Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

EAG2 potassium channel with evolutionarily conserved function as a brain tumor target

Abstract

Over 20% of the drugs for treating human diseases target ion channels, but no cancer drug approved by the US Food and Drug Administration (FDA) is intended to target an ion channel. We found that the EAG2 (Ether-a-go-go 2) potassium channel has an evolutionarily conserved function for promoting brain tumor growth and metastasis, delineate downstream pathways, and uncover a mechanism for different potassium channels to functionally cooperate and regulate mitotic cell volume and tumor progression. EAG2 potassium channel was enriched at the trailing edge of migrating medulloblastoma (MB) cells to regulate local cell volume dynamics, thereby facilitating cell motility. We identified the FDA-approved antipsychotic drug thioridazine as an EAG2 channel blocker that reduces xenografted MB growth and metastasis, and present a case report of repurposing thioridazine for treating a human patient. Our findings illustrate the potential of targeting ion channels in cancer treatment.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Drosophila Eag channel deficiency reduces brain tumor growth and metastasis.
Figure 2: Cross-species transcriptomic studies identified KCNT2 as a potassium channel that is enriched in SHH-MB and regulates tumor growth.
Figure 3: KCNT2 and EAG2 functionally cooperate to regulate mitotic cell volume.
Figure 4: EAG2 channel promotes MB metastasis and enriches to the trailing edge to regulate MB cell motility.
Figure 5: Thioridazine is an EAG2 blocker that reduces MB cell viability and mobility.
Figure 6: Thioridazine inhibition of EAG2 channel reduces intracranial MB growth and metastasis.
Figure 7: A subset of metastasized MB tumors display EAG2 upregulation compared with matched primary tumors.
Figure 8: A case report of thioridazine treatment of a patient with metastatic SHH-MB.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Thompson, M.C. et al. Genomics identifies medulloblastoma subgroups that are enriched for specific genetic alterations. J. Clin. Oncol. 24, 1924–1931 (2006).

    CAS  PubMed  Google Scholar 

  2. Kool, M. et al. Integrated genomics identifies five medulloblastoma subtypes with distinct genetic profiles, pathway signatures and clinicopathological features. PLoS ONE 3, e3088 (2008).

    PubMed  PubMed Central  Google Scholar 

  3. Cho, Y.J. et al. Integrative genomic analysis of medulloblastoma identifies a molecular subgroup that drives poor clinical outcome. J. Clin. Oncol. 29, 1424–1430 (2011).

    PubMed  Google Scholar 

  4. Northcott, P.A. et al. Medulloblastoma comprises four distinct molecular variants. J. Clin. Oncol. 29, 1408–1414 (2011).

    PubMed  Google Scholar 

  5. Goodrich, L.V., Milenkovic, L., Higgins, K.M. & Scott, M.P. Altered neural cell fates and medulloblastoma in mouse patched mutants. Science 277, 1109–1113 (1997).

    CAS  PubMed  Google Scholar 

  6. Yang, Z.J. et al. Medulloblastoma can be initiated by deletion of Patched in lineage-restricted progenitors or stem cells. Cancer Cell 14, 135–145 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Schüller, U. et al. Acquisition of granule neuron precursor identity is a critical determinant of progenitor cell competence to form Shh-induced medulloblastoma. Cancer Cell 14, 123–134 (2008).

    PubMed  PubMed Central  Google Scholar 

  8. Hatton, B.A. et al. The Smo/Smo model: hedgehog-induced medulloblastoma with 90% incidence and leptomeningeal spread. Cancer Res. 68, 1768–1776 (2008).

    CAS  PubMed  Google Scholar 

  9. Romer, J. & Curran, T. Targeting medulloblastoma: small-molecule inhibitors of the Sonic Hedgehog pathway as potential cancer therapeutics. Cancer Res. 65, 4975–4978 (2005).

    CAS  PubMed  Google Scholar 

  10. Rudin, C.M. et al. Treatment of medulloblastoma with hedgehog pathway inhibitor GDC-0449. N. Engl. J. Med. 361, 1173–1178 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Yauch, R.L. et al. Smoothened mutation confers resistance to a Hedgehog pathway inhibitor in medulloblastoma. Science 326, 572–574 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Wu, X. et al. Clonal selection drives genetic divergence of metastatic medulloblastoma. Nature 482, 529–533 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Schönherr, R. Clinical relevance of ion channels for diagnosis and therapy of cancer. J. Membr. Biol. 205, 175–184 (2005).

    PubMed  Google Scholar 

  14. Pardo, L.A. Voltage-gated potassium channels in cell proliferation. Physiology (Bethesda) 19, 285–292 (2004).

    CAS  Google Scholar 

  15. Fraser, S.P. & Pardo, L.A. Ion channels: functional expression and therapeutic potential in cancer. Colloquium on ion channels and cancer. EMBO Rep. 9, 512–515 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Prevarskaya, N., Skryma, R. & Shuba, Y. Ion channels and the hallmarks of cancer. Trends Mol. Med. 16, 107–121 (2010).

    CAS  PubMed  Google Scholar 

  17. Huang, X. & Jan, L.Y. Targeting potassium channels in cancer. J. Cell Biol. 206, 151–162 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Homem, C.C. & Knoblich, J.A. Drosophila neuroblasts: a model for stem cell biology. Development 139, 4297–4310 (2012).

    CAS  PubMed  Google Scholar 

  19. Zhu, S. et al. The bHLH repressor Deadpan regulates the self-renewal and specification of Drosophila larval neural stem cells independently of Notch. PLoS ONE 7, e46724 (2012).

    PubMed  PubMed Central  Google Scholar 

  20. Bowman, S.K. et al. The tumor suppressors Brat and Numb regulate transit-amplifying neuroblast lineages in Drosophila. Dev. Cell 14, 535–546 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Richter, C., Oktaba, K., Steinmann, J., Muller, J. & Knoblich, J.A. The tumour suppressor L(3)mbt inhibits neuroepithelial proliferation and acts on insulator elements. Nat. Cell Biol. 13, 1029–1039 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Northcott, P.A. et al. Multiple recurrent genetic events converge on control of histone lysine methylation in medulloblastoma. Nat. Genet. 41, 465–472 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Gonzalez, C. Drosophila melanogaster: a model and a tool to investigate malignancy and identify new therapeutics. Nat. Rev. Cancer 13, 172–183 (2013).

    CAS  PubMed  Google Scholar 

  24. Huang, X. et al. Voltage-gated potassium channel EAG2 controls mitotic entry and tumor growth in medulloblastoma via regulating cell volume dynamics. Genes Dev. 26, 1780–1796 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Saini, N. & Reichert, H. Neural stem cells in Drosophila: molecular genetic mechanisms underlying normal neural proliferation and abnormal brain tumor formation. Stem Cells Int. 2012, 486169 (2012).

    PubMed  PubMed Central  Google Scholar 

  26. Reimand, J., Arak, T. & Vilo, J. g:Profiler–a web server for functional interpretation of gene lists (2011 update). Nucleic Acids Res. 39, W307–W315 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Merico, D., Isserlin, R., Stueker, O., Emili, A. & Bader, G.D. Enrichment map: a network-based method for gene-set enrichment visualization and interpretation. PLoS ONE 5, e13984 (2010).

    PubMed  PubMed Central  Google Scholar 

  28. Bhattacharjee, A. et al. Slick (Slo2.1), a rapidly-gating sodium-activated potassium channel inhibited by ATP. J. Neurosci. 23, 11681–11691 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Kaczmarek, L.K. Slack, Slick and sodium-activated potassium channels. ISRN Neurosci. 2013, 354262 (2013).

    PubMed  PubMed Central  Google Scholar 

  30. Hage, T.A. & Salkoff, L. Sodium-activated potassium channels are functionally coupled to persistent sodium currents. J. Neurosci. 32, 2714–2721 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Rutkowski, S. et al. Survival and prognostic factors of early childhood medulloblastoma: an international meta-analysis. J. Clin. Oncol. 28, 4961–4968 (2010).

    PubMed  Google Scholar 

  32. García-Ferreiro, R.E. et al. Mechanism of block of hEag1 K+ channels by imipramine and astemizole. J. Gen. Physiol. 124, 301–317 (2004).

    PubMed  PubMed Central  Google Scholar 

  33. Herzberg, I.M., Trudeau, M.C. & Robertson, G.A. Transfer of rapid inactivation and sensitivity to the class III antiarrhythmic drug E-4031 from HERG to M-eag channels. J. Physiol. (Lond.) 511, 3–14 (1998).

    CAS  Google Scholar 

  34. Preskorn, S.H. Antipsychotic drug development in the pre-human-genome era: a full circle. J. Psychiatr. Pract. 7, 209–213 (2001).

    CAS  PubMed  Google Scholar 

  35. Vanderheeren, F.A. & Muusze, R.G. Plasma levels and half lives of thioridazine and some of its metabolites. I. High doses in young acute schizophrenics. Eur. J. Clin. Pharmacol. 11, 135–140 (1977).

    CAS  PubMed  Google Scholar 

  36. Svendsen, C.N., Hrbek, C.C., Casendino, M., Nichols, R.D. & Bird, E.D. Concentration and distribution of thioridazine and metabolites in schizophrenic post-mortem brain tissue. Psychiatry Res. 23, 1–10 (1988).

    CAS  PubMed  Google Scholar 

  37. Willoughby, L.F. et al. An in vivo large-scale chemical screening platform using Drosophila for anti-cancer drug discovery. Dis. Model. Mech. 6, 521–529 (2013).

    CAS  PubMed  Google Scholar 

  38. Habela, C.W. & Sontheimer, H. Cytoplasmic volume condensation is an integral part of mitosis. Cell Cycle 6, 1613–1620 (2007).

    CAS  PubMed  Google Scholar 

  39. Boucrot, E. & Kirchhausen, T. Mammalian cells change volume during mitosis. PLoS ONE 3, e1477 (2008).

    PubMed  PubMed Central  Google Scholar 

  40. Schwab, A., Fabian, A., Hanley, P.J. & Stock, C. Role of ion channels and transporters in cell migration. Physiol. Rev. 92, 1865–1913 (2012).

    CAS  PubMed  Google Scholar 

  41. Jakab, M. & Ritter, M. Cell volume regulatory ion transport in the regulation of cell migration. Contrib. Nephrol. 152, 161–180 (2006).

    CAS  PubMed  Google Scholar 

  42. Schwab, A., Hanley, P., Fabian, A. & Stock, C. Potassium channels keep mobile cells on the go. Physiology (Bethesda) 23, 212–220 (2008).

    CAS  Google Scholar 

  43. Schwab, A. et al. Polarized ion transport during migration of transformed Madin-Darby canine kidney cells. Pflugers Arch. 430, 802–807 (1995).

    CAS  PubMed  Google Scholar 

  44. Schneider, S.W. et al. Volume dynamics in migrating epithelial cells measured with atomic force microscopy. Pflugers Arch. 439, 297–303 (2000).

    CAS  PubMed  Google Scholar 

  45. Schwab, A. et al. Subcellular distribution of calcium-sensitive potassium channels (IK1) in migrating cells. J. Cell. Physiol. 206, 86–94 (2006).

    CAS  PubMed  Google Scholar 

  46. Ward, R.J. et al. Multipotent CD15+ cancer stem cells in patched-1-deficient mouse medulloblastoma. Cancer Res. 69, 4682–4690 (2009).

    CAS  PubMed  Google Scholar 

  47. Huang, X., Ketova, T., Litingtung, Y. & Chiang, C. Isolation, enrichment, and maintenance of medulloblastoma stem cells. J. Vis. Exp. published online, 2086 (1 September 2010).

  48. Berman, D.M. et al. Medulloblastoma growth inhibition by hedgehog pathway blockade. Science 297, 1559–1561 (2002).

    CAS  PubMed  Google Scholar 

  49. Corno, D. et al. Gene signatures associated with mouse postnatal hindbrain neural stem cells and medulloblastoma cancer stem cells identify novel molecular mediators and predict human medulloblastoma molecular classification. Cancer Discov. 2, 554–568 (2012).

    PubMed  Google Scholar 

  50. Lee, H.Y., Greene, L.A., Mason, C.A. & Manzini, M.C. Isolation and culture of post-natal mouse cerebellar granule neuron progenitor cells and neurons. J. Vis. Exp. published online, doi:10.3791/99010.3791/990 (16 January 2009).

  51. Wallace, K., Liu, T.H. & Vaessin, H. The pan-neural bHLH proteins DEADPAN and ASENSE regulate mitotic activity and cdk inhibitor dacapo expression in the Drosophila larval optic lobes. Genesis 26, 77–85 (2000).

    CAS  PubMed  Google Scholar 

  52. Wang, Z., Wilson, G.F. & Griffith, L.C. Calcium/calmodulin-dependent protein kinase II phosphorylates and regulates the Drosophila eag potassium channel. J. Biol. Chem. 277, 24022–24029 (2002).

    CAS  PubMed  Google Scholar 

  53. Broughton, S.J., Kitamoto, T. & Greenspan, R.J. Excitatory and inhibitory switches for courtship in the brain of Drosophila melanogaster. Curr. Biol. 14, 538–547 (2004).

    CAS  PubMed  Google Scholar 

  54. Vahasoyrinki, M., Niven, J.E., Hardie, R.C., Weckstrom, M. & Juusola, M. Robustness of neural coding in Drosophila photoreceptors in the absence of slow delayed rectifier K+ channels. J. Neurosci. 26, 2652–2660 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Janic, A., Mendizabal, L., Llamazares, S., Rossell, D. & Gonzalez, C. Ectopic expression of germline genes drives malignant brain tumor growth in Drosophila. Science 330, 1824–1827 (2010).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank Y. Song, W.-P. Ge, S.-B. Yang, H. Yang, C. Peters, B. Piggott, S. Morrissy, D. Shih, B.K. Shoichet and all of the members of the Jan laboratory for constructive suggestions. We thank C. Chiang for critical reading of the manuscript. This work was supported by the GEMS-CTSI postdoctoral award from the Howard Hughes Medical Institute and UCSF, and the Damon Runyon Cancer Research Foundation Fellowship to X.H. (Kandis Ann Ulrich-Carleton Fellow), NIH grant P41 GM103504 to G.D.B., the Pediatric Brain Tumor Foundation and R01 grants CA133091, CA148699 and CA159859 to W.A.W. and M.D.T., the Garron Family Chair in Childhood Cancer Research at The Hospital for Sick Children and The University of Toronto, operating funds from the Canadian Institutes of Health Research, the Terry Fox Research Institute, and the Pediatric Brain Tumor Foundation to M.D.T., NIH grants R37NS040929 to Y.N.J., and R37MH065334 and R01CA185039 to L.Y.J. Y.N.J. and L.Y.J. are Howard Hughes Medical Institute investigators.

Author information

Authors and Affiliations

Authors

Contributions

X.H. and L.Y.J. conceived and designed the study. X.H., Y.H., A.M.D., R.H., W.Z., J.R., H.Y., T.A.W., S.J.S., S.Y., S.B., S.Z., M.K.C., J.P., V.R., L.G., X.W., M.R., C.M.F., C.C.K., G.D.B. and S.M. contributed to methodology and data acquisition. X.H., Y.H., A.M.D., W.Z., J.R., S.Y., J.P., V.R., C.M.F., C.C.K., W.A.W., C.D.J., M.A.S., G.D.B., S.M., M.D.T., Y.N.J. and L.Y.J. analyzed and interpreted the data. X.H., Y.N.J. and L.Y.J. wrote and revised the manuscript.

Corresponding authors

Correspondence to Yuh Nung Jan or Lily Yeh Jan.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 eag loss-of-function mutation reduces the growth of brain tumors induced by reduction of tumor suppressor Brat activity, and transplantation assay to study Drosophila brain tumor metastasis

a, External view of the control brains, insc-Gal4-driven bratRNAi-induced brain tumors and bratRNAi-induced brain tumors with eag loss-of-function (eag1) at 25oC (n = 9 and 8 for bratRNAi and bratRNAi; eag1, respectively, P = 0.0002, two-tailed Student’s t-test) and 29oC (n = 10 and 10 for bratRNAi and bratRNAi; eag1, respectively, P < 0.0001, two-tailed Student’s t-test). b, GFP-labeled, dpn-overexpressing primary brain lobe tumors were retrieved from 3rd instar larvae. The tumors were micro-dissected into small pieces with equal volumes. Young female adult hosts were anesthetized with CO2 and a piece of larval brain was picked up with the tip of a glass needle and injected tangentially into the mid-ventral part of the abdomen. Implanted hosts were kept at 25°C for 10 days and monitored for tumor growth in the abdomen and metastasis into the brain. Tumor formation or metastasis was detected in hosts transplanted with dpn-overexpressing tumor cells, and the tumor-transplanted hosts displayed significantly shortened survival compared to the hosts transplanted with normal brain cells retrieved from w- control larvae (n = 12 and 20 for control and dpnO/E, respectively, log-rank Kaplan-Meier test).

Supplementary Figure 2 Mutation status of KCNT2 across different cancer types and analysis of KCNT2 knockdown in MB cells

a, Analysis of large-scale cancer genomics datasets shows frequent KCNT2 gene amplification and mutations in different cancer types. Data analysis and visualization are performed using the cBioPortal Cancer Genomics website (http://www.cbioportal.org). b, Lentivirus-mediated RNAi knockdown of KCNT2 reduces the clonogenic growth of MB cells. c, KCNT2 RNAi treatment in MB cells significantly reduces KCNT2 transcript level (n = 3 independent cultures and qPCR experiments, two-tailed Student’s t-test). Error bars show ± SEM.

Supplementary Figure 3 Working model: co-option of potassium channel activity regulates mitotic cell volume in MB

Left: Potassium channel activity is increased in MB cells during late G2 and mitotic phases, which regulates cell volume reduction. Right: EAG2, which localizes to the plasma membrane at late G2 and during mitosis, regulates KCNT2 mRNA expression. KCNT2 channel enriches to the plasma membrane beginning at metaphase to cooperate with EAG2 and regulate mitotic volume dynamics. EAG2 channel block by astemizole or thioridazine, or indirect inhibition of KCNT2 channel using riluzole, represents viable strategy to inhibit MB tumor growth by targeting potassium channels.

Supplementary Figure 4 EAG2 channel localizes to the trailing membrane during MB cell migration and regulates migratory polarity and cell motility

Immunofluorescence staining using EAG2 antibodies obtained from different commercial sources shows cell rear and trailing membrane enrichment during migration of multiple human MB cell lines, including SF8953-MB, DAOY, Vandy-MB-11, Vandy-MB-6 (at passage 3) and Vandy-MB-10 (at passage 3) cells. White arrows indicate migratory direction.

Supplementary Figure 5 KCNT2 channel does not enrich to the trailing edge during MB cell migration and does not regulate migratory polarity or cell motility

a, EAG2, but not KCNT2, displays cell rear and trailing edge enrichment during MB cell migration. b, KCNT2 knockdown does not considerably affect MB cell migratory polarization or the formation of lamellipodia (n = 482 and 391 randomly selected cells for Scr. shRNA and KCNT2 shRNA, respectively). c, 3-D reconstruction shows rear cell body contraction in control Vandy-MB-11 cells and cells with KCNT2 knockdown. d, Control Vandy-MB-11 cells and cells with KCNT2 knockdown show comparable migratory velocity (n = 20 for each group, two-tailed Student’s t-test). Each colored trace represents the migratory route of an individual Vandy-MB-11 cell over 5 hours. e, 5 μM thioridazine treatment for 3 days results in significantly reduced KCNT2 expression in Vandy-MB-11 cells (n = 4 independent cultures and qPCR experiments, two-tailed Student’s t-test). Error bars show ± SEM.

Supplementary Figure 6 Promazine blocks EAG2 channel and inhibits MB cell growth

a, Chemical structure of promazine. b, Promazine treatment markedly inhibits MB cell growth at 10 μM. c, Patch clamp recording shows that promazine significantly blocks EAG2 channel at 10 μM (n = 3, two-tailed Student’s t-test). Error bars show ± SEM.

Supplementary Figure 7 Working model: localized EAG2 potassium channel activity at the trailing edge regulates MB cell motility

EAG2 channel enriched at the trailing edge regulates local potassium efflux, which accompanies water loss to promote cell rear contraction and cell motility.

Supplementary Figure 8 Astemizole treatment of mice bearing subcutaneously allografted Math1-Cre; SmoM2 MB tumors

a, Astemizole treatment reduces MB cell clonal growth as neurospheres (Math1-Cre; SmoM2 cells) or monolayer colonies (Vandy-MB-11, DAOY and Ptch1+/-; p53-/- cells). b, Experimental design for acute allografting of Math1-Cre; SmoM2 MB tumors into the flanks of nu/nu BALB/C mice and astemizole treatment by oral gavage. Once palpable tumors are detected one-week post implantation, daily oral gavage of 50 mg/kg astemizole to the mice was performed for two weeks. c, 14 days of daily oral gavage of astemizole reduces proliferation and increases apoptosis in subcutaneously allografted Math1-Cre; SmoM2 tumors. d, Quantifications of tumor volumes show significantly inhibited tumor growth with astemizole treatment (n = 9 for vehicle and astemizole group, Two-Way ANOVA). Error bars show ± SEM.

Supplementary Figure 9 Full-length Western blotting images for cropped images shown in Fig. 6b

The full-length Western blotting images for the cropped images in Fig. 6b are shown with molecular weights indicated by protein ladder. The same membrane was first blotted with EAG2 antibody, followed by antibody stripping and re-blotting with α tubulin antibody. Chemiluminescence was imaged using Molecular Imager@VersaDoc MP4000 system (Bio-Rad).

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Huang, X., He, Y., Dubuc, A. et al. EAG2 potassium channel with evolutionarily conserved function as a brain tumor target. Nat Neurosci 18, 1236–1246 (2015). https://doi.org/10.1038/nn.4088

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.4088

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer