Voltage-gated potassium channel EAG2 controls mitotic entry and tumor growth in medulloblastoma via regulating cell volume dynamics

  1. Lily Yeh Jan1,2,3,13
  1. 1Howard Hughes Medical Institute,
  2. 2Department of Physiology,
  3. 3Department of Biophysics and Biochemistry, University of California at San Francisco, San Francisco, California 94158, USA;
  4. 4Division of Neurosurgery, Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada;
  5. 5Department of Neurological Surgery,
  6. 6Department of Urology, UCSF Helen Diller Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94158, USA;
  7. 7Department of Cell and Developmental Biology,
  8. 8Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA;
  9. 9Department of Pathology, UCSF Helen Diller Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94158, USA;
  10. 10Howard Hughes Medical Institute,
  11. 11Department of Neurological Surgery, University of California at San Francisco, San Francisco, California 94143, USA;
  12. 12Department of Medicine, UCSF Helen Diller Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94158, USA

    Abstract

    Medulloblastoma (MB) is the most common pediatric CNS malignancy. We identify EAG2 as an overexpressed potassium channel in MBs across different molecular and histological subgroups. EAG2 knockdown not only impairs MB cell growth in vitro, but also reduces tumor burden in vivo and enhances survival in xenograft studies. Mechanistically, we demonstrate that EAG2 protein is confined intracellularly during interphase but is enriched in the plasma membrane during late G2 phase and mitosis. Disruption of EAG2 expression results in G2 arrest and mitotic catastrophe associated with failure of premitotic cytoplasmic condensation. While the tumor suppression function of EAG2 knockdown is independent of p53 activation, DNA damage checkpoint activation, or changes in the AKT pathway, this defective cell volume control is specifically associated with hyperactivation of the p38 MAPK pathway. Inhibition of the p38 pathway significantly rescues the growth defect and G2 arrest. Strikingly, ectopic membrane expression of EAG2 in cells at interphase results in cell volume reduction and mitotic-like morphology. Our study establishes the functional significance of EAG2 in promoting MB tumor progression via regulating cell volume dynamics, the perturbation of which activates the tumor suppressor p38 MAPK pathway, and provides clinical relevance for targeting this ion channel in human MBs.

    Keywords

    Footnotes

    • Received April 6, 2012.
    • Accepted July 9, 2012.
    | Table of Contents

    Life Science Alliance