Skip to main content
Log in

Role of Inositol 1,4,5-Trishosphate Receptors in Pathogenesis of Huntington’s Disease and Spinocerebellar Ataxias

  • Original Paper
  • Published:
Neurochemical Research Aims and scope Submit manuscript

Abstract

Huntington’s disease (HD) and spinocerebellar ataxias (SCAs) are autosomal-dominant neurodegenerative disorders. HD is caused by polyglutamine (polyQ) expansion in the amino-terminal region of a protein huntingtin (Htt) and primarily affects medium spiny striatal neurons (MSN). Many SCAs are caused by polyQ-expansion in ataxin proteins and primarily affect cerebellar Purkinje cells. The reasons for neuronal dysfunction and death in HD and SCAs remain poorly understood and no cure is available for the patients. Our laboratory discovered that mutant huntingtin, ataxin-2 and ataxin-3 proteins specifically bind to the carboxy-terminal region of the type 1 inositol 1,4,5-trisphosphate receptor (IP3R1), an intracellular Ca2+ release channel. Moreover, we found that association of mutant huntingtin or ataxins with IP3R1 causes sensitization of IP3R1 to activation by IP3 in planar lipid bilayers and in neuronal cells. These results suggested that deranged neuronal Ca2+ signaling might play an important role in pathogenesis of HD, SCA2 and SCA3. In support of this idea, we demonstrated a connection between abnormal Ca2+ signaling and neuronal cell death in experiments with HD, SCA2 and SCA3 transgenic mouse models. Additional data in the literature indicate that abnormal neuronal Ca2+ signaling may also play an important role in pathogenesis of SCAl, SCA5, SCA6, SCA14 and SCA15/16. Based on these results I propose that IP3R and other Ca2+ signaling proteins should be considered as potential therapeutic targets for treatment of HD and SCAs.

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

Access this article

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

Instant access to the full article PDF.

Institutional subscriptions

Similar content being viewed by others

References

  1. Vonsattel JP, DiFiglia M (1998) Huntington disease. J Neuropathol Exp Neurol 57:369–384

    PubMed  CAS  Google Scholar 

  2. Vonsattel JP, Myers RH, Stevens TJ, Ferrante RJ, Bird ED, Richardson EP Jr (1985) Neuropathological classification of Huntington’s disease. J Neuropathol Exp Neurol 44:559–577

    PubMed  CAS  Google Scholar 

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

    Google Scholar 

  4. Langbehn DR, Brinkman RR, Falush D, Paulsen JS, Hayden MR (2004) A new model for prediction of the age of onset and penetrance for Huntington’s disease based on CAG length. Clin Genet 65:267–277

    PubMed  CAS  Google Scholar 

  5. Li SH, Schilling G, Young WS III, Li XJ, Margolis RL, Stine OC, Wagster MV, Abbott MH, Franz ML, Ranen NG et al (1993) Huntington’s disease gene (IT15) is widely expressed in human and rat tissues. Neuron 11:985–993

    PubMed  CAS  Google Scholar 

  6. Strong TV, Tagle DA, Valdes JM, Elmer LW, Boehm K, Swaroop M, Kaatz KW, Collins FS, Albin RL (1993) Widespread expression of the human and rat Huntington’s disease gene in brain and nonneural tissues. Nat Genet 5:259–265

    PubMed  CAS  Google Scholar 

  7. Sharp AH, Loev SJ, Schilling G, Li SH, Li XJ, Bao J, Wagster MV, Kotzuk JA, Steiner JP, Lo A et al (1995) Widespread expression of Huntington’s disease gene (IT15) protein product. Neuron 14:1065–1074

    PubMed  CAS  Google Scholar 

  8. Nasir J, Floresco SB, O’Kusky JR, Diewert VM, Richman JM, Zeisler J, Borowski A, Marth JD, Phillips AG, Hayden MR (1995) Targeted disruption of the Huntington’s disease gene results in embryonic lethality and behavioral and morphological changes in heterozygotes. Cell 81:811–823

    PubMed  CAS  Google Scholar 

  9. Duyao MP, Auerbach AB, Ryan A, Persichetti F, Barnes GT, McNeil SM, Ge P, Vonsattel JP, Gusella JF, Joyner AL et al (1995) Inactivation of the mouse Huntington’s disease gene homolog Hdh. Science 269:407–410

    PubMed  CAS  Google Scholar 

  10. MacDonald ME (2003) Huntingtin: alive and well and working in middle management. Sci STKE 2003: pe48

  11. Heng MY, Detloff PJ, Albin RL (2008) Rodent genetic models of Huntington disease. Neurobiol Dis 32:1–9

    PubMed  CAS  Google Scholar 

  12. Andre VM, Cepeda C, Levine MS (2010) Dopamine and glutamate in Huntington’s disease: a balancing act. CNS Neurosci Ther 16:163–178

    PubMed  CAS  Google Scholar 

  13. Milnerwood AJ, Raymond LA (2010) Early synaptic pathophysiology in neurodegeneration: insights from Huntington’s disease. Trends Neurosci 33:513–523

    PubMed  CAS  Google Scholar 

  14. Miller BR, Bezprozvanny I (2010) Corticostriatal circuit dysfunction in Huntington’s disease: intersection of glutamate, dopamine, and calcium. Future Neurol 5:735–756

    CAS  Google Scholar 

  15. Filla A, De Michele G, Santoro L, Calabrese O, Castaldo I, Giuffrida S, Restivo D, Serlenga L, Condorelli DF, Bonuccelli U, Scala R, Coppola G, Caruso G, Cocozza S (1999) Spinocerebellar ataxia type 2 in southern Italy: a clinical and molecular study of 30 families. J Neurol 246:467–471

    PubMed  CAS  Google Scholar 

  16. Schols L, Bauer P, Schmidt T, Schulte T, Riess O (2004) Autosomal dominant cerebellar ataxias: clinical features, genetics, and pathogenesis. Lancet Neurol 3:291–304

    PubMed  Google Scholar 

  17. Lastres-Becker I, Rub U, Auburger G (2008) Spinocerebellar ataxia 2 (SCA2). Cerebellum 7:115–124

    PubMed  CAS  Google Scholar 

  18. Hara K, Shiga A, Nozaki H, Mitsui J, Takahashi Y, Ishiguro H, Yomono H, Kurisaki H, Goto J, Ikeuchi T, Tsuji S, Nishizawa M, Onodera O (2008) Total deletion and a missense mutation of ITPR1 in Japanese SCA15 families. Neurology 71:547–551

    PubMed  CAS  Google Scholar 

  19. Iwaki A, Kawano Y, Miura S, Shibata H, Matsuse D, Li W, Furuya H, Ohyagi Y, Taniwaki T, Kira J, Fukumaki Y (2008) Heterozygous deletion of ITPR1, but not SUMF1, in spinocerebellar ataxia type 16. J Med Genet 45:32–35

    PubMed  CAS  Google Scholar 

  20. Paulson HL (2009) The spinocerebellar ataxias. J Neuroophthalmol 29:227–237

    PubMed  Google Scholar 

  21. Matilla-Duenas A, Sanchez I, Corral-Juan M, Davalos A, Alvarez R, Latorre P (2010) Cellular and molecular pathways triggering neurodegeneration in the spinocerebellar ataxias. Cerebellum 9:148–166

    PubMed  CAS  Google Scholar 

  22. Carlson KM, Andresen JM, Orr HT (2009) Emerging pathogenic pathways in the spinocerebellar ataxias. Curr Opin Genet Dev 19:247–253

    PubMed  CAS  Google Scholar 

  23. Bezprozvanny I, Klockgether T (2010) Therapeutic prospects for spinocerebellar ataxia type 2 and 3. Drugs Future 34: 991–999

    Google Scholar 

  24. Kasumu A, Bezprozvanny I (2010) Deranged calcium signaling in purkinje cells and pathogenesis in spinocerebellar ataxia 2 (SCA2) and other ataxias. Cerebellum [Epub ahead of print]

  25. Pirker W, Back C, Gerschlager W, Laccone F, Alesch F (2003) Chronic thalamic stimulation in a patient with spinocerebellar ataxia type 2. Mov Disord 18:222–225

    PubMed  Google Scholar 

  26. Berridge MJ (1998) Neuronal calcium signaling. Neuron 21:13–26

    PubMed  CAS  Google Scholar 

  27. Furuichi T, Kohda K, Miyawaki A, Mikoshiba K (1994) Intracellular channels. Curr Opinion Neurobiol 4:294–303

    CAS  Google Scholar 

  28. Matsumoto M, Nakagawa T, Inoue T, Nagata E, Tanaka K, Takano H, Minowa O, Kuno J, Sakakibara S, Yamada M, Yoneshima H, Miyawaki A, Fukuuchi Y, Furuichi T, Okano H, Mikoshiba K, Noda T (1996) Ataxia and epileptic seizures in mice lacking type 1 inositol 1,4,5-trisphosphate receptor. Nature 379:168–171

    PubMed  CAS  Google Scholar 

  29. Street VA, Bosma MM, Demas VP, Regan MR, Lin DD, Robinson LC, Agnew WS, Tempel BL (1997) The type 1 inositol 1, 4, 5-trisphosphate receptor gene is altered in the opisthotonos mouse. J Neurosci 17:635–645

    PubMed  CAS  Google Scholar 

  30. Bezprozvanny I (2009) Calcium signaling and neurodegenerative diseases. Trends Mol Med 15:89–100

    PubMed  CAS  Google Scholar 

  31. Bezprozvanny I, Hayden MR (2004) Deranged neuronal calcium signaling and Huntington disease. Biochem Biophys Res Commun 322:1310–1317

    PubMed  CAS  Google Scholar 

  32. Tang TS, Tu H, Chan EY, Maximov A, Wang Z, Wellington CL, Hayden MR, Bezprozvanny I (2003) Huntingtin and huntingtin-associated protein 1 influence neuronal calcium signaling mediated by inositol-(1,4,5) triphosphate receptor type 1. Neuron 39:227–239

    PubMed  CAS  Google Scholar 

  33. Kaltenbach LS, Romero E, Becklin RR, Chettier R, Bell R, Phansalkar A, Strand A, Torcassi C, Savage J, Hurlburt A, Cha GH, Ukani L, Chepanoske CL, Zhen Y, Sahasrabudhe S, Olson J, Kurschner C, Ellerby LM, Peltier JM, Botas J, Hughes RE (2007) Huntingtin interacting proteins are genetic modifiers of neurodegeneration. PLoS Genet 3: e82

  34. Tang TS, Slow E, Lupu V, Stavrovskaya IG, Sugimori M, Llinas R, Kristal BS, Hayden MR, Bezprozvanny I (2005) Disturbed Ca2+ signaling and apoptosis of medium spiny neurons in Huntington’s disease. Proc Natl Acad Sci USA 102:2602–2607

    PubMed  CAS  Google Scholar 

  35. Zhang H, Li Q, Graham RK, Slow E, Hayden MR, Bezprozvanny I (2008) 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 31:80–88

    PubMed  CAS  Google Scholar 

  36. Tang TS, Guo C, Wang H, Chen X, Bezprozvanny I (2009) Neuroprotective effects of inositol 1,4,5-trisphosphate receptor C-terminal fragment in a Huntington’s disease mouse model. J Neurosci 29:1257–1266

    PubMed  CAS  Google Scholar 

  37. Mao L, Wang JQ (2001) Upregulation of preprodynorphin and preproenkephalin mRNA expression by selective activation of group I metabotropic glutamate receptors in characterized primary cultures of rat striatal neurons. Brain Res Mol Brain Res 86:125–137

    PubMed  CAS  Google Scholar 

  38. Mao L, Wang JQ (2002) Glutamate cascade to cAMP response element-binding protein phosphorylation in cultured striatal neurons through calcium-coupled group I metabotropic glutamate receptors. Mol Pharmacol 62:473–484

    PubMed  CAS  Google Scholar 

  39. Tallaksen-Greene SJ, Kaatz KW, Romano C, Albin RL (1998) Localization of mGluR1a-like immunoreactivity and mGluR5-like immunoreactivity in identified populations of striatal neurons. Brain Res 780:210–217

    PubMed  CAS  Google Scholar 

  40. Kerner JA, Standaert DG, Penney JB Jr, Young AB, Landwehrmeyer GB (1997) Expression of group one metabotropic glutamate receptor subunit mRNAs in neurochemically identified neurons in the rat neostriatum, neocortex, and hippocampus. Brain Res Mol Brain Res 48:259–269

    PubMed  CAS  Google Scholar 

  41. Testa CM, Standaert DG, Landwehrmeyer GB, Penney JB Jr, Young AB (1995) Differential expression of mGluR5 metabotropic glutamate receptor mRNA by rat striatal neurons. J Comp Neurol 354:241–252

    PubMed  CAS  Google Scholar 

  42. Cross AJ, Crow TJ, Johnson JA, Dawson JM, Peters TJ (1985) Loss of endoplasmic reticulum-associated enzymes in affected brain regions in Huntington’s disease and Alzheimer-type dementia. J Neurol Sci 71:137–143

    PubMed  CAS  Google Scholar 

  43. Higo T, Hamada K, Hisatsune C, Nukina N, Hashikawa T, Hattori M, Nakamura T, Mikoshiba K (2010) Mechanism of ER stress-induced brain damage by IP3 receptor. Neuron 68(5):865–878

    PubMed  CAS  Google Scholar 

  44. Landwehrmeyer GB, Standaert DG, Testa CM, Penney JB Jr, Young AB (1995) NMDA receptor subunit mRNA expression by projection neurons and interneurons in rat striatum. J Neurosci 15:5297–5307

    PubMed  CAS  Google Scholar 

  45. Monyer H, Burnashev N, Laurie DJ, Sakmann B, Seeburg PH (1994) Developmental and regional expression in the rat brain and functional properties of four NMDA receptors. Neuron 12:529–540

    PubMed  CAS  Google Scholar 

  46. Portera-Cailliau C, Price DL, Martin LJ (1996) N-methyl-D-aspartate receptor proteins NR2A and NR2B are differentially distributed in the developing rat central nervous system as revealed by subunit-specific antibodies. J Neurochem 66:692–700

    PubMed  CAS  Google Scholar 

  47. Chen N, Luo T, Wellington C, Metzler M, McCutcheon K, Hayden MR, Raymond LA (1999) Subtype-specific enhancement of NMDA receptor currents by mutant huntingtin. J Neurochem 72:1890–1898

    PubMed  CAS  Google Scholar 

  48. Zeron MM, Chen N, Moshaver A, Lee AT, Wellington CL, Hayden MR, Raymond LA (2001) Mutant huntingtin enhances excitotoxic cell death. Mol Cell Neurosci 17:41–53

    PubMed  CAS  Google Scholar 

  49. Zeron MM, Hansson O, Chen N, Wellington CL, Leavitt BR, Brundin P, Hayden MR, Raymond LA (2002) Increased sensitivity to N-methyl-D-aspartate receptor-mediated excitotoxicity in a mouse model of Huntington’s disease. Neuron 33:849–860

    PubMed  CAS  Google Scholar 

  50. Ali NJ, Levine MS (2006) Changes in expression of N-methyl-D-aspartate receptor subunits occur early in the R6/2 mouse model of Huntington’s disease. Dev Neurosci 28:230–238

    PubMed  CAS  Google Scholar 

  51. Li JH, Wang YH, Wolfe BB, Krueger KE, Corsi L, Stocca G, Vicini S (1998) Developmental changes in localization of NMDA receptor subunits in primary cultures of cortical neurons. Eur J Neurosci 10:1704–1715

    PubMed  CAS  Google Scholar 

  52. Stocca G, Vicini S (1998) Increased contribution of NR2A subunit to synaptic NMDA receptors in developing rat cortical neurons. J Physiol 507(Pt 1):13–24

    PubMed  CAS  Google Scholar 

  53. Tovar KR, Westbrook GL (1999) The incorporation of NMDA receptors with a distinct subunit composition at nascent hippocampal synapses in vitro. J Neurosci 19:4180–4188

    PubMed  CAS  Google Scholar 

  54. Papadia S, Hardingham GE (2007) The dichotomy of NMDA receptor signaling. Neuroscientist 13:572–579

    PubMed  CAS  Google Scholar 

  55. Vanhoutte P, Bading H (2003) Opposing roles of synaptic and extrasynaptic NMDA receptors in neuronal calcium signalling and BDNF gene regulation. Curr Opin Neurobiol 13:366–371

    PubMed  CAS  Google Scholar 

  56. Gouix E, Leveille F, Nicole O, Melon C, Had-Aissouni L, Buisson A (2009) Reverse glial glutamate uptake triggers neuronal cell death through extrasynaptic NMDA receptor activation. Mol Cell Neurosci 40:463–473

    PubMed  CAS  Google Scholar 

  57. Hardingham GE, Fukunaga Y, Bading H (2002) Extrasynaptic NMDARs oppose synaptic NMDARs by triggering CREB shut-off and cell death pathways. Nat Neurosci 5:405–414

    PubMed  CAS  Google Scholar 

  58. Papadia S, Soriano FX, Leveille F, Martel MA, Dakin KA, Hansen HH, Kaindl A, Sifringer M, Fowler J, Stefovska V, McKenzie G, Craigon M, Corriveau R, Ghazal P, Horsburgh K, Yankner BA, Wyllie DJ, Ikonomidou C, Hardingham GE (2008) Synaptic NMDA receptor activity boosts intrinsic antioxidant defenses. Nat Neurosci 11:476–487

    PubMed  CAS  Google Scholar 

  59. Milnerwood AJ, Gladding CM, Pouladi MA, Kaufman AM, Hines RM, Boyd JD, Ko RW, Vasuta OC, Graham RK, Hayden MR, Murphy TH, Raymond LA (2010) Early increase in extrasynaptic NMDA receptor signaling and expression contributes to phenotype onset in Huntington’s disease mice. Neuron 65:178–190

    PubMed  CAS  Google Scholar 

  60. Li L, Murphy TH, Hayden MR, Raymond LA (2004) Enhanced striatal NR2B-containing N-methyl-D-aspartate receptor-mediated synaptic currents in a mouse model of Huntington disease. J Neurophysiol 92:2738–2746

    PubMed  CAS  Google Scholar 

  61. Heng MY, Detloff PJ, Wang PL, Tsien JZ, Albin RL (2009) In vivo evidence for NMDA receptor-mediated excitotoxicity in a murine genetic model of Huntington disease. J Neurosci 29:3200–3205

    PubMed  CAS  Google Scholar 

  62. Milnerwood AJ, Raymond LA (2006) Synaptic abnormalities associated with Huntington’s disease. In: AE-HaA Dityatev (ed) In molecular mechanisms of synaotogenesis. Springer, New York, pp 457–469

    Google Scholar 

  63. Sun Y, Savanenin A, Reddy PH, Liu YF (2001) Polyglutamine-expanded huntingtin promotes sensitization of N-methyl-D- aspartate receptors via post-synaptic density 95. J Biol Chem 276:24713–24718

    PubMed  CAS  Google Scholar 

  64. Song C, Zhang Y, Parsons CG, Liu YF (2003) Expression of polyglutamine-expanded huntingtin induces tyrosine phosphorylation of N-methyl-D-aspartate receptors. J Biol Chem 278:33364–33369

    PubMed  CAS  Google Scholar 

  65. Fan J, Cowan CM, Zhang LY, Hayden MR, Raymond LA (2009) Interaction of postsynaptic density protein-95 with NMDA receptors influences excitotoxicity in the yeast artificial chromosome mouse model of Huntington’s disease. J Neurosci 29:10928–10938

    PubMed  CAS  Google Scholar 

  66. Fan MM, Fernandes HB, Zhang LY, Hayden MR, Raymond LA (2007) Altered NMDA receptor trafficking in a yeast artificial chromosome transgenic mouse model of Huntington’s disease. J Neurosci 27:3768–3779

    PubMed  CAS  Google Scholar 

  67. Okamoto S, Pouladi MA, Talantova M, Yao D, Xia P, Ehrnhoefer DE, Zaidi R, Clemente A, Kaul M, Graham RK, Zhang D, Vincent Chen HS, Tong G, Hayden MR, Lipton SA (2009) Balance between synaptic versus extrasynaptic NMDA receptor activity influences inclusions and neurotoxicity of mutant huntingtin. Nat Med 15:1407–1413

    PubMed  CAS  Google Scholar 

  68. Wu J, Tang T, Bezprozvanny I (2006) Evaluation of clinically relevant glutamate pathway inhibitors in in vitro model of Huntington’s disease. Neurosci Lett 407:219–223

    PubMed  CAS  Google Scholar 

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

    PubMed  Google Scholar 

  70. Coyle JT, Schwarcz R (1976) Lesion of striatal neurones with kainic acid provides a model for Huntington’s chorea. Nature 263:244–246

    PubMed  CAS  Google Scholar 

  71. McGeer EG, McGeer PL (1976) Duplication of biochemical changes of Huntington’s chorea by intrastriatal injections of glutamic and kainic acids. Nature 263:517–519

    PubMed  CAS  Google Scholar 

  72. McGeer EG, McGeer PL, Singh K (1978) Kainate-induced degeneration of neostriatal neurons: dependency upon corticostriatal tract. Brain Res 139:381–383

    PubMed  CAS  Google Scholar 

  73. Beal MF, Kowall NW, Ellison DW, Mazurek MF, Swartz KJ, Martin JB (1986) Replication of the neurochemical characteristics of Huntington’s disease by quinolinic acid. Nature 321:168–171

    PubMed  CAS  Google Scholar 

  74. Hantraye P, Riche D, Maziere M, Isacson O (1990) A primate model of Huntington’s disease: behavioral and anatomical studies of unilateral excitotoxic lesions of the caudate-putamen in the baboon. Exp Neurol 108:91–104

    PubMed  CAS  Google Scholar 

  75. Beal MF, Ferrante RJ, Swartz KJ, Kowall NW (1991) Chronic quinolinic acid lesions in rats closely resemble Huntington’s disease. J Neurosci 11:1649–1659

    PubMed  CAS  Google Scholar 

  76. Slow EJ, van Raamsdonk J, Rogers D, Coleman SH, Graham RK, Deng Y, Oh R, Bissada N, Hossain SM, Yang YZ, Li XJ, Simpson EM, Gutekunst CA, Leavitt BR, Hayden MR (2003) Selective striatal neuronal loss in a YAC128 mouse model of Huntington disease. Hum Mol Genet 12:1555–1567

    PubMed  CAS  Google Scholar 

  77. Hodgson JG, Agopyan N, Gutekunst CA, Leavitt BR, LePiane F, Singaraja R, Smith DJ, Bissada N, McCutcheon K, Nasir J, Jamot L, Li XJ, Stevens ME, Rosemond E, Roder JC, Phillips AG, Rubin EM, Hersch SM, Hayden MR (1999) A YAC mouse model for Huntington’s disease with full-length mutant huntingtin, cytoplasmic toxicity, and selective striatal neurodegeneration. Neuron 23:181–192

    PubMed  CAS  Google Scholar 

  78. Maruyama T, Kanaji T, Nakade S, Kanno T, Mikoshiba K (1997) 2APB, 2-aminoethoxydiphenyl borate, a membrane-penetrable modulator of Ins(1,4,5)P3-induced Ca2+ release. J Biochem (Tokyo) 122:498–505

    CAS  Google Scholar 

  79. Pulst SM, Nechiporuk A, Nechiporuk T, Gispert S, Chen XN, Lopes-Cendes I, Pearlman S, Starkman S, Orozco-Diaz G, Lunkes A, DeJong P, Rouleau GA, Auburger G, Korenberg JR, Figueroa C, Sahba S (1996) Moderate expansion of a normally biallelic trinucleotide repeat in spinocerebellar ataxia type 2. Nat Genet 14:269–276

    PubMed  CAS  Google Scholar 

  80. Imbert G, Saudou F, Yvert G, Devys D, Trottier Y, Garnier JM, Weber C, Mandel JL, Cancel G, Abbas N, Durr A, Didierjean O, Stevanin G, Agid Y, Brice A (1996) Cloning of the gene for spinocerebellar ataxia 2 reveals a locus with high sensitivity to expanded CAG/glutamine repeats. Nat Genet 14:285–291

    PubMed  CAS  Google Scholar 

  81. Sanpei K, Takano H, Igarashi S, Sato T, Oyake M, Sasaki H, Wakisaka A, Tashiro K, Ishida Y, Ikeuchi T, Koide R, Saito M, Sato A, Tanaka T, Hanyu S, Takiyama Y, Nishizawa M, Shimizu N, Nomura Y, Segawa M, Iwabuchi K, Eguchi I, Tanaka H, Takahashi H, Tsuji S (1996) Identification of the spinocerebellar ataxia type 2 gene using a direct identification of repeat expansion and cloning technique, DIRECT. Nat Genet 14:277–284

    PubMed  CAS  Google Scholar 

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

    PubMed  CAS  Google Scholar 

  83. Kiehl TR, Shibata H, Pulst SM (2000) The ortholog of human ataxin-2 is essential for early embryonic patterning in C. elegans. J Mol Neurosci 15:231–241

    PubMed  CAS  Google Scholar 

  84. Satterfield TF, Jackson SM, Pallanck LJ (2002) A drosophila homolog of the polyglutamine disease gene SCA2 is a dosage-sensitive regulator of actin filament formation. Genetics 162:1687–1702

    PubMed  CAS  Google Scholar 

  85. Kiehl TR, Nechiporuk A, Figueroa KP, Keating MT, Huynh DP, Pulst SM (2006) Generation and characterization of Sca2 (ataxin-2) knockout mice. Biochem Biophys Res Commun 339:17–24

    PubMed  CAS  Google Scholar 

  86. Pulst SM, Santos N, Wang D, Yang H, Huynh D, Velazquez L, Figueroa KP (2005) Spinocerebellar ataxia type 2: polyQ repeat variation in the CACNA1A calcium channel modifies age of onset. Brain 128:2297–2303

    PubMed  Google Scholar 

  87. Liu J, Tang TS, Tu H, Nelson O, Herndon E, Huynh DP, Pulst SM, Bezprozvanny I (2009) Deranged calcium signaling and neurodegeneration in spinocerebellar ataxia type 2. J Neurosci 29:9148–9162

    PubMed  CAS  Google Scholar 

  88. Furuichi T, Simon-Chazottes D, Fujino I, Yamada N, Hasegawa M, Miyawaki A, Yoshikawa S, Guenet JL, Mikoshiba K (1993) Widespread expression of inositol 1, 4, 5-trisphosphate receptor type 1 gene (Insp3r1) in the mouse central nervous system. Recept Channels 1:11–24

    PubMed  CAS  Google Scholar 

  89. Sharp AH, Nucifora FC Jr, Blondel O, Sheppard CA, Zhang C, Snyder SH, Russell JT, Ryugo DK, Ross CA (1999) Differential cellular expression of isoforms of inositol 1,4,5-triphosphate receptors in neurons and glia in brain. J Comp Neurol 406:207–220

    PubMed  CAS  Google Scholar 

  90. van de Loo S, Eich F, Nonis D, Auburger G, Nowock J (2009) Ataxin-2 associates with rough endoplasmic reticulum. Exp Neurol 215:110–118

    PubMed  Google Scholar 

  91. Stevanin G, Durr A, Brice A (2000) Clinical and molecular advances in autosomal dominant cerebellar ataxias: from genotype to phenotype and physiopathology. Eur J Hum Genet 8:4–18

    PubMed  CAS  Google Scholar 

  92. Coutinho P, Andrade C (1978) Autosomal dominant system degeneration in Portuguese families of the Azores Islands. A new genetic disorder involving cerebellar, pyramidal, extrapyramidal and spinal cord motor functions. Neurology 28:703–709

    PubMed  CAS  Google Scholar 

  93. 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

    PubMed  CAS  Google Scholar 

  94. Scheel H, Tomiuk S, Hofmann K (2003) Elucidation of ataxin-3 and ataxin-7 function by integrative bioinformatics. Hum Mol Genet 12:2845–2852

    PubMed  CAS  Google Scholar 

  95. Albrecht M, Golatta M, Wullner U, Lengauer T (2004) Structural and functional analysis of ataxin-2 and ataxin-3. Eur J Biochem 271:3155–3170

    PubMed  CAS  Google Scholar 

  96. 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

    PubMed  CAS  Google Scholar 

  97. 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

    PubMed  CAS  Google Scholar 

  98. 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

    PubMed  CAS  Google Scholar 

  99. 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

    PubMed  CAS  Google Scholar 

  100. 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

    PubMed  CAS  Google Scholar 

  101. 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

    PubMed  CAS  Google Scholar 

  102. Schorge S, van de Leemput J, Singleton A, Houlden H, Hardy J (2010) Human ataxias: a genetic dissection of inositol triphosphate receptor (ITPR1)-dependent signaling. Trends Neurosci 33:211–219

    PubMed  CAS  Google Scholar 

  103. Zoghbi HY, Orr HT (2009) Pathogenic mechanisms of a polyglutamine-mediated neurodegenerative disease, spinocerebellar ataxia type 1. J Biol Chem 284:7425–7429

    PubMed  CAS  Google Scholar 

  104. Klement IA, Skinner PJ, Kaytor MD, Yi H, Hersch SM, Clark HB, Zoghbi HY, Orr HT (1998) Ataxin-1 nuclear localization and aggregation: role in polyglutamine-induced disease in SCA1 transgenic mice. Cell 95:41–53

    PubMed  CAS  Google Scholar 

  105. Serra HG, Duvick L, Zu T, Carlson K, Stevens S, Jorgensen N, Lysholm A, Burright E, Zoghbi HY, Clark HB, Andresen JM, Orr HT (2006) RORalpha-mediated Purkinje cell development determines disease severity in adult SCA1 mice. Cell 127:697–708

    PubMed  CAS  Google Scholar 

  106. Goold R, Hubank M, Hunt A, Holton J, Menon RP, Revesz T, Pandolfo M, Matilla-Duenas A (2007) Down-regulation of the dopamine receptor D2 in mice lacking ataxin 1. Hum Mol Genet 16:2122–2134

    PubMed  CAS  Google Scholar 

  107. Lin X, Antalffy B, Kang D, Orr HT, Zoghbi HY (2000) Polyglutamine expansion down-regulates specific neuronal genes before pathologic changes in SCA1. Nat Neurosci 3:157–163

    PubMed  CAS  Google Scholar 

  108. Vig PJ, Subramony SH, Qin Z, McDaniel DO, Fratkin JD (2000) Relationship between ataxin-1 nuclear inclusions and Purkinje cell specific proteins in SCA-1 transgenic mice. J Neurol Sci 174:100–110

    PubMed  CAS  Google Scholar 

  109. Serra HG, Byam CE, Lande JD, Tousey SK, Zoghbi HY, Orr HT (2004) Gene profiling links SCA1 pathophysiology to glutamate signaling in Purkinje cells of transgenic mice. Hum Mol Genet 13:2535–2543

    PubMed  CAS  Google Scholar 

  110. Inoue T, Lin X, Kohlmeier KA, Orr HT, Zoghbi HY, Ross WN (2001) Calcium dynamics and electrophysiological properties of cerebellar Purkinje cells in SCA1 transgenic mice. J Neurophysiol 85:1750–1760

    PubMed  CAS  Google Scholar 

  111. Piedras-Renteria ES, Watase K, Harata N, Zhuchenko O, Zoghbi HY, Lee CC, Tsien RW (2001) Increased expression of alpha 1A Ca2+ channel currents arising from expanded trinucleotide repeats in spinocerebellar ataxia type 6. J Neurosci 21:9185–9193

    PubMed  CAS  Google Scholar 

  112. Watase K, Barrett CF, Miyazaki T, Ishiguro T, Ishikawa K, Hu Y, Unno T, Sun Y, Kasai S, Watanabe M, Gomez CM, Mizusawa H, Tsien RW, Zoghbi HY (2008) Spinocerebellar ataxia type 6 knockin mice develop a progressive neuronal dysfunction with age-dependent accumulation of mutant CaV2.1 channels. Proc Natl Acad Sci USA 105:11987–11992

    PubMed  CAS  Google Scholar 

  113. Ikeda Y, Dick KA, Weatherspoon MR, Gincel D, Armbrust KR, Dalton JC, Stevanin G, Durr A, Zuhlke C, Burk K, Clark HB, Brice A, Rothstein JD, Schut LJ, Day JW, Ranum LP (2006) Spectrin mutations cause spinocerebellar ataxia type 5. Nat Genet 38:184–190

    PubMed  CAS  Google Scholar 

  114. Kose A, Saito N, Ito H, Kikkawa U, Nishizuka Y, Tanaka C (1988) Electron microscopic localization of type I protein kinase C in rat Purkinje cells. J Neurosci 8:4262–4268

    PubMed  CAS  Google Scholar 

  115. Adachi N, Kobayashi T, Takahashi H, Kawasaki T, Shirai Y, Ueyama T, Matsuda T, Seki T, Sakai N, Saito N (2008) Enzymological analysis of mutant protein kinase Cgamma causing spinocerebellar ataxia type 14 and dysfunction in Ca2+ homeostasis. J Biol Chem 283:19854–19863

    PubMed  CAS  Google Scholar 

  116. Zhu LP, Yu XD, Ling S, Brown RA, Kuo TH (2000) Mitochondrial Ca(2+)homeostasis in the regulation of apoptotic and necrotic cell deaths. Cell Calcium 28:107–117

    PubMed  CAS  Google Scholar 

  117. van de Leemput J, Chandran J, Knight MA, Holtzclaw LA, Scholz S, Cookson MR, Houlden H, Gwinn-Hardy K, Fung HC, Lin X, Hernandez D, Simon-Sanchez J, Wood NW, Giunti P, Rafferty I, Hardy J, Storey E, Gardner RJ, Forrest SM, Fisher EM, Russell JT, Cai H, Singleton AB (2007) Deletion at ITPR1 underlies ataxia in mice and spinocerebellar ataxia 15 in humans. PLoS Genet 3:e108

  118. Miyoshi Y, Yamada T, Tanimura M, Taniwaki T, Arakawa K, Ohyagi Y, Furuya H, Yamamoto K, Sakai K, Sasazuki T, Kira J (2001) A novel autosomal dominant spinocerebellar ataxia (SCA16) linked to chromosome 8q22.1–24.1. Neurology 57:96–100

    PubMed  CAS  Google Scholar 

  119. Miura S, Shibata H, Furuya H, Ohyagi Y, Osoegawa M, Miyoshi Y, Matsunaga H, Shibata A, Matsumoto N, Iwaki A, Taniwaki T, Kikuchi H, Kira J, Fukumaki Y (2006) The contactin 4 gene locus at 3p26 is a candidate gene of SCA16. Neurology 67:1236–1241

    PubMed  CAS  Google Scholar 

  120. Tanaka E, Maruyama H, Morino H, Nakajima E, Kawakami H (2008) The CNTN4 c.4256C > T mutation is rare in Japanese with inherited spinocerebellar ataxia. J Neurol Sci 266:180–181

    PubMed  CAS  Google Scholar 

  121. Hisatsune C, Kuroda Y, Akagi T, Torashima T, Hirai H, Hashikawa T, Inoue T, Mikoshiba K (2006) Inositol 1,4,5-trisphosphate receptor type 1 in granule cells, not in Purkinje cells, regulates the dendritic morphology of Purkinje cells through brain-derived neurotrophic factor production. J Neurosci 26:10916–10924

    PubMed  CAS  Google Scholar 

  122. Inoue T, Kato K, Kohda K, Mikoshiba K (1998) Type 1 inositol 1,4,5-trisphosphate receptor is required for induction of long-term depression in cerebellar Purkinje neurons. J Neurosci 18:5366–5373

    PubMed  CAS  Google Scholar 

  123. Fujii S, Matsumoto M, Igarashi K, Kato H, Mikoshiba K (2000) Synaptic plasticity in hippocampal CA1 neurons of mice lacking type 1 inositol-1,4,5-trisphosphate receptors. Learn Mem 7:312–320

    PubMed  CAS  Google Scholar 

  124. Nagase T, Ito KI, Kato K, Kaneko K, Kohda K, Matsumoto M, Hoshino A, Inoue T, Fujii S, Kato H, Mikoshiba K (2003) Long-term potentiation and long-term depression in hippocampal CA1 neurons of mice lacking the IP(3) type 1 receptor. Neuroscience 117:821–830

    PubMed  CAS  Google Scholar 

  125. Harper SQ, Staber PD, He X, Eliason SL, Martins IH, Mao Q, Yang L, Kotin RM, Paulson HL, Davidson BL (2005) RNA interference improves motor and neuropathological abnormalities in a Huntington’s disease mouse model. Proc Natl Acad Sci USA 102:5820–5825

    PubMed  CAS  Google Scholar 

  126. Xia H, Mao Q, Eliason SL, Harper SQ, Martins IH, Orr HT, Paulson HL, Yang L, Kotin RM, Davidson BL (2004) RNAi suppresses polyglutamine-induced neurodegeneration in a model of spinocerebellar ataxia. Nat Med 10:816–820

    PubMed  CAS  Google Scholar 

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

    PubMed  CAS  Google Scholar 

  128. Shao J, Diamond MI (2007) Polyglutamine diseases: emerging concepts in pathogenesis and therapy. Hum Mol Genet 16 (2): R115–R123

    Google Scholar 

  129. Warrick JM, Chan HY, Gray-Board GL, Chai Y, Paulson HL, Bonini NM (1999) Suppression of polyglutamine-mediated neurodegeneration in Drosophila by the molecular chaperone HSP70. Nat Genet 23:425–428

    PubMed  CAS  Google Scholar 

  130. Wyttenbach A, Sauvageot O, Carmichael J, Diaz-Latoud C, Arrigo AP, Rubinsztein DC (2002) Heat shock protein 27 prevents cellular polyglutamine toxicity and suppresses the increase of reactive oxygen species caused by huntingtin. Hum Mol Genet 11:1137–1151

    PubMed  CAS  Google Scholar 

  131. Wacker JL, Zareie MH, Fong H, Sarikaya M, Muchowski PJ (2004) Hsp70 and Hsp40 attenuate formation of spherical and annular polyglutamine oligomers by partitioning monomer. Nat Struct Mol Biol 11:1215–1222

    PubMed  CAS  Google Scholar 

  132. Heiser V, Engemann S, Brocker W, Dunkel I, Boeddrich A, Waelter S, Nordhoff E, Lurz R, Schugardt N, Rautenberg S, Herhaus C, Barnickel G, Bottcher H, Lehrach H, Wanker EE (2002) Identification of benzothiazoles as potential polyglutamine aggregation inhibitors of Huntington’s disease by using an automated filter retardation assay. Proc Natl Acad Sci USA 99(Suppl 4):16400–16406

    PubMed  CAS  Google Scholar 

  133. Sanchez I, Mahlke C, Yuan J (2003) Pivotal role of oligomerization in expanded polyglutamine neurodegenerative disorders. Nature 421:373–379

    PubMed  CAS  Google Scholar 

  134. Zhang X, Smith DL, Meriin AB, Engemann S, Russel DE, Roark M, Washington SL, Maxwell MM, Marsh JL, Thompson LM, Wanker EE, Young AB, Housman DE, Bates GP, Sherman MY, Kazantsev AG (2005) A potent small molecule inhibits polyglutamine aggregation in Huntington’s disease neurons and suppresses neurodegeneration in vivo. Proc Natl Acad Sci USA 102:892–897

    PubMed  CAS  Google Scholar 

  135. Wang J, Gines S, MacDonald ME, Gusella JF (2005) Reversal of a full-length mutant huntingtin neuronal cell phenotype by chemical inhibitors of polyglutamine-mediated aggregation. BMC Neurosci 6:1

    PubMed  Google Scholar 

  136. Ehrnhoefer DE, Duennwald M, Markovic P, Wacker JL, Engemann S, Roark M, Legleiter J, Marsh JL, Thompson LM, Lindquist S, Muchowski PJ, Wanker EE (2006) Green tea (-)-epigallocatechin-gallate modulates early events in huntingtin misfolding and reduces toxicity in Huntington’s disease models. Hum Mol Genet 15:2743–2751

    PubMed  CAS  Google Scholar 

  137. Chopra V, Fox JH, Lieberman G, Dorsey K, Matson W, Waldmeier P, Housman DE, Kazantsev A, Young AB, Hersch S (2007) 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 104:16685–16689

    PubMed  CAS  Google Scholar 

  138. Nagai Y, Tucker T, Ren H, Kenan DJ, Henderson BS, Keene JD, Strittmatter WJ, Burke JR (2000) Inhibition of polyglutamine protein aggregation and cell death by novel peptides identified by phage display screening. J Biol Chem 275:10437–10442

    PubMed  CAS  Google Scholar 

  139. Nagai Y, Fujikake N, Ohno K, Higashiyama H, Popiel HA, Rahadian J, Yamaguchi M, Strittmatter WJ, Burke JR, Toda T (2003) Prevention of polyglutamine oligomerization and neurodegeneration by the peptide inhibitor QBP1 in Drosophila. Hum Mol Genet 12:1253–1259

    PubMed  CAS  Google Scholar 

  140. Popiel HA, Nagai Y, Fujikake N, Toda T (2007) Protein transduction domain-mediated delivery of QBP1 suppresses polyglutamine-induced neurodegeneration in vivo. Mol Ther 15:303–309

    PubMed  CAS  Google Scholar 

  141. Popiel HA, Nagai Y, Fujikake N, Toda T (2009) Delivery of the aggregate inhibitor peptide QBP1 into the mouse brain using PTDs and its therapeutic effect on polyglutamine disease mice. Neurosci Lett 449:87–92

    PubMed  CAS  Google Scholar 

  142. Kim MW, Chelliah Y, Kim SW, Otwinowski Z, Bezprozvanny I (2009) Secondary structure of Huntingtin amino-terminal region. Structure 17:1205–1212

    PubMed  CAS  Google Scholar 

  143. Lee ST, Chu K, Park JE, Kang L, Ko SY, Jung KH, Kim M (2006) Memantine reduces striatal cell death with decreasing calpain level in 3-nitropropionic model of Huntington’s disease. Brain Res 1118:199–207

    PubMed  CAS  Google Scholar 

  144. Landwehrmeyer GB, Dubois B, de Yebenes JG, Kremer B, Gaus W, Kraus PH, Przuntek H, Dib M, Doble A, Fischer W, Ludolph AC (2007) Riluzole in Huntington’s disease: a 3-year, randomized controlled study. Ann Neurol 62:262–272

    PubMed  CAS  Google Scholar 

  145. Chan CS, Guzman JN, Ilijic E, Mercer JN, Rick C, Tkatch T, Meredith GE, Surmeier DJ (2007) ‘Rejuvenation’ protects neurons in mouse models of Parkinson’s disease. Nature 447:1081–1086

    PubMed  CAS  Google Scholar 

  146. Becker C, Jick SS, Meier CR (2008) Use of antihypertensives and the risk of Parkinson disease. Neurology 70:1438–1444

    PubMed  CAS  Google Scholar 

  147. Chaturvedi RK, Beal MF (2008) Mitochondrial approaches for neuroprotection. Ann N Y Acad Sci 1147:395–412

    PubMed  CAS  Google Scholar 

Download references

Acknowledgments

I would like to thank members of my laboratory for their hard work. I am a holder of Carla Cocke Francis Professorship in Alzheimer’s Research and supported by the McKnight Neuroscience of Brain Disorders Award. Our work on HD, SCA2 and SCA3 was supported by CHDI, Hereditary Disease Foundation, the National Organization for Rare Disorders, National Ataxia Foundation, Ataxia MJD Research Project, and the National Institutes of Health grants R01NS38082 and R01NS056224.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Ilya Bezprozvanny.

Additional information

Special Issue: In Honor of Dr. Mikoshiba.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Bezprozvanny, I. Role of Inositol 1,4,5-Trishosphate Receptors in Pathogenesis of Huntington’s Disease and Spinocerebellar Ataxias. Neurochem Res 36, 1186–1197 (2011). https://doi.org/10.1007/s11064-010-0393-y

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11064-010-0393-y

Keywords

Navigation