Skip to main content

Molecular Mechanisms and Cellular Pathways Implicated in Machado-Joseph Disease Pathogenesis

  • Chapter
  • First Online:
Polyglutamine Disorders

Abstract

Machado-Joseph disease (MJD) is a dominantly inherited disorder originally described in people of Portuguese descent, and associated with the expansion of a CAG tract in the coding region of the causative gene MJD1/ATX3. The CAG repeats range from 10 to 51 in the normal population and from 55 to 87 in SCA3/MJD patients. MJD1 encodes ataxin-3, a protein whose physiological function has been linked to ubiquitin-mediated proteolysis. Despite the identification of the causative mutation, the pathogenic process leading to the neurodegeneration observed in the disease is not yet completely understood. In the past years, several studies identified different molecular mechanisms and cellular pathways as being impaired or deregulated in MJD. Autophagy, proteolysis or post-translational modifications, among other processes, were implicated in MJD pathogenesis. From these studies it was possible to identify new targets for therapeutic intervention, which in some cases proved successful in models of disease.

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

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 109.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 139.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 199.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Paulson HL, Das SS, Crino PB, Perez MK, Patel SC, Gotsdiner D, Fischbeck KH, Pittman RN (1997) Machado-Joseph disease gene product is a cytoplasmic protein widely expressed in brain. Ann Neurol 41:453–462

    Article  CAS  PubMed  Google Scholar 

  2. Schmidt T et al (1998) An isoform of ataxin-3 accumulates in the nucleus of neuronal cells in affected brain regions of SCA3 patients. Brain Pathol 8:669–679

    Article  CAS  PubMed  Google Scholar 

  3. Masino L, Musi V, Menon RP, Fusi P, Kelly G, Frenkiel TA, Trottier Y, Pastore A (2003) Domain architecture of the polyglutamine protein ataxin-3: a globular domain followed by a flexible tail. FEBS Lett 549:21–25

    Article  CAS  PubMed  Google Scholar 

  4. Nagai Y, Inui T, Popiel HA, Fujikake N, Hasegawa K, Urade Y, Goto Y, Naiki H, Toda T (2007) A toxic monomeric conformer of the polyglutamine protein. Nat Struct Mol Biol 14:332–340

    Article  CAS  PubMed  Google Scholar 

  5. Paulson HL, Perez MK, Trottier Y, Trojanowski JQ, Subramony SH, Das SS, Vig P, Mandel JL, Fischbeck KH, Pittman RN (1997) Intranuclear inclusions of expanded polyglutamine protein in spinocerebellar ataxia type 3. Neuron 19:333–344

    Article  CAS  PubMed  Google Scholar 

  6. Bichelmeier U et al (2007) Nuclear localization of ataxin-3 is required for the manifestation of symptoms in SCA3: in vivo evidence. J Neurosci 27:7418–7428

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Yang W, Dunlap JR, Andrews RB, Wetzel R (2002) Aggregated polyglutamine peptides delivered to nuclei are toxic to mammalian cells. Hum Mol Genet 11:2905–2917

    Article  CAS  PubMed  Google Scholar 

  8. Boy J et al (2010) A transgenic mouse model of spinocerebellar ataxia type 3 resembling late disease onset and gender-specific instability of CAG repeats. Neurobiol Dis 37:284–293

    Article  CAS  PubMed  Google Scholar 

  9. Silva-Fernandes A et al (2010) Motor uncoordination and neuropathology in a transgenic mouse model of Machado-Joseph disease lacking intranuclear inclusions and ataxin-3 cleavage products. Neurobiol Dis 40:163–176

    Article  CAS  PubMed  Google Scholar 

  10. Trottier Y, Cancel G, An-Gourfinkel I, Lutz Y, Weber C, Brice A, Hirsch E, Mandel JL (1998) Heterogeneous intracellular localization and expression of ataxin-3. Neurobiol Dis 5:335–347

    Article  CAS  PubMed  Google Scholar 

  11. Warrick JM, Paulson HL, Gray-Board GL, Bui QT, Fischbeck KH, Pittman RN, Bonini NM (1998) Expanded polyglutamine protein forms nuclear inclusions and causes neural degeneration in Drosophila. Cell 93:939–949

    Article  CAS  PubMed  Google Scholar 

  12. Matos CA, de Macedo-Ribeiro S, Carvalho AL (2011) Polyglutamine diseases: the special case of ataxin-3 and Machado-Joseph disease. Prog Neurobiol 95:26–48

    Article  CAS  PubMed  Google Scholar 

  13. Costa Mdo C, Paulson HL (2012) Toward understanding Machado-Joseph disease. Prog Neurobiol 97:239–257

    Article  CAS  PubMed  Google Scholar 

  14. Evers MM, Toonen LJ, van Roon-Mom WM (2014) Ataxin-3 protein and RNA toxicity in spinocerebellar ataxia type 3: current insights and emerging therapeutic strategies. Mol Neurobiol 49:1513–1531

    CAS  PubMed  Google Scholar 

  15. Carvalho DR, La Rocque-Ferreira A, Rizzo IM, Imamura EU, Speck-Martins CE (2008) Homozygosity enhances severity in spinocerebellar ataxia type 3. Pediatr Neurol 38:296–299

    Article  PubMed  Google Scholar 

  16. Rodrigues AJ, Coppola G, Santos C, Costa Mdo C, Ailion M, Sequeiros J, Geschwind DH, Maciel P (2007) Functional genomics and biochemical characterization of the C. elegans orthologue of the Machado-Joseph disease protein ataxin-3. FASEB J 21:1126–1136

    Article  CAS  PubMed  Google Scholar 

  17. Schmitt I, Linden M, Khazneh H, Evert BO, Breuer P, Klockgether T, Wuellner U (2007) Inactivation of the mouse Atxn3 (ataxin-3) gene increases protein ubiquitination. Biochem Biophys Res Commun 362:734–739

    Article  CAS  PubMed  Google Scholar 

  18. Chai Y, Koppenhafer SL, Bonini NM, Paulson HL (1999) Analysis of the role of heat shock protein (Hsp) molecular chaperones in polyglutamine disease. J Neurosci 19:10338–10347

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Chai Y, Wu L, Griffin JD, Paulson HL (2001) The role of protein composition in specifying nuclear inclusion formation in polyglutamine disease. J Biol Chem 276:44889–44897

    Article  CAS  PubMed  Google Scholar 

  20. McCampbell A et al (2000) CREB-binding protein sequestration by expanded polyglutamine. Hum Mol Genet 9:2197–2202

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  22. Yoshizawa T, Yamagishi Y, Koseki N, Goto J, Yoshida H, Shibasaki F, Shoji S, Kanazawa I (2000) Cell cycle arrest enhances the in vitro cellular toxicity of the truncated Machado-Joseph disease gene product with an expanded polyglutamine stretch. Hum Mol Genet 9:69–78

    Article  CAS  PubMed  Google Scholar 

  23. Goti D 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 24:10266–10279

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Hubener J et al (2011) N-terminal ataxin-3 causes neurological symptoms with inclusions, endoplasmic reticulum stress and ribosomal dislocation. Brain 134:1925–1942

    Article  PubMed  Google Scholar 

  25. Berke SJ, Schmied FA, Brunt ER, Ellerby LM, Paulson HL (2004) Caspase-mediated proteolysis of the polyglutamine disease protein ataxin-3. J Neurochem 89:908–918

    Article  CAS  PubMed  Google Scholar 

  26. Breuer P, Haacke A, Evert BO, Wullner U (2010) Nuclear aggregation of polyglutamine-expanded ataxin-3: fragments escape the cytoplasmic quality control. J Biol Chem 285:6532–6537

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Haacke A, Broadley SA, Boteva R, Tzvetkov N, Hartl FU, Breuer P (2006) Proteolytic cleavage of polyglutamine-expanded ataxin-3 is critical for aggregation and sequestration of non-expanded ataxin-3. Hum Mol Genet 15:555–568

    Article  CAS  PubMed  Google Scholar 

  28. Jung J, Xu K, Lessing D, Bonini NM (2009) Preventing Ataxin-3 protein cleavage mitigates degeneration in a Drosophila model of SCA3. Hum Mol Genet 18:4843–4852

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Liman J, Deeg S, Voigt A, Vossfeldt H, Dohm CP, Karch A, Weishaupt J, Schulz JB, Bahr M, Kermer P (2014) CDK5 protects from caspase-induced Ataxin-3 cleavage and neurodegeneration. J Neurochem 129:1013–1023

    Article  CAS  PubMed  Google Scholar 

  30. Wellington CL 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

    Article  CAS  PubMed  Google Scholar 

  31. Haacke A, Hartl FU, Breuer P (2007) Calpain inhibition is sufficient to suppress aggregation of polyglutamine-expanded ataxin-3. J Biol Chem 282:18851–18856

    Article  CAS  PubMed  Google Scholar 

  32. Hubener J et al (2013) Calpain-mediated ataxin-3 cleavage in the molecular pathogenesis of spinocerebellar ataxia type 3 (SCA3). Hum Mol Genet 22:508–518

    Article  PubMed  CAS  Google Scholar 

  33. Koch P et al (2011) Excitation-induced ataxin-3 aggregation in neurons from patients with Machado-Joseph disease. Nature 480:543–546

    Article  CAS  PubMed  Google Scholar 

  34. Simoes AT, Goncalves N, Koeppen A, Deglon N, Kugler S, Duarte CB, Pereira de Almeida L (2012) Calpastatin-mediated inhibition of calpains in the mouse brain prevents mutant ataxin 3 proteolysis, nuclear localization and aggregation, relieving Machado-Joseph disease. Brain 135:2428–2439

    Article  PubMed  Google Scholar 

  35. Simoes AT, Goncalves N, Nobre RJ, Duarte CB, Pereira de Almeida L (2014) Calpain inhibition reduces ataxin-3 cleavage alleviating neuropathology and motor impairments in mouse models of Machado-Joseph disease. Hum Mol Genet 23:4932–4944

    Article  CAS  PubMed  Google Scholar 

  36. Chen X, Tang TS, Tu H, Nelson O, Pook M, Hammer R, Nukina N, Bezprozvanny I (2008) Deranged calcium signaling and neurodegeneration in spinocerebellar ataxia type 3. J Neurosci 28:12713–12724

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Kim YJ, Yi Y, Sapp E, Wang Y, Cuiffo B, Kegel KB, Qin ZH, Aronin N, DiFiglia M (2001) Caspase 3-cleaved N-terminal fragments of wild-type and mutant huntingtin are present in normal and Huntington’s disease brains, associate with membranes, and undergo calpain-dependent proteolysis. Proc Natl Acad Sci U S A 98:12784–12789

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Colomer Gould VF, Goti D, Pearce D, Gonzalez GA, Gao H, Bermudez de Leon M, Jenkins NA, Copeland NG, Ross CA, Brown DR (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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Antony PM, Mantele S, Mollenkopf P, Boy J, Kehlenbach RH, Riess O, Schmidt T (2009) Identification and functional dissection of localization signals within ataxin-3. Neurobiol Dis 36:280–292

    Article  CAS  PubMed  Google Scholar 

  40. Chen S, Peng GH, Wang X, Smith AC, Grote SK, Sopher BL, La Spada AR (2004) Interference of Crx-dependent transcription by ataxin-7 involves interaction between the glutamine regions and requires the ataxin-7 carboxy-terminal region for nuclear localization. Hum Mol Genet 13:53–67

    Article  CAS  PubMed  Google Scholar 

  41. Goehler H et al (2004) A protein interaction network links GIT1, an enhancer of huntingtin aggregation, to Huntington’s disease. Mol Cell 15:853–865

    Article  CAS  PubMed  Google Scholar 

  42. Lim J et al (2006) A protein-protein interaction network for human inherited ataxias and disorders of Purkinje cell degeneration. Cell 125:801–814

    Article  CAS  PubMed  Google Scholar 

  43. Steffan JS, Kazantsev A, Spasic-Boskovic O, Greenwald M, Zhu YZ, Gohler H, Wanker EE, Bates GP, Housman DE, Thompson LM (2000) The Huntington’s disease protein interacts with p53 and CREB-binding protein and represses transcription. Proc Natl Acad Sci U S A 97:6763–6768

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Tsuda H et al (2005) The AXH domain of Ataxin-1 mediates neurodegeneration through its interaction with Gfi-1/senseless proteins. Cell 122:633–644

    Article  CAS  PubMed  Google Scholar 

  45. Takahashi T, Katada S, Onodera O (2010) Polyglutamine diseases: where does toxicity come from? what is toxicity? where are we going? J Mol Cell Biol 2:180–191

    Article  CAS  PubMed  Google Scholar 

  46. 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

    CAS  PubMed  Google Scholar 

  47. Nobrega C, Carmo-Silva S, Albuquerque D, Vasconcelos-Ferreira A, Vijayakumar UG, Mendonca L, Hirai H, de Almeida LP (2015) Re-establishing ataxin-2 downregulates translation of mutant ataxin-3 and alleviates Machado-Joseph disease. Brain 138:3537–3554

    Article  PubMed  Google Scholar 

  48. Clague MJ, Coulson JM, Urbe S (2012) Cellular functions of the DUBs. J Cell Sci 125:277–286

    Article  CAS  PubMed  Google Scholar 

  49. Kleiger G, Mayor T (2014) Perilous journey: a tour of the ubiquitin-proteasome system. Trends Cell Biol 24:352–359

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Reyes-Turcu FE, Ventii KH, Wilkinson KD (2009) Regulation and cellular roles of ubiquitin-specific deubiquitinating enzymes. Annu Rev Biochem 78:363–397

    Article  CAS  PubMed  Google Scholar 

  51. Suresh B, Lee J, Kim H, Ramakrishna S (2016) Regulation of pluripotency and differentiation by deubiquitinating enzymes. Cell Death Differ 23:1257–1264

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Mori H, Kondo J, Ihara Y (1987) Ubiquitin is a component of paired helical filaments in Alzheimer’s disease. Science 235:1641–1644

    Article  CAS  PubMed  Google Scholar 

  53. Chow MK, Mackay JP, Whisstock JC, Scanlon MJ, Bottomley SP (2004) Structural and functional analysis of the Josephin domain of the polyglutamine protein ataxin-3. Biochem Biophys Res Commun 322:387–394

    Article  CAS  PubMed  Google Scholar 

  54. Cummings CJ, Mancini MA, Antalffy B, DeFranco DB, Orr HT, Zoghbi HY (1998) Chaperone suppression of aggregation and altered subcellular proteasome localization imply protein misfolding in SCA1. Nat Genet 19:148–154

    Article  CAS  PubMed  Google Scholar 

  55. Schmidt T, Lindenberg KS, Krebs A, Schols L, Laccone F, Herms J, Rechsteiner M, Riess O, Landwehrmeyer GB (2002) Protein surveillance machinery in brains with spinocerebellar ataxia type 3: redistribution and differential recruitment of 26S proteasome subunits and chaperones to neuronal intranuclear inclusions. Ann Neurol 51:302–310

    Article  CAS  PubMed  Google Scholar 

  56. Zander C, Takahashi J, El Hachimi KH, Fujigasaki H, Albanese V, Lebre AS, Stevanin G, Duyckaerts C, Brice A (2001) Similarities between spinocerebellar ataxia type 7 (SCA7) cell models and human brain: proteins recruited in inclusions and activation of caspase-3. Hum Mol Genet 10:2569–2579

    Article  CAS  PubMed  Google Scholar 

  57. Seidel K, den Dunnen WF, Schultz C, Paulson H, Frank S, de Vos RA, Brunt ER, Deller T, Kampinga HH, Rub U (2010) Axonal inclusions in spinocerebellar ataxia type 3. Acta Neuropathol 120:449–460

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Berke SJ, Chai Y, Marrs GL, Wen H, Paulson HL (2005) Defining the role of ubiquitin-interacting motifs in the polyglutamine disease protein, ataxin-3. J Biol Chem 280:32026–32034

    Article  CAS  PubMed  Google Scholar 

  59. Mao Y, Senic-Matuglia F, Di Fiore PP, Polo S, Hodsdon ME, De Camilli P (2005) Deubiquitinating function of ataxin-3: insights from the solution structure of the Josephin domain. Proc Natl Acad Sci U S A 102:12700–12705

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Nicastro G, Menon RP, Masino L, Knowles PP, McDonald NQ, Pastore A (2005) The solution structure of the Josephin domain of ataxin-3: structural determinants for molecular recognition. Proc Natl Acad Sci U S A 102:10493–10498

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Burnett B, Li F, Pittman RN (2003) The polyglutamine neurodegenerative protein ataxin-3 binds polyubiquitylated proteins and has ubiquitin protease activity. Hum Mol Genet 12:3195–3205

    Article  CAS  PubMed  Google Scholar 

  62. Chai Y, Berke SS, Cohen RE, Paulson HL (2004) Poly-ubiquitin binding by the polyglutamine disease protein ataxin-3 links its normal function to protein surveillance pathways. J Biol Chem 279:3605–3611

    Article  CAS  PubMed  Google Scholar 

  63. Winborn BJ, Travis SM, Todi SV, Scaglione KM, Xu P, Williams AJ, Cohen RE, Peng J, Paulson HL (2008) The deubiquitinating enzyme ataxin-3, a polyglutamine disease protein, edits Lys63 linkages in mixed linkage ubiquitin chains. J Biol Chem 283:26436–26443

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Boeddrich A et al (2006) An arginine/lysine-rich motif is crucial for VCP/p97-mediated modulation of ataxin-3 fibrillogenesis. EMBO J 25:1547–1558

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Donaldson KM, Li W, Ching KA, Batalov S, Tsai CC, Joazeiro CA (2003) Ubiquitin-mediated sequestration of normal cellular proteins into polyglutamine aggregates. Proc Natl Acad Sci U S A 100:8892–8897

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Doss-Pepe EW, Stenroos ES, Johnson WG, Madura K (2003) Ataxin-3 interactions with rad23 and valosin-containing protein and its associations with ubiquitin chains and the proteasome are consistent with a role in ubiquitin-mediated proteolysis. Mol Cell Biol 23:6469–6483

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Durcan TM, Kontogiannea M, Thorarinsdottir T, Fallon L, Williams AJ, Djarmati A, Fantaneanu T, Paulson HL, Fon EA (2011) The Machado-Joseph disease-associated mutant form of ataxin-3 regulates parkin ubiquitination and stability. Hum Mol Genet 20:141–154

    Article  CAS  PubMed  Google Scholar 

  68. Jana NR, Dikshit P, Goswami A, Kotliarova S, Murata S, Tanaka K, Nukina N (2005) Co-chaperone CHIP associates with expanded polyglutamine protein and promotes their degradation by proteasomes. J Biol Chem 280:11635–11640

    Article  CAS  PubMed  Google Scholar 

  69. Matsumoto M, Yada M, Hatakeyama S, Ishimoto H, Tanimura T, Tsuji S, Kakizuka A, Kitagawa M, Nakayama KI (2004) Molecular clearance of ataxin-3 is regulated by a mammalian E4. EMBO J 23:659–669

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Scaglione KM et al (2011) Ube2w and ataxin-3 coordinately regulate the ubiquitin ligase CHIP. Mol Cell 43:599–612

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Tsai YC, Fishman PS, Thakor NV, Oyler GA (2003) Parkin facilitates the elimination of expanded polyglutamine proteins and leads to preservation of proteasome function. J Biol Chem 278:22044–22055

    Article  CAS  PubMed  Google Scholar 

  72. Wang Q, Li L, Ye Y (2006) Regulation of retrotranslocation by p97-associated deubiquitinating enzyme ataxin-3. J Cell Biol 174:963–971

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Ying Z, Wang H, Fan H, Zhu X, Zhou J, Fei E, Wang G (2009) Gp78, an ER associated E3, promotes SOD1 and ataxin-3 degradation. Hum Mol Genet 18:4268–4281

    Article  CAS  PubMed  Google Scholar 

  74. Zhong X, Pittman RN (2006) Ataxin-3 binds VCP/p97 and regulates retrotranslocation of ERAD substrates. Hum Mol Genet 15:2409–2420

    Article  CAS  PubMed  Google Scholar 

  75. Durcan TM, Fon EA (2013) Ataxin-3 and its e3 partners: implications for machado-joseph disease. Front Neurol 4:46

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  76. Laco MN, Cortes L, Travis SM, Paulson HL, Rego AC (2012) Valosin-containing protein (VCP/p97) is an activator of wild-type ataxin-3. PLoS ONE 7:e43563

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Laco MN, Oliveira CR, Paulson HL, Rego AC (2012) Compromised mitochondrial complex II in models of Machado-Joseph disease. Biochim Biophys Acta 1822:139–149

    Article  CAS  PubMed  Google Scholar 

  78. Bence NF, Sampat RM, Kopito RR (2001) Impairment of the ubiquitin-proteasome system by protein aggregation. Science 292:1552–1555

    Article  CAS  PubMed  Google Scholar 

  79. Chen ZJ, Sun LJ (2009) Nonproteolytic functions of ubiquitin in cell signaling. Mol Cell 33:275–286

    Article  CAS  PubMed  Google Scholar 

  80. Neves-Carvalho A et al (2015) Dominant negative effect of polyglutamine expansion perturbs normal function of ataxin-3 in neuronal cells. Hum Mol Genet 24:100–117

    Article  CAS  PubMed  Google Scholar 

  81. Cuervo AM (2004) Autophagy: in sickness and in health. Trends Cell Biol 14:70–77

    Article  PubMed  CAS  Google Scholar 

  82. Cuervo AM (2004) Autophagy: many paths to the same end. Mol Cell Biochem 263:55–72

    Article  CAS  PubMed  Google Scholar 

  83. Williams A, Jahreiss L, Sarkar S, Saiki S, Menzies FM, Ravikumar B, Rubinsztein DC (2006) Aggregate-prone proteins are cleared from the cytosol by autophagy: therapeutic implications. Curr Top Dev Biol 76:89–101

    Article  CAS  PubMed  Google Scholar 

  84. Berger Z et al (2006) Rapamycin alleviates toxicity of different aggregate-prone proteins. Hum Mol Genet 15:433–442

    Article  CAS  PubMed  Google Scholar 

  85. Ravikumar B, Duden R, Rubinsztein DC (2002) Aggregate-prone proteins with polyglutamine and polyalanine expansions are degraded by autophagy. Hum Mol Genet 11:1107–1117

    Article  CAS  PubMed  Google Scholar 

  86. Menzies FM, Huebener J, Renna M, Bonin M, Riess O, Rubinsztein DC (2010) Autophagy induction reduces mutant ataxin-3 levels and toxicity in a mouse model of spinocerebellar ataxia type 3. Brain 133:93–104

    Article  PubMed  CAS  Google Scholar 

  87. Bilen J, Bonini NM (2007) Genome-wide screen for modifiers of ataxin-3 neurodegeneration in Drosophila. PLoS Genet 3:1950–1964

    Article  CAS  PubMed  Google Scholar 

  88. Nascimento-Ferreira I et al (2011) Overexpression of the autophagic beclin-1 protein clears mutant ataxin-3 and alleviates Machado-Joseph disease. Brain 134:1400–1415

    Article  PubMed  Google Scholar 

  89. Ashkenazi A, Bento CF, Ricketts T, Vicinanza M, Siddiqi F, Pavel M, Squitieri F, Hardenberg MC, Imarisio S, Menzies FM, Rubinsztein DC (2017) Polyglutamine tracts regulate beclin 1-dependent autophagy. Nature 545:108–111

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Knott AB, Perkins G, Schwarzenbacher R, Bossy-Wetzel E (2008) Mitochondrial fragmentation in neurodegeneration. Nat Rev Neurosci 9:505–518

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Browne SE, Bowling AC, MacGarvey U, Baik MJ, Berger SC, Muqit MM, Bird ED, Beal MF (1997) Oxidative damage and metabolic dysfunction in Huntington’s disease: selective vulnerability of the basal ganglia. Ann Neurol 41:646–653

    Article  CAS  PubMed  Google Scholar 

  92. Lou S, Lepak VC, Eberly LE, Roth B, Cui W, Zhu XH, Oz G, Dubinsky JM (2016) Oxygen consumption deficit in Huntington disease mouse brain under metabolic stress. Hum Mol Genet

    Google Scholar 

  93. Orr AL, Li S, Wang CE, Li H, Wang J, Rong J, Xu X, Mastroberardino PG, Greenamyre JT, Li XJ (2008) N-terminal mutant huntingtin associates with mitochondria and impairs mitochondrial trafficking. J Neurosci 28:2783–2792

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Panov AV, Gutekunst CA, Leavitt BR, Hayden MR, Burke JR, Strittmatter WJ, Greenamyre JT (2002) Early mitochondrial calcium defects in Huntington’s disease are a direct effect of polyglutamines. Nat Neurosci 5:731–736

    Article  CAS  PubMed  Google Scholar 

  95. Beauchemin AM, Gottlieb B, Beitel LK, Elhaji YA, Pinsky L, Trifiro MA (2001) Cytochrome c oxidase subunit Vb interacts with human androgen receptor: a potential mechanism for neurotoxicity in spinobulbar muscular atrophy. Brain Res Bull 56:285–297

    Article  CAS  PubMed  Google Scholar 

  96. Lodi R, Schapira AH, Manners D, Styles P, Wood NW, Taylor DJ, Warner TT (2000) Abnormal in vivo skeletal muscle energy metabolism in Huntington’s disease and dentatorubropallidoluysian atrophy. Ann Neurol 48:72–76

    Article  CAS  PubMed  Google Scholar 

  97. Kish SJ, Mastrogiacomo F, Guttman M, Furukawa Y, Taanman JW, Dozic S, Pandolfo M, Lamarche J, DiStefano L, Chang LJ (1999) Decreased brain protein levels of cytochrome oxidase subunits in Alzheimer’s disease and in hereditary spinocerebellar ataxia disorders: a nonspecific change? J Neurochem 72:700–707

    Article  CAS  PubMed  Google Scholar 

  98. Matsuishi T, Sakai T, Naito E, Nagamitsu S, Kuroda Y, Iwashita H, Kato H (1996) Elevated cerebrospinal fluid lactate/pyruvate ratio in Machado-Joseph disease. Acta Neurol Scand 93:72–75

    Article  CAS  PubMed  Google Scholar 

  99. Chou AH, Yeh TH, Kuo YL, Kao YC, Jou MJ, Hsu CY, Tsai SR, Kakizuka A, Wang HL (2006) Polyglutamine-expanded ataxin-3 activates mitochondrial apoptotic pathway by upregulating Bax and downregulating Bcl-xL. Neurobiol Dis 21:333–345

    Article  CAS  PubMed  Google Scholar 

  100. Tsai HF, Tsai HJ, Hsieh M (2004) Full-length expanded ataxin-3 enhances mitochondrial-mediated cell death and decreases Bcl-2 expression in human neuroblastoma cells. Biochem Biophys Res Commun 324:1274–1282

    Article  CAS  PubMed  Google Scholar 

  101. Yu YC, Kuo CL, Cheng WL, Liu CS, Hsieh M (2009) Decreased antioxidant enzyme activity and increased mitochondrial DNA damage in cellular models of Machado-Joseph disease. J Neurosci Res 87:1884–1891

    Article  CAS  PubMed  Google Scholar 

  102. Pozzi C, Valtorta M, Tedeschi G, Galbusera E, Pastori V, Bigi A, Nonnis S, Grassi E, Fusi P (2008) Study of subcellular localization and proteolysis of ataxin-3. Neurobiol Dis 30:190–200

    Article  CAS  PubMed  Google Scholar 

  103. Kazachkova N, Raposo M, Montiel R, Cymbron T, Bettencourt C, Silva-Fernandes A, Silva S, Maciel P, Lima M (2013) Patterns of mitochondrial DNA damage in blood and brain tissues of a transgenic mouse model of Machado-Joseph disease. Neurodegener Dis 11:206–214

    Article  CAS  PubMed  Google Scholar 

  104. Ramos A, Kazachkova N, Silva F, Maciel P, Silva-Fernandes A, Duarte-Silva S, Santos C, Lima M (2015) Differential mtDNA damage patterns in a transgenic mouse model of Machado-Joseph disease (MJD/SCA3). J Mol Neurosci 55:449–453

    Article  CAS  PubMed  Google Scholar 

  105. Caviston JP, Ross JL, Antony SM, Tokito M, Holzbaur EL (2007) Huntingtin facilitates dynein/dynactin-mediated vesicle transport. Proc Natl Acad Sci U S A 104:10045–10050

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Gunawardena S, Her LS, Brusch RG, Laymon RA, Niesman IR, Gordesky-Gold B, Sintasath L, Bonini NM, Goldstein LS (2003) Disruption of axonal transport by loss of huntingtin or expression of pathogenic polyQ proteins in Drosophila. Neuron 40:25–40

    Article  CAS  PubMed  Google Scholar 

  107. Szebenyi G et al (2003) Neuropathogenic forms of huntingtin and androgen receptor inhibit fast axonal transport. Neuron 40:41–52

    Article  CAS  PubMed  Google Scholar 

  108. Klockgether T et al (1999) Age related axonal neuropathy in spinocerebellar ataxia type 3/Machado-Joseph disease (SCA3/MJD). J Neurol Neurosurg Psychiatry 66:222–224

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. D’Abreu A, Franca MC Jr, Paulson HL, Lopes-Cendes I (2010) Caring for Machado-Joseph disease: current understanding and how to help patients. Parkinsonism Relat Disord 16:2–7

    Article  PubMed  Google Scholar 

  110. Bading H (2013) Nuclear calcium signalling in the regulation of brain function. Nat Rev Neurosci 14:593–608

    Article  CAS  PubMed  Google Scholar 

  111. Brini M, Cali T, Ottolini D, Carafoli E (2014) Neuronal calcium signaling: function and dysfunction. Cell Mol Life Sci 71:2787–2814

    Article  CAS  PubMed  Google Scholar 

  112. Clapham DE (2007) Calcium signaling. Cell 131:1047–1058

    Article  CAS  PubMed  Google Scholar 

  113. Zhivotovsky B, Orrenius S (2011) Calcium and cell death mechanisms: a perspective from the cell death community. Cell Calcium 50:211–221

    Article  CAS  PubMed  Google Scholar 

  114. Pellistri F et al (2013) Different ataxin-3 amyloid aggregates induce intracellular Ca(2+) deregulation by different mechanisms in cerebellar granule cells. Biochim Biophys Acta 1833:3155–3165

    Article  CAS  PubMed  Google Scholar 

  115. Lim J, Crespo-Barreto J, Jafar-Nejad P, Bowman AB, Richman R, Hill DE, Orr HT, Zoghbi HY (2008) Opposing effects of polyglutamine expansion on native protein complexes contribute to SCA1. Nature 452:713–718

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Yamada M, Tsuji S, Takahashi H (2000) Pathology of CAG repeat diseases. Neuropathology 20:319–325

    Article  CAS  PubMed  Google Scholar 

  117. Kazantsev A, Preisinger E, Dranovsky A, Goldgaber D, Housman D (1999) Insoluble detergent-resistant aggregates form between pathological and nonpathological lengths of polyglutamine in mammalian cells. Proc Natl Acad Sci U S A 96:11404–11409

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Perez MK, Paulson HL, Pendse SJ, Saionz SJ, Bonini NM, Pittman RN (1998) Recruitment and the role of nuclear localization in polyglutamine-mediated aggregation. J Cell Biol 143:1457–1470

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Evert BO, Araujo J, Vieira-Saecker AM, de Vos RA, Harendza S, Klockgether T, Wullner U (2006) Ataxin-3 represses transcription via chromatin binding, interaction with histone deacetylase 3, and histone deacetylation. J Neurosci 26:11474–11486

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Li F, Macfarlan T, Pittman RN, Chakravarti D (2002) Ataxin-3 is a histone-binding protein with two independent transcriptional corepressor activities. J Biol Chem 277:45004–45012

    Article  CAS  PubMed  Google Scholar 

  121. Araujo J, Breuer P, Dieringer S, Krauss S, Dorn S, Zimmermann K, Pfeifer A, Klockgether T, Wuellner U, Evert BO (2011) FOXO4-dependent upregulation of superoxide dismutase-2 in response to oxidative stress is impaired in spinocerebellar ataxia type 3. Hum Mol Genet 20:2928–2941

    Article  CAS  PubMed  Google Scholar 

  122. Chou AH, Yeh TH, Ouyang P, Chen YL, Chen SY, Wang HL (2008) Polyglutamine-expanded ataxin-3 causes cerebellar dysfunction of SCA3 transgenic mice by inducing transcriptional dysregulation. Neurobiol Dis 31:89–101

    Article  CAS  PubMed  Google Scholar 

  123. Evert BO, Vogt IR, Kindermann C, Ozimek L, de Vos RA, Brunt ER, Schmitt I, Klockgether T, Wullner U (2001) Inflammatory genes are upregulated in expanded ataxin-3-expressing cell lines and spinocerebellar ataxia type 3 brains. J Neurosci 21:5389–5396

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Evert BO, Vogt IR, Vieira-Saecker AM, Ozimek L, de Vos RA, Brunt ER, Klockgether T, Wullner U (2003) Gene expression profiling in ataxin-3 expressing cell lines reveals distinct effects of normal and mutant ataxin-3. J Neuropathol Exp Neurol 62:1006–1018

    Article  CAS  PubMed  Google Scholar 

  125. Carmona V, Cunha-Santos J, Onofre I, Simões AT, Vijayakumar U, Davidson BL, de Almeida Pereira (2017) Unravelling endogenous microRNA system dysfunction as a new pathophysiological mechanism in Mahcado-Joseph disease. Mol Ther 25:1028–1055

    Article  CAS  Google Scholar 

  126. Fei E, Jia N, Zhang T, Ma X, Wang H, Liu C, Zhang W, Ding L, Nukina N, Wang G (2007) Phosphorylation of ataxin-3 by glycogen synthase kinase 3 beta at serine 256 regulates the aggregation of ataxin-3. Biochem Biophys Res Commun 357:487–492

    Article  CAS  PubMed  Google Scholar 

  127. Mueller T, Breuer P, Schmitt I, Walter J, Evert BO, Wullner U (2009) CK2-dependent phosphorylation determines cellular localization and stability of ataxin-3. Hum Mol Genet 18:3334–3343

    Article  CAS  PubMed  Google Scholar 

  128. Matos CA, Nóbrega C, Louros SR, Almeida B, Ferreiro E, Valero J, Pereira de Almeida L, Macedo-Ribeiro S, Carvalho AL (2016) Ataxin-3 phosphorylation decreases neuronal defects in spinocerebellar ataxia type 3 models. J Cell Biol 212(4):465–480

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Todi SV, Winborn BJ, Scaglione KM, Blount JR, Travis SM, Paulson HL (2009) Ubiquitination directly enhances activity of the deubiquitinating enzyme ataxin-3. EMBO J 28:372–382

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Todi SV, Scaglione KM, Blount JR, Basrur V, Conlon KP, Pastore A, Elenitoba-Johnson K, Paulson HL (2010) Activity and cellular functions of the deubiquitinating enzyme and polyglutamine disease protein ataxin-3 are regulated by ubiquitination at lysine 117. J Biol Chem 285:39303–39313

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Tsou WL, Burr AA, Ouyang M, Blount JR, Scaglione KM, Todi SV (2013) Ubiquitination regulates the neuroprotective function of the deubiquitinase ataxin-3 in vivo. J Biol Chem 288:34460–34469

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Almeida B, Abreu IA, Matos CA, Fraga JS, Fernandes S, Macedo MG, Gutierrez-Gallego R, Pereira PJ, Carvalho AL, Macedo-Ribeiro S (2015) SUMOylation of the brain-predominant Ataxin-3 isoform modulates its interaction with p97. Biochim Biophys Acta 1852:1950–1959

    Article  CAS  PubMed  Google Scholar 

  133. Zhou YF et al (2013) SUMO-1 modification on K166 of polyQ-expanded ataxin-3 strengthens its stability and increases its cytotoxicity. PLoS ONE 8:e54214

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Farooqui T, Farooqui AA (2009) Aging: an important factor for the pathogenesis of neurodegenerative diseases. Mech Ageing Dev 130:203–215

    Article  CAS  PubMed  Google Scholar 

  135. Lopez-Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G (2013) The hallmarks of aging. Cell 153:1194–1217

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Carrard G, Bulteau AL, Petropoulos I, Friguet B (2002) Impairment of proteasome structure and function in aging. Int J Biochem Cell Biol 34:1461–1474

    Article  CAS  PubMed  Google Scholar 

  137. Ferrington DA, Husom AD, Thompson LV (2005) Altered proteasome structure, function, and oxidation in aged muscle. FASEB J 19:644–646

    Article  CAS  PubMed  Google Scholar 

  138. Keller JN, Dimayuga E, Chen Q, Thorpe J, Gee J, Ding Q (2004) Autophagy, proteasomes, lipofuscin, and oxidative stress in the aging brain. Int J Biochem Cell Biol 36:2376–2391

    Article  CAS  PubMed  Google Scholar 

  139. Boveris A, Navarro A (2008) Brain mitochondrial dysfunction in aging. IUBMB Life 60:308–314

    Article  CAS  PubMed  Google Scholar 

  140. Chang KH et al (2011) Nuclear envelope dispersion triggered by deregulated Cdk5 precedes neuronal death. Mol Biol Cell 22:1452–1462

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  141. Lee KY, Clark AW, Rosales JL, Chapman K, Fung T, Johnston RN (1999) Elevated neuronal Cdc2-like kinase activity in the Alzheimer disease brain. Neurosci Res 34:21–29

    Article  CAS  PubMed  Google Scholar 

  142. Shelton SB, Johnson GV (2004) Cyclin-dependent kinase-5 in neurodegeneration. J Neurochem 88:1313–1326

    Article  CAS  PubMed  Google Scholar 

  143. Bishop NA, Lu T, Yankner BA (2010) Neural mechanisms of ageing and cognitive decline. Nature 464:529–535

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Kirkwood TB (2005) Understanding the odd science of aging. Cell 120:437–447

    Article  CAS  PubMed  Google Scholar 

  145. Cohen HY, Miller C, Bitterman KJ, Wall NR, Hekking B, Kessler B, Howitz KT, Gorospe M, de Cabo R, Sinclair DA (2004) Calorie restriction promotes mammalian cell survival by inducing the SIRT1 deacetylase. Science 305:390–392

    Article  CAS  PubMed  Google Scholar 

  146. Cunha-Santos J, Duarte-Neves J, Carmona V, Guarente L, Pereira de Almeida L, Cavadas C (2016) Caloric restriction blocks neuropathology and motor deficits in Machado-Joseph disease mouse models through SIRT1 pathway. Nat Commun 7:11445

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Clévio Nóbrega or Luís Pereira de Almeida .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2018 Springer International Publishing AG

About this chapter

Check for updates. Verify currency and authenticity via CrossMark

Cite this chapter

Nóbrega, C. et al. (2018). Molecular Mechanisms and Cellular Pathways Implicated in Machado-Joseph Disease Pathogenesis . In: Nóbrega, C., Pereira de Almeida, L. (eds) Polyglutamine Disorders. Advances in Experimental Medicine and Biology, vol 1049. Springer, Cham. https://doi.org/10.1007/978-3-319-71779-1_18

Download citation

Publish with us

Policies and ethics