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:

Prophylactic treatment with sialic acid metabolites precludes the development of the myopathic phenotype in the DMRV-hIBM mouse model

Abstract

Distal myopathy with rimmed vacuoles (DMRV)–hereditary inclusion body myopathy (hIBM) is an adult-onset, moderately progressive autosomal recessive myopathy; eventually, affected individuals become wheelchair bound1. It is characterized clinically by skeletal muscle atrophy and weakness, and pathologically by rimmed vacuoles, which are actually accumulations of autophagic vacuoles2,3,4, scattered angular fibers and intracellular accumulation of amyloid and other proteins5. To date, no therapy is available for this debilitating myopathy, primarily because the disease pathomechanism has been enigmatic. It is known that the disease gene underlying DMRV-hIBM is GNE, encoding glucosamine (UDP-N-acetyl)-2-epimerase and N-acetylmannosamine kinase6,7,8—two essential enzymes in sialic acid biosynthesis9. It is still unclear, however, whether decreased sialic acid production causes muscle degeneration, as GNE has been proposed to have roles other than for sialic acid biosynthesis10,11,12. By showing that muscle atrophy and weakness are completely prevented in a mouse model of DMRV-hIBM after treatment with sialic acid metabolites orally, we provide evidence that hyposialylation is indeed one of the key factors in the pathomechanism of DMRV-hIBM. These results support the notion that DMRV-hIBM can potentially be treated simply by giving sialic acids, a strategy that could be applied in clinical trials in the near future.

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: Favorable effects in the myopathic phenotype of DRMV-hIBM mice are seen after administration of ManNAc but are not correlated with dose.
Figure 2: Low-dose administration of ManNAc, NeuAc or sialyllactose increases the sialic acid concentration in serum and muscle and leads to an improvement in survival and favorable effects on overall motor performance.
Figure 3: Oral administration of sialic acid metabolites notably prevents atrophy in skeletal muscles and increases generation of force.
Figure 4: Muscle degeneration in DMRV-hIBM mice is virtually abrogated after oral treatment with sialic acid metabolites.

Similar content being viewed by others

References

  1. Nonaka, I., Noguchi, S. & Nishino, I. Distal myopathy with rimmed vacuoles and hereditary inclusion body myopathy. Curr. Neurol. Neurosci. Rep. 5, 61–65 (2005).

    Article  CAS  Google Scholar 

  2. Nishino, I. et al. Molecular pathomechanism of distal myopathy with rimmed vacuoles. Acta Myol. 24, 80–83 (2005).

    CAS  PubMed  Google Scholar 

  3. Tsuruta, Y. et al. Expression of the lysosome-associated membrane proteins in myopathies with rimmed vacuoles. Acta Neuropathol. 101, 579–584 (2001).

    CAS  PubMed  Google Scholar 

  4. Malicdan, M.C., Noguchi, S. & Nishino, I. Autophagy in a mouse model of distal myopathy with rimmed vacuoles or hereditary inclusion body myopathy. Autophagy 3, 396–398 (2007).

    Article  CAS  Google Scholar 

  5. Askanas, V. & Engel, W.K. Hereditary inclusion myopathies. in. The molecular and genetic basis of neurologic and psychiatric disease (eds. Rosenberg, R.N., Prusiner, S.B., DiMauro, S., Barchi, R.L. & Nestler, E.J.) 501–509 (Butterworth–Heinemann, Woburn, Massachusetts, 2003).

    Google Scholar 

  6. Eisenberg, I. et al. The UDP–N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase gene is mutated in recessive hereditary inclusion body myopathy. Nat. Genet. 29, 83–87 (2001).

    Article  CAS  Google Scholar 

  7. Ikeuchi, T. et al. Gene locus for autosomal recessive distal myopathy with rimmed vacuoles maps to chromosome 9. Ann. Neurol. 41, 432–437 (1997).

    Article  CAS  Google Scholar 

  8. Nishino, I. et al. Distal myopathy with rimmed vacuoles is allelic to hereditary inclusion body myopathy. Neurology 59, 1689–1693 (2002).

    Article  CAS  Google Scholar 

  9. Keppler, O.T. et al. UDP–GlcNAc 2-epimerase: a regulator of cell surface sialylation. Science 284, 1372–1376 (1999).

    Article  CAS  Google Scholar 

  10. Krause, S. et al. Localization of UDP–GlcNAc 2-epimerase/ManAc kinase (GNE) in the Golgi complex and the nucleus of mammalian cells. Exp. Cell Res. 304, 365–379 (2005).

    Article  CAS  Google Scholar 

  11. Wang, Z., Sun, Z., Li, A.V. & Yarema, K.J. Roles for GNE outside of sialic acid biosynthesis: modulation of sialyltransferase and BiP expression, GM3 and GD3 biosynthesis, proliferation and apoptosis, and ERK1/2 phosphorylation. J. Biol. Chem. 281, 27016–27028 (2006).

    Article  CAS  Google Scholar 

  12. Amsili, S. et al. UDP–N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase (GNE) binds to α-actinin 1: novel pathways in skeletal muscle? PLoS One 3, e2477 (2008).

    Article  Google Scholar 

  13. Noguchi, S. et al. Reduction of UDP–N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase activity and sialylation in distal myopathy with rimmed vacuoles. J. Biol. Chem. 279, 11402–11407 (2004).

    Article  CAS  Google Scholar 

  14. Broccolini, A. et al. Hyposialylation of neprilysin possibly affects its expression and enzymatic activity in hereditary inclusion–body myopathy muscle. J. Neurochem. 105, 971–981 (2008).

    Article  CAS  Google Scholar 

  15. Malicdan, M.C., Noguchi, S., Nonaka, I., Hayashi, Y.K. & Nishino, I. A Gne knockout mouse expressing human GNE D176V mutation develops features similar to distal myopathy with rimmed vacuoles or hereditary inclusion body myopathy. Hum. Mol. Genet. 16, 2669–2682 (2007).

    Article  CAS  Google Scholar 

  16. Malicdan, M.C., Noguchi, S., Hayashi, Y.K. & Nishino, I. Muscle weakness correlates with muscle atrophy and precedes the development of inclusion body or rimmed vacuoles in the mouse model of DMRV/hIBM. Physiol. Genomics 35, 106–115 (2008).

    Article  CAS  Google Scholar 

  17. Comb, D.G. & Roseman, S. Composition and enzymatic synthesis of N-acetylneuraminic (sialic) acid. J. Am. Chem. Soc. 80, 497–499 (1958).

    Article  CAS  Google Scholar 

  18. Thomas, G.H., Scocca, J., Miller, C. & Reynolds, L.W. Accumulation of N-acetylneuraminic acid (sialic acid) in human fibroblasts cultured in the presence of N-mannosamine. Biochim. Biophys. Acta 846, 37–43 (1985).

    Article  CAS  Google Scholar 

  19. Galeano, B. et al. Mutation in the key enzyme of sialic acid biosynthesis causes severe glomerular proteinuria and is rescued by N-acetylmannosamine. J. Clin. Invest. 117, 1585–1594 (2007).

    Article  CAS  Google Scholar 

  20. Robitaille, G., Ng-Kwai-Hang, K.F. & Monardes, H.G. Association of κ-casein glycosylation with milk production and composition in holsteins. J. Dairy Sci. 74, 3314–3317 (1991).

    Article  CAS  Google Scholar 

  21. Clarke, B.A. et al. The E3 ligase MuRF1 degrades myosin heavy chain protein in dexamethasone-treated skeletal muscle. Cell Metab. 6, 376–385 (2007).

    Article  CAS  Google Scholar 

  22. Kabeya, Y. et al. LC3, GABARAP and GATE16 localize to autophagosomal membrane depending on form-II formation. J. Cell Sci. 117, 2805–2812 (2004).

    Article  CAS  Google Scholar 

  23. Mizushima, N. & Yoshimori, T. How to interpret LC3 immunoblotting. Autophagy 3, 542–545 (2007).

    Article  CAS  Google Scholar 

  24. Kumamoto, T. et al. Proteasomes in distal myopathy with rimmed vacuoles. Intern. Med. 37, 746–752 (1998).

    Article  CAS  Google Scholar 

  25. Corfield, A.P. & Schauer, R. Occurrence of sialic acids. in Sialic Acids. Chemistry, Metabolism, and Function (ed. Schauer, R.) 5–50 (Springer, Wien, New York, 1982).

    Chapter  Google Scholar 

  26. Helenius, A. How N-linked oligosaccharides affect glycoprotein folding in the endoplasmic reticulum. Mol. Biol. Cell 5, 253–265 (1994).

    Article  CAS  Google Scholar 

  27. Iijima, R., Takahashi, H., Namme, R., Ikegami, S. & Yamazaki, M. Novel biological function of sialic acid (N-acetylneuraminic acid) as a hydrogen peroxide scavenger. FEBS Lett. 561, 163–166 (2004).

    Article  CAS  Google Scholar 

  28. Askanas, V. & Engel, W.K. Inclusion-body myositis: a myodegenerative conformational disorder associated with Aβ, protein misfolding, and proteasome inhibition. Neurology 66 (suppl 1), S39–S48 (2006).

    Article  CAS  Google Scholar 

  29. Spuler, S. et al. Dysferlin-deficient muscular dystrophy features amyloidosis. Ann. Neurol. 63, 323–328 (2008).

    Article  CAS  Google Scholar 

  30. Lynch, G.S., Hinkle, R.T., Chamberlain, J.S., Brooks, S.V. & Faulkner, J. Force and power output of fast and slow skeletal muscles from mdx mice 6–28 months old. J. Physiol. (Lond.) 535, 591–600 (2001).

    Article  CAS  Google Scholar 

  31. Malicdan, M.C., Noguchi, S. & Nishino, I. Monitoring autophagy in muscle disaseases. Methods Enzymol. 453, 379–396 (2009).

    Article  CAS  Google Scholar 

  32. Gagiannis, D., Gossrau, R., Reutter, W., Zimmermann–Kordmann, M. & Horstkorte, R. Engineering the sialic acid in organs of mice using N-propanoylmannosamine. Biochim. Biophys. Acta 1770, 297–306 (2007).

    Article  CAS  Google Scholar 

  33. Hara, S. et al. Determination of mono-O-acetylated N-acetylneuraminic acids in human and rat sera by fluorometric high-performance liquid chromatography. Anal. Biochem. 179, 162–166 (1989).

    Article  CAS  Google Scholar 

  34. Schmidt, S., Jiang, Y., Nixon, R. & Mathews, P. Amyloid proteins methods and protocol. in. Methods in Molecular Biology Vol. 299 (ed. Sigurdsson, E.M.) 408 (Humana Press, New York, 2004).

    Google Scholar 

  35. Kotani, K., Maekawa, M. & Kanno, T. Reestimation of aspartate aminotransferase (AST)/alanine aminotransferase (ALT) ratio based on JSCC consensus method—changes of criteria for a differential diagnosis of hepatic disorders following the alteration from Karmen method to JSCC method [in Japanese]. Nippon Shokakibyo Gakkai Zasshi 91, 154–161 (1994).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors acknowledge the valuable assistance offered by F. Funato and R. Hoshi in motor performance evaluation of mice, toxicology assays and sialic acid measurement. This study is supported partly by the Research on Psychiatric and Neurological Diseases and Mental Health grant from the Japanese Health Sciences Foundation; the Program for Promotion of Fundamental Studies in Health Sciences of the Japanese National Institute of Biomedical Innovation the Research Grant (19A–7) for Nervous and Mental Disorders from the Ministry of Health Labour and Welfare in Japan, the Kato Memorial Trust for Nambyo Research and the Neuromuscular Disease Foundation.

Author information

Authors and Affiliations

Authors

Contributions

M.C.V.M. conducted most of the experiments and wrote the manuscript; S.N. supervised all aspects of this study, including study design, execution and interpretation and manuscript preparation, and participated in the analysis of the in vitro muscle data and sialic acid measurement; I. Nonaka, Y.K.H. and I. Nishino were involved in analyzing and interpreting all the data and also supervised the study design, execution and interpretation and manuscript preparation.

Corresponding author

Correspondence to Satoru Noguchi.

Ethics declarations

Competing interests

The National Center of Neurology and Psychiatry (Japan), where the authors are employed, along with the Japan Health Science Foundation have applied to the Japan Patent Office for a patent on the use of sialic acid metabolites and their derivatives in DMRV-hIBM treatment.

Supplementary information

Supplementary Text and Figures

Supplementary Figs. 1–9, Supplementary Table 1 and Supplementary Methods (PDF 1125 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Malicdan, M., Noguchi, S., Hayashi, Y. et al. Prophylactic treatment with sialic acid metabolites precludes the development of the myopathic phenotype in the DMRV-hIBM mouse model. Nat Med 15, 690–695 (2009). https://doi.org/10.1038/nm.1956

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.1956

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