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 ISPD cause Walker-Warburg syndrome and defective glycosylation of α-dystroglycan

Abstract

Walker-Warburg syndrome (WWS) is an autosomal recessive multisystem disorder characterized by complex eye and brain abnormalities with congenital muscular dystrophy (CMD) and aberrant α-dystroglycan glycosylation. Here we report mutations in the ISPD gene (encoding isoprenoid synthase domain containing) as the second most common cause of WWS. Bacterial IspD is a nucleotidyl transferase belonging to a large glycosyltransferase family, but the role of the orthologous protein in chordates is obscure to date, as this phylum does not have the corresponding non-mevalonate isoprenoid biosynthesis pathway. Knockdown of ispd in zebrafish recapitulates the human WWS phenotype with hydrocephalus, reduced eye size, muscle degeneration and hypoglycosylated α-dystroglycan. These results implicate ISPD in α-dystroglycan glycosylation in maintaining sarcolemma integrity in vertebrates.

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: Overview of genetic data in the cohort of individuals with WWS.
Figure 2: Magnetic resonance images and muscle staining of subject WWS-160.
Figure 3: Knockdown of zebrafish ispd recapitulates pathological defects of human WWS.
Figure 4: Hypoglycosylation of α-dystroglycan and disrupted sarcolemma integrity in ispd MO1–injected zebrafish embryos.

Similar content being viewed by others

References

  1. van Reeuwijk, J., Brunner, H.G. & van Bokhoven, H. Glyc-O-genetics of Walker-Warburg syndrome. Clin. Genet. 67, 281–289 (2005).

    Article  CAS  PubMed  Google Scholar 

  2. Kobayashi, K. et al. An ancient retrotransposal insertion causes Fukuyama-type congenital muscular dystrophy. Nature 394, 388–392 (1998).

    Article  CAS  PubMed  Google Scholar 

  3. Yoshida, A. et al. Muscular dystrophy and neuronal migration disorder caused by mutations in a glycosyltransferase, POMGnT1. Dev. Cell 1, 717–724 (2001).

    Article  CAS  PubMed  Google Scholar 

  4. Beltrán-Valero de Bernabé, D. et al. Mutations in the O-mannosyltransferase gene POMT1 give rise to the severe neuronal migration disorder Walker-Warburg syndrome. Am. J. Hum. Genet. 71, 1033–1043 (2002).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Brockington, M. et al. Mutations in the fukutin-related protein gene (FKRP) cause a form of congenital muscular dystrophy with secondary laminin α2 deficiency and abnormal glycosylation of α-dystroglycan. Am. J. Hum. Genet. 69, 1198–1209 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Longman, C. et al. Mutations in the human LARGE gene cause MDC1D, a novel form of congenital muscular dystrophy with severe mental retardation and abnormal glycosylation of α-dystroglycan. Hum. Mol. Genet. 12, 2853–2861 (2003).

    Article  CAS  PubMed  Google Scholar 

  7. Beltrán-Valero de Bernabé, D. et al. Mutations in the FKRP gene can cause muscle-eye-brain disease and Walker-Warburg syndrome. J. Med. Genet. 41, e61 (2004).

    Article  PubMed  Google Scholar 

  8. van Reeuwijk, J. et al. POMT2 mutations cause α-dystroglycan hypoglycosylation and Walker-Warburg syndrome. J. Med. Genet. 42, 907–912 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. van Reeuwijk, J. et al. Intragenic deletion in the LARGE gene causes Walker-Warburg syndrome. Hum. Genet. 121, 685–690 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Silan, F. et al. A new mutation of the fukutin gene in a non-Japanese patient. Ann. Neurol. 53, 392–396 (2003).

    Article  CAS  PubMed  Google Scholar 

  11. Hara, Y. et al. A dystroglycan mutation associated with limb-girdle muscular dystrophy. N. Engl. J. Med. 364, 939–946 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Lefeber, D.J. et al. Deficiency of Dol-P-Man synthase subunit DPM3 bridges the congenital disorders of glycosylation with the dystroglycanopathies. Am. J. Hum. Genet. 85, 76–86 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Godfrey, C. et al. Refining genotype phenotype correlations in muscular dystrophies with defective glycosylation of dystroglycan. Brain 130, 2725–2735 (2007).

    Article  PubMed  Google Scholar 

  14. Jimenez-Mallebrera, C. et al. A comparative study of α-dystroglycan glycosylation in dystroglycanopathies suggests that the hypoglycosylation of α-dystroglycan does not consistently correlate with clinical severity. Brain Pathol. 19, 596–611 (2009).

    Article  CAS  PubMed  Google Scholar 

  15. Lin, Y.Y. et al. Zebrafish Fukutin family proteins link the unfolded protein response with dystroglycanopathies. Hum. Mol. Genet. 20, 1763–1775 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Zhang, Z., Zhang, P. & Hu, H. LARGE expression augments the glycosylation of glycoproteins in addition to α-dystroglycan conferring laminin binding. PLoS ONE 6, e19080 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Bleckmann, C. et al. O-glycosylation pattern of CD24 from mouse brain. Biol. Chem. 390, 627–645 (2009).

    Article  CAS  PubMed  Google Scholar 

  18. Gilissen, C. et al. Exome sequencing identifies WDR35 variants involved in Sensenbrenner syndrome. Am. J. Hum. Genet. 87, 418–423 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Liu, J. & Mushegian, A. Three monophyletic superfamilies account for the majority of the known glycosyltransferases. Protein Sci. 12, 1418–1431 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Michele, D.E. et al. Post-translational disruption of dystroglycan-ligand interactions in congenital muscular dystrophies. Nature 418, 417–422 (2002).

    Article  CAS  PubMed  Google Scholar 

  21. Snow, C.J. et al. Time-lapse analysis and mathematical characterization elucidate novel mechanisms underlying muscle morphogenesis. PLoS Genet. 4, e1000219 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Parsons, M.J. et al. Zebrafish mutants identify an essential role for laminins in notochord formation. Development 129, 3137–3146 (2002).

    CAS  PubMed  Google Scholar 

  23. Bassett, D.I. et al. Dystrophin is required for the formation of stable muscle attachments in the zebrafish embryo. Development 130, 5851–5860 (2003).

    Article  CAS  PubMed  Google Scholar 

  24. Straub, V., Rafael, J.A., Chamberlain, J.S. & Campbell, K.P. Animal models for muscular dystrophy show different patterns of sarcolemmal disruption. J. Cell Biol. 139, 375–385 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Richard, S.B. et al. Kinetic analysis of Escherichia coli 2-C-methyl-D-erythritol-4-phosphate cytidyltransferase, wild type and mutants, reveals roles of active site amino acids. Biochemistry 43, 12189–12197 (2004).

    Article  CAS  PubMed  Google Scholar 

  26. Baur, S., Marles-Wright, J., Buckenmaier, S., Lewis, R.J. & Vollmer, W. Synthesis of CDP-activated ribitol for teichoic acid precursors in Streptococcus pneumoniae. J. Bacteriol. 191, 1200–1210 (2009).

    Article  CAS  PubMed  Google Scholar 

  27. MacLeod, H. et al. A novel FKRP mutation in congenital muscular dystrophy disrupts the dystrophin glycoprotein complex. Neuromuscul. Disord. 17, 285–289 (2007).

    Article  PubMed  Google Scholar 

  28. Wood, A.J. et al. Abnormal vascular development in zebrafish models for fukutin and FKRP deficiency. Hum. Mol. Genet. 20, 4879–4890 (2011).

    Article  CAS  PubMed  Google Scholar 

  29. McMullan, D.J. et al. Molecular karyotyping of patients with unexplained mental retardation by SNP arrays: a multicenter study. Hum. Mutat. 30, 1082–1092 (2009).

    Article  CAS  PubMed  Google Scholar 

  30. Woods, C.G. et al. Quantification of homozygosity in consanguineous individuals with autosomal recessive disease. Am. J. Hum. Genet. 78, 889–896 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Parsons, M.J., Campos, I., Hirst, E.M. & Stemple, D.L. Removal of dystroglycan causes severe muscular dystrophy in zebrafish embryos. Development 129, 3505–3512 (2002).

    CAS  PubMed  Google Scholar 

  32. Link, V., Shevchenko, A. & Heisenberg, C.P. Proteomics of early zebrafish embryos. BMC Dev. Biol. 6, 1 (2006).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Robu, M.E. et al. p53 activation by knockdown technologies. PLoS Genet. 3, e78 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Ciruna, B., Jenny, A., Lee, D., Mlodzik, M. & Schier, A.F. Planar cell polarity signalling couples cell division and morphogenesis during neurulation. Nature 439, 220–224 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Hall, T.E. et al. The zebrafish candyfloss mutant implicates extracellular matrix adhesion failure in laminin α2–deficient congenital muscular dystrophy. Proc. Natl. Acad. Sci. USA 104, 7092–7097 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Venselaar, H., Te Beek, T.A., Kuipers, R.K., Hekkelman, M.L. & Vriend, G. Protein structure analysis of mutations causing inheritable diseases. An e-Science approach with life scientist friendly interfaces. BMC Bioinformatics 11, 548 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Kemp, L.E., Bond, C.S. & Hunter, W.N. Structure of a tetragonal crystal form of Escherichia coli 2-C-methyl-D-erythritol 4-phosphate cytidylyltransferase. Acta Crystallogr. D Biol. Crystallogr. 59, 607–610 (2003).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We thank all family members who participated in this study. We would also like to thank A. Charon for referring an affected individual, G. Powell and S. Gerety for critical comments on the manuscript and R. Schot for study of the deletion in family WWS-161. This project was supported by the Large-Scale Integrating Project GENCODYS-Genetic and Epigenetic Networks in COgnitive DYSfunction (241995), which is funded by the European Union Framework Programme 7 (FP7) Health program (to H.v.B.), the Australian National Health and Medical Research Council (NHMRC) with an overseas postdoctoral fellowship (to T.R.), The Prinses Beatrix Fund (grant W.OR09-15 to D.L. and H.v.B.), the Hersenstichting Nederland (to H.v.B.) and a European Molecular Biology Organization (EMBO) Long-Term Fellowship (ALTF 805-2009 to K.B.). All zebrafish work was sponsored by the Wellcome Trust (grant WT 077047/Z/05/Z and WT 077037/Z/05/Z). Next-generation sequencing experiments were financially supported by the Department of Human Genetics at Nijmegen, as well as by the Netherlands Organisation for Health Research and Development (ZonMW; grant 916-86-016 to L.E.L.M.V.).

Author information

Authors and Affiliations

Authors

Contributions

The study was designed and results were interpreted by T.R., K.B., D.L.S., Y.-Y.L., H.G.B., D.J.L. and H.v.B. Subject ascertainment and recruitment were carried out by I.M., U.A., B.G., G.M.S.M., P.D., M.A.W., D.P.R., D.C., C.B., E.S., E.A.J.P., G.M.B.T.-S., C.E.d.D.-S., K.D., H.K., O.A.E.-F.E.-H. and H.v.B. Sequencing, CNV analysis and genotyping were carried out and interpreted by T.R., E.-J.K., K.B., I.M., J.v.R., C.v.d.E., E.v.B., M.R., R.P., L.E.L.M.V., M.S., M.F.B., H.Z., J.A.V., C.G., G.M.S.M. and H.v.B. Zebrafish studies were designed and carried out by K.B., D.L.S. and Y.-Y.L. Bioinformatic data analysis and protein modeling were performed by T.R., J.v.R., C.G. and D.J.L. The manuscript was drafted by T.R., K.B., D.J.L., Y.-Y.L. and H.v.B. All authors contributed to the final version of the paper.

Corresponding authors

Correspondence to Yung-Yao Lin or Hans van Bokhoven.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 and Supplementary Tables 1 and 2 (PDF 2560 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Roscioli, T., Kamsteeg, EJ., Buysse, K. et al. Mutations in ISPD cause Walker-Warburg syndrome and defective glycosylation of α-dystroglycan. Nat Genet 44, 581–585 (2012). https://doi.org/10.1038/ng.2253

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.2253

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