Identification of a DNA-binding site and transcriptional target for the EWS–WT1(+KTS) oncoprotein

  1. Paul A. Reynolds1,
  2. Gromoslaw A. Smolen1,
  3. Rachel E. Palmer1,
  4. Dennis Sgroi2,
  5. Vijay Yajnik1,
  6. William L. Gerald3, and
  7. Daniel A. Haber1,4
  1. 1 Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, Massachusetts 02129, USA
  2. 2 Molecular Pathology Unit, Massachusetts General Hospital, Charlestown, Massachusetts 02129, USA
  3. 3 Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA

Abstract

Desmoplastic small round cell tumor (DSRCT) is defined by a chimeric transcription factor, resulting from fusion of the N-terminal domain of the Ewing's sarcoma gene EWS to the three C-terminal zinc fingers of the Wilms' tumor suppressor WT1. Although DNA-binding sites have been defined for the uninterrupted WT1 zinc finger domains, the most prevalent isoforms of both WT1 and EWS–WT1 have an insertion of three amino acids [lysine, threonine, and serine (KTS)], which abrogates binding to known consensus sequences and transactivation of known target genes. Here, we used cDNA subtractive hybridization to identify an endogenous gene, LRRC15, which is specifically up-regulated after inducible expression of EWS–WT1(+KTS) in cancer cell lines, and is expressed within primary DSRCT cells. The chimeric protein binds in vitro and in vivo to a specific element upstream of LRRC15, leading to dramatic transcriptional activation. Mutagenesis studies define the optimal binding site of the (+KTS) isoform of EWS–WT1 as 5′-GGAGG(A/G)-3′. LRRC15 encodes a leucine-rich transmembrane protein, present at the leading edge of migrating cells, the expression of which in normal tissues is restricted to the invasive cytotrophoblast layer of the placenta; small interfering (siRNA)-mediated suppression of LRRC15 expression in breast cancer cells leads to abrogation of invasiveness in vitro. Together, these observations define the consequence of (KTS) insertion within WT1-derived zinc fingers, and identify a novel EWS–WT1 transcriptional target implicated in tumor invasiveness.

Keywords

Footnotes

  • Article published online ahead of print. Article and publication date are at http://www.genesdev.org/cgi/doi/10.1101/gad.1110703.

  • 4 Corresponding author.

    4 E-MAIL Haber{at}helix.mgh.harvard.edu; FAX (617) 724-6919.

    • Accepted July 1, 2003.
    • Received May 7, 2003.
| Table of Contents

Life Science Alliance