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.

  • Letter
  • Published:

The structural mechanism of KCNH-channel regulation by the eag domain

Abstract

The KCNH voltage-dependent potassium channels (ether-à-go-go, EAG; EAG-related gene, ERG; EAG-like channels, ELK) are important regulators of cellular excitability1,2,3 and have key roles in diseases such as cardiac long QT syndrome type 2 (LQT2)4, epilepsy5, schizophrenia6 and cancer7. The intracellular domains of KCNH channels are structurally distinct from other voltage-gated channels. The amino-terminal region contains an eag domain, which is composed of a Per-Arnt-Sim (PAS) domain and a PAS-cap domain8, whereas the carboxy-terminal region contains a cyclic nucleotide-binding homology domain (CNBHD), which is connected to the pore through a C-linker domain. Many disease-causing mutations localize to these specialized intracellular domains, which underlie the unique gating and regulation of KCNH channels9. It has been suggested that the eag domain may regulate the channel by interacting with either the S4–S5 linker or the CNBHD8,10. Here we present a 2 Å resolution crystal structure of the eag domain–CNBHD complex of the mouse EAG1 (also known as KCNH1) channel. It displays extensive interactions between the eag domain and the CNBHD, indicating that the regulatory mechanism of the eag domain primarily involves the CNBHD. Notably, the structure reveals that a number of LQT2 mutations at homologous positions in human ERG, in addition to cancer-associated mutations in EAG channels, localize to the eag domain–CNBHD interface. Furthermore, mutations at the interface produced marked effects on channel gating, demonstrating the important physiological role of the eag domain–CNBHD interaction. Our structure of the eag domain–CNBHD complex of mouse EAG1 provides unique insights into the physiological and pathophysiological mechanisms of KCNH channels.

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: Structure of the eag domain–CNBHD complex of mEAG1.
Figure 2: The various interfaces between the eag domain and CNBHD, and different complex types in the asymmetric unit.
Figure 3: A salt bridge between the eag domain and the CNBHD.
Figure 4: PAS-cap interaction with the CNBHD, and predicted tetramer of the eag domain–CNBHD complex.

Similar content being viewed by others

Accession codes

Accessions

Protein Data Bank

Data deposits

Atomic coordinates and structure factors for the reported crystal structures have been deposited with the Protein Data Bank under accession code 4LLO.

References

  1. Warmke, J., Drysdale, R. & Ganetzky, B. A distinct potassium channel polypeptide encoded by the Drosophila eag locus. Science 252, 1560–1562 (1991)

    Article  ADS  CAS  Google Scholar 

  2. Ganetzky, B., Robertson, G. A., Wilson, G. F., Trudeau, M. C. & Titus, S. A. The eag family of K+ channels in Drosophila and mammals. Ann. NY Acad. Sci. 868, 356–369 (1999)

    Article  ADS  CAS  Google Scholar 

  3. Bauer, C. K. & Schwarz, J. R. Physiology of EAG K+ channels. J. Membr. Biol. 182, 1–15 (2001)

    Article  CAS  Google Scholar 

  4. Sanguinetti, M. C. & Tristani-Firouzi, M. hERG potassium channels and cardiac arrhythmia. Nature 440, 463–469 (2006)

    Article  ADS  CAS  Google Scholar 

  5. Zhang, X. et al. Deletion of the potassium channel Kv12.2 causes hippocampal hyperexcitability and epilepsy. Nature Neurosci. 13, 1056–1058 (2010)

    Article  CAS  Google Scholar 

  6. Huffaker, S. J. et al. A primate-specific, brain isoform of KCNH2 affects cortical physiology, cognition, neuronal repolarization and risk of schizophrenia. Nature Med. 15, 509–518 (2009)

    Article  CAS  Google Scholar 

  7. Camacho, J. Ether à go-go potassium channels and cancer. Cancer Lett. 233, 1–9 (2006)

    Article  CAS  Google Scholar 

  8. Morais Cabral, J. H. et al. Crystal structure and functional analysis of the HERG potassium channel N terminus: a eukaryotic PAS domain. Cell 95, 649–655 (1998)

    Article  CAS  Google Scholar 

  9. Gustina, A. S. & Trudeau, M. C. HERG potassium channel regulation by the N-terminal eag domain. Cell. Signal. 24, 1592–1598 (2012)

    Article  CAS  Google Scholar 

  10. Li, Q. et al. NMR solution structure of the N-terminal domain of hERG and its interaction with the S4–S5 linker. Biochem. Biophys. Res. Commun. 403, 126–132 (2010)

    Article  CAS  Google Scholar 

  11. Brelidze, T. I., Carlson, A. E., Sankaran, B. & Zagotta, W. N. Structure of the carboxy-terminal region of a KCNH channel. Nature 481, 530–533 (2012)

    Article  ADS  CAS  Google Scholar 

  12. Marques-Carvalho, M. J. et al. Structural, biochemical, and functional characterization of the cyclic nucleotide binding homology domain from the mouse EAG1 potassium channel. J. Mol. Biol. 423, 34–46 (2012)

    Article  CAS  Google Scholar 

  13. Brelidze, T. I., Carlson, A. E. & Zagotta, W. N. Absence of direct cyclic nucleotide modulation of mEAG1 and hERG1 channels revealed with fluorescence and electrophysiological methods. J. Biol. Chem. 284, 27989–27997 (2009)

    Article  CAS  Google Scholar 

  14. Stevens, L., Ju, M. & Wray, D. Roles of surface residues of intracellular domains of heag potassium channels. Eur. Biophys. J. 38, 523–532 (2009)

    Article  CAS  Google Scholar 

  15. Bamford, S. et al. The COSMIC (Catalogue of Somatic Mutations in Cancer) database and website. Br. J. Cancer 91, 355–358 (2004)

    Article  CAS  Google Scholar 

  16. Chen, J., Zou, A., Splawski, I., Keating, M. T. & Sanguinetti, M. C. Long QT syndrome-associated mutations in the Per-Arnt-Sim (PAS) domain of HERG potassium channels accelerate channel deactivation. J. Biol. Chem. 274, 10113–10118 (1999)

    Article  CAS  Google Scholar 

  17. Splawski, I. et al. Spectrum of mutations in long-QT syndrome genes KVLQT1, HERG, SCN5A, KCNE1, and KCNE2. Circulation 102, 1178–1185 (2000)

    Article  CAS  Google Scholar 

  18. Wang, J., Trudeau, M. C., Zappia, A. M. & Robertson, G. A. Regulation of deactivation by an amino terminal domain in human ether-à-go-go-related gene potassium channels. J. Gen. Physiol. 112, 637–647 (1998)

    Article  CAS  Google Scholar 

  19. Muskett, F. W. et al. Mechanistic insight into human ether-à-go-go-related gene (hERG) K+ channel deactivation gating from the solution structure of the EAG domain. J. Biol. Chem. 286, 6184–6191 (2011)

    Article  CAS  Google Scholar 

  20. Brelidze, T. I., Gianulis, E. C., Dimaio, F., Trudeau, M. C. & Zagotta, W. N. Structure of the C-terminal region of an ERG channel and functional implications. Proc. Natl Acad. Sci. USA 110, 11648–11653 (2013)

    Article  ADS  CAS  Google Scholar 

  21. Adaixo, R., Harley, C. A., Castro-Rodrigues, A. F. & Morais-Cabral, J. H. Structural properties of PAS domains from the KCNH potassium channels. PLoS ONE 8, e59265 (2013)

    Article  ADS  CAS  Google Scholar 

  22. Ng, C. A. et al. The N-terminal tail of hERG contains an amphipathic α-helix that regulates channel deactivation. PLoS ONE 6, e16191 (2011)

    Article  ADS  CAS  Google Scholar 

  23. Napolitano, C. et al. Genetic testing in the long QT syndrome: development and validation of an efficient approach to genotyping in clinical practice. J. Am. Med. Assoc. 294, 2975–2980 (2005)

    Article  CAS  Google Scholar 

  24. 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  Google Scholar 

  25. Sun, X. X., Hodge, J. J., Zhou, Y., Nguyen, M. & Griffith, L. C. The eag potassium channel binds and locally activates calcium/calmodulin-dependent protein kinase II. J. Biol. Chem. 279, 10206–10214 (2004)

    Article  CAS  Google Scholar 

  26. Ziechner, U. et al. Inhibition of human ether à go-go potassium channels by Ca2+/calmodulin binding to the cytosolic N- and C-termini. FEBS J. 273, 1074–1086 (2006)

    Article  CAS  Google Scholar 

  27. Wang, J., Myers, C. D. & Robertson, G. A. Dynamic control of deactivation gating by a soluble amino-terminal domain in HERG K+channels. J. Gen. Physiol. 115, 749–758 (2000)

    Article  CAS  Google Scholar 

  28. Tester, D. J., Will, M. L., Haglund, C. M. & Ackerman, M. J. Compendium of cardiac channel mutations in 541 consecutive unrelated patients referred for long QT syndrome genetic testing. Heart Rhythm 2, 507–517 (2005)

    Article  Google Scholar 

  29. Zagotta, W. N. et al. Structural basis for modulation and agonist specificity of HCN pacemaker channels. Nature 425, 200–205 (2003)

    Article  ADS  CAS  Google Scholar 

  30. Taraska, J. W., Puljung, M. C., Olivier, N. B., Flynn, G. E. & Zagotta, W. N. Mapping the structure and conformational movements of proteins with transition metal ion FRET. Nature Methods 6, 532–537 (2009)

    Article  CAS  Google Scholar 

  31. Kawate, T. & Gouaux, E. Fluorescence-detection size-exclusion chromatography for precrystallization screening of integral membrane proteins. Structure 14, 673–681 (2006)

    Article  CAS  Google Scholar 

  32. Rossi, A. M. & Taylor, C. W. Analysis of protein-ligand interactions by fluorescence polarization. Nature Protocols 6, 365–387 (2011)

    Article  CAS  Google Scholar 

  33. Collaborative Computational Project, 4. The CCP4 suite: programs for protein crystallography. Acta Crystallogr. D 50, 760–763 (1994)

    Article  Google Scholar 

  34. McCoy, A. J. Solving structures of protein complexes by molecular replacement with Phaser. Acta Crystallogr. D 63, 32–41 (2007)

    Article  CAS  Google Scholar 

  35. Adams, P. D. et al. PHENIX: a comprehensive Python-based system for macromolecular structure solution. Acta Crystallogr. D 66, 213–221 (2010)

    CAS  PubMed  Google Scholar 

  36. Emsley, P. & Cowtan, K. Coot: model-building tools for molecular graphics. Acta Crystallogr. D 60, 2126–2132 (2004)

    Article  Google Scholar 

  37. Zagotta, W. N., Hoshi, T. & Aldrich, R. W. Gating of single Shaker potassium channels in Drosophila muscle and in Xenopus oocytes injected with Shaker mRNA. Proc. Natl Acad. Sci. USA 86, 7243–7247 (1989)

    Article  ADS  CAS  Google Scholar 

  38. Hamill, O. P., Marty, A., Neher, E., Sakmann, B. & Sigworth, F. J. Improved patch-clamp techniques for high-resolution current recording from cells and cell-free membrane patches. Pflugers Arch. 391, 85–100 (1981)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank S. Cunnington and S. Camp for technical assistance. We thank the beamline staff at the Advanced Light Source (ALS). We also thank G. Flynn, J. Bankston, M. Puljung and S. Gordon for helpful discussions and critical reading of the manuscript. This work was supported by the National Institutes of Health (NIH) grant R01 EY010329 (W.N.Z.), The International Human Frontier Science Program Organization (HFSPO) long-term postdoctoral fellowship LT-001025/2011 (Y.H.), and NIH grant F32 HL095241 (A.E.C.). The Berkeley Center for Structural Biology is supported in part by the National Institutes of Health, National Institute of General Medical Sciences, and the Howard Hughes Medical Institute. The Advanced Light Source is supported by the Director, Office of Science, Office of Basic Energy Sciences, of the US Department of Energy under Contract No. DE-AC02-05CH11231.

Author information

Authors and Affiliations

Authors

Contributions

Y.H. and W.N.Z. conceived the experiments. Y.H. performed the biochemical and crystallographic experiments and data analysis. A.E.C. conceived the PAS-cap-related experiments and performed all the electrophysiology experiments and data analysis. Y.H. and W.N.Z. wrote the manuscript.

Corresponding author

Correspondence to William N. Zagotta.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Information

This file contains Supplementary Figures 1-7 and Supplementary Tables 1-4. (PDF 3384 kb)

Morph of the two confomers of the mEAG1 intracellular complex .

A 'type I' eag domain-CNBHD complex (chains A and B) was morphed with a 'type II' complex (chains C and D). Color-coding as in Fig. 1b. Distance measurement is shown between residues D47 of the eag domain and E578 of the CNBHD. Angle measurement was performed between S94 (eag domain), V706 (CNBHD) and H570 (CNBHD). (MOV 3451 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Haitin, Y., Carlson, A. & Zagotta, W. The structural mechanism of KCNH-channel regulation by the eag domain. Nature 501, 444–448 (2013). https://doi.org/10.1038/nature12487

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature12487

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing