Skip to main content
Neurotherapeutics logoLink to Neurotherapeutics
. 2008 Apr;5(2):260–269. doi: 10.1016/j.nurt.2008.01.005

Advances in gene therapy for movement disorders

Hideki Mochizuki 1,2, Toru Yasuda 1, M Maral Mouradian 3,
PMCID: PMC5084168  PMID: 18394568

Summary

After nearly 20 years of preclinical experimentation with various gene delivery approaches in animal models of Parkinson’s disease (PD), clinical trials are finally underway. The risk/benefit ratio for these procedures is now generally considered acceptable under approved protocols. The current vehicle for gene delivery to the human brain is recombinant adeno-associated viral vector, which is nonpathogenic and non—self-amplifying. Candidate genes tested in PD patients encode 1) glutamic acid decarboxylase, which is injected into the subthalamic nucleus to catalyze biosynthesis of the inhibitory neurotransmitter γ-aminobutyric acid and so essentially mimic deep brain stimulation of this nucleus; 2) aromatic l-amino acid decarboxylase, which converts l-dopa to dopamine; and 3) neurturin, a member of the glial cell line-derived neurotrophic factor family. Unraveling the genetic underpinnings of PD could allow gene therapy to go beyond modulating neurotransmission or providing trophic effects to dopaminergic neurons by delivering a specific missing or defective gene. For example, the parkin gene (PARK2) is linked to recessively inherited PD due to loss of function mutations; it prevents α-synuclein-induced degeneration of nigral dopaminergic neurons in rats and nonhuman primates. On the other hand, for dominantly inherited Huntington’s disease (HD), in which an expanded polyglutamine tract imparts to the protein huntingtin a toxic gain of function, repressing expression of the mutant allele in the striatum using RNA interference technology mitigates pathology and delays the phenotype in a mouse model. Here we review the current state of preclinical and clinical gene therapy studies conducted in PD and HD.

Key Words: Movement disorders, neurodegeneration, Parkinson’s disease, Huntington’s disease, adeno-associated viral vectors, gene therapy, parkin protein, α-synuclein

References

  • 1.Mouradian MM, Chase TN. Gene therapy for Parkinson’s disease: current knowledge and future perspective. Gene Ther. 1997;4:504–506. doi: 10.1038/sj.gt.3300449. [DOI] [PubMed] [Google Scholar]
  • 2.Linden RM, Ward P, Giraud C, Winocour E, Berns KI. Site-specific integration by adeno-associated virus. Proc Natl Acad Sci U S A. 1996;93:11288–11294. doi: 10.1073/pnas.93.21.11288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Gao G, Vandenberghe LH, Wilson JM. New recombinant sero-types of AAV vectors. Curr Gene Ther. 2005;5:285–297. doi: 10.2174/1566523054065057. [DOI] [PubMed] [Google Scholar]
  • 4.Freed WJ, Geller HM, Poltorak M, et al. Genetically altered and defined cell lines for transplantation in animal models of Parkinson’s disease. Prog Brain Res. 1990;82:11–21. doi: 10.1016/s0079-6123(08)62585-6. [DOI] [PubMed] [Google Scholar]
  • 5.Wolff JA, Fisher LJ, Xu L, et al. Grafting fibroblasts genetically modified to produce l-dopa in a rat model of Parkinson disease. Proc Natl Acad Sci U S A. 1989;86:9011–9014. doi: 10.1073/pnas.86.22.9011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Sajadi A, Bensadoun JC, Schneider BL, Lo Bianco C, Aebischer P. Transient striatal delivery of GDNF via encapsulated cells leads to sustained behavioral improvement in a bilateral model of Parkinson disease. Neurobiol Dis. 2006;22:119–129. doi: 10.1016/j.nbd.2005.10.006. [DOI] [PubMed] [Google Scholar]
  • 7.Kishima H, Poyot T, Bloch J, et al. Encapsulated GDNF-producing C2C12 cells for Parkinson’s disease: a pre-clinical study in chronic MPTP-treated baboons. Neurobiol Dis. 2004;16:428–439. doi: 10.1016/j.nbd.2004.03.012. [DOI] [PubMed] [Google Scholar]
  • 8.Prasad KN, Clarkson ED, La Rosa FG, Edwards-Prasad J, Freed CR. Efficacy of grafted immortalized dopamine neurons in an animal model of parkinsonism: a review. Mol Genet Metab. 1998;65:1–9. doi: 10.1006/mgme.1998.2726. [DOI] [PubMed] [Google Scholar]
  • 9.Anton R, Kordower JH, Maidment NT, et al. Neural-targeted gene therapy for rodent and primate hemiparkinsonism. Exp Neurol. 1994;127:207–218. doi: 10.1006/exnr.1994.1097. [DOI] [PubMed] [Google Scholar]
  • 10.Rohrer DC, Nilaver G, Nipper V, Machida CA. Genetically modified PC12 brain grafts: Survivability and inducible nerve growth factor expression. Cell Transplant. 1996;5:57–68. doi: 10.1177/096368979600500111. [DOI] [PubMed] [Google Scholar]
  • 11.Kawaja MD, Gage FH. Morphological and neurochemical features of cultured primary skin fibroblasts of Fischer 344 rats following striatal implantation. J Comp Neurol. 1992;317:102–116. doi: 10.1002/cne.903170108. [DOI] [PubMed] [Google Scholar]
  • 12.Lee WY, Lee EA, Jeon MY, Kang HY, Park YG. Vesicular monoamine transporter-2 and aromatic l-amino acid decarboxylase gene therapy prevents development of motor complications in parkinsonian rats after chronic intermittent l-3,4-dihydroxyphenylalanine administration. Exp Neurol. 2006;197:215–224. doi: 10.1016/j.expneurol.2005.09.012. [DOI] [PubMed] [Google Scholar]
  • 13.Bencsics C, Wachtel SR, Milstien S, Hatakeyama K, Becker JB, Kang UJ. Double transduction with GTP cyclohydrolase I and tyrosine hydroxylase is necessary for spontaneous synthesis of l-DOPA by primary fibroblasts. J Neurosci. 1996;16:4449–4456. doi: 10.1523/JNEUROSCI.16-14-04449.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Ridet JL, Corti O, Pencalet P, et al. Toward autologous ex vivo gene therapy for the central nervous system with human adult astrocytes. Hum Gene Ther. 1999;10:271–280. doi: 10.1089/10430349950019057. [DOI] [PubMed] [Google Scholar]
  • 15.Yoshimoto Y, Lin Q, Collier TJ, Frim DM, Breakefield XO, Bohn MC. Astrocytes retrovirally transduced with BDNF elicit behavioral improvement in a rat model of Parkinson’s disease. Brain Res. 1995;691:25–36. doi: 10.1016/0006-8993(95)00596-i. [DOI] [PubMed] [Google Scholar]
  • 16.Ljungberg MC, Stern G, Wilkin GP. Survival of genetically engineered, adult-derived rat astrocytes grafted into the 6-hydroxydopamine lesioned adult rat striatum. Brain Res. 1999;816:29–37. doi: 10.1016/s0006-8993(98)01061-0. [DOI] [PubMed] [Google Scholar]
  • 17.Lundberg C, Horellou P, Mallet J, Bjorklund A. Generation of DOPA-producing astrocytes by retroviral transduction of the human tyrosine hydroxylase gene: in vivo characterization and in vivo effects in the rat Parkinson model. Exp Neurol. 1996;139:39–53. doi: 10.1006/exnr.1996.0079. [DOI] [PubMed] [Google Scholar]
  • 18.Tornatore C, Baker-Cairns B, Yadid G, et al. Expression of tyrosine hydroxylase in an immortalized human fetal astrocyte cell line: in vitro characterization and engraftment into the rodent striatum. Cell Transplant. 1996;5:145–163. doi: 10.1177/096368979600500206. [DOI] [PubMed] [Google Scholar]
  • 19.Ericson C, Georgievska B, Lundberg C. Ex vivo gene delivery of GDNF using primary astrocytes transduced with a lentiviral vector provides neuroprotection in a rat model of Parkinson’s disease. Eur J Neurosci. 2005;22:2755–2764. doi: 10.1111/j.1460-9568.2005.04503.x. [DOI] [PubMed] [Google Scholar]
  • 20.Brazelton TR, Rossi FM, Keshet GI, Blau HM. From marrow to brain: expression of neuronal phenotypes in adult mice. Science. 2000;290:1775–1779. doi: 10.1126/science.290.5497.1775. [DOI] [PubMed] [Google Scholar]
  • 21.Nakano K, Migita M, Mochizuki H, Shimada T. Differentiation of transplanted bone marrow cells in the adult mouse brain. Transplantation. 2001;71:1735–1740. doi: 10.1097/00007890-200106270-00006. [DOI] [PubMed] [Google Scholar]
  • 22.Azizi SA, Stokes D, Augelli BJ, DiGirolamo C, Prockop DJ. Engraftment and migration of human bone marrow stromal cells implanted in the brains of albino rats: similarities to astrocyte grafts. Proc Natl Acad Sci U S A. 1998;95:3908–3913. doi: 10.1073/pnas.95.7.3908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Munoz JR, Stoutenger BR, Robinson AP, Spees JL, Prockop DJ. Human stem/progenitor cells from bone marrow promote neurogenesis of endogenous neural stem cells in the hippocampus of mice. Proc Natl Acad Sci U S A. 2005;102:18171–18176. doi: 10.1073/pnas.0508945102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Dezawa M, Kanno H, Hoshino M, et al. Specific induction of neuronal cells from bone marrow stromal cells and application for autologous transplantation. J Clin Invest. 2004;113:1701–1710. doi: 10.1172/JCI20935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Eglitis MA, Mezey E. Hematopoietic cells differentiate into both microglia and macroglia in the brains of adult mice. Proc Natl Acad Sci U S A. 1997;94:4080–4085. doi: 10.1073/pnas.94.8.4080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Mezey E, Chandross KJ, Harta G, Maki RA, McKercher SR. Turning blood into brain: cells bearing neuronal antigens generated in vivo from bone marrow. Science. 2000;290:1779–1782. doi: 10.1126/science.290.5497.1779. [DOI] [PubMed] [Google Scholar]
  • 27.Eglitis MA, Dawson D, Park KW, Mouradian MM. Targeting of marrow-derived astrocytes to the ischemic brain. Neuroreport. 1999;10:1289–1292. doi: 10.1097/00001756-199904260-00025. [DOI] [PubMed] [Google Scholar]
  • 28.Park KW, Eglitis MA, Mouradian MM. Protection of nigral neurons by GDNF-engineered marrow cell transplantation. Neurosci Res. 2001;40:315–323. doi: 10.1016/s0168-0102(01)00242-5. [DOI] [PubMed] [Google Scholar]
  • 29.Mouradian MM, Chase TN. Gene therapy of Parkinson’s disease: An approach to the prevention or palliation of levodopa-associated motor complications. Exp Neurol. 1997;144:51–57. doi: 10.1006/exnr.1996.6388. [DOI] [PubMed] [Google Scholar]
  • 30.Espejo EF, Montoro RJ, Armengol JA, Lopez-Barneo J. Cellular and functional recovery of parkinsonian rats after intrastriatal transplantation of carotid body cell aggregates. Neuron. 1998;20:197–206. doi: 10.1016/s0896-6273(00)80449-3. [DOI] [PubMed] [Google Scholar]
  • 31.Toledo-Aral JJ, Méndez-Ferrer S, Pardal R, Echevama M, López-Barneo J. Trophic restoration of the nigrostriatal dopaminergic pathway in long-term carotid body-grafted parkinsonian rats. J Neurosci. 2003;23:141–148. doi: 10.1523/JNEUROSCI.23-01-00141.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Arjona V, Míguez-Castellanos A, Montoro RJ, et al. Autotransplantation of human carotid body cell aggregates for treatment of Parkinson’s disease. Neurosurgery. 2003;53:321–328. doi: 10.1227/01.neu.0000073315.88827.72. [DOI] [PubMed] [Google Scholar]
  • 33.Mínguez-Castellanos A, Escamilla-Sevilla F, Hotton GR, et al. Carotid body autotransplantation in Parkinson disease: a clinical and positron emission tomography study. J Neurol Neurosurg Psychiatry. 2007;78:825–831. doi: 10.1136/jnnp.2006.106021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.During MJ, Naegele JR, O’Malley KL, Geller AI. Long-term behavioral recovery in parkinsonian rats by an HSV vector expressing tyrosine hydroxylase. Science. 1994;266:1399–1403. doi: 10.1126/science.266.5189.1399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Kaplitt MG, Leone P, Samulski RJ, et al. Long-term gene expression and phenotypic collection using adeno-associated virus vectors in the mammalian brain. Nat Genet. 1994;8:148–154. doi: 10.1038/ng1094-148. [DOI] [PubMed] [Google Scholar]
  • 36.Rivière C, Danos O, Douar AM. Long-term expression and repeated administration of AAV type 1, 2 and 5 vectors in skeletal muscle of immunocompetent adult mice. Gene Ther. 2006;13:1300–1308. doi: 10.1038/sj.gt.3302766. [DOI] [PubMed] [Google Scholar]
  • 37.Horellou P, Mallet J. Gene therapy for Parkinson’s disease. Mol Neurobiol. 1997;15:241–256. doi: 10.1007/BF02740636. [DOI] [PubMed] [Google Scholar]
  • 38.Uchida K, Tsuzaki N, Nagatsu T, Kohsaka S. Tetrahydrobiopterin-dependent functional recovery in 6-hydroxydopamine-treated rats by intracerebral grafting of fibroblasts transfected with tyrosine hydroxylase cDNA. Dev Neurosci. 1992;14:173–180. doi: 10.1159/000111661. [DOI] [PubMed] [Google Scholar]
  • 39.Mandel RJ, Rendahl KG, Spratt SK, Snyder RO, Cohen LK, Leff SE. Characterization of intrastriatal recombinant adeno-associated virus-mediated gene transfer of human tyrosine hydroxylase and human GTP-cyclohydrolase I in a rat model of Parkinson’s disease. J Neurosci. 1998;18:4271–4284. doi: 10.1523/JNEUROSCI.18-11-04271.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Sánchez-Pernaute R, Harvey-White J, Cunningham J, Bankiewicz KS. Functional effect of adeno-associated virus mediated gene transfer of aromatic l-amino acid decarboxylase into the striatum of 6-OHDA-lesioned rats. Mol Ther. 2001;4:324–330. doi: 10.1006/mthe.2001.0466. [DOI] [PubMed] [Google Scholar]
  • 41.Bankiewicz KS, Forsayeth J, Eberling JL, et al. Long-term clinical improvement in MPTP-lesioned primates after gene therapy with AAV-hAADC. Mol Ther. 2006;14:564–570. doi: 10.1016/j.ymthe.2006.05.005. [DOI] [PubMed] [Google Scholar]
  • 42.Fan DS, Ogawa M, Fujimoto KI, et al. Behavioral recovery in 6-hydroxydopamine-lesioned rats by cotransduction of striatum with tyrosine hydroxylase and aromatic l-amino acid decarboxylase genes using two separate adeno-associated virus vectors. Hum Gene Ther. 1998;9:2527–2535. doi: 10.1089/hum.1998.9.17-2527. [DOI] [PubMed] [Google Scholar]
  • 43.During MJ, Samulski RJ, Elsworth JD, et al. In vivo expression of therapeutic human genes for dopamine production in the caudates of MPTP-treated monkeys using an AAV vector. Gene Ther. 1998;5:820–827. doi: 10.1038/sj.gt.3300650. [DOI] [PubMed] [Google Scholar]
  • 44.Wachtel SR, Bencsics C, Kang UJ. Role of aromatic l-amino acid decarboxylase for dopamine replacement by genetically modified fibroblasts in a rat model of Parkinson’s disease. J Neurochem. 1997;9:2055–2063. doi: 10.1046/j.1471-4159.1997.69052055.x. [DOI] [PubMed] [Google Scholar]
  • 45.Moffat M, Harmon S, Haycock J, O’Malley KL. l-Dopa and dopamine-producing gene cassettes for gene therapy approaches to Parkinson’s disease. Exp Neurol. 1997;144:69–73. doi: 10.1006/exnr.1996.6390. [DOI] [PubMed] [Google Scholar]
  • 46.During MJ, Kaplitt MG, Stern MB, Eidelberg D. Subthalamic GAD gene transfer in Parkinson disease patients who are candidates for deep brain stimulation. Hum Gene Ther. 2001;12:1589–1591. [PubMed] [Google Scholar]
  • 47.Luo J, Kaplitt MG, Fitzsimons HL, et al. Subthalamic GAD gene therapy in a Parkinson’s disease rat model. Science. 2002;298:425–429. doi: 10.1126/science.1074549. [DOI] [PubMed] [Google Scholar]
  • 48.Emborg ME, Carbon M, Holden JE, et al. Subthalamic glutamic acid decarboxylase gene therapy: changes in motor function and cortical metabolism. J Cereb Blood Flow Metab. 2007;27:501–509. doi: 10.1038/sj.jcbfm.9600364. [DOI] [PubMed] [Google Scholar]
  • 49.Olson L. The coming of age of the GDNF family and its receptors: gene delivery in a rat Parkinson model may have clinical implications. Trends Neurol Sci. 1997;20:277–279. doi: 10.1016/s0166-2236(97)01098-9. [DOI] [PubMed] [Google Scholar]
  • 50.Bohn MC. A commentary on glial cell line-derived neurotrophic factor (GDNF): from a glial secreted molecule to gene therapy. Biochem Pharmacol. 1999;57:135–142. doi: 10.1016/s0006-2952(98)00280-9. [DOI] [PubMed] [Google Scholar]
  • 51.Choi-Lundberg DL, Lin Q, Chang YN, et al. Dopaminergic neurons protected from degeneration by GDNF gene therapy. Science. 1997;75:838–841. doi: 10.1126/science.275.5301.838. [DOI] [PubMed] [Google Scholar]
  • 52.Bilang-Bleuel A, Revah F, Colin P, et al. Intrastriatal injection of an adenoviral vector expressing glial-cell-line-derived neurotrophic factor prevents dopaminergic neuron degeneration and behavioral impairment in a rat model of Parkinson’s disease. Proc Natl Acad Sci U S A. 1997;94:8818–8823. doi: 10.1073/pnas.94.16.8818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Choi-Lundberg DL, Lin Q, Schallert T, et al. Behavioral and cellular protection of rat dopaminergic neurons by an adenoviral vector encoding glial cell line-derived neurotrophic factor. Exp Neurol. 1998;154:261–275. doi: 10.1006/exnr.1998.6887. [DOI] [PubMed] [Google Scholar]
  • 54.Lapchak PA, Araujo DM, Hilt DC, Sheng J, Jiao S. Adenoviral vector-mediated GDNF gene therapy in a rodent lesion model of late stage Parkinson’s disease. Brain Res. 1997;777:153–160. doi: 10.1016/s0006-8993(97)01100-1. [DOI] [PubMed] [Google Scholar]
  • 55.Kojima H, Abiru Y, Sakajiri K, et al. Adenovirus-mediated transduction with human glial cell line-derived neurotrophic factor gene prevents 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced dopamine depletion in striatum of mouse brain. Biochem Biophys Res Commun. 1997;238:569–573. doi: 10.1006/bbrc.1997.7183. [DOI] [PubMed] [Google Scholar]
  • 56.Mandel RJ, Spratt SK, Snyder RO, Leff SE. Midbrain injection of recombinant adeno-associated virus encoding rat glial cell line-derived neurotrophic factor protects nigral neurons in a progressive 6-hydroxydopamine-induced degeneration model of Parkinson’s disease in rats. Proc Natl Acad Sci U S A. 1997;94:14083–14088. doi: 10.1073/pnas.94.25.14083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Kotzbauer PT, Lampe PA, Heuckeroth RO, et al. Neurturin, a relative of glial-cell-line-derived neurotrophic factor. Nature. 1996;384:467–470. doi: 10.1038/384467a0. [DOI] [PubMed] [Google Scholar]
  • 58.Horger BA, Nishimura MC, Armanini MP, et al. Neurturin exerts potent actions on survival and function of midbrain dopaminergic neurons. J Neurosci. 1998;18:4929–4937. doi: 10.1523/JNEUROSCI.18-13-04929.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Rosenblad C, Kirik D, Devaux B, Moffat B, Phillips HS, Björklund A. Protection and regeneration of nigral dopaminergic neurons by neurturin or GDNF in a partial lesion model of Parkinson’s disease after administration into the striatum or the lateral ventricle. Eur J Neurosci. 1999;11:1554–66. doi: 10.1046/j.1460-9568.1999.00566.x. [DOI] [PubMed] [Google Scholar]
  • 60.Kordower JH, Herzog CD, Dass B, et al. Delivery of neurturin by AAV2 (CERE-120)-mediated gene transfer provides structural and functional neuroprotection and neurorestoration in MPTP-treated monkeys. Ann Neurol. 2006;60:706–715. doi: 10.1002/ana.21032. [DOI] [PubMed] [Google Scholar]
  • 61.Herzog CD, Dass B, Holden JE, et al. Striatal delivery of CERE-120, an AAV2 vector encoding human neurturin, enhances activity of the dopaminergic nigrostriatal system in aged monkeys. Mov Disord. 2007;22:1124–1132. doi: 10.1002/mds.21503. [DOI] [PubMed] [Google Scholar]
  • 62.Gasmi M, Brandon EP, Herzog CD, et al. AAV2-mediated delivery of human neurturin to the rat nigrostriatal system: long-term efficacy and tolerability of CERE-120 for Parkinson’s disease. Neurobiol Dis. 2007;27:67–76. doi: 10.1016/j.nbd.2007.04.003. [DOI] [PubMed] [Google Scholar]
  • 63.Mochizuki H, Hayakawa H, Migita M, et al. An AAV-derived Apaf-1 dominant negative inhibitor prevents MPTP toxicity as antiapoptotic gene therapy for Parkinson’s disease. Proc Natl Acad Sci U S A. 2001;98:10918–10923. doi: 10.1073/pnas.191107398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Eberhardt O, Coelln RV, Kugler S, et al. Protection by synergistic effects of adenovirus-mediated X-chromosome-linked inhibitor of apoptosis and glial cell line—derived neurotrophic factor gene transfer in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine model of Parkinson’s disease. J Neurosci. 2000;20:9126–9134. doi: 10.1523/JNEUROSCI.20-24-09126.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Dedmon MM, Christodoulou J, Wilson MR, Dobson CM. Heat shock protein 70 inhibits α-synuclein fibril formation via preferential binding to prefibrillar species. J Biol Chem. 2005;280:14733–14740. doi: 10.1074/jbc.M413024200. [DOI] [PubMed] [Google Scholar]
  • 66.Junn E, Ronchetti RD, Quezado MM, Kim SY, Mouradian MM. Tissue transglutaminase-induced aggregation of α-synuclein: implications for Lewy body formation in Parkinson’s disease and dementia with Lewy bodies. Proc Natl Acad Sci U S A. 2003;100:2047–2052. doi: 10.1073/pnas.0438021100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Hayashita-Kinoh H, Yamada M, Yokota T, Mizuno Y, Mochizuki H. Down-regulation of α-synuclein expression can rescue dopaminergic cells from cell death in the substantia nigra of Parkinson’s disease rat model. Biochem Biophys Res Commun. 2006;341:1088–1095. doi: 10.1016/j.bbrc.2006.01.057. [DOI] [PubMed] [Google Scholar]
  • 68.Masliah E, Rockenstein E, Adame A, et al. Effects of α-synuclein immunization in a mouse model of Parkinson’s disease. Neuron. 2005;46:857–868. doi: 10.1016/j.neuron.2005.05.010. [DOI] [PubMed] [Google Scholar]
  • 69.Ridet JL, Bensadoun JC, Déglon N, Aebischer P, Zum AD. Lentivirus-mediated expression of glutathione peroxidase: neuroprotection in murine models of Parkinson’s disease. Neurobiol Dis. 2006;21:29–34. doi: 10.1016/j.nbd.2005.06.003. [DOI] [PubMed] [Google Scholar]
  • 70.Farrer MJ. Genetics of Parkinson disease: paradigm shifts and future prospects. Nat Rev Genet. 2006;7:306–318. doi: 10.1038/nrg1831. [DOI] [PubMed] [Google Scholar]
  • 71.Kim RH, Smith PD, Aleyasin H, et al. Hypersensitivity of DJ-1-deficient mice to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyrindine (MPTP) and oxidative stress. Proc Natl Acad Sci U S A. 2005;102:5215–5220. doi: 10.1073/pnas.0501282102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Yamada M, Iwatsubo T, Mizuno Y, Mochizuki H. Overexpression of α-synuclein in rat substantia nigra results in loss of dopaminergic neurons, phosphorylation of α-synuclein and activation of caspase-9: resemblance to pathogenetic changes in Parkinson’s disease. J Neurochem. 2004;91:451–461. doi: 10.1111/j.1471-4159.2004.02728.x. [DOI] [PubMed] [Google Scholar]
  • 73.Kitada T, Asakawa S, Hattori N, et al. Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism. Nature. 1998;392:605–608. doi: 10.1038/33416. [DOI] [PubMed] [Google Scholar]
  • 74.Shimura H, Hattori N, Kubo S, et al. Familial Parkinson disease gene product, parkin, is a ubiquitin-protein ligase. Nat Genet. 2000;25:302–305. doi: 10.1038/77060. [DOI] [PubMed] [Google Scholar]
  • 75.Chung KK, Thomas B, Li X, et al. S-nitrosylation of parkin regulates ubiquitination and compromises parkin’s protective function. Science. 2004;304:1328–1331. doi: 10.1126/science.1093891. [DOI] [PubMed] [Google Scholar]
  • 76.Yao D, Gu Z, Nakamura T, et al. Nitrosative stress linked to sporadic Parkinson’s disease: S-nitrosylation of parkin regulates its E3 ubiquitin ligase activity. Proc Natl Acad Sci U S A. 2004;101:10810–10814. doi: 10.1073/pnas.0404161101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.LaVoie MJ, Ostaszewski BL, Weihofen A, Schlossmacher MG, Selkoe DJ. Dopamine covalently modifies and functionally inactivates parkin. Nat Med. 2005;11:1214–1221. doi: 10.1038/nm1314. [DOI] [PubMed] [Google Scholar]
  • 78.Li W, Lesuisse C, Xu Y, Troncoso JC, Price DL, Lee MK. Stabilization of α-synuclein protein with aging and familial Parkinson’s disease-linked A53T mutation. J Neurosci. 2004;24:7400–7409. doi: 10.1523/JNEUROSCI.1370-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Chu Y, Kordower JH. Age-associated increases of α-synuclein in monkeys and humans are associated with nigrostriatal dopamine depletion: Is this the target for Parkinson’s disease? Neurobiol Dis. 2007;25:134–149. doi: 10.1016/j.nbd.2006.08.021. [DOI] [PubMed] [Google Scholar]
  • 80.Yamada M, Mizuno Y, Mochizuki H. Parkin gene therapy for α-synucleinopathy: a rat model of Parkinson’s disease. Hum Gene Ther. 2005;16:262–270. doi: 10.1089/hum.2005.16.262. [DOI] [PubMed] [Google Scholar]
  • 81.Lo Bianco C, Schneider BL, Bauer M, et al. Lentiviral vector delivery of parkin prevents dopaminergic degeneration in an α-synuclein rat model of Parkinson’s disease. Proc Natl Acad Sci U S A. 2004;101:17510–17515. doi: 10.1073/pnas.0405313101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Yasuda T, Miyachi S, Kitagawa R, et al. Neuronal specificity of α-synuclein toxicity and effect of parkin co-expression in primates. Neuroscience. 2007;144:743–753. doi: 10.1016/j.neuroscience.2006.09.052. [DOI] [PubMed] [Google Scholar]
  • 83.Park J, Lee SB, Lee S, et al. Mitochondrial dysfunction in Drosophila PINK1 mutants is complemented by parkin. Nature. 2006;441:1157–1161. doi: 10.1038/nature04788. [DOI] [PubMed] [Google Scholar]
  • 84.Clark IE, Dodson MW, Jiang C, et al. Drosophila PINK1 is required for mitochondrial function and interacts genetically with parkin. Nature. 2006;441:1162–1166. doi: 10.1038/nature04779. [DOI] [PubMed] [Google Scholar]
  • 85.Yang Y, Gehrke S, Imai Y, et al. Mitochondrial pathology and muscle and dopaminergic neuron degeneration caused by inactivation of Drosophila Pink1 is rescued by parkin. Proc Natl Acad Sci U S A. 2006;103:10793–10798. doi: 10.1073/pnas.0602493103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Vercammen L, Van der Perren A, Vaudano E, et al. Parkin protects against neurotoxicity in the 6-hydroxydopamine rat model for Parkinson’s disease. Mol Ther. 2006;14:716–723. doi: 10.1016/j.ymthe.2006.06.009. [DOI] [PubMed] [Google Scholar]
  • 87.Paterna JC, Leng A, Weber E, Feldon J, Bueler H. DJ-1 and parkin modulate dopamine-dependent behavior and inhibit MPTP-induced nigral dopamine neuron loss in mice. Mol Ther. 2007;15:698–704. doi: 10.1038/sj.mt.6300067. [DOI] [PubMed] [Google Scholar]
  • 88.Kaplitt MG, Feigin A, Tang C, et al. Safety and tolerability of gene therapy with an adeno-associated virus (AAV) borne GAD gene for Parkinson’s disease: an open label, phase I trial. Lancet. 2007;369:2097–2105. doi: 10.1016/S0140-6736(07)60982-9. [DOI] [PubMed] [Google Scholar]
  • 89.Mandel RJ, Burger C. Clinical trials in neurological disorders using AAV vectors: promises and challenges. Curr Opin Mol Ther. 2004;6:482–490. [PubMed] [Google Scholar]
  • 90.Fjord-Larsen L, Johansen JL, Kusk P, et al. Efficient in vivo protection of nigral dopaminergic neurons by lentiviral gene transfer of a modified neurturin construct. Exp Neurol. 2005;195:49–60. doi: 10.1016/j.expneurol.2005.03.006. [DOI] [PubMed] [Google Scholar]
  • 91.Check E. Second chance. Nat Med. 2007;13:770–771. doi: 10.1038/nm0707-770. [DOI] [PubMed] [Google Scholar]
  • 92.Yamamoto A, Lucas JJ, Hen R. Reversal of neuropathology and motor dysfunction in a conditional model of Huntington’s disease. Cell. 2000;101:57–66. doi: 10.1016/S0092-8674(00)80623-6. [DOI] [PubMed] [Google Scholar]
  • 93.Wang YL, Liu W, Wada E, Murata M, Wada K, Kanazawa I. Clinico-pathological rescue of a model mouse of Huntington’s disease by siRNA. Neurosci Res. 2005;53:241–249. doi: 10.1016/j.neures.2005.06.021. [DOI] [PubMed] [Google Scholar]
  • 94.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;12:618–633. doi: 10.1016/j.ymthe.2005.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Zuccato C, Ciammola A, Rigamonti D, et al. Loss of huntingtin-mediated BDNF gene transcription in Huntington’s disease. Science. 2001;293:493–498. doi: 10.1126/science.1059581. [DOI] [PubMed] [Google Scholar]
  • 96.Kells AP, Fong DM, Dragunow M, During MJ, Young D, Connor B. AAV-mediated gene delivery of BDNF or GDNF is neuroprotective in a model of Huntington disease. Mol Ther. 2004;9:682–688. doi: 10.1016/j.ymthe.2004.02.016. [DOI] [PubMed] [Google Scholar]
  • 97.Pérez-Navarro E, Akerud P, Marco S, et al. Neurturin protects striatal projection neurons but not intemeurons in a rat model of Huntington’s disease. Neuroscience. 2000;98:89–96. doi: 10.1016/s0306-4522(00)00074-9. [DOI] [PubMed] [Google Scholar]
  • 98.Ramaswamy S, McBride JL, Herzog CD, et al. Neurturin gene therapy improves motor function and prevents death of striatal neurons in a 3-nitropropionic acid rat model of Huntington’s disease. Neurobiol Dis. 2007;26:375–384. doi: 10.1016/j.nbd.2007.01.003. [DOI] [PubMed] [Google Scholar]
  • 99.Curtis MA, Penney EB, Pearson AG, et al. Increased cell proliferation and neurogenesis in the adult human Huntington’s disease brain. Proc Natl Acad Sci U S A. 2003;100:9023–9027. doi: 10.1073/pnas.1532244100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Cho SR, 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;117:2889–2902. doi: 10.1172/JCI31778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Bloch J, Bachoud-Levi AC, Deglon N, et al. Neuroprotective gene therapy for Huntington’s disease, using polymer-encapsulated cells engineered to secrete human ciliary neurotrophic factor: results of a phase I study. Hum Gene Ther. 2004;15:968–975. doi: 10.1089/hum.2004.15.968. [DOI] [PubMed] [Google Scholar]
  • 102.Lang AE, Gill S, Patel NK, et al. Randomized controlled trial of intraputamenal glial cell line-derived neurotrophic factor infusion in Parkinson disease. Ann Neurol. 2006;59:459–466. doi: 10.1002/ana.20737. [DOI] [PubMed] [Google Scholar]
  • 103.Gill SS, Patel NK, Hotton GR, et al. Direct brain infusion of glial cell line-derived neurotrophic factor in Parkinson disease. Nat Med. 2003;9:589–595. doi: 10.1038/nm850. [DOI] [PubMed] [Google Scholar]
  • 104.de la Fuente-Femández R, Ruth TJ, Sossi V, Schulzer M, Calne DB, Stoessl AJ. Expectation and dopamine release: mechanism of the placebo effect in Parkinson’s disease. Science. 2001;293:1164–1166. doi: 10.1126/science.1060937. [DOI] [PubMed] [Google Scholar]
  • 105.Benedetti F, Colloca L, Torre E, et al. Placebo-responsive Parkinson patients show decreased activity in single neurons of subthalamic nucleus. Nat Neurosci. 2004;7:587–588. doi: 10.1038/nn1250. [DOI] [PubMed] [Google Scholar]
  • 106.Frank S, Kieburtz K, Holloway R, Kim SY. What is the risk of sham surgery in Parkinson disease clinical trials? A review of published reports. Neurology. 2005;65:1101–1103. doi: 10.1212/01.wnl.0000171957.90640.b5. [DOI] [PubMed] [Google Scholar]
  • 107.Kim SY, Frank S, Holloway R, Zimmerman C, Wilson R, Kieburtz K. Science and ethics of sham surgery: a survey of Parkinson disease clinical researchers. Arch Neurol. 2005;62:1357–1360. doi: 10.1001/archneur.62.9.1357. [DOI] [PubMed] [Google Scholar]

Articles from Neurotherapeutics are provided here courtesy of Elsevier

RESOURCES