Genome-wide RNAi screens in human brain tumor isolates reveal a novel viability requirement for PHF5A

  1. Patrick J. Paddison2,14
  1. 1Clinical Research Division,
  2. 2Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA;
  3. 3Molecular and Cellular Biology Program, University of Washington, Seattle, Washington 98195, USA;
  4. 4Computational Biology Program, Public Health Sciences Division,
  5. 5Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA;
  6. 6Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado 80523, USA;
  7. 7Computational Biology Shared Resource, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA;
  8. 8Department of Pharmacology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98195, USA;
  9. 9Genomics Shared Resource, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA;
  10. 10High Throughput Chemistry Facility, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA;
  11. 11Cancer Institute, University College London, London WC1E 6BT, United Kingdom;
  12. 12Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44192, USA
    1. 13 These authors contributed equally to this work.

    Abstract

    To identify key regulators of human brain tumor maintenance and initiation, we performed multiple genome-wide RNAi screens in patient-derived glioblastoma multiforme (GBM) stem cells (GSCs). These screens identified the plant homeodomain (PHD)-finger domain protein PHF5A as differentially required for GSC expansion, as compared with untransformed neural stem cells (NSCs) and fibroblasts. Given PHF5A's known involvement in facilitating interactions between the U2 snRNP complex and ATP-dependent helicases, we examined cancer-specific roles in RNA splicing. We found that in GSCs, but not untransformed controls, PHF5A facilitates recognition of exons with unusual C-rich 3′ splice sites in thousands of essential genes. PHF5A knockdown in GSCs, but not untransformed NSCs, astrocytes, or fibroblasts, inhibited splicing of these genes, leading to cell cycle arrest and loss of viability. Notably, pharmacologic inhibition of U2 snRNP activity phenocopied PHF5A knockdown in GSCs and also in NSCs or fibroblasts overexpressing MYC. Furthermore, PHF5A inhibition compromised GSC tumor formation in vivo and inhibited growth of established GBM patient-derived xenograft tumors. Our results demonstrate a novel viability requirement for PHF5A to maintain proper exon recognition in brain tumor-initiating cells and may provide new inroads for novel anti-GBM therapeutic strategies.

    Keywords

    Footnotes

    • Received December 19, 2012.
    • Accepted April 4, 2013.
    | Table of Contents

    Life Science Alliance