Abstract

We previously reported overexpression of Prostate derived Ets transcription factor (PDEF) in breast cancer and its role in breast cancer progression, supporting PDEF as an attractive target in this cancer. The goal of this research was to identify specific PDEF induced molecules that, like PDEF, show overexpression in breast tumors and a role in breast tumor progression. PDEF expression was down regulated by shRNA in MCF-7 human breast tumor cell line, and probes from PDEF down-regulated and control MCF-7 cells were used to screen the HG-U133A human gene chips. These analyses identified 1318 genes that were induced two-fold or higher by PDEF in MCF-7 cells. Further analysis of three of these genes, namely CEACAM6, S100A7 and B7-H4, in relation to PDEF in primary breast tumors showed that in 82% of ER+, 67% of Her2 overexpressing and 24% of triple-negative breast tumors both PDEF and CEACAM6 expression was elevated 10-fold or higher in comparison to normal breast tissue. Overall, 72% (94 of 131) of the primary breast tumors showed 10-fold or higher expression of both PDEF and CEACAM6. In contrast, S100A7 and B7-H4 failed to show concordant elevated expression with PDEF in primary tumors. To determine the significance of elevated PDEF and CEACAM6 expression to tumor phenotype, their expression was down regulated by specific siRNAs in human breast tumor cell lines. This resulted in the loss of viability of tumor cells in vitro, supporting an oncogenic role for both PDEF and CEACAM6 in breast cancer. Together, these findings show that PDEF-CEACAM6 is a highly active oncogenic axis in breast cancer and suggest that targeting of these molecules should provide novel treatments for most breast cancer patients.

Highlights

  • Targeted treatments of ER+ and Her2 overexpressing breast tumors have significantly improved the clinical outcomes for most breast cancer patients [1, 2]

  • Prostate derived Ets transcription factor (PDEF) expression was stably down-regulated in MCF-7 breast tumor cell line by transfection with a plasmid encoding a PDEF specific small hairpin RNA (shRNA) sequence

  • The down-regulation of PDEF expression was confirmed by RT/PCR and the data are shown in Figure 1, Panel 1A

Read more

Summary

Introduction

Targeted treatments of ER+ and Her overexpressing breast tumors have significantly improved the clinical outcomes for most breast cancer patients [1, 2]. Transfection of PDEF into the immortalized MCF-10A and MCF-12A breast epithelial cell lines led to increased clonogenicity in vitro and tumorigenicity in immunodeficient mice; and meta-analysis of PDEF expression in relation to clinical outcome showed a significant association of high PDEF expression with poor disease-free and overall survival in independent patient cohorts [16, 18]. These observations established PDEF as a novel oncogene and an attractive target in breast cancer

Objectives
Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call