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:

Misregulated alternative splicing of BIN1 is associated with T tubule alterations and muscle weakness in myotonic dystrophy

Abstract

Myotonic dystrophy is the most common muscular dystrophy in adults and the first recognized example of an RNA-mediated disease. Congenital myotonic dystrophy (CDM1) and myotonic dystrophy of type 1 (DM1) or of type 2 (DM2) are caused by the expression of mutant RNAs containing expanded CUG or CCUG repeats, respectively. These mutant RNAs sequester the splicing regulator Muscleblind-like-1 (MBNL1), resulting in specific misregulation of the alternative splicing of other pre-mRNAs. We found that alternative splicing of the bridging integrator-1 (BIN1) pre-mRNA is altered in skeletal muscle samples of people with CDM1, DM1 and DM2. BIN1 is involved in tubular invaginations of membranes and is required for the biogenesis of muscle T tubules, which are specialized skeletal muscle membrane structures essential for excitation-contraction coupling. Mutations in the BIN1 gene cause centronuclear myopathy, which shares some histopathological features with myotonic dystrophy. We found that MBNL1 binds the BIN1 pre-mRNA and regulates its alternative splicing. BIN1 missplicing results in expression of an inactive form of BIN1 lacking phosphatidylinositol 5-phosphate–binding and membrane-tubulating activities. Consistent with a defect of BIN1, muscle T tubules are altered in people with myotonic dystrophy, and membrane structures are restored upon expression of the normal splicing form of BIN1 in muscle cells of such individuals. Finally, reproducing BIN1 splicing alteration in mice is sufficient to promote T tubule alterations and muscle weakness, a predominant feature of myotonic dystrophy.

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: BIN1 exon 11 is skipped in humans with myotonic dystrophy.
Figure 2: MBNL1 stimulates inclusion of BIN1 exon 11.
Figure 3: The splicing form of BIN1 expressed in myotonic dystrophic patients is inactive.
Figure 4: Skipping of Bin1 exon 11 is sufficient to induce muscle weakness in mice.

Similar content being viewed by others

Accession codes

Accessions

Gene Expression Omnibus

References

  1. Brook, J.D. et al. Molecular basis of myotonic dystrophy: expansion of a trinucleotide (CTG) repeat at the 3′ end of a transcript encoding a protein kinase family member. Cell 68, 799–808 (1992).

    Article  CAS  Google Scholar 

  2. Fu, Y.H. et al. An unstable triplet repeat in a gene related to myotonic muscular dystrophy. Science 255, 1256–1258 (1992).

    Article  CAS  Google Scholar 

  3. Mahadevan, M. et al. Myotonic dystrophy mutation: an unstable CTG repeat in the 3′ untranslated region of the gene. Science 255, 1253–1255 (1992).

    Article  CAS  Google Scholar 

  4. Liquori, C.L. et al. Myotonic dystrophy type 2 caused by a CCTG expansion in intron 1 of ZNF9. Science 293, 864–867 (2001).

    Article  CAS  Google Scholar 

  5. Philips, A.V., Timchenko, L.T. & Cooper, T.A. Disruption of splicing regulated by a CUG-binding protein in myotonic dystrophy. Science 280, 737–741 (1998).

    Article  CAS  Google Scholar 

  6. Mankodi, A. et al. Myotonic dystrophy in transgenic mice expressing an expanded CUG repeat. Science 289, 1769–1773 (2000).

    Article  CAS  Google Scholar 

  7. Seznec, H. et al. Mice transgenic for the human myotonic dystrophy region with expanded CTG repeats display muscular and brain abnormalities. Hum. Mol. Genet. 10, 2717–2726 (2001).

    Article  CAS  Google Scholar 

  8. Miller, J.W. et al. Recruitment of human muscleblind proteins to (CUG)n expansions associated with myotonic dystrophy. EMBO J. 19, 4439–4448 (2000).

    Article  CAS  Google Scholar 

  9. Kanadia, R.N. et al. A muscleblind knockout model for myotonic dystrophy. Science 302, 1978–1980 (2003).

    Article  CAS  Google Scholar 

  10. Ho, T.H. et al. Muscleblind proteins regulate alternative splicing. EMBO J. 23, 3103–3112 (2004).

    Article  CAS  Google Scholar 

  11. Kuyumcu-Martinez, N.M., Wang, G.S. & Cooper, T.A. Increased steady-state levels of CUGBP1 in myotonic dystrophy 1 are due to PKC-mediated hyperphosphorylation. Mol. Cell 28, 68–78 (2007).

    Article  CAS  Google Scholar 

  12. Savkur, R.S., Philips, A.V. & Cooper, T.A. Aberrant regulation of insulin receptor alternative splicing is associated with insulin resistance in myotonic dystrophy. Nat. Genet. 29, 40–47 (2001).

    Article  CAS  Google Scholar 

  13. Mankodi, A. et al. Expanded CUG repeats trigger aberrant splicing of ClC-1 chloride channel pre-mRNA and hyperexcitability of skeletal muscle in myotonic dystrophy. Mol. Cell 10, 35–44 (2002).

    Article  CAS  Google Scholar 

  14. Charlet-Berguerand, N. et al. Loss of the muscle-specific chloride channel in type 1 myotonic dystrophy due to misregulated alternative splicing. Mol. Cell 10, 45–53 (2002).

    Article  Google Scholar 

  15. Jungbluth, H., Wallgren-Pettersson, C. & Laporte, J. Centronuclear (myotubular) myopathy. Orphanet J. Rare Dis. 3, 26 (2008).

    Article  Google Scholar 

  16. Laporte, J. et al. A gene mutated in X-linked myotubular myopathy defines a new putative tyrosine phosphatase family conserved in yeast. Nat. Genet. 13, 175–182 (1996).

    Article  CAS  Google Scholar 

  17. Bitoun, M. et al. Mutations in dynamin 2 cause dominant centronuclear myopathy. Nat. Genet. 37, 1207–1209 (2005).

    Article  CAS  Google Scholar 

  18. Nicot, A.S. et al. Mutations in amphiphysin 2 (BIN1) disrupt interaction with dynamin 2 and cause autosomal recessive centronuclear myopathy. Nat. Genet. 39, 1134–1139 (2007).

    Article  CAS  Google Scholar 

  19. Lee, E. et al. Amphiphysin 2 (BIN1) and T tubule biogenesis in muscle. Science 297, 1193–1196 (2002).

    Article  CAS  Google Scholar 

  20. Peter, B.J. et al. BAR domains as sensors of membrane curvature: the amphiphysin BAR structure. Science 303, 495–499 (2004).

    Article  CAS  Google Scholar 

  21. Razzaq, A. et al. Amphiphysin is necessary for organization of the excitation-contraction coupling machinery of muscles, but not for synaptic vesicle endocytosis in Drosophila. Genes Dev. 15, 2967–2979 (2001).

    Article  CAS  Google Scholar 

  22. Dansithong, W. et al. MBNL1 is the primary determinant of focus formation and aberrant insulin receptor splicing in DM1. J. Biol. Chem. 280, 5773–5780 (2005).

    Article  CAS  Google Scholar 

  23. Hino, S. et al. Molecular mechanisms responsible for aberrant splicing of SERCA1 in myotonic dystrophy type 1. Hum. Mol. Genet. 16, 2834–2843 (2007).

    Article  CAS  Google Scholar 

  24. Yuan, Y. et al. Muscleblind-like 1 interacts with RNA hairpins in splicing target and pathogenic RNAs. Nucleic Acids Res. 35, 5474–5486 (2007).

    Article  CAS  Google Scholar 

  25. Kino, Y. et al. MBNL and CELF proteins regulate alternative splicing of the skeletal muscle chloride channel CLCN1. Nucleic Acids Res. 37, 6477–6490 (2009).

    Article  CAS  Google Scholar 

  26. Kalsotra, A. et al. A postnatal switch of CELF and MBNL proteins reprograms alternative splicing in the developing heart. Proc. Natl. Acad. Sci. USA 105, 20333–20338 (2008).

    Article  CAS  Google Scholar 

  27. Kanadia, R.N. et al. Reversal of RNA missplicing and myotonia after muscleblind overexpression in a mouse poly(CUG) model for myotonic dystrophy. Proc. Natl. Acad. Sci. USA 103, 11748–11753 (2006).

    Article  CAS  Google Scholar 

  28. Teplova, M. & Patel, D.J. Structural insights into RNA recognition by the alternative-splicing regulator muscleblind-like MBNL1. Nat. Struct. Mol. Biol. 15, 1343–1351 (2008).

    Article  CAS  Google Scholar 

  29. Tronchère, H. et al. Production of phosphatidylinositol 5-phosphate by the phosphoinositide 3-phosphatase myotubularin in mammalian cells. J. Biol. Chem. 279, 7304–7312 (2004).

    Article  Google Scholar 

  30. Ueda, H. et al. Decreased expression of myotonic dystrophy protein kinase and disorganization of sarcoplasmic reticulum in skeletal muscle of myotonic dystrophy. J. Neurol. Sci. 162, 38–50 (1999).

    Article  CAS  Google Scholar 

  31. Goyenvalle, A. et al. Rescue of dystrophic muscle through U7 snRNA-mediated exon skipping. Science 306, 1796–1799 (2004).

    Article  CAS  Google Scholar 

  32. Muller, A.J. et al. Targeted disruption of the murine Bin1/amphiphysin II gene does not disable endocytosis but results in embryonic cardiomyopathy with aberrant myofibril formation. Mol. Cell. Biol. 23, 4295–4306 (2003).

    Article  CAS  Google Scholar 

  33. Al-Qusairi, L. et al. T tubule disorganization and defective excitation-contraction coupling in muscle fibers lacking myotubularin lipid phosphatase. Proc. Natl. Acad. Sci. USA 106, 18763–18768 (2009).

    Article  CAS  Google Scholar 

  34. Kimura, T. et al. Altered mRNA splicing of the skeletal muscle ryanodine receptor and sarcoplasmic/endoplasmic reticulum Ca2+-ATPase in myotonic dystrophy type 1. Hum. Mol. Genet. 14, 2189–2200 (2005).

    Article  CAS  Google Scholar 

  35. Osborne, R.J. et al. Transcriptional and post-transcriptional impact of toxic RNA in myotonic dystrophy. Hum. Mol. Genet. 18, 1471–1481 (2009).

    Article  CAS  Google Scholar 

  36. Yuan, S. et al. Biogenesis of transverse tubules: immunocytochemical localization of a transverse tubular protein (TS28) and a sarcolemmal protein (SL50) in rabbit skeletal muscle developing in situ. J. Cell Biol. 110, 1187–1198 (1990).

    Article  CAS  Google Scholar 

  37. Dowling, J.J. et al. Loss of myotubularin function results in T tubule disorganization in zebrafish and human myotubular myopathy. PLoS Genet. 5, e1000372 (2009).

    Article  Google Scholar 

  38. Toussaint, A. et al. Defects in amphiphysin 2 (BIN1) and triads in several forms of centronuclear myopathies. Acta Neuropathol. 121, 253–266 (2010).

    Article  Google Scholar 

  39. Wilmshurst, J.M. et al. RYR1 mutations are a common cause of congenital myopathies with central nuclei. Ann. Neurol. 68, 717–726 (2010).

    Article  CAS  Google Scholar 

  40. Mouisel, E. et al. Outcome of acetylcholinesterase deficiency for neuromuscular functioning. Neurosci. Res. 55, 389–396 (2006).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank T. Cooper (Baylor College of Medicine) for the gift of the β-globin 4.11.12, DMPKS, DT960 and tgCUGBP1 plasmids, L. Ranum (University of Minnesota) for the gift of the CCTG300 expression plasmid, C. Branlant (CNRS) for the gift of the pGEX-MBNL1Δ101 vector, M. Swanson (University of Florida) for the gift of the pGEX-6P-MBNL1-His vector, P. Hawkins (Babraham Research Campus) for the gift of the GFP-PH domain of PLCg and GFP-PX domain of P40 constructs, O. Gozani (Stanford University) for the gift of the GFP-PHD domain of ING2 vector, Z. Xue (Université Paris 7) for the gift of the synemin-specific antibody, I. Marty (INSERM) for the gift of the RYR1-specific antibody, M. Swanson (University of Florida), G. Gourdon (INSERM) and C. Thornton (University of Rochester) for the gift of RNA from skeletal muscles of MBNL1ΔE3, DM300 and HSALR mice, respectively, the IGBMC facilities for assistance, J. Lainé, G. Butler-Browne and all members of the French DM Network for fruitful discussion. This work was supported by INSERM AVENIR (N.C.-B.), Agence nationale de la recherche GENOPAT 07-942 (N.C.-B.), 08-005 (J.L.) and BLANC 07-065 (J.L.), Association Française contre les Myopathies MNM1 12982 (N.C.-B.), 12570 and 14269 (D.F.), 12576 and 14058 (J.L.), Fondation pour la Recherche Médicale 20071210538 (J.L.), Japan Society for the Promotion of Science KAKENHI 20590998 (M.P.T.) and a European Molecular Biology Organization long-term fellowship (Y.I.).

Author information

Authors and Affiliations

Authors

Contributions

Experiments were carried out by C.F., A.F.K., C.H., S.V., Y.I., A.T., V.T., A.V., A.F., N.M., Y.K., R.T., V.F., G.P., C.B.-L. F.D. and M.-C.H. Bioinformatic analyses were done by P.d.l.G., D.D., C.T. and D.A. Clinical samples and patient data were from A.L.d.M., N.S., A.L., B.U., B.S., M.P.T., I.N., G.B. and D.F. Data were collected and analyzed by N.M., C.T., D.A., L.G., P.Z., I.N., M.P.T., B.U., B.S., G.B., J.L., D.F. and N.C.-B. The study was designed, coordinated and written by D.F. and N.C.-B.

Corresponding author

Correspondence to Nicolas Charlet-Berguerand.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–5 and Supplementary Methods (PDF 1360 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Fugier, C., Klein, A., Hammer, C. et al. Misregulated alternative splicing of BIN1 is associated with T tubule alterations and muscle weakness in myotonic dystrophy. Nat Med 17, 720–725 (2011). https://doi.org/10.1038/nm.2374

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.2374

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