross MMTV-Her2 transgenic mice with IL-1 and IL-6 knockout mice models to study the functions of these two cytokines in Her2/neu-induced tumor onset and progression. Successful completion of this study will help us understand the mechanism and functions of HER2-induced IL1 and IL6 cytokines in the generation and maintenance of the CSC population both in vitro and in vivo.

Public Health Relevance

Genomic amplification or overexpression of HER2 occurs in 20-30% of breast cancers and correlates with poor prognosis. HER2-targeted drugs, such as HER2 antibody and kinase inhibitors, are effective in treating HER2-postitive breast cancer. However, most patients ultimately develop resistance to such drugs, necessitating the identification of new therapeutic strategies, such as targeting cancer stem cells. Our results indicate that the IL-1 and IL-6 cytokines play a critical role in HER2-induced expansion of cancer stem cells and tumorigenesis. Thus, the targeting of IL-1 or IL-6 offers an opportunity for the treatment of HER2-positive breast cancer.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA178386-03
Application #
8850406
Study Section
Tumor Cell Biology Study Section (TCB)
Program Officer
Yassin, Rihab R
Project Start
2013-08-08
Project End
2016-05-31
Budget Start
2015-06-01
Budget End
2016-05-31
Support Year
3
Fiscal Year
2015
Total Cost
Indirect Cost
Name
University of South Carolina at Columbia
Department
Biology
Type
Schools of Arts and Sciences
DUNS #
041387846
City
Columbia
State
SC
Country
United States
Zip Code
29208
Patel, Y; Soni, M; Awgulewitsch, A et al. (2018) Overexpression of miR-489 derails mammary hierarchy structure and inhibits HER2/neu-induced tumorigenesis. Oncogene :
Liu, Shou; Lee, Ji Shin; Jie, Chunfa et al. (2018) HER2 Overexpression Triggers an IL1? Proinflammatory Circuit to Drive Tumorigenesis and Promote Chemotherapy Resistance. Cancer Res 78:2040-2051
Soni, Mithil; Patel, Yogin; Markoutsa, Eleni et al. (2018) Autophagy, Cell Viability, and Chemoresistance Are Regulated By miR-489 in Breast Cancer. Mol Cancer Res 16:1348-1360
He, Huacheng; Altomare, Diego; Ozer, Ufuk et al. (2016) Cancer cell-selective killing polymer/copper combination. Biomater Sci 4:115-20
Patel, Yogin; Lee, Ji S; Chen, Hexin (2016) Clinicopathological Analysis of miRNA Expression in Breast Cancer Tissues by Using miRNA In Situ Hybridization. J Vis Exp :
Patel, Yogin; Shah, Nirav; Lee, Ji Shin et al. (2016) A novel double-negative feedback loop between miR-489 and the HER2-SHP2-MAPK signaling axis regulates breast cancer cell proliferation and tumor growth. Oncotarget 7:18295-308
Liu, Shou; Jin, Kideok; Hui, Yvonne et al. (2015) HOXB7 promotes malignant progression by activating the TGF? signaling pathway. Cancer Res 75:709-19
Tashkandi, Hossam; Shah, Nirav; Patel, Yogin et al. (2015) Identification of new miRNA biomarkers associated with HER2-positive breast cancers. Oncoscience 2:924-9
Remant, Bahadur K C; Chandrashekaran, Varun; Cheng, Bei et al. (2014) Redox potential ultrasensitive nanoparticle for the targeted delivery of camptothecin to HER2-positive cancer cells. Mol Pharm 11:1897-905
Johnson, Sara; Chen, Hexin; Lo, Pang-Kuo (2013) Isolation of Cancer Epithelial Cells from Mouse Mammary Tumors. Bio Protoc 3:

Showing the most recent 10 out of 11 publications