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:

Mutations in the voltage-gated potassium channel gene KCNH1 cause Temple-Baraitser syndrome and epilepsy

A Corrigendum to this article was published on 25 February 2015

This article has been updated

Abstract

Temple-Baraitser syndrome (TBS) is a multisystem developmental disorder characterized by intellectual disability, epilepsy, and hypoplasia or aplasia of the nails of the thumb and great toe1,2. Here we report damaging de novo mutations in KCNH1 (encoding a protein called ether à go-go, EAG1 or KV10.1), a voltage-gated potassium channel that is predominantly expressed in the central nervous system (CNS), in six individuals with TBS. Characterization of the mutant channels in both Xenopus laevis oocytes and human HEK293T cells showed a decreased threshold of activation and delayed deactivation, demonstrating that TBS-associated KCNH1 mutations lead to deleterious gain of function. Consistent with this result, we find that two mothers of children with TBS, who have epilepsy but are otherwise healthy, are low-level (10% and 27%) mosaic carriers of pathogenic KCNH1 mutations. Consistent with recent reports3,4,5,6,7,8, this finding demonstrates that the etiology of many unresolved CNS disorders, including epilepsies, might be explained by pathogenic mosaic mutations.

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: Phenotypic features of TBS.
Figure 2: KCNH1 mutations in subjects with TBS.
Figure 3: Mutant KCNH1 channels show a decreased activation threshold and delayed deactivation.

Similar content being viewed by others

Accession codes

Accessions

NCBI Reference Sequence

Change history

  • 06 February 2015

    In the version of this article initially published, in Figure 2a, the order of the protein alterations for variants c.1465C>T and c.1480A>G was inverted. The correct protein alterations for these two variants are p.Leu489Phe and p.Ile494Val, respectively. This error has been corrected in the HTML and PDF versions of the article.

References

  1. Temple, I.K. & Baraitser, M. Severe mental retardation and absent nails of hallux and pollex. Am. J. Med. Genet. 41, 173–175 (1991).

    Article  CAS  PubMed  Google Scholar 

  2. Gabbett, M.T., Clark, R.C. & McGaughran, J.M. A second case of severe mental retardation and absent nails of hallux and pollex (Temple-Baraitser syndrome). Am. J. Med. Genet. 146A, 450–452 (2008).

    Article  PubMed  Google Scholar 

  3. Depienne, C. et al. Mechanisms for variable expressivity of inherited SCN1A mutations causing Dravet syndrome. J. Med. Genet. 47, 404–410 (2010).

    Article  CAS  PubMed  Google Scholar 

  4. Marini, C., Mei, D., Helen Cross, J. & Guerrini, R. Mosaic SCN1A mutation in familial severe myoclonic epilepsy of infancy. Epilepsia 47, 1737–1740 (2006).

    Article  CAS  PubMed  Google Scholar 

  5. Selmer, K.K. et al. Parental SCN1A mutation mosaicism in familial Dravet syndrome. Clin. Genet. 76, 398–403 (2009).

    Article  CAS  PubMed  Google Scholar 

  6. Kato, M. et al. Clinical spectrum of early onset epileptic encephalopathies caused by KCNQ2 mutation. Epilepsia 54, 1282–1287 (2013).

    Article  CAS  PubMed  Google Scholar 

  7. Weckhuysen, S. et al. KCNQ2 encephalopathy: emerging phenotype of a neonatal epileptic encephalopathy. Ann. Neurol. 71, 15–25 (2012).

    Article  CAS  PubMed  Google Scholar 

  8. Jamuar, S.S. et al. Somatic mutations in cerebral cortical malformations. N. Engl. J. Med. 371, 733–743 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Yesil, G., Guler, S., Yuksel, A. & Alanay, Y. Report of a patient with Temple-Baraitser syndrome. Am. J. Med. Genet. 164A, 848–851 (2014).

    Article  PubMed  Google Scholar 

  10. Jacquinet, A. et al. Temple-Baraitser syndrome: a rare and possibly unrecognized condition. Am. J. Med. Genet. 152A, 2322–2326 (2010).

    Article  PubMed  Google Scholar 

  11. Adzhubei, I.A. et al. A method and server for predicting damaging missense mutations. Nat. Methods 7, 248–249 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Kumar, P., Henikoff, S. & Ng, P.C. Predicting the effects of coding non-synonymous variants on protein function using the SIFT algorithm. Nat. Protoc. 4, 1073–1081 (2009).

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  14. Urrego, D., Tomczak, A.P., Zahed, F., Stühmer, W. & Pardo, L.A. Potassium channels in cell cycle and cell proliferation. Phil. Trans. R. Soc. Lond. B 369, 20130094 (2014).

    Article  Google Scholar 

  15. Gómez-Varela, D. et al. Monoclonal antibody blockade of the human Eag1 potassium channel function exerts antitumor activity. Cancer Res. 67, 7343–7349 (2007).

    Article  PubMed  Google Scholar 

  16. Zhang, J. et al. Regulation of cell proliferation of human induced pluripotent stem cell–derived mesenchymal stem cells via ether-à-go-go 1 (hEAG1) potassium channel. Am. J. Physiol. Cell Physiol. 303, C115–C125 (2012).

    Article  CAS  PubMed  Google Scholar 

  17. Ouadid-Ahidouch, H. & Le Bourhis, X. 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 (2000).

    Google Scholar 

  18. Petrovski, S., Wang, Q., Heinzen, E.L., Allen, A.S. & Goldstein, D.B. Genic intolerance to functional variation and the interpretation of personal genomes. PLoS Genet. 9, e1003709 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Tian, C. et al. Potassium channels: structures, diseases, and modulators. Chem. Biol. Drug Des. 83, 1–26 (2014).

    Article  CAS  PubMed  Google Scholar 

  20. 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 

  21. D'Adamo, M.C., Catacuzzeno, L., Di Giovanni, G., Franciolini, F. & Pessia, M. K+ channelepsy: progress in the neurobiology of potassium channels and epilepsy. Front. Cell Neurosci. 7, 134 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Barcia, G. et al. De novo gain-of-function KCNT1 channel mutations cause malignant migrating partial seizures of infancy. Nat. Genet. 44, 1255–1259 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Heron, S.E. et al. Missense mutations in the sodium-gated potassium channel gene KCNT1 cause severe autosomal dominant nocturnal frontal lobe epilepsy. Nat. Genet. 44, 1188–1190 (2012).

    Article  CAS  PubMed  Google Scholar 

  24. Browne, D.L. et al. Episodic ataxia/myokymia syndrome is associated with point mutations in the human potassium channel gene, KCNA1. Nat. Genet. 8, 136–140 (1994).

    Article  CAS  PubMed  Google Scholar 

  25. Lee, Y.-C. et al. Mutations in KCND3 cause spinocerebellar ataxia type 22. Ann. Neurol. 72, 859–869 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Coucke, P.J. et al. Mutations in the KCNQ4 gene are responsible for autosomal dominant deafness in four DFNA2 families. Hum. Mol. Genet. 8, 1321–1328 (1999).

    Article  CAS  PubMed  Google Scholar 

  27. Tawil, R. et al. Andersen's syndrome: potassium-sensitive periodic paralysis, ventricular ectopy, and dysmorphic features. Ann. Neurol. 35, 326–330 (1994).

    Article  CAS  PubMed  Google Scholar 

  28. Epi4K Consortium & Epilepsy Phenome/Genome Project. De novo mutations in epileptic encephalopathies. Nature 501, 217–221 (2013).

  29. Poduri, A., Evrony, G.D., Cai, X. & Walsh, C.A. Somatic mutation, genomic variation, and neurological disease. Science 341, 1237758 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Cleary, J.G. et al. Joint variant and de novo mutation identification on pedigrees from high-throughput sequencing data. J. Comput. Biol. 21, 405–419 (2014).

    Article  CAS  PubMed  Google Scholar 

  31. Cingolani, P. et al. A program for annotating and predicting the effects of single nucleotide polymorphisms, SnpEff: SNPs in the genome of Drosophila melanogaster strain w1118; iso-2; iso-3. Fly (Austin) 6, 80–92 (2012).

    Article  CAS  Google Scholar 

  32. Simons, C. et al. A de novo mutation in the β-tubulin gene TUBB4A results in the leukoencephalopathy hypomyelination with atrophy of the basal ganglia and cerebellum. Am. J. Hum. Genet. 92, 767–773 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Bolger, A.M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 25, 1754–1760 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Koboldt, D.C. et al. VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res. 22, 568–576 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Schroeder, C.I. et al. Chemical synthesis, 3D structure, and ASIC binding site of the toxin mambalgin-2. Angew. Chem. Int. Edn Engl. 53, 1017–1020 (2014).

    Article  CAS  Google Scholar 

  37. Garg, V., Sachse, F.B. & Sanguinetti, M.C. Tuning of EAG K+ channel inactivation: molecular determinants of amplification by mutations and a small molecule. J. Gen. Physiol. 140, 307–324 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We would like to thank the patients and their families for their participation in this study. Computational support was provided by the NeCTAR Genomics Virtual Laboratory and QRIScloud programs. We thank the Queensland Centre for Medical Genomics and Institute for Molecular Bioscience sequencing facility teams for their assistance with this project and J. Lynch for providing access to his patch-clamp facilities. We also wish to thank J. Vandenberg for his helpful discussion of results. R.J.T. was supported by an Australian Research Council Discovery Early-Career Research Award. L.M. is supported by an Australian National Health and Medical Research Council (NHMRC) C.J. Martin fellowship. A.J. and F.-G.D. were supported by a Fund Invest for Scientific Research (FIRS) grant from Centre Hospitalier Universitaire of Liége, Belgium. This work was supported in part by a University of Queensland Foundation Research Excellence Award and a donation by Joan and Graham J. Crawford.

Author information

Authors and Affiliations

Authors

Contributions

C.S., R.J.T., M.T.G. and J.M. conceived and designed the project. M.T.G., J.M., Y.A., A.J., F.-G.D., A.V., J.S., G.Y., S.G. and A.Y. phenotyped the cases, provided clinical samples and provided clinical information. D.M. and S.M.G. were responsible for exome sequencing. C.S. performed all exome and variant analysis, with input from R.J.T. J.G.C. provided software and advice for sequence analysis. J.C., B.C.-A. and K.R. performed Sanger sequencing and constructed the expression vectors. L.D.R., L.M. and G.F.K. conceived the electrophysiology experiments, which were performed by L.D.R., B.C.-A. and L.M. J.C. prepared samples for targeted amplicon sequencing. Amplicon data analysis was performed by C.S. and G.J.B. All data were reviewed and synthesized by C.S. and R.J.T. The manuscript was drafted by C.S. and R.J.T. with input from M.T.G. and L.D.R. All authors discussed the results and commented on the manuscript.

Corresponding authors

Correspondence to Cas Simons or Ryan J Taft.

Ethics declarations

Competing interests

R.J.T. became an employee of Illumina, Inc., during the course of this project.

Integrated supplementary information

Supplementary Figure 1 Activation, deactivation and Inactivation of wild-type and mutant KCNH1 channels expressed in Xenopus oocytes.

(a) Representative families of whole-cell currents showing voltage-dependent activation of wild-type (WT) and mutant human KCNH1 channels expressed in Xenopus oocytes. Currents were elicited by steps from –120 mV to +80 mV in 20-mV increments, using a holding potential of –100 mV. Inset in the final panel: schematic of the pulse protocol used for the activation experiments. (b) Representative families of whole-cell currents showing only minor steady-state inactivation of WT and mutant KCNH1 channels expressed in Xenopus oocytes. Inset in the final panel: schematic of the pulse protocol used for the inactivation experiments. (c) Activation current-voltage relationship (± s.e.m.) for WT, p.Lys217Asn, p.Leu489Phe, p.Ile494Val and p.Gln503Arg channels expressed in Xenopus oocytes. Currents were measured at the end of the activation test pulse. (d) Fast and slow time constants (τ) of deactivation for WT and mutant KCNH1 channels expressed in Xenopus oocytes, analyzed from the tail current at –90 mV following a maximally activating prepulse to +80 mV. (e) Steady-state inactivation current-voltage relationship (± s.e.m.) for WT and mutant KCNH1 channels expressed in Xenopus oocytes. Currents were measured at the end of the +30 mV activation test pulse. Data are presented as mean ± s.e.m. with the numbers of experiments indicated in parentheses. P values were calculated in comparison to WT using an unpaired t test with Welch’s correction. *P < 0.05, **P < 0.01, ***P < 0.001.

Supplementary information

Supplementary Text and Figures

Supplementary Figure 1, Supplementary Tables 1–3 and Supplementary Note. (PDF 681 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Simons, C., Rash, L., Crawford, J. et al. Mutations in the voltage-gated potassium channel gene KCNH1 cause Temple-Baraitser syndrome and epilepsy. Nat Genet 47, 73–77 (2015). https://doi.org/10.1038/ng.3153

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3153

This article is cited by

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