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.

  • Article
  • Published:

PNPLA1 mutations cause autosomal recessive congenital ichthyosis in golden retriever dogs and humans

Abstract

Ichthyoses comprise a heterogeneous group of genodermatoses characterized by abnormal desquamation over the whole body, for which the genetic causes of several human forms remain unknown. We used a spontaneous dog model in the golden retriever breed, which is affected by a lamellar ichthyosis resembling human autosomal recessive congenital ichthyoses (ARCI), to carry out a genome-wide association study. We identified a homozygous insertion-deletion (indel) mutation in PNPLA1 that leads to a premature stop codon in all affected golden retriever dogs. We subsequently found one missense and one nonsense mutation in the catalytic domain of human PNPLA1 in six individuals with ARCI from two families. Further experiments highlighted the importance of PNPLA1 in the formation of the epidermal lipid barrier. This study identifies a new gene involved in human ichthyoses and provides insights into the localization and function of this yet uncharacterized member of the PNPLA protein family.

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: Identification of the PNPLA1 mutation in affected golden retriever dogs.
Figure 2: Identification of PNPLA1 mutations in humans with autosomal recessive congenital ichthyosis.
Figure 3: Histological analysis of skin biopsies from golden retriever dogs and human subjects.
Figure 4: Localization of wild-type PNPLA1 protein in human skin.
Figure 5: Transmission electron micrographs of fixed fresh skin biopsies from golden retriever dogs and humans.
Figure 6: Protein blotting of PNPLA1 in normal and mutant human keratinocytes, before differentiation and at 3 and 7 d after induction of differentiation.
Figure 7: Triglyceride hydrolase activity and lipid profiles of wild-type and PNPLA1-deficient human keratinocytes in cell culture.

Similar content being viewed by others

Accession codes

Accessions

Swiss-Prot

References

  1. Oji, V. et al. Revised nomenclature and classification of inherited ichthyoses: results of the First Ichthyosis Consensus Conference in Soreze 2009. J. Am. Acad. Dermatol. 63, 607–641 (2010).

    Article  Google Scholar 

  2. Fischer, J. Autosomal recessive congenital ichthyosis. J. Invest. Dermatol. 129, 1319–1321 (2009).

    Article  CAS  Google Scholar 

  3. Israeli, S. et al. A mutation in LIPN, encoding epidermal lipase N, causes a late-onset form of autosomal-recessive congenital ichthyosis. Am. J. Hum. Genet. 88, 482–487 (2011).

    Article  CAS  PubMed Central  Google Scholar 

  4. Lefèvre, C. et al. Mutations in CGI-58, the gene encoding a new protein of the esterase/lipase/thioesterase subfamily, in Chanarin-Dorfman syndrome. Am. J. Hum. Genet. 69, 1002–1012 (2001).

    Article  PubMed Central  Google Scholar 

  5. Demerjian, M., Crumrine, D.A., Milstone, L.M., Williams, M.L. & Elias, P.M. Barrier dysfunction and pathogenesis of neutral lipid storage disease with ichthyosis (Chanarin-Dorfman syndrome). J. Invest. Dermatol. 126, 2032–2038 (2006).

    Article  CAS  Google Scholar 

  6. Yamaguchi, T. & Osumi, T. Chanarin-Dorfman syndrome: deficiency in CGI-58, a lipid droplet-bound coactivator of lipase. Biochim. Biophys. Acta 1791, 519–523 (2009).

    Article  CAS  Google Scholar 

  7. Zimmermann, R., Lass, A., Haemmerle, G. & Zechner, R. Fate of fat: the role of adipose triglyceride lipase in lipolysis. Biochim. Biophys. Acta 1791, 494–500 (2009).

    Article  CAS  Google Scholar 

  8. Schweiger, M. et al. The C-terminal region of human adipose triglyceride lipase affects enzyme activity and lipid droplet binding. J. Biol. Chem. 283, 17211–17220 (2008).

    Article  CAS  Google Scholar 

  9. Fischer, J. et al. The gene encoding adipose triglyceride lipase (PNPLA2) is mutated in neutral lipid storage disease with myopathy. Nat. Genet. 39, 28–30 (2007).

    Article  CAS  Google Scholar 

  10. Ohkuma, A. et al. Distal lipid storage myopathy due to PNPLA2 mutation. Neuromuscul. Disord. 18, 671–674 (2008).

    Article  Google Scholar 

  11. Akiyama, M. et al. Novel duplication mutation in the patatin domain of adipose triglyceride lipase (PNPLA2) in neutral lipid storage disease with severe myopathy. Muscle Nerve 36, 856–859 (2007).

    Article  CAS  PubMed Central  Google Scholar 

  12. Lake, A.C. et al. Expression, regulation, and triglyceride hydrolase activity of Adiponutrin family members. J. Lipid Res. 46, 2477–2487 (2005).

    Article  CAS  Google Scholar 

  13. Wilson, P.A., Gardner, S.D., Lambie, N.M., Commans, S.A. & Crowther, D.J. Characterization of the human patatin-like phospholipase family. J. Lipid Res. 47, 1940–1949 (2006).

    Article  CAS  Google Scholar 

  14. Kienesberger, P.C., Oberer, M., Lass, A. & Zechner, R. Mammalian patatin domain containing proteins: a family with diverse lipolytic activities involved in multiple biological functions. J. Lipid Res. 50 (suppl.), S63–S68 (2009).

    Article  PubMed Central  Google Scholar 

  15. Baulande, S. & Langlois, C. Proteins sharing PNPLA domain, a new family of enzymes regulating lipid metabolism. Med. Sci. (Paris) 26, 177–184 (2010).

    Article  Google Scholar 

  16. Romeo, S. et al. Genetic variation in PNPLA3 confers susceptibility to nonalcoholic fatty liver disease. Nat. Genet. 40, 1461–1465 (2008).

    Article  CAS  PubMed Central  Google Scholar 

  17. Tian, C., Stokowski, R.P., Kershenobich, D., Ballinger, D.G. & Hinds, D.A. Variant in PNPLA3 is associated with alcoholic liver disease. Nat. Genet. 42, 21–23 (2010).

    Article  CAS  Google Scholar 

  18. Rainier, S. et al. Neuropathy target esterase gene mutations cause motor neuron disease. Am. J. Hum. Genet. 82, 780–785 (2008).

    Article  CAS  PubMed Central  Google Scholar 

  19. Mubaidin, A. et al. Karak syndrome: a novel degenerative disorder of the basal ganglia and cerebellum. J. Med. Genet. 40, 543–546 (2003).

    Article  CAS  PubMed Central  Google Scholar 

  20. Tan, E.K., Ho, P., Tan, L., Prakash, K.M. & Zhao, Y. PLA2G6 mutations and Parkinson's disease. Ann. Neurol. 67, 148 (2010).

    Article  CAS  Google Scholar 

  21. Gregory, A. et al. Neurodegeneration associated with genetic defects in phospholipase A(2). Neurology 71, 1402–1409 (2008).

    Article  CAS  PubMed Central  Google Scholar 

  22. Sutter, N.B. & Ostrander, E.A. Dog star rising: the canine genetic system. Nat. Rev. Genet. 5, 900–910 (2004).

    Article  CAS  Google Scholar 

  23. Karlsson, E.K. et al. Efficient mapping of mendelian traits in dogs through genome-wide association. Nat. Genet. 39, 1321–1328 (2007).

    Article  CAS  Google Scholar 

  24. Galibert, F. & Andre, C. The dog: a powerful model for studying genotype-phenotype relationships. Comp. Biochem. Physiol. Part D Genomics Proteomics 3, 67–77 (2008).

    Article  Google Scholar 

  25. Cadieu, E. et al. Coat variation in the domestic dog is governed by variants in three genes. Science 326, 150–153 (2009).

    Article  CAS  PubMed Central  Google Scholar 

  26. Parker, H.G. et al. An expressed fgf4 retrogene is associated with breed-defining chondrodysplasia in domestic dogs. Science 325, 995–998 (2009).

    Article  CAS  PubMed Central  Google Scholar 

  27. Merveille, A.C. et al. CCDC39 is required for assembly of inner dynein arms and the dynein regulatory complex and for normal ciliary motility in humans and dogs. Nat. Genet. 43, 72–78 (2011).

    Article  CAS  Google Scholar 

  28. Credille, K.M., Barnhart, K.F., Minor, J.S. & Dunstan, R.W. Mild recessive epidermolytic hyperkeratosis associated with a novel keratin 10 donor splice-site mutation in a family of Norfolk terrier dogs. Br. J. Dermatol. 153, 51–58 (2005).

    Article  CAS  Google Scholar 

  29. Credille, K.M. et al. Transglutaminase 1-deficient recessive lamellar ichthyosis associated with a LINE-1 insertion in Jack Russell terrier dogs. Br. J. Dermatol. 161, 265–272 (2009).

    Article  CAS  Google Scholar 

  30. Akiyama, M. & Shimizu, H. An update on molecular aspects of the non-syndromic ichthyoses. Exp. Dermatol. 17, 373–382 (2008).

    Article  CAS  Google Scholar 

  31. Huber, M. et al. Mutations of keratinocyte transglutaminase in lamellar ichthyosis. Science 267, 525–528 (1995).

    Article  CAS  Google Scholar 

  32. Parmentier, L. et al. Autosomal recessive lamellar ichthyosis: identification of a new mutation in transglutaminase 1 and evidence for genetic heterogeneity. Hum. Mol. Genet. 4, 1391–1395 (1995).

    Article  CAS  Google Scholar 

  33. Russell, L.J. et al. Mutations in the gene for transglutaminase 1 in autosomal recessive lamellar ichthyosis. Nat. Genet. 9, 279–283 (1995).

    Article  CAS  Google Scholar 

  34. Mauldin, E.A., Credille, K.M., Dunstan, R.W. & Casal, M.L. Clinical, histopathological and ultrastructural analysis of golden retriever ichthyosis. Vet. Dermatol. 18, 187 (2007).

    Article  Google Scholar 

  35. Guaguere, E., Bensignor, E., Muller, A., Degorce-Rubiales, F. & Andre, C. Epidemiological, clinical, histopathological and ultrastructural aspects of ichthyosis in golden retrievers: a report of 50 cases. Vet. Dermatol. 18, 382–383 (2007).

    Google Scholar 

  36. Cadiergues, M.C. et al. Cornification defect in the golden retriever: clinical, histopathological, ultrastructural and genetic characterisation. Vet. Dermatol. 19, 120–129 (2008).

    Article  Google Scholar 

  37. Mauldin, E.A., Credille, K.M., Dunstan, R.W. & Casal, M.L. The clinical and morphologic features of nonepidermolytic ichthyosis in the golden retriever. Vet. Pathol. 45, 174–180 (2008).

    Article  CAS  PubMed Central  Google Scholar 

  38. Guaguere, E. et al. Clinical, histopathological and genetic data of ichthyosis in the golden retriever: a prospective study. J. Small Anim. Pract. 50, 227–235 (2009).

    Article  CAS  Google Scholar 

  39. Purcell, S. et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am. J. Hum. Genet. 81, 559–575 (2007).

    Article  CAS  PubMed Central  Google Scholar 

  40. Adzhubei, I.A. et al. A method and server for predicting damaging missense mutations. Nat. Methods 7, 248–249 (2010).

    Article  CAS  PubMed Central  Google Scholar 

  41. Akiyama, M. et al. CGI-58 is an alpha/beta-hydrolase within lipid transporting lamellar granules of differentiated keratinocytes. Am. J. Pathol. 173, 1349–1360 (2008).

    Article  CAS  PubMed Central  Google Scholar 

  42. Eckert, R.L. et al. Regulation of involucrin gene expression. J. Invest. Dermatol. 123, 13–22 (2004).

    Article  CAS  Google Scholar 

  43. Hitomi, K. Transglutaminases in skin epidermis. Eur. J. Dermatol. 15, 313–319 (2005).

    CAS  PubMed  Google Scholar 

  44. Toulza, E. et al. Large-scale identification of human genes implicated in epidermal barrier function. Genome Biol. 8, R107 (2007).

    Article  PubMed Central  Google Scholar 

  45. Elias, P.M., Williams, M.L., Holleran, W.M., Jiang, Y.J. & Schmuth, M. Pathogenesis of permeability barrier abnormalities in the ichthyoses: inherited disorders of lipid metabolism. J. Lipid Res. 49, 697–714 (2008).

    Article  CAS  PubMed Central  Google Scholar 

  46. Laiho, E. et al. Clinical and morphological correlations for transglutaminase 1 gene mutations in autosomal recessive congenital ichthyosis. Eur. J. Hum. Genet. 7, 625–632 (1999).

    Article  CAS  Google Scholar 

  47. Klar, J. et al. Mutations in the fatty acid transport protein 4 gene cause the ichthyosis prematurity syndrome. Am. J. Hum. Genet. 85, 248–253 (2009).

    Article  CAS  PubMed Central  Google Scholar 

  48. Ziblat, R., Leiserowitz, L. & Addadi, L. Crystalline domain structure and cholesterol crystal nucleation in single hydrated DPPC:cholesterol:POPC bilayers. J. Am. Chem. Soc. 132, 9920–9927 (2010).

    Article  CAS  Google Scholar 

  49. Johansson, L.E. et al. Genetic variance in the adiponutrin gene family and childhood obesity. PLoS ONE 4, e5327 (2009).

    Article  PubMed Central  Google Scholar 

  50. Gao, J.G., Shih, A., Gruber, R., Schmuth, M. & Simon, M. GS2 as a retinol transacylase and as a catalytic dyad independent regulator of retinylester accretion. Mol. Genet. Metab. 96, 253–260 (2009).

    Article  CAS  Google Scholar 

  51. Breiden, B., Gallala, H., Doering, T. & Sandhoff, K. Optimization of submerged keratinocyte cultures for the synthesis of barrier ceramides. Eur. J. Cell Biol. 86, 657–673 (2007).

    Article  CAS  Google Scholar 

  52. Slot, J.W. & Geuze, H.J. Cryosectioning and immunolabeling. Nat. Protoc. 2, 2480–2491 (2007).

    Article  CAS  Google Scholar 

  53. Lass, A. et al. Adipose triglyceride lipase-mediated lipolysis of cellular fat stores is activated by CGI-58 and defective in Chanarin-Dorfman Syndrome. Cell Metab. 3, 309–319 (2006).

    Article  CAS  Google Scholar 

  54. Zimmermann, R. et al. Fat mobilization in adipose tissue is promoted by adipose triglyceride lipase. Science 306, 1383–1386 (2004).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank A.S. Lequarré, an excellent coordinator for the European FP7 LUPA project, A. Boland and D. Zelenika for the genotyping performed at CNG (Evry, France); dermatologists, especially F. Caux, and Généthon, for patient DNA processing; J. Abadie (AMaROC research unit, Oniris, Ecole Nationale Vétérinaire de Nantes, France) and practitioners T. Bord, X. Langon, P. Prelaud, M.D. Vaillant, A. Muller and other veterinarians for providing us with clinical data and samples, as well as dog owners and breeders, especially J. Robidou, B. Facq, V. d'Alcantara and C. de Vinck. We thank P. Roosje and T. Leeb (University of Bern, Switzerland) for providing six Swiss golden retriever samples. We thank A. Fautrel and P. Bellaud, from the histopathology platform H2P2, IFR140 Biogenouest, (Rennes, France), M.D. Vignon-Pennamen from the anatomopathology laboratory of Saint Louis Hospital (Paris, France) and M. Werner from the Institute of Pathology at the University Hospital of Freiburg (Freiburg, Germany) who kindly provided paraffin human skin sections, as well as the Vébiotel laboratory (Arcueil, France) for dog sample biochemical analyses. We are grateful to G. Queney (Antagene, Lyon, France) and P. Quignon, G. Rabut and E. Watrin (Institut de Génétique et Développement de Rennes, France) for helpful discussions. Finally, we warmly thank S. Cure from Genoscope (Evry, France) for her several careful readings and English corrections and her kind availability, as well as D. Morris-Rosendahl (Institute for Human Genetics, Freiburg, Germany). This study was supported by CNRS, the European Commission (FP7-LUPA, GA-201370). R. Zechner and R. Zimmermann were supported by the FWF F30 SFB Lipotox, Z136 Wittgenstein, the GEN-AU project GOLD by the Austrian Ministry of Science and Research and FFG. I.H. was supported by the NIRK Network (German BMBF 01GM0904).

Author information

Authors and Affiliations

Authors

Contributions

C.A., E.G. and F.G. designed the genetic aspects of the dog experiments. A.G., S.P., C.H., M.L.G., L.L. and S.K. performed the genetic and functional experiments for the dog studies. J. Fischer designed the human genetic analyses and supervised the functional studies on humans. E. Bourrat provided patient material and data. C.D. and G.-J.K. performed the genetic and microscopy experiments for the human studies. I.H. performed light and electron microscopy as well as immunoelectron microscopy investigations. F.D.-R. did H&E staining for histological diagnosis and investigations in dogs. S.G., F.P.W.R., R. Zimmermann and R. Zechner performed functional studies E.G., E. Bensignor, J. Fontaine and D.P., veterinarians specializing in dermatology, collected dog samples and interpreted clinical and biological data. A.T. provided 400 dog DNA samples and performed validation of the mutation in dogs. C.A., A.G., J. Fischer, F.G., C.H., M.L. and I.H.. contributed to the writing of the manuscript.

Corresponding authors

Correspondence to Catherine André or Judith Fischer.

Ethics declarations

Competing interests

CNRS and Université Rennes 1 (including C.A., E.G. and S.P.) have applied for an international patent (Catherine André et al., PCT/EP2010/067569) covering the use of the canine PNPLA1 mutation for the genetic screening of ichthyosis in dogs. The Antagene laboratory has the international license for providing the ichthyosis DNA test in dogs.

Supplementary information

Supplementary Text and Figures

Supplementary Note, Supplementary Tables 1–5 and Supplementary Figures 1–4 (PDF 6871 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Grall, A., Guaguère, E., Planchais, S. et al. PNPLA1 mutations cause autosomal recessive congenital ichthyosis in golden retriever dogs and humans. Nat Genet 44, 140–147 (2012). https://doi.org/10.1038/ng.1056

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.1056

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