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.

Mutations in GRIN2A and GRIN2B encoding regulatory subunits of NMDA receptors cause variable neurodevelopmental phenotypes

Abstract

N-methyl-D-aspartate (NMDA) receptors mediate excitatory neurotransmission in the mammalian brain. Two glycine-binding NR1 subunits and two glutamate-binding NR2 subunits each form highly Ca2+-permeable cation channels which are blocked by extracellular Mg2+ in a voltage-dependent manner1. Either GRIN2B or GRIN2A, encoding the NMDA receptor subunits NR2B and NR2A, was found to be disrupted by chromosome translocation breakpoints in individuals with mental retardation and/or epilepsy. Sequencing of GRIN2B in 468 individuals with mental retardation revealed four de novo mutations: a frameshift, a missense and two splice-site mutations. In another cohort of 127 individuals with idiopathic epilepsy and/or mental retardation, we discovered a GRIN2A nonsense mutation in a three-generation family. In a girl with early-onset epileptic encephalopathy, we identified the de novo GRIN2A mutation c.1845C>A predicting the amino acid substitution p.N615K. Analysis of NR1-NR2AN615K (NR2A subunit with the p.N615K alteration) receptor currents revealed a loss of the Mg2+ block and a decrease in Ca2+ permeability. Our findings suggest that disturbances in the neuronal electrophysiological balance during development result in variable neurological phenotypes depending on which NR2 subunit of NMDA receptors is affected.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: Disruption of either GRIN2A or GRIN2B in subjects with chromosome translocations and different neurodevelopmental phenotypes.
Figure 2: Transcript analysis for the mutations c.411+1G>A, c.2360-2A>G and c.803_804delCA in GRIN2B and c.652C>T in GRIN2A.
Figure 3: Structural and functional consequences of missense mutations in GRIN2B and GRIN2A found in subjects with mental retardation and/or epilepsy.

Similar content being viewed by others

Accession codes

Accessions

GenBank/EMBL/DDBJ

Protein Data Bank

References

  1. Cull-Candy, S., Brickley, S. & Farrant, M. NMDA receptor subunits: diversity, development and disease. Curr. Opin. Neurobiol. 11, 327–335 (2001).

    Google Scholar 

  2. Cull-Candy, S.G. & Leszkiewicz, D.N. Role of distinct NMDA receptor subtypes at central synapses. Sci. STKE 2004, re16 (2004).

    Google Scholar 

  3. Lau, C.G. & Zukin, R.S. NMDA receptor trafficking in synaptic plasticity and neuropsychiatric disorders. Nat. Rev. Neurosci. 8, 413–426 (2007).

    Google Scholar 

  4. Kurotaki, N. et al. Haploinsufficiency of NSD1 causes Sotos syndrome. Nat. Genet. 30, 365–366 (2002).

    Google Scholar 

  5. Najm, J. et al. Mutations of CASK cause an X-linked brain malformation phenotype with microcephaly and hypoplasia of the brainstem and cerebellum. Nat. Genet. 40, 1065–1067 (2008).

    Google Scholar 

  6. Ray, P.N. et al. Cloning of the breakpoint of an X;21 translocation associated with Duchenne muscular dystrophy. Nature 318, 672–675 (1985).

    Google Scholar 

  7. Tonkin, E.T. et al. NIPBL, encoding a homolog of fungal Scc2-type sister chromatid cohesion proteins and fly Nipped-B, is mutated in Cornelia de Lange syndrome. Nat. Genet. 36, 636–641 (2004).

    Google Scholar 

  8. Akashi, K. et al. NMDA receptor GluN2B (GluR epsilon 2/NR2B) subunit is crucial for channel function, postsynaptic macromolecular organization, and actin cytoskeleton at hippocampal CA3 synapses. J. Neurosci. 29, 10869–10882 (2009).

    Google Scholar 

  9. Tang, Y.P. et al. Genetic enhancement of learning and memory in mice. Nature 401, 63–69 (1999).

    Google Scholar 

  10. Wang, D. et al. Genetic enhancement of memory and long-term potentiation but not CA1 long-term depression in NR2B transgenic rats. PLoS ONE 4, e7486 (2009).

    Google Scholar 

  11. Reutlinger, C. et al. Deletions in 16p13 including GRIN2A in patients with intellectual disability, various dysmorphic features and seizure disorder of the rolandic region. Epilepsia published online, doi:10.1111/j.1528-1167.2010.02555.x (2 April 2010).

  12. Sakimura, K. et al. Reduced hippocampal LTP and spatial learning in mice lacking NMDA receptor epsilon 1 subunit. Nature 373, 151–155 (1995).

    Google Scholar 

  13. Kiyama, Y. et al. Increased thresholds for long-term potentiation and contextual learning in mice lacking the NMDA-type glutamate receptor epsilon1 subunit. J. Neurosci. 18, 6704–6712 (1998).

    Google Scholar 

  14. Kutsuwada, T. et al. Impairment of suckling response, trigeminal neuronal pattern formation, and hippocampal LTD in NMDA receptor epsilon 2 subunit mutant mice. Neuron 16, 333–344 (1996).

    Google Scholar 

  15. Wang, G.S. & Cooper, T.A. Splicing in disease: disruption of the splicing code and the decoding machinery. Nat. Rev. Genet. 8, 749–761 (2007).

    Google Scholar 

  16. Laube, B. et al. Molecular determinants of agonist discrimination by NMDA receptor subunits: analysis of the glutamate binding site on the NR2B subunit. Neuron 18, 493–503 (1997).

    Google Scholar 

  17. Laube, B., Schemm, R. & Betz, H. Molecular determinants of ligand discrimination in the glutamate-binding pocket of the NMDA receptor. Neuropharmacology 47, 994–1007 (2004).

    Google Scholar 

  18. Mayer, M.L. Glutamate receptors at atomic resolution. Nature 440, 456–462 (2006).

    Google Scholar 

  19. Wollmuth, L.P., Kuner, T. & Sakmann, B. Adjacent asparagines in the NR2-subunit of the NMDA receptor channel control the voltage-dependent block by extracellular Mg2+. J. Physiol. (Lond.) 506, 13–32 (1998).

    Google Scholar 

  20. Yashiro, K. & Philpot, B.D. Regulation of NMDA receptor subunit expression and its implications for LTD, LTP, and metaplasticity. Neuropharmacology 55, 1081–1094 (2008).

    Google Scholar 

  21. Ding, Y.X. et al. A possible association of responsiveness to adrenocorticotropic hormone with specific GRIN1 haplotypes in infantile spasms. Dev. Med. Child Neurol. published online, doi:10.1111/j.1469-8749.2010.03746.x (16 August 2010).

  22. Barrett, M.T. et al. Comparative genomic hybridization using oligonucleotide microarrays and total genomic DNA. Proc. Natl. Acad. Sci. USA 101, 17765–17770 (2004).

    Google Scholar 

  23. Spitz, R. et al. Oligonucleotide array-based comparative genomic hybridization (aCGH) of 90 neuroblastomas reveals aberration patterns closely associated with relapse pattern and outcome. Genes Chromosom. Cancer 45, 1130–1142 (2006).

    Google Scholar 

  24. Chen, W. et al. CGHPRO–a comprehensive data analysis tool for array CGH. BMC Bioinformatics 6, 85 (2005).

    Google Scholar 

  25. Desmet, F.O. et al. Human Splicing Finder: an online bioinformatics tool to predict splicing signals. Nucleic Acids Res. 37, e67 (2009).

    Google Scholar 

  26. Reese, M.G., Eeckman, F.H., Kulp, D. & Haussler, D. Improved splice site detection in Genie. J. Comput. Biol. 4, 311–323 (1997).

    Google Scholar 

  27. Hebsgaard, S.M. et al. Splice site prediction in Arabidopsis thaliana pre-mRNA by combining local and global sequence information. Nucleic Acids Res. 24, 3439–3452 (1996).

    Google Scholar 

  28. 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).

    Google Scholar 

  29. Ramensky, V., Bork, P. & Sunyaev, S. Human non-synonymous SNPs: server and survey. Nucleic Acids Res. 30, 3894–3900 (2002).

    Google Scholar 

  30. Bromberg, Y. & Rost, B. SNAP: predict effect of non-synonymous polymorphisms on function. Nucleic Acids Res. 35, 3823–3835 (2007).

    Google Scholar 

  31. Thomas, P.D. et al. PANTHER: a library of protein families and subfamilies indexed by function. Genome Res. 13, 2129–2141 (2003).

    Google Scholar 

  32. Sobolevsky, A.I., Rosconi, M.P. & Gouaux, E. X-ray structure, symmetry and mechanism of an AMPA-subtype glutamate receptor. Nature 462, 745–756 (2009).

    Google Scholar 

  33. Furukawa, H., Singh, S.K., Mancusso, R. & Gouaux, E. Subunit arrangement and function in NMDA receptors. Nature 438, 185–192 (2005).

    Google Scholar 

  34. Fiser, A. & Sali, A. Modeller: generation and refinement of homology-based protein structure models. Methods Enzymol. 374, 461–491 (2003).

    Google Scholar 

  35. Kleywegt, G.J. Use of non-crystallographic symmetry in protein structure refinement. Acta Crystallogr. D Biol. Crystallogr. 52, 842–857 (1996).

    Google Scholar 

  36. Schüler, T. et al. Formation of NR1/NR2 and NR1/NR3 heterodimers constitutes the initial step in N-methyl-D-aspartate receptor assembly. J. Biol. Chem. 283, 37–46 (2008).

    Google Scholar 

  37. Haeger, S. et al. An intramembrane aromatic network determines pentameric assembly of Cys-loop receptors. Nat. Struct. Mol. Biol. 17, 90–98 (2010).

    Google Scholar 

  38. Madry, C., Betz, H., Geiger, J.R. & Laube, B. Potentiation of glycine-gated NR1/NR3A NMDA receptors relieves Ca-dependent outward rectification. Front. Mol. Neurosci. 3, 6 (2010).

    Google Scholar 

Download references

Acknowledgements

We thank all subjects and healthy (control) individuals for their participation in this study; P. De Jonghe for providing DNA samples of individuals with idiopathic epilepsy; A. Gal and B. Horsthemke for continuous support; H.-H. Richardt and H. Petri for clinical evaluation of subjects 3 and 1; A. Tzschach and M. Hoeltzenbein for initial help with collecting clinical data of subject 2; S. Fuchs for chromosome analysis; B. Lübker and C. Menzel for FISH analysis; R. Ullmann and A. Ahmed for array comparative genomic hybridization (CGH) on subject 2, O. Riess and M. Bonin for copy number variation analysis on subjects 8 and 9, and I. Göhring and A.B. Ekici for microarray analysis of the Erlanger cohort; S. Berkel, J. Hoyer and K. Cremer for support with data maintenance; and A. Diem, S. Freier, I. Jantke, S. Meien and K. Ziegler for skillfull technical assistance. This work was part of the German Mental Retardation Network (MRNET) funded through a grant from the German Ministry of Research and Education to A. Rauch and A. Reis (01GS08160), G. Rappold (01GS08168-9), H.-H.R. (01GS08161) and D.W. (01GS08164). M.M. was supported by the Fondation Française pour la Recherche sur l'Epilepsie, B.L. was supported by the Gemeinnützige Hertie-Stiftung, and G. Rosenberger and K.K. were supported by a grant from the Deutsche Forschungsgemeinschaft (FOR 885/IRP5).

Author information

Authors and Affiliations

Authors

Contributions

Mutation analysis: S.E., B.P. and G. Rosenberger. Transcript analysis: K.K. and G. Rosenberger. Functional analysis of NMDA receptors: K.G. and B.L. NMDA receptor modeling: C.T. and B.L. Subject ascertainment: Y.H., L.V.M., M.M., U.M., G. Rappold, A. Rauch, S.v.S., I.S., N.V., L.V., D.W., B.Z. and M.Z. FISH analysis and breakpoint mapping: A.F., V.M.K., F.K., F.K.P. and H.-H.R. Array CGH analysis: J.K. and H.T. Manuscript writing: K.K., V.M.K., B.L., L.V.M., A. Reis, G. Rosenberger and D.W. Study design: K.K., L.V.M., A. Reis, G. Rosenberger and D.W. All authors contributed to the final version of the paper.

Corresponding author

Correspondence to Kerstin Kutsche.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–5, Supplementary Tables 1–8 and Supplementary Note (PDF 520 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Endele, S., Rosenberger, G., Geider, K. et al. Mutations in GRIN2A and GRIN2B encoding regulatory subunits of NMDA receptors cause variable neurodevelopmental phenotypes. Nat Genet 42, 1021–1026 (2010). https://doi.org/10.1038/ng.677

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.677

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