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:

Germline loss-of-function mutations in SPRED1 cause a neurofibromatosis 1–like phenotype

Abstract

We report germline loss-of-function mutations in SPRED1 in a newly identified autosomal dominant human disorder. SPRED1 is a member of the SPROUTY/SPRED family1 of proteins that act as negative regulators of RAS->RAF interaction and mitogen-activated protein kinase (MAPK) signaling2. The clinical features of the reported disorder resemble those of neurofibromatosis type 1 and consist of multiple café-au-lait spots, axillary freckling and macrocephaly. Melanocytes from a café-au-lait spot showed, in addition to the germline SPRED1 mutation, an acquired somatic mutation in the wild-type SPRED1 allele, indicating that complete SPRED1 inactivation is needed to generate a café-au-lait spot in this syndrome. This disorder is yet another member of the recently characterized group of phenotypically overlapping syndromes caused by mutations in the genes encoding key components of the RAS-MAPK pathway3,4. To our knowledge, this is the first report of mutations in the SPRY (SPROUTY)/SPRED family of genes in human disease.

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: Pedigrees and clinical photographs.
Figure 2: Exon-intron structure of human SPRED1 showing noncoding sequences as open boxes and protein-coding exons as filled black boxes.
Figure 3: Effect of SPRED1 on the RAS-MAPK pathway.

Similar content being viewed by others

References

  1. Bundschu, K., Walter, U. & Schuh, K. The VASP-Spred-Sprouty domain puzzle. J. Biol. Chem. 281, 36477–36481 (2006).

    Article  CAS  Google Scholar 

  2. Wakioka, T. et al. Spred is a Sprouty-related suppressor of Ras signalling. Nature 412, 647–651 (2001).

    Article  CAS  Google Scholar 

  3. Shannon, K. & Bollag, G. Sending out an SOS. Nat. Genet. 39, 8–9 (2007).

    Article  CAS  Google Scholar 

  4. Bentires-Alj, M., Kontaridis, M.I. & Neel, B.G. Stops along the RAS pathway in human genetic disease. Nat. Med. 12, 283–285 (2006).

    Article  CAS  Google Scholar 

  5. Legius, E., Marchuk, D.A., Collins, F.S. & Glover, T.W. Somatic deletion of the neurofibromatosis type 1 gene in a neurofibrosarcoma supports a tumour suppressor gene hypothesis. Nat. Genet. 3, 122–126 (1993).

    Article  CAS  Google Scholar 

  6. Cichowski, K. & Jacks, T. NF1 tumor suppressor gene function: narrowing the GAP. Cell 104, 593–604 (2001).

    Article  CAS  Google Scholar 

  7. Tartaglia, M. et al. Mutations in PTPN11, encoding the protein tyrosine phosphatase SHP-2, cause Noonan syndrome. Nat. Genet. 29, 465–468 (2001).

    Article  CAS  Google Scholar 

  8. Schubbert, S. et al. Germline KRAS mutations cause Noonan syndrome. Nat. Genet. 38, 331–336 (2006).

    Article  CAS  Google Scholar 

  9. Roberts, A.E. et al. Germline gain-of-function mutations in SOS1 cause Noonan syndrome. Nat. Genet. 39, 70–74 (2007).

    Article  CAS  Google Scholar 

  10. Tartaglia, M. et al. Gain-of-function SOS1 mutations cause a distinctive form of Noonan syndrome. Nat. Genet. 39, 75–79 (2007).

    Article  CAS  Google Scholar 

  11. Digilio, M.C. et al. Grouping of multiple-lentigines/LEOPARD and Noonan syndromes on the PTPN11 gene. Am. J. Hum. Genet. 71, 389–394 (2002).

    Article  CAS  Google Scholar 

  12. Legius, E. et al. PTPN11 mutations in LEOPARD syndrome. J. Med. Genet. 39, 571–574 (2002).

    Article  CAS  Google Scholar 

  13. Aoki, Y. et al. Germline mutations in HRAS proto-oncogene cause Costello syndrome. Nat. Genet. 37, 1038–1040 (2005).

    Article  CAS  Google Scholar 

  14. Niihori, T. et al. Germline KRAS and BRAF mutations in cardio-facio-cutaneous syndrome. Nat. Genet. 38, 294–296 (2006).

    Article  CAS  Google Scholar 

  15. Rodriguez-Viciana, P. et al. Germline mutations in genes within the MAPK pathway cause cardio-facio-cutaneous syndrome. Science 311, 1287–1290 (2006).

    Article  CAS  Google Scholar 

  16. Messiaen, L.M. et al. Exhaustive mutation analysis of the NF1 gene allows identification of 95% of mutations and reveals a high frequency of unusual splicing defects. Hum. Mutat. 15, 541–555 (2000).

    Article  CAS  Google Scholar 

  17. De Schepper, S. et al. Cafe-au-lait spots in neurofibromatosis type 1 and in healthy control individuals: hyperpigmentation of a different kind? Arch. Dermatol. Res. 297, 439–449 (2006).

    Article  Google Scholar 

  18. Gille, H. et al. ERK phosphorylation potentiates Elk-1-mediated ternary complex formation and transactivation. EMBO J. 14, 951–962 (1995).

    Article  CAS  Google Scholar 

  19. King, J.A.J. et al. Distinct requirements for the Sprouty domain for functional activity of Spred proteins. Biochem. J. 388, 445–454 (2005).

    Article  CAS  Google Scholar 

  20. Miyoshi, K. et al. The Sprouty-related protein, Spred, inhibits cell motility, metastasis, and Rho-mediated actin reorganization. Oncogene 23, 5567–5576 (2004).

    Article  CAS  Google Scholar 

  21. Engelhardt, C.M. et al. Expression and subcellular localization of Spred proteins in mouse and human tissues. Histochem. Cell Biol. 122, 527–538 (2004).

    Article  CAS  Google Scholar 

  22. Inoue, H. et al. Spred-1 negatively regulates allergen-induced airway eosinophilia and hyperresponsiveness. J. Exp. Med. 201, 73–82 (2005).

    Article  CAS  Google Scholar 

  23. Stumpf, D. et al. Neurofibromatosis conference statement. National Institutes of Health Consensus Development Conference. Arch. Neurol. 45, 575–578 (1988).

    Article  Google Scholar 

  24. Maertens, O. et al. Molecular dissection of isolated disease features in mosaic neurofibromatosis type 1. Am. J. Hum. Genet. 81, 243–251 (2007).

    Article  CAS  Google Scholar 

  25. Taniguchi, K. et al. Spreds are essential for embryonic lymphangiogenesis by regulating vascular endothelial growth factor receptor 3 signaling. Mol. Cell. Biol. 27, 4541–4550 (2007).

    Article  CAS  Google Scholar 

  26. Sasaki, A. et al. Identification of a dominant negative mutant of Sprouty that potentiates fibroblast growth factor- but not epidermal growth factor-induced ERK activation. J. Biol. Chem. 276, 36804–36808 (2001).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors thank T. de Ravel for critically reading the manuscript, M. De Mil for technical assistance in melanocyte cell culture and F. Okamoto for technical assistance in cell culture and plasmid preparation. H.B. is supported by the Institute for the Promotion of Innovation through Science and Technology in Flanders (IWT-Vlaanderen). M.C. was supported by the Marie Curie European Community fellowship (contract HPMT-CT2001-00273). E.D. is a predoctoral researcher (Aspirant of the Fonds voor Wetenschappelijk Onderzoek-Vlaanderen), J.C. is a postdoctoral researcher and E.L. is a part-time clinical researcher of the Fonds voor Wetenschappelijk Onderzoek-Vlaanderen (FWO). This work is also supported by research grants from the Fonds voor Wetenschappelijk Onderzoek Vlaanderen (G.0096.02, E.L.) and (G.0507.04, P.M.); by the Interuniversity Attraction Poles (IAP) granted by the Federal Office for Scientific, Technical and Cultural Affairs, Belgium (2002–2006; P5/25) (P.M. and E.L.); by a Concerted Action Grant from the Catholic University Leuven; by the Federation des Maladies Genetiques Orphelines (G.T.) and by grants-in-aid from the Ministry of Education, Science, Technology, Sports, and Culture of Japan (A.Y.). This work was made possible by the Centre of Excellence SymBioSys (Research Council, Catholic University Leuven EF/05/007) (E.L.).

Author information

Authors and Affiliations

Authors

Contributions

The study was coordinated by H.B., G.T., P.M., A.Y. and E.L.; patient phenotyping was performed by J.-P.F. and E.L. and clinical data collected by E.D.; linkage analysis was done by M.S. and G.T.; NF1 mutation analysis was conducted by L.M. and SPRED1 mutation analysis by H.B., M.C. and E.D.; skin biopsies were carried out by S.D.S. and melanocytes cultured by H.B. and S.D.S.; biochemical analysis was performed by H.B., M.C., E.D., K.T., R.S. and J.C.; mouse characterization was performed by R.K.; the manuscript was written by H.B., M.C., E.D., L.M., S.D.S., A.Y., J.-P.F., J.C., P.M., G.T. and E.L.

Corresponding author

Correspondence to Eric Legius.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Table 1, Supplementary Figures 1–7 (PDF 635 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Brems, H., Chmara, M., Sahbatou, M. et al. Germline loss-of-function mutations in SPRED1 cause a neurofibromatosis 1–like phenotype. Nat Genet 39, 1120–1126 (2007). https://doi.org/10.1038/ng2113

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng2113

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