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.

Deep sequencing reveals 50 novel genes for recessive cognitive disorders

Abstract

Common diseases are often complex because they are genetically heterogeneous, with many different genetic defects giving rise to clinically indistinguishable phenotypes. This has been amply documented for early-onset cognitive impairment, or intellectual disability, one of the most complex disorders known and a very important health care problem worldwide. More than 90 different gene defects have been identified for X-chromosome-linked intellectual disability alone, but research into the more frequent autosomal forms of intellectual disability is still in its infancy. To expedite the molecular elucidation of autosomal-recessive intellectual disability, we have now performed homozygosity mapping, exon enrichment and next-generation sequencing in 136 consanguineous families with autosomal-recessive intellectual disability from Iran and elsewhere. This study, the largest published so far, has revealed additional mutations in 23 genes previously implicated in intellectual disability or related neurological disorders, as well as single, probably disease-causing variants in 50 novel candidate genes. Proteins encoded by several of these genes interact directly with products of known intellectual disability genes, and many are involved in fundamental cellular processes such as transcription and translation, cell-cycle control, energy metabolism and fatty-acid synthesis, which seem to be pivotal for normal brain development and function.

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: Known and novel intellectual disability genes form protein and regulatory networks.

Accession codes

Primary accessions

Sequence Read Archive

Data deposits

Raw sequencing data can be retrieved from the Sequence Read Archive (SRA), accession number SRA036250.

References

  1. Ropers, H. H. Genetics of early onset cognitive impairment. Annu. Rev. Genomics Hum. Genet. 11, 161–187 (2010)

    Article  CAS  Google Scholar 

  2. Hamdan, F. F. et al. Excess of de novo deleterious mutations in genes associated with glutamatergic systems in nonsyndromic intellectual disability. Am. J. Hum. Genet. 88, 306–316 (2011)

    Article  CAS  Google Scholar 

  3. Hamdan, F. F. et al. Mutations in SYNGAP1 in autosomal nonsyndromic mental retardation. N. Engl. J. Med. 360, 599–605 (2009)

    Article  CAS  Google Scholar 

  4. Vissers, L. E. et al. A de novo paradigm for mental retardation. Nature Genet. 42, 1109–1112 (2010)

    Article  CAS  Google Scholar 

  5. Najmabadi, H. et al. Homozygosity mapping in consanguineous families reveals extreme heterogeneity of non-syndromic autosomal recessive mental retardation and identifies 8 novel gene loci. Hum. Genet. 121, 43–48 (2007)

    Article  CAS  Google Scholar 

  6. Kuss, A. W. et al. Autosomal recessive mental retardation: homozygosity mapping identifies 27 single linkage intervals, at least 14 novel loci and several mutation hotspots. Hum. Genet. 129, 141–148 (2011)

    Article  Google Scholar 

  7. Kaufman, L., Ayub, M. & Vincent, J. B. The genetic basis of non-syndromic intellectual disability: a review. J. Neurodev. Disord. 2, 182–209 (2010)

    Article  Google Scholar 

  8. Ng, S. B. et al. Exome sequencing identifies MLL2 mutations as a cause of Kabuki syndrome. Nature Genet. 42, 790–793 (2010)

    Article  CAS  Google Scholar 

  9. Ng, S. B. et al. Exome sequencing identifies the cause of a mendelian disorder. Nature Genet. 42, 30–35 (2010)

    Article  CAS  Google Scholar 

  10. Lupski, J. R. et al. Whole-genome sequencing in a patient with Charcot-Marie-Tooth neuropathy. N. Engl. J. Med. 362, 1181–1191 (2010)

    Article  CAS  Google Scholar 

  11. Cantagrel, V. et al. SRD5A3 is required for converting polyprenol to dolichol and is mutated in a congenital glycosylation disorder. Cell 142, 203–217 (2010)

    Article  CAS  Google Scholar 

  12. Kahrizi, K. et al. Next generation sequencing in a family with autosomal recessive Kahrizi syndrome (OMIM 612713) reveals a homozygous frameshift mutation in SRD5A3 . Eur. J. Hum. Genet. 19, 115–117 (2011)

    Article  CAS  Google Scholar 

  13. Raas-Rothschild, A. et al. A PEX6-defective peroxisomal biogenesis disorder with severe phenotype in an infant, versus mild phenotype resembling Usher syndrome in the affected parents. Am. J. Hum. Genet. 70, 1062–1068 (2002)

    Article  CAS  Google Scholar 

  14. Steinfeld, R. et al. Folate receptor alpha defect causes cerebral folate transport deficiency: a treatable neurodegenerative disorder associated with disturbed myelin metabolism. Am. J. Hum. Genet. 85, 354–363 (2009)

    Article  CAS  Google Scholar 

  15. Shoichet, S. A. et al. Mutations in the ZNF41 gene are associated with cognitive deficits: identification of a new candidate for X-linked mental retardation. Am. J. Hum. Genet. 73, 1341–1354 (2003)

    Article  CAS  Google Scholar 

  16. Jensen, L. R. et al. Mutations in the JARID1C gene, which is involved in transcriptional regulation and chromatin remodeling, cause X-linked mental retardation. Am. J. Hum. Genet. 76, 227–236 (2005)

    Article  CAS  Google Scholar 

  17. Freude, K. et al. Mutations in the FTSJ1 gene coding for a novel S-adenosylmethionine-binding protein cause nonsyndromic X-linked mental retardation. Am. J. Hum. Genet. 75, 305–309 (2004)

    Article  CAS  Google Scholar 

  18. Ramser, J. et al. A splice site mutation in the methyltransferase gene FTSJ1 in Xp11.23 is associated with non-syndromic mental retardation in a large Belgian family (MRX9). J. Med. Genet. 41, 679–683 (2004)

    Article  CAS  Google Scholar 

  19. Klein, U., Ramirez, M. T., Kobilka, B. K. & von Zastrow, M. A novel interaction between adrenergic receptors and the α-subunit of eukaryotic initiation factor 2B. J. Biol. Chem. 272, 19099–19102 (1997)

    Article  CAS  Google Scholar 

  20. Schwarz, J. M., Rodelsperger, C., Schuelke, M. & Seelow, D. MutationTaster evaluates disease-causing potential of sequence alterations. Nature Methods 7, 575–576 (2010)

    Article  CAS  Google Scholar 

  21. Geiduschek, E. P. & Kassavetis, G. A. The RNA polymerase III transcription apparatus. J. Mol. Biol. 310, 1–26 (2001)

    Article  CAS  Google Scholar 

  22. Hurov, K. E., Cotta-Ramusino, C. & Elledge, S. J. A genetic screen identifies the Triple T complex required for DNA damage signaling and ATM and ATR stability. Genes Dev. 24, 1939–1950 (2010)

    Article  CAS  Google Scholar 

  23. Zamponi, G. W., Lory, P. & Perez-Reyes, E. Role of voltage-gated calcium channels in epilepsy. Pflugers Arch. 460, 395–403 (2010)

    Article  CAS  Google Scholar 

  24. Singh, B. et al. Mutational analysis of CACNA1G in idiopathic generalized epilepsy. Hum. Mutat. 28, 524–525 (2007)

    Article  Google Scholar 

  25. Södersten, E., Lilja, T. & Hermanson, O. The novel BTB/POZ and zinc finger factor Zbtb45 is essential for proper glial differentiation of neural and oligodendrocyte progenitor cells. Cell Cycle 9, 4866–4875 (2010)

    Article  Google Scholar 

  26. Zhu, X. et al. Disruption of PC1/3 expression in mice causes dwarfism and multiple neuroendocrine peptide processing defects. Proc. Natl Acad. Sci. USA 99, 10293–10298 (2002)

    Article  CAS  ADS  Google Scholar 

  27. Billuart, P. et al. Oligophrenin-1 encodes a rhoGAP protein involved in X-linked mental retardation. Nature 392, 923–926 (1998)

    Article  CAS  ADS  Google Scholar 

  28. D’Adamo, P. et al. Mutations in GDI1 are responsible for X-linked non-specific mental retardation. Nature Genet. 19, 134–139 (1998)

    Article  Google Scholar 

  29. Jaffe, A. B., Aspenstrom, P. & Hall, A. Human CNK1 acts as a scaffold protein, linking Rho and Ras signal transduction pathways. Mol. Cell. Biol. 24, 1736–1746 (2004)

    Article  CAS  Google Scholar 

  30. Boison, D. et al. Neonatal hepatic steatosis by disruption of the adenosine kinase gene. Proc. Natl Acad. Sci. USA 99, 6985–6990 (2002)

    Article  CAS  ADS  Google Scholar 

  31. Tarpey, P. S. et al. A systematic, large-scale resequencing screen of X-chromosome coding exons in mental retardation. Nature Genet. 41, 535–543 (2009)

    Article  CAS  Google Scholar 

  32. Pak, C. et al. Mutation of the conserved polyadenosine RNA binding protein, ZC3H14/dNab2, impairs neural function in Drosophila and humans. Proc. Natl Acad. Sci. USA 108, 12390–12395 (2011)

    Article  CAS  ADS  Google Scholar 

  33. Garshasbi, M. et al. A defect in the TUSC3 gene is associated with autosomal recessive mental retardation. Am. J. Hum. Genet. 82, 1158–1164 (2008)

    Article  CAS  Google Scholar 

  34. Mir, A. et al. Identification of mutations in TRAPPC9, which encodes the NIK- and IKK-β-binding protein, in nonsyndromic autosomal-recessive mental retardation. Am. J. Hum. Genet. 85, 909–915 (2009)

    Article  CAS  Google Scholar 

  35. Moheb, L. A. et al. Identification of a nonsense mutation in the very low-density lipoprotein receptor gene (VLDLR) in an Iranian family with dysequilibrium syndrome. Eur. J. Hum. Genet. 16, 270–273 (2008)

    Article  CAS  Google Scholar 

  36. Motazacker, M. M. et al. A defect in the ionotropic glutamate receptor 6 gene (GRIK2) is associated with autosomal recessive mental retardation. Am. J. Hum. Genet. 81, 792–798 (2007)

    Article  CAS  Google Scholar 

  37. Laumonnier, F., Cuthbert, P. C. & Grant, S. G. The role of neuronal complexes in human X-linked brain diseases. Am. J. Hum. Genet. 80, 205–220 (2007)

    Article  CAS  Google Scholar 

  38. Ullmann, R. et al. Array CGH identifies reciprocal 16p13.1 duplications and deletions that predispose to autism and/or mental retardation. Hum. Mutat. 28, 674–682 (2007)

    Article  CAS  Google Scholar 

  39. Yildirim, Y. et al. A frameshift mutation of ERLIN2 in recessive intellectual disability, motor dysfunction and multiple joint contractures. Hum. Mol. Genet. 20, 1886–1892 (2011)

    Article  CAS  Google Scholar 

  40. Alazami, A. M., Adly, N., Al Dhalaan, H. & Alkuraya, F. S. A nullimorphic ERLIN2 mutation defines a complicated hereditary spastic paraplegia locus (SPG18). Neurogenetics 10.1007/s10048-011-0291-8 (2011)

  41. Abou Jamra, R. et al. Adaptor protein complex 4 deficiency causes severe autosomal-recessive intellectual disability, progressive spastic paraplegia, shy character, and short stature. Am. J. Hum. Genet. 88, 788–795 (2011)

    Article  CAS  Google Scholar 

  42. Rafiq, M. A. et al. Mutations in the alpha 1,2-mannosidase gene, MAN1B1, cause autosomal-recessive intellectual disability. Am. J. Hum. Genet. 89, 176–182 (2011)

    Article  CAS  Google Scholar 

  43. Putoux, A. et al. KIF7 mutations cause fetal hydrolethalus and acrocallosal syndromes. Nature Genet. 43, 601–606 (2011)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We express our gratitude to the patients and their families for their participation in the study. We thank S. Nakhee and K. Javan for their support; S. Arzhangi, S. Banihashemi, M. Kasiri, H. Khodaee, M. Schlicht and M. Gerloff for contributing to this project in various ways; G. Eder for her assistance with the preparation of the manuscript; and S. Shoichet for critically reading the manuscript. This work was supported by the Max Planck Innovation Funds, the German Federal Ministry of Education and Research through the MRNET (grant 01GS08161, to H.H.R.), the Iranian National Science Foundation and the EU-FP7 project GENCODYS.

Author information

Authors and Affiliations

Authors

Contributions

H.H.R. and H.N. initiated and directed this study. H.H., M.G., W.C., S.H., K.W., V.K., R.U., K.K. and A.W.K. contributed to its design and coordination. H.N., K.K., A.T., P.J., V.H., D.W., M.C., I.R., F.M., C.H., A.D., A.R., M.J.S.B., M.F. and H.D. recruited patients and families, and K.K. and A.T. were responsible for the clinical investigations. F.B., S.G.F. and R.K. did the karyotyping. M.G. performed the linkage analyses and together with R.W. and H.H., he established data management tools. R.U. and I.M. performed exon enrichments, and C.J. and M.B. did the deep sequencing experiments. H.H. analysed the sequencing data and provided bioinformatics support. M.G., S.S.A., M.H., A.Z., M.M., L.P., L.N.V., L.A.M., F.L., B.L., S.E.-N., Z.F., J.H., L.M. and A.W.K. participated in the validation of the results and the segregation analyses. T.Z. performed the pathway analyses. H.H., M.G., A.Z., L.P., R.W., T.Z., L.M., A.W.K., A.T., K.K., H.N. and H.H.R. evaluated and interpreted the results, and H.H.R., H.H. and M.G. wrote the manuscript.

Corresponding authors

Correspondence to Kimia Kahrizi or H. Hilger Ropers.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Information

The file contains Supplementary Methods, Supplementary Figures 1-2 with legends and additional references. (PDF 264 kb)

Supplementary Table 1

The table shows families information. (XLS 138 kb)

Supplementary Table 2

The table shows mutations information. (XLS 83 kb)

Supplementary Table 3

The table shows population specific control screening in novel (candidate) genes for ID. (XLS 25 kb)

Supplementary Table 4

The table shows a list of truncating mutations that did not co-segregate with ID. (XLS 17 kb)

Supplementary Table 5

The table shows table coverage for 136 ARMR samples in the targeted exons. (XLS 18 kb)

Supplementary Table 6

The table shows homozygous variants detected by SNP array and NGS in the targeted exons. (XLS 16 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Najmabadi, H., Hu, H., Garshasbi, M. et al. Deep sequencing reveals 50 novel genes for recessive cognitive disorders. Nature 478, 57–63 (2011). https://doi.org/10.1038/nature10423

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature10423

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