Skip to main content

Advertisement

Log in

Spinocerebellar Ataxia Type 2: Clinical Presentation, Molecular Mechanisms, and Therapeutic Perspectives

  • Published:
Molecular Neurobiology Aims and scope Submit manuscript

Abstract

Spinocerebellar ataxia type 2 (SCA2) is an autosomal dominant genetic disease characterized by cerebellar dysfunction associated with slow saccades, early hyporeflexia, severe tremor of postural or action type, peripheral neuropathy, cognitive disorders, and other multisystemic features. SCA2, one of the most common ataxias worldwide, is caused by the expansion of a CAG triplet repeat located in the N-terminal coding region of the ATXN2 gene, which results in the incorporation of a segment of polyglutamines in the mutant protein, being longer expansions associated with earlier onset and more sever disease in subsequent generations. In this review, we offer a detailed description of the clinical manifestations of SCA2 and compile the experimental evidence showing the participation of ataxin-2 in crucial cellular processes, including messenger RNA maturation and translation, and endocytosis. In addition, we discuss in the light of present data the potential molecular mechanisms underlying SCA2 pathogenesis. The mutant protein exhibits a toxic gain of function that is mainly attributed to the generation of neuronal inclusions of phosphorylated and/or proteolytic cleaved mutant ataxin-2, which might alter normal ataxin-2 function, leading to cell dysfunction and death of target cells. In the final part of this review, we discuss the perspectives of development of therapeutic strategies for SCA2. Based on previous experience with other polyglutamine disorders and considering the molecular basis of SCA2 pathogenesis, a nuclei-acid-based strategy focused on the specific silencing of the dominant disease allele that preserves the expression of the wild-type allele is highly desirable and might prevent toxic neurodegenerative sequelae.

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

Similar content being viewed by others

Abbreviations

SCA2:

Spinocerebellar ataxia type 2

polyQ:

Polyglutamine

SBMA:

Spinal bulbar muscular atrophy

HD:

Huntington’s disease

SCA:

Spinocerebellar ataxia

SCA1:

Spinocerebellar ataxia type 1

SCA3:

Spinocerebellar ataxia type 3

SCA6:

Spinocerebellar ataxia type 6

SCA7:

Spinocerebellar ataxia type 7

SCA17:

Spinocerebellar ataxia type 17

SCA10:

Spinocerebellar ataxia type 10

OPCA:

Olivopontocerebellar atrophy

REM:

Rapid eye movement

ALS:

Amyotrophic lateral sclerosis

PD:

Parkinson disease

MSA:

Multiple system atrophy

Lsm:

Like Sm domain

LsmAD:

Lsm-associated domain

PAM2:

PABPC interacting motif-2

A2D:

Ataxin-2 domain protein

PABP:

Polyadenylate-binding protein

Pbp1:

Ataxin-2 homolog in yeast

Pab1:

PABP homolog in yeast

A2BP1:

Ataxin-2-binding protein

RNP:

Ribonucleoprotein

LTP:

Long-term potentiation

RBM9:

RNA-binding motif protein 9

RBPMS:

RNA-binding protein with multiple splicing

InsP3R1:

Type 1 inositol (1,4,5)-triphosphate receptor

RyanR1:

Ryanodine receptor

DRPLA:

Dentatorubral–pallidoluysian atrophy

UPS:

Ubiquitin–proteasome system

shRNA:

Short-hairpin RNA

AAV1:

Adeno-associated virus serotype 1

RNAi:

RNA interference

AON:

Antisense oligonucleotide

CNS:

Central nervous system

References

  1. Orr HT, Zoghbi HY (2007) Trinucleotide repeat disorders. Annu Rev Neurosci 30:575–621. doi:10.1146/annurev.neuro.29.051605.113042

    Article  PubMed  CAS  Google Scholar 

  2. Ellegren H (2000) Heterogeneous mutation processes in human microsatellite DNA sequences. Nat Genet 24:400–2. doi:10.1038/74249

    Article  PubMed  CAS  Google Scholar 

  3. Bauer PO, Nukina N (2009) The pathogenic mechanisms of polyglutamine diseases and current therapeutic strategies. J Neurochem 110:1737–1765. doi:10.1111/j.1471-4159.2009.06302.x

    Article  PubMed  CAS  Google Scholar 

  4. Harding AE (1982) The clinical features and classification of the late onset autosomal dominant cerebellar ataxias. A study of 11 families, including descendants of the Drew family of Walworth. Brain 105:1–28. doi:10.1093/brain/105.1.1

    Article  PubMed  CAS  Google Scholar 

  5. Durr A (2010) Autosomal dominant cerebellar ataxias: polyglutamine expansions and beyond. Lancet Neurol 9:885–94. doi:10.1016/S1474-4422(10)70183-6

    Article  PubMed  CAS  Google Scholar 

  6. Paulson HL (2009) The spinocerebellar ataxias. J Neuroophthalmol 29:227–37. doi:10.1097/WNO0b013e3181b416de

    Article  PubMed  Google Scholar 

  7. Manto MU (2005) The wide spectrum of spinocerebellar ataxias (SCAs). Cerebellum 4:2–6. doi:10.1080/14734220510007914

    Article  PubMed  CAS  Google Scholar 

  8. Teive HA (2009) Spinocerebellar ataxias. Arq Neuropsiquiatr 67(4):1133–42. doi:10.1590/S0004-282X2009000600035

    Article  PubMed  Google Scholar 

  9. Trott A, Houenou LJ (2012) Mini-review: spinocerebellar ataxias: an update of SCA genes. Recent Pat DNA Gene Seq 6:115–121

    Article  PubMed  CAS  Google Scholar 

  10. Pearson CE, Nichol-Edamura K, Cleary JD (2005) Repeat instability: mechanisms of dynamic mutations. Nat Rev Genet 6:729–42. doi:10.1083/nrg1689

    Article  PubMed  CAS  Google Scholar 

  11. Velázquez Pérez L, Cruz GS, Santos Falcón N, Enrique Almaguer Mederos L, Escalona Batallan K, Rodríguez Labrada R et al (2009) Molecular epidemiology of spinocerebellar ataxias in Cuba: insights into SCA2 founder effect in Holguín. Neurosci Lett 454:157–60. doi:10.1016/j.neulet.2009.03.015

    Article  PubMed  CAS  Google Scholar 

  12. Saleem Q, Choudhry S, Mukerji M, Bashyam L, Padma MV, Chakravarthy A et al (2000) Molecular analysis of autosomal dominant hereditary ataxias in the Indian population: high frequency of SCA2 and evidence for a common founder mutation. Hum Genet 106:179–87. doi:10.1007/s004390051026

    Article  PubMed  CAS  Google Scholar 

  13. Velázquez Pérez L, Rodríguez Labrada R, García Rodríguez JC, Almaguer Mederos LE, Cruz-Mariño T, Laffita-Mesa JM (2011) A comprehensive review of spinocerebellar ataxia type 2 in Cuba. Cerebellum 10(2):184–198. doi:10.1007/s12311-011-0265-2

    Article  PubMed  Google Scholar 

  14. Trott A, Jardim LB, Ludwig HT, Saute JA, Artigalás O, Kieling C, Wanderley HY et al (2006) Spinocerebellar ataxias in 114 Brazilian families: clinical and molecular findings. Clin Genet 70:173–176. doi:10.1111/j.1399-0004.2006.00656.x

    Article  PubMed  CAS  Google Scholar 

  15. Velázquez-Pérez L, García R, Santos FN, Paneque HM, Medina HE, Hechavarría PR (2001) Epidemiology of Cuban hereditary ataxias. Rev Neurol 32:606–11

    PubMed  Google Scholar 

  16. Alonso E, Martínez-Ruano L, De Biase I, Mader C, Ochoa A, Yescas P et al (2007) Distinct distribution of autosomal dominant spinocerebellar ataxia in the Mexican population. Mov Disord 22:1050–3. doi:10.1002/mds.21470

    Article  PubMed  Google Scholar 

  17. Wadia NH, Swami RK (1971) A new form of heredo-familiar spinocerebellar degeneration with slow eye movements (nine families). Brain 94:359–74. doi:10.1093/brain/94.2.359

    Article  PubMed  CAS  Google Scholar 

  18. Velázquez-Pérez L, Seifried C, Santos-Falcón N, Abele M, Ziemann U, Almaguer LE et al (2004) Saccade velocity is controlled by polyglutamine size in spinocerebellar ataxia 2. Ann Neurol 56:444–7. doi:10.1002/ana.20220

    Article  PubMed  Google Scholar 

  19. Velázquez-Pérez L, Seifried C, Abele M, Wirjatijasa F, Rodríguez-Labrada R, Santos-Falcón N et al (2009) Saccade velocity is reduced in presymptomatic spinocerebellar ataxia type 2. Clin Neurophysiol 120:632–5. doi:10.1016/j.clinph.2008.12.040

    Article  PubMed  Google Scholar 

  20. Velázquez Pérez L, Sánchez Cruz G, Canales Ochoa N, Rodríguez Labrada R, Rodríguez Díaz J, Almaguer Mederos L et al (2007) Electrophysiological features in patients and presymptomatic relatives with spinocerebellar ataxia type 2. J Neurol Sci 263:158–64. doi:10.1016/j.jns.2007.07.013

    Article  PubMed  Google Scholar 

  21. Velázquez-Pérez L, Rodríguez-Labrada R, Canales-Ochoa N, Sánchez-Cruz G, Fernández-Ruiz J, Montero JM et al (2010) Progression markers of spinocerebellar ataxia 2. A twenty years neurophysiological follow up study. J Neurol Sci 290:22–6. doi:10.1016/j.jns.2009.12.013

    Article  PubMed  CAS  Google Scholar 

  22. Rodríguez-Labrada R, Velázquez-Pérez L, Canales-Ochoa N, Galicia-Polo L, Haro-Valencia R, Sánchez-Cruz G et al (2011) Subtle REM sleep abnormalities in presymptomatic spinocerebellar ataxia type 2 gene carriers. Mov Disord 26:347–50. doi:10.1002/mds.23409

    Article  PubMed  Google Scholar 

  23. Geiner S, Horn AK, Wadia NH, Sakai H, Büttner-Ennever JA (2008) The neuroanatomical basis of slow saccades in spinocerebellar ataxia type 2 (Wadia-subtype). Prog Brain Res 171:575–81. doi:10.1016/S0079-6123(08)00683-3

    Article  PubMed  CAS  Google Scholar 

  24. Rosa AL, Molina I, Kowaljow V, Conde CB (2006) Brisk deep-tendon reflexes as a distinctive phenotype in an Argentinean spinocerebellar ataxia type 2 pedigree. Mov Disord 21:66–68. doi:10.1002/mds.20636

    Article  PubMed  Google Scholar 

  25. Furtado S, Payami H, Lockhart PJ, Hanson M, Nutt JG, Singleton AA et al (2004) Profile of families with parkinsonism predominant spinocerebellar ataxia type 2 (SCA2). Mov Disord 19:622–9. doi:10.1002/mds.20074

    Article  PubMed  Google Scholar 

  26. Simon-Sánchez J, Hanson M, Singleton A, Hernández D, McInerney A, Nussbaum R et al (2005) Analysis of SCA-2 and SCA-3 repeats in parkinsonism: evidence of SCA-2 expansion in a family with autosomal dominant Parkinson’s disease. Neurosci Lett 382:191–4. doi:10.1016/j.neulet.2005.03.015

    Article  PubMed  CAS  Google Scholar 

  27. Velázquez-Pérez L, Fernández-Ruiz J, Díaz R, González RP, Ochoa NC, Cruz GS et al (2006) Spinocerebellar ataxia type 2 olfactory impairment shows a pattern similar to other major neurodegenerative diseases. J Neurol 253:1165–9. doi:10.1007/s00415-006-0183-2

    Article  PubMed  Google Scholar 

  28. Reynaldo-Armiñán RD, Reynaldo-Hernández R, Paneque-Herrera M, Prieto-Ávila L, Pérez-Ruiz E (2002) Mental disorders in patients with spinocerebellar ataxia type 2 in Cuba. Rev Neurol 35:818–21

    PubMed  Google Scholar 

  29. Fernández-Ruiz J, Velásquez-Pérez L, Díaz R, Drucker-Colín R, Pérez-González R, Canales N (2007) Prism adaptation in spinocerebellar ataxia type 2. Neuropsychologia 45:2692–8

    Article  PubMed  Google Scholar 

  30. Babovic-Vuksanovic D, Snow K, Patterson MC, Michels W (1998) Spinocerebellar ataxia type 2 (SCA2) in an infant with extreme CAG repeat expansion. Am J Med Genet 79:383–7. doi:10.1002/(SICI)1096-8628(19981012)7

    Article  PubMed  CAS  Google Scholar 

  31. Rufa A, Dotti MT, Gali L, Orrico A, Sicurelli F, Federico A (2002) Spinocerebellar ataxia type 2 (SCA2) associated with retinal pigmentary degeneration. Eur Neurol 47:128–9. doi:10.1159/000047968

    Article  PubMed  Google Scholar 

  32. Mao R, Aylsworth AS, Potter N, Wilson WG, Breningstall G, Wick MJ et al (2002) Childhood-onset ataxia: testing for large CAG-repeats in SCA2 and SCA7. Am J Med Genet 110:338–45. doi:10.1002/ajmg.10467

    Article  PubMed  Google Scholar 

  33. Moretti P, Blazo M, Garcia L, Amstrong D, Lewis RA, Roa B et al (2004) Spinocerebellar ataxia type 2 (SCA2) presenting with ophthalmoplegia and developmental delay in infancy. Am J Genet A 124A:392–6. doi:10.1002/ajmg.a.20428

    Article  Google Scholar 

  34. Parciorkowski AR, Shafrir Y, Hrivnak J, Patterson MC, Tennison MB, Clark HB et al (2011) Massive expansion of SCA2 with autonomic dysfunction, retinitis pigmentosa, and infantile spasms. Neurology 77:1055–60

    Article  CAS  Google Scholar 

  35. Di Fabio R, Santorelli F, Bertini E, Balestri M, Cursi L, Tessa A et al (2012) Infantile childhood onset of spinocerebellar ataxia type 2. Cerebellum 11:526–30. doi:10.1007/s12311-011-0315-9

    Article  PubMed  Google Scholar 

  36. Ramocki MB, Chapieski L, McDonald RO, Fernandez F, Malphrus AD (2008) Spinocerebellar ataxia type 2 presenting with cognitive regression in childhood. J Child Neurol 23:999–1001. doi:10.1177/0883073808315622

    Article  PubMed  Google Scholar 

  37. Abdel-Aleem A, Zaki MS (2008) Spinocerebellar ataxia type 2 in an Egyptian family presenting with polyphagia and marked CAG expansion in infancy. J Neurol 255:413–9. doi:10.1007/s00415-008-0690-4

    Article  PubMed  CAS  Google Scholar 

  38. Estrada R, Galarraga J, Orozco G, Nodarse A, Auburger G (1999) Spinocerebellar ataxia 2 (SCA2): morphometric analyses in 11 autopsies. Acta Neuropathol (Berl) 97:306–10. doi:10.1007/s004010050989

    Article  CAS  Google Scholar 

  39. Gierga K, Bürk K, Bauer M, Orozco Diaz G, Auburger G, Schultz C et al (2005) Involvement of the cranial nerves and their nuclei in spinocerebellar ataxia type 2 (SCA2). Acta Neuropathol 109:617–31. doi:10.1007/s00401-005-1014-8

    Article  PubMed  CAS  Google Scholar 

  40. Hoche F, Seidel K, Brunt ER, Auburger G, Schols L, Burk K et al (2008) Involvement of the auditory brainstem system in spinocerebellar ataxia type 2 (SCA2), type 3 (SCA3) and type 7 (SCA7). Neuropathol Appl Neurobiol 34:479–91. doi:10.1111/j.1365-2990.2007.00933.x

    Article  PubMed  CAS  Google Scholar 

  41. Koyano S, Iwabuchi K, Yagishita S, Kuroiwa Y, Uchihara T (2002) Paradoxical absence of nuclear inclusion in cerebellar purkinje cells of hereditary ataxias linked to CAG expansion. J Neuro Neurosurg Psychiatry 73:450–2. doi:10.1136/jnnp.73.4.450

    Article  CAS  Google Scholar 

  42. Pang JT, Giunti P, Chamberlain S, An SF, Vitaliani R, Scaravilli T et al (2002) Neuronal intranuclear inclusions in SCA2: a genetic, morphological and immunohistochemical study of two cases. Brain 125:656–63. doi:10.1093/brain/awf060

    Article  PubMed  Google Scholar 

  43. Rub U, Del Turco D, Burk K, Díaz GO, Auburger G, Mittelbronn M et al (2005) Extended pathoanatomical studies point to a consistent affection of the thalamus in spinocerebellar ataxia type 2. Neuropathol Appl Neurobiol 31:127–40. doi:10.1111/j.1365-2990.2004.00617.x

    Article  PubMed  CAS  Google Scholar 

  44. Rub U, Seidel K, Ozerden I, Gierga K, Brunt ER, Schols L et al (2007) Consistent affection of the central somatosensory system in spinocerebellar ataxia type 2 and type 3 and its significance for clinical symptoms and rehabilitative therapy. Brain Res Rev 53:235–49. doi:10.1016/j.brainresrev.2006.08.003

    Article  PubMed  CAS  Google Scholar 

  45. Huynh DP, Del Bigio MR, Ho DH, Pulst SM (1999) Expression of ataxin-2 in brains from normal individuals and patients with Alzheimer’s disease and spinocerebellar ataxia 2. Ann Neurol 45:232–41

    Article  PubMed  CAS  Google Scholar 

  46. Rub U, Schultz C, Del Tredici K, Gierga K, Reifenberger G, de Vos RA et al (2003) Anatomically based guidelines for systematic investigation of the central somatosensory system and their application to a spinocerebellar ataxia type 2 (SCA2) patient. Neuropathol Appl Neurobiol 29:418–33. doi:10.1046/j.1365-2990.2003.00504.x

    Article  PubMed  CAS  Google Scholar 

  47. Ying SH, Choi SI, Lee M, Perlman SL, Baloh RW, Toga AW et al (2005) Relative atrophy of the flocculus and ocular motor dysfunction in SCA2 and SCA6. Ann N Y Acad Sci 1039:430–5. doi:10.1196/annals.1325.040

    Article  PubMed  CAS  Google Scholar 

  48. Imbert G, Saudou F, Yvert G, Devys D, Trottier Y, Garnier JM et al (1996) Cloning of the gene for spinocerebellar ataxia 2 reveals a locus with high sensitivity to expanded CAG/glutamine repeats. Nat Genet 14:285–91. doi:10.1038/ng1196-285

    Article  PubMed  CAS  Google Scholar 

  49. Pulst MS, Nechiporuk A, Nechiporuk T, Gispert S, Chen XN, Lopes-Cendes I et al (1996) Moderate expansion of a normally biallelic trinucleotide repeat in spinocerebellar ataxia type 2. Nat Genet 14:269–76. doi:10.1038/ng1196-269

    Article  PubMed  CAS  Google Scholar 

  50. Sanpei K, Takano H, Igarashi S, Sato T, Oyake M, Sasaki H et al (1996) Identification of the spinocerebellar ataxia type 2 gene using a direct identification of repeat expansion and cloning technique, DIRECT. Nat Genet 14:277–84. doi:10.1038/ng1196-277

    Article  PubMed  CAS  Google Scholar 

  51. Tan EK, Ashisawa T (2001) Genetic testing in spinocerebellar ataxias: defining a clinical role. Arch Neurol 58:191–5. doi:10.1001/pubs.ArchNeurol.-ISSN-0003-9942-58-2-nnr00011

    Article  PubMed  CAS  Google Scholar 

  52. Sahba S, Nechiporuk A, Figueroa KP, Nechiporuk T, Pulst SM (1998) Genomic structure of the human gene for spinocerebellar ataxia 2 (SCA2) on chromosome 12q24.1. Genomics 47:359–64. doi:10.1006/geno.1997.5131

    Article  PubMed  CAS  Google Scholar 

  53. Mao R, Aylsworth AS, Potter N, Wilson WG, Breningstall G, Wick MJ et al (2002) Childhood-onset ataxia: testing for large CAG-repeats in SCA2 and SCA7. Am J Med Genet 110:338–45. doi:10.1002/ajmg.10467

    Article  PubMed  Google Scholar 

  54. Andrés AM, Lao O, Soldevila M, Calafell F, Bertranpetit J (2002) Dynamics of CAG repeat loci revealed by the analysis of their variability. Hum Mutat 21:61–70. doi:10.1002/humu.10151

    Article  CAS  Google Scholar 

  55. Magaña JJ, Leyva-García N, Cisneros B (2009) Pathogenesis of myotonic dystrophy type 1. Gac Med Mex 145:331–7

    PubMed  Google Scholar 

  56. Fernández M, McClain ME, Martínez RA, Snow K, Lipe H, Ravits J et al (2000) Late-onset SCA2:33 CAG repeats are sufficient to cause disease. Neurology 55:569–72

    Article  PubMed  Google Scholar 

  57. Dorschner MO, Barden D, Stephens K (2002) Diagnosis of five spinocerebellar ataxia disorders by multiplex amplification and capillary electrophoresis. J Mol Diagn 4:108–13. doi:10.1016/S1525-1578(10)60689-7

    Article  PubMed  CAS  Google Scholar 

  58. Giunti P, Sabbadini G, Sweeney MG, Davis MB, Veneziano L, Mantuano E et al (1998) The role of SCA2 trinucleotide repeat expansion in 89 autosomal dominant cerebellar ataxia families: frequency, clinical and genetics correlates. Brain 121:459–67. doi:10.1093/brain/121.3.459

    Article  PubMed  Google Scholar 

  59. Pulst SM, Santos N, Wang D, Yang H, Huynh D, Velázquez L et al (2005) Spinocerebellar ataxia type 2: polyQ repeat variation in the CACNA1A calcium channel modifies age of onset. Brain 128:2297–303. doi:10.1093/brain/awh586

    Article  PubMed  Google Scholar 

  60. Simon DK, Zheng K, Velázquez L, Figueroa KP, Falcón N, Almaguer LE, Pulst SM (2007) Mitochondrial complex I gene variant associated with early age of onset in SCA2. Arch Neurol 64:1042–4. doi:10.1001/archneur.64.7.1042

    Article  PubMed  Google Scholar 

  61. Hayes S, Turecki G, Brisebois K, Lopes-Cendes I, Gaspar C, Riess O et al (2000) CAG repeat length in RAI1 is associated with age at onset variability in spinocerebellar ataxia type 2 (SCA2). Hum Mol Genet 9:1753–8. doi:10.1093/hmg/9.12.1753

    Article  PubMed  CAS  Google Scholar 

  62. Van Damme P, Veldink JH, van Blitterswijk M, Corveleyn A, van Vught PW, Thijs V et al (2011) Expanded ATXN2 CAG repeat size in ALS identifies genetic overlap between ALS and SCA2. Neurology 76:2066–2072

    Article  PubMed  CAS  Google Scholar 

  63. Lee T, Li YR, Ingre C, Weber M, Grehl T, Gredal O et al (2011) Ataxin-2 intermediate-length polyglutamine expansions in European ALS patients. Hum Mol Genet 20:1697–1700. doi:10.1093/hmg/ddr045

    Article  PubMed  CAS  Google Scholar 

  64. Yu Z, Zhu Y, Chen-Plotkin AS, Clay-Falcone D, McCluskey L, Elman L et al (2011) PolyQ repeat expansions in ATXN2 associated with ALS are CAA interrupted repeats. PLoS One 6:e17951. doi:10.1371/journal.pone.0017951

    Article  PubMed  CAS  Google Scholar 

  65. Elden AC, Kim HJ, Hart MP, Chen-Plotkin AS, Johnson BS, Fang X et al (2010) Ataxin-2 intermediate-length polyglutamine expansions are associated with increased risk for ALS. Nature 466:1069–1075. doi:10.1038/nature09320

    Article  PubMed  CAS  Google Scholar 

  66. Chen Y, Huang R, Tang Y, Chen K, Song W, Pan P et al (2011) Ataxin-2 intermediate-length polyglutamine: a possible risk factor for Chinese patients with amyotrophic lateral sclerosis. Neurobiol Aging 32:1925.e1–1925.e5. doi:10.1016/j.neurobiolaging.2011.05.015

    Article  CAS  Google Scholar 

  67. Daoud H, Belzil V, Martins S, Sabbagh M, Provencher P, Lacomblez L et al (2011) Association of long ATXN2 CAG repeat sizes with increased risk of amyotrophic lateral sclerosis. Arch Neurol 68:739–742. doi:10.1001/archneurol.2011.111

    Article  PubMed  Google Scholar 

  68. Gispert S, Kurz A, Waibel S, Bauer P, Liepelt I, Geisen C et al (2012) The modulation of amyotrophic lateral sclerosis risk by ataxin-2 intermediate polyglutamine expansions is a specific effect. Neurobiol Dis 45:356–361. doi:10.1016/j.nbd.2011.08.021

    Article  PubMed  CAS  Google Scholar 

  69. Charles P, Camuzat A, Benammar N, Sellal F, Destée A, Bonnet AM et al (2007) French Parkinson’s Disease Genetic Study Group. Are interrupted SCA2 CAG repeat expansions responsible for parkinsonism? Neurology 69:1970–1975

    Article  PubMed  CAS  Google Scholar 

  70. Albrecht M, Golatta M, Wullner U, Lengauer T (2004) Structural and functional analysis of ataxin-2 and ataxin-3. Eur J Biochem 271:3155–70. doi:10.1111/j.1432-1033.2004.04245.x

    Article  PubMed  CAS  Google Scholar 

  71. van de Loo S, Eich F, Nonis D, Auburger G, Nowock J (2009) Ataxin-2 associates with rough endoplasmic reticulum. Exp Neurol 215:110–8. doi:10.1016/j.expneurol.2008.09.020

    Article  PubMed  CAS  Google Scholar 

  72. Neuwald AF, Koonin EV (1998) Ataxin-2, global regulators of bacterial gene expression, and spliceosomal snRNP proteins share a conserved domain. J Mol Med 76:3–5. doi:10.1007/s001090050184

    Article  PubMed  CAS  Google Scholar 

  73. Satterfield TF, Pallanck LJ (2006) Ataxin-2 and its Drosophila homolog, ATX2, physically assemble with polyribosomes. Hum Mol Genet 15:2523–32. doi:10.1093/hmg/ddl173

    Article  PubMed  CAS  Google Scholar 

  74. Lastres-Becker I, Rüb U, Auburger G (2008) Spinocerebellar ataxia 2 (SCA2). Cerebellum 7:115–124. doi:10.1007/s12311-008-0019-y

    Article  PubMed  CAS  Google Scholar 

  75. Meunier C, Bordereaux D, Porteu F, Gisselbrecht S, Chretien S, Courtois G (2002) Cloning and characterization of a family of proteins associated with Mp1. J Biol Chem 277:9139–47. doi:10.1074/jbc.M105970200

    Article  PubMed  CAS  Google Scholar 

  76. Tharun S (2009) Roles of eukaryotic Lsm proteins in the regulation of mRNA function. Int Rev Cell Mol Biol 272:149–89. doi:10.1016/S1937-6448(08)01604-3

    Article  PubMed  CAS  Google Scholar 

  77. Ralser M, Albrecht M, Nonhoff U, Lengauer T, Lehrach H, Krobitsch S (2005) An integrative approach to gain insights into the cellular function of human ataxin-2. J Mol Biol 346:203–14. doi:10.1016/j.jmb.2004.11.024

    Article  PubMed  CAS  Google Scholar 

  78. Bravo J, Aguilar-Henonin L, Olmedo G, Guzmán P (2005) Four distinct classes of proteins as interaction partners of the PABC domain of Arabidopsis thaliana Poly(A)-binding proteins. Mol Genet Genomics 272:651–65. doi:10.1007/s00438-004-1090-9

    Article  PubMed  CAS  Google Scholar 

  79. Shibata H, Huynh DP, Pulst SM (2000) A novel protein with RNA-binding motifs interacts with ataxin-2. Hum Mol Genet 9:1303–13. doi:10.1093/hmg/9.9.1303

    Article  PubMed  CAS  Google Scholar 

  80. Lee JA, Tang ZZ, Black DL (2009) An inducible change in Fox-1/A2BP1 splicing modulates the alternative splicing of downstream neuronal target exons. Genes Dev 23:2284–93. doi:10.1101/gad.1837009

    Article  PubMed  CAS  Google Scholar 

  81. Tsien JZ, Huerta PT, Tonegawa S (1996) The essential role of hippocampal CA1 NMDA receptor-dependent synaptic plasticity in spatial memory. Cell 87:1327–38. doi:10.1016/S0092-8674(00)81827-9

    Article  PubMed  CAS  Google Scholar 

  82. Lim J, Hao T, Shaw C, Patel AJ, Szabo G, Rual JF et al (2006) A protein-protein interaction network for human inherited ataxias and disorders of Purkinje cell degeneration. Cell 125:801–14

    Article  PubMed  CAS  Google Scholar 

  83. Villace P, Marion RM, Ortin J (2004) The composition of Staufen-containing RNA granules from human cells indicates their role in the regulated transport and translation of messenger RNAs. Nucleic Acids Res 32:2411–20. doi:10.1093/nar/jkh552

    Article  PubMed  CAS  Google Scholar 

  84. Kozlov G, Safaee N, Rosenauer A, Gehring K (2010) Structural basis of binding of P-body-associated proteins GW182 and ataxin-2 by the MLLE domain of poly(A)-binding protein. J Biol Chem 285:13599–606. doi:10.1074/jbc.M109.089540

    Article  PubMed  CAS  Google Scholar 

  85. Ciosk R, DePalma M, Priess JR (2004) ATX-2, the C. elegans ortholog of ataxin 2, functions in translational regulation in the germline. Development 131:4831–41. doi:10.1242/dev.01352

    Article  PubMed  CAS  Google Scholar 

  86. Kedersha N, Anderson P (2002) Stress granules: sites of mRNA triage that regulate mRNA stability and translatability. Biochem Soc Trans 30:963–9

    Article  PubMed  CAS  Google Scholar 

  87. Vernet C, Artzt K (1997) STAR, a gene family involved in signal transduction and activation of RNA. Trends Genet 13:479–84. doi:10.1016/S0168-9525(97)01269-9

    Article  PubMed  CAS  Google Scholar 

  88. Swisher KD, Parker R (2010) Localization to, and effects of Pbp1, Pbp4, Lsm12, Dhh1, and Pab1 on stress granules in Saccharomyces cerevisiae. PLoS One 5:e10006. doi:10.1371/journal.pone.0010006

    Article  PubMed  CAS  Google Scholar 

  89. Ralser M, Nonhoff U, Albrecht M, Lengauer T, Wanker EE, Lehrach H et al (2005) Ataxin-2 and huntingtin interact with endophilin-A complexes to function in plastin-associated pathways. Hum Mol Genet 14:2893–909. doi:10.1093/hmg/ddi321

    Article  PubMed  CAS  Google Scholar 

  90. Huynh DP, Scoles DR, Nguyen D, Pulst SM (2003) The autosomal recessive juvenile Parkinson disease gene product, parkin, interacts with and ubiquitinates synaptotagmin XI. Hum Mol Genet 12:2587–97. doi:10.1093/hmg/ddg269

    Article  PubMed  CAS  Google Scholar 

  91. Imai Y, Soda M, Inoue H, Hattori N, Mizuno Y, Takahashi R (2001) An unfolded putative transmembrane polypeptide, which can lead to endoplasmic reticulum stress, is a substrate of parkin. Cell 105:891–902. doi:10.1016/S0092-8674(01)00407-X

    Article  PubMed  CAS  Google Scholar 

  92. Soubeyran P, Kowanetz K, Szymkiewicz I, Langdon WY, Dikic I (2002) Cbl-CIN85-endophilin complex mediates ligand induced downregulation of EGF receptors. Nature 416:183–7. doi:10.1038/416183a

    Article  PubMed  CAS  Google Scholar 

  93. Satterfield TF, Jackson SM, Pallanck LJ (2002) A Drosophila homolog of the polyglutamine disease gene SCA2 is a dosage-sensitive regulator of actin filament formation. Genetics 162:1687–702

    PubMed  CAS  Google Scholar 

  94. Huynh DP, Figueroa K, Hoang N, Pulst SM (2000) Nuclear localization or inclusion body formation of ataxin-2 are not necessary for SCA2 pathogenesis in mouse or human. Nat Genet 26:44–50. doi:10.1038/79162

    Article  PubMed  CAS  Google Scholar 

  95. Matilla-Dueñas A, Sánchez I, Corral-Juan M, Dávalos A, Alvarez R, Latorre P (2010) Cellular and molecular pathways triggering neurodegeneration in the spinocerebellar ataxias. Cerebellum 9(2):148–66. doi:10.1007/s12311-009-0144-2

    Article  PubMed  CAS  Google Scholar 

  96. Davies SW, Turmaine M, Cozens BA, DiFiglia M, Sharp AH, Ross CA et al (1997) Formation of neuronal intranuclear inclusions underlies the neurological dysfunction in mice transgenic for the HD mutation. Cell 90:537–548. doi:10.1016/S0092-8674(00)80513-9

    Article  PubMed  CAS  Google Scholar 

  97. DiFiglia M, Sapp E, Chase KO, Davies SW, Bates GP, Vonsattel JP et al (1997) Aggregation of huntingtin in neuronal intranuclear inclusions and dystrophic neurites in brain. Science 277:1990–1993

    Article  PubMed  CAS  Google Scholar 

  98. Huynh DP, Yang HT, Vakharia H, Nguyen D, Pulst SM (2003) Expansion of the polyQ repeat in ataxin-2 alters its Golgi localization, disrupt the Golgi complex and causes cell death. Hum Mol Genet 12:1485–1496. doi:10.1093/hmg/ddg175

    Article  PubMed  CAS  Google Scholar 

  99. Koyano S, Uchihara T, Fujigasaki H, Nakamura A, Yagishita S, Iwabuchi K (1999) Neuronal intranuclear inclusions in spinocerebellar ataxia type 2: triple-labeling immunofluorescent study. Neurosci Lett 57:805–810. doi:10.1016/S0304-3940(99)00656-4

    CAS  Google Scholar 

  100. Uchihara T, Fujigasaki H, Koyano S, Nakamura A, Yagishita S, Iwabuchi K (2001) Non-expanded polyglutamine proteins in intranuclear inclusions of hereditary ataxias-triple-labeling immunofluorescence study. Acta Neuropathol 102:149–152. doi:10.1007/s004010100364

    PubMed  CAS  Google Scholar 

  101. Saudou F, Finkbeiner S, Devys D, Greenberg ME (1998) Huntingtin acts in the nucleus to induce apoptosis but death does not correlate with the formation of intranuclear inclusions. Cell 95:55–66. doi:10.1016/S0092-8674(00)81782-1

    Article  PubMed  CAS  Google Scholar 

  102. Lunkes A, Mandel JL (1998) A cellular model that recapitulates major pathogenic steps of Hungtington’s disease. Hum Mol Genet 7:1355–1361. doi:10.1093/hmg/7.9.1355

    Article  PubMed  CAS  Google Scholar 

  103. Lunkes A, Trottier Y, Fagart J, Schultz P, Zeder-Lutz G, Moras D et al (1999) Properties of polyglutamine expansion in vitro and in a cellular model for Huntington’s disease. Philos Trans R Soc Lond B Biol Sci 354:1013–1019

    Article  PubMed  CAS  Google Scholar 

  104. Mangiarini L, Sathasivam K, Seller M, Cozens B, Harper A, Hetherington C et al (1996) Exon 1 of HD gene with an expanded CAG repeat is sufficient to cause a progressive neurological phenotype in transgenic mice. Cell 87:493–506. doi:10.1016/S0092-8674(00)81369-0

    Article  PubMed  CAS  Google Scholar 

  105. Aronin N, Kim M, Laforet G, DiFiglia M (1999) Are there multiple pathways in the pathogenesis of Huntington’s disease? Philos Trans R Soc Lond B Biol Sci 354:995–1003

    Article  PubMed  CAS  Google Scholar 

  106. Reddy PH, Charles V, Williams M, Miller G, Whetsell WO Jr, Tagle DA (1999) Transgenic mice expressing mutated full-length HD cDNA: a paradigm for locomotor changes and selective neuronal loss in Huntington’s disease. Philos Trans R Soc Lond B Biol Sci 354:1035–1045

    Article  PubMed  CAS  Google Scholar 

  107. Tarlac V, Turnbull V, Stefani D, Kelly L, Walsh R, Storey E (2007) Inclusion formation by ataxins-1,-2,-3, and-7. Int J Neurosci 117:1289–1314. doi:10.1080/00207450600936668

    Article  PubMed  CAS  Google Scholar 

  108. Aguiar J, Fernández J, Aguilar A, Mendoza Y, Vázquez M, Suárez J et al (2006) Ubiquitous expression of human SCA2 gene under the regulation of the SCA2 self promoter causes specific Purkinje cell degeneration in transgenic mice. Neurosci Lett 392:202–6. doi:10.1016/j.neulet.2005.09.020

    Article  PubMed  CAS  Google Scholar 

  109. Turnbull VJ, Storey E, Tarlac V, Walsh R, Stefani D, Clark R et al (2004) Different ataxin-2 antibodies display different immunoreactive profiles. Brain Res 1027:103–116. doi:10.1016/j.brainres.2004.08.044

    Article  PubMed  CAS  Google Scholar 

  110. Paulson HL, Perez MK, Trottier Y, Trojanowski JQ, Subramony SH, Das SS et al (1997) Intranuclear inclusions of expanded polyglutamine protein in spinocerebellar ataxia type 3. Neuron 93:939–949. doi:10.1016/S0896-6273(00)80943-5

    Google Scholar 

  111. Ikeda H, Yamaguchi M, Sugai S, Aze Y, Narumiya S, Kakizuka A (1996) Expanded polyglutamine in the Machado-Joseph disease protein induce cell death in vitro and in vivo. Nat Genet 13:196–202. doi:10.1038/ng0696-196

    Article  PubMed  CAS  Google Scholar 

  112. Merry DE, Kobayashi Y, Bailey CK, Taye AA, Fischbeck KH (1998) Cleavage, aggregation and toxicity of the expanded androgen receptor in spinal and bulbar muscular atrophy. Hum Mol Genet 7:693–701. doi:10.1093/hmg/7.4.693

    Article  PubMed  CAS  Google Scholar 

  113. Wellington CL, Ellerby LM, Hackam AS, Margolis RL, Trifiro MA, Singaraja R et al (1998) Caspase cleavage of gene products associated with triplet expansion disorders generates truncated fragments containing the polyglutamine tract. J Biol Chem 273:9158–9167. doi:10.1074/jbc.273.15.9158

    Article  PubMed  CAS  Google Scholar 

  114. Graham RK, Deng Y, Slow EJ, haigh B, Bissada N, Lu G et al (2006) Cleavage at the caspase-6 site is required for neuronal dysfunction and degeneration due to mutant huntingtin. Cell 125:1179–1191. doi:10.1016/j.cell.2006.04.026

    Article  PubMed  CAS  Google Scholar 

  115. Goti D, Katzen SM, Mez J, Kurtis N, Kiluk J, Ben-Haïem L et al (2004) A mutant ataxin-3 putative-cleavage fragment in brains of Machado-Joseph disease patients and transgenic mice is cytotoxic above a critical concentration. J Neurosci 24:10266–79. doi:10.1523/JNEUROSCI.2734-04.2004

    Article  PubMed  CAS  Google Scholar 

  116. Colomer Gould VF, Goti D, Pearce D, Gonzalez GA, Gao H, Bermudez de Leon M et al (2007) A mutant ataxin-3 fragment results from processing at a site N-terminal to amino acid 190 in brain of Machado-Joseph disease like transgenic mice. Neurobiol Dis 27:362–369. doi:10.1016/j.nbd.2007.06.005

    Article  PubMed  CAS  Google Scholar 

  117. Berke SJ, Schmied FA, Brunt ER, Ellerby LM, Paulson HL (2004) Caspase-mediated proteolysis of polyglutamine disease protein ataxin-3. J Neurochem 89:908–918. doi:10.1111/j.1471-4159.2004.02369.x

    Article  PubMed  CAS  Google Scholar 

  118. Warby SC, Doty CN, Graham RK, Shively J, Singaraja RR, Hayden MR (2009) Phosphorylation of huntingtin reduces the accumulation of its nuclear fragments. Mol Cell Neurosci 40:121–127. doi:10.1016/j.mcn.2008.09.007

    Article  PubMed  CAS  Google Scholar 

  119. Humbert S, Bryson EA, Cordelieres FP, Connors NC, Datta SR, Finkbeiner S et al (2002) The IGF-1/Akt pathway is neuroprotective in Huntington’s disease and involves Huntingtin phosphorylation by Akt. Dev Cell 2:831–837. doi:10.1016/S1534-5807(02)00188-0

    Article  PubMed  CAS  Google Scholar 

  120. Rangone H, Poizat G, Troncoso J, Ross CA, McDonald ME, Saudou F et al (2004) The serum-and glucocorticoid-induced kinase SGK inhibits mutant huntingtin-induced toxicity by phosphorylating serine 421 of huntingtin. Eur J Neurosci 19:273–279. doi:10.1111/j.0953-816X.2003.03131.x

    Article  PubMed  Google Scholar 

  121. Schilling B, Gafni J, Torcassi C, Cong X, Row RH, LaFevre-Bernt MA et al (2006) Huntingtin phosphorylation sites mapped by mass spectrometry. Modulation of cleavage and toxicity. J Biol Chem 281:23686–23697. doi:10.1074/jbc.M513507200

    Article  PubMed  CAS  Google Scholar 

  122. Gafni J, Hermel E, Young JE, Wellington CL, Hayden MR, Ellerby LM (2004) Inhibition of calpain cleavage of huntingtin reduces toxicity: accumulation of calpain/caspase fragments in the nucleus. J Biol Chem 279:20211–20220. doi:10.1074/jbc.M401267200

    Article  PubMed  CAS  Google Scholar 

  123. Fei E, Jia N, Zhang T, Ma X, Wang H, Liu C et al (2007) Phosphorylation of ataxin-3 by glycogen synthase kinase 3beta at serine 256 regulates the aggregation of ataxin-3. Biochem Biophys Res Commun 357:487–492. doi:10.1016/j.bbrc.2007.03.160

    Article  PubMed  CAS  Google Scholar 

  124. Mueller T, Breuer P, Schmitt I, Walter J, Evert BO, Wüllner U (2009) CK2-dependent phosphorylation determines cellular localization and stability of ataxin-3. Hum Mol Genet 18:121–136. doi:10.1093/hmg/ddp274

    Article  CAS  Google Scholar 

  125. Muchowski PJ, Wacker JL (2005) Modulation of neurodegeneration by molecular chaperones. Nat Rev Neurosci 6:11–22. doi:10.1038/nrn1587

    Article  PubMed  CAS  Google Scholar 

  126. Sherman MY, Goldberg AL (2001) Cellular defenses against unfolded proteins: a cell biologist thinks about neurodegenerative diseases. Neuron 29:15–32. doi:10.1016/S0896-6273(01)00177-5

    Article  PubMed  CAS  Google Scholar 

  127. Pirker W, Back C, Gerschlager W, Laccone F, Alesch F (2003) Chronic thalamic stimulation in a patient with spinocerebellar ataxia type 2. Mov Disord 18:222–5. doi:10.1002/mds.10192

    Article  PubMed  Google Scholar 

  128. Freund HJ, Barnikol UB, Nolte D, Treuer H, Auburger G, Tass PA et al (2007) Subthalamic-thalamic DBS in a case with spinocerebellar ataxia type 2 and severe tremor-A unusual clinical benefit. Mov Disord 22:732–5. doi:10.1002/mds.21338

    Article  PubMed  Google Scholar 

  129. Ristori G, Romano S, Visconti A, Cannoni S, Spadaro M, Frontali M et al (2010) Riluzole in cerebellar ataxia: a randomized, double-blind, placebo-controlled pilot trial (CME) (LOE Classification). Neurology 74:839–45

    Article  PubMed  CAS  Google Scholar 

  130. Velázquez-Pérez L, Rodríguez-Chanfrau J, García-Rodríguez JC, Sánchez-Cruz G, Aguilera-Rodríguez R, Rodríguez-Labrada R et al (2011) Oral zinc sulphate supplementation for six months in SCA2 patients: a randomized, double-blind, placebo-controlled trial. Neurochem Res 36(10):1793–800. doi:10.1007/s11064-011-0496-0

    Article  PubMed  CAS  Google Scholar 

  131. Xia H, Mao Q, Eliason SL, Harper SQ, Martins IH, Orr HT et al (2004) RNAi suppresses polyglutamine-induced neurodegeneration in a model of spinocerebellar ataxia. Nat Med 10:816–20

    Article  PubMed  CAS  Google Scholar 

  132. Harper SQ, Staber PD, He X, Eliason SL, Martins IH, Mao Q et al (2005) RNA interference improves motor and neuropathological abnormalities in a Huntington’s disease mouse model. Proc Natl Acad Sci USA 102:5820–5825. doi:10.1073/pnas.0501507102

    Article  PubMed  CAS  Google Scholar 

  133. Rodriguez-Lebron E, Denovan-Wrigth EM, Nash K, Lewin AS, Mandel RJ (2005) Intrastratial rAAV-mediated delivery of anti-huntingtin shRNAs induces partial reversal of disease progression in R6/1 Huntington’s disease transgenic mice. Mol Ther 12:618–633. doi:10.1016/j.ymthe.2005.05.006

    Article  PubMed  CAS  Google Scholar 

  134. Franich NR, Fitzsimons HL, Fong DM, Klugmann M, During MJ, Young D (2008) AAV vector-mediated RNAi of mutant huntingtin expression is neuroprotective in a novel genetic rat model of Huntington’s disease. Mol Ther 16:947–956. doi:10.1038/mt.2008.50

    Article  PubMed  CAS  Google Scholar 

  135. Machida Y, Okada T, Kurosawa M, Oyama F, Ozawa K, Nukina N (2006) rAAV-mediated shRNA ameliorated neuropathology in Huntington disease model mouse. Biochem Biophys Res Commun 343:190–197. doi:10.1016/j.bbrc.2006.02.141

    Article  PubMed  CAS  Google Scholar 

  136. Wang YL, Liu W, Wada E, Murata M, Wada K, Kanazawa I (2005) Clinico-pathological rescue of a model mouse of Huntington’s disease by siRNA. Neurosci Res 53:241–249. doi:10.1016/j.neures.2005.06.021

    Article  PubMed  CAS  Google Scholar 

  137. Caplen NJ, Taylor JP, Statham VS, Tanaka F, Fire A, Morgan RA (2002) Rescue of polyglutamine-mediated cytotoxicity by double-stranded RNA-mediated RNA interference. Hum Mol Genet 11:175–184. doi:10.1093/hmg/11.2.175

    Article  PubMed  CAS  Google Scholar 

  138. Koutsilieri E, Rethwilm A, Scheller C (2007) The therapeutic potential of siRNA in gene therapy of neurodegenerative disorders. J Neural Transm Suppl 72:43–49

    Article  PubMed  CAS  Google Scholar 

  139. Bonini NM, La Spada AR (2005) Silencing polyglutamine degeneration with RNAi. Neuron 48:715–8. doi:10.1016/jneuron.2005.11.008

    Article  PubMed  CAS  Google Scholar 

  140. Miller VM, Xia H, Marrs GL, Gouvion CM, Lee G, Davidson BL et al (2003) Allele-specific silencing of dominant disease genes. Proc Natl Acad Sci USA 100:7195–7200. doi:10.1073/pnas.1231012100

    Article  PubMed  CAS  Google Scholar 

  141. Li Y, Yokota T, Matsumura R, Taira K, Misuzawa H (2004) Sequence dependent and independent inhibition specific for mutant ataxin-3 by small interfering RNA. Ann Neurol 56:124–129. doi:10.1002/ana.20141

    Article  PubMed  CAS  Google Scholar 

  142. Alves S, Nascimento-Ferreira I, Auregan G, Hassig R, Dufour N, Brouillet E et al (2008) Allele-specific RNA silencing of mutant ataxin 3 mediates protection in a rat model of Machado-Joseph disease. PLoS One 3:e3341. doi:10.1371/journal.pone.0003341

    Article  PubMed  CAS  Google Scholar 

  143. Scholefield J, Greenberg LJ, Weinberg MS, Arbuthnot PB, Abdelgany A, Wood MJ (2009) Design of RNAi hairpins for mutation-specific silencing of ataxin-7 and correlation of a SCA7 phenotype. PLoS One 4:e7232. doi:10.1371/journal.pone.0007232

    Article  PubMed  CAS  Google Scholar 

  144. Zhang Y, Engelman J, Friedlander RM (2009) Allele-specific silencing of mutant Huntington’s disease gene. J Neurochem 108:82–90. doi:10.1111/j.1471-4159.2008.05734.x

    Article  PubMed  CAS  Google Scholar 

  145. Gagnon KT, Pendergraff HM, Deleavey GF, Swayze EE, Potier P, Randolph J, Roesch EB, Chattopadhyaya J, Damha MJ, Bennett CF et al (2010) Allele-selective inhibition of mutant huntingtin expression with antisense oligonucleotides targeting the expanded CAG repeat. Biochemistry 49:10166–10178. doi:10.1021/bi101208k

    Article  PubMed  CAS  Google Scholar 

  146. Hu J, Matsui M, Gagnon KT, Schwartz JC, Gabillet S, Arar K, Wu J, Bezprozvanny I, Corey DR (2009) Allele-specific silencing of mutant huntingtin and ataxin-3 genes by targeting expanded CAG repeats in mRNAs. Nat Biotechnol 27:478–484. doi:10.1038/nbt.1539

    Article  PubMed  CAS  Google Scholar 

  147. Kubodera T, Yokota T, Ishikawa K, Mizusawa H (2005) New RNAi strategy for selective suppression of a mutant allele in polyglutamine disease. Oligonucleotides 15:298–302. doi:10.1089/oli.2005.15.298

    Article  PubMed  CAS  Google Scholar 

  148. Davidson BL, Breakefield XO (2003) Viral vectors for gene delivery to the nervous system. Nat Rev Neurosci 4:353–364. doi:10.1038/nrn1104

    Article  PubMed  CAS  Google Scholar 

  149. Tang Y, Cummins J, Huard J, Wang B (2010) AAV-directed muscular dystrophy gene therapy. Expert Opin Biol Ther 10:395–408. doi:10.1517/14712591003604690

    Article  PubMed  CAS  Google Scholar 

  150. Sarkar S, Perlstein EO, Imarisio S, Pineau S, Cordenier A, Maglathlin RL et al (2007) Small molecules enhance autophagy and reduce toxicity in Huntington’s disease models. Nat Chem Biol 3:331–338. doi:10.1038/nchembio883

    Article  PubMed  CAS  Google Scholar 

  151. Williams A, Sarkar S, Cuddon P, Ttofi EK, Saiki S, Siddiqui FH et al (2008) Novel targets for Huntington’s disease in an mTOR independent autophagy pathway. Nat Chem Biol 4:295–305. doi:10.1038/nchembio.79

    Article  PubMed  CAS  Google Scholar 

  152. Wong HK, Bauer PO, Kurosawa M et al (2008) Blocking acid-sensing ion channel 1 alleviates Huntington’s disease pathology via an ubiquitin-proteasome system-dependent mechanism. Hum Mol Genet 17:3223–35. doi:10.1093/hmg/ddn218

    Article  PubMed  CAS  Google Scholar 

  153. Pollitt SK, Pallos J, Shao J, Desai UA, Ma AA, Thompson LM, Marsh JL, Diamond MI (2003) A rapid cellular FRET assay of polyglutamine aggregation identifies a novel inhibitor. Neuron 40:685–94. doi:10.1016/S0896-6273(03)00697-4

    Article  PubMed  CAS  Google Scholar 

  154. Tanaka S, Dan C, Kawano H, Omoto M, Ishihara T (2008) Pathological study amyloidosis in Cygnus olor (mute swan) and other waterfowl. Med Mol Morphol 41:99–108. doi:10.1007/s00795-008-0401-3

    Article  PubMed  Google Scholar 

  155. Sánchez I, Mahlke C, Yuan J (2003) Pivotal role of oligomerization in expanded polyglutamine neurodegenerative disorders. Nature 421:373–379. doi:10.1038/nature01301

    Article  PubMed  CAS  Google Scholar 

  156. Wood NI, Pallier PN, Wanderer J, Morton AJ (2007) Systemic administration of Congo red does not improve motor or cognitive function in R6/2 mice. Neurobiol Dis 25:342–53. doi:10.1016/j.nbd.2006.09.015

    Article  PubMed  CAS  Google Scholar 

  157. Nagai Y, Fujikake N, Ohno K, Higashiyama H, Popiel HA, Rahadian J et al (2003) Prevention of polyglutamine oligomerization and neurodegeneration by the peptide inhibitor QBP1 in Drosophila. Hum Mol Genet 12:1253–1259. doi:10.1093/hmg/ddg144

    Article  PubMed  CAS  Google Scholar 

  158. Popiel HA, Nagai Y, Fujikake N, Toda T. Protein transduction domain-mediated delivery of QBP1 supresses polyglutamine-induced neurodegeneration in vivo. Mol Ther 15:303-309. doi:10.1038/sj.mt.6300045

  159. Karpuj MV, Becher MW, Springer JE, Chabas D, Youssef S, Pedotti R et al (2002) Prolonged survival and decreased abnormal movements in transgenic model of Huntington disease, with administration of the transglutaminase inhibitor cystamine. Nat Med 8:143–9. doi:10.1038/nm0202-143

    Article  PubMed  CAS  Google Scholar 

  160. Dedeoglu A, Kubilus JK, Jeitner TM et al (2002) Therapeutic effects of cystamine in a murine model of Huntington’s disease. J Neurosci 22:8942–50

    PubMed  CAS  Google Scholar 

  161. Igarashi S, Koide R, Shimohata T, Yamada M, Hayashi Y, Takano H et al (1998) Suppression of aggregate formation and apoptosis by tansglutaminase inhibitors in cells expressing truncated DRPLA protein with an expanded polyglutamine stretch. Nat Genet 18:111–7. doi:10.1038/ng0298-111

    Article  PubMed  CAS  Google Scholar 

  162. Friedman MJ, Wang CE, Li XJ, Li S (2008) Polyglutamine expansion reduces the association of TATA-binding protein with DNA and induces DNA binding-independent neurotoxicity. J Biol Chem 283:8283–8290. doi:10.1074/jbc.M709674200

    Article  PubMed  CAS  Google Scholar 

  163. Kim SJ, Kim TS, Hong S, Rhim H, Kim IY, Kang S (2003) Oxidative stimuli affect polyglutamine aggregation and cell death in human mutant ataxin-1 expressing cells. Neurosci Lett 348:21–24. doi:10.1016/S0304-3940(03)00657-8

    Article  PubMed  CAS  Google Scholar 

  164. Ferrante RJ, Andreassen OA, Dedeoglu A, Ferrante KL, Jenkins BG, Hersch SM et al (2002) Therapeutic effects of coenzyme Q10 and remacemide in transgenic mouse models of Huntington’s disease. J Neurosci 22:1592–9

    PubMed  CAS  Google Scholar 

  165. Pérez-Ávila I, Fernández-Vieitez JA, Martínez-Góngora E, Ochoa-Mastrapa R, Velázquez-Manresa MG (2004) Effects of a physical training program on quantitative neurological indices in mild stage type 2 spinocerebellar ataxia patients. Rev Neurol 39:907–10

    PubMed  Google Scholar 

Download references

Acknowledgments

This work was supported by CONACyT-México, Grant No. 128418 (B.C.).

Conflict of interest

The authors declare that there are no conflicts of interest.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to B. Cisneros.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Magaña, J.J., Velázquez-Pérez, L. & Cisneros, B. Spinocerebellar Ataxia Type 2: Clinical Presentation, Molecular Mechanisms, and Therapeutic Perspectives. Mol Neurobiol 47, 90–104 (2013). https://doi.org/10.1007/s12035-012-8348-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12035-012-8348-8

Keywords

Navigation