Skip to main content
Log in

ε-Sarcoglycan: Unraveling the Myoclonus-Dystonia Gene

  • Published:
Molecular Neurobiology Aims and scope Submit manuscript

Abstract

Myoclonus-dystonia (MD) is a rare childhood-onset movement disorder, with an estimated prevalence of about 2 per 1,000,.000 in Europe, characterized by myoclonic jerks in combination with focal or segmental dystonia. Pathogenic variants in the gene encoding ε-sarcoglycan (SGCE), a maternally imprinted gene, are the most frequent genetic cause of MD. To date, the exact role of ε-sarcoglycan and the pathogenic mechanisms that lead to MD are still unknown. However, there are more than 40 reported isoforms of human ε-sarcoglycan, pointing to a complex biology of this protein. Additionally, some of these are brain-specific isoforms, which may suggest an important role within the central nervous system. In the present review, we aim to provide an overview of the current state of knowledge of ε-sarcoglycan. We will focus on the genetic landscape of SGCE and the presence and plausible role of ε-sarcoglycan in the brain. Finally, we discuss the importance of the brain-specific isoforms and hypothesize that SGCE may play essential roles in normal synaptic functioning and their alteration will be strongly related to MD.

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.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

Data Availability

Not applicable.

References

  1. Asmus F, Gasser T (2010) Dystonia-plus syndromes. Eur J Neurol 17(Suppl 1):37–45. https://doi.org/10.1111/j.1468-1331.2010.03049.x

    Article  PubMed  Google Scholar 

  2. Roze E, Lang AE, Vidailhet M (2018) Myoclonus-dystonia: classification, phenomenology, pathogenesis, and treatment. Curr Opin Neurol 31:484–490. https://doi.org/10.1097/WCO.0000000000000577

    Article  PubMed  Google Scholar 

  3. Nardocci N, Zorzi G, Barzaghi C, Zibordi F, Ciano C, Ghezzi D, Garavaglia B (2008) Myoclonus-dystonia syndrome: clinical presentation, disease course, and genetic features in 11 families. Mov Disord 23:28–34. https://doi.org/10.1002/mds.21715

    Article  PubMed  Google Scholar 

  4. Timmers ER, Smit M, Kuiper A, Bartels AL, van der Veen S, van der Stouwe AMM, Santens P, Bergmans B et al (2019) Myoclonus-dystonia: distinctive motor and non-motor phenotype from other dystonia syndromes. Parkinsonism Relat Disord 69:85–90. https://doi.org/10.1016/j.parkreldis.2019.10.015

    Article  PubMed  Google Scholar 

  5. Wang X, Yu X (2021) Deep brain stimulation for myoclonus dystonia syndrome: a meta-analysis with individual patient data. Neurosurg Rev 44:451–462. https://doi.org/10.1007/s10143-019-01233-x

    Article  PubMed  Google Scholar 

  6. Kosutzka Z, Tisch S, Bonnet C, Ruiz M, Hainque E, Welter ML, Viallet F, Karachi C et al (2019) Long-term GPi-DBS improves motor features in myoclonus-dystonia and enhances social adjustment. Mov Disord 34:87–94. https://doi.org/10.1002/mds.27474

    Article  PubMed  Google Scholar 

  7. Hess CW, Raymond D, Aguiar Pde C, Frucht S, Shriberg J, Heiman GA, Kurlan R, Klein C et al (2007) Myoclonus-dystonia, obsessive-compulsive disorder, and alcohol dependence in SGCE mutation carriers. Neurology. 68:522–524. https://doi.org/10.1212/01.wnl.0000253188.76092.06

    Article  CAS  PubMed  Google Scholar 

  8. Weissbach A, Werner E, Bally JF, Tunc S, Löns S, Timmann D, Zeuner KE, Tadic V et al (2017) Alcohol improves cerebellar learning deficit in myoclonus-dystonia: a clinical and electrophysiological investigation. Ann Neurol 82:543–553. https://doi.org/10.1002/ana.25035

    Article  CAS  PubMed  Google Scholar 

  9. Nygaard TG, Raymond D, Chen C, Nishino I, Greene PE, Jennings D, Heiman GA, Klein C et al (1999) Localization of a gene for myoclonus-dystonia to chromosome 7q21-q31. Ann Neurol 46:794–798. https://doi.org/10.1002/1531-8249(199911)46:5<794::aid-ana19>3.0.co;2-2

    Article  CAS  PubMed  Google Scholar 

  10. Zimprich A, Grabowski M, Asmus F, Naumann M, Berg D, Bertram M, Scheidtmann K, Kern P et al (2001) Mutations in the gene encoding epsilon-sarcoglycan cause myoclonus-dystonia syndrome. Nat Genet 29:66–69. https://doi.org/10.1038/ng709

    Article  CAS  PubMed  Google Scholar 

  11. Carecchio M, Magliozzi M, Copetti M, Ferraris A, Bernardini L, Bonetti M, Defazio G, Edwards MJ et al (2013) Defining the epsilon-sarcoglycan (SGCE) gene phenotypic signature in myoclonus-dystonia: a reappraisal of genetic testing criteria. Mov Disord 28:787–794. https://doi.org/10.1002/mds.25506

    Article  CAS  PubMed  Google Scholar 

  12. Ghosh D, Indulkar S (2013) Primary myoclonus-dystonia: a diagnosis often missed in children. J Child Neurol 28:1418–1422. https://doi.org/10.1177/0883073813488677

    Article  PubMed  Google Scholar 

  13. Peall KJ, Kurian MA, Wardle M, Waite AJ, Hedderly T, Lin JP, Smith M, Whone A et al (2014) SGCE and myoclonus dystonia: motor characteristics, diagnostic criteria and clinical predictors of genotype. J Neurol 261:2296–2304. https://doi.org/10.1007/s00415-014-7488-3

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Vanegas MI, Marcé-Grau A, Martí-Sánchez L, Mellid S, Baide-Mairena H, Correa-Vela M, Cazurro A, Rodríguez C et al (2020) Delineating the motor phenotype of SGCE-myoclonus dystonia syndrome. Parkinsonism Relat Disord 80:165–174. https://doi.org/10.1016/j.parkreldis.2020.09.023

    Article  PubMed  Google Scholar 

  15. Ettinger AJ, Feng G, Sanes JR (1997) Epsilon-Sarcoglycan, a broadly expressed homologue of the gene mutated in limb-girdle muscular dystrophy 2D. J Biol Chem 272:32534–32538. https://doi.org/10.1074/jbc.272.51.32534

    Article  CAS  PubMed  Google Scholar 

  16. McNally EM, Ly CT, Kunkel LM (1998) Human epsilon-sarcoglycan is highly related to alpha-sarcoglycan (adhalin), the limb girdle muscular dystrophy 2D gene. FEBS Lett 422:27–32. https://doi.org/10.1016/s0014-5793(97)01593-7

    Article  CAS  PubMed  Google Scholar 

  17. Rachad L, El Kadmiri N, Slassi I, El Otmani H, Nadifi S (2017) Genetic aspects of myoclonus-dystonia syndrome (MDS). Mol Neurobiol 54:939–942. https://doi.org/10.1007/s12035-016-9712-x

    Article  CAS  PubMed  Google Scholar 

  18. Grabowski M, Zimprich A, Lorenz-Depiereux B, Kalscheuer V, Asmus F, Gasser T, Meitinger T, Strom TM (2003) The epsilon-sarcoglycan gene (SGCE), mutated in myoclonus-dystonia syndrome, is maternally imprinted. Eur J Hum Genet 11:138–144. https://doi.org/10.1038/sj.ejhg.5200938

    Article  CAS  PubMed  Google Scholar 

  19. Grütz K, Seibler P, Weissbach A, Lohmann K, Carlisle FA, Blake DJ, Westenberger A, Klein C et al (2017) Faithful SGCE imprinting in iPSC-derived cortical neurons: an endogenous cellular model of myoclonus-dystonia. Sci Rep 7:41156. https://doi.org/10.1038/srep41156

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Yokoi F, Dang MT, Mitsui S, Li Y (2005) Exclusive paternal expression and novel alternatively spliced variants of epsilon-sarcoglycan mRNA in mouse brain. FEBS Lett 579:4822–4828. https://doi.org/10.1016/j.febslet.2005.07.065

    Article  CAS  PubMed  Google Scholar 

  21. Ritz K, van Schaik BD, Jakobs ME, van Kampen AH, Aronica E, Tijssen MA, Baas F (2011) SGCE isoform characterization and expression in human brain: implications for myoclonus-dystonia pathogenesis? Eur J Hum Genet 19:438–444. https://doi.org/10.1038/ejhg.2010.206

    Article  CAS  PubMed  Google Scholar 

  22. Nishiyama A, Endo T, Takeda S, Imamura M (2004) Identification and characterization of epsilon-sarcoglycans in the central nervous system. Brain Res Mol Brain Res 125:1–12. https://doi.org/10.1016/j.molbrainres.2004.01.012

    Article  CAS  PubMed  Google Scholar 

  23. Chawla G, Lin CH, Han A, Shiue L, Ares M Jr, Black DL (2009) Sam68 regulates a set of alternatively spliced exons during neurogenesis. Mol Cell Biol 29:201–213. https://doi.org/10.1128/MCB.01349-08

    Article  CAS  PubMed  Google Scholar 

  24. Grünewald A, Djarmati A, Lohmann-Hedrich K, Farrell K, Zeller JA, Allert N, Papengut F, Petersen B et al (2008) Myoclonus-dystonia: significance of large SGCE deletions. Hum Mutat 29:331–332. https://doi.org/10.1002/humu.9521

    Article  PubMed  Google Scholar 

  25. Fokkema IF, den Dunnen JT, Taschner PE (2005) LOVD: easy creation of a locus-specific sequence variation database using an "LSDB-in-a-box" approach. Hum Mutat 26:63–68. https://doi.org/10.1002/humu.20201

    Article  CAS  PubMed  Google Scholar 

  26. Esapa CT, Waite A, Locke M, Benson MA, Kraus M, McIlhinney RA, Sillitoe RV, Beesley PW et al (2007) SGCE missense mutations that cause myoclonus-dystonia syndrome impair epsilon-sarcoglycan trafficking to the plasma membrane: modulation by ubiquitination and torsinA. Hum Mol Genet 16:327–342. https://doi.org/10.1093/hmg/ddl472

    Article  CAS  PubMed  Google Scholar 

  27. Asmus F, Salih F, Hjermind LE, Ostergaard K, Munz M, Kühn AA, Dupont E, Kupsch A et al (2005) Myoclonus-dystonia due to genomic deletions in the epsilon-sarcoglycan gene. Ann Neurol 58:792–797. https://doi.org/10.1002/ana.20661

    Article  CAS  PubMed  Google Scholar 

  28. Asmus F, Hjermind LE, Dupont E, Wagenstaller J, Haberlandt E, Munz M, Strom TM, Gasser T (2007) Genomic deletion size at the epsilon-sarcoglycan locus determines the clinical phenotype. Brain. 130:2736–2745. https://doi.org/10.1093/brain/awm209

    Article  PubMed  Google Scholar 

  29. Vado Y, Pereda A, Llano-Rivas I, Gorria-Redondo N, Díez I, Perez de Nanclares G (2020) Novel variant in PLAG1 in a familial case with Silver-Russell Syndrome suspicion. Genes (Basel) 11:1461. https://doi.org/10.3390/genes11121461

    Article  CAS  Google Scholar 

  30. Sheridan MB, Bytyci Telegrafi A, Stinnett V, Umeh CC, Mari Z, Dawson TM, Bodurtha J, Batista DA (2013) Myoclonus-dystonia and Silver-Russell syndrome resulting from maternal uniparental disomy of chromosome 7. Clin Genet 84:368–372. https://doi.org/10.1111/cge.12075

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Guettard E, Portnoi MF, Lohmann-Hedrich K, Keren B, Rossignol S, Winkler S, El Kamel I, Leu S et al (2008) Myoclonus-dystonia due to maternal uniparental disomy. Arch Neurol 65:1380–1385. https://doi.org/10.1001/archneur.65.10.1380

    Article  PubMed  Google Scholar 

  32. Stark Z, Ryan MM, Bruno DL, Burgess T, Savarirayan R (2010) Atypical Silver-Russell phenotype resulting from maternal uniparental disomy of chromosome 7. Am J Med Genet A 152A:2342–2345. https://doi.org/10.1002/ajmg.a.33590

    Article  PubMed  Google Scholar 

  33. Augustine EF, Blackburn J, Pellegrino JE, Miller R, Mink JW (2013) Myoclonus-dystonia syndrome associated with Russell Silver syndrome. Mov Disord 28:841–842. https://doi.org/10.1002/mds.25483

    Article  PubMed  PubMed Central  Google Scholar 

  34. Martins J, Gabriel D, Borges T, Soares G, Temudo T (2020) Child neurology: myoclonus-dystonia in Russell-Silver syndrome: two syndromes caused by one genetic defect. Neurology 95:e936–e938. https://doi.org/10.1212/WNL.0000000000010076

    Article  PubMed  Google Scholar 

  35. Menozzi E, Balint B, Latorre A, Valente EM, Rothwell JC, Bhatia KP (2019) Twenty years on: myoclonus-dystonia and ε-sarcoglycan - neurodevelopment, channel, and signaling dysfunction. Mov Disord 34:1588–1601. https://doi.org/10.1002/mds.27822

    Article  CAS  PubMed  Google Scholar 

  36. Online Mendelian Inheritance in Man, OMIM® (2021) McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University (Baltimore, MD). https://omim.org/. Accessed 28th January

  37. UniProt Consortium (2019) UniProt: a worldwide hub of protein knowledge. Nucleic Acids Res 47(D1):D506–D515. https://doi.org/10.1093/nar/gky1049

    Article  CAS  Google Scholar 

  38. Sandonà D, Betto R (2009) Sarcoglycanopathies: molecular pathogenesis and therapeutic prospects. Expert Rev Mol Med 11:e28. https://doi.org/10.1017/S1462399409001203

    Article  PubMed  PubMed Central  Google Scholar 

  39. Gao QQ, McNally EM (2015) The dystrophin complex: structure, function, and implications for therapy. Compr Physiol 5:1223–1239. https://doi.org/10.1002/cphy.c140048

    Article  PubMed  PubMed Central  Google Scholar 

  40. Waite A, De Rosa MC, Brancaccio A, Blake DJ (2011) A gain-of-glycosylation mutation associated with myoclonus-dystonia syndrome affects trafficking and processing of mouse ε-sarcoglycan in the late secretory pathway. Hum Mutat 32:1246–1258. https://doi.org/10.1002/humu.21561

    Article  CAS  PubMed  Google Scholar 

  41. Xiao J, LeDoux MS (2003) Cloning, developmental regulation and neural localization of rat epsilon-sarcoglycan. Brain Res Mol Brain Res 119:132–143. https://doi.org/10.1016/j.molbrainres.2003.09.004

    Article  CAS  PubMed  Google Scholar 

  42. Hunt SE, McLaren W, Gil L, Thormann A, Schuilenburg H, Sheppard D, Parton A, Armean IM et al (2018) Ensembl variation resources. Database (Oxford). https://doi.org/10.1093/database/bay119

  43. Sheng M, Sala C (2001) PDZ domains and the organization of supramolecular complexes. Annu Rev Neurosci 24:1–29. https://doi.org/10.1146/annurev.neuro.24.1.1

    Article  CAS  PubMed  Google Scholar 

  44. Imamura M, Araishi K, Noguchi S, Ozawa E (2000) A sarcoglycan-dystroglycan complex anchors Dp116 and utrophin in the peripheral nervous system. Hum Mol Genet 9:3091–3100. https://doi.org/10.1093/hmg/9.20.3091

    Article  CAS  PubMed  Google Scholar 

  45. Maltese M, Martella G, Imbriani P, Schuermans J, Billion K, Sciamanna G, Farook F, Ponterio G et al (2017) Abnormal striatal plasticity in a DYT11/SGCE myoclonus dystonia mouse model is reversed by adenosine A2A receptor inhibition. Neurobiol Dis 108:128–139. https://doi.org/10.1016/j.nbd.2017.08.007

    Article  CAS  PubMed  Google Scholar 

  46. Waite AJ, Carlisle FA, Chan YM, Blake DJ (2016) Myoclonus dystonia and muscular dystrophy: ɛ-sarcoglycan is part of the dystrophin-associated protein complex in brain. Mov Disord 31:1694–1703. https://doi.org/10.1002/mds.26738

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Xiao J, Vemula SR, Xue Y, Khan MM, Carlisle FA, Waite AJ, Blake DJ, Dragatsis I et al (2017) Role of major and brain-specific Sgce isoforms in the pathogenesis of myoclonus-dystonia syndrome. Neurobiol Dis 98:52–65. https://doi.org/10.1016/j.nbd.2016.11.003

    Article  CAS  PubMed  Google Scholar 

  48. Cai H, Erdman RA, Zweier L, Chen J, Shaw JH 4th, Baylor KA, Stecker MM, Carey DJ et al (2007) The sarcoglycan complex in Schwann cells and its role in myelin stability. Exp Neurol 205:257–269. https://doi.org/10.1016/j.expneurol.2007.02.015

    Article  CAS  PubMed  Google Scholar 

  49. Lancioni A, Rotundo IL, Kobayashi YM, D'Orsi L, Aurino S, Nigro G, Piluso G, Acampora D et al (2011) Combined deficiency of alpha and epsilon sarcoglycan disrupts the cardiac dystrophin complex. Hum Mol Genet 20:4644–4654. https://doi.org/10.1093/hmg/ddr398

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Waite A, Tinsley CL, Locke M, Blake DJ (2009) The neurobiology of the dystrophin-associated glycoprotein complex. Ann Med 41:344–359. https://doi.org/10.1080/07853890802668522

    Article  CAS  PubMed  Google Scholar 

  51. Shi W, Chen Z, Schottenfeld J, Stahl RC, Kunkel LM, Chan YM (2004) Specific assembly pathway of sarcoglycans is dependent on beta- and delta-sarcoglycan. Muscle Nerve 29:409–419. https://doi.org/10.1002/mus.10566

    Article  CAS  PubMed  Google Scholar 

  52. Tarakci H, Berger J (2016) The sarcoglycan complex in skeletal muscle. Front Biosci (Landmark Ed) 21:744–756. https://doi.org/10.2741/4418

    Article  CAS  Google Scholar 

  53. Allikian MJ, McNally EM (2007) Processing and assembly of the dystrophin glycoprotein complex. Traffic. 8:177–183. https://doi.org/10.1111/j.1600-0854.2006.00519.x

    Article  CAS  PubMed  Google Scholar 

  54. Waite A, Brown SC, Blake DJ (2012) The dystrophin-glycoprotein complex in brain development and disease. Trends Neurosci 35:487–496. https://doi.org/10.1016/j.tins.2012.04.004

    Article  CAS  PubMed  Google Scholar 

  55. Yokoi F, Dang MT, Zhou T, Li Y (2012) Abnormal nuclear envelopes in the striatum and motor deficits in DYT11 myoclonus-dystonia mouse models. Hum Mol Genet 21:916–925. https://doi.org/10.1093/hmg/ddr528

    Article  CAS  PubMed  Google Scholar 

  56. Shang S, Hua F, Hu ZW (2017) The regulation of β-catenin activity and function in cancer: therapeutic opportunities. Oncotarget 8:33972–33989. https://doi.org/10.18632/oncotarget.15687

    Article  PubMed  PubMed Central  Google Scholar 

  57. Valenta T, Hausmann G, Basler K (2012) The many faces and functions of β-catenin. EMBO J 31:2714–2736. https://doi.org/10.1038/emboj.2012.150

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Blake DJ, Hawkes R, Benson MA, Beesley PW (1999) Different dystrophin-like complexes are expressed in neurons and glia. J Cell Biol 147:645–658. https://doi.org/10.1083/jcb.147.3.645

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Tozawa T, Itoh K, Yaoi T, Tando S, Umekage M, Dai H, Hosoi H, Fushiki S (2012) The shortest isoform of dystrophin (Dp40) interacts with a group of presynaptic proteins to form a presumptive novel complex in the mouse brain. Mol Neurobiol 45:287–297. https://doi.org/10.1007/s12035-012-8233-5

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Yokoi F, Dang MT, Li J, Li Y (2006) Myoclonus, motor deficits, alterations in emotional responses and monoamine metabolism in epsilon-sarcoglycan deficient mice. J Biochem 140:141–146. https://doi.org/10.1093/jb/mvj138

    Article  CAS  PubMed  Google Scholar 

  61. Yokoi F, Yang G, Li J, DeAndrade MP, Zhou T, Li Y (2010) Earlier onset of motor deficits in mice with double mutations in Dyt1 and Sgce. J Biochem 148:459–466. https://doi.org/10.1093/jb/mvq078

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Washburn S, Fremont R, Moreno-Escobar MC, Angueyra C, Khodakhah K (2019) Acute cerebellar knockdown of Sgce reproduces salient features of myoclonus-dystonia (DYT11) in mice. Elife. 8:e52101. https://doi.org/10.7554/eLife.52101

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Li J, Liu Y, Li Q, Huang X, Zhou D, Xu H, Zhao F, Mi X et al (2020) Mutation in ε-sarcoglycan induces a myoclonus-dystonia syndrome-like movement disorder in mice. Neurosci Bull 37:311–322. https://doi.org/10.1007/s12264-020-00612-5

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Zhang L, Yokoi F, Parsons DS, Standaert DG, Li Y (2012) Alteration of striatal dopaminergic neurotransmission in a mouse model of DYT11 myoclonus-dystonia. PLoS One 7:e33669. https://doi.org/10.1371/journal.pone.0033669

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Yokoi F, Dang MT, Yang G, Li J, Doroodchi A, Zhou T, Li Y (2012) Abnormal nuclear envelope in the cerebellar Purkinje cells and impaired motor learning in DYT11 myoclonus-dystonia mouse models. Behav Brain Res 227:12–20. https://doi.org/10.1016/j.bbr.2011.10.024

    Article  CAS  PubMed  Google Scholar 

  66. Raymond D, Saunders-Pullman R, de Carvalho AP, Schule B, Kock N, Friedman J, Harris J, Ford B et al (2008) Phenotypic spectrum and sex effects in eleven myoclonus-dystonia families with epsilon-sarcoglycan mutations. Mov Disord 23:588–592. https://doi.org/10.1002/mds.21785

    Article  PubMed  Google Scholar 

  67. Anderson JL, Head SI, Morley JW (2003) Altered inhibitory input to Purkinje cells of dystrophin-deficient mice. Brain Res 982:280–283. https://doi.org/10.1016/s0006-8993(03)03018-x

    Article  CAS  PubMed  Google Scholar 

  68. Anderson JL, Head SI, Morley JW (2004) Long-term depression is reduced in cerebellar Purkinje cells of dystrophin-deficient mdx mice. Brain Res 1019:289–292. https://doi.org/10.1016/j.brainres.2004.06.011

    Article  CAS  PubMed  Google Scholar 

  69. Briatore F, Pregno G, Di Angelantonio S, Frola E, De Stefano ME, Vaillend C, Sassoè-Pognetto M, Patrizi A (2020) Dystroglycan mediates clustering of essential GABAergic components in cerebellar Purkinje cells. Front Mol Neurosci 13:164. https://doi.org/10.3389/fnmol.2020.00164

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Roze E, Apartis E, Clot F, Dorison N, Thobois S, Guyant-Marechal L, Tranchant C, Damier P et al (2008) Myoclonus-dystonia: clinical and electrophysiologic pattern related to SGCE mutations. Neurology. 70:1010–1016. https://doi.org/10.1212/01.wnl.0000297516.98574.c0

    Article  CAS  PubMed  Google Scholar 

  71. Hubsch C, Vidailhet M, Rivaud-Péchoux S, Pouget P, Brochard V, Degos B, Pélisson D, Golmard JL et al (2011) Impaired saccadic adaptation in DYT11 dystonia. J Neurol Neurosurg Psychiatry 82:1103–1106. https://doi.org/10.1136/jnnp.2010.232793

    Article  PubMed  Google Scholar 

  72. Popa T, Milani P, Richard A, Hubsch C, Brochard V, Tranchant C, Sadnicka A, Rothwell J et al (2014) The neurophysiological features of myoclonus-dystonia and differentiation from other dystonias. JAMA Neurol 71:612–619. https://doi.org/10.1001/jamaneurol.2014.99

    Article  PubMed  Google Scholar 

  73. Carbon M, Raymond D, Ozelius L, Saunders-Pullman R, Frucht S, Dhawan V, Bressman S, Eidelberg D (2013) Metabolic changes in DYT11 myoclonus-dystonia. Neurology. 80:385–391. https://doi.org/10.1212/WNL.0b013e31827f0798

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Welter ML, Grabli D, Karachi C, Jodoin N, Fernandez-Vidal S, Brun Y, Navarro S, Rogers A et al (2015) Pallidal activity in myoclonus dystonia correlates with motor signs. Mov Disord 30:992–996. https://doi.org/10.1002/mds.26244

    Article  PubMed  Google Scholar 

  75. Liu X, Griffin IC, Parkin SG, Miall RC, Rowe JG, Gregory RP, Scott RB, Aziz TZ et al (2002) Involvement of the medial pallidum in focal myoclonic dystonia: a clinical and neurophysiological case study. Mov Disord 17:346–353. https://doi.org/10.1002/mds.10038

    Article  PubMed  Google Scholar 

  76. Foncke EM, Bour LJ, Speelman JD, Koelman JH, Tijssen MA (2007) Local field potentials and oscillatory activity of the internal globus pallidus in myoclonus-dystonia. Mov Disord 22:369–376. https://doi.org/10.1002/mds.21284

    Article  PubMed  Google Scholar 

  77. Tewari A, Fremont R, Khodakhah K (2017) It's not just the basal ganglia: cerebellum as a target for dystonia therapeutics. Mov Disord 32:1537–1545. https://doi.org/10.1002/mds.27123

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Chen CH, Fremont R, Arteaga-Bracho EE, Khodakhah K (2014) Short latency cerebellar modulation of the basal ganglia. Nat Neurosci 17:1767–1775. https://doi.org/10.1038/nn.3868

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Kurosaki T, Popp MW, Maquat LE (2019) Quality and quantity control of gene expression by nonsense-mediated mRNA decay. Nat Rev Mol Cell Biol 20:406–420. https://doi.org/10.1038/s41580-019-0126-2

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by a Grant from Myoclonus Dystonia Syndrome Spanish association (ALUDME) (grant PIC/124/16), grant PI18/01319 from the Plan Nacional de I +D + I, award AC17/00005 by the Carlos III Health Institute (ISCIII) through AES2017 and within the NEURON framework, Career Integration Grant (ref. 304111), Ramón y Cajal Fellowship (RYC-2011-08391p), IEDI-2017-00822 by the Spanish Ministry of Economy and Competitiveness, and Agency for Management of University and Research Grants (AGAUR) (SGR14-297 and 2017 SGR 1776).

Funding

This work was supported by a Grant from Myoclonus Dystonia Syndrome Spanish association (ALUDME) (grant PIC/124/16) and grant PI18/01319 from the Plan Nacional de I +D + I.

AB. financial support was provided by: Award AC17/00005 by ISCIII through AES2017 and within the NEURON framework, Career Integration Grant (ref. 304111), Ramón y Cajal Fellowship (RYC-2011-08391p), IEDI-2017-00822, and AGAUR (SGR14-297 and 2017 SGR 1776).

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to drafting and critically reviewing the manuscript.

Corresponding author

Correspondence to Belén Pérez-Dueñas.

Ethics declarations

Ethical standards

Not applicable.

Consent to participate

Not applicable.

Consent for publication

Not applicable.

Conflict of interest

The authors declare no competing interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Supplementary Table 1

Updated list of SGCE (NM_003919) variants associated to MD; last revised March 17th, 2021. Clinical significance is established according to the American College of Medical Genetics and Genomics (ACMG) guidelines. Abbreviations: P, pathogenic; LP, likely pathogenic; VUS, variant of uncertain significance; B, benign (DOCX 45.4 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cazurro-Gutiérrez, A., Marcé-Grau, A., Correa-Vela, M. et al. ε-Sarcoglycan: Unraveling the Myoclonus-Dystonia Gene. Mol Neurobiol 58, 3938–3952 (2021). https://doi.org/10.1007/s12035-021-02391-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12035-021-02391-0

Keywords

Navigation