Oncotarget

Research Papers:

The silencing of the SWI/SNF subunit and anticancer gene BRM in Rhabdoid tumors

Bhaskar Kahali, Jinlong Yu, Stefanie B. Marquez, Kenneth W. Thompson, Shermi Y. Liang, Li Lu and David Reisman _

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2014; 5:3316-3332. https://doi.org/10.18632/oncotarget.1945

Metrics: PDF 2456 views  |   HTML 3778 views  |   ?  


Abstract

Bhaskar Kahali1, Jinlong Yu1, Stefanie B. Marquez1, Kenneth. W. Thompson1, Shermi Y. Liang1, Li Lu2 and David Reisman1

1 Division of Hematology/Oncology, Department of Medicine, University of Florida, Florida, USA

2 Department of Pathology, University of Florida, Florida, USA

Correspondence:

David Reisman, email:

Keywords: SWI/SNF, Brahma-Related Gene 1, chromatin remodeling, Brahma, Rhabdoid

Received: March 18, 2014 Accepted: May 3, 2014 Published: May 4, 2014

Abstract

Rhabdoid sarcomas are highly malignant tumors that usually occur in young children. A key to the genesis of this tumor is the mutational loss of the BAF47 gene as well as the widespread epigenetic suppression of other key anticancer genes. The BRM gene is one such epigenetically silenced gene in Rhabdoid tumors. This gene codes for an ATPase catalytic subunit that shifts histones and opens the chromatin. We show that BRM is an epigenetically silenced gene in 10/11 Rhabdoid cell lines and in 70% of Rhabdoid tumors. Moreover, BRM can be induced by BAF47 re-expression and by Flavopiridol. By selective shRNAi knockdown of BRM, we show that BRM re-expression is necessary for growth inhibition by BAF47 re-expression or Flavopiridol application. Similar to lung cancer cell lines, we found that HDAC3, HDAC9, MEF2D and GATA3 controlled BRM silencing and that HDAC9 was overexpressed in Rhabdoid cancer cell lines. In primary BRM-deficient Rhabdoid tumors, HDAC9 was also found to be highly overexpressed. Two insertional BRM promoter polymorphisms contribute to BRM silencing, but only the -1321 polymorphism correlated with BRM silencing in Rhabdoid cell lines. To determine how these polymorphisms were tied to BRM silencing, we conducted ChIP assays and found that both HDAC9 and MEF2D bound to the BRM promoter at or near these polymorphic sites. Using BRM promoter swap experiments, we indirectly showed that both HDAC9 and MEF2D bound to these polymorphic sites. Together, these data show that the mechanism of BRM silencing contributes to the pathogenesis of Rhabdoid tumors and appears to be conserved among tumor types.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 1945