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 EFHC1 cause juvenile myoclonic epilepsy

Abstract

Juvenile myoclonic epilepsy (JME) is the most frequent cause of hereditary grand mal seizures1,2. We previously mapped and narrowed a region associated with JME on chromosome 6p12–p11 (EJM1)3,4,5. Here, we describe a new gene in this region, EFHC1, which encodes a protein with an EF-hand motif. Mutation analyses identified five missense mutations in EFHC1 that cosegregated with epilepsy or EEG polyspike wave in affected members of six unrelated families with JME and did not occur in 382 control individuals. Overexpression of EFHC1 in mouse hippocampal primary culture neurons induced apoptosis that was significantly lowered by the mutations. Apoptosis was specifically suppressed by SNX-482, an antagonist of R-type voltage-dependent Ca2+ channel (Cav2.3). EFHC1 and Cav2.3 immunomaterials overlapped in mouse brain, and EFHC1 coimmunoprecipitated with the Cav2.3 C terminus. In patch-clamp analysis, EFHC1 specifically increased R-type Ca2+ currents that were reversed by the mutations associated with JME.

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 and expression of EFHC1.
Figure 2: EFHC1 mutations segregating with epilepsy traits in families with JME.
Figure 3: Activation of R-type VDCC (Cav2.3)-dependent apoptosis by EFHC1 and its reversal by mutations associated with JME.
Figure 4: Colocalization of Efhc1 and Cav2.3.
Figure 5: Potentiation of R-type VDCC (Cav2.3) activity by wild-type and its reversal by mutant EFHC1.
Figure 6: Immunoprecipitation assays of EFHC1 and VDCCs.

Similar content being viewed by others

Accession codes

Accessions

GenBank/EMBL/DDBJ

References

  1. Janz, D.C.W. Impusive-petit mal. J. Neurol. 176, 344–386 (1957).

    Article  Google Scholar 

  2. Delgado-Escueta, A.V. et al. Mapping and positional cloning of common idiopathic generalized epilepsies: juvenile myoclonus epilepsy and childhood absence epilepsy. Adv. Neurol. 79, 351–374 (1999).

    CAS  PubMed  Google Scholar 

  3. Liu, A.W. et al. Juvenile myoclonic epilepsy locus in chromosome 6p21.2-p11: linkage to convulsions and electroencephalography trait. Am. J. Hum. Genet. 57, 368–381 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Liu, A.W. et al. Juvenile myoclonic epilepsy in chromosome 6p12-p11: locus heterogeneity and recombinations. Am. J. Med. Genet. 63, 438–446 (1996).

    Article  CAS  Google Scholar 

  5. Bai, D. et al. Juvenile myoclonic epilepsy: Linkage to chromosome 6p12 in Mexico families. Am. J. Med. Genet. 113, 268–274 (2002).

    Article  Google Scholar 

  6. Sander, T. et al. Refined mapping of the epilepsy susceptibility locus EJM1 on chromosome 6. Neurology 49, 842–847 (1997).

    Article  CAS  Google Scholar 

  7. Greenberg, D.A. et al. Reproducibility and complications in gene searches: linkage on chromosome 6, heterogeneity, association, and maternal inheritance in juvenile myoclonic epilepsy. Am. J. Hum. Genet. 66, 508–516 (2000).

    Article  CAS  Google Scholar 

  8. Pal, D.K. et al. BRD2 (RING3) is a probable major susceptibility gene for common juvenile myoclonic epilepsy. Am. J. Hum. Genet. 73, 261–270 (2003).

    Article  CAS  Google Scholar 

  9. Pinto, D. et al. Evidence for linkage between juvenile myoclonic epilepsy-related idiopathic generalized epilepsy and 6p11-12 in Dutch families. Epilepsia 45, 211–217 (2004).

    Article  Google Scholar 

  10. Suzuki, T. et al. Identification and mutational analysis of candidate genes for juvenile myoclonic epilepsy on 6p11-p12: LRRC1, GCLC, KIAA0057 and CLIC5. Epilepsy Res. 50, 265–275 (2002).

    Article  CAS  Google Scholar 

  11. Braunewell, K.H. & Gundelfinger, E.D. Intracellular neuronal calcium sensor proteins: a family of EF-hand calcium-binding proteins in search of a function. Cell Tissue Res. 295, 1–12 (1999).

    Article  CAS  Google Scholar 

  12. Escayg, A. et al. Coding and noncoding variation of the human calcium-channel beta 4-subunit gene CACNB4 in patients with idiopathic generalized epilepsy and episodic ataxia. Am. J. Hum. Genet. 66, 1531–1539 (2000).

    Article  CAS  Google Scholar 

  13. Jouvenceau, A. et al. Human epilepsy associated with dysfunction of the brain P/Q-type calcium channel. Lancet 358, 801–807 (2001).

    Article  CAS  Google Scholar 

  14. Fletcher, C.F. et al. Absence epilepsy in tottering mutant mice is associated with calcium channel defects. Cell 87, 607–617 (1996).

    Article  CAS  Google Scholar 

  15. Burgess, D.L., Jones, J.M., Meisler, M.H. & Noebels, J.L. Mutation of the Ca2+ channel beta subunit gene Cchb4 is associated with ataxia and seizures in the lethargic (lh) mouse. Cell 88, 385–392 (1997).

    Article  CAS  Google Scholar 

  16. Letts, V.A. et al. The mouse stargazer gene encodes a neuronal Ca2+-channel gamma subunit. Nat. Genet. 19, 340–347 (1998).

    Article  CAS  Google Scholar 

  17. Newcomb, R. et al. Selective peptide antagonist of the class E calcium channel from the venom of the tarantula Hysterocrates gigas. Biochemistry 37, 15353–15362 (1998).

    Article  CAS  Google Scholar 

  18. Yokoyama, C.T. et al. Biochemical properties and subcellular distribution of the neuronal class E calcium channel alpha 1 subunit. J. Neurosci. 15, 6419–6432 (1995).

    Article  CAS  Google Scholar 

  19. Wakamori, M. et al. Single tottering mutations responsible for the neuropathic phenotype of the P-type calcium channel. J. Biol. Chem. 273, 34857–34867 (1998).

    Article  CAS  Google Scholar 

  20. Catterall, W.A. Structure and regulation of voltage-gated Ca2+ channels. Annu. Rev. Cell Dev. Biol. 16, 521–555 (2000).

    Article  CAS  Google Scholar 

  21. Woermann, F.G., Sisosiya, S.M., Free, S.L. & Duncan, J.S. Quantitative MRI in patients with idiopathic generalized epilepsy - Evidence of widespread cerebral structural changes. Brain 121, 1661–1667 (1998).

    Article  Google Scholar 

  22. Meencke, H.J. & Veith, G. The relevance of slight migrational disturbances (microdysgenesis) to the etiology of the epilepsies. in Jasper's Basic Mechanisms of the Epilepsies, 3rd Edition, Advances in Neurology vol. 79 (eds. Delgado-Escueta, A.V., Wilson, W.A., Olsen, R.W. and Porter, R.J.) 123–131 (Lippincott Williams and Wilkins, Philadelphia, 1999).

    Google Scholar 

  23. Saegusa, H. et al. Altered responses in mice lacking α1E subunit of the voltage-dependent Ca2+ channel. Proc. Natl. Acad. Sci. USA 97, 6132–6137 (2000).

    Article  CAS  Google Scholar 

  24. Pennacchio, L.A. et al. Mutations in the gene encoding cystatin B in progressive myoclonus epilepsy (EPM1). Science 271, 1731–1734 (1996).

    Article  CAS  Google Scholar 

  25. Virtaneva, K. et al. Unstable minisatellite expansion causing recessively inherited myoclonus epilepsy, EPM1. Nat. Genet. 15, 393–396 (1997).

    Article  CAS  Google Scholar 

  26. Sugimoto, Y. et al. Childhood absence epilepsy in 8q24: refinement of candidate region and construction of physical map. Genomics 68, 264–272 (2000).

    Article  CAS  Google Scholar 

  27. Morita, R. et al. Exclusion of the JRK/JH8 gene as a candidate for human childhood absence epilepsy mapped on 8q24. Epilepsy Res. 37, 151–158 (1999).

    Article  CAS  Google Scholar 

  28. Niidome, T., Kim, M.S., Friedrich, T. & Mori, Y. Molecular cloning and characterization of a novel calcium channel from rabbit brain. FEBS Lett. 308, 7–13 (1992).

    Article  CAS  Google Scholar 

  29. Chen, Y. et al. Formation of an endophilin-Ca2+ channel complex is critical for clathrin-mediated synaptic vesicle endocytosis. Cell 115, 37–48 (2003).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank the family members for participating in this study; Y. Itsukaichi, K. Yamada, Y. Tsutsumi, K. Shoda, E. Mazaki, A. Nitta, N. Okamura, C. Uchikawa, M. Hishinuma and S.G. Pietsch for their help; and the Research Resources Center of RIKEN Brain Science Institute for DNA sequencing analysis and generation of rabbit polyclonal antibodies. This work was supported in part by a grant from RIKEN Brain Science Institute, Japan. A.V.D.E. is partly supported by a grant from the US National Institutes of Health.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Antonio V Delgado-Escueta or Kazuhiro Yamakawa.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Fig. 1

Amino acid sequence of EFHC1 and evolutionary conservation of amino acids mutated in JME. (PDF 25 kb)

Supplementary Fig. 2

Expression of mouse Efhc1 transcripts and proteins. (PDF 500 kb)

Supplementary Fig. 3

No effect of VDCC antagonists on neurons expressing GFP-controls and binding of EFHC1 mutants to CaV2.3. (PDF 642 kb)

Supplementary Table 1

Modulation of activation and inactivation properties of CaV2.3 by EFH1 proteins. (PDF 397 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Suzuki, T., Delgado-Escueta, A., Aguan, K. et al. Mutations in EFHC1 cause juvenile myoclonic epilepsy. Nat Genet 36, 842–849 (2004). https://doi.org/10.1038/ng1393

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng1393

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