Skip to main content

Huntington’s Disease

  • Chapter

Part of the book series: Advances in Experimental Medicine and Biology ((AEMB,volume 685))

Abstract

Huntington’s disease (HD) is one of the most frequently found neurodegenerative disorders. Its main clinical manifestations arc chorea, cognitive impairment and psychiatric disorders. It is an autosomal-dominant disorder with almost complete penetrance. The mutation responsible for HD, unstable expansion of a CAG repcat, is located in the 5′ tcrminal section of the gene (ITJS) that encodes huntingtin protein (Htt). The pathophysiology of HD is not entirely clear. One intriguing characteristic of HD is the special vulnerability of the striatum tomutated Htt, despite similar expression of the mutated protcin in other brain regions. Aggregation of mutated Htt, transcriptional dysregulation, altered energy metabolism, excitotoxicity, impaired axonal transport and altered synaptic transmission culminate in neuronal dysfianction and death. There is currently no way ofpreventing or slowing down the disease progression and death usually occurs at about 20 years after dia

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

Buying options

Chapter
USD   29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD   129.00
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD   169.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD   169.99
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

Learn about institutional subscriptions

Preview

Unable to display preview. Download preview PDF.

Unable to display preview. Download preview PDF.

References

  1. Huntington G. On chorea. Med Surg Rep 1872:317–321.

    Google Scholar 

  2. Okun MS, Thommi N. Anserico Negrette (1924 to 2003): diagnosing Huntington disease in Venezuela. Neurology 2004; 63:340–343.

    PubMed  Google Scholar 

  3. Gusella JF, Wexler NS, Conneally PM et al. A polymorphic DNA markergenetically linked to Huntington’s disease. Nature 1983; 306:234–238

    Article  CAS  PubMed  Google Scholar 

  4. A novel gene containing a trinudeotide repeat that is expanded and unstable on Huntington’s disease chromosomes. The Huntington’s Disease Collaborative Research Group. Cell 1993; 72:971–983.

    Google Scholar 

  5. Paradisi I, Heenandez A, Arias S. Huntington disease mutation in Venezuela: age of onset, haplotype analyses and geographic aregation. J Hum Genet 2008; 53:127–135.

    Article  PubMed  Google Scholar 

  6. Harper PS. The epidemiology of Huntington’s disease. Hum Genet 1992; 89:365–376.

    Article  CAS  PubMed  Google Scholar 

  7. Foroud T, Gray J, Ivashina J et al.. Differences in duration of Huntington’s disease based on age at onset. Journal of neurology, neurosurgery and psychiatry 1999; 66:52–56.

    Article  CAS  Google Scholar 

  8. Mahant N, McCusker EA, Byth K et al. Huntington’s disease: clinical correlates of disability and progression. Neurology 2003; 61:1085–1092.

    CAS  PubMed  Google Scholar 

  9. Rao AK, Muratori L, Louis ED et al.. Spectrum of gait impainnents in presymptomatic and symptomatic Huntington’s disease. Mov Disoed 2008; 23:1100–1107.

    Article  Google Scholar 

  10. Hicks SL, Robert MP, Golding CV et al.. Oculomotor deficits indicate the progression of Huntington’s disease. Prog Brain Res 2008: 171:555–558.

    Article  PubMed  Google Scholar 

  11. Paulsen JS, Langbehn DR, Stout JC et al.. Detection of Huntington’s disease decades before diagnosis: the Predict-HD study. Journal of neurology, neurosurgery and psychiatry 2008; 79:874–880.

    Article  CAS  Google Scholar 

  12. Julien CL, Thompson JC, Wild S et al.. Psychiatric disorders in preclinical Huntington’s disease. Journal of neurology, neurosurgery and psychiatry 2007; 78:939–943.

    Article  Google Scholar 

  13. Kingma EM, van Duijn E, Timman R et al.. Behavioural problems in Huntington’s disease using the Problem Behaviours Assessment. Gen Hosp Psychiatry 2008; 30:155–161.

    Article  PubMed  Google Scholar 

  14. Beglinger U, Langbehn DR, Duff K et al.. Probability of obsessive and compulsive symptoms in Hunting ton’s disease. Biol Psychiatry 2007; 61:415–418.

    Article  PubMed  Google Scholar 

  15. Gargiulo M, Lejeune S, Tanguy ML et al.. Long-term outcome of presysnpromatic testing in Huntington disease. EurJ Hum Genet 2009; 17:165–171.

    Article  Google Scholar 

  16. Quinn N, Schrag A. Huntington’s disease and other choreas. J Neurol 1998; 245:709–716.

    Article  CAS  PubMed  Google Scholar 

  17. Penney JB Jr, Young AB, Shoulson l et al.. Huntington’s disease in Venezuela: 7 ears of follow-up on symptomatic and asynsptosnatic individuals. Mov Disord 1990; 5:93–99.

    Article  PubMed  Google Scholar 

  18. Kremer B, Weber B, Hayden MR. New insights into the clinical features, pathogenesis and molecular genetics of Huntington disease. Brain Pathol 1992; 2:321–335.

    Article  CAS  PubMed  Google Scholar 

  19. Busse ME, Wiles CM, Rosser AE. Mobility and falls in people with Huntington’s disease. Journal of neurology, neurosurgery and psychiatry 2009; 80:88–90.

    Article  CAS  Google Scholar 

  20. Wheelock VL, Tempkin T, Marder K et al.. Predictors of nursing home placensent in Huntington disease. Neurology 2003; 60:998–1001.

    CAS  PubMed  Google Scholar 

  21. Kerbeshian J, Burd L, Leech C et al.. Huntington disease and childhood-onsetTouretre syndrome. Am J Med Gener 1991; 39:1–3.

    Article  CAS  Google Scholar 

  22. Jankovic J, Beach J, Ashizawa T. Emotional and functional impact of DNA testing on patients with synsptoms of Huntingtoils disease. J Med Genet 1995: 32:516–518

    Article  CAS  PubMed  Google Scholar 

  23. Vogel CM, Drury I, Terry LC et al.. Myoclonus in adult Huntington’s disease. Ann Neurol 1991; 29:213–215.

    Article  CAS  PubMed  Google Scholar 

  24. Catella F, Scaioli V, Ciano C et al.. Adult onset myoclonic Huntington’s disease. Mov Disord 1993; 8:201–205.

    Article  Google Scholar 

  25. Thompson PD, Bhatia KP, Brown P et al. Cortical myoclonus in Huntington’s disease. Mov Disord 1994; 9:633–641.

    Article  CAS  PubMed  Google Scholar 

  26. Siesling S, Vegter-van der Vlis M, Roos RA. Juvenile Huntington disease in the Netherlands. Pediatr Neurol 1997; 17:37–43.

    Article  CAS  PubMed  Google Scholar 

  27. Gonzalez-Alegre P, Afifi AK. Clinical characteristics of childhood-onset (juvenile) Huntington disease: report of 12 patients and review of the literature. J Child Neurol 2006; 21:223–229.

    PubMed  Google Scholar 

  28. Gambardella A, Muglia M, Labate A et al.. Juvenile Huntington’s disease presenting as progressive myoclonic epilepsy. Neurology 2001; 57:708–711.

    PubMed  Google Scholar 

  29. MacMillan JC, Morrison PJ, Nevin NC et al.. Identification ofan expanded CAG repeat in the Huntington’s disease gene (IT15) in a family reporred to have benign hereditary chotea. J Med Genet 1993; 30:1012–1013.

    Article  CAS  PubMed  Google Scholar 

  30. Britton JW, Uitti RJ, Ahlskog JE et al.. Heteditary late-onset chorea without significant dementia: genetic evidence for substantial phenotypic variation in Huntington’s disease. Neurology 1995; 45:443–447.

    CAS  PubMed  Google Scholar 

  31. Ho AK, Sahakian BJ, Brown RG et al.. Profile of cognitive progression in early Huntington’s disease. Neurology 2003; 61:1702–1706.

    CAS  PubMed  Google Scholar 

  32. Caine ED, Hunt RD, Weingartner H et al.. Huntington’s dementia. Clinical and neuropsychological featutes. Arch Gen Psychiatry 1978; 35:377–384.

    CAS  PubMed  Google Scholar 

  33. Bamford KA, Caine ED, Kido DK et al.. A prospective evaluation of cognitive decline in early Huntington’s disease: functional and radiographic correlates. Neurology 1995; 45:1867–1873.

    CAS  PubMed  Google Scholar 

  34. Peinemann A, Schuller S, Pohl C et al.. Executive dysfunction in early stages of Huntington’s disease is associated with striatal and insular atrophy: a neuropsychological and voxel-based morphometric study. J Neurol Sci 2005; 239:11–19.

    Article  PubMed  Google Scholar 

  35. Caine ED, Ebert MH, Weingartner H. An outline for the analysis of dementia. The memory disorder of Huntingrons disease. Neurology 1977; 27:1087–1092.

    CAS  PubMed  Google Scholar 

  36. Hodges JR, Salmon DP, Butters N. Differential impairment of semantic and episodic memory in Alzheimer’s and Huntington’s diseases: a controlled prospective study. Journal of neurology, neurosurgery and psychiatry 1990; 53:1089–1095.

    Article  CAS  Google Scholar 

  37. Pillon B, Deweer B, Agid Y et al.. Explicit memory in Alzheimer’s, Huntington’s and Parkinson’s diseases. Arch Neurol 1993; 50:374–379.

    CAS  PubMed  Google Scholar 

  38. Pillon B, Deweer B, Michon A et al.. Are explicit memory disorders of progressive supranuclear palsy related to damage to striatofrontal circuits? Comparison with Alzheimer’s, Parkinson’s and Huntington’s diseases. Neurology 1994; 44:1264–1270.

    CAS  PubMed  Google Scholar 

  39. Shiwach RS, Norbury CG. A controlled psychiatric study of individuals at risk for Huntington’s disease. Br J Psychiatry 1994; 165:500–505.

    Article  CAS  PubMed  Google Scholar 

  40. Kirkwood SC, Siemers E, Viken R et al.. Longimdinal personality changes among presympromatic Hun tington disease gene carriers. Neuropsychiarry Neuropsychol Behav Neurol 2002; 15:192–197.

    Google Scholar 

  41. Cummings JL, Cunningham K. Obsessive-compulsive disorder in Huntington’s disease. Biol Psychiatry 1992; 31:263–270.

    Article  CAS  PubMed  Google Scholar 

  42. Patzold T, Brune M. Obsessive compulsive disorder in Huntington disease: a case of isolated obsessions successfully treated with serrraline. Neuropsychiatry Neuropsychol Behav Neurol 2002; 15:216–219.

    PubMed  Google Scholar 

  43. Fedoroff JP, Peyser C, Franz ML et al.. Sexual disorders in Huntington’s disease. J Neuropsychiatry Clin Neurosci 1994; 6:147–153.

    CAS  PubMed  Google Scholar 

  44. Pflanz S, Besson JA, Ebmeier KP et al.. The clinical manifestation of mental disorder in Huntington’s disease: a retrospective case record study of disease progression. Acra Psychiatr Scand 1991; 83:53–60.

    Article  CAS  Google Scholar 

  45. Paulsen JS, Ready RE, Hamilton JM et al.. Neuropsychiatric aspects ofHuntington’s disease. Journal of neurology, neurosurgery and psychiatry 2001; 71:310–314.

    Article  CAS  Google Scholar 

  46. Caine ED, Shoulson I. Psychiatric syndromes in Huntington’s disease. Am J Psychiatry 1983; 140:728–733.

    CAS  PubMed  Google Scholar 

  47. Folstein SE, Franz ML, Jensen BA et al.. Conduct disorder and affective disorder among the offspring of patients with Huntington’s disease. Psychol Med 1983; 13:45–52.

    Article  CAS  PubMed  Google Scholar 

  48. Di Maio L, Squitieri F, Napolitano C et al.. Onset symptoms in 510 patients with Huntington’s disease. J Med Genet 1993; 30:289–292.

    Article  PubMed  Google Scholar 

  49. Cummings JL. Behavioral and psychiatric symptoms associated with Huntington’s disease. Adv Neurol 1995; 65:179–186.

    CAS  PubMed  Google Scholar 

  50. Jensen P, Fenger K, Bolwig TG et al.. Crime in Huntington’s disease: a study of registered offences among patients, relatives and controls. Journal of neurology, neurosurgery and psychiatry 1998: 65:467–471.

    Article  CAS  Google Scholar 

  51. Mendez MF. Mania in neurologic disorders. Curr Psychiatry Rep 2000; 2:440–445.

    Article  CAS  PubMed  Google Scholar 

  52. Rosenblatt A, Leroi I. Neuropsychiatry of Huntington’s disease and other basal ganglia disorders. Psychosomatics 2000; 41:24–30.

    CAS  PubMed  Google Scholar 

  53. Farrer LA. Suicide and anempted suicide in Huntington disease implications for preclinical testing of persons at risk. Am J Med Genet 1986; 24:305–311.

    Article  CAS  PubMed  Google Scholar 

  54. Paulsen JS, Hoth KF, Nehl C et al.. Critical periods of suicide risk in Huntington’s disease. Am J Psychiatry 2005; 162:725–731.

    Article  PubMed  Google Scholar 

  55. Lipe H, Schultz A, Bird TD. Risk factors for suicide in Huntingtons disease: a retrospective case controlled study. Ans J Med Genet 1993; 48:231–233.

    Article  CAS  Google Scholar 

  56. Taylor N, Bramble D. Sleep disturbance and Huntingdon’s disease. Br J Psychiatry 1997; 171:393.

    Article  CAS  PubMed  Google Scholar 

  57. Morton AJ, Wood NI, Hastings MH et al.. Disintegration of the sleep-wake cycle and circadian timing in Huntington’s disease. J Neurosci 2005; 25:157–163.

    Article  CAS  PubMed  Google Scholar 

  58. Amulf I, Nielsen J, Lohmann R et al.. Rapid eye movement sleep disturbances in Huntington disease. Arch Neurol 2008; 65:482–488.

    Article  Google Scholar 

  59. Videnovic A, Leurgans S, Fan W et al.. Daytime somnolence and nocturnsal sleep disturbances in Huntington disease. Parkinsonism Relat Disord 2008.

    Google Scholar 

  60. Aziz NA, van der Burg JM, Landwehrmeyer GB et al.. Weight loss in Huntington disease increases with higher CAG repeat number. Neurology 2008; 71:1506–1513.

    Article  CAS  PubMed  Google Scholar 

  61. Pratley RE, Salbe AD, Ravussio E et al.. Higher sedentary energy expenditure in patients with Huntington’s disease. Ann Neurol 2000; 47:64–70.

    Article  CAS  PubMed  Google Scholar 

  62. Kenney C, Powell S, Jankovic J. Autopsy-proven Huntington’s disease with 29 trinucleotide repeats. Mov Disord 2007; 22:127–130.

    Article  PubMed  Google Scholar 

  63. Semaka A, Warby S, Leavitt BR et al.. Re: Autopsy-proven Huntington’s disease with 29 trinucleotide repeats. Mov Disord 2008; 23:1794–1795: author reply 1793.

    Article  PubMed  Google Scholar 

  64. Reynolds N. Re: Autopsy-proven Huntington’s disease with 29 ttinucleotide repeats. Mov Disotd 2008; 23:1795–1796; author reply 1793.

    Article  Google Scholar 

  65. Goldberg YP, Rommeos JM, Andrew SE et al.. Identification of an Alu retrotransposition event in close proximity to a strong candidate gene for Huntington’s disease. Nature 1993; 362:370–373.

    Article  CAS  PubMed  Google Scholar 

  66. Davis MB, Bateman D, Qion NP et al.. Mutation analysis in patients with postible but apparendy sporadic Huntington’s disease. Lancet 1994; 344:714–717.

    Article  CAS  PubMed  Google Scholar 

  67. Zuhlke C, Riess O, Bockel B et al.. Mitotic stability and meiotic variability of the (CAG)n repeat in the Huntington disease gene. Hum Mol Genet 1993; 2:2063–2067.

    Article  CAS  PubMed  Google Scholar 

  68. Ranen NG, Stine OC, Abbott MH et al.. Anticipation and instability of IT-is (CAG)n repeats in parent-offspring pairs with Huntington disease. Am J Hum Genet 1995; 57:593–602.

    CAS  PubMed  Google Scholar 

  69. Nance MA, Westphal B, Nugent S. Diagnosis of patients presenting to a Huntington disease (HD) clinic without a family history of HD. Neurology 1996; 47:1578–1580.

    CAS  PubMed  Google Scholar 

  70. Myets RH, MacDonald ME, Koroshetz WJ et al.. Dc novo expansion of a (CAG)n repeat in sporadic Huntington’s disease. Nat Genet 1993; 5:168–173.

    Article  Google Scholar 

  71. Alfotd RL, Ashizawa T, Jankovic J et al.. Molecular detection of new mutations, resolution of ambiguous resuIts and complex genetic counselingissues in Huntington disease. Am J Med Genet 1996: 66:281–286.

    Article  Google Scholar 

  72. Wexler NS, Lorimer J, Porter J et al.. Venezuelan kindreds reveal that genetic and environmental factors modulate Huntington’s disease age of onset. Proc NatI Acad Sci USA 2004: 101:3498–3503.

    Article  CAS  Google Scholar 

  73. Andtesen JM, Gayan J, Djousse L et al.. The relationship between CAG repeat length and age of onset differs for Huntington’s disease patients with Juvenile onset or Adult onset. Ann Hum Genet 2007: 71:295–301.

    Article  CAS  Google Scholar 

  74. Snell RG, MacMillan JC, Cheadle JP et al.. Relationship between ttinucleotid e repeat expansion and phenotypic variation in Huntington’s disease. Nat Genet 1993; 4:393–397.

    Article  CAS  PubMed  Google Scholar 

  75. Andrew SE, Goldberg YP, Kremer B et al.. The relationship between trioucleotide (CAG) repeat length and clinical features of Huntington’s disease. Nat Genet 1993; 4:398–403.

    Article  CAS  PubMed  Google Scholar 

  76. Duyao M, Ambrose C, Myers R et al.. Trinucleotide repeat length instability and age of onset in Hunting ton’s disease. Nat Genet 1993; 4:387–392.

    Article  CAS  PubMed  Google Scholar 

  77. Marder K, SandIer S, Lechich A et al.. Relationship between CAG repeat length and late-stage outcomes in Huntington’s disease. Neurology 2002; 59:1622–1624.

    Article  CAS  PubMed  Google Scholar 

  78. Ashizawa T, Wong U, Richards CS et al.. CAG repeat tize and clinical presentation in Huntington’s disease. Neurology 1994: 44:1137–1143.

    CAS  PubMed  Google Scholar 

  79. Illatioshkin SN, Igarashi S, Onodera O et al.. Trinucleotide repeat length and rate of progression of Hun tington’s disease. Ann Neurol 1994; 36:630–635.

    Article  Google Scholar 

  80. Kieburtz K, MacDonald M, Shih C et al.. Trinucleotide repeat length and progression of illness in Hun tiogton’s disease. J Med Genet 1994; 31:872–874.

    Article  CAS  PubMed  Google Scholar 

  81. Claes S, Van Zand K, Legius R et al.. Correlations between triplet repeat expansion and clinical features in Huntington’s disease. Arch Neurol 1995; 52:749–753.

    CAS  PubMed  Google Scholar 

  82. Brandt J, Byisma FW, Gross R et al.. Trinucleotide repeat length and clinical progression in Huntington’s disease. Neurology 1996; 46:527–531.

    CAS  PubMed  Google Scholar 

  83. Penney JB Jr, Vonsattel JP, MacDonald ME et al.. CAG repeat number governs the development rate of pathology in Huntington’s disease. Ann Neutol 1997; 41:689–692.

    Article  Google Scholar 

  84. Squitieri F, Cannella M, Simonelli M. CAG mutation effect on rate of progression in Huntington’s disease. Neurol Sci 2002; 23(Suppl 2):S107–108.

    Article  Google Scholar 

  85. Telenins H, Kremer B, Goldberg YP et al.. Somatic and gnnadal mosaicism of the Huntington diseasegene CAG repeat in brain and sperm. Nat Genet 1994; 6:409–414.

    Article  Google Scholar 

  86. Aronin N, Chase K, Young C et al.. CAG expansion affects the expression of mutant Huntingtin in the Huntington’s disease brain. Neuron 1995; 15:1193–1201.

    Article  CAS  PubMed  Google Scholar 

  87. Kahlem P, Djian P. The expanded CAG repeat associated with juvenile Huntington disease shows a corn mon origin of most orall neurons and glia in human cerebrum. Neurosci Len 2000; 286:203–207.

    Article  CAS  Google Scholar 

  88. Ishiguro H, Yamada K, Sawada H et al.. Age-dependent and tissue-specific CAG repeat instability occurs in mouse knock-in for a mutant Huntington’s disease gene. J Neurosci Res 2001; 65:289–297.

    Article  CAS  PubMed  Google Scholar 

  89. Shelbourne PF, Keller-McGandy C, Bi WL et al.. Triplet repeat mutation length gains correlate with cell-type specific vulnerability in Huntington disease brain. Hum Mol Genet 2007; 16:1133–1142.

    Article  CAS  PubMed  Google Scholar 

  90. Farrer LA, Cupples LA, Wiater P et al.. The normal Huntington disease (HD) allele, or a closely linked gene, influences age at onset of HD. Am J Hum Genet 1993; 53:125–130.

    CAS  PubMed  Google Scholar 

  91. Vuillaunse I, Vermersch P, Destee A et al.. Genetic polymorphisms adjacent to the CAG repeat influence clinical fearutes at onset in Huntington’s disease. Journal of neurologyc neurosurgery and psychiatry 1998; 64:758–762.

    Article  Google Scholar 

  92. Chatropadhyay B, Ghosh S, Gangopadhyay PK et al.. Modulation of age at onset in Huntington’s disease and spinocerebellar ataxia type 2 patients originated from eastern India. Neurosci Lert 2003; 345:93–96.

    Article  Google Scholar 

  93. Rubinsztein DC, Leggo J, Chiano M et al.. Genotypes at the GIuR6 kainate receptor locus are associated with variation in the age of onset of Huntington disease. Proc Natl Acad Sci USA 1997; 94:3872–3876.

    Article  CAS  PubMed  Google Scholar 

  94. MacDonald ME, Vonsatrel JP, Shrinidhi J et al.. Evidence for the GIuR6 gene associated with younger onset age of Huntington’s disease. Neurology 1999; 53:1330–1332.

    CAS  PubMed  Google Scholar 

  95. Naze P, Vuillaume I, Destee A et al.. Mutation analysis and association studies of the ubiquitin carboxy-terminal hydrolase L1 gene in Huntingtoils disease. Neurosci Len 2002; 328:1–4.

    Article  CAS  Google Scholar 

  96. Panas M, Avramopoulos D, Karadima G et al.. Apolipoprotein E and presenilin-1 genotypes in Huntington’s disease. J Neurol 1999; 246:574–577.

    Article  CAS  PubMed  Google Scholar 

  97. Kehoe P, Krawczak M, Harper PS et al.. Age of onset in Huntington disease: sex specific influence of apolipoprotein E genotype and normal CAG repeat length. J Med Genet 1999; 36:108–111.

    CAS  PubMed  Google Scholar 

  98. Holbert S, Denghien I, Kiechle T et al.. The Gln-Ala repeat nansci-iptional activator CA15O interacts with huntingtin: neuropathologic and genetic evidence for a role in Huntington’s disease pathogenesis. Proc Natl Acad Sci USA 2001; 98:1811–1816.

    Article  CAS  PubMed  Google Scholar 

  99. Chattopadhyay B, Baksi K, Mukhopadhyay S et al.. Modulation of age at onset of Huntington disease patients byvariations in TP53 and human caspase activated DNase (hCAD) genes. Neurosci Len 2005; 374:81–86.

    Article  CAS  Google Scholar 

  100. Arning L, Kraus PH, Valenrin S et al.. NR2A and NR2B receptor gene variations modify age at onset in Huntington disease. Neurogenetics 2005; 6:25–28.

    Article  CAS  PubMed  Google Scholar 

  101. Arning L, Saft C, Wieczorek S et al.. NR2A and NR2B receptor gene variations modify age at onset in Huntington disease in a sex-specific manner. Hum Gener 2007.

    Google Scholar 

  102. Merzger S, Rong J, Nguyen HP et al.. Huntingtin-associated protein-1 is a modifier of the age-at-onset of Huntington’s disease. Hum Mol Genet 2008; 17:1137–1146.

    Article  CAS  Google Scholar 

  103. Andresen JM, Gayan J, Cherny SS et al.. Replication of twelve association studies for Huntington’s disease residual age of onset in large Venezuelan kindreds. J Med Genet 2007; 44:44–50.

    Article  CAS  PubMed  Google Scholar 

  104. Vonsattel JP, Myers RH, Stevens TJ et al.. Neuropathoiogical classification of Huntington’s disease. J Neuropathol Exp Neurol 1985; 44:559–577.

    Article  CAS  PubMed  Google Scholar 

  105. Richfield EK, Herkenham M. Selective vulnerability in Huntington’s disease: preferential loss of cannabinoid receptors in lateral globus pallidus. Ann Neurol 1994; 36:577–584.

    Article  CAS  PubMed  Google Scholar 

  106. Maninez-Mir MI, Probst A, Palacios JM. Adenosine A2 receptors: selective localization in the human basal ganglia and alterations with disease. Neuroscience 1991; 42:697–706.

    Article  Google Scholar 

  107. Richfield EK, O’Brien CF, Eskin T et al.. Heterogeneous dopamine receptor changes in early and late Huntington’s disease. Neurosci Lett 1991; 132:121–126.

    Article  CAS  PubMed  Google Scholar 

  108. Goto S, Hirano A, Rojas-Corona RR. Immunohistochemicai visualization of afferent nerve terminals in human globus pallidus and its alteration in neostriaral neurodegenerative disorders. Acta Neuropathol (Berl) 1989; 78:543–550.

    Article  CAS  Google Scholar 

  109. Dawbarn D, De Quidt ME Emson PC. Survival of basal ganglia neuropeptide Y-somatostatin neurones in Huntington’s disease. Brain Res 1985; 340:251–260.

    Article  CAS  PubMed  Google Scholar 

  110. Ferrante RJ, Kowall NW, Beal MF et al. Selective sparing of a class of striatal neurons in Huntington’s disease. Science 1985; 230:561–563.

    Article  CAS  PubMed  Google Scholar 

  111. Ferrante RJ, Kowall NW, Beal MF et al. Morphologic and histochemical characteristics of a spared subset of striatal neurons in Huntington’s disease. J Neuropathol Exp Neurol 1987; 46:12–27.

    Article  CAS  PubMed  Google Scholar 

  112. Ferrante RJ, Kowall NW, Richardson EP Jr. Proliferative and degenerative changes in striatal spiny neurons in Huntington’s disease: a combined study using the section-Golgi method and calbindin D28k immunocytochemistry. J Neurosci 1991; 11:3877–3887.

    CAS  PubMed  Google Scholar 

  113. Lange H, Thorner G, Hopf A et al. Morphometric studies of the neuropathological changes in choreatic diseases. J Neurol Sci 1976; 28:401–425.

    Article  CAS  PubMed  Google Scholar 

  114. DiFiglia M, Sapp E, Chase KO et al. Aggregation of huntingtin in neuronal intranuclear inclusions and dystrophic neurites in brain. Science 1997; 277:1990–1993.

    Article  CAS  PubMed  Google Scholar 

  115. Graveland GA, Williams RS, DiFiglia M. Evidence for degenerative and regenerative changes in neostriatal spiny neurons in Huntington’s disease. Science 1985; 227:770–773.

    Article  CAS  PubMed  Google Scholar 

  116. de la Monte SM, Vonsattel JP, Richardson EP Jr. Morphometric demonstration of atrophic changes in the cerebral cortex, white matter and neostriatum in Huntington’s disease. J Neuropathol Exp Neurol 1988; 47:516–525.

    Article  PubMed  Google Scholar 

  117. Sotrel A, Paskevich PA, Kiely DK et al. Morphometric analysis of the prefrontal cortex in Huntington’s disease. Neurology 1991; 41:1117–1123.

    CAS  PubMed  Google Scholar 

  118. Heinsen H, Strik M, Bauer M et al. Cortical and striatal neurone number in Huntington’s disease. Acta Neuropathol (Berl) 1994; 88:320–333.

    Article  CAS  Google Scholar 

  119. Selemon LD, Rajkowska G, Goldman-Rakic PS. Evidence for progression in frontal cortical pathology in late-stage Huntington’s disease. J Comp Neurol 2004; 468:190–204.

    Article  PubMed  Google Scholar 

  120. Oyanagi K, Takeda S, Takahashi H et al. A quantitative investigation of the substantia nigra in Huntington’s disease. Ann Neurol 1989; 26:13–19.

    Article  CAS  PubMed  Google Scholar 

  121. Heinsen H, Rub U, Bauer M et al. Nerve cell loss in the thalamic mediodorsal nucleus in Huntington’s disease. Acta Neuropathol (Berl) 1999; 97:613–622.

    Article  CAS  Google Scholar 

  122. Heinsen H, Rub U, Gangnus D et al. Nerve cell loss in the thalamic centromedian-parafascicular complex in patients with Huntington’s disease. Acta Neuropathol (Berl) 1996; 91:161–168.

    Article  CAS  Google Scholar 

  123. Kremer HP, Roos RA, Dingjan G et al. Atrophy of the hypothalamic lateral tuberal nucleus in Huntington’s disease. J Neuropathol Exp Neurol 1990; 49:371–382.

    Article  CAS  PubMed  Google Scholar 

  124. Jeste DV, Barban L, Parisi J. Reduced Purkinje cell density in Huntington’s disease. Exp Neurol 1984; 85:78–86.

    Article  CAS  PubMed  Google Scholar 

  125. Rodda RA. Cerebellar atrophy in Huntington’s disease. J Neurol Sci 1981; 50:147–157.

    Article  CAS  PubMed  Google Scholar 

  126. Hoffner G, Island ML, Djian P. Purification of neuronal inclusions of patients with Huntington’s disease reveals a broad range of N-terminal fragments of expanded huntingtin and insoluble polymers. J Neurochem 2005; 95:125–136.

    Article  CAS  PubMed  Google Scholar 

  127. Gutekunst CA, Li SH, Yi H et al. Nuclear and neuropil aggregates in Huntington’s disease: relationship to neuropathology. J Neurosci 1999; 19:2522–2534.

    CAS  PubMed  Google Scholar 

  128. Scherzinger E, Lurz R, Turmaine M et al. Huntingtin-encoded polyglutamine expansions form amyloid-like protein aggregates in vitro and in vivo. Cell 1997; 90:549–558.

    Article  CAS  PubMed  Google Scholar 

  129. Huang CC, Faber PW, Persichetti F et al. Amyloid formation by mutant huntingtin: threshold, progressivity and recruitment of normal polyglutamine proteins. Somat Cell Mol Genet 1998; 24:217–233.

    Article  CAS  PubMed  Google Scholar 

  130. Diguet E, Petit F, Escartin C et al. Normal aging modulates the neurotoxicity of mutant huntingtin. PLoS ONE 2009; 4:e4637.

    Article  PubMed  CAS  Google Scholar 

  131. Kegel KB, Meloni AR, Yi Y et al. Huntingtin is present in the nucleus, interacts with the transcriptional corepressor C-terminal binding protein and represses transcription. J Biol Chem 2002; 277:7466–7476.

    Article  CAS  PubMed  Google Scholar 

  132. Kegel KB, Sapp E, Yoder J et al. Huntingtin associates with acidic phospholipids at the plasma membrane. J Biol Chem 2005; 280:36464–36473.

    Article  CAS  PubMed  Google Scholar 

  133. Rockabrand E, Slepko N, Pantalone A et al. The first 17 amino acids of Huntingtin modulate its sub-cellular localization, aggregation and effects on calcium homeostasis. Hum Mol Genet 2007; 16:61–77.

    Article  CAS  PubMed  Google Scholar 

  134. Caviston JP, Ross JL, Antony SM et al. Huntingtin facilitates dynein/dynactin-mediated vesicle transport. Proc Natl Acad Sci USA 2007; 104:10045–10050.

    Article  CAS  PubMed  Google Scholar 

  135. Strehlow AN, Li JZ, Myers RM. Wild-type huntingtin participates in protein trafficking between the Golgi and the extracellular space. Hum Mol Genet 2007; 16:391–409.

    Article  CAS  PubMed  Google Scholar 

  136. Harjes P, Wanker EE. The hunt for huntingtin function: interaction partners tell many different stories. Trends Biochem Sci 2003; 28:425–433.

    Article  CAS  PubMed  Google Scholar 

  137. Li SH, Li XJ. Huntingtin-protein interactions and the pathogenesis of Huntington’s disease. Trends Genet 2004; 20:146–154.

    Article  PubMed  CAS  Google Scholar 

  138. Sugars KL, Rubinsztein DC. Transcriptional abnormalities in Huntington disease. Trends Genet 2003; 19:233–238.

    Article  CAS  PubMed  Google Scholar 

  139. Zhai W, Jeong H, Cui L et al. In vitro analysis of huntingtin-mediated transcriptional repression reveals multiple transcription factor targets. Cell 2005; 123:1241–1253.

    Article  CAS  PubMed  Google Scholar 

  140. Sipione S, Rigamonti D, Valenza M et al. Early transcriptional profiles in huntingtin-inducible striatal cells by microarray analyses. Hum Mol Genet 2002; 11:1953–1965.

    Article  CAS  PubMed  Google Scholar 

  141. Luthi-Carter R, Hanson SA, Strand AD et al. Dysregulation of gene expression in the R6/2 model of polyglutamine disease: parallel changes in muscle and brain. Hum Mol Genet 2002; 11:1911–1926.

    Article  CAS  PubMed  Google Scholar 

  142. Luthi-Carter R, Strand A, Peters NL et al. Decreased expression of striatal signaling genes in a mouse model of Huntington’s disease. Hum Mol Genet 2000; 9:1259–1271.

    Article  CAS  PubMed  Google Scholar 

  143. Luthi-Carter R, Strand AD, Hanson SA et al. Polyglutamine and transcription: gene expression changes shared by DRPLA and Huntington’s disease mouse models reveal context-independent effects. Hum Mol Genet 2002; 11:1927–1937.

    Article  CAS  PubMed  Google Scholar 

  144. Kuhn A, Goldstein DR, Hodges A et al. Mutant huntingtin’s effects on striatal gene expression in mice recapitulate changes observed in human Huntington’s disease brain and do not differ with mutant huntingtin length or wild-type huntingtin dosage. Hum Mol Genet 2007.

    Google Scholar 

  145. Duyao MP, Auerbach AB, Ryan A et al. Inactivation of the mouse Huntington’s disease gene homolog Hdh. Science 1995; 269:407–410.

    Article  CAS  PubMed  Google Scholar 

  146. Zeitlin S, Liu JP, Chapman DL et al. Increased apoptosis and early embryonic lethality in mice nullizygous for the Huntington’s disease gene homologue. Nat Genet 1995; 11:155–163.

    Article  CAS  PubMed  Google Scholar 

  147. Dragatsis I, Dietrich P, Zeitlin S. Expression of the Huntingtin-associated protein 1 gene in the developing and adult mouse. Neurosci Lett 2000; 282:37–40.

    Article  CAS  PubMed  Google Scholar 

  148. Perutz MF, Pope BJ, Owen D et al. Aggregation of proteins with expanded glutamine and alanine repeats of the glutamine-rich and asparagine-rich domains of Sup35 and of the amyloid beta-peptide of amyloid plaques. Proc Natl Acad Sci USA 2002; 99:5596–5600.

    Article  CAS  PubMed  Google Scholar 

  149. Weiss A, Klein C, Woodman B et al. Sensitive biochemical aggregate detection reveals aggregation onset before symptom development in cellular and murine models of Huntington’s disease. J Neurochem 2007.

    Google Scholar 

  150. Martindale D, Hackam A, Wieczorek A et al. Length of huntingtin and its polyglutamine tract influences localization and frequency of intracellular aggregates. Nat Genet 1998; 18:150–154.

    Article  CAS  PubMed  Google Scholar 

  151. Kazantsev A, Preisinger E, Dranovsky A et al. Insoluble detergent-resistant aggregates form between pathological and nonpathological lengths of polyglutamine in mammalian cells. Proc Natl Acad Sci USA 1999; 96:11404–11409.

    Article  CAS  PubMed  Google Scholar 

  152. Nucifora FC, Jr, Sasaki M, Peters MF et al. Interference by huntingtin and atrophin-1 with cbp-mediated transcription leading to cellular toxicity. Science 2001; 291:2423–2428.

    Article  CAS  PubMed  Google Scholar 

  153. Steffan JS, Kazantsev A, Spasic-Boskovic O et al. The Huntington’s disease protein interacts with p53 and CREB-binding protein and represses transcription. Proc Natl Acad Sci USA 2000; 97:6763–6768.

    Article  CAS  PubMed  Google Scholar 

  154. Gunawardena S, Her LS, Brusch RG et al. Disruption of axonal transport by loss of huntingtin or expression of pathogenic polyQ proteins in Drosophila. Neuron 2003; 40:25–40.

    Article  CAS  PubMed  Google Scholar 

  155. Trushina E, Dyer RB, Badger JD 2nd et al. Mutant huntingtin impairs axonal trafficking in mammalian neurons in vivo and in vitro. Mol Cell Biol 2004; 24:8195–8209.

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  157. Verhoef LG, Lindsten K, Masucci MG et al. Aggregate formation inhibits proteasomal degradation of polyglutamine proteins. Hum Mol Genet 2002; 11:2689–2700.

    Article  CAS  PubMed  Google Scholar 

  158. Venkatraman P, Wetzel R, Tanaka M et al. Eukaryotic proteasomes cannot digest polyglutamine sequences and release them during degradation of polyglutamine-containing proteins. Mol Cell 2004; 14:95–104.

    Article  CAS  PubMed  Google Scholar 

  159. Jana NR, Zemskov EA, Wang G et al. Altered proteasomal function due to the expression of polyglutamine-expanded truncated N-terminal huntingtin induces apoptosis by caspase activation through mitochondrial cytochrome c release. Hum Mol Genet 2001; 10:1049–1059.

    Article  CAS  PubMed  Google Scholar 

  160. Bennett EJ, Shaler TA, Woodman B et al. Global changes to the ubiquitin system in Huntington’s disease. Nature 2007; 448:704–708.

    Article  CAS  PubMed  Google Scholar 

  161. Hunter JM, Lesort M, Johnson GV. Ubiquitin-proteasome system alterations in a striatal cell model of Huntington’s disease. J Neurosci Res 2007.

    Google Scholar 

  162. Tydlacka S, Wang CE, Wang X et al. Differential activities of the ubiquitin-proteasome system in neurons versus glia may account for the preferential accumulation of misfolded proteins in neurons. J Neurosci 2008; 28:13285–13295.

    Article  CAS  PubMed  Google Scholar 

  163. Saudou F, Finkbeiner S, Devys D et al. Huntingtin acts in the nucleus to induce apoptosis but death does not correlate with the formation of intranuclear inclusions. Cell 1998; 95:55–66.

    Article  CAS  PubMed  Google Scholar 

  164. Slow EJ, Graham RK, Osmand AP et al. Absence of behavioral abnormalities and neurodegeneration in vivo despite widespread neuronal huntingtin inclusions. Proc Natl Acad Sci USA 2005; 102:11402–11407.

    Article  CAS  PubMed  Google Scholar 

  165. Arrasate M, Mitra S, Schweitzer ES et al. Inclusion body formation reduces levels of mutant huntingtin and the risk of neuronal death. Nature 2004; 431:805–810.

    Article  CAS  PubMed  Google Scholar 

  166. Sawada H, Ishiguro H, Nishii K et al. Characterization of neuron-specific huntingtin aggregates in human huntingtin knock-in mice. Neurosci Res 2007; 57:559–573.

    Article  CAS  PubMed  Google Scholar 

  167. Mitra S, Tsvetkov AS, Finkbeiner S. Single neuron ubiquitin-proteasome dynamics accompanying inclusion body formation in Huntington disease. J Biol Chem 2009; 284:4398–4403.

    Article  CAS  PubMed  Google Scholar 

  168. Gong B, Lim MC, Wanderer J et al. Time-lapse analysis of aggregate formation in an inducible PC12 cell model of Huntington’s disease reveals time-dependent aggregate formation that transiently delays cell death. Brain Res Bull 2008; 75:146–157.

    Article  CAS  PubMed  Google Scholar 

  169. Li H, Li SH, Yu ZX et al. Huntingtin aggregate-associated axonal degeneration is an early pathological event in Huntington’s disease mice. J Neurosci 2001; 21:8473–8481.

    CAS  PubMed  Google Scholar 

  170. Li H, Li SH, Johnston H et al. Amino-terminal fragments of mutant huntingtin show selective accumulation in striatal neurons and synaptic toxicity. Nat Genet 2000; 25:385–389.

    Article  CAS  PubMed  Google Scholar 

  171. Lee WC, Yoshihara M, Littleton JT. Cytoplasmic aggregates trap polyglutamine-containing proteins and block axonal transport in a Drosophila model of Huntington’s disease. Proc Natl Acad Sci USA 2004; 101:3224–3229.

    Article  CAS  PubMed  Google Scholar 

  172. DiFiglia M. Clinical Genetics, II. Huntington’s disease: from the gene to pathophysiology. Am J Psychiatry 1997; 154:1046.

    CAS  PubMed  Google Scholar 

  173. Herbst M, Wanker EE. Small molecule inducers of heat-shock response reduce polyQ-mediated huntingtin aggregation. A possible therapeutic strategy. Neuro-degenerative diseases 2007; 4:254–260.

    CAS  PubMed  Google Scholar 

  174. Jana NR, Tanaka M, Wang G et al. Polyglutamine length-dependent interaction of Hsp40 and Hsp70 family chaperones with truncated N-terminal huntingtin: their role in suppression of aggregation and cellular toxicity. Hum Mol Genet 2000; 9:2009–2018.

    Article  CAS  PubMed  Google Scholar 

  175. Carmichael J, Chatellier J, Woolfson A et al. Bacterial and yeast chaperones reduce both aggregate formation and cell death in mammalian cell models of Huntington’s disease. Proc Natl Acad Sci USA 2000; 97:9701–9705.

    Article  CAS  PubMed  Google Scholar 

  176. Vacher C, Garcia-Oroz L, Rubinsztein DC. Overexpression of yeast hsp104 reduces polyglutamine aggregation and prolongs survival of a transgenic mouse model of Huntington’s disease. Hum Mol Genet 2005; 14:3425–3433.

    Article  CAS  PubMed  Google Scholar 

  177. Perrin V, Regulier E, Abbas-Terki T et al. Neuroprotection by Hsp104 and Hsp27 in Lentiviral-based rat models of Huntington’s disease. Mol Ther 2007; 15:903–911.

    Article  CAS  PubMed  Google Scholar 

  178. Seo H, Sonntag KC, Kim W et al. Proteasome activator enhances survival of Huntington’s disease neuronal model cells. PLoS ONE 2007; 2:e238.

    Article  PubMed  CAS  Google Scholar 

  179. Ravikumar B, Berger Z, Vacher C et al. Rapamycin pretreatment protects against apoptosis. Hum Mol Genet 2006; 15:1209–1216.

    Article  CAS  PubMed  Google Scholar 

  180. Sarkar S, Perlstein EO, Imarisio S et al. Small molecules enhance autophagy and reduce toxicity in Huntington’s disease models. Nature Chemical Biology 2007.

    Google Scholar 

  181. Chopra V, Fox JH, Lieberman G et al. A small-molecule therapeutic lead for Huntington’s disease: preclinical pharmacology and efficacy of C2-8 in the R6/2 transgenic mouse. Proc Natl Acad Sci USA 2007; 104:16685–16689.

    Article  CAS  PubMed  Google Scholar 

  182. Augood SJ, Faull RL, Emson PC. Dopamine D1 and D2 receptor gene expression in the striatum in Huntington’s disease. Ann Neurol 1997; 42:215–221.

    Article  CAS  PubMed  Google Scholar 

  183. Augood SJ, Faull RL, Love DR et al. Reduction in enkephalin and substance P messenger RNA in the striatum of early grade Huntington’s disease: a detailed cellular in situ hybridization study. Neuroscience 1996; 72:1023–1036.

    Article  CAS  PubMed  Google Scholar 

  184. Cha JH, Kosinski CM, Kerner JA et al. Altered brain neurotransmitter receptors in transgenic mice expressing a portion of an abnormal human Huntington disease gene. Proc Natl Acad Sci USA 1998; 95:6480–6485.

    Article  CAS  PubMed  Google Scholar 

  185. Cha JH, Frey AS, Alsdorf SA et al. Altered neurotransmitter receptor expression in transgenic mouse models of Huntington’s disease. Philos Trans R Soc Lond B Biol Sci 1999; 354:981–989.

    Article  CAS  PubMed  Google Scholar 

  186. Cha JH. Transcriptional signatures in Huntington’s disease. Prog Neurobiol 2007.

    Google Scholar 

  187. Runne H, Kuhn A, Wild EJ et al. Analysis of potential transcriptomic biomarkers for Huntington’s disease in peripheral blood. Proc Natl Acad Sci USA 2007; 104:14424–14429.

    Article  CAS  PubMed  Google Scholar 

  188. Borovecki F, Lovrecic L, Zhou J et al. Genome-wide expression profiling of human blood reveals biomarkers for Huntington’s disease. Proc Natl Acad Sci USA 2005; 102:11023–11028.

    Article  CAS  PubMed  Google Scholar 

  189. Dunah AW, Jeong H, Griffin A et al. Sp1 and TAFII130 transcriptional activity disrupted in early Huntington’s disease. Science 2002; 296:2238–2243.

    Article  CAS  PubMed  Google Scholar 

  190. Li SH, Cheng AL, Zhou H et al. Interaction of Huntington disease protein with transcriptional activator Sp1. Mol Cell Biol 2002; 22:1277–1287.

    Article  CAS  PubMed  Google Scholar 

  191. Shimohata T, Nakajima T, Yamada M et al. Expanded polyglutamine stretches interact with TAFII130, interfering with CREB-dependent transcription. Nat Genet 2000; 26:29–36.

    Article  CAS  PubMed  Google Scholar 

  192. Suhr ST, Senut MC, Whitelegge JP et al. Identities of sequestered proteins in aggregates from cells with induced polyglutamine expression. J Cell Biol 2001; 153:283–294.

    Article  CAS  PubMed  Google Scholar 

  193. Bae BI, Xu H, Igarashi S et al. p53 mediates cellular dysfunction and behavioral abnormalities in Huntington’s disease. Neuron 2005; 47:29–41.

    Article  CAS  PubMed  Google Scholar 

  194. Arango M, Holbert S, Zala D et al. CA150 expression delays striatal cell death in overexpression and knock-in conditions for mutant huntingtin neurotoxicity. J Neurosci 2006; 26:4649–4659.

    Article  CAS  PubMed  Google Scholar 

  195. Boutell JM, Thomas P, Neal JW et al. Aberrant interactions of transcriptional repressor proteins with the Huntington’s disease gene product, huntingtin. Hum Mol Genet 1999; 8:1647–1655.

    Article  CAS  PubMed  Google Scholar 

  196. Ferrer I, Goutan E, Marin C et al. Brain-derived neurotrophic factor in Huntington disease. Brain Res 2000; 866:257–261.

    Article  CAS  PubMed  Google Scholar 

  197. Zuccato C, Ciammola A, Rigamonti D et al. Loss of huntingtin-mediated BDNF gene transcription in Huntington’s disease. Science 2001; 293:493–498.

    Article  CAS  PubMed  Google Scholar 

  198. Strand AD, Baquet ZC, Aragaki AK et al. Expression profiling of Huntington’s disease models suggests that brain-derived neurotrophic factor depletion plays a major role in striatal degeneration. J Neurosci 2007; 27:11758–11768.

    Article  CAS  PubMed  Google Scholar 

  199. Lynch G, Kramar EA, Rex CS et al. Brain-derived neurotrophic factor restores synaptic plasticity in a knock-in mouse model of Huntington’s disease. J Neurosci 2007; 27:4424–4434.

    Article  CAS  PubMed  Google Scholar 

  200. Zuccato C, Tartari M, Crotti A et al. Huntingtin interacts with REST/NRSF to modulate the transcription of NRSE-controlled neuronal genes. Nat Genet 2003; 35:76–83.

    Article  CAS  PubMed  Google Scholar 

  201. Zuccato C, Belyaev N, Conforti P et al. Widespread disruption of repressor element-1 silencing transcription factor/neuron-restrictive silencer factor occupancy at its target genes in Huntington’s disease. J Neurosci 2007; 27:6972–6983.

    Article  CAS  PubMed  Google Scholar 

  202. Rigamonti D, Bolognini D, Mutti C et al. Loss of huntingtin function complemented by small molecules acting as re1/nrse silencer modulators. J Biol Chem 2007.

    Google Scholar 

  203. Simmons DA, Rex CS, Palmer L et al. Up-regulating BDNF with an ampakine rescues synaptic plasticity and memory in Huntington’s disease knockin mice. Proc Natl Acad Sci USA 2009; 106:4906–4911.

    Article  CAS  PubMed  Google Scholar 

  204. Taniura H, Sng JC, Yoneda Y. Histone modifications in the brain. Neurochemistry International 2007; 51:85–91.

    Article  CAS  PubMed  Google Scholar 

  205. Steffan JS, Bodai L, Pallos J et al. Histone deacetylase inhibitors arrest polyglutamine-dependent neurodegeneration in Drosophila. Nature 2001; 413:739–743.

    Article  CAS  PubMed  Google Scholar 

  206. Hockly E, Richon VM, Woodman B et al. Suberoylanilide hydroxamic acid, a histone deacetylase inhibitor, ameliorates motor deficits in a mouse model of Huntington’s disease. Proc Natl Acad Sci USA 2003; 100:2041–2046.

    Article  CAS  PubMed  Google Scholar 

  207. Ferrante RJ, Kubilus JK, Lee J et al. Histone deacetylase inhibition by sodium butyrate chemotherapy ameliorates the neurodegenerative phenotype in Huntington’s disease mice. J Neurosci 2003; 23:9418–9427.

    CAS  PubMed  Google Scholar 

  208. Gardian G, Browne SE, Choi DK et al. Neuroprotective effects of phenylbutyrate in the N171-82Q transgenic mouse model of Huntington’s disease. J Biol Chem 2005; 280:556–563.

    CAS  PubMed  Google Scholar 

  209. Thomas EA, Coppola G, Desplats PA et al. The HDAC inhibitor 4b ameliorates the disease phenotype and transcriptional abnormalities in Huntington’s disease transgenic mice. Proc Natl Acad Sci USA 2008; 105:15564–15569.

    Article  CAS  PubMed  Google Scholar 

  210. Pallos J, Bodai L, Lukacsovich T et al. Inhibition of specific HDACs and sirtuins suppresses pathogenesis in a Drosophila model of Huntington’s disease. Hum Mol Genet 2008; 17:3767–3775.

    Article  CAS  PubMed  Google Scholar 

  211. Sadri-Vakili G, Bouzou B, Benn CL et al. Histones associated with downregulated genes are hypo-acetylated in Huntington’s disease models. Hum Mol Genet 2007.

    Google Scholar 

  212. Ryu H, Lee J, Hagerty SW et al. ESET/SETDB1 gene expression and histone H3 (K9) trimethylation in Huntington’s disease. Proc Natl Acad Sci USA 2006; 103:19176–19181.

    Article  CAS  PubMed  Google Scholar 

  213. Yang L, Mei Q, Zielinska-Kwiatkowska A et al. An ERG (ets-related gene)-associated histone methyltransferase interacts with histone deacetylases 1/2 and transcription corepressors mSin3A/B. Biochem J 2003; 369:651–657.

    Article  CAS  PubMed  Google Scholar 

  214. Stack EC, Del Signore SJ, Luthi-Carter R et al. Modulation of Nucleosome Dynamics in Huntington’s Disease. Hum Mol Genet 2007.

    Google Scholar 

  215. Roze E, Betuing S, Deyts C et al. Mitogen-and stress-activated protein kinase-1 deficiency is involved in expanded-huntingtin-induced transcriptional dysregulation and striatal death. FASEB J 2008; 22:1083–1093.

    Article  CAS  PubMed  Google Scholar 

  216. Kim MO, Chawla P, Overland RP et al. Altered histone monoubiquitylation mediated by mutant huntingtin induces transcriptional dysregulation. J Neurosci 2008; 28:3947–3957.

    Article  CAS  PubMed  Google Scholar 

  217. Rothman SM, Olney JW. Excitotoxicity and the NMDA receptor-still lethal after eight years. Trends Neurosci 1995; 18:57–58.

    Article  CAS  PubMed  Google Scholar 

  218. Cull-Candy S, Brickley S, Farrant M. NMDA receptor subunits: diversity, development and disease. Curr Opin Neurobiol 2001; 11:327–335.

    Article  CAS  PubMed  Google Scholar 

  219. Dingledine R, Borges K, Bowie D et al. The glutamate receptor ion channels. Pharmacol Rev 1999; 51:7–61.

    CAS  PubMed  Google Scholar 

  220. Beal MF, Kowall NW, Ellison DW et al. Replication of the neurochemical characteristics of Huntington’s disease by quinolinic acid. Nature 1986; 321:168–171.

    Article  CAS  PubMed  Google Scholar 

  221. Sanberg PR, Calderon SF, Giordano M et al. The quinolinic acid model of Huntington’s disease: locomotor abnormalities. Exp Neurol 1989; 105:45–53.

    Article  CAS  PubMed  Google Scholar 

  222. Ferrante RJ, Kowall NW, Cipolloni PB et al. Excitotoxin lesions in primates as a model for Huntington’s disease: histopathologic and neurochemical characterization. Exp Neurol 1993; 119:46–71.

    Article  CAS  PubMed  Google Scholar 

  223. Cepeda C, Hurst RS, Calvert CR et al. Transient and progressive electrophysiological alterations in the corticostriatal pathway in a mouse model of Huntington’s disease. J Neurosci 2003; 23:961–969.

    CAS  PubMed  Google Scholar 

  224. Stack EC, Dedeoglu A, Smith KM et al. Neuroprotective effects of synaptic modulation in Huntington’s disease R6/2 mice. J Neurosci 2007; 27:12908–12915.

    Article  CAS  PubMed  Google Scholar 

  225. Behrens PF, Franz P, Woodman B et al. Impaired glutamate transport and glutamate-glutamine cycling: downstream effects of the Huntington mutation. Brain 2002; 125:1908–1922.

    Article  CAS  PubMed  Google Scholar 

  226. Shin JY, Fang ZH, Yu ZX et al. Expression of mutant huntingtin in glial cells contributes to neuronal excitotoxicity. J Cell Biol 2005; 171:1001–1012.

    Article  CAS  PubMed  Google Scholar 

  227. Hassel B, Tessler S, Faull RL et al. Glutamate uptake is reduced in prefrontal cortex in Huntington’s disease. Neurochem Res 2007.

    Google Scholar 

  228. Zeron MM, Hansson O, Chen N et al. Increased sensitivity to N-methyl-D-aspartate receptor-mediated excitotoxicity in a mouse model of Huntington’s disease. Neuron 2002; 33:849–860.

    Article  CAS  PubMed  Google Scholar 

  229. Starling AJ, Andre VM, Cepeda C et al. Alterations in N-methyl-D-aspartate receptor sensitivity and magnesium blockade occur early in development in the R6/2 mouse model of Huntington’s disease. J Neurosci Res 2005; 82:377–386.

    Article  CAS  PubMed  Google Scholar 

  230. Panov AV, Burke JR, Strittmatter WJ et al. In vitro effects of polyglutamine tracts on Ca2+-dependent depolarization of rat and human mitochondria: relevance to Huntington’s disease. Archives of Biochemistry and Biophysics 2003; 410:1–6.

    Article  CAS  PubMed  Google Scholar 

  231. Panov AV, Gutekunst CA, Leavitt BR et al. Early mitochondrial calcium defects in Huntington’s disease are a direct effect of polyglutamines. Nature Neuroscience 2002; 5:731–736.

    CAS  PubMed  Google Scholar 

  232. Oliveira JM, Jekabsons MB, Chen S et al. Mitochondrial dysfunction in Huntington’s disease: the bioenergetics of isolated and in situ mitochondria from transgenic mice. J Neurochem 2007; 101:241–249.

    Article  CAS  PubMed  Google Scholar 

  233. Sun Y, Savanenin A, Reddy PH et al. Polyglutamine-expanded huntingtin promotes sensitization of N-methyl-D-aspartate receptors via postsynaptic density 95. J Biol Chem 2001; 276:24713–24718.

    Article  CAS  PubMed  Google Scholar 

  234. Roche KW, Standley S, McCallum J et al. Molecular determinants of NMDA receptor internalization. Nature Neuroscience 2001; 4:794–802.

    Article  CAS  PubMed  Google Scholar 

  235. Fan MM, Fernandes HB, Zhang LY et al. Altered NMDA receptor trafficking in a yeast artificial chromosome transgenic mouse model of Huntington’s disease. J Neurosci 2007; 27:3768–3779.

    Article  CAS  PubMed  Google Scholar 

  236. Metzler M, Gan L, Wong TP et al. NMDA receptor function and NMDA receptor-dependent phosphorylation of huntingtin is altered by the endocytic protein HIP1. J Neurosci 2007; 27:2298–2308.

    Article  CAS  PubMed  Google Scholar 

  237. Tang TS, Tu H, Chan EY et al. Huntingtin and huntingtin-associated protein 1 influence neuronal calcium signaling mediated by inositol-(1,4,5) triphosphate receptor type 1. Neuron 2003; 39:227–239.

    Article  CAS  PubMed  Google Scholar 

  238. Tang TS, Tu H, Orban PC et al. HAP1 facilitates effects of mutant huntingtin on inositol 1,4,5-trisphosphate-induced Ca release in primary culture of striatal medium spiny neurons. Eur J Neurosci 2004; 20:1779–1787.

    Article  PubMed  Google Scholar 

  239. Tang TS, Guo C, Wang H et al. Neuroprotective effects of inositol 1,4,5-trisphosphate receptor C-terminal fragment in a Huntington’s disease mouse model. J Neurosci 2009; 29:1257–1266.

    Article  CAS  PubMed  Google Scholar 

  240. Landwehrmeyer GB, Dubois B, de Yebenes JG et al. Riluzole in Huntington’s disease: a 3-year, randomized controlled study. Ann Neurol 2007; 62:262–272.

    Article  CAS  PubMed  Google Scholar 

  241. O’Suilleabhain P, Dewey RB Jr. A randomized trial of amantadine in Huntington disease. Arch Neurol 2003; 60:996–998.

    Article  PubMed  Google Scholar 

  242. Ondo WG, Mejia NI, Hunter CB. A pilot study of the clinical efficacy and safety of memantine for Huntington’s disease. Parkinsonism Relat Disord 2007; 13:453–454.

    Article  PubMed  Google Scholar 

  243. Shen YC. Lamotrigine in motor and mood symptoms of Huntington’s disease. World J Biol Psychiatry 2008; 9:147–149.

    Article  PubMed  Google Scholar 

  244. Reynolds DS, Carter RJ, Morton AJ. Dopamine modulates the susceptibility of striatal neurons to 3-nitropropionic acid in the rat model of Huntington’s disease. J Neurosci 1998; 18:10116–10127.

    CAS  PubMed  Google Scholar 

  245. Benchoua A, Trioulier Y, Diguet E et al. Dopamine determines the vulnerability of striatal neurons to the N-terminal fragment of mutant huntingtin through the regulation of mitochondrial complex II. Hum Mol Genet 2008; 17:1446–1456.

    Article  CAS  PubMed  Google Scholar 

  246. Cyr M, Sotnikova TD, Gainetdinov RR et al. Dopamine enhances motor and neuropathological consequences of polyglutamine expanded huntingtin. FASEB J 2006; 20:2541–2543.

    Article  CAS  PubMed  Google Scholar 

  247. Charvin D, Vanhoutte P, Pages C et al. Unraveling a role for dopamine in Huntington’s disease: the dual role of reactive oxygen species and D2 receptor stimulation. Proc Natl Acad Sci USA 2005; 102:12218–12223.

    Article  CAS  PubMed  Google Scholar 

  248. Tang TS, Chen X, Liu J et al. Dopaminergic signaling and striatal neurodegeneration in Huntington’s disease. J Neurosci 2007; 27:7899–7910.

    Article  CAS  PubMed  Google Scholar 

  249. Charvin D, Roze E, Perrin V et al. Haloperidol protects striatal neurons from dysfunction induced by mutated huntingtin in vivo. Neurobiol Dis 2008; 29:22–29.

    Article  CAS  PubMed  Google Scholar 

  250. Jakel RJ, Maragos WF. Neuronal cell death in Huntington’s disease: a potential role for dopamine. Trends Neurosci 2000; 23:239–245.

    Article  CAS  PubMed  Google Scholar 

  251. Petersen AA, Larsen KE, Behr GG et al. Brain-derived neurotrophic factor inhibits apoptosis and dopamine-induced free radical production in striatal neurons but does not prevent cell death. Brain Res Bull 2001; 56:331–335.

    Article  CAS  Google Scholar 

  252. Garcia M, Charvin D, Caboche J. Expanded huntingtin activates the c-Jun terminal kinase/c-Jun pathway prior to aggregate formation in striatal neurons in culture. Neuroscience 2004; 127:859–870.

    Article  CAS  PubMed  Google Scholar 

  253. Apostol BL, Simmons DA, Zuccato C et al. CEP-1347 reduces mutant huntingtin-associated neurotoxicity and restores BDNF levels in R6/2 mice. Mol Cell Neurosci 2008; 39:8–20.

    Article  CAS  PubMed  Google Scholar 

  254. Zhang H, Li Q, Graham RK et al. Full length mutant huntingtin is required for altered Ca2??signaling and apoptosis of striatal neurons in the YAC mouse model of Huntington’s disease. Neurobiol Dis 2008; 31:80–88.

    Article  CAS  PubMed  Google Scholar 

  255. Cepeda C, Ariano MA, Calvert CR et al. NMDA receptor function in mouse models of Huntington disease. J Neurosci Res 2001; 66:525–539.

    Article  CAS  PubMed  Google Scholar 

  256. Zeron MM, Fernandes HB, Krebs C et al. Potentiation of NMDA receptor-mediated excitotoxicity linked with intrinsic apoptotic pathway in YAC transgenic mouse model of Huntington’s disease. Mol Cell Neurosci 2004; 25:469–479.

    Article  CAS  PubMed  Google Scholar 

  257. Paoletti P, Vila I, Rife M et al. Dopaminergic and glutamatergic signaling crosstalk in Huntington’s disease neurodegeneration: the role of p25/cyclin-dependent kinase 5. J Neurosci 2008; 28:10090–10101.

    Article  CAS  PubMed  Google Scholar 

  258. Anne SL, Saudou F, Humbert S. Phosphorylation of huntingtin by cyclin-dependent kinase 5 is induced by DNA damage and regulates wild-type and mutant huntingtin toxicity in neurons. J Neurosci 2007; 27:7318–7328.

    Article  CAS  PubMed  Google Scholar 

  259. Luo S, Vacher C, Davies JE et al. Cdk5 phosphorylation of huntingtin reduces its cleavage by caspases: implications for mutant huntingtin toxicity. J Cell Biol 2005; 169:647–656.

    Article  CAS  PubMed  Google Scholar 

  260. Kaminosono S, Saito T, Oyama F et al. Suppression of mutant Huntingtin aggregate formation by Cdk5/ p35 through the effect on microtubule stability. J Neurosci 2008; 28:8747–8755.

    Article  CAS  PubMed  Google Scholar 

  261. de Almeida LP, Ross CA, Zala D et al. Lentiviral-mediated delivery of mutant huntingtin in the striatum of rats induces a selective neuropathology modulated by polyglutamine repeat size, huntingtin expression levels and protein length. J Neurosci 2002; 22:3473–3483.

    PubMed  Google Scholar 

  262. Gauthier LR, Charrin BC, Borrell-Pages M et al. Huntingtin controls neurotrophic support and survival of neurons by enhancing BDNF vesicular transport along microtubules. Cell 2004; 118:127–138.

    Article  CAS  PubMed  Google Scholar 

  263. Sinadinos C, Burbidge-King T, Soh D et al. Live axonal transport disruption by mutant huntingtin fragments in Drosophila motor neuron axons. Neurobiol Dis 2009.

    Google Scholar 

  264. Szebenyi G, Morfini GA, Babcock A et al. Neuropathogenic forms of huntingtin and androgen receptor inhibit fast axonal transport. Neuron 2003; 40:41–52.

    Article  CAS  PubMed  Google Scholar 

  265. Borrell-Pages M, Zala D, Humbert S et al. Huntington’s disease: from huntingtin function and dysfunction to therapeutic strategies. Cell Mol Life Sci 2006; 63:2642–2660.

    Article  CAS  PubMed  Google Scholar 

  266. Smith R, Brundin P, Li JY. Synaptic dysfunction in Huntington’s disease: a new perspective. Cell Mol Life Sci 2005; 62:1901–1912.

    Article  CAS  PubMed  Google Scholar 

  267. Colin E, Zala D, Liot G et al. Huntingtin phosphorylation acts as a molecular switch for anterograde/ retrograde transport in neurons. EMBO J 2008; 27:2124–2134.

    Article  CAS  PubMed  Google Scholar 

  268. Dompierre JP, Godin JD, Charrin BC et al. Histone deacetylase 6 inhibition compensates for the transport deficit in Huntington’s disease by increasing tubulin acetylation. J Neurosci 2007; 27:3571–3583.

    Article  CAS  PubMed  Google Scholar 

  269. Borrell-Pages M, Canals JM, Cordelieres FP et al. Cystamine and cysteamine increase brain levels of BDNF in Huntington disease via HSJ1b and transglutaminase. J Clin Invest 2006; 116:1410–1424.

    Article  CAS  PubMed  Google Scholar 

  270. del Toro D, Canals JM, Gines S et al. Mutant huntingtin impairs the postGolgi trafficking of brain-derived neurotrophic factor but not its Val66Met polymorphism. J Neurosci 2006; 26:12748–12757.

    Article  PubMed  CAS  Google Scholar 

  271. Yanai A, Huang K, Kang R et al. Palmitoylation of huntingtin by HIP14 is essential for its trafficking and function. Nature Neuroscience 2006; 9:824–831.

    Article  CAS  PubMed  Google Scholar 

  272. Ohyama T, Verstreken P, Ly CV et al. Huntingtin-interacting protein 14, a palmitoyl transferase required for exocytosis and targeting of CSP to synaptic vesicles. J Cell Biol 2007; 179:1481–1496.

    Article  CAS  PubMed  Google Scholar 

  273. Romero E, Cha GH, Verstreken P et al. Suppression of neurodegeneration and increased neurotransmission caused by expanded full-length huntingtin accumulating in the cytoplasm. Neuron 2008; 57:27–40.

    Article  CAS  PubMed  Google Scholar 

  274. Hamilton JM, Wolfson T, Peavy GM et al. Rate and correlates of weight change in Huntington’s disease. Journal of Neurology, Neurosurgery and Psychiatry 2004; 75:209–212.

    Article  CAS  Google Scholar 

  275. Djousse L, Knowlton B, Cupples LA et al. Weight loss in early stage of Huntington’s disease. Neurology 2002; 59:1325–1330.

    CAS  PubMed  Google Scholar 

  276. Arenas J, Campos Y, Ribacoba R et al. Complex I defect in muscle from patients with Huntington’s disease. Ann Neurol 1998; 43:397–400.

    Article  CAS  PubMed  Google Scholar 

  277. Turner C, Cooper JM, Schapira AH. Clinical correlates of mitochondrial function in Huntington’s disease muscle. Mov Disord 2007.

    Google Scholar 

  278. Jenkins BG, Rosas HD, Chen YC et al. 1H NMR spectroscopy studies of Huntington’s disease: correlations with CAG repeat numbers. Neurology 1998; 50:1357–1365.

    CAS  PubMed  Google Scholar 

  279. Mochel F, Charles P, Seguin F et al. Early energy deficit in Huntington disease: identification of a plasma biomarker traceable during disease progression. PLoS ONE 2007; 2:e647.

    Article  PubMed  CAS  Google Scholar 

  280. Beal MF, Brouillet E, Jenkins BG et al. Neurochemical and histologic characterization of striatal excitotoxic lesions produced by the mitochondrial toxin 3-nitropropionic acid. J Neurosci 1993; 13:4181–4192.

    CAS  PubMed  Google Scholar 

  281. Benchoua A, Trioulier Y, Zala D et al. Involvement of mitochondrial complex II defects in neuronal death produced by N-terminus fragment of mutated huntingtin. Mol Biol Cell 2006; 17:1652–1663.

    Article  CAS  PubMed  Google Scholar 

  282. Fernandes HB, Baimbridge KG, Church J et al. Mitochondrial sensitivity and altered calcium handling underlie enhanced NMDA-induced apoptosis in YAC128 model of Huntington’s disease. J Neurosci 2007; 27:13614–13623.

    Article  CAS  PubMed  Google Scholar 

  283. Cui L, Jeong H, Borovecki F et al. Transcriptional repression of PGC-1alpha by mutant huntingtin leads to mitochondrial dysfunction and neurodegeneration. Cell 2006; 127:59–69.

    Article  CAS  PubMed  Google Scholar 

  284. Weydt P, Pineda VV, Torrence AE et al. Thermoregulatory and metabolic defects in Huntington’s disease transgenic mice implicate PGC-1alpha in Huntington’s disease neurodegeneration. Cell Metab 2006; 4:349–362.

    Article  CAS  PubMed  Google Scholar 

  285. Chang DT, Rintoul GL, Pandipati S et al. Mutant huntingtin aggregates impair mitochondrial movement and trafficking in cortical neurons. Neurobiol Dis 2006; 22:388–400.

    Article  CAS  PubMed  Google Scholar 

  286. Orr AL, Li S, Wang CE et al. N-terminal mutant huntingtin associates with mitochondria and impairs mitochondrial trafficking. J Neurosci 2008; 28:2783–2792.

    Article  CAS  PubMed  Google Scholar 

  287. Wang H, Lim PJ, Karbowski M et al. Effects of overexpression of huntingtin proteins on mitochondrial integrity. Hum Mol Genet 2009; 18:737–752.

    Article  CAS  PubMed  Google Scholar 

  288. Lesort M, Chun W, Johnson GV et al. Tissue transglutaminase is increased in Huntington’s disease brain. J Neurochem 1999; 73:2018–2027.

    CAS  PubMed  Google Scholar 

  289. Karpuj MV, Garren H, Slunt H et al. Transglutaminase aggregates huntingtin into nonamyloidogenic polymers and its enzymatic activity increases in Huntington’s disease brain nuclei. Proc Natl Acad Sci USA 1999; 96:7388–7393.

    Article  CAS  PubMed  Google Scholar 

  290. Dedeoglu A, Kubilus JK, Jeitner TM et al. Therapeutic effects of cystamine in a murine model of Huntington’s disease. J Neurosci 2002; 22:8942–8950.

    CAS  PubMed  Google Scholar 

  291. Battaglia G, Farrace MG, Mastroberardino PG et al. Transglutaminase 2 ablation leads to defective function of mitochondrial respiratory complex I affecting neuronal vulnerability in experimental models of extrapyramidal disorders. J Neurochem 2007; 100:36–49.

    Article  CAS  PubMed  Google Scholar 

  292. Mastroberardino PG, Iannicola C, Nardacci R et al ‘Tissue’ transglutaminase ablation reduces neuronal death and prolongs survival in a mouse model of Huntington’s disease. Cell Death Differ 2002; 9:873–880.

    Article  CAS  PubMed  Google Scholar 

  293. Bailey CD, Johnson GV. Tissue transglutaminase contributes to disease progression in the R6/2 Huntington’s disease mouse model via aggregate-independent mechanisms. J Neurochem 2005; 92:83–92.

    Article  CAS  PubMed  Google Scholar 

  294. Lee JM, Ivanova EV, Seong IS et al. Unbiased gene expression analysis implicates the huntingtin polyglutamine tract in extra-mitochondrial energy metabolism. PLoS Genet 2007; 3:e135.

    Article  PubMed  CAS  Google Scholar 

  295. Schilling G, Coonfield ML, Ross CA et al. Coenzyme Q10 and remacemide hydrochloride ameliorate motor deficits in a Huntington’s disease transgenic mouse model. Neurosci Lett 2001; 315:149–153.

    Article  CAS  PubMed  Google Scholar 

  296. Smith KM, Matson S, Matson WR et al. Dose ranging and efficacy study of high-dose coenzyme Q10 formulations in Huntington’s disease mice. Biochim Biophys Acta 2006; 1762:616–626.

    CAS  PubMed  Google Scholar 

  297. Ferrante RJ, Andreassen OA, Dedeoglu A et al. Therapeutic effects of coenzyme Q10 and remacemide in transgenic mouse models of Huntington’s disease. J Neurosci 2002; 22:1592–1599.

    CAS  PubMed  Google Scholar 

  298. Dedeoglu A, Kubilus JK, Yang L et al. Creatine therapy provides neuroprotection after onset of clinical symptoms in Huntington’s disease transgenic mice. J Neurochem 2003; 85:1359–1367.

    Article  CAS  PubMed  Google Scholar 

  299. Andreassen OA, Dedeoglu A, Ferrante RJ et al. Creatine increase survival and delays motor symptoms in a transgenic animal model of Huntington’s disease. Neurobiol Dis 2001; 8:479–491.

    Article  CAS  PubMed  Google Scholar 

  300. A randomized, placebo-controlled trial of coenzyme Q10 and remacemide in Huntington’s disease. Neurology 2001; 57:397–404.

    Google Scholar 

  301. Puri BK, Leavitt BR, Hayden MR et al. Ethyl-EPA in Huntington disease: a double-blind, randomized, placebo-controlled trial. Neurology 2005; 65:286–292.

    Article  CAS  PubMed  Google Scholar 

  302. Verbessem P, Lemiere J, Eijnde BO et al. Creatine supplementation in Huntington’s disease: a placebo-controlled pilot trial. Neurology 2003; 61:925–930.

    CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Emmanuel Roze .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2010 Landes Bioscience and Springer Science+Business Media

About this chapter

Cite this chapter

Roze, E., Bonnet, C., Betuing, S., Caboche, J. (2010). Huntington’s Disease. In: Ahmad, S.I. (eds) Diseases of DNA Repair. Advances in Experimental Medicine and Biology, vol 685. Springer, New York, NY. https://doi.org/10.1007/978-1-4419-6448-9_5

Download citation

Publish with us

Policies and ethics