Skip to main content

Advertisement

Log in

Metabolic Myopathies

  • Published:
Current Rheumatology Reports Aims and scope Submit manuscript

Abstract

We consider recent developments in disorders affecting three areas of metabolism: glycogen, fatty acids, and the mitochondrial respiratory chain. Among the glycogenoses, new attention has been directed to defects of glycogen synthesis resulting in absence rather than excess of muscle glycogen (“aglycogenosis”). These include defects of glycogen synthetase and defects of glycogenin, the primer of glycogen synthesis. Considerable progress also has been made in our understanding of alterations of glycogen metabolism that result in polyglucosan storage. Among the disorders of lipid metabolism, mutations in the genes encoding two triglyceride lipases acting hand in hand cause severe generalized lipid storage myopathy, one associated with ichthyosis (Chanarin-Dorfman syndrome), the other dominated by juvenile-onset weakness. For the mitochondrial myopathies, we discuss the importance of homoplasmic mitochondrial DNA mutations and review the rapid progress made in our understanding of the coenzyme Q10 deficiencies, which are often treatable.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

References

Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance

  1. Kollberg G, Tulinius M, Gilljam T, et al.: Cardiomyopathy and exercise intolerance in muscle glycogen storage disease 0. N Engl J Med 2007, 357:1507–1514.

    Article  CAS  PubMed  Google Scholar 

  2. McCue ME, Valberg SJ, Miller MB, et al.: Glycogen synthase (GYS1) mutation causes a novel skeletal muscle glycogenosis. Genomics 2008, 91:458–466.

    Article  CAS  PubMed  Google Scholar 

  3. Pitcher J, Smythe C, Cohen P: Glycogenin is the priming glucosyltransferase required for the initiation of glycogen biogenesis in rabbit skeletal muscle. Eur J Biochem 1988, 176:391–395.

    Article  CAS  PubMed  Google Scholar 

  4. •• Moslemi A-R, Lindberg C, Nilsson J, et al.: Glycogenin-1 deficiency and inactivated priming of glycogen synthesis. N Engl J Med 2010, 362:1203–1210. These are the long-awaited first mutations in the muscle glycogenin gene and the second example of “glycogenosis 0” in human muscle.

    Article  CAS  PubMed  Google Scholar 

  5. •• Stojkovic T, Vissing J, Petit F, et al.: Muscle glycogenosis due to phosphoglucomutase 1 deficiency. N Engl J Med 2009, 361:425–427. This is a new glycolytic defect showing the “typical” syndrome of exercise intolerance, cramps, and recurrent myoglobinuria.

    Article  CAS  PubMed  Google Scholar 

  6. Sugie H, Kobayashi J, Sugie Y, et al.: Infantile muscle glycogen storage disease: phosphoglucomutase deficiency with decreased muscle and serum carnitine. Neurology 1988, 38:602–605.

    CAS  PubMed  Google Scholar 

  7. Nadaj-Pakleza AA, Vincitorio CM, Laforet P, et al.: Permanent muscle weakness in McArdle disease. Muscle Nerve 2009, 40:350–357.

    Article  PubMed  Google Scholar 

  8. • Vissing J, Duno M, Schwartz M, Haller RG: Splice mutations preserve myophosphorylase activity that ameliorates the phenotype in McArdle disease. Brain 2009, 132:1545–1552. This is an interesting genotype–phenotype association documenting that even a very small amount of residual enzyme activity ameliorates the clinical presentation.

    Article  PubMed  Google Scholar 

  9. Orngreen MC, Jeppesen TD, Tvede Andersen S, et al.: Fat metabolism during exercise in patients with McArdle diseae. Neurology 2009, 72:718–724.

    Article  CAS  PubMed  Google Scholar 

  10. Quinlivan R, Beynon RJ: Pharmacological and nutritional treatment for McArdle’s disease (glycogen storage disease type V). Cochrane Database Syst Rev 2004, 3:CD003458.

  11. • van der Ploeg AT, Clemens PR, Corzo D, et al.: A randomized study of alglucosidase alfa in late-onset Pompe’s disease. N Engl J Med 2010, 362:1396–1406. This is the first (but probably not the last) multicentric study of enzyme replacement therapy in glycogenosis type II.

    Article  PubMed  Google Scholar 

  12. Kishnani PS, Goldenberg PC, DeArmey SL, et al.: Cross-reacting immunological material status affects treatment outcomes in Pompe disease infants. Mol Genet Metab 2010, 99:26–33.

    Article  CAS  PubMed  Google Scholar 

  13. Mendelsohn NJ, Messinger YH, Rosenberg A, Kishnani P: Elimination of antibodies to recombinant enzyme in Pompe’s disease. N Engl J Med 2009, 360:194–195.

    Article  CAS  PubMed  Google Scholar 

  14. Parenti G: Treating lysosomal storage diseases with pharmacological chaperones: from concept to clinics. EMBO Mol Med 2009, 1:268–279.

    Article  CAS  PubMed  Google Scholar 

  15. • Ramachandran N, Girard J-M, Turnbull J, Minassian B: The autosomal recessively inherited progressive myoclonus epilepsies and their genes. Epilepsia 2009, 50(Suppl 5):29–36. This is an excellent critical review of Lafora’s disease and other progressive myoclonic epilepsies.

    Article  CAS  PubMed  Google Scholar 

  16. • Ohkuma A, Noguchi S, Sugie H, et al.: Clinical and genetic analysis of lipid storage myopathies. Muscle Nerve 2009, 39:333–342. This is an excellent systematic analysis of a large cohort of patients with LSM and a critical review of the subject.

    Article  PubMed  Google Scholar 

  17. el-Hattab AW, Li F-Y, Shen J, et al.: Maternal systemic primary carnitine deficiency uncovered by newborn screening: clinical, biochemical, and molecular aspects. Genet Med 2010, 12:19–24.

    Article  CAS  PubMed  Google Scholar 

  18. • Bruno C, Bertini E, Di Rocco M, et al.: Clinical and genetic characterization of Chanarin-Dorfman syndrome. Biochem Biophys Res Comm 2008, 369:1125–1128. This is an excellent review of Chanarin-Dorfman syndrome.

    Article  CAS  PubMed  Google Scholar 

  19. •• Akman HO, Davidzon G, Tanji K, et al.: Neutral lipid storage disease with subclinical myopathy due to a retrotransposal insertion in the PNPLA2 gene. Neuromuscul Disord 2010, 20:397–402. This article discussed an interesting presymptomatic patient with an unusual mutation and a problematic disconnect between muscle biopsy and normal strength.

    Article  PubMed  Google Scholar 

  20. Gempel K, Topaloglu H, Talim B, et al.: The myopathic form of coenzyme Q10 deficiency is caused by mutations in the electron-transferring-flavoprotein dehydrogenase (ETFDH) gene. Brain 2007, 130:2037–2044.

    Article  PubMed  Google Scholar 

  21. Liang W-C, Ohkuma A, Hayashi YK, et al.: ETFDH mutations, CoQ10 levels, and respiratory chain activities in patients with riboflavin-responsive multiple acyl-CoA dehydrogenase deficiency. Neuromuscul Disord 2009, 19:212–216.

    Article  PubMed  Google Scholar 

  22. • Bonnefont J-P, Bastin J, Behin A, Djouadi F: Bezafibrate for an inborn mitochondrial beta-oxidation defect. N Engl J Med 2009, 360:838–840. This article discusses a convincing and promising therapeutic approach to an inborn error of metabolism.

    Article  CAS  PubMed  Google Scholar 

  23. Roe CR, Yang B-Z, Brunengraber H, et al.: Carnitine palmitoyltransferase II deficiency: successful anaplerotic diet therapy. Neurology 2008, 71:260–264.

    Article  CAS  PubMed  Google Scholar 

  24. Schaefer AM, McFarland R, Blakely E, et al.: Prevalence of mitochondrial DNA disease in adults. Ann Neurol 2008, 63:35–39.

    Article  CAS  PubMed  Google Scholar 

  25. •• Elliott HR, Samuels DC, Eden JA, et al.: Pathogenic mitochondrial DNA mutations are common in the general population. Am J Hum Genet 2008, 83:254–260. This article presented astonishing results of a simple but long overdue survey of healthy newborns showing an unexpectedly high prevalence of mtDNA pathogenic mutations.

    Article  CAS  PubMed  Google Scholar 

  26. • Kaufmann P, Engelstad K, Wei Y, et al.: Protean phenotypic features of the A3243G mitochondrial DNA mutation. Arch Neurol 2009, 66:85–91. This study provided interesting documentation of the clinical spectrum of the common MELAS mutation in a large cohort of mutation carriers.

    Article  PubMed  Google Scholar 

  27. • Davidson MM, Walker WF, Hernandez-Rosa E: The m.3243A>G mtDNA mutation is pathogenic in an in vitro model of the human blood brain barrier. Mitochondrion 2009, 9:463–470. This article presented elegant in vitro modeling of the human BBB and a promising tool for studies of pathogenesis and therapeutic strategies.

    Article  CAS  PubMed  Google Scholar 

  28. •• Horvath R, Kemp JP, Tuppen HAL, et al.: Molecular basis of infantile reversible cytochrome c oxidase deficiency. Brain 2009, 132:3165–3174. This was a fascinating example of the complex interaction between the nuclear and mitochondrial genomes.

    Article  PubMed  Google Scholar 

  29. Hoefs SJG, Skjeldal OH, Rodenburg RJ, et al.: Novel mutations in the NDUFS1 gene cause low residual activities in human complex I deficiencies. Mol Gen Metab 2010, 100:251–256.

    Article  CAS  Google Scholar 

  30. Lagier-Tourenne C, Tazir M, Lopez LC, et al.: ADSK3, an ancestral kinase, is mutated in a form of recessive ataxia associated with coenzyme Q10 deficiency. Am J Hum Genet 2008, 82:661–672.

    Article  CAS  PubMed  Google Scholar 

  31. Mollet J, Delahodde A, Serre V, et al.: CABC1 gene mutations cause ubiquinone deficiency with cerebellar ataxia and seizures. Am J Hum Genet 2008, 82:623–630.

    Article  CAS  PubMed  Google Scholar 

  32. Duncan AJ, Bitner-Glindziez M, Meunier B, et al.: A nonsense mutation in COQ9 causes autosomal recessive neonatal-onset primary coenzyme Q10 deficiency: a potentially treatable form of mitochondrial disease. Am J Hum Genet 2009, 84:558–566.

    Article  CAS  PubMed  Google Scholar 

  33. • Sacconi S, Trevisson E, Salviati L, et al.: Coenzyme Q10 is frequently reduced in muscle of patients with mitochondrial myopathy. Neuromuscul Disord 2010, 20:44–48. This was an interesting collaborative study documenting secondary CoQ 10 deficiency in patients with primary mitochondrial myopathy.

    Article  PubMed  Google Scholar 

  34. Cordero MD, Moreno-Fernandez AM, deMiguel M, et al.: Coenzyme Q10 distribution is altered in patients with fibromyalgia. Clin Biochem 2009, 42:732–735.

    Article  CAS  PubMed  Google Scholar 

  35. •• Massa V, Fernandez-Vizarra E, Alshahwan S, et al.: Severe infantile encephalomyopathy caused by a mutation in COX6B1, a nucleus-encoded subunit of cytochrome c oxidase. Am J Hum Genet 2008, 82:1281–1289. This study was a historical “first” showing that mutations affecting COX subunits directly do occur.

    Article  CAS  PubMed  Google Scholar 

  36. Sugiana C, Pagliarini DJ, McKenzie M, et al.: Mutation of C20orf7 disrupts complex I assembly and causes lethal neonatal mitochondrial disease. Am J Hum Genet 2008, 83:468–478.

    Article  CAS  PubMed  Google Scholar 

  37. Saada A, Edvardson S, Rapaport M, et al.: C6ORF66 is an assembly factor of mitochondrial complex I. Am J Hum Genet 2008, 82:32–38.

    Article  CAS  PubMed  Google Scholar 

  38. •• Weraarpachai W, Antonicka H, Sasarman F, et al.: Mutation in TACO1, encoding a translational activator of COX I, results in cytochrome c oxidase deficiency and late-onset Leigh syndrome. Nat Genet 2009, 41:833–837. This was the first demonstration that “indirect hits” can affect isolated subunits of RC complexes.

    Article  CAS  PubMed  Google Scholar 

  39. Cizkova A, Stranecky V, Mayr JA, et al.: TMEM70 mutations cause isolated ATP synthase deficiency and neonatal mitochondrial encephalomyopathy. Nat Genet 2009, 40:1288–1290.

    Article  Google Scholar 

  40. •• Yu-Wai-Man P, Griffiths PG, Gorman GS, et al.: Multi-system neurological disease is common in patients with OPA1 mutations. Brain 2010, 133:771–786. This was an important demonstration of the protean phenotypic expression of OPA1 mutations.

    Article  CAS  PubMed  Google Scholar 

  41. • Smits P, Smeitink JAM, van den Heuvel B: Mitochondrial translation and beyond: processes implicated in combined oxidative phosphorylation deficiencies. J Biomed Biotechnol 2010 Apr 13 (Epub ahead of print). This is a comprehensive and comprehensible review of a complex subject.

  42. • Chen H, Chan DC: Mitochondrial dynamics—fusion, fission, movement, and mitophagy—in neurodegenerative diseases. Hum Mol Genet 2009, 18:R169–R176. This is a lucid review of mitochondrial dynamics gone awry.

    Article  CAS  PubMed  Google Scholar 

  43. Muller-Hocker J: Mitochondria and ageing. Brain Pathol 1992, 2:149–158.

    Article  CAS  PubMed  Google Scholar 

  44. •• Wenz T, Diaz F, Spiegelman BM, Moraes CT: Activation of the PPAR/PGC-1alpha pathway prevents a bioenergetic deficit and effectively improves a mitochondrial myopathy phenotype. Cell Metab 2008, 8:249–255. This is an elegant demonstration of how enhancing mitochondrial biogenesis can benefit an RC defect.

    Article  CAS  PubMed  Google Scholar 

  45. Wenz T, Rossi S, Rotundo RL, et al.: Increased muscle PGC-1alpha expression protects from sarcopenia and metabolic disease during aging. Proc Natl Acad Sci U S A 2009, 106:20405–20410.

    Article  CAS  PubMed  Google Scholar 

  46. • Wenz T, Diaz F, Hernandez D, Moraes CT: Endurance exercise is protective for mice with mitochondrial myopathy. J Appl Physiol 2009, 106:1712–1719. This article explains why aerobic exercise is beneficial in mitochondrial myopathies.

    Article  CAS  PubMed  Google Scholar 

  47. Civitarese AE, Carling S, Heilbronn LK, et al.: Calorie restriction increases muscle mitochondrial biogenesis in healthy humans. PLoS Med 2007, 4:e76.

    Article  PubMed  Google Scholar 

  48. Mathew SS, Mao X, Keegan KA, et al.: Ventricular cerebrospinal fluid lactate is increased in chronic fatigue syndrome compared with generalized anxiety disorder: an in vivo 3.0 T 1H MRS imaging study. NMR Biomed 2009; 22:251–258.

    Article  CAS  PubMed  Google Scholar 

  49. Myhill S, Booth NE, McLaren-Howard J: Chronic fatigue syndrome and mitochondrial dysfunction. Int J Clin Exp Med 2009, 2:1–16.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgments

This work was supported by grants from the National Institute of Health (HD 32062) and from the Marriott Mitochondrial Disorder Clinical Research Fund.

Disclosure

No potential conflicts of interest relevant to this article were reported.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Salvatore DiMauro.

Rights and permissions

Reprints and permissions

About this article

Cite this article

DiMauro, S., Garone, C. & Naini, A. Metabolic Myopathies. Curr Rheumatol Rep 12, 386–393 (2010). https://doi.org/10.1007/s11926-010-0119-9

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11926-010-0119-9

Keywords

Navigation