Skip to main content
Top
Published in: Current Neurology and Neuroscience Reports 3/2012

Open Access 01-06-2012 | Genetics (V Bonifati, Section Editor)

Modeling Parkinson’s Disease Using Induced Pluripotent Stem Cells

Authors: Blake Byers, Hsiao-lu Lee, Renee Reijo Pera

Published in: Current Neurology and Neuroscience Reports | Issue 3/2012

Login to get access

Abstract

Our understanding of the underlying molecular mechanism of Parkinson’s disease (PD) is hampered by a lack of access to affected human dopaminergic (DA) neurons on which to base experimental research. Fortunately, the recent development of a PD disease model using induced pluripotent stem cells (iPSCs) provides access to cell types that were previously unobtainable in sufficient quantity or quality, and presents exciting promises for the elucidation of PD etiology and the development of potential therapeutics. To more effectively model PD, we generated two patient-derived iPSC lines: a line carrying a homozygous p.G2019S mutation in the leucine-rich repeat kinase 2 (LRRK2) gene and another carrying a full gene triplication of the α-synuclein encoding gene, SNCA. We demonstrated that these PD-linked pluripotent lines were able to differentiate into DA neurons and that these neurons exhibited increased expression of key oxidative stress response genes and α-synuclein protein. Moreover, when compared to wild-type DA neurons, LRRK2-G2019S iPSC-derived DA neurons were more sensitive to caspase-3 activation caused by exposure to hydrogen peroxide, MG-132, and 6-hydroxydopamine. In addition, SNCA-triplication iPSC-derived DA neurons formed early ubiquitin-positive puncta and were more sensitive to peak toxicity from hydrogen peroxide-induced stress. These aforementioned findings suggest that LRRK2-G2019S and SNCA-triplication iPSC-derived DA neurons exhibit early phenotypes linked to PD. Given the high penetrance of the homozygous LRRK2 mutation, the expression of wild-type α-synuclein protein in the SNCA-triplication line, and the clinical resemblance of patients afflicted with these familial disorders to sporadic PD patients, these iPSC-derived neurons may be unique and valuable models for disease diagnostics and development of novel pharmacological agents for alleviation of relevant disease phenotypes.
Literature
1.
go back to reference Hughes AJ, Daniel SE, Kilford L, Lees AJ. Accuracy of clinical diagnosis of idiopathic Parkinson’s disease: a clinico-pathological study of 100 cases. J Neurol Neurosurg Psychiatry. 1992;55:181–4.PubMedCrossRef Hughes AJ, Daniel SE, Kilford L, Lees AJ. Accuracy of clinical diagnosis of idiopathic Parkinson’s disease: a clinico-pathological study of 100 cases. J Neurol Neurosurg Psychiatry. 1992;55:181–4.PubMedCrossRef
2.
go back to reference Wakabayashi K, Takahashi H. Neuropathology of autonomic nervous system in Parkinson’s disease. Eur Neurol. 1997;38 Suppl 2:2–7.PubMedCrossRef Wakabayashi K, Takahashi H. Neuropathology of autonomic nervous system in Parkinson’s disease. Eur Neurol. 1997;38 Suppl 2:2–7.PubMedCrossRef
3.
go back to reference Langston JW. The Parkinson’s complex: parkinsonism is just the tip of the iceberg. Ann Neurol. 2006;59:591–6.PubMedCrossRef Langston JW. The Parkinson’s complex: parkinsonism is just the tip of the iceberg. Ann Neurol. 2006;59:591–6.PubMedCrossRef
4.
go back to reference Lewy F. in Pathologische Anatomie. In: Handbuch der Neurologie. (ed M Lewandowsky) Ch. 920–933, 13 Springer-Verlag; 1912. Lewy F. in Pathologische Anatomie. In: Handbuch der Neurologie. (ed M Lewandowsky) Ch. 920–933, 13 Springer-Verlag; 1912.
5.
go back to reference Lewy F. Zur pathologischen Anatomie der Paralysis Agitans. Deutsche Zeitschrift fur Nervenheilkunde. 1913;50:50–5. Lewy F. Zur pathologischen Anatomie der Paralysis Agitans. Deutsche Zeitschrift fur Nervenheilkunde. 1913;50:50–5.
8.
go back to reference Dawson TM, Dawson VL. Molecular pathways of neurodegeneration in Parkinson’s disease. Science. 2003;302:819–22.PubMedCrossRef Dawson TM, Dawson VL. Molecular pathways of neurodegeneration in Parkinson’s disease. Science. 2003;302:819–22.PubMedCrossRef
9.
go back to reference Ross CA, Poirier MA. Protein aggregation and neurodegenerative disease. Nat Med. 2004;10(Suppl):S10–7.PubMedCrossRef Ross CA, Poirier MA. Protein aggregation and neurodegenerative disease. Nat Med. 2004;10(Suppl):S10–7.PubMedCrossRef
10.
go back to reference Kidd M. Paired helical filaments in electron microscopy of Alzheimer’s disease. Nature. 1963;197:192–3.PubMedCrossRef Kidd M. Paired helical filaments in electron microscopy of Alzheimer’s disease. Nature. 1963;197:192–3.PubMedCrossRef
11.
go back to reference Duffy PE, Tennyson VM. Phase and electron microscopic observations of Lewy bodies and melanin granules in the substantia nigra and locus caeruleus in Parkinson’s disease. J Neuropathol Exp Neurol. 1965;24:398.CrossRef Duffy PE, Tennyson VM. Phase and electron microscopic observations of Lewy bodies and melanin granules in the substantia nigra and locus caeruleus in Parkinson’s disease. J Neuropathol Exp Neurol. 1965;24:398.CrossRef
12.
go back to reference Nuytemans K, Theuns J, Cruts M, Van Broeckhoven C. Genetic etiology of Parkinson disease associated with mutations in the SNCA, PARK2, PINK1, PARK7, and LRRK2 genes: a mutation update. Hum Mutat. 2010;31:763–80.PubMedCrossRef Nuytemans K, Theuns J, Cruts M, Van Broeckhoven C. Genetic etiology of Parkinson disease associated with mutations in the SNCA, PARK2, PINK1, PARK7, and LRRK2 genes: a mutation update. Hum Mutat. 2010;31:763–80.PubMedCrossRef
13.
go back to reference Lesage S, Brice A. Parkinson's disease: from monogenic forms to genetic susceptibility factors. Hum Mol Genet. 2009;18(R1):R48–59.PubMedCrossRef Lesage S, Brice A. Parkinson's disease: from monogenic forms to genetic susceptibility factors. Hum Mol Genet. 2009;18(R1):R48–59.PubMedCrossRef
14.
go back to reference Dodson MW, Guo M. Pink1, Parkin, DJ-1 and mitochondrial dysfunction in Parkinson’s disease. Curr Opin Neurobiol. 2007;17:331–7.PubMedCrossRef Dodson MW, Guo M. Pink1, Parkin, DJ-1 and mitochondrial dysfunction in Parkinson’s disease. Curr Opin Neurobiol. 2007;17:331–7.PubMedCrossRef
15.
go back to reference Chartier-Harlin MC, et al. Alpha-synuclein locus duplication as a cause of familial Parkinson’s disease. Lancet. 2004;364:1167–9.PubMedCrossRef Chartier-Harlin MC, et al. Alpha-synuclein locus duplication as a cause of familial Parkinson’s disease. Lancet. 2004;364:1167–9.PubMedCrossRef
16.
go back to reference Polymeropoulos MH, et al. Mutation in the alpha-synuclein gene identified in families with Parkinson’s disease. Science. 1997;276:2045–7.PubMedCrossRef Polymeropoulos MH, et al. Mutation in the alpha-synuclein gene identified in families with Parkinson’s disease. Science. 1997;276:2045–7.PubMedCrossRef
17.
go back to reference Singleton AB, et al. alpha-Synuclein locus triplication causes Parkinson’s disease. Science. 2003;302:841.PubMedCrossRef Singleton AB, et al. alpha-Synuclein locus triplication causes Parkinson’s disease. Science. 2003;302:841.PubMedCrossRef
18.
go back to reference Polymeropoulos MH, et al. Mapping of a gene for Parkinson’s disease to chromosome 4q21-q23. Science. 1996;274(5290):1197–9.PubMedCrossRef Polymeropoulos MH, et al. Mapping of a gene for Parkinson’s disease to chromosome 4q21-q23. Science. 1996;274(5290):1197–9.PubMedCrossRef
19.
go back to reference Uversky VN. A protein-chameleon: conformational plasticity of alpha-synuclein, a disordered protein involved in neurodegenerative disorders. J Biomol Struct Dyn. 2003;21:211.PubMed Uversky VN. A protein-chameleon: conformational plasticity of alpha-synuclein, a disordered protein involved in neurodegenerative disorders. J Biomol Struct Dyn. 2003;21:211.PubMed
20.
go back to reference •• Bartels T, Choi JG, Selkoe DJ. α-Synuclein occurs physiologically as a helically folded tetramer that resists aggregation. Nature 2011, 477(7362):107–110. Using endogenous α-synuclein isolated and analyzed under nondenaturing conditions from neuronal and non-neuronal cell lines, brain tissue, and living human cells, Bartels et al. report that α-synuclein occurs natively as a folded tetramer of about 58 kDa. PubMedCrossRef •• Bartels T, Choi JG, Selkoe DJ. α-Synuclein occurs physiologically as a helically folded tetramer that resists aggregation. Nature 2011, 477(7362):107–110. Using endogenous α-synuclein isolated and analyzed under nondenaturing conditions from neuronal and non-neuronal cell lines, brain tissue, and living human cells, Bartels et al. report that α-synuclein occurs natively as a folded tetramer of about 58 kDa. PubMedCrossRef
21.
go back to reference Jenco JM, Rawlingson A, Daniels B, Morris A. J. Regulation of phospholipase D2: selective inhibition of mammalian phospholipase D isoenzymes by α- and β-synucleins. Biochemistry. 1998;37:4901–9.PubMedCrossRef Jenco JM, Rawlingson A, Daniels B, Morris A. J. Regulation of phospholipase D2: selective inhibition of mammalian phospholipase D isoenzymes by α- and β-synucleins. Biochemistry. 1998;37:4901–9.PubMedCrossRef
22.
go back to reference Davidson WS, Jonas A, Clayton DF, George JM. Stabilization of α-synuclein secondary structure upon binding to synthetic membranes. J Biol Chem. 1998;273:9443.PubMedCrossRef Davidson WS, Jonas A, Clayton DF, George JM. Stabilization of α-synuclein secondary structure upon binding to synthetic membranes. J Biol Chem. 1998;273:9443.PubMedCrossRef
23.
go back to reference McLean PJ, Kawamata H, Ribich S, Hyman BT. Membrane association and protein conformation of α-synuclein in intact neurons. J Biol Chem. 2000;275:8812.PubMedCrossRef McLean PJ, Kawamata H, Ribich S, Hyman BT. Membrane association and protein conformation of α-synuclein in intact neurons. J Biol Chem. 2000;275:8812.PubMedCrossRef
24.
go back to reference Nemani VM, et al. Increased expression of α-synuclein reduces neurotransmitter release by inhibiting synaptic vesicle reclustering after endocytosis. Neuron. 2010;65:66–79.PubMedCrossRef Nemani VM, et al. Increased expression of α-synuclein reduces neurotransmitter release by inhibiting synaptic vesicle reclustering after endocytosis. Neuron. 2010;65:66–79.PubMedCrossRef
25.
go back to reference Chandra S, Gallardo G, Fernández-Chacón R, Schlüter OM, Südhof TC. α-Synuclein Cooperates with CSPα in Preventing Neurodegeneration. Cell. 2005;123:383–96.PubMedCrossRef Chandra S, Gallardo G, Fernández-Chacón R, Schlüter OM, Südhof TC. α-Synuclein Cooperates with CSPα in Preventing Neurodegeneration. Cell. 2005;123:383–96.PubMedCrossRef
26.
go back to reference Burré J, Sharma M, Tsetsenis T, Buchman V, Etherton MR, Südhof TC. alpha-Synuclein promotes SNARE-complex assembly in vivo and in vitro. Science. 2010;329:1663–7.PubMedCrossRef Burré J, Sharma M, Tsetsenis T, Buchman V, Etherton MR, Südhof TC. alpha-Synuclein promotes SNARE-complex assembly in vivo and in vitro. Science. 2010;329:1663–7.PubMedCrossRef
27.
go back to reference Drolet RE, Behrouz B, Lookingland KJ, Goudreau JL. Mice lacking α-synuclein have an attenuated loss of striatal dopamine following prolonged chronic MPTP Administration. Neurotoxicology. 2004;25:761–9.PubMedCrossRef Drolet RE, Behrouz B, Lookingland KJ, Goudreau JL. Mice lacking α-synuclein have an attenuated loss of striatal dopamine following prolonged chronic MPTP Administration. Neurotoxicology. 2004;25:761–9.PubMedCrossRef
28.
go back to reference Klivenyi P, et al. Mice lacking alpha-synuclein are resistant to mitochondrial toxins. Neurobiol Dis. 2006;21:541–8.PubMedCrossRef Klivenyi P, et al. Mice lacking alpha-synuclein are resistant to mitochondrial toxins. Neurobiol Dis. 2006;21:541–8.PubMedCrossRef
29.
go back to reference Greten-Harrison B, et al. αβγ-Synuclein triple knockout mice reveal age-dependent neuronal dysfunction. Proc Natl Acad Sci USA. 2010;107(45):19573–8.PubMedCrossRef Greten-Harrison B, et al. αβγ-Synuclein triple knockout mice reveal age-dependent neuronal dysfunction. Proc Natl Acad Sci USA. 2010;107(45):19573–8.PubMedCrossRef
30.
go back to reference MacLeod D, et al. The familial Parkinsonism gene LRRK2 regulates neurite process morphology. Neuron. 2006;52:587–93.PubMedCrossRef MacLeod D, et al. The familial Parkinsonism gene LRRK2 regulates neurite process morphology. Neuron. 2006;52:587–93.PubMedCrossRef
31.
go back to reference Smith WW, et al. Kinase activity of mutant LRRK2 mediates neuronal toxicity. Nat Neurosci. 2006;9:1231–3.PubMedCrossRef Smith WW, et al. Kinase activity of mutant LRRK2 mediates neuronal toxicity. Nat Neurosci. 2006;9:1231–3.PubMedCrossRef
32.
go back to reference West AB, et al. Parkinson’s disease-associated mutations in leucine-rich repeat kinase 2 augment kinase activity. Proc Natl Acad Sci USA. 2005;102:16842–7.PubMedCrossRef West AB, et al. Parkinson’s disease-associated mutations in leucine-rich repeat kinase 2 augment kinase activity. Proc Natl Acad Sci USA. 2005;102:16842–7.PubMedCrossRef
33.
go back to reference Heo HY, et al. LRRK2 enhances oxidative stress-induced neurotoxicity via its kinase activity. Exp Cell Res. 2010;316:649–56.PubMedCrossRef Heo HY, et al. LRRK2 enhances oxidative stress-induced neurotoxicity via its kinase activity. Exp Cell Res. 2010;316:649–56.PubMedCrossRef
34.
go back to reference Milosevic J, et al. Emerging role of LRRK2 in human neural progenitor cell cycle progression, survival and differentiation. Mol neurodegener. 2009;4:25.PubMedCrossRef Milosevic J, et al. Emerging role of LRRK2 in human neural progenitor cell cycle progression, survival and differentiation. Mol neurodegener. 2009;4:25.PubMedCrossRef
35.
go back to reference Lin X, et al. Leucine-rich repeat kinase 2 regulates the progression of neuropathology induced by Parkinson’s-disease-related mutant alpha-synuclein. Neuron. 2009;64:807–27.PubMedCrossRef Lin X, et al. Leucine-rich repeat kinase 2 regulates the progression of neuropathology induced by Parkinson’s-disease-related mutant alpha-synuclein. Neuron. 2009;64:807–27.PubMedCrossRef
36.
go back to reference Betarbet R, Sherer TB, Greenamyre JT. Animal models of Parkinson's disease. Bioessays. 2002;24:308–18.PubMedCrossRef Betarbet R, Sherer TB, Greenamyre JT. Animal models of Parkinson's disease. Bioessays. 2002;24:308–18.PubMedCrossRef
37.
go back to reference Ng CH, et al. Parkin protects against LRRK2 G2019S mutant-induced dopaminergic neurodegeneration in Drosophila. J Neurosci. 2009;29:11257–62.PubMedCrossRef Ng CH, et al. Parkin protects against LRRK2 G2019S mutant-induced dopaminergic neurodegeneration in Drosophila. J Neurosci. 2009;29:11257–62.PubMedCrossRef
38.
go back to reference Paisan-Ruiz C, et al. Cloning of the gene containing mutations that cause PARK8-linked Parkinson’s disease. Neuron. 2004;44:595–600.PubMedCrossRef Paisan-Ruiz C, et al. Cloning of the gene containing mutations that cause PARK8-linked Parkinson’s disease. Neuron. 2004;44:595–600.PubMedCrossRef
39.
go back to reference Schüle B, Reijo Pera RA, Langston JW. Can cellular models revolutionize drug discovery in Parkinson's disease? Biochim Biophys Acta. 2009;1792:1043–51.PubMed Schüle B, Reijo Pera RA, Langston JW. Can cellular models revolutionize drug discovery in Parkinson's disease? Biochim Biophys Acta. 2009;1792:1043–51.PubMed
40.
go back to reference Wakabayashi K, Takahashi H, Ohama E, Ikuta F. Parkinson’s disease: an immunohistochemical study of Lewy body-containing neurons in the enteric nervous system. Acta Neuropathol. 1990;79:581–3.PubMedCrossRef Wakabayashi K, Takahashi H, Ohama E, Ikuta F. Parkinson’s disease: an immunohistochemical study of Lewy body-containing neurons in the enteric nervous system. Acta Neuropathol. 1990;79:581–3.PubMedCrossRef
41.
go back to reference Zimprich A, et al. Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology. Neuron. 2004;44:601–7.PubMedCrossRef Zimprich A, et al. Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology. Neuron. 2004;44:601–7.PubMedCrossRef
42.
go back to reference Haass C, Selkoe DJ. Soluble protein oligomers in neurodegeneration: lessons from the Alzheimer’s amyloid beta-peptide. Nat Rev Mol Cell Biol. 2007;8:101–12.PubMedCrossRef Haass C, Selkoe DJ. Soluble protein oligomers in neurodegeneration: lessons from the Alzheimer’s amyloid beta-peptide. Nat Rev Mol Cell Biol. 2007;8:101–12.PubMedCrossRef
43.
go back to reference Masliah E, et al. Dopaminergic loss and inclusion body formation in α-synuclein mice: implications for neurodegenerative disorders. Science. 2000;287:1265.PubMedCrossRef Masliah E, et al. Dopaminergic loss and inclusion body formation in α-synuclein mice: implications for neurodegenerative disorders. Science. 2000;287:1265.PubMedCrossRef
44.
go back to reference van der Putten H, et al. Neuropathology in mice expressing human α-synuclein. J Neurosci. 2000;20:6021–9.PubMed van der Putten H, et al. Neuropathology in mice expressing human α-synuclein. J Neurosci. 2000;20:6021–9.PubMed
45.
go back to reference Kahle PJ, et al. Subcellular localization of wild-type and Parkinson’s disease-associated mutant α-synuclein in human and transgenic mouse brain. J Neurosci. 2000;20:6365–73.PubMed Kahle PJ, et al. Subcellular localization of wild-type and Parkinson’s disease-associated mutant α-synuclein in human and transgenic mouse brain. J Neurosci. 2000;20:6365–73.PubMed
46.
go back to reference Li Y, Liu W, Oo TF, Wang L, Tang Y, Jackson-Lewis V, et al. Mutant LRRK2(R1441G) BAC transgenic mice recapitulate cardinal features of Parkinson’s disease. Nat Neurosci. 2009;12:826–8.PubMedCrossRef Li Y, Liu W, Oo TF, Wang L, Tang Y, Jackson-Lewis V, et al. Mutant LRRK2(R1441G) BAC transgenic mice recapitulate cardinal features of Parkinson’s disease. Nat Neurosci. 2009;12:826–8.PubMedCrossRef
47.
go back to reference Marti MJ, Tolosa E, Campdelacreu J. Clinical overview of the synucleinopathies. Mov Disord. 2003;18 Suppl 6:S21–7.PubMedCrossRef Marti MJ, Tolosa E, Campdelacreu J. Clinical overview of the synucleinopathies. Mov Disord. 2003;18 Suppl 6:S21–7.PubMedCrossRef
48.
go back to reference Kirik D, et al. Nigrostriatal alpha-synucleinopathy induced by viral vector-mediated overexpression of human alpha-synuclein: a new primate model of Parkinson’s disease. Proc Natl Acad Sci USA. 2003;100:2884–9.PubMedCrossRef Kirik D, et al. Nigrostriatal alpha-synucleinopathy induced by viral vector-mediated overexpression of human alpha-synuclein: a new primate model of Parkinson’s disease. Proc Natl Acad Sci USA. 2003;100:2884–9.PubMedCrossRef
49.
go back to reference Maries E, Dass B, Collier TJ, Kordower JH, Steece-Collier K. The role of alpha-synuclein in Parkinson’s disease: insights from animal models. Nat Rev Neurosci. 2003;4:727–38.PubMedCrossRef Maries E, Dass B, Collier TJ, Kordower JH, Steece-Collier K. The role of alpha-synuclein in Parkinson’s disease: insights from animal models. Nat Rev Neurosci. 2003;4:727–38.PubMedCrossRef
50.
go back to reference Takahashi K, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131:861–72.PubMedCrossRef Takahashi K, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131:861–72.PubMedCrossRef
51.
go back to reference Yu J, et al. Induced pluripotent stem cell lines derived from human somatic cells. Science. 2007;318:1917–20.PubMedCrossRef Yu J, et al. Induced pluripotent stem cell lines derived from human somatic cells. Science. 2007;318:1917–20.PubMedCrossRef
52.
go back to reference Paisan-Ruiz C, et al. LRRK2 gene in Parkinson disease: mutation analysis and case control association study. Neurology. 2005;65:696–700.PubMedCrossRef Paisan-Ruiz C, et al. LRRK2 gene in Parkinson disease: mutation analysis and case control association study. Neurology. 2005;65:696–700.PubMedCrossRef
53.
go back to reference Hernandez D, et al. The dardarin G 2019 S mutation is a common cause of Parkinson’s disease but not other neurodegenerative diseases. Neurosci lett. 2005;389:137–9.PubMedCrossRef Hernandez D, et al. The dardarin G 2019 S mutation is a common cause of Parkinson’s disease but not other neurodegenerative diseases. Neurosci lett. 2005;389:137–9.PubMedCrossRef
54.
go back to reference Nichols WC, et al. Genetic screening for a single common LRRK2 mutation in familial Parkinson’s disease. Lancet. 2005;365:410–2.PubMed Nichols WC, et al. Genetic screening for a single common LRRK2 mutation in familial Parkinson’s disease. Lancet. 2005;365:410–2.PubMed
55.
go back to reference Kachergus J, et al. Identification of a novel LRRK2 mutation linked to autosomal dominant parkinsonism: evidence of a common founder across European populations. Am J Hum Genet. 2005;76:672–80.PubMedCrossRef Kachergus J, et al. Identification of a novel LRRK2 mutation linked to autosomal dominant parkinsonism: evidence of a common founder across European populations. Am J Hum Genet. 2005;76:672–80.PubMedCrossRef
56.
go back to reference Aarsland D, Marsh L, Schrag A. Neuropsychiatric symptoms in Parkinson’s disease. Mov Disord. 2009;24:2175–86.PubMedCrossRef Aarsland D, Marsh L, Schrag A. Neuropsychiatric symptoms in Parkinson’s disease. Mov Disord. 2009;24:2175–86.PubMedCrossRef
57.
go back to reference Ibanez P, et al. Causal relation between alpha-synuclein gene duplication and familial Parkinson’s disease. Lancet. 2004;364:1169–71.PubMedCrossRef Ibanez P, et al. Causal relation between alpha-synuclein gene duplication and familial Parkinson’s disease. Lancet. 2004;364:1169–71.PubMedCrossRef
58.
go back to reference Waters CH, Miller CA. Autosomal dominant Lewy body parkinsonism in a four-generation family. Ann Neurol. 1994;35:59–64.PubMedCrossRef Waters CH, Miller CA. Autosomal dominant Lewy body parkinsonism in a four-generation family. Ann Neurol. 1994;35:59–64.PubMedCrossRef
59.
60.
go back to reference Soldner F, et al. Parkinson’s disease patient-derived induced pluripotent stem cells free of viral reprogramming factors. Cell. 2009;136:964–77.PubMedCrossRef Soldner F, et al. Parkinson’s disease patient-derived induced pluripotent stem cells free of viral reprogramming factors. Cell. 2009;136:964–77.PubMedCrossRef
61.
go back to reference •• Israel MA, et al. Probing sporadic and familial Alzheimer’s disease using induced pluripotent stem cells. Nature 2012, 482:216–220. In this recent study, scientists have created stem cell–derived, in vitro models of sporadic and hereditary AD, using iPSCs from patients with the neurodegenerative disorder. Their finding suggests that sporadic AD patients may have genomes that generate strong neuronal phenotypes. Therefore, future iPSC studies have the potential to provide great insight into the underlying mechanisms behind the observed heterogeneity in sporadic neurodegenerative disease pathogenesis. PubMed •• Israel MA, et al. Probing sporadic and familial Alzheimer’s disease using induced pluripotent stem cells. Nature 2012, 482:216–220. In this recent study, scientists have created stem cell–derived, in vitro models of sporadic and hereditary AD, using iPSCs from patients with the neurodegenerative disorder. Their finding suggests that sporadic AD patients may have genomes that generate strong neuronal phenotypes. Therefore, future iPSC studies have the potential to provide great insight into the underlying mechanisms behind the observed heterogeneity in sporadic neurodegenerative disease pathogenesis. PubMed
62.
go back to reference • Byers B, et al. SNCA triplication Parkinson’s patient’s iPSC-derived DA neurons accumulate α-synuclein and are susceptible to oxidative stress. PLoS One 2011, 6:e26159. Scientistis succeeded in the generation of iPSC-derived mDA neurons from a patient with a triplication in the α-synuclein gene (SNCA). These iPSCs readily differentiated into functional neurons and exhibited cardinal disease-related phenotypes in culture. These phenotypes include the accumulation of α-synuclein, inherent overexpression of markers of oxidative stress, and sensitivity to peroxide-induced oxidative stress. These findings show that the SNCA-triplication mutation can affect cellular function in culture and corroborate a cell autonomous etiology for PD that is independent of complex entirety of the diseased brain. PubMedCrossRef • Byers B, et al. SNCA triplication Parkinson’s patient’s iPSC-derived DA neurons accumulate α-synuclein and are susceptible to oxidative stress. PLoS One 2011, 6:e26159. Scientistis succeeded in the generation of iPSC-derived mDA neurons from a patient with a triplication in the α-synuclein gene (SNCA). These iPSCs readily differentiated into functional neurons and exhibited cardinal disease-related phenotypes in culture. These phenotypes include the accumulation of α-synuclein, inherent overexpression of markers of oxidative stress, and sensitivity to peroxide-induced oxidative stress. These findings show that the SNCA-triplication mutation can affect cellular function in culture and corroborate a cell autonomous etiology for PD that is independent of complex entirety of the diseased brain. PubMedCrossRef
63.
go back to reference • Nguyen HN, et al. LRRK2 mutant iPSC-derived DA neurons demonstrate increased susceptibility to oxidative stress. Cell Stem Cell 2011, 8:267–280. Scientists successfully derived DA neurons from iPSCs carrying a G2019S point mutation in LRKK2. These neurons exhibited increased expression of key oxidative stress-response genes and α-synuclein protein; they are also more sensitive to caspase-3 activation and cell death caused by exposure to stress agents, such as hydrogen peroxide, MG-132, and 6-hydroxydopamine, than wild-type DA neurons. This observed heightened stress sensitivity is consistent with existing understanding of early PD phenotypes and presents a potential therapeutic target. PubMedCrossRef • Nguyen HN, et al. LRRK2 mutant iPSC-derived DA neurons demonstrate increased susceptibility to oxidative stress. Cell Stem Cell 2011, 8:267–280. Scientists successfully derived DA neurons from iPSCs carrying a G2019S point mutation in LRKK2. These neurons exhibited increased expression of key oxidative stress-response genes and α-synuclein protein; they are also more sensitive to caspase-3 activation and cell death caused by exposure to stress agents, such as hydrogen peroxide, MG-132, and 6-hydroxydopamine, than wild-type DA neurons. This observed heightened stress sensitivity is consistent with existing understanding of early PD phenotypes and presents a potential therapeutic target. PubMedCrossRef
64.
go back to reference Dimos JT, et al. Induced pluripotent stem cells generated from patients with ALS can be differentiated into motor neurons. Science. 2008;321:1218–21.PubMedCrossRef Dimos JT, et al. Induced pluripotent stem cells generated from patients with ALS can be differentiated into motor neurons. Science. 2008;321:1218–21.PubMedCrossRef
65.
go back to reference Ebert AD, et al. Induced pluripotent stem cells from a spinal muscular atrophy patient. Nature. 2009;457:277–80.PubMedCrossRef Ebert AD, et al. Induced pluripotent stem cells from a spinal muscular atrophy patient. Nature. 2009;457:277–80.PubMedCrossRef
Metadata
Title
Modeling Parkinson’s Disease Using Induced Pluripotent Stem Cells
Authors
Blake Byers
Hsiao-lu Lee
Renee Reijo Pera
Publication date
01-06-2012
Publisher
Current Science Inc.
Published in
Current Neurology and Neuroscience Reports / Issue 3/2012
Print ISSN: 1528-4042
Electronic ISSN: 1534-6293
DOI
https://doi.org/10.1007/s11910-012-0270-y

Other articles of this Issue 3/2012

Current Neurology and Neuroscience Reports 3/2012 Go to the issue

Genetics (V Bonifati, Section Editor)

Schizophrenia Genetics: Putting All the Pieces Together

Genetics (V Bonifati, Section Editor)

Recent Advances in the Genetics of the ALS-FTLD Complex

Neuro-Oncology (LE Abrey, Section Editor)

Chemotherapy-Related Cognitive Dysfunction