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.

  • Review Article
  • Published:

The roles of K+ channels in cancer

Key Points

  • Ion transport molecules are involved in many physiological and pathological processes.

  • Among ion transporters, potassium channels show the highest variability and the most frequently altered expression in many tumour types.

  • Cell cycle and proliferation, cell migration, invasion and apoptosis are all processes that can be modified by the expression of potassium channels.

  • Permeation-dependent and permeation-independent mechanisms contribute to the roles of potassium channels in oncological processes.

  • The experience gained with pharmacological manipulation of potassium channels in other pathologies might facilitate the use of potassium channels as cancer targets.

Abstract

Potassium channels are transmembrane proteins that selectively facilitate the flow of potassium ions down an electrochemical gradient. These molecules have been studied in great detail in the context of cell excitability, but their roles in less cell type-specific functions, such as cell proliferation, angiogenesis or cell migration, have only recently been assessed. Moreover, the importance of these channels for tumour biology has become evident. This, coupled with the fact that they are accessible proteins and that their pharmacology is well characterized, has increased the interest in investigating potassium channels as therapeutic targets in cancer patients.

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: Schematic structure of potassium channels.
Figure 2: Summary of the roles of K+ channels in oncology and the mechanisms responsible for their aberrant expression.
Figure 3: Summary of the roles of Kv10.1 in oncology and the mechanisms that are responsible for its aberrant expression in tumour cells.

Similar content being viewed by others

References

  1. Wulff, H., Castle, N. A. & Pardo, L. A. Voltage-gated potassium channels as therapeutic targets. Nature Rev. Drug Discov. 8, 982–1001 (2009).

    Article  CAS  Google Scholar 

  2. Downie, B. R. et al. Eag1 expression interferes with hypoxia homeostasis and induces angiogenesis in tumors. J. Biol. Chem. 283, 36234–36240 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Hegle, A. P., Marble, D. D. & Wilson, G. F. A voltage-driven switch for ion-independent signaling by ether-à-go-go K+ channels. Proc. Natl Acad. Sci. USA 103, 2886–2891 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Gutman, G. A. et al. International Union of Pharmacology. LIII. Nomenclature and molecular relationships of voltage-gated potassium channels. Pharmacol. Rev. 57, 473–508 (2005).

    Article  CAS  PubMed  Google Scholar 

  5. Kaczmarek, L. K. Non-conducting functions of voltage-gated ion channels. Nature Rev. Neurosci. 7, 761–771 (2006). This review brings into focus the non-canonical functions of ion channels as more than just a curious phenomenon and as the biological basis of many functions of ion channels.

    Article  CAS  Google Scholar 

  6. Huber, S. M. Oncochannels. Cell Calcium 53, 241–255 (2013).

    Article  CAS  PubMed  Google Scholar 

  7. Choi, M. et al. K+ channel mutations in adrenal aldosterone-producing adenomas and hereditary hypertension. Science 331, 768–772 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Hofmann, G. et al. HERG K+ channels activation during β1 integrin-mediated adhesion to fibronectin induces an up-regulation of αvβ3 integrin in the preosteoclastic leukemia cell line FLG 29.1. J. Biol. Chem. 276, 4923–4931 (2001).

    Article  CAS  PubMed  Google Scholar 

  9. Munoz, C. et al. Stimulation of HERG channel activity by β-catenin. PLoS ONE 7, e43353 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Wu, X. et al. p38 MAPK regulates the expression of ether a go-go potassium channel in human osteosarcoma cells. Radiol Oncol. 47, 42–49 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Alvarez-Baron, C. P., Jonsson, P., Thomas, C., Dryer, S. E. & Williams, C. The two-pore domain potassium channel KCNK5: induction by estrogen receptor α and role in proliferation of breast cancer cells. Mol. Endocrinol. 25, 1326–1336 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Cho, Y. G. et al. Genetic and expression analysis of the KCNRG gene in hepatocellular carcinomas. Exp. Mol. Med. 38, 247–255 (2006).

    Article  CAS  PubMed  Google Scholar 

  13. Ivanov, D. V. et al. A new human gene KCNRG encoding potassium channel regulating protein is a cancer suppressor gene candidate located in 13q14.3. FEBS Lett. 539, 156–160 (2003).

    Article  CAS  PubMed  Google Scholar 

  14. Usman, H. & Mathew, M. K. Potassium channel regulator KCNRG regulates surface expression of Shaker-type potassium channels. Biochem. Biophys. Res. Commun. 391, 1301–1305 (2010).

    Article  CAS  PubMed  Google Scholar 

  15. Brevet, M. et al. Deregulation of 2 potassium channels in pancreas adenocarcinomas: implication of KV1.3 gene promoter methylation. Pancreas 38, 649–654 (2009).

    Article  CAS  PubMed  Google Scholar 

  16. Brevet, M., Haren, N., Sevestre, H., Merviel, P. & Ouadid-Ahidouch, H. DNA methylation of KV1.3 potassium channel gene promoter is associated with poorly differentiated breast adenocarcinoma. Cell Physiol. Biochem. 24, 25–32 (2009).

    Article  CAS  PubMed  Google Scholar 

  17. Feng, Q. et al. DNA methylation in tumor and matched normal tissues from non-small cell lung cancer patients. Cancer Epidemiol. Biomarkers Prev. 17, 645–654 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Cicek, M. S. et al. Epigenome-wide ovarian cancer analysis identifies a methylation profile differentiating clear-cell histology with epigenetic silencing of the HERG K+ channel. Hum. Mol. Genet. 22, 3038–3047 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Menendez, S. T. et al. Frequent aberrant expression of the human ether à go-go (hEAG1) potassium channel in head and neck cancer: pathobiological mechanisms and clinical implications. J. Mol. Med. 90, 1173–1184 (2012).

    Article  CAS  PubMed  Google Scholar 

  20. Bianchi, L. et al. herg encodes a K+ current highly conserved in tumors of different histogenesis: a selective advantage for cancer cells? Cancer Res. 58, 815–822 (1998). This study shows the first direct evidence of the relevance of a potassium channel for tumour progression in different tumour types.

    CAS  PubMed  Google Scholar 

  21. Cheong, A. et al. Downregulated REST transcription factor is a switch enabling critical potassium channel expression and cell proliferation. Mol. Cell 20, 45–52 (2005).

    Article  CAS  PubMed  Google Scholar 

  22. Lin, H. et al. Transcriptional and post- transcriptional mechanisms for oncogenic overexpression of ether à go-go K+ channel. PLoS ONE 6, e20362 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Diaz, L. et al. Estrogens and human papilloma virus oncogenes regulate human ether-a-go-go-1 potassium channel expression. Cancer Res. 69, 3300–3307 (2009).

    Article  CAS  PubMed  Google Scholar 

  24. Liu, R. G., Wang, W. J., Song, N., Chen, Y. Q. & Li, L. H. Diazoxide preconditioning alleviates apoptosis of hippocampal neurons induced by anoxia-reoxygenation in vitro through up-regulation of Bcl-2/Bax protein ratio. Sheng Li Xue Bao 58, 345–350 (2006).

    CAS  PubMed  Google Scholar 

  25. Scheffner, M., Werness, B. A., Huibregtse, J. M., Levine, A. J. & Howley, P. M. The E6 oncoprotein encoded by human papillomavirus type-16 and type-18 promotes the degradation of p53. Cell 63, 1129–1136 (1990).

    Article  CAS  PubMed  Google Scholar 

  26. Hwang, S. G., Lee, D. Y., Kim, J. Y., Seo, T. G. & Choe, J. H. Human papillomavirus type 16 E7 binds to E2F1 and activates E2F1-driven transcription in a retinoblastoma protein-independent manner. J. Biol. Chem. 277, 2923–2930 (2002).

    Article  CAS  PubMed  Google Scholar 

  27. Borowiec, A. S. et al. IGF-1 activates hEAG K+ channels through an Akt-dependent signaling pathway in breast cancer cells: role in cell proliferation. J. Cell. Physiol. 212, 690–701 (2007).

    Article  CAS  PubMed  Google Scholar 

  28. Sabbatini, P. & McCormick, F. Phosphoinositide 3-OH kinase (PI3K) and PKB/Akt delay the onset of p53-mediated, transcriptionally dependent apoptosis. J. Biol. Chem. 274, 24263–24269 (1999).

    Article  CAS  PubMed  Google Scholar 

  29. Bai, Y. et al. MiR-296-3p regulates cell growth and multi-drug resistance of human glioblastoma by targeting ether-a-go-go (EAG1). Eur. J. Cancer 49, 710–724 (2013).

    Article  CAS  PubMed  Google Scholar 

  30. Yoon, A.-R. et al. MicroRNA-296 is enriched in cancer cells and downregulates p21WAF1 mRNA expression via interaction with its 3′ untranslated region. Nucleic Acids Res. 39, 8078–8091 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Ousingsawat, J. et al. Expression of voltage-gated potassium channels in human and mouse colonic carcinoma. Clin. Cancer Res. 13, 824–831 (2007).

    Article  CAS  PubMed  Google Scholar 

  32. Pardo, L. A. et al. Oncogenic potential of EAG K+ channels. EMBO J. 18, 5540–5547 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Ufartes, R. et al. Behavioural and functional characterization of Kv10.1 (Eag1) knockout mice. Hum. Mol. Genet. 22, 2247–2262 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Martin, S. et al. Eag1 potassium channel immunohistochemistry in the CNS of adult rat and selected regions of human brain. Neuroscience 155, 833–844 (2008).

    Article  CAS  PubMed  Google Scholar 

  35. Hemmerlein, B. et al. Overexpression of Eag1 potassium channels in clinical tumours. Mol. Cancer 5, 41 (2006).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. de Queiroz, F. M., Suarez-Kurtz, G., Stühmer, W. & Pardo, L. A. Ether a go-go potassium channel expression in soft tissue sarcoma patients. Mol. Cancer 5, 42 (2006).

    Article  CAS  Google Scholar 

  37. Agarwal, J. R., Griesinger, F., Stühmer, W. & Pardo, L. A. The potassium channel Ether à go-go is a novel prognostic factor with functional relevance in acute myeloid leukemia. Mol. Cancer 9, 18 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Asher, V. et al. The Eag potassium channel as a new prognostic marker in ovarian cancer. Diagn. Pathol. 5, 78 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Ding, X. W., Luo, H. S., Jin, X., Yan, J. J. & Ai, Y. W. Aberrant expression of Eag1 potassium channels in gastric cancer patients and cell lines. Med. Oncol. 24, 345–350 (2007).

    Article  CAS  PubMed  Google Scholar 

  40. Ding, X. W., Yan, J. J., An, P., Lu, P. & Luo, H. S. Aberrant expression of ether a go-go potassium channel in colorectal cancer patients and cell lines. World J. Gastroenterol. 13, 1257–1261 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Wadhwa, S., Wadhwa, P., Dinda, A. K. & Gupta, N. P. Differential expression of potassium ion channels in human renal cell carcinoma. Int. Urol. Nephrol. 41, 251–257 (2009).

    Article  CAS  PubMed  Google Scholar 

  42. Gómez-Varela, D. et al. Monoclonal antibody blockade of the human Eag1 potassium channel function exerts antitumor activity. Cancer Res. 67, 7343–7349 (2007). This study shows the in vivo efficacy of a functional monoclonal antibody against a potassium channel.

    Article  PubMed  CAS  Google Scholar 

  43. Hartung, F., Stühmer, W. & Pardo, L. A. Tumor cell-selective apoptosis induction through targeting of KV10.1 via bifunctional TRAIL antibody. Mol. Cancer 10, 109 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Becchetti, A. & Arcangeli, A. Integrins and ion channels in cell migration: implications for neuronal development, wound healing and metastatic spread. Adv. Exp. Med. Biol. 674, 107–123 (2010).

    Article  CAS  PubMed  Google Scholar 

  45. Schwab, A., Fabian, A., Hanley, P. J. & Stock, C. Role of ion channels and transporters in cell migration. Physiol. Rev. 92, 1865–1913 (2012). This is a thorough review of the roles of ion transport molecules in cell migration.

    Article  CAS  PubMed  Google Scholar 

  46. Rezzonico, R. et al. Focal adhesion kinase pp125FAK interacts with the large conductance calcium-activated hSlo potassium channel in human osteoblasts: potential role in mechanotransduction. J. Bone Miner. Res. 18, 1863–1871 (2003).

    Article  CAS  PubMed  Google Scholar 

  47. Wei, J. F. et al. Formation of Kv2.1-FAK complex as a mechanism of FAK activation, cell polarization and enhanced motility. J. Cell. Physiol. 217, 544–557 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Cheng, L., Yung, A., Covarrubias, M. & Radice, G. L. Cortactin is required for N-cadherin regulation of Kv1.5 channel function. J. Biol. Chem. 286, 20478–20489 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Hattan, D., Nesti, E., Cachero, T. G. & Morielli, A. D. Tyrosine phosphorylation of Kv1.2 modulates its interaction with the actin-binding protein cortactin. J. Biol. Chem. 277, 38596–38606 (2002).

    Article  CAS  PubMed  Google Scholar 

  50. Bittner, S. et al. The TASK1 channel inhibitor A293 shows efficacy in a mouse model of multiple sclerosis. Exp. Neurol. 238, 149–155 (2012).

    Article  CAS  PubMed  Google Scholar 

  51. Tian, L. et al. A noncanonical SH3 domain binding motif links BK channels to the actin cytoskeleton via the SH3 adapter cortactin. FASEB J. 20, 2588–2590 (2006).

    Article  CAS  PubMed  Google Scholar 

  52. Williams, M. R., Markey, J. C., Doczi, M. A. & Morielli, A. D. An essential role for cortactin in the modulation of the potassium channel Kv1.2. Proc. Natl Acad. Sci. USA 104, 17412–17417 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Levite, M. et al. Extracellular K+ and opening of voltage-gated potassium channels activate T cell integrin function: Physical and functional association between Kv1.3 channels and β1 integrins. J. Exp. Med. 191, 1167–1176 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Cherubini, A. et al. Human ether-a-go-go-related gene 1 channels are physically linked to β1 integrins and modulate adhesion-dependent signaling. Mol. Biol. Cell 16, 2972–2983 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Pillozzi, S. et al. VEGFR-1 (FLT-1), β1 integrin, and hERG K+ channel for a macromolecular signaling complex in acute myeloid leukemia: role in cell migration and clinical outcome. Blood 110, 1238–1250 (2007).

    Article  CAS  PubMed  Google Scholar 

  56. Pillozzi, S. & Arcangeli, A. Physical and functional interaction between integrins and hERG1 channels in cancer cells. Adv. Exp. Med. Biol. 674, 55–67 (2010).

    Article  CAS  PubMed  Google Scholar 

  57. Li, H. et al. The role of hERG1 K+ channels and a functional link between hERG1 K+ channels and SDF-1 in acute leukemic cell migration. Exp. Cell Res. 315, 2256–2264 (2009).

    Article  CAS  PubMed  Google Scholar 

  58. Afrasiabi, E. et al. Expression and significance of HERG (KCNH2) potassium channels in the regulation of MDA-MB-435S melanoma cell proliferation and migration. Cell Signal 22, 57–64 (2010).

    Article  CAS  PubMed  Google Scholar 

  59. Asghar, M. Y., Viitanen, T., Kemppainen, K. & Tornquist, K. Sphingosine 1-phosphate and human ether-à-go-go-related gene potassium channels modulate migration in human anaplastic thyroid cancer cells. Endocr. Relat. Cancer 19, 667–680 (2012).

    Article  CAS  PubMed  Google Scholar 

  60. Arcangeli, A. et al. A novel inward-rectifying K+ current with a cell-cycle dependence governs the resting potential of mammalian neuroblastoma cells. J. Physiol. 489, 455–471 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Arcangeli, A. et al. HERG- and IRK-like inward rectifier currents are sequentially expressed during neuronal development of neural crest cells and their derivatives. Eur. J. Neurosci. 9, 2596–2604 (1997).

    Article  CAS  PubMed  Google Scholar 

  62. Pillozzi, S. et al. HERG potassium channels are constitutively expressed in primary human acute myeloid leukemias and regulate cell proliferation of normal and leukemic hemopoietic progenitors. Leukemia 16, 1791–1798 (2002).

    Article  CAS  PubMed  Google Scholar 

  63. Smith, G. A. M. et al. Functional up-regulation of HERG K+ channels in neoplastic hematopoietic cells. J. Biol. Chem. 277, 18528–18534 (2002).

    Article  CAS  PubMed  Google Scholar 

  64. Lastraioli, E. et al. herg1 gene and HERG1 protein are overexpressed in colorectal cancers and regulate cell invasion of tumor cells. Cancer Res. 64, 606–611 (2004).

    Article  CAS  PubMed  Google Scholar 

  65. Shao, X. D. et al. Expression and significance of HERG protein in gastric cancer. Cancer Biol. Ther. 7, 45–50 (2008).

    Article  CAS  PubMed  Google Scholar 

  66. Ding, X. W. et al. Prognostic significance of hERG1 expression in gastric cancer. Dig. Dis. Sci. 55, 1004–1010 (2010).

    Article  CAS  PubMed  Google Scholar 

  67. Dolderer, J. H. et al. HERG1 gene expression as a specific tumor marker in colorectal tissues. Ejso 36, 72–77 (2010).

    Article  CAS  PubMed  Google Scholar 

  68. Glassmeier, G. et al. Inhibition of HERG1 K+ channel protein expression decreases cell proliferation of human small cell lung cancer cells. Pflugers Arch. 463, 365–376 (2012).

    Article  CAS  PubMed  Google Scholar 

  69. Menendez, S. T. et al. Role of HERG1 potassium channel in both malignant transformation and disease progression in head and neck carcinomas. Mod. Pathol. 25, 1069–1078 (2012).

    Article  CAS  PubMed  Google Scholar 

  70. Lastraioli, E. et al. hERG1 channels in human esophagus: evidence for their aberrant expression in the malignant progression of Barrett's esophagus. J. Cell. Physiol. 209, 398–404 (2006).

    Article  CAS  PubMed  Google Scholar 

  71. Masi, A. et al. hERG1 channels are overexpressed in glioblastoma multiforme and modulate VEGF secretion in glioblastoma cell lines. Br. J. Cancer 93, 781–792 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Crociani, O. et al. Cell cycle-dependent expression of HERG1 and HERG1B isoforms in tumor cells. J. Biol. Chem. 278, 2947–2955 (2003).

    Article  CAS  PubMed  Google Scholar 

  73. Shao, X. D. et al. The potent inhibitory effects of cisapride, a specific blocker for human ether-a-go-go-related gene (HERG) channel, on gastric cancer cells. Cancer Biol. Ther. 4, 295–301 (2005).

    Article  CAS  PubMed  Google Scholar 

  74. Zhao, J., Wei, X. L., Jia, Y. S. & Zheng, J. Q. Silencing of herg gene by shRNA inhibits SH-SY5Y cell growth in vitro and in vivo. Eur. J. Pharmacol. 579, 50–57 (2008).

    Article  CAS  PubMed  Google Scholar 

  75. Vandenberg, J. I. et al. hERG K+ channels: structure, function, and clinical significance. Physiol. Rev. 92, 1393–1478 (2012).

    Article  CAS  PubMed  Google Scholar 

  76. Guasti, L. et al. Identification of a posttranslational mechanism for the regulation of hERG1 K+ channel expression and hERG1 current density in tumor cells. Mol. Cell. Biol. 28, 5043–5060 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  Google Scholar 

  78. Schwab, A., Wojnowski, L., Gabriel, K. & Oberleithner, H. Oscillating activity of a Ca2+-sensitive K+ channel. A prerequisite for migration of transformed Madin-Darby canine kidney focus cells. J. Clin. Invest. 93, 1631–1636 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Reinhardt, J., Golenhofen, N., Pongs, O., Oberleithner, H. & Schwab, A. Migrating transformed mdck cells are able to structurally polarize a voltage-activated K+ channel. Proc. Natl Acad. Sci. USA 95, 5378–5382 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Schwab, A., Reinhardt, J., Schneider, S. W., Gassner, B. & Schuricht, B. K+ channel-dependent migration of fibroblasts and human melanoma cells. Cell Physiol. Biochem. 9, 126–132 (1999).

    Article  CAS  PubMed  Google Scholar 

  81. Schwab, A., Schuricht, B., Seeger, P., Reinhardt, J. & Dartsch, P. C. Migration of transformed renal epithelial cells is regulated by K+ channel modulation of actin cytoskeleton and cell volume. Pflugers Arch. 438, 330–337 (1999).

    Article  CAS  PubMed  Google Scholar 

  82. Liu, D., Lu, C., Wan, R., Auyeung, W. W. & Mattson, M. P. Activation of mitochondrial ATP-dependent potassium channels protects neurons against ischemia-induced death by a mechanism involving suppression of Bax translocation and cytochrome c release. J. Cereb. Blood Flow Metab. 22, 431–443 (2002).

    Article  CAS  PubMed  Google Scholar 

  83. Ransom, C. B., Liu, X. & Sontheimer, H. BK channels in human glioma cells have enhanced calcium sensitivity. Glia 38, 281–291 (2002).

    Article  PubMed  Google Scholar 

  84. Weaver, A. K., Liu, X. & Sontheimer, H. Role for calcium-activated potassium channels (BK) in growth control of human malignant glioma cells. J. Neurosci. Res. 78, 224–234 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Sciaccaluga, M. et al. CXCL12-induced glioblastoma cell migration requires intermediate conductance Ca2+-activated K+ channel activity. Am. J. Physiol. Cell Physiol. 299, C175–C184 (2010).

    Article  CAS  PubMed  Google Scholar 

  86. Catacuzzeno, L., Fioretti, B. & Franciolini, F. Expression and role of the intermediate-conductance calcium-activated potassium channel KCa3.1 in glioblastoma. J. Signal Trans. 2012, 421564 (2012).

    Google Scholar 

  87. Ruggieri, P. et al. The inhibition of KCa3.1 channels activity reduces cell motility in glioblastoma derived cancer stem cells. PLoS ONE 7, e47825 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Steinle, M. et al. Ionizing radiation induces migration of glioblastoma cells by activating BK K+ channels. Radiother. Oncol. 101, 122–126 (2011). This study shows that, radiotherapy can increase the migration of glioma cells by the activation of potassium channels and it can thus be counterproductive.

    Article  CAS  PubMed  Google Scholar 

  89. Potier, M. et al. Altered SK3/KCa2.3-mediated migration in adenomatous polyposis coli (Apc) mutated mouse colon epithelial cells. Biochem. Biophys. Res. Commun. 397, 42–47 (2010).

    Article  CAS  PubMed  Google Scholar 

  90. Chantome, A. et al. KCa2.3 channel-dependent hyperpolarization increases melanoma cell motility. Exp. Cell Res. 315, 3620–3630 (2009).

    Article  CAS  PubMed  Google Scholar 

  91. Potier, M. et al. Identification of SK3 channel as a new mediator of breast cancer cell migration. Mol. Cancer Ther. 5, 2946–2953 (2006).

    Article  CAS  PubMed  Google Scholar 

  92. Girault, A. et al. New alkyl-lipid blockers of SK3 channels reduce cancer cell migration and occurrence of metastasis. Curr. Cancer Drug Targets 11, 1111–1125 (2011).

    Article  CAS  PubMed  Google Scholar 

  93. Hammadi, M. et al. Human ether a-gogo K+ channel 1 (hEag1) regulates MDA-MB-231 breast cancer cell migration through Orai1-dependent calcium entry. J. Cell. Physiol. 227, 3837–3846 (2012).

    Article  CAS  PubMed  Google Scholar 

  94. Chantome, A. et al. Pivotal role of the lipid raft SK3-Orai1 complex in human cancer cell migration and bone metastases. Cancer Res. 73, 4852–4861 (2013). This study shows the relevance of the coordinated action of a Ca2+ channel and a K+ channel in migration and provides evidence showing that the inhibition of a KCa2.3–ORAI1 complex reduces the number of metastases in vivo.

    Article  CAS  PubMed  Google Scholar 

  95. Hoffmann, E. K. & Pedersen, S. F. Cell volume homeostatic mechanisms: effectors and signalling pathways. Acta Physiol. (Oxf.) 202, 465–485 (2011).

    Article  CAS  Google Scholar 

  96. Hoffmann, E. K. Ion channels involved in cell volume regulation: effects on migration, proliferation, and programmed cell death in non adherent EAT cells and adherent ELA cells. Cell Physiol. Biochem. 28, 1061–1078 (2011).

    Article  CAS  PubMed  Google Scholar 

  97. Völkl, H., Paulmichl, M. & Lang, F. Cell volume regulation in renal cortical cells. Ren Physiol. Biochem. 11, 158–173 (1988).

    PubMed  Google Scholar 

  98. Rouzaire-Dubois, B., Gerard, V. & Dubois, J. M. Involvement of K+ channels in the quercetin-induced inhibition of neuroblastoma cell growth. Pflugers Arch. 423, 202–205 (1993).

    Article  CAS  PubMed  Google Scholar 

  99. Dubois, J.-M. & Rouzaire-Dubois, B. The influence of cell volume changes on tumour cell proliferation. Eur. Biophys. J. 33, 227–232 (2004).

    Article  PubMed  Google Scholar 

  100. 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). This study gives a mechanistic insight into the consequences of overexpression of a potassium channel for volume control of tumour cells and its relevance for mitosis.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Neylon, C. B., Avdonin, P. V., Larsen, M. A. & Bobik, A. Rat aortic smooth muscle cells expressing charybdotoxin-sensitive potassium channels exhibit enhanced proliferative responses. Clin. Exp. Pharmacol. Physiol. 21, 117–120 (1994).

    Article  CAS  PubMed  Google Scholar 

  102. Wiecha, J. et al. Blockade of Ca2+-activated K+ channels inhibits proliferation of human endothelial cells induced by basic fibroblast growth factor. J. Vasc. Res. 35, 363–371 (1998).

    Article  CAS  PubMed  Google Scholar 

  103. Faehling, M., Koch, E. D., Raithel, J., Trischler, G. & Waltenberger, J. Vascular endothelial growth factor-a activates Ca2+-activated K+ channels in human endothelial cells in culture. Int. J. Biochem. Cell Biol. 33, 337–346 (2001).

    Article  CAS  PubMed  Google Scholar 

  104. Neylon, C. B. Potassium channels and vascular proliferation. Vasc. Pharmacol. 38, 35–41 (2002).

    Article  CAS  Google Scholar 

  105. Miguel-Velado, E. et al. Cell cycle-dependent expression of Kv3.4 channels modulates proliferation of human uterine artery smooth muscle cells. Cardiovasc. Res. 86, 383–391 (2010).

    Article  CAS  PubMed  Google Scholar 

  106. Cheong, A. et al. Potent suppression of vascular smooth muscle cell migration and human neointimal hyperplasia by KV1.3 channel blockers. Cardiovasc. Res. 89, 282–289 (2011).

    Article  CAS  PubMed  Google Scholar 

  107. Cidad, P. et al. Kv1.3 channels can modulate cell proliferation during phenotypic switch by an ion-flux independent mechanism. Arterioscler. Thromb. Vasc. Biol. 32, 1299–1307 (2012).

    Article  CAS  PubMed  Google Scholar 

  108. Bi, D. et al. The intermediate-conductance calcium-activated potassium channel KCa3.1 regulates vascular smooth muscle cell proliferation via controlling calcium-dependent signaling. J Biol Chem, 15843–15853 (2013).

  109. Zhao, L. M., Su, X. L., Wang, Y., Li, G. R. & Deng, X. L. KCa3.1 channels mediate the increase of cell migration and proliferation by advanced glycation endproducts in cultured rat vascular smooth muscle cells. Lab. Invest. 93, 159–167 (2013).

    Article  CAS  PubMed  Google Scholar 

  110. Ouadid-Ahidouch, H. et al. Changes in the K+ current-density of MCF-7 cells during progression through the cell cycle: Possible involvement of a h-ether.a-gogo K+ channel. Recept. Channels 7, 345–356 (2001).

    CAS  PubMed  Google Scholar 

  111. Benito, J. et al. Pronounced hypoxia in models of murine and human leukemia: High efficacy of hypoxia-activated prodrug PR-104. PLoS ONE 6, e23108 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011).

    Article  CAS  PubMed  Google Scholar 

  113. Urrego-Blanco, D., Tomczak, A. P., Zahed, F., Stühmer, W. & Pardo, L. A. Potassium channels in cell cycle and cell proliferation. Philos Trans B (in the press).

  114. DeCoursey, T. E., Chandy, K. G., Gupta, S. & Cahalan, M. D. Voltage-gated K+ channels in human T lymphocytes: a role in mitogenesis? Nature 307, 465–468 (1984). This paper describes seminal work on the relevance of potassium channels for cell proliferation.

    Article  CAS  PubMed  Google Scholar 

  115. Blackiston, D. J., McLaughlin, K. A. & Levin, M. Bioelectric controls of cell proliferation: ion channels, membrane voltage and the cell cycle. Cell Cycle 8, 3519–3528 (2009).

    Article  Google Scholar 

  116. Cahalan, M. D. & Chandy, K. G. The functional network of ion channels in T lymphocytes. Immunol. Rev. 231, 59–87 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Faouzi, M., Chopin, V., Ahidouch, A. & Ouadid-Ahidouch, H. Intermediate Ca2+-sensitive K+ channels are necessary for prolactin-induced proliferation in breast cancer cells. J. Membr. Biol. 234, 47–56 (2010).

    Article  CAS  PubMed  Google Scholar 

  118. Lallet-Daher, H. et al. Intermediate-conductance Ca2+-activated K+ channels (IKCa1) regulate human prostate cancer cell proliferation through a close control of calcium entry. Oncogene 28, 1792–1806 (2009).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  120. Skelding, K. A., Rostas, J. A. P. & Verrills, N. M. Controlling the cell cycle: The role of calcium/calmodulin-stimulated protein kinases I and II. Cell Cycle 10, 631–639 (2011).

    Article  CAS  PubMed  Google Scholar 

  121. Kahl, C. R. & Means, A. R. Regulation of cell cycle progression by calcium/calmodulin-dependent pathways. Endocr. Rev. 24, 719–736 (2003).

    Article  CAS  PubMed  Google Scholar 

  122. Patel, A. J. & Lazdunski, M. The 2P-domain K+ channels: role in apoptosis and tumorigenesis. Pflugers Arch. 448, 261–273 (2004).

    Article  CAS  PubMed  Google Scholar 

  123. McFerrin, M. B., Turner, K. L., Cuddapah, V. A. & Sontheimer, H. Differential role of IK and BK potassium channels as mediators of intrinsic and extrinsic apoptotic cell death. Am. J. Physiol. Cell Physiol. 303, C1070–C1078 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Borjesson, S. I., Englund, U. H., Asif, M. H., Willander, M. & Elinder, F. Intracellular K+ concentration decrease is not obligatory for apoptosis. J. Biol. Chem. 286, 39823–39828 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  125. Benitez-Rangel, E., Garcia, L., Namorado, M. C., Reyes, J. L. & Guerrero-Hernandez, A. Ion channel inhibitors block caspase activation by mechanisms other than restoring intracellular potassium concentration. Cell Death Dis. 2, e113 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Nagata, K. et al. Nicorandil inhibits oxidative stress-induced apoptosis in cardiac myocytes through activation of mitochondrial ATP-sensitive potassium channels and a nitrate-like effect. J. Mol. Cell Cardiol 35, 1505–1512 (2003).

    Article  CAS  PubMed  Google Scholar 

  127. Qian, X. et al. Glibenclamide exerts an antitumor activity through reactive oxygen species-c-jun NH2-terminal kinase pathway in human gastric cancer cell line MGC-803. Biochem. Pharmacol. 76, 1705–1715 (2008).

    Article  CAS  PubMed  Google Scholar 

  128. Wang, L. et al. The protective roles of mitochondrial ATP-sensitive potassium channels during hypoxia–ischemia–reperfusion in brain. Neurosci. Lett. 491, 63–67 (2011).

    Article  CAS  PubMed  Google Scholar 

  129. Foster, D. B. et al. Mitochondrial ROMK channel is a molecular component of mitoKATP . Circ. Res. 111, 446–454 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Kim, Y., Bang, H. & Kim, D. TASK-3, a new member of the tandem pore K+ channel family. J. Biol. Chem. 275, 9340–9347 (2000).

    Article  CAS  PubMed  Google Scholar 

  131. Mu, D. et al. Genomic amplification and oncogenic properties of the KCNK9 potassium channel gene. Cancer Cell 3, 297–302 (2003).

    Article  CAS  PubMed  Google Scholar 

  132. Lee, G. W. et al. Reduction of breast cancer cell migration via up-regulation of TASK-3 two-pore domain K+ channel. Acta Physiol. (Oxf.) 204, 513–524 (2012).

    Article  CAS  Google Scholar 

  133. Pei, L. et al. Oncogenic potential of TASK3 (Kcnk9) depends on K+ channel function. Proc. Natl Acad. Sci. USA 100, 7803–7807 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Lauritzen, I. et al. K+-dependent cerebellar granule neuron apoptosis. Role of TASK leak K+ channels. J. Biol. Chem. 278, 32068–32076 (2003).

    Article  CAS  PubMed  Google Scholar 

  135. Meuth, S. G. et al. The two-pore domain potassium channel TASK3 functionally impacts glioma cell death. J. Neurooncol 87, 263–270 (2008).

    Article  CAS  PubMed  Google Scholar 

  136. Rusznak, Z. et al. Mitochondrial expression of the two-pore domain TASK-3 channels in malignantly transformed and non-malignant human cells. Virchows Arch. 452, 415–426 (2008).

    Article  CAS  PubMed  Google Scholar 

  137. Innamaa, A. et al. Expression and prognostic significance of the oncogenic K2P potassium channel KCNK9 (TASK-3) in ovarian carcinoma. Anticancer Res. 33, 1401–1408 (2013).

    CAS  PubMed  Google Scholar 

  138. Bock, J., Szabo, I., Jekle, A. & Gulbins, E. Actinomycin D-induced apoptosis involves the potassium channel Kv1.3. Biochem. Biophys. Res. Commun. 295, 526–531 (2002).

    Article  CAS  PubMed  Google Scholar 

  139. Szabo, I. et al. Mitochondrial potassium channel Kv1.3 mediates Bax-induced apoptosis in lymphocytes. Proc. Natl Acad. Sci. USA 105, 14861–14866 (2008). This study highlights the relevance of potassium channels that are expressed in intracellular organelles (and not at the plasma membrane) for fundamental cellular functions.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Gulbins, E., Sassi, N., Grassme, H., Zoratti, M. & Szabo, I. Role of Kv1.3 mitochondrial potassium channel in apoptotic signalling in lymphocytes. Biochim. Biophys. Acta 1797, 1251–1259 (2010).

    Article  CAS  PubMed  Google Scholar 

  141. Leanza, L. et al. Inhibitors of mitochondrial Kv1.3 channels induce Bax/Bak-independent death of cancer cells. EMBO Mol. Med. 4, 577–593 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Leanza, L., Zoratti, M., Gulbins, E. & Szabo, I. Induction of apoptosis in macrophages via Kv1.3 and Kv1.5 potassium channels. Curr. Med. Chem. 19, 5394–5404 (2012).

    Article  CAS  PubMed  Google Scholar 

  143. Sassi, N. et al. An investigation of the occurrence and properties of the mitochondrial intermediate-conductance Ca2+-activated K+ channel mtKCa3.1. Biochim. Biophys. Acta 1797, 1260–1267 (2010).

    Article  CAS  PubMed  Google Scholar 

  144. Pillozzi, S. et al. Chemotherapy resistance in acute lymphoblastic leukemia requires hERG1 channels and is overcome by hERG1 blockers. Blood 117, 902–914 (2011). This study gives a strategy for using the targeted inhibition of a potassium channel to overcome resistance to chemotherapeutic agents.

    Article  CAS  PubMed  Google Scholar 

  145. Suzuki, Y. et al. Depolarization potentiates TRAIL-induced apoptosis in human melanoma cells: role for ATP-sensitive K+ channels and endoplasmic reticulum stress. Int. J. Oncol. 41, 465–475 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Gong, J. H., Liu, X. J., Shang, B. Y., Chen, S. Z. & Zhen, Y. S. HERG K+ channel related chemosensitivity to sparfloxacin in colon cancer cells. Oncol. Rep. 23, 1747–1756 (2010).

    CAS  PubMed  Google Scholar 

  147. Ganapathi, S. B., Kester, M. & Elmslie, K. S. State-dependent block of HERG potassium channels by R-roscovitine: implications for cancer therapy. Am. J. Physiol. Cell Physiol. 296, C701–C710 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Ortiz, C. S. et al. Eag1 potassium channels as markers of cervical dysplasia. Oncol. Rep. 26, 1377–1383 (2011).

    PubMed  Google Scholar 

  149. Chen, S. Z., Jiang, M. & Zhen, Y. S. HERG K+ channel expression-related chemosensitivity in cancer cells and its modulation by erythromycin. Cancer Chemother. Pharmacol. 56, 212–220 (2005).

    Article  CAS  PubMed  Google Scholar 

  150. Zhang, R. et al. Human ether-a-go-go-related gene expression is essential for cisplatin to induce apoptosis in human gastric cancer. Oncol. Rep. 27, 433–440 (2012).

    CAS  PubMed  Google Scholar 

  151. Asher, V., Sowter, H., Shaw, R., Bali, A. & Khan, R. Eag and HERG potassium channels as novel therapeutic targets in cancer. World J. Surg. Oncol. 8, 113 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  152. Sontheimer, H. An unexpected role for ion channels in brain tumor metastasis. Exp. Biol. Med. (Maywood) 233, 779–791 (2008).

    Article  CAS  Google Scholar 

  153. Jahchan, N. S. et al. A drug repositioning approach identifies tricyclic antidepressants as inhibitors of small cell lung cancer and other neuroendocrine tumors. Cancer Discovery http://dx.doi.org/10.1158/2159-8290.CD-13-0183 (2013).

  154. Ninkovic, M., Mitkovski, M., Kohl, T., Stühmer, W. & Pardo, L. A. Physical and functional interaction of KV10.1 with Rabaptin-5 impacts ion channel trafficking. FEBS Lett. 586, 3077–3084 (2012).

    Article  CAS  PubMed  Google Scholar 

  155. Herrmann, S., Ninkovic, M., Kohl, T., Lörinczi, E. & Pardo, L. A. Cortactin controls surface expression of the voltage-gated potassium channel KV10.1. J. Biol. Chem. 287, 44151–44163 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Farias, L. M. B. et al. Ether à go-go potassium channels as human cervical cancer markers. Cancer Res. 64, 6996–7001 (2004).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank the Max Planck Society and the past and present members of their department.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Luis A. Pardo or Walter Stühmer.

Ethics declarations

Competing interests

L.A.P. and W.S. are shareholders of iOnGen AG.

PowerPoint slides

Glossary

Permeation path

The path formed by a fourfold repeat of the pore-forming loop (P-loop) that is provided by each of the four subunits that constitutes the ion channel. Residues in the P-loop are responsible for the selectivity for specific ions.

Fratricide

The induction of apoptotic death in nearby cells, which normally occurs via a death receptor and its ligand. This occurs naturally in the immune system and other systems, and it can also be induced by chimeric ligands.

Long QT syndrome

The prolongation of the ventricular action potential, which can be diagnosed by a large time interval between the Q and T waves in an electrocardiogram. It is mainly caused by genetic mutations in mechanisms that are responsible for repolarization, such as the Kv11.1-related ion channel ERG.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Pardo, L., Stühmer, W. The roles of K+ channels in cancer. Nat Rev Cancer 14, 39–48 (2014). https://doi.org/10.1038/nrc3635

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrc3635

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