메뉴 건너뛰기




Volumn 2, Issue 4, 2013, Pages 405-436

Surveying the landscape of huntington’s disease mechanisms, measurements, and medicines

Author keywords

Aggregation; Chaperones; Disease mechanisms; Excitotoxicity; Gene therapy; Huntington s disease; Neurodegeneration; Therapeutics

Indexed keywords

AGGREGATION; ANIMAL; CHAPERONES; DISEASE MECHANISMS; EXCITOTOXICITY; GENE THERAPY; GENETICS; HUMAN; HUNTINGTON CHOREA; NERVE DEGENERATION; PATHOPHYSIOLOGY; REVIEW; THERAPY;

EID: 84906924042     PISSN: 18796397     EISSN: 18796400     Source Type: Journal    
DOI: 10.3233/JHD-130072     Document Type: Article
Times cited : (9)

References (310)
  • 1
    • 0027480960 scopus 로고
    • A novel gene containing a trinucleotide repeat that is expanded and unstable on Huntington’s disease chromosomes
    • Group THDCR. A novel gene containing a trinucleotide repeat that is expanded and unstable on Huntington’s disease chromosomes. Cell. 1993;72:971-83.
    • (1993) Cell. , vol.72 , pp. 971-983
  • 2
    • 0027327418 scopus 로고
    • Trinucleotide repeat elongation in the Huntingtin gene in Huntington disease patients from 71 Danish families
    • Nørremølle A, Riess O, Epplen JT, Fenger K, Hasholt L, Sørensen SA. Trinucleotide repeat elongation in the Huntingtin gene in Huntington disease patients from 71 Danish families. Hum Mol Genet. 1993;2:1475-6.
    • (1993) Hum Mol Genet. , vol.2 , pp. 1475-1476
    • Nørremølle, A.1    Riess, O.2    Epplen, J.T.3    Fenger, K.4    Hasholt, L.5    Sørensen, S.A.6
  • 7
    • 79551655290 scopus 로고    scopus 로고
    • Huntington’s disease: Can mice lead theway to treatment?
    • 2011
    • Crook ZR, Housman D (2011) Huntington’s disease: can mice lead theway to treatment? Neuron. 2011;69(3):423-35.
    • (2011) Neuron. , vol.69 , Issue.3 , pp. 423-435
    • Crook, Z.R.1    Housman, D.2
  • 10
    • 36949022900 scopus 로고    scopus 로고
    • CAG repeat disorder models and human neuropathology: Similarities and differences
    • Yamada M, Sato T, Tsuji S, Takahashi H. CAG repeat disorder models and human neuropathology: Similarities and differences. Acta Neuropathol. 2008;115:71-86.
    • (2008) Acta Neuropathol. , vol.115 , pp. 71-86
    • Yamada, M.1    Sato, T.2    Tsuji, S.3    Takahashi, H.4
  • 14
    • 0001388128 scopus 로고    scopus 로고
    • Expression of polyglutamine-expanded Huntingtin activates the SEK1-JNK pathway and induces apoptosis in a hippocampal neuronal cell line
    • Liu YF. Expression of polyglutamine-expanded Huntingtin activates the SEK1-JNK pathway and induces apoptosis in a hippocampal neuronal cell line. J Biol Chem. 1998;273:28873-7.
    • (1998) J Biol Chem. , vol.273 , pp. 28873-28877
    • Liu, Y.F.1
  • 17
    • 84863990252 scopus 로고    scopus 로고
    • Physical chemistry of polyglutamine: Intriguing tales of a monotonous sequence
    • Wetzel R. Physical chemistry of polyglutamine: Intriguing tales of a monotonous sequence. J Mol Biol. 2012;421:466-90.
    • (2012) J Mol Biol. , vol.421 , pp. 466-490
    • Wetzel, R.1
  • 19
    • 0037174879 scopus 로고    scopus 로고
    • Huntingtin spheroids and protofibrils as precursors in polyglutamine fibrilization
    • Poirier MA, Li H, Macosko J, Cai S, Amzel M, Ross CA. Huntingtin spheroids and protofibrils as precursors in polyglutamine fibrilization. J Biol Chem. 2002;277:41032-7.
    • (2002) J Biol Chem. , vol.277 , pp. 41032-41037
    • Poirier, M.A.1    Li, H.2    Macosko, J.3    Cai, S.4    Amzel, M.5    Ross, C.A.6
  • 20
    • 33846295583 scopus 로고    scopus 로고
    • Systemic administration of Congo red does not improve motor or cognitive function in R6/2 mice
    • Wood NI, Pallier PN, Wanderer J, Morton AJ. Systemic administration of Congo red does not improve motor or cognitive function in R6/2 mice. Neurobiology of Disease. 2007;25:342-53.
    • (2007) Neurobiology of Disease. , vol.25 , pp. 342-353
    • Wood, N.I.1    Pallier, P.N.2    Wanderer, J.3    Morton, A.J.4
  • 21
    • 0037461730 scopus 로고    scopus 로고
    • Pivotal role of oligomerization in expanded polyglutamine neurodegenerative disorders
    • Sánchez I, Mahlke C, Yuan J. Pivotal role of oligomerization in expanded polyglutamine neurodegenerative disorders. Nature. 2003;421:373-9.
    • (2003) Nature. , vol.421 , pp. 373-379
    • Sánchez, I.1    Mahlke, C.2    Yuan, J.3
  • 22
    • 7244236320 scopus 로고    scopus 로고
    • Inclusion body formation reduces levels of mutant huntingtin and the risk of neuronal death
    • Arrasate M, Mitra S, Schweitzer ES, Segal MR, Finkbeiner S. Inclusion body formation reduces levels of mutant huntingtin and the risk of neuronal death. Nature. 2004;431:805-10.
    • (2004) Nature. , vol.431 , pp. 805-810
    • Arrasate, M.1    Mitra, S.2    Schweitzer, E.S.3    Segal, M.R.4    Finkbeiner, S.5
  • 23
    • 14644419638 scopus 로고    scopus 로고
    • Neuronal dysfunction in a polyglutamine disease model occurs in the absence of ubiquitin-proteasome system impairment and inversely correlates with the degree of nuclear inclusion formation
    • Bowman AB, Yoo S-Y, Dantuma NP, Zoghbi HY. Neuronal dysfunction in a polyglutamine disease model occurs in the absence of ubiquitin-proteasome system impairment and inversely correlates with the degree of nuclear inclusion formation. Hum Mol Genet. 2005;14:679-91.
    • (2005) Hum Mol Genet. , vol.14 , pp. 679-691
    • Bowman, A.B.1    Yoo, S.-Y.2    Dantuma, N.P.3    Zoghbi, H.Y.4
  • 24
    • 37049029416 scopus 로고    scopus 로고
    • R6/2 neurons with intranuclear inclusions survive for prolonged periods in the brains of chimeric mice
    • Reiner A, del Mar N, Deng Y-P, Meade CA, Sun Z, Goldowitz D. R6/2 neurons with intranuclear inclusions survive for prolonged periods in the brains of chimeric mice. J Comp Neurol. 2007;505:603-29.
    • (2007) J Comp Neurol. , vol.505 , pp. 603-629
    • Reiner, A.1    del Mar, N.2    Deng, Y.-P.3    Meade, C.A.4    Sun, Z.5    Goldowitz, D.6
  • 25
    • 0035503677 scopus 로고    scopus 로고
    • Differential susceptibility of cultured cell lines to aggregate formation and cell death produced by the truncated Machado-Joseph disease gene product with an expanded polyglutamine stretch
    • Yoshizawa T, Yoshida H, Shoji S. Differential susceptibility of cultured cell lines to aggregate formation and cell death produced by the truncated Machado-Joseph disease gene product with an expanded polyglutamine stretch. Brain Res Bull. 2001;56:349-52.
    • (2001) Brain Res Bull. , vol.56 , pp. 349-352
    • Yoshizawa, T.1    Yoshida, H.2    Shoji, S.3
  • 27
    • 79953123196 scopus 로고    scopus 로고
    • Premature death and neurologic abnormalities in transgenic mice expressing a mutant huntingtin exon-2 fragment
    • Tebbenkamp ATN, Swing D, Tessarollo L, Borchelt DR. Premature death and neurologic abnormalities in transgenic mice expressing a mutant huntingtin exon-2 fragment. Hum Mol Genet. 2011;20:1633-42.
    • (2011) Hum Mol Genet. , vol.20 , pp. 1633-1642
    • Tebbenkamp, A.T.N.1    Swing, D.2    Tessarollo, L.3    Borchelt, D.R.4
  • 35
    • 35348877164 scopus 로고    scopus 로고
    • Transcriptional signatures in Huntington’s disease
    • Cha J-HJ. Transcriptional signatures in Huntington’s disease. Prog Neurobiol. 2007;83:228-48.
    • (2007) Prog Neurobiol. , vol.83 , pp. 228-248
    • Cha, J.-H.J.1
  • 36
    • 0037090927 scopus 로고    scopus 로고
    • Huntingtin inclusions do not deplete polyglutamine-containing transcription factors in HD mice
    • Yu Z-X, Li S-H, Nguyen HP, Li X-J. Huntingtin inclusions do not deplete polyglutamine-containing transcription factors in HD mice. Hum Mol Genet. 2002;11:905-14.
    • (2002) Hum Mol Genet. , vol.11 , pp. 905-914
    • Yu, Z.-X.1    Li, S.-H.2    Nguyen, H.P.3    Li, X.-J.4
  • 38
    • 0034646426 scopus 로고    scopus 로고
    • Effects of heat shock, heat shock protein 40 (HDJ-2), and proteasome inhibition on protein aggregation in cellular models of Huntington’s disease
    • Wyttenbach A, Carmichael J, Swartz J, Furlong RA, Narain Y, Rankin J, Rubinsztein DC. Effects of heat shock, heat shock protein 40 (HDJ-2), and proteasome inhibition on protein aggregation in cellular models of Huntington’s disease. Proc Natl Acad Sci U S A. 2000;97:2898-903.
    • (2000) Proc Natl Acad Sci U S A. , vol.97 , pp. 2898-2903
    • Wyttenbach, A.1    Carmichael, J.2    Swartz, J.3    Furlong, R.A.4    Narain, Y.5    Rankin, J.6    Rubinsztein, D.C.7
  • 39
    • 0034641589 scopus 로고    scopus 로고
    • Polyglutamine length-dependent interaction of Hsp40 and Hsp70 family chaperones with truncated N-terminal huntingtin: Their role in suppression of aggregation and cellular toxicity
    • Jana NR, Tanaka M, Wang GH, Nukina N. Polyglutamine length-dependent interaction of Hsp40 and Hsp70 family chaperones with truncated N-terminal huntingtin: Their role in suppression of aggregation and cellular toxicity. HumMol Genet. 2000;9:2009-18.
    • (2000) HumMol Genet. , vol.9 , pp. 2009-2018
    • Jana, N.R.1    Tanaka, M.2    Wang, G.H.3    Nukina, N.4
  • 41
    • 84855478434 scopus 로고    scopus 로고
    • A screen for enhancers of clearance identifies huntingtin as an heat shock protein 90 (Hsp90) client protein
    • Baldo B, Weiss A, Parker CN, Bibel M, Paganetti P, Kaupmann K. A screen for enhancers of clearance identifies huntingtin as an heat shock protein 90 (Hsp90) client protein. Journal of Biological Chemistry. 2012;287(2):1406-14.
    • (2012) Journal of Biological Chemistry. , vol.287 , Issue.2 , pp. 1406-1414
    • Baldo, B.1    Weiss, A.2    Parker, C.N.3    Bibel, M.4    Paganetti, P.5    Kaupmann, K.6
  • 43
    • 0032727617 scopus 로고    scopus 로고
    • Suppression of polyglutamine-mediated neurodegeneration in Drosophila by the molecular chaperone HSP70
    • Warrick JM, ChanHY, Gray-Board GL, ChaiY, Paulson HL, Bonini NM. Suppression of polyglutamine-mediated neurodegeneration in Drosophila by the molecular chaperone HSP70. Nat Genet. 1999;23:425-8.
    • (1999) Nat Genet. , vol.23 , pp. 425-428
    • Warrick, J.M.1    Chan, H.Y.2    Gray-Board, G.L.3    Chai, Y.4    Paulson, H.L.5    Bonini, N.M.6
  • 44
    • 0036850456 scopus 로고    scopus 로고
    • Genetic modulation of polyglutamine toxicity by protein conjugation pathways in Drosophila
    • Chan HYE, Warrick JM, Andriola I, Merry D, Bonini NM. Genetic modulation of polyglutamine toxicity by protein conjugation pathways in Drosophila. Hum Mol Genet. 2002;11:2895-904.
    • (2002) Hum Mol Genet. , vol.11 , pp. 2895-2904
    • Chan, H.Y.E.1    Warrick, J.M.2    Andriola, I.3    Merry, D.4    Bonini, N.M.5
  • 47
    • 66749167799 scopus 로고    scopus 로고
    • Rhes, a striatal specific protein, mediates mutant-huntingtin cytotoxicity
    • Subramaniam S, Sixt KM, Barrow R, Snyder SH. Rhes, a striatal specific protein, mediates mutant-huntingtin cytotoxicity. Science. 2009;324:1327-30.
    • (2009) Science. , vol.324 , pp. 1327-1330
    • Subramaniam, S.1    Sixt, K.M.2    Barrow, R.3    Snyder, S.H.4
  • 48
    • 0036848793 scopus 로고    scopus 로고
    • Purification of polyglutamine aggregates and identification of elongation factor-1alpha and heat shock protein 84 as aggregateinteracting proteins
    • Mitsui K, Nakayama H, Akagi T, Nekooki M, Ohtawa K, Takio K, Hashikawa T, Nukina N. Purification of polyglutamine aggregates and identification of elongation factor-1alpha and heat shock protein 84 as aggregateinteracting proteins. J Neurosci. 2002;22:9267-77.
    • (2002) J Neurosci. , vol.22 , pp. 9267-9277
    • Mitsui, K.1    Nakayama, H.2    Akagi, T.3    Nekooki, M.4    Ohtawa, K.5    Takio, K.6    Hashikawa, T.7    Nukina, N.8
  • 49
    • 0347928859 scopus 로고    scopus 로고
    • Overexpression of heat shock protein 70 in R6/2 Huntington’s disease mice has only modest effects on disease progression
    • Hansson O, Nylandsted J, Castilho RF, Leist M, Jäättelä M, Brundin P. Overexpression of heat shock protein 70 in R6/2 Huntington’s disease mice has only modest effects on disease progression. Brain Res. 2003;970:47-57.
    • (2003) Brain Res. , vol.970 , pp. 47-57
    • Hansson, O.1    Nylandsted, J.2    Castilho, R.F.3    Leist, M.4    Jäättelä, M.5    Brundin, P.6
  • 50
    • 3242695184 scopus 로고    scopus 로고
    • Progressive decrease in chaperone protein levels in a mouse model of Huntington’s disease and induction of stress proteins as a therapeutic approach
    • Hay DG, Sathasivam K, Tobaben S, Stahl B, Marber M, Mestril R, Mahal A, Smith DL, Woodman B, Bates GP. Progressive decrease in chaperone protein levels in a mouse model of Huntington’s disease and induction of stress proteins as a therapeutic approach. Hum Mol Genet. 2004;13:1389-405.
    • (2004) Hum Mol Genet. , vol.13 , pp. 1389-1405
    • Hay, D.G.1    Sathasivam, K.2    Tobaben, S.3    Stahl, B.4    Marber, M.5    Mestril, R.6    Mahal, A.7    Smith, D.L.8    Woodman, B.9    Bates, G.P.10
  • 51
    • 27144524290 scopus 로고    scopus 로고
    • Active HSF1 significantly suppresses polyglutamine aggregate formation in cellular and mouse models
    • Fujimoto M, Takaki E, Hayashi T, Kitaura Y, Tanaka Y, Inouye S, Nakai A. Active HSF1 significantly suppresses polyglutamine aggregate formation in cellular and mouse models. J Biol Chem. 2005;280:34908-16.
    • (2005) J Biol Chem. , vol.280 , pp. 34908-34916
    • Fujimoto, M.1    Takaki, E.2    Hayashi, T.3    Kitaura, Y.4    Tanaka, Y.5    Inouye, S.6    Nakai, A.7
  • 52
    • 84875519144 scopus 로고    scopus 로고
    • Targeting the molecular chaperone heat shock protein 90 (HSP90): Lessons learned and future directions
    • Hong DS, Banerji U, Tavana B, George GC, Aaron J, Kurzrock R. Targeting the molecular chaperone heat shock protein 90 (HSP90): Lessons learned and future directions. Cancer Treat Rev. 2013;39:375-87.
    • (2013) Cancer Treat Rev. , vol.39 , pp. 375-387
    • Hong, D.S.1    Banerji, U.2    Tavana, B.3    George, G.C.4    Aaron, J.5    Kurzrock, R.6
  • 53
    • 84861563268 scopus 로고    scopus 로고
    • Cancer incidence in patients with polyglutamine diseases: A population-based study in Sweden
    • Ji J, Sundquist K, Sundquist J. Cancer incidence in patients with polyglutamine diseases: A population-based study in Sweden. Lancet Oncol. 2012.
    • (2012) Lancet Oncol.
    • Ji, J.1    Sundquist, K.2    Sundquist, J.3
  • 54
    • 7444231693 scopus 로고    scopus 로고
    • Mechanism of the eukaryotic chaperonin: Protein folding in the chamber of secrets
    • Spiess C, Meyer AS, Reissmann S, Frydman J. Mechanism of the eukaryotic chaperonin: Protein folding in the chamber of secrets. Trends Cell Biol. 2004;14:598-604.
    • (2004) Trends Cell Biol. , vol.14 , pp. 598-604
    • Spiess, C.1    Meyer, A.S.2    Reissmann, S.3    Frydman, J.4
  • 56
    • 71449084004 scopus 로고    scopus 로고
    • The chaperonin TRiC blocks a huntingtin sequence element that promotes the conformational switch to aggregation
    • TamS, Spiess C, AuyeungW, Joachimiak L, Chen B, Poirier MA, Frydman J. The chaperonin TRiC blocks a huntingtin sequence element that promotes the conformational switch to aggregation. Nat Struct Mol Biol. 2009;16:1279-85.
    • (2009) Nat Struct Mol Biol. , vol.16 , pp. 1279-1285
    • Tam, S.1    Spiess, C.2    Auyeung, W.3    Joachimiak, L.4    Chen, B.5    Poirier, M.A.6    Frydman, J.7
  • 58
    • 0029895292 scopus 로고    scopus 로고
    • Striatal D1 and D2 dopamine receptor loss in asymptomatic mutation carriers of Huntington’s disease
    • Weeks RA, Piccini P, Harding AE, Brooks DJ. Striatal D1 and D2 dopamine receptor loss in asymptomatic mutation carriers of Huntington’s disease. Ann Neurol. 1996;40:49-54.
    • (1996) Ann Neurol. , vol.40 , pp. 49-54
    • Weeks, R.A.1    Piccini, P.2    Harding, A.E.3    Brooks, D.J.4
  • 59
    • 0030612119 scopus 로고    scopus 로고
    • Dopamine D1 and D2 receptor gene expression in the striatum in Huntington’s disease
    • Augood SJ, Faull RL, Emson PC. Dopamine D1 and D2 receptor gene expression in the striatum in Huntington’s disease. Ann Neurol. 1997;42:215-21.
    • (1997) Ann Neurol. , vol.42 , pp. 215-221
    • Augood, S.J.1    Faull, R.L.2    Emson, P.C.3
  • 60
    • 0029926199 scopus 로고    scopus 로고
    • Reduction in enkephalin and substance P messenger RNA in the striatum of early grade Huntington’s disease: A detailed cellular in situ hybridization study
    • Augood SJ, Faull RL, Love DR, Emson PC. Reduction in enkephalin and substance P messenger RNA in the striatum of early grade Huntington’s disease: A detailed cellular in situ hybridization study. NSC. 1996;72:1023-36.
    • (1996) NSC. , vol.72 , pp. 1023-1036
    • Augood, S.J.1    Faull, R.L.2    Love, D.R.3    Emson, P.C.4
  • 61
    • 0029966584 scopus 로고    scopus 로고
    • Decreased neuronal nitric oxide synthase messenger RNA and somatostatin messenger RNA in the striatum of Huntington’s disease
    • Norris PJ, Waldvogel HJ, Faull RL, Love DR, Emson PC. Decreased neuronal nitric oxide synthase messenger RNA and somatostatin messenger RNA in the striatum of Huntington’s disease. NSC. 1996;72:1037-47.
    • (1996) NSC. , vol.72 , pp. 1037-1047
    • Norris, P.J.1    Waldvogel, H.J.2    Faull, R.L.3    Love, D.R.4    Emson, P.C.5
  • 64
    • 58049197863 scopus 로고    scopus 로고
    • Huntingtin regulates RE1-silencing transcription factor/neuron-restrictive silencer factor (REST/NRSF) nuclear trafficking indirectly through a complex with REST/NRSF-interacting LIM domain protein (RILP) and dynactin p150 Glued
    • Shimojo M. Huntingtin regulates RE1-silencing transcription factor/neuron-restrictive silencer factor (REST/NRSF) nuclear trafficking indirectly through a complex with REST/NRSF-interacting LIM domain protein (RILP) and dynactin p150 Glued. J Biol Chem. 2008;283:34880-6.
    • (2008) J Biol Chem. , vol.283 , pp. 34880-34886
    • Shimojo, M.1
  • 69
    • 41749121925 scopus 로고    scopus 로고
    • Brain gene expression correlates with changes in behavior in the R6/1 mouse model of Huntington’s disease
    • Hodges A, Hughes G, Brooks S, Elliston L, Holmans P, Dunnett SB, Jones L. Brain gene expression correlates with changes in behavior in the R6/1 mouse model of Huntington’s disease. Genes Brain Behav. 2008;7:288-99.
    • (2008) Genes Brain Behav. , vol.7 , pp. 288-299
    • Hodges, A.1    Hughes, G.2    Brooks, S.3    Elliston, L.4    Holmans, P.5    Dunnett, S.B.6    Jones, L.7
  • 71
    • 70350142444 scopus 로고    scopus 로고
    • Gene dysregulation in Huntington’s disease: REST, microRNAs and beyond
    • Johnson R, Buckley NJ. Gene dysregulation in Huntington’s disease: REST, microRNAs and beyond. Neuromol Med. 2009;11:183-99.
    • (2009) Neuromol Med. , vol.11 , pp. 183-199
    • Johnson, R.1    Buckley, N.J.2
  • 73
    • 34047130812 scopus 로고    scopus 로고
    • Role of brain-derived neurotrophic factor in Huntington’s disease
    • Zuccato C, Cattaneo E. Role of brain-derived neurotrophic factor in Huntington’s disease. Prog Neurobiol. 2007;81:294-330.
    • (2007) Prog Neurobiol. , vol.81 , pp. 294-330
    • Zuccato, C.1    Cattaneo, E.2
  • 74
    • 34347364706 scopus 로고    scopus 로고
    • Widespread disruption of repressor element-1 silencing transcription factor/neuron-restrictive silencer factor occupancy at its target genes in Huntington’s disease
    • Zuccato C, Belyaev N, Conforti P, Ooi L, Tartari M, Papadimou E, MacDonald M, Fossale E, Zeitlin S, Buckley N, Cattaneo E. 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;27:6972-83.
    • (2007) J Neurosci. , vol.27 , pp. 6972-6983
    • Zuccato, C.1    Belyaev, N.2    Conforti, P.3    Ooi, L.4    Tartari, M.5    Papadimou, E.6    MacDonald, M.7    Fossale, E.8    Zeitlin, S.9    Buckley, N.10    Cattaneo, E.11
  • 75
    • 33749042331 scopus 로고    scopus 로고
    • Transcriptional repression of PGC-1alpha by mutant huntingtin leads to mitochondrial dysfunction and neurodegeneration
    • Cui L, Jeong H, Borovecki F, Parkhurst CN, Tanese N, Krainc D. Transcriptional repression of PGC-1alpha by mutant huntingtin leads to mitochondrial dysfunction and neurodegeneration. Cell. 2006;127:59-69.
    • (2006) Cell. , vol.127 , pp. 59-69
    • Cui, L.1    Jeong, H.2    Borovecki, F.3    Parkhurst, C.N.4    Tanese, N.5    Krainc, D.6
  • 76
    • 77955017449 scopus 로고    scopus 로고
    • Impairment of PGC-1alpha expression, neuropathology and hepatic steatosis in a transgenic mouse model of Huntington’s disease following chronic energy deprivation
    • Chaturvedi RK, Calingasan NY, Yang L, Hennessey T, Johri A, Beal MF. Impairment of PGC-1alpha expression, neuropathology and hepatic steatosis in a transgenic mouse model of Huntington’s disease following chronic energy deprivation. Hum Mol Genet. 2010;19:3190-205.
    • (2010) Hum Mol Genet. , vol.19 , pp. 3190-3205
    • Chaturvedi, R.K.1    Calingasan, N.Y.2    Yang, L.3    Hennessey, T.4    Johri, A.5    Beal, M.F.6
  • 78
    • 70349640049 scopus 로고    scopus 로고
    • Protein turnover and inclusion body formation
    • Mitra S, Tsvetkov AS, Finkbeiner S. Protein turnover and inclusion body formation. Autophagy. 2009;5: 1037-8.
    • (2009) Autophagy. , vol.5 , pp. 1037-1038
    • Mitra, S.1    Tsvetkov, A.S.2    Finkbeiner, S.3
  • 79
    • 0034754875 scopus 로고    scopus 로고
    • Accumulation of mutant huntingtin fragments in aggresome-like inclusion bodies as a result of insufficient protein degradation
    • Waelter S, Boeddrich A, Lurz R, Scherzinger E, Lueder G, Lehrach H, Wanker EE. Accumulation of mutant huntingtin fragments in aggresome-like inclusion bodies as a result of insufficient protein degradation. Mol Biol Cell. 2001;12:1393-407.
    • (2001) Mol Biol Cell. , vol.12 , pp. 1393-1407
    • Waelter, S.1    Boeddrich, A.2    Lurz, R.3    Scherzinger, E.4    Lueder, G.5    Lehrach, H.6    Wanker, E.E.7
  • 80
    • 1842766144 scopus 로고    scopus 로고
    • Eukaryotic proteasomes cannot digest polyglutamine sequences and release them during degradation of polyglutamine-containing proteins
    • Venkatraman P, Wetzel R, Tanaka M, Nukina N, Goldberg AL. Eukaryotic proteasomes cannot digest polyglutamine sequences and release them during degradation of polyglutamine-containing proteins. Mol Cell. 2004;14:95-104.
    • (2004) Mol Cell. , vol.14 , pp. 95-104
    • Venkatraman, P.1    Wetzel, R.2    Tanaka, M.3    Nukina, N.4    Goldberg, A.L.5
  • 82
    • 80053563760 scopus 로고    scopus 로고
    • Induction of autophagy with catalytic mTOR inhibitors reduces huntingtin aggregates in a neuronal cell model
    • Roscic A, Baldo B, Crochemore C, Marcellin D, Paganetti P. Induction of autophagy with catalytic mTOR inhibitors reduces huntingtin aggregates in a neuronal cell model. J Neurochem. 2011;119:398-407.
    • (2011) J Neurochem. , vol.119 , pp. 398-407
    • Roscic, A.1    Baldo, B.2    Crochemore, C.3    Marcellin, D.4    Paganetti, P.5
  • 83
    • 0037418339 scopus 로고    scopus 로고
    • Dietary restriction normalizes glucose metabolism and BDNF levels, slows disease progression, and increases survival in huntingtin mutant mice
    • Duan W, Guo Z, Jiang H, Ware M, Li X-J, Mattson MP. Dietary restriction normalizes glucose metabolism and BDNF levels, slows disease progression, and increases survival in huntingtin mutant mice. Proc Natl Acad Sci U S A. 2003;100:2911-6.
    • (2003) Proc Natl Acad Sci U S A. , vol.100 , pp. 2911-2916
    • Duan, W.1    Guo, Z.2    Jiang, H.3    Ware, M.4    Li, X.-J.5    Mattson, M.P.6
  • 84
    • 35448994487 scopus 로고    scopus 로고
    • Huntingtin has a membrane association signal that can modulate huntingtin aggregation, nuclear entry and toxicity
    • Atwal RS, Xia J, Pinchev D, Taylor J, Epand RM, Truant R. Huntingtin has a membrane association signal that can modulate huntingtin aggregation, nuclear entry and toxicity. Hum Mol Genet. 2007;16:2600-15.
    • (2007) Hum Mol Genet. , vol.16 , pp. 2600-2615
    • Atwal, R.S.1    Xia, J.2    Pinchev, D.3    Taylor, J.4    Epand, R.M.5    Truant, R.6
  • 85
    • 0034307476 scopus 로고    scopus 로고
    • Huntingtin expression stimulates endosomal-lysosomal activity, endosome tubulation, and autophagy
    • Kegel KB, Kim M, Sapp E, McIntyre C, Castaño JG, Aronin N, DiFiglia M. Huntingtin expression stimulates endosomal-lysosomal activity, endosome tubulation, and autophagy. J Neurosci. 2000;20:7268-78.
    • (2000) J Neurosci. , vol.20 , pp. 7268-7278
    • Kegel, K.B.1    Kim, M.2    Sapp, E.3    McIntyre, C.4    Castaño, J.G.5    Aronin, N.6    DiFiglia, M.7
  • 88
    • 77957565855 scopus 로고    scopus 로고
    • Age at onset in Huntington’s disease is modified by the autophagy pathway: Implication of the V471A polymorphism in Atg7
    • Metzger S, Saukko M, Van Che H, Tong L, Puder Y, Riess O, Nguyen HP. Age at onset in Huntington’s disease is modified by the autophagy pathway: Implication of the V471A polymorphism in Atg7. Hum Genet. 2010;128:453-9.
    • (2010) Hum Genet. , vol.128 , pp. 453-459
    • Metzger, S.1    Saukko, M.2    Van Che, H.3    Tong, L.4    Puder, Y.5    Riess, O.6    Nguyen, H.P.7
  • 92
    • 0033812145 scopus 로고    scopus 로고
    • Effect of exogenous and endogenous antioxidants on 3- nitropionic acid-induced in vivo oxidative stress and striatal lesions: Insights into Huntington’s disease
    • Fontaine MA, Geddes JW, Banks A, Butterfield DA. Effect of exogenous and endogenous antioxidants on 3- nitropionic acid-induced in vivo oxidative stress and striatal lesions: insights into Huntington’s disease. J Neurochem. 2000;75:1709-15.
    • (2000) J Neurochem. , vol.75 , pp. 1709-1715
    • Fontaine, M.A.1    Geddes, J.W.2    Banks, A.3    Butterfield, D.A.4
  • 93
    • 84872855026 scopus 로고    scopus 로고
    • 3-Nitropropionic acid induces autophagy by forming mitochondrial permeability transition pores rather than activating the mitochondrial fission pathway
    • Solesio ME, Saez-Atienzar S, Jordan J, Galindo MF. 3-Nitropropionic acid induces autophagy by forming mitochondrial permeability transition pores rather than activating the mitochondrial fission pathway. Br J Pharmacol. 2013;168(1):63-75.
    • (2013) Br J Pharmacol. , vol.168 , Issue.1 , pp. 63-75
    • Solesio, M.E.1    Saez-Atienzar, S.2    Jordan, J.3    Galindo, M.F.4
  • 94
    • 1442274934 scopus 로고    scopus 로고
    • Mitochondrial calcium, oxidative stress and apoptosis in a neurodegenerative disease model induced by 3-nitropropionic acid
    • Rosenstock TR, Carvalho ACP, Jurkiewicz A, Frussa-Filho R, Smaili SS. Mitochondrial calcium, oxidative stress and apoptosis in a neurodegenerative disease model induced by 3-nitropropionic acid. J Neurochem. 2004;88:1220-8.
    • (2004) J Neurochem. , vol.88 , pp. 1220-1228
    • Rosenstock, T.R.1    Carvalho, A.C.P.2    Jurkiewicz, A.3    Frussa-Filho, R.4    Smaili, S.S.5
  • 95
    • 79960079717 scopus 로고    scopus 로고
    • The striatum is highly susceptible to mitochondrial oxidative phosphorylation dysfunctions
    • Pickrell AM, Fukui H, Wang X, Pinto M, Moraes CT. The striatum is highly susceptible to mitochondrial oxidative phosphorylation dysfunctions. J Neurosci. 2011;31:9895-904.
    • (2011) J Neurosci. , vol.31 , pp. 9895-9904
    • Pickrell, A.M.1    Fukui, H.2    Wang, X.3    Pinto, M.4    Moraes, C.T.5
  • 97
    • 0034994207 scopus 로고    scopus 로고
    • Early degenerative changes in transgenic mice expressing mutant huntingtin involve dendritic abnormalities but no impairment of mitochondrial energy production
    • Guidetti P, Charles V, Chen EY, Reddy PH, Kordower JH, Whetsell WO, Schwarcz R, Tagle DA. Early degenerative changes in transgenic mice expressing mutant huntingtin involve dendritic abnormalities but no impairment of mitochondrial energy production. Experimental Neurology. 2001;169:340-50.
    • (2001) Experimental Neurology. , vol.169 , pp. 340-350
    • Guidetti, P.1    Charles, V.2    Chen, E.Y.3    Reddy, P.H.4    Kordower, J.H.5    Whetsell, W.O.6    Schwarcz, R.7    Tagle, D.A.8
  • 101
    • 0035668684 scopus 로고    scopus 로고
    • Increased oxidative damage to DNA in a transgenic mouse model of Huntington’s disease
    • Bogdanov MB, Andreassen OA, Dedeoglu A, Ferrante RJ, Beal MF. Increased oxidative damage to DNA in a transgenic mouse model of Huntington’s disease. J Neurochem. 2001;79:1246-9.
    • (2001) J Neurochem. , vol.79 , pp. 1246-1249
    • Bogdanov, M.B.1    Andreassen, O.A.2    Dedeoglu, A.3    Ferrante, R.J.4    Beal, M.F.5
  • 102
    • 22544469454 scopus 로고    scopus 로고
    • Increased glutathione levels in cortical and striatal mitochondria of the R6/2 Huntington’s disease mouse model
    • Choo YS, Mao Z, Johnson GVW, Lesort M. Increased glutathione levels in cortical and striatal mitochondria of the R6/2 Huntington’s disease mouse model. Neurosci Lett. 2005;386:63-8.
    • (2005) Neurosci Lett. , vol.386 , pp. 63-68
    • Choo, Y.S.1    Mao, Z.2    Johnson, G.V.W.3    Lesort, M.4
  • 103
    • 0031797267 scopus 로고    scopus 로고
    • Increased vulnerability to 3-nitropropionic acid in an animal model of Huntington’s disease
    • Bogdanov MB, Ferrante RJ, Kuemmerle S, Klivenyi P, Beal MF. Increased vulnerability to 3-nitropropionic acid in an animal model of Huntington’s disease. J Neurochem. 1998;71:2642-4.
    • (1998) J Neurochem. , vol.71 , pp. 2642-2644
    • Bogdanov, M.B.1    Ferrante, R.J.2    Kuemmerle, S.3    Klivenyi, P.4    Beal, M.F.5
  • 109
    • 0035166814 scopus 로고    scopus 로고
    • Dynamin-related protein Drp1 is required for mitochondrial division in mammalian cells
    • Smirnova E, Griparic L, Shurland DL, van der Bliek AM. Dynamin-related protein Drp1 is required for mitochondrial division in mammalian cells. Mol Biol Cell. 2001;12:2245-56.
    • (2001) Mol Biol Cell. , vol.12 , pp. 2245-2256
    • Smirnova, E.1    Griparic, L.2    Shurland, D.L.3    van der Bliek, A.M.4
  • 111
    • 79952585486 scopus 로고    scopus 로고
    • Abnormal mitochondrial dynamics, mitochondrial loss and mutant huntingtin oligomers in Huntington’s disease: Implications for selective neuronal damage
    • Shirendeb U, Reddy AP, Manczak M, Calkins MJ, Mao P, Tagle DA, Reddy PH. Abnormal mitochondrial dynamics, mitochondrial loss and mutant huntingtin oligomers in Huntington’s disease: Implications for selective neuronal damage. Hum Mol Genet. 2011;20:1438-55.
    • (2011) Hum Mol Genet. , vol.20 , pp. 1438-1455
    • Shirendeb, U.1    Reddy, A.P.2    Manczak, M.3    Calkins, M.J.4    Mao, P.5    Tagle, D.A.6    Reddy, P.H.7
  • 112
    • 84855395163 scopus 로고    scopus 로고
    • Mutant huntingtin" s interaction with mitochondrial protein Drp1 impairs mitochondrial biogenesis and causes defective axonal transport and synaptic degeneration in Huntington"s disease
    • Shirendeb UP, Calkins MJ, Manczak M, Anekonda V, Dufour B, McBride JL, Mao P, Reddy PH. Mutant huntingtin" s interaction with mitochondrial protein Drp1 impairs mitochondrial biogenesis and causes defective axonal transport and synaptic degeneration in Huntington"s disease. Hum Mol Genet. 2012;21:406-20.
    • (2012) Hum Mol Genet. , vol.21 , pp. 406-420
    • Shirendeb, U.P.1    Calkins, M.J.2    Manczak, M.3    Anekonda, V.4    Dufour, B.5    McBride, J.L.6    Mao, P.7    Reddy, P.H.8
  • 115
    • 58949099388 scopus 로고    scopus 로고
    • Effects of overexpression of huntingtin proteins on mitochondrial integrity
    • Wang H, Lim PJ, Karbowski M, Monteiro MJ. Effects of overexpression of huntingtin proteins on mitochondrial integrity. Hum Mol Genet. 2009;18:737-52.
    • (2009) Hum Mol Genet. , vol.18 , pp. 737-752
    • Wang, H.1    Lim, P.J.2    Karbowski, M.3    Monteiro, M.J.4
  • 116
    • 34249092704 scopus 로고    scopus 로고
    • Calcium and neurodegeneration
    • Mattson MP. Calcium and neurodegeneration. Aging Cell. 2007;6:337-50.
    • (2007) Aging Cell. , vol.6 , pp. 337-350
    • Mattson, M.P.1
  • 118
    • 37249083913 scopus 로고    scopus 로고
    • Mitochondrial sensitivity and altered calcium handling underlie enhancedNMDA-induced apoptosis in YAC128 model of Huntington’s disease
    • Fernandes HB, Baimbridge KG, Church J, Hayden MR, Raymond LA. Mitochondrial sensitivity and altered calcium handling underlie enhancedNMDA-induced apoptosis in YAC128 model of Huntington’s disease. J Neurosci. 2007;27:13614-23.
    • (2007) J Neurosci. , vol.27 , pp. 13614-13623
    • Fernandes, H.B.1    Baimbridge, K.G.2    Church, J.3    Hayden, M.R.4    Raymond, L.A.5
  • 119
    • 0033382256 scopus 로고    scopus 로고
    • Ca(2+)-dependent permeability transition and complex I activity in lymphoblast mitochondria from normal individuals and patients with Huntington"s or Alzheimer"s disease
    • Panov A, Obertone T, Bennett-Desmelik J, Greenamyre JT. Ca(2+)-dependent permeability transition and complex I activity in lymphoblast mitochondria from normal individuals and patients with Huntington"s or Alzheimer"s disease. Ann N Y Acad Sci. 1999;893:365-8.
    • (1999) Ann N Y Acad Sci. , vol.893 , pp. 365-368
    • Panov, A.1    Obertone, T.2    Bennett-Desmelik, J.3    Greenamyre, J.T.4
  • 120
    • 0016230519 scopus 로고
    • Kainic acid: A powerful neurotoxic analogue of glutamate
    • Olney JW, Rhee V, Ho OL. Kainic acid: A powerful neurotoxic analogue of glutamate. Brain Res. 1974;77:507-12.
    • (1974) Brain Res. , vol.77 , pp. 507-512
    • Olney, J.W.1    Rhee, V.2    Ho, O.L.3
  • 121
    • 0017167057 scopus 로고
    • Lesion of striatal neurones with kainic acid provides a model for Huntington’s chorea
    • Coyle JT, Schwarcz R. Lesion of striatal neurones with kainic acid provides a model for Huntington’s chorea. Nature. 1976;263:244-6.
    • (1976) Nature. , vol.263 , pp. 244-246
    • Coyle, J.T.1    Schwarcz, R.2
  • 122
    • 0022446150 scopus 로고
    • Replication of the neurochemical characteristics of Huntington’s disease by quinolinic acid
    • Beal MF, Kowall NW, Ellison DW, Mazurek MF, Swartz KJ, Martin JB. Replication of the neurochemical characteristics of Huntington’s disease by quinolinic acid. Nature. 1986;321:168-71.
    • (1986) Nature. , vol.321 , pp. 168-171
    • Beal, M.F.1    Kowall, N.W.2    Ellison, D.W.3    Mazurek, M.F.4    Swartz, K.J.5    Martin, J.B.6
  • 123
    • 0026043565 scopus 로고
    • Chronic quinolinic acid lesions in rats closely resemble Huntington’s disease
    • Beal MF, Ferrante RJ, Swartz KJ, Kowall NW. Chronic quinolinic acid lesions in rats closely resemble Huntington’s disease. J Neurosci. 1991;11:1649-59.
    • (1991) J Neurosci. , vol.11 , pp. 1649-1659
    • Beal, M.F.1    Ferrante, R.J.2    Swartz, K.J.3    Kowall, N.W.4
  • 124
    • 0027396021 scopus 로고
    • Excitotoxin lesions in primates as a model for Huntington’s disease: Histopathologic and neurochemical characterization
    • Ferrante RJ, Kowall NW, Cipolloni PB, Storey E, Beal MF. Excitotoxin lesions in primates as a model for Huntington’s disease: Histopathologic and neurochemical characterization. Experimental Neurology. 1993;119:46-71.
    • (1993) Experimental Neurology. , vol.119 , pp. 46-71
    • Ferrante, R.J.1    Kowall, N.W.2    Cipolloni, P.B.3    Storey, E.4    Beal, M.F.5
  • 127
    • 58149477890 scopus 로고    scopus 로고
    • Hypoxic/ischemic conditions induce expression of the putative pro-death gene Clca1 via activation of extrasynaptic N-methyl-D-aspartate receptors
    • Wahl A-S, Buchthal B, Rode F, Bomholt SF, Freitag HE, Hardingham GE, Rønn LCB, Bading H. Hypoxic/ischemic conditions induce expression of the putative pro-death gene Clca1 via activation of extrasynaptic N-methyl-D-aspartate receptors. NSC. 2009;158:344-52.
    • (2009) NSC. , vol.158 , pp. 344-352
    • Wahl, A.-S.1    Buchthal, B.2    Rode, F.3    Bomholt, S.F.4    Freitag, H.E.5    Hardingham, G.E.6    Rønn, L.C.B.7    Bading, H.8
  • 128
    • 14844314896 scopus 로고    scopus 로고
    • Enhanced striatal NR2B-containing N-methyl-D-aspartate receptormediated synaptic currents in a mouse model of Huntington disease
    • Li L, Murphy TH, Hayden MR, Raymond LA. Enhanced striatal NR2B-containing N-methyl-D-aspartate receptormediated synaptic currents in a mouse model of Huntington disease. J Neurophysiol. 2004;92:2738-46.
    • (2004) J Neurophysiol. , vol.92 , pp. 2738-2746
    • Li, L.1    Murphy, T.H.2    Hayden, M.R.3    Raymond, L.A.4
  • 131
    • 63849154126 scopus 로고    scopus 로고
    • In Vivo Evidence for NMDA Receptor-Mediated Excitotoxicity in aMurine Genetic Model of Huntington Disease
    • HengMY, Detloff PJ, Wang PL, Tsien JZ, Albin RL. In Vivo Evidence for NMDA Receptor-Mediated Excitotoxicity in aMurine Genetic Model of Huntington Disease. J Neurosci. 2009;29:3200-5.
    • (2009) J Neurosci. , vol.29 , pp. 3200-3205
    • Heng, M.Y.1    Detloff, P.J.2    Wang, P.L.3    Tsien, J.Z.4    Albin, R.L.5
  • 132
    • 84865078592 scopus 로고    scopus 로고
    • Calpain and STriatal-Enriched protein tyrosine phosphatase (STEP) activation contribute to extrasynaptic NMDA receptor localization in a Huntington’s disease mouse model
    • Gladding CM, Sepers MD, Xu J, Zhang LYJ, Milnerwood AJ, Lombroso PJ, Raymond LA. Calpain and STriatal-Enriched protein tyrosine phosphatase (STEP) activation contribute to extrasynaptic NMDA receptor localization in a Huntington’s disease mouse model. Hum Mol Genet. 2012;21:3739-52.
    • (2012) Hum Mol Genet. , vol.21 , pp. 3739-3752
    • Gladding, C.M.1    Sepers, M.D.2    Xu, J.3    Zhang, L.Y.J.4    Milnerwood, A.J.5    Lombroso, P.J.6    Raymond, L.A.7
  • 135
    • 58149373434 scopus 로고    scopus 로고
    • Polyglutamine-Modulated Striatal Calpain Activity in YAC Transgenic Huntington Disease Mouse Model: Impact on NMDA Receptor Function andToxicity
    • Cowan CM, Fan MMY, Fan J, Shehadeh J, Zhang LYJ, Graham RK, Hayden MR, Raymond LA. Polyglutamine-Modulated Striatal Calpain Activity in YAC Transgenic Huntington Disease Mouse Model: Impact on NMDA Receptor Function andToxicity. J Neurosci. 2008;28:12725-35.
    • (2008) J Neurosci. , vol.28 , pp. 12725-12735
    • Cowan, C.M.1    Fan, M.M.Y.2    Fan, J.3    Shehadeh, J.4    Zhang, L.Y.J.5    Graham, R.K.6    Hayden, M.R.7    Raymond, L.A.8
  • 136
    • 0036315706 scopus 로고    scopus 로고
    • Mice overexpressing 70-kDa heat shock protein show increased resistance to malonate and 3-nitropropionic acid
    • Dedeoglu A, Ferrante RJ, Andreassen OA, Dillmann WH, Beal MF. Mice overexpressing 70-kDa heat shock protein show increased resistance to malonate and 3-nitropropionic acid. Experimental Neurology. 2002;176:262-5.
    • (2002) Experimental Neurology. , vol.176 , pp. 262-265
    • Dedeoglu, A.1    Ferrante, R.J.2    Andreassen, O.A.3    Dillmann, W.H.4    Beal, M.F.5
  • 137
    • 79551655290 scopus 로고    scopus 로고
    • Huntington’s Disease: Can Mice Lead the Way to Treatment?
    • Crook ZR, Housman DE. Huntington’s Disease: Can Mice Lead the Way to Treatment? Neuron. 2011;69:423-35.
    • (2011) Neuron. , vol.69 , pp. 423-435
    • Crook, Z.R.1    Housman, D.E.2
  • 140
    • 0346749473 scopus 로고    scopus 로고
    • Enhanced Akt signaling is an early pro-survival response that reflects N-methyl-D-aspartate receptor activation in Huntington’s disease knock-in striatal cells
    • Gines S, Ivanova E, Seong IS, Saura CA, Macdonald ME. Enhanced Akt signaling is an early pro-survival response that reflects N-methyl-D-aspartate receptor activation in Huntington’s disease knock-in striatal cells. J Biol Chem. 2003;278:50514-22.
    • (2003) J Biol Chem. , vol.278 , pp. 50514-50522
    • Gines, S.1    Ivanova, E.2    Seong, I.S.3    Saura, C.A.4    Macdonald, M.E.5
  • 144
    • 45049085513 scopus 로고    scopus 로고
    • Full length mutant huntingtin is required for altered Ca2+ signaling and apoptosis of striatal neurons in the YAC mouse model of Huntington’s disease
    • Zhang H, Li Q, Graham RK, Slow E, Hayden MR, Bezprozvanny I. Full length mutant huntingtin is required for altered Ca2+ signaling and apoptosis of striatal neurons in the YAC mouse model of Huntington’s disease. Neurobiology of Disease. 2008;31:80-8.
    • (2008) Neurobiology of Disease. , vol.31 , pp. 80-88
    • Zhang, H.1    Li, Q.2    Graham, R.K.3    Slow, E.4    Hayden, M.R.5    Bezprozvanny, I.6
  • 147
    • 78049342155 scopus 로고    scopus 로고
    • Phosphorylation of huntingtin at Ser421 in YAC128 neurons is associated with protection of YAC128 neurons fromNMDA-mediated excitotoxicity and is modulated by PP1 and PP2A
    • Metzler M, Gan L, Mazarei G, Graham RK, Liu L, Bissada N, Lu G, Leavitt BR, Hayden MR. Phosphorylation of huntingtin at Ser421 in YAC128 neurons is associated with protection of YAC128 neurons fromNMDA-mediated excitotoxicity and is modulated by PP1 and PP2A. J Neurosci. 2010;30:14318-29.
    • (2010) J Neurosci. , vol.30 , pp. 14318-14329
    • Metzler, M.1    Gan, L.2    Mazarei, G.3    Graham, R.K.4    Liu, L.5    Bissada, N.6    Lu, G.7    Leavitt, B.R.8    Hayden, M.R.9
  • 148
    • 43549092044 scopus 로고    scopus 로고
    • Calcineurin is involved in the early activation of NMDA-mediated cell death in mutant huntingtin knock-in striatal cells
    • Xifró X, García-Martínez JM, del Toro D, Alberch J, Pérez-Navarro E. Calcineurin is involved in the early activation of NMDA-mediated cell death in mutant huntingtin knock-in striatal cells. J Neurochem. 2008;105:1596-612.
    • (2008) J Neurochem. , vol.105 , pp. 1596-1612
    • Xifró, X.1    García-Martínez, J.M.2    del Toro, D.3    Alberch, J.4    Pérez-Navarro, E.5
  • 149
    • 57049184027 scopus 로고    scopus 로고
    • Phosphorylation of mutant huntingtin at S421 restores anterograde and retrograde transport in neurons
    • Zala D, Colin E, Rangone H, Liot G, Humbert S, Saudou F. Phosphorylation of mutant huntingtin at S421 restores anterograde and retrograde transport in neurons. Hum Mol Genet. 2008;17:3837-46.
    • (2008) Hum Mol Genet. , vol.17 , pp. 3837-3846
    • Zala, D.1    Colin, E.2    Rangone, H.3    Liot, G.4    Humbert, S.5    Saudou, F.6
  • 151
    • 49149112606 scopus 로고    scopus 로고
    • Huntingtin phosphorylation acts as a molecular switch for anterograde/retrograde transport in neurons
    • Colin E, Zala D, Liot G, Rangone H, Borrell-Pagès M, Li X-J, Saudou F, Humbert S. Huntingtin phosphorylation acts as a molecular switch for anterograde/retrograde transport in neurons. EMBO J. 2008;27:2124-34.
    • (2008) EMBO J. , vol.27 , pp. 2124-2134
    • Colin, E.1    Zala, D.2    Liot, G.3    Rangone, H.4    Borrell-Pagès, M.5    Li, X.-J.6    Saudou, F.7    Humbert, S.8
  • 152
    • 32544432052 scopus 로고    scopus 로고
    • Inhibition of calcineurin by FK506 protects against polyglutamine-huntingtin toxicity through an increase of huntingtin phosphorylation at S421
    • Pardo R, Colin E, Régulier E, Aebischer P, Déglon N, Humbert S, Saudou F. Inhibition of calcineurin by FK506 protects against polyglutamine-huntingtin toxicity through an increase of huntingtin phosphorylation at S421. J Neurosci. 2006;26:1635-45.
    • (2006) J Neurosci. , vol.26 , pp. 1635-1645
    • Pardo, R.1    Colin, E.2    Régulier, E.3    Aebischer, P.4    Déglon, N.5    Humbert, S.6    Saudou, F.7
  • 153
    • 78049525220 scopus 로고    scopus 로고
    • BDNFoverexpression in the forebrain rescues Huntington’s disease phenotypes in YAC128 mice
    • XieY, Hayden MR, Xu B. BDNFoverexpression in the forebrain rescues Huntington’s disease phenotypes in YAC128 mice. J Neurosci. 2010;30:14708-18.
    • (2010) J Neurosci. , vol.30 , pp. 14708-14718
    • Xie, Y.1    Hayden, M.R.2    Xu, B.3
  • 154
    • 72949098095 scopus 로고    scopus 로고
    • Genetic and pharmacological inhibition of calcineurin corrects the BDNF transport defect in Huntington’s disease
    • Pineda JR, Pardo R, Zala D, Yu H, Humbert S, Saudou F. Genetic and pharmacological inhibition of calcineurin corrects the BDNF transport defect in Huntington’s disease. Mol Brai. 2009;2:33.
    • (2009) Mol Brai. , vol.2 , pp. 33
    • Pineda, J.R.1    Pardo, R.2    Zala, D.3    Yu, H.4    Humbert, S.5    Saudou, F.6
  • 155
    • 4444350918 scopus 로고    scopus 로고
    • Brain-derived neurotrophic factor regulates the onset and severity of motor dysfunction associated with enkephalinergic neuronal degeneration in Huntington’s disease
    • Canals JM, Pineda JR, Torres-Peraza JF, Bosch M, Martín-Ibañez R, Muñoz MT, Mengod G, Ernfors P, Alberch J. Brain-derived neurotrophic factor regulates the onset and severity of motor dysfunction associated with enkephalinergic neuronal degeneration in Huntington’s disease. J Neurosci. 2004;24:7727-39.
    • (2004) J Neurosci. , vol.24 , pp. 7727-7739
    • Canals, J.M.1    Pineda, J.R.2    Torres-Peraza, J.F.3    Bosch, M.4    Martín-Ibañez, R.5    Muñoz, M.T.6    Mengod, G.7    Ernfors, P.8    Alberch, J.9
  • 156
    • 34948855498 scopus 로고    scopus 로고
    • Induction of neostriatal neurogenesis slows disease progression in a transgenic murine model of Huntington disease
    • Cho S-R, Benraiss A, Chmielnicki E, Samdani A, Economides A, Goldman SA. Induction of neostriatal neurogenesis slows disease progression in a transgenic murine model of Huntington disease. J Clin Invest. 2007;117:2889-902.
    • (2007) J Clin Invest. , vol.117 , pp. 2889-2902
    • Cho, S.-R.1    Benraiss, A.2    Chmielnicki, E.3    Samdani, A.4    Economides, A.5    Goldman, S.A.6
  • 157
    • 41749083721 scopus 로고    scopus 로고
    • Brainderived neurotrophic factor over-expression in the forebrain ameliorates Huntington’s disease phenotypes in mice
    • Gharami K, Xie Y, An JJ, Tonegawa S, Xu B. Brainderived neurotrophic factor over-expression in the forebrain ameliorates Huntington’s disease phenotypes in mice. J Neurochem. 2008;105:369-79.
    • (2008) J Neurochem. , vol.105 , pp. 369-379
    • Gharami, K.1    Xie, Y.2    An, J.J.3    Tonegawa, S.4    Xu, B.5
  • 158
    • 85009332113 scopus 로고    scopus 로고
    • Unified Huntington’s Disease Rating Scale: Reliability and consistency. Huntington Study Group
    • Group THS. Unified Huntington’s Disease Rating Scale: Reliability and consistency. Huntington Study Group. Mov Disord. 1996;11:136-42.
    • (1996) Mov Disord. , vol.11 , pp. 136-142
    • Group, T.H.S.1
  • 159
    • 2642666489 scopus 로고    scopus 로고
    • Cognitive deficits in Huntington’s disease are predicted by dopaminergic PET markers and brain volumes
    • Bäckman L, Robins-Wahlin TB, Lundin A, Ginovart N, Farde L. Cognitive deficits in Huntington’s disease are predicted by dopaminergic PET markers and brain volumes. Brain. 1997;120(Pt 12):2207-17.
    • (1997) Brain. , vol.120 , pp. 2207-2217
    • Bäckman, L.1    Robins-Wahlin, T.B.2    Lundin, A.3    Ginovart, N.4    Farde, L.5
  • 160
  • 161
    • 0031594146 scopus 로고    scopus 로고
    • [11C]raclopride-PET studies of the Huntington’s disease rate of progression: Relevance of the trinucleotide repeat length
    • Antonini A, Leenders KL, Eidelberg D. [11C]raclopride-PET studies of the Huntington’s disease rate of progression: Relevance of the trinucleotide repeat length. Ann Neurol. 1998;43:253-5.
    • (1998) Ann Neurol. , vol.43 , pp. 253-255
    • Antonini, A.1    Leenders, K.L.2    Eidelberg, D.3
  • 165
    • 33747602312 scopus 로고    scopus 로고
    • Hypothalamic-endocrine aspects in Huntington’s disease
    • Petersén A, Björkqvist M. Hypothalamic-endocrine aspects in Huntington’s disease. Eur J Neurosci. 2006;24: 961-7.
    • (2006) Eur J Neurosci. , vol.24 , pp. 961-967
    • Petersén, A.1    Björkqvist, M.2
  • 167
    • 84924914420 scopus 로고    scopus 로고
    • New Perspectives on the Neuropathology in Huntington’s Disease in the Human Brain and its Relation to Symptom Variation
    • Waldvogel HJ, Kim EH, Thu DC, Tippett LJ, Faull RL. New Perspectives on the Neuropathology in Huntington’s Disease in the Human Brain and its Relation to Symptom Variation. J Huntingtons Dis. 2012;1:143-53.
    • (2012) J Huntingtons Dis. , vol.1 , pp. 143-153
    • Waldvogel, H.J.1    Kim, E.H.2    Thu, D.C.3    Tippett, L.J.4    Faull, R.L.5
  • 168
    • 84875940946 scopus 로고    scopus 로고
    • Correlations of Behavioral Deficits with Brain Pathology Assessed through Longitudinal MRI and Histopathology in the R6/2 Mouse Model of HD
    • Rattray I, Smith E, Gale R, Matsumoto K, Bates GP, Modo M. Correlations of Behavioral Deficits with Brain Pathology Assessed through Longitudinal MRI and Histopathology in the R6/2 Mouse Model of HD. PLoS ONE. 2013;8: e60012.
    • (2013) PLoS ONE. , vol.8 , pp. e60012
    • Rattray, I.1    Smith, E.2    Gale, R.3    Matsumoto, K.4    Bates, G.P.5    Modo, M.6
  • 169
    • 84862806580 scopus 로고    scopus 로고
    • Spatiotemporal mapping of brain atrophy in mouse models of Huntington’s disease using longitudinal in vivo magnetic resonance imaging
    • Aggarwal M, Duan W, Hou Z, Rakesh N, Peng Q, Ross CA, Miller MI, Mori S, Zhang J. Spatiotemporal mapping of brain atrophy in mouse models of Huntington’s disease using longitudinal in vivo magnetic resonance imaging. Neuroimage. 2012;60:2086-95.
    • (2012) Neuroimage. , vol.60 , pp. 2086-2095
    • Aggarwal, M.1    Duan, W.2    Hou, Z.3    Rakesh, N.4    Peng, Q.5    Ross, C.A.6    Miller, M.I.7    Mori, S.8    Zhang, J.9
  • 173
    • 84860835246 scopus 로고    scopus 로고
    • 8OHdG as a marker for Huntington disease progression
    • Long JD, Matson WR, Juhl AR, Leavitt BR, Paulsen JS, the PREDICT-HD Investigators and Coordinators of the Huntington Study Group. 8OHdG as a marker for Huntington disease progression. Neurobiology of Disease. 2012;46(3):625-34.
    • (2012) Neurobiology of Disease. , vol.46 , Issue.3 , pp. 625-634
    • Long, J.D.1    Matson, W.R.2    Juhl, A.R.3    Leavitt, B.R.4    Paulsen, J.S.5
  • 178
    • 0022904060 scopus 로고
    • Attitudes of persons at risk for Huntington disease toward predictive testing
    • Kessler S, Field T, Worth L, Mosbarger H. Attitudes of persons at risk for Huntington disease toward predictive testing. Am J Med Genet. 1987;26:259-70.
    • (1987) Am J Med Genet. , vol.26 , pp. 259-270
    • Kessler, S.1    Field, T.2    Worth, L.3    Mosbarger, H.4
  • 186
    • 84859480672 scopus 로고    scopus 로고
    • Striatal volume contributes to the prediction of onset of Huntington disease in incident cases
    • Aylward EH, Liu D, Nopoulos PC, Ross CA, Pierson RK, Mills JA, Long JD, Paulsen JS, Predict-HD Investigators and Coordinators of the Huntington Study Group. Striatal volume contributes to the prediction of onset of Huntington disease in incident cases. Biol Psychiatry. 2012;71:822-8.
    • (2012) Biol Psychiatry. , vol.71 , pp. 822-828
    • Aylward, E.H.1    Liu, D.2    Nopoulos, P.C.3    Ross, C.A.4    Pierson, R.K.5    Mills, J.A.6    Long, J.D.7    Paulsen, J.S.8
  • 188
    • 84862658038 scopus 로고    scopus 로고
    • (18)F-FDG PET uptake in the pre-Huntington disease caudate affects the time-to-onset independently of CAG expansion size
    • Ciarmiello A, Giovacchini G, Orobello S, Bruselli L, Elifani F, Squitieri F. (18)F-FDG PET uptake in the pre-Huntington disease caudate affects the time-to-onset independently of CAG expansion size. Eur J Nucl Med Mol Imaging. 2012;39(6):1030-36.
    • (2012) Eur J Nucl Med Mol Imaging. , vol.39 , Issue.6 , pp. 1030-1036
    • Ciarmiello, A.1    Giovacchini, G.2    Orobello, S.3    Bruselli, L.4    Elifani, F.5    Squitieri, F.6
  • 189
    • 36549036813 scopus 로고    scopus 로고
    • Psychiatric symptoms in Huntington’s disease before diagnosis: The predict-HD study
    • Duff K, Paulsen JS, Beglinger LJ, Langbehn DR, Stout JC, PREDICT-HD Investigators of the Huntington Study Group. Psychiatric symptoms in Huntington’s disease before diagnosis: The predict-HD study. Biol Psychiatry. 2007;62:1341-6.
    • (2007) Biol Psychiatry. , vol.62 , pp. 1341-1346
    • Duff, K.1    Paulsen, J.S.2    Beglinger, L.J.3    Langbehn, D.R.4    Stout, J.C.5
  • 192
    • 84862829257 scopus 로고    scopus 로고
    • Cognitive domains that predict time to diagnosis in prodromal Huntington disease
    • Harrington DL, Smith MM, Zhang Y, Carlozzi NE, Paulsen JS, the PREDICT-HD Investigators of the Huntington Study Group. Cognitive domains that predict time to diagnosis in prodromal Huntington disease. J Neurol Neurosurg Psychiatr. 2012;83(6):612-19.
    • (2012) J Neurol Neurosurg Psychiatr. , vol.83 , Issue.6 , pp. 612-619
    • Harrington, D.L.1    Smith, M.M.2    Zhang, Y.3    Carlozzi, N.E.4    Paulsen, J.S.5
  • 195
    • 0035740965 scopus 로고    scopus 로고
    • Neuroprotective therapy for Huntington’s disease: New prospects and challenges
    • Hersch SM, Rosas HD. Neuroprotective therapy for Huntington’s disease: New prospects and challenges. Expert review of neurotherapeutics. 2001;1:111-8.
    • (2001) Expert review of neurotherapeutics. , vol.1 , pp. 111-118
    • Hersch, S.M.1    Rosas, H.D.2
  • 196
    • 84855922033 scopus 로고    scopus 로고
    • Pharmacologic approaches to the treatment of Huntington’s disease
    • Venuto CS, McGarry A, Ma Q, Kieburtz K. Pharmacologic approaches to the treatment of Huntington’s disease. Mov Disord. 2012;27:31-41.
    • (2012) Mov Disord. , vol.27 , pp. 31-41
    • Venuto, C.S.1    McGarry, A.2    Ma, Q.3    Kieburtz, K.4
  • 197
    • 34250659732 scopus 로고    scopus 로고
    • Huntington’s disease: Pathological mechanisms and therapeutic strategies
    • Ramaswamy SS, Shannon KMK, Kordower JHJ. Huntington’s disease: Pathological mechanisms and therapeutic strategies. Cell Transplant. 2007;16:301-12.
    • (2007) Cell Transplant. , vol.16 , pp. 301-312
    • Ramaswamy, S.S.1    Shannon, K.M.K.2    Kordower, J.H.J.3
  • 199
    • 33645798913 scopus 로고    scopus 로고
    • Tetrabenazine as antichorea therapy in Huntington disease: A randomized controlled trial
    • Huntington Study, Group. Tetrabenazine as antichorea therapy in Huntington disease: A randomized controlled trial. Neurology. 2006;66:366-72.
    • (2006) Neurology. , vol.66 , pp. 366-372
  • 200
    • 74549128476 scopus 로고    scopus 로고
    • Tetrabenazine as anti-chorea therapy in Huntington disease: An open-label continuation study. Huntington Study Group/TETRA-HD Investigators
    • Frank S. Tetrabenazine as anti-chorea therapy in Huntington disease: An open-label continuation study. Huntington Study Group/TETRA-HD Investigators. BMC Neuro. 2009;9:62.
    • (2009) BMC Neuro. , vol.9 , pp. 62
    • Frank, S.1
  • 201
    • 33645872123 scopus 로고    scopus 로고
    • Tetrabenazine in the treatment of hyperkinetic movement disorders
    • Kenney C, Jankovic J. Tetrabenazine in the treatment of hyperkinetic movement disorders. Expert review of neurotherapeutics. 2006;6:7-17.
    • (2006) Expert review of neurotherapeutics. , vol.6 , pp. 7-17
    • Kenney, C.1    Jankovic, J.2
  • 202
    • 0019963368 scopus 로고
    • Tetrabenazine has properties of a dopamine receptor antagonist
    • Login IS, Cronin MJ, MacLeod RM. Tetrabenazine has properties of a dopamine receptor antagonist. Ann Neurol. 1982;12:257-62.
    • (1982) Ann Neurol. , vol.12 , pp. 257-262
    • Login, I.S.1    Cronin, M.J.2    MacLeod, R.M.3
  • 203
    • 0021251067 scopus 로고
    • Tetrabenazineinduced depletion of brain monoamines: Characterization and interaction with selected antidepressants
    • Pettibone DJ, Totaro JA, Pflueger AB. Tetrabenazineinduced depletion of brain monoamines: Characterization and interaction with selected antidepressants. Eur. J. Pharmacol. 1984;102:425-30.
    • (1984) Eur. J. Pharmacol. , vol.102 , pp. 425-430
    • Pettibone, D.J.1    Totaro, J.A.2    Pflueger, A.B.3
  • 204
    • 84884724757 scopus 로고    scopus 로고
    • A randomized, double-blind, placebo-controlled trial of pridopidine in Huntington’s disease
    • The Huntington Study Group HART Investigators. A randomized, double-blind, placebo-controlled trial of pridopidine in Huntington’s disease. Mov Disord. 2013;28(10):1407-15.
    • (2013) Mov Disord. , vol.28 , Issue.10 , pp. 1407-1415
  • 205
    • 84860385532 scopus 로고    scopus 로고
    • An evidence-based approach in the treatment of Huntington’s disease
    • Mestre TA, Ferreira JJ. An evidence-based approach in the treatment of Huntington’s disease. Parkinsonism Relat Disord. 2012;18:316-20.
    • (2012) Parkinsonism Relat Disord. , vol.18 , pp. 316-320
    • Mestre, T.A.1    Ferreira, J.J.2
  • 206
  • 211
    • 85009226418 scopus 로고    scopus 로고
    • A randomized, placebocontrolled trial of coenzyme Q10 and remacemide in Huntington’s disease
    • Huntington Study, Group. A randomized, placebocontrolled trial of coenzyme Q10 and remacemide in Huntington’s disease. Neurology. 2001;57:397-404.
    • (2001) Neurology. , vol.57 , pp. 397-404
  • 214
    • 0037148140 scopus 로고    scopus 로고
    • A randomised, placebo-controlled, double blind study of treatment of Huntington’s disease with unsaturated fatty acids
    • Vaddadi KS, Soosai E, Chiu E, Dingjan P. A randomised, placebo-controlled, double blind study of treatment of Huntington’s disease with unsaturated fatty acids. NeuroReport. 2002;13:29-33.
    • (2002) NeuroReport. , vol.13 , pp. 29-33
    • Vaddadi, K.S.1    Soosai, E.2    Chiu, E.3    Dingjan, P.4
  • 215
    • 33646248382 scopus 로고    scopus 로고
    • CYTE-I-HD: Phase I dose finding and tolerability study of cysteamine (Cystagon) in Huntington’s disease
    • Dubinsky R, Gray C. CYTE-I-HD: Phase I dose finding and tolerability study of cysteamine (Cystagon) in Huntington’s disease. Mov Disord. 2006;21:530-3.
    • (2006) Mov Disord. , vol.21 , pp. 530-533
    • Dubinsky, R.1    Gray, C.2
  • 216
    • 0036172346 scopus 로고    scopus 로고
    • Prolonged survival and decreased abnormal movements in transgenic model of Huntington disease, with administration of the transglutaminase inhibitor cystamine
    • Karpuj MV, Becher MW, Springer JE, Chabas D, Youssef S, Pedotti R, Mitchell D, Steinman L. Prolonged survival and decreased abnormal movements in transgenic model of Huntington disease, with administration of the transglutaminase inhibitor cystamine. Nat Med. 2002;8:143-9.
    • (2002) Nat Med. , vol.8 , pp. 143-149
    • Karpuj, M.V.1    Becher, M.W.2    Springer, J.E.3    Chabas, D.4    Youssef, S.5    Pedotti, R.6    Mitchell, D.7    Steinman, L.8
  • 219
    • 0035363805 scopus 로고    scopus 로고
    • Geldanamycin activates a heat shock response and inhibits huntingtin aggregation in a cell culture model of Huntington’s disease
    • Sittler A, Lurz R, Lueder G, Priller J, Lehrach H, Hayer-Hartl MK, Hartl FU, Wanker EE. Geldanamycin activates a heat shock response and inhibits huntingtin aggregation in a cell culture model of Huntington’s disease. Hum Mol Genet. 2001;10:1307-15.
    • (2001) Hum Mol Genet. , vol.10 , pp. 1307-1315
    • Sittler, A.1    Lurz, R.2    Lueder, G.3    Priller, J.4    Lehrach, H.5    Hayer-Hartl, M.K.6    Hartl, F.U.7    Wanker, E.E.8
  • 221
    • 0029812759 scopus 로고    scopus 로고
    • Polyubiquitination and proteasomal degradation of the p185c-erbB-2 receptor protein-tyrosine kinase induced by geldanamycin
    • Mimnaugh EG, Chavany C, Neckers L. Polyubiquitination and proteasomal degradation of the p185c-erbB-2 receptor protein-tyrosine kinase induced by geldanamycin. J Biol Chem. 1996;271:22796-801.
    • (1996) J Biol Chem. , vol.271 , pp. 22796-22801
    • Mimnaugh, E.G.1    Chavany, C.2    Neckers, L.3
  • 222
    • 2342625412 scopus 로고    scopus 로고
    • Ubiquitination and proteasomal degradation of nucleophosminanaplastic lymphoma kinase induced by 17-allylaminodemethoxygeldanamycin: Role of the co-chaperone carboxyl heat shock protein 70-interacting protein
    • Bonvini P, Dalla Rosa H, Vignes N, Rosolen A. Ubiquitination and proteasomal degradation of nucleophosminanaplastic lymphoma kinase induced by 17-allylaminodemethoxygeldanamycin: Role of the co-chaperone carboxyl heat shock protein 70-interacting protein. Cancer Res. 2004;64:3256-64.
    • (2004) Cancer Res. , vol.64 , pp. 3256-3264
    • Bonvini, P.1    Dalla Rosa, H.2    Vignes, N.3    Rosolen, A.4
  • 223
    • 54449101793 scopus 로고    scopus 로고
    • Heat shock transcription factor 1-activating compounds suppress polyglutamine-induced neurodegeneration through induction of multiple molecular chaperones
    • Fujikake N, Nagai Y, Popiel HA, Okamoto Y, Yamaguchi M, Toda T. Heat shock transcription factor 1-activating compounds suppress polyglutamine-induced neurodegeneration through induction of multiple molecular chaperones. J Biol Chem. 2008;283:26188-97.
    • (2008) J Biol Chem. , vol.283 , pp. 26188-26197
    • Fujikake, N.1    Nagai, Y.2    Popiel, H.A.3    Okamoto, Y.4    Yamaguchi, M.5    Toda, T.6
  • 226
    • 77953665285 scopus 로고    scopus 로고
    • The mTOR kinase inhibitor Everolimus decreases S6 kinase phosphorylation but fails to reduce mutant huntingtin levels in brain and is not neuroprotective in the R6/2 mouse model of Huntington’s disease
    • Fox JH, Connor T, Chopra V, Dorsey K, Kama JA, Bleckmann D, Betschart C, Hoyer D, Frentzel S, Difiglia M, Paganetti P, Hersch SM. The mTOR kinase inhibitor Everolimus decreases S6 kinase phosphorylation but fails to reduce mutant huntingtin levels in brain and is not neuroprotective in the R6/2 mouse model of Huntington’s disease. Mol Neurodegene. 2010;5:26.
    • (2010) Mol Neurodegene. , vol.5 , pp. 26
    • Fox, J.H.1    Connor, T.2    Chopra, V.3    Dorsey, K.4    Kama, J.A.5    Bleckmann, D.6    Betschart, C.7    Hoyer, D.8    Frentzel, S.9    Difiglia, M.10    Paganetti, P.11    Hersch, S.M.12
  • 231
    • 10744228913 scopus 로고    scopus 로고
    • Minocycline is protective in a mouse model of Huntington’s disease
    • 841-author reply
    • Hersch S, Fink K, Vonsattel J-P, Friedlander RM. Minocycline is protective in a mouse model of Huntington’s disease. Ann Neurol. 2003;54:841-author reply 842-3.
    • (2003) Ann Neurol. , vol.54 , pp. 842-843
    • Hersch, S.1    Fink, K.2    Vonsattel, J.-P.3    Friedlander, R.M.4
  • 232
    • 78649369122 scopus 로고    scopus 로고
    • Afutility study of minocycline in Huntington’s disease
    • Huntington Study Group DOMINO, Investigators. Afutility study of minocycline in Huntington’s disease. Mov Disord. 2010;25:2219-24.
    • (2010) Mov Disord. , vol.25 , pp. 2219-2224
  • 235
    • 77955643169 scopus 로고    scopus 로고
    • Molecular mechanisms and potential therapeutical targets in Huntington’s disease
    • Zuccato C, Valenza M, Cattaneo E. Molecular mechanisms and potential therapeutical targets in Huntington’s disease. Physiol Rev. 2010;90:905-81.
    • (2010) Physiol Rev. , vol.90 , pp. 905-981
    • Zuccato, C.1    Valenza, M.2    Cattaneo, E.3
  • 239
    • 66749119970 scopus 로고    scopus 로고
    • Genetic knock-down of HDAC7 does not ameliorate disease pathogenesis in the R6/2 mouse model of Huntington’s disease
    • Benn CL, Butler R, Mariner L, Nixon J, Moffitt H, Mielcarek M, Woodman B, Bates GP. Genetic knock-down of HDAC7 does not ameliorate disease pathogenesis in the R6/2 mouse model of Huntington’s disease. PLoS ONE. 2009;4:e5747.
    • (2009) PLoS ONE. , vol.4 , pp. e5747
    • Benn, C.L.1    Butler, R.2    Mariner, L.3    Nixon, J.4    Moffitt, H.5    Mielcarek, M.6    Woodman, B.7    Bates, G.P.8
  • 240
    • 79958064073 scopus 로고    scopus 로고
    • Hdac6 knock-out increases tubulin acetylation but does not modify disease progression in the R6/2 mouse model of Huntington’s disease
    • Bobrowska A, Paganetti P, Matthias P, Bates GP. Hdac6 knock-out increases tubulin acetylation but does not modify disease progression in the R6/2 mouse model of Huntington’s disease. PLoS ONE. 2011;6:e20696.
    • (2011) PLoS ONE. , vol.6 , pp. e20696
    • Bobrowska, A.1    Paganetti, P.2    Matthias, P.3    Bates, G.P.4
  • 241
    • 84856710176 scopus 로고    scopus 로고
    • Genetic knock-down of HDAC3 does not modify diseaserelated phenotypes in a mouse model of Huntington’s disease
    • Moumné L, Campbell K, Howland D, Ouyang Y, Bates GP. Genetic knock-down of HDAC3 does not modify diseaserelated phenotypes in a mouse model of Huntington’s disease. PLoS ONE. 2012;7:e31080.
    • (2012) PLoS ONE. , vol.7 , pp. e31080
    • Moumné, L.1    Campbell, K.2    Howland, D.3    Ouyang, Y.4    Bates, G.P.5
  • 242
    • 34548814154 scopus 로고    scopus 로고
    • Apilot study of the clinical efficacy and safety of memantine for Huntington’s disease
    • OndoWG, Mejia NI, Hunter CB. Apilot study of the clinical efficacy and safety of memantine for Huntington’s disease. Parkinsonism Relat Disord. 2007;13:453-4.
    • (2007) Parkinsonism Relat Disord. , vol.13 , pp. 453-454
    • Ondo, W.G.1    Mejia, N.I.2    Hunter, C.B.3
  • 243
    • 0030764219 scopus 로고    scopus 로고
    • Riluzole protects from motor deficits and striatal degeneration produced by systemic 3-nitropropionic acid intoxication in rats
    • Guyot MC, Palfi S, Stutzmann JM, Mazière M, Hantraye P, Brouillet E. Riluzole protects from motor deficits and striatal degeneration produced by systemic 3-nitropropionic acid intoxication in rats. NSC. 1997;81:141-9.
    • (1997) NSC. , vol.81 , pp. 141-149
    • Guyot, M.C.1    Palfi, S.2    Stutzmann, J.M.3    Mazière, M.4    Hantraye, P.5    Brouillet, E.6
  • 245
    • 0028803390 scopus 로고
    • Effect of riluzole on quinolinate-induced neuronal damage in rats: Comparison with blockers of glutamatergic neurotransmission
    • Mary V, Wahl F, Stutzmann JM. Effect of riluzole on quinolinate-induced neuronal damage in rats: Comparison with blockers of glutamatergic neurotransmission. Neurosci Lett. 1995;201:92-6.
    • (1995) Neurosci Lett. , vol.201 , pp. 92-96
    • Mary, V.1    Wahl, F.2    Stutzmann, J.M.3
  • 246
    • 0345600893 scopus 로고    scopus 로고
    • Dosage effects of riluzole in Huntington’s disease: A multicenter placebo-controlled study
    • Huntington Study, Group. Dosage effects of riluzole in Huntington’s disease: A multicenter placebo-controlled study. Neurology. 2003;61:1551-6.
    • (2003) Neurology. , vol.61 , pp. 1551-1556
  • 247
    • 84902330133 scopus 로고    scopus 로고
    • Large Genetic Animal Models of Huntington’s Disease
    • Morton AJ, Howland DS. Large Genetic Animal Models of Huntington’s Disease. J Huntingtons Dis. 2013;2(1):3-19.
    • (2013) J Huntingtons Dis. , vol.2 , Issue.1 , pp. 3-19
    • Morton, A.J.1    Howland, D.S.2
  • 254
    • 84864628471 scopus 로고    scopus 로고
    • Induced Pluripotent Stem Cells from Patients with Huntington’s Disease Show CAGRepeat-Expansion-Associated Phenotypes
    • The Hd Ipsc Consortium. Induced Pluripotent Stem Cells from Patients with Huntington’s Disease Show CAGRepeat-Expansion-Associated Phenotypes. Cell Stem Cell. 2012;11(2):264-78.
    • (2012) Cell Stem Cell. , vol.11 , Issue.2 , pp. 264-278
  • 256
    • 79952163901 scopus 로고    scopus 로고
    • Neuronal degeneration in striatal transplants and Huntington’s disease: Potential mechanisms and clinical implications
    • Cicchetti F, Soulet D, Freeman TB. Neuronal degeneration in striatal transplants and Huntington’s disease: Potential mechanisms and clinical implications. Brain. 2011;134:641-52.
    • (2011) Brain. , vol.134 , pp. 641-652
    • Cicchetti, F.1    Soulet, D.2    Freeman, T.B.3
  • 257
    • 0034811810 scopus 로고    scopus 로고
    • Neuroprotection of striatal neurons against kainate excitotoxicity by neurotrophins and GDNF family members
    • In
    • Gratac`os E, Pérez-Navarro E, Tolosa E, Arenas E, Alberch J. Neuroprotection of striatal neurons against kainate excitotoxicity by neurotrophins and GDNF family members. In, 2001;1287-96.
    • (2001) , pp. 1287-1296
    • E.1    Gratac2    Pérez-Navarro, E.4    Tolosa, E.5    Arenas, E.6    Alberch, J.7
  • 259
    • 0028962436 scopus 로고
    • Brain-derived neurotrophic factortransduced fibroblasts: Production of BDNF and effects of grafting to the adult rat brain
    • Lucidi-Phillipi CA, Gage FH, Shults CW, Jones KR, Reichardt LF, Kang UJ. Brain-derived neurotrophic factortransduced fibroblasts: Production of BDNF and effects of grafting to the adult rat brain. JCompNeurol. 1995;354:361-76.
    • (1995) JCompNeurol. , vol.354 , pp. 361-376
    • Lucidi-Phillipi, C.A.1    Gage, F.H.2    Shults, C.W.3    Jones, K.R.4    Reichardt, L.F.5    Kang, U.J.6
  • 260
    • 0033789409 scopus 로고    scopus 로고
    • Brain-derived neurotrophic factor, neurotrophin-3, and neurotrophin-4/5 prevent the death of striatal projection neurons in a rodent model of Huntington’s disease
    • Pérez-Navarro E, Canudas AM, Akerund P, Alberch J, Arenas E. Brain-derived neurotrophic factor, neurotrophin-3, and neurotrophin-4/5 prevent the death of striatal projection neurons in a rodent model of Huntington’s disease. J Neurochem. 2000;75:2190-9.
    • (2000) J Neurochem. , vol.75 , pp. 2190-2199
    • Pérez-Navarro, E.1    Canudas, A.M.2    Akerund, P.3    Alberch, J.4    Arenas, E.5
  • 262
    • 0442276435 scopus 로고    scopus 로고
    • Brain-derived neurotrophic factor (BDNF) gene delivery into the CNS using bone marrow cells as vehicles in mice
    • Makar TK, Trisler D, Eglitis MA, Mouradian MM, Dhib-Jalbut S. Brain-derived neurotrophic factor (BDNF) gene delivery into the CNS using bone marrow cells as vehicles in mice. Neurosci Lett. 2004;356:215-9.
    • (2004) Neurosci Lett. , vol.356 , pp. 215-219
    • Makar, T.K.1    Trisler, D.2    Eglitis, M.A.3    Mouradian, M.M.4    Dhib-Jalbut, S.5
  • 265
    • 78650416363 scopus 로고    scopus 로고
    • Mesenchymal stem cell transplantation and DMEM administration in a 3NP rat model of Huntington’s disease: Morphological and behavioral outcomes
    • Rossignol J, Boyer C, Lévèque X, Fink KD, Thinard R, Blanchard F, Dunbar GL, Lescaudron L. Mesenchymal stem cell transplantation and DMEM administration in a 3NP rat model of Huntington’s disease: Morphological and behavioral outcomes. Behav Brain Res. 2011;217: 369-78.
    • (2011) Behav Brain Res. , vol.217 , pp. 369-378
    • Rossignol, J.1    Boyer, C.2    Lévèque, X.3    Fink, K.D.4    Thinard, R.5    Blanchard, F.6    Dunbar, G.L.7    Lescaudron, L.8
  • 266
    • 79952182383 scopus 로고    scopus 로고
    • Potential therapeutic uses of BDNF in neurological and psychiatric disorders
    • Nagahara AH, Tuszynski MH. Potential therapeutic uses of BDNF in neurological and psychiatric disorders. Nat Rev Drug Discov. 2011;10:209-19.
    • (2011) Nat Rev Drug Discov. , vol.10 , pp. 209-219
    • Nagahara, A.H.1    Tuszynski, M.H.2
  • 267
    • 84878481329 scopus 로고    scopus 로고
    • Small-molecule TrkB receptor agonists improve motor function and extend survival in a mouse model of Huntington’s disease
    • Jiang M, Peng Q, Liu X, Jin J, Hou Z, Zhang J, Mori S, Ross CA, Ye K, Duan W. Small-molecule TrkB receptor agonists improve motor function and extend survival in a mouse model of Huntington’s disease. Hum Mol Genet. 2013;22(12):2462-70.
    • (2013) Hum Mol Genet. , vol.22 , Issue.12 , pp. 2462-2470
    • Jiang, M.1    Peng, Q.2    Liu, X.3    Jin, J.4    Hou, Z.5    Zhang, J.6    Mori, S.7    Ross, C.A.8    Ye, K.9    Duan, W.10
  • 269
    • 77953701829 scopus 로고    scopus 로고
    • Ex vivo delivery of GDNF maintains motor function and prevents neuronal loss in a transgenic mouse model of Huntington’s disease
    • Ebert AD, Barber AE, Heins BM, Svendsen CN. Ex vivo delivery of GDNF maintains motor function and prevents neuronal loss in a transgenic mouse model of Huntington’s disease. Experimental Neurology. 2010;224:155-62.
    • (2010) Experimental Neurology. , vol.224 , pp. 155-162
    • Ebert, A.D.1    Barber, A.E.2    Heins, B.M.3    Svendsen, C.N.4
  • 270
    • 0030601657 scopus 로고    scopus 로고
    • Glial cell line-derived neurotrophic factor protects striatal calbindin-immunoreactive neurons from excitotoxic damage
    • Pérez-Navarro E, Arenas E, Reiriz J, Calvo N, Alberch J. Glial cell line-derived neurotrophic factor protects striatal calbindin-immunoreactive neurons from excitotoxic damage. NSC. 1996;75:345-52.
    • (1996) NSC. , vol.75 , pp. 345-352
    • Pérez-Navarro, E.1    Arenas, E.2    Reiriz, J.3    Calvo, N.4    Alberch, J.5
  • 271
    • 0032921749 scopus 로고    scopus 로고
    • Intrastriatal grafting of a GDNF-producing cell line protects striatonigral neurons from quinolinic acid excitotoxicity in vivo
    • Pérez-Navarro E, Arenas E, Marco S, Alberch J. Intrastriatal grafting of a GDNF-producing cell line protects striatonigral neurons from quinolinic acid excitotoxicity in vivo. Eur J Neurosci. 1999;11:241-9.
    • (1999) Eur J Neurosci. , vol.11 , pp. 241-249
    • Pérez-Navarro, E.1    Arenas, E.2    Marco, S.3    Alberch, J.4
  • 272
    • 0343091556 scopus 로고    scopus 로고
    • Neurturin protects striatal projection neurons but not interneurons in a rat model of Huntington’s disease
    • Pérez-Navarro E, Akerud P, Marco S, Canals JM, Tolosa E, Arenas E, Alberch J. Neurturin protects striatal projection neurons but not interneurons in a rat model of Huntington’s disease. NSC. 2000;98:89-96.
    • (2000) NSC. , vol.98 , pp. 89-96
    • Pérez-Navarro, E.1    Akerud, P.2    Marco, S.3    Canals, J.M.4    Tolosa, E.5    Arenas, E.6    Alberch, J.7
  • 273
    • 0029761786 scopus 로고    scopus 로고
    • Implants of encapsulated human CNTFproducing fibroblasts prevent behavioral deficits and striatal degeneration in a rodent model of Huntington’s disease
    • Emerich DF, Lindner MD, Winn SR, Chen EY, Frydel BR, Kordower JH. Implants of encapsulated human CNTFproducing fibroblasts prevent behavioral deficits and striatal degeneration in a rodent model of Huntington’s disease. J Neurosci. 1996;16:5168-81.
    • (1996) J Neurosci. , vol.16 , pp. 5168-5181
    • Emerich, D.F.1    Lindner, M.D.2    Winn, S.R.3    Chen, E.Y.4    Frydel, B.R.5    Kordower, J.H.6
  • 276
    • 84862140284 scopus 로고    scopus 로고
    • Sustained induction of neuronal addition to the adult rat neostriatum by AAV4-delivered noggin and BDNF
    • Benraiss A, Bruel-Jungerman E, Lu G, Economides AN, Davidson B, Goldman SA. Sustained induction of neuronal addition to the adult rat neostriatum by AAV4-delivered noggin and BDNF. Gene Ther. 2011;19:483-93.
    • (2011) Gene Ther. , vol.19 , pp. 483-493
    • Benraiss, A.1    Bruel-Jungerman, E.2    Lu, G.3    Economides, A.N.4    Davidson, B.5    Goldman, S.A.6
  • 277
    • 0037762555 scopus 로고    scopus 로고
    • Structural and functional neuroprotection in a rat model of Huntington’s disease by viral gene transfer of GDNF
    • McBride JL, During MJ, Wuu J, Chen EY, Leurgans SE, Kordower JH. Structural and functional neuroprotection in a rat model of Huntington’s disease by viral gene transfer of GDNF. Experimental Neurology. 2003;181:213-23.
    • (2003) Experimental Neurology. , vol.181 , pp. 213-223
    • McBride, J.L.1    During, M.J.2    Wuu, J.3    Chen, E.Y.4    Leurgans, S.E.5    Kordower, J.H.6
  • 281
    • 84870885663 scopus 로고    scopus 로고
    • AAV2-neurturin (CERE-120) for Parkinson’s disease
    • Hickey P, Stacy M. AAV2-neurturin (CERE-120) for Parkinson’s disease. Expert Opin Biol Ther. 2013;13:137-45.
    • (2013) Expert Opin Biol Ther. , vol.13 , pp. 137-145
    • Hickey, P.1    Stacy, M.2
  • 283
    • 34147166160 scopus 로고    scopus 로고
    • Neurturin gene therapy improves motor function and prevents death of striatal neurons in a 3-nitropropionic acid rat model of Huntington’s disease
    • Ramaswamy S, McBride JL, Herzog CD, Brandon E, Gasmi M, Bartus RT, Kordower JH. Neurturin gene therapy improves motor function and prevents death of striatal neurons in a 3-nitropropionic acid rat model of Huntington’s disease. Neurobiology of Disease. 2007;26: 375-84.
    • (2007) Neurobiology of Disease. , vol.26 , pp. 375-384
    • Ramaswamy, S.1    McBride, J.L.2    Herzog, C.D.3    Brandon, E.4    Gasmi, M.5    Bartus, R.T.6    Kordower, J.H.7
  • 284
    • 54049111928 scopus 로고    scopus 로고
    • Intrabodies binding the proline-rich domains of mutant huntingtin increase its turnover and reduce neurotoxicity
    • Southwell AL, Khoshnan A, Dunn DE, Bugg CW, Lo DC, Patterson PH. Intrabodies binding the proline-rich domains of mutant huntingtin increase its turnover and reduce neurotoxicity. J Neurosci. 2008;28:9013-20.
    • (2008) J Neurosci. , vol.28 , pp. 9013-9020
    • Southwell, A.L.1    Khoshnan, A.2    Dunn, D.E.3    Bugg, C.W.4    Lo, D.C.5    Patterson, P.H.6
  • 285
    • 70350543879 scopus 로고    scopus 로고
    • Intrabody gene therapy ameliorates motor, cognitive, and neuropathological symptoms in multiple mouse models of Huntington’s disease
    • Southwell AL, Ko J, Patterson PH. Intrabody gene therapy ameliorates motor, cognitive, and neuropathological symptoms in multiple mouse models of Huntington’s disease. J Neurosci. 2009;29:13589-602.
    • (2009) J Neurosci. , vol.29 , pp. 13589-13602
    • Southwell, A.L.1    Ko, J.2    Patterson, P.H.3
  • 286
    • 44649113841 scopus 로고    scopus 로고
    • Suppression of neuropil aggregates and neurological symptoms by an intracellular antibody implicates the cytoplasmic toxicity of mutant huntingtin
    • Wang C-E, Zhou H, McGuire JR, Cerullo V, Lee B, Li S-H, Li X-J. Suppression of neuropil aggregates and neurological symptoms by an intracellular antibody implicates the cytoplasmic toxicity of mutant huntingtin. The Journal of Cell Biology. 2008;181:803-16.
    • (2008) The Journal of Cell Biology. , vol.181 , pp. 803-816
    • Wang, C.-E.1    Zhou, H.2    McGuire, J.R.3    Cerullo, V.4    Lee, B.5    Li, S.-H.6    Li, X.-J.7
  • 288
    • 84993912315 scopus 로고
    • Increased apoptosis and early embryonic lethality in mice nullizygous for the Huntington’s disease gene homologue
    • Zeitlin S, Liu JP, Chapman DL, Papaioannou VE, Efstratiadis A. Increased apoptosis and early embryonic lethality in mice nullizygous for the Huntington’s disease gene homologue. Nat Genet. 1995;11:155-63.
    • (1995) Nat Genet. , vol.11 , pp. 155-163
    • Zeitlin, S.1    Liu, J.P.2    Chapman, D.L.3    Papaioannou, V.E.4    Efstratiadis, A.5
  • 289
    • 84862643627 scopus 로고    scopus 로고
    • "Huntingtin holiday": Progress toward an antisense therapy for Huntington’s disease
    • Lu X-H, Yang XW. "Huntingtin holiday": Progress toward an antisense therapy for Huntington’s disease. Neuron. 2012;74:964-6.
    • (2012) Neuron. , vol.74 , pp. 964-966
    • Lu, X.-H.1    Yang, X.W.2
  • 290
    • 25144464388 scopus 로고    scopus 로고
    • Intrastriatal rAAV-mediated delivery of anti-huntingtin shRNAs induces partial reversal of disease progression in R6/1 Huntington’s disease transgenic mice
    • Rodriguez-Lebron E, Denovan-Wright EM, Nash K, Lewin AS, Mandel RJ. Intrastriatal rAAV-mediated delivery of anti-huntingtin shRNAs induces partial reversal of disease progression in R6/1 Huntington’s disease transgenic mice. Mol Ther. 2005;12:618-33.
    • (2005) Mol Ther. , vol.12 , pp. 618-633
    • Rodriguez-Lebron, E.1    Denovan-Wright, E.M.2    Nash, K.3    Lewin, A.S.4    Mandel, R.J.5
  • 291
    • 82955233704 scopus 로고    scopus 로고
    • Rational Design of Therapeutic siRNAs: Minimizing Off-targeting Potential to Improve the Safety of RNAi Therapy for Huntington’s Disease
    • Boudreau RL, Spengler RM, Davidson BL. Rational Design of Therapeutic siRNAs: Minimizing Off-targeting Potential to Improve the Safety of RNAi Therapy for Huntington’s Disease. Mol Ther. 2011;19(12):2169-77.
    • (2011) Mol Ther. , vol.19 , Issue.12 , pp. 2169-2177
    • Boudreau, R.L.1    Spengler, R.M.2    Davidson, B.L.3
  • 292
    • 82955199935 scopus 로고    scopus 로고
    • Preclinical Safety of RNAi-Mediated HTT Suppression in the Rhesus Macaque as a Potential Therapy for Huntington’s Disease
    • McBride JL, Pitzer MR, Boudreau RL, Dufour B, Hobbs T, Ojeda SR, Davidson BL. Preclinical Safety of RNAi-Mediated HTT Suppression in the Rhesus Macaque as a Potential Therapy for Huntington’s Disease. Mol Ther. 2011;19:2152-62.
    • (2011) Mol Ther. , vol.19 , pp. 2152-2162
    • McBride, J.L.1    Pitzer, M.R.2    Boudreau, R.L.3    Dufour, B.4    Hobbs, T.5    Ojeda, S.R.6    Davidson, B.L.7
  • 295
    • 42549134402 scopus 로고    scopus 로고
    • AAV vector-mediated RNAi of mutant huntingtin expression is neuroprotective in a novel genetic rat model of Huntington’s disease
    • Franich NR, Fitzsimons HL, Fong DM, Klugmann M, During MJ, Young D. AAV vector-mediated RNAi of mutant huntingtin expression is neuroprotective in a novel genetic rat model of Huntington’s disease. Mol Ther. 2008;16:947-56.
    • (2008) Mol Ther. , vol.16 , pp. 947-956
    • Franich, N.R.1    Fitzsimons, H.L.2    Fong, D.M.3    Klugmann, M.4    During, M.J.5    Young, D.6
  • 296
    • 67349100160 scopus 로고    scopus 로고
    • Nonallele-specific silencing of mutant and wild-type huntingtin demonstrates therapeutic efficacy in Huntington’s disease mice
    • Boudreau RL, McBride JL, Martins I, Shen S, XingY, Carter BJ, Davidson BL. Nonallele-specific silencing of mutant and wild-type huntingtin demonstrates therapeutic efficacy in Huntington’s disease mice. Mol Ther. 2009;17:1053-63.
    • (2009) Mol Ther. , vol.17 , pp. 1053-1063
    • Boudreau, R.L.1    McBride, J.L.2    Martins, I.3    Shen, S.4    Xing, Y.5    Carter, B.J.6    Davidson, B.L.7
  • 299
    • 82955237522 scopus 로고    scopus 로고
    • Potent and selective antisense oligonucleotides targeting single-nucleotide polymorphisms in the Huntington disease gene/allele-specific silencing of mutant huntingtin
    • Carroll JB, Warby SC, Southwell AL, Doty CN, Greenlee S, Skotte N, Hung G, Bennett CF, Freier SM, Hayden MR. Potent and selective antisense oligonucleotides targeting single-nucleotide polymorphisms in the Huntington disease gene/allele-specific silencing of mutant huntingtin. Mol Ther. 2011;19:2178-85.
    • (2011) Mol Ther. , vol.19 , pp. 2178-2185
    • Carroll, J.B.1    Warby, S.C.2    Southwell, A.L.3    Doty, C.N.4    Greenlee, S.5    Skotte, N.6    Hung, G.7    Bennett, C.F.8    Freier, S.M.9    Hayden, M.R.10
  • 302
    • 0032555066 scopus 로고    scopus 로고
    • Coenzyme Q10 administration increases brain mitochondrial concentrations and exerts neuroprotective effects
    • Matthews RT, Yang L, Browne S, Baik M, Beal MF. Coenzyme Q10 administration increases brain mitochondrial concentrations and exerts neuroprotective effects. Proc Natl Acad Sci U S A. 1998;95:8892-7.
    • (1998) Proc Natl Acad Sci U S A. , vol.95 , pp. 8892-8897
    • Matthews, R.T.1    Yang, L.2    Browne, S.3    Baik, M.4    Beal, M.F.5
  • 309
    • 82155182012 scopus 로고    scopus 로고
    • SAHA decreases HDAC 2 and 4 levels in vivo and improves molecular phenotypes in the R6/2 mouse model of Huntington’s disease
    • Mielcarek M, Benn CL, Franklin SA, Smith DL, Woodman B, Marks PA, Bates GP. SAHA decreases HDAC 2 and 4 levels in vivo and improves molecular phenotypes in the R6/2 mouse model of Huntington’s disease. PLoS ONE. 2011;6:e27746.
    • (2011) PLoS ONE. , vol.6 , pp. e27746
    • Mielcarek, M.1    Benn, C.L.2    Franklin, S.A.3    Smith, D.L.4    Woodman, B.5    Marks, P.A.6    Bates, G.P.7
  • 310
    • 84860812619 scopus 로고    scopus 로고
    • Dysregulation of dopamine receptor D2 as a sensitive measure for Huntington disease pathology in model mice
    • Crook ZR, Housman DE. Dysregulation of dopamine receptor D2 as a sensitive measure for Huntington disease pathology in model mice. Proc Natl Acad Sci U S A. 2012;109:7487-92.
    • (2012) Proc Natl Acad Sci U S A. , vol.109 , pp. 7487-7492
    • Crook, Z.R.1    Housman, D.E.2


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.