Skip to main content

Advertisement

Log in

α-Synucleinopathy models and human neuropathology: similarities and differences

  • Review
  • Published:
Acta Neuropathologica Aims and scope Submit manuscript

Abstract

Point mutations and genomic multiplications in the α-synuclein (αSYN) gene cause autosomal-dominant Parkinson’s disease. Moreover, αSYN fibrils are the major component of Lewy bodies, the neuropathological hallmarks of Parkinson’s disease and dementia with Lewy bodies as well as of glial cytoplasmic inclusions in multiple system atrophy. These diseases are collectively referred to as α-synucleinopathies. Cellular mechanisms regulating αSYN fibril formation and toxicity are intensely studied in vitro, and in cell culture and diverse animal models. Specific neuropathology was achieved in transgenic mouse models using several promoters to express human wild-type and mutant αSYN in brain regions affected by the various α-synucleinopathies. Somatodendritic accumulation of the transgenic αSYN with neuritic distortions was a common finding. The nigrostriatal dopaminergic projections were surprisingly resistant to α-synucleinopathy in transgenic mice, although they tended to be more vulnerable to neurotoxins. In a few mouse models, αSYN aggregated in an age-dependent manner into genuine fibrillar amyloid. Brain region selective αSYN neuropathology correlated with specific behavioral impairments, such as locomotor dysfunction and cognitive decline. Thus, the αSYN fibrillization process is tightly linked to neuropathology. The role and thus therapeutic potential of post-translational modifications (ubiquitinylation, oxidation, phosphorylation, truncation) and modifier genes on αSYN neuropathology can now be assessed in valid transgenic mouse models of α-synucleinopathies.

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

Access this article

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

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

References

  1. Abeliovich A, Schmitz Y, Fariñas I, Choi-Lundberg D, Ho W-H, Castillo PE, Shinsky N, Garcia Verdugo JM, Armanini M, Ryan A, Hynes M, Phillips H, Sulzer D, Rosenthal A (2000) Mice lacking α-synuclein display functional deficits in the nigrostriatal dopamine system. Neuron 25:239–252

    Article  PubMed  CAS  Google Scholar 

  2. Anderson JP, Walker DE, Goldstein JM, de Laat R, Banducci K, Caccavello RJ, Barbour R, Huang J, Kling K, Lee M, Diep L, Keim PS, Shen X, Chataway T, Schlossmacher MG, Seubert P, Schenk D, Sinha S, Gai WP, Chilcote TJ (2006) Phosphorylation of Ser-129 is the dominant pathological modification of α-synuclein in familial and sporadic Lewy body disease. J Biol Chem 281:29739–29752

    Article  PubMed  CAS  Google Scholar 

  3. Arima K, Uéda K, Sunohara N, Arakawa K, Hirai S, Nakamura M, Tonozuka-Uehara H, Kawai M (1998) NACP/α-synuclein immunoreactivity in fibrillary components of neuronal and oligodendroglial cytoplasmic inclusions in the pontine nuclei in multiple system atrophy. Acta Neuropathol 96:439–444

    Article  PubMed  CAS  Google Scholar 

  4. Baba M, Nakajo S, Tu P-H, Tomita T, Nakaya K, Lee VM-Y, Trojanowski JQ, Iwatsubo T (1998) Aggregation of α-synuclein in Lewy bodies of sporadic Parkinson’s disease and dementia with Lewy bodies. Am J Pathol 152:879–884

    PubMed  CAS  Google Scholar 

  5. Chandra S, Gallardo G, Fernández-Chacón R, Schlüter OM, Südhof TC (2005) α-Synuclein cooperates with CSPα in preventing neurodegeneration. Cell 123:383–396

    Article  PubMed  CAS  Google Scholar 

  6. Chartier-Harlin M-C, Kachergus J, Roumier C, Mouroux V, Douay X, Lincoln S, Levecque C, Larvor L, Andrieux J, Hulihan M, Waucquier N, Defebvre L, Amouyel P, Farrer M, Destée A (2004) α-Synuclein locus duplication as a cause of familial Parkinson’s disease. Lancet 364:1167–1169

    Article  PubMed  CAS  Google Scholar 

  7. Cooper AA, Gitler AD, Cashikar A, Haynes CM, Hill KJ, Bhullar B, Liu K, Xu K, Strathearn KE, Liu F, Cao S, Caldwell KA, Caldwell GA, Marsischky G, Kolodner RD, LaBaer J, Rochet J-C, Bonini NM, Lindquist S (2006) α-Synuclein blocks ER-Golgi traffic and Rab1 rescues neuron loss in Parkinson’s models. Science 313:324–328

    Article  PubMed  CAS  Google Scholar 

  8. Dalfó E, Gómez-Isla T, Rosa JL, Nieto Bodelón M, Cuadrado Tejedor M, Barrachina M, Ambrosio S, Ferrer I (2004) Abnormal α-synuclein interactions with Rab proteins in α-synuclein A30P transgenic mice. J Neuropathol Exp Neurol 63:302–313

    PubMed  Google Scholar 

  9. Di Rosa G, Puzzo D, Sant’Angelo A, Trinchese F, Arancio O (2003) α-Synuclein: between synaptic function and dysfunction. Histol Histopathol 18:1257–1266

    PubMed  CAS  Google Scholar 

  10. Dufty BM, Warner LR, Hou ST, Jiang SX, Gomez-Isla T, Leenhouts KM, Oxford JT, Feany MB, Masliah E, Rohn TT (2007) Calpain-cleavage of α-synuclein: connecting proteolytic processing to disease-linked aggregation. Am J Pathol 170:1725–1738

    Article  PubMed  CAS  Google Scholar 

  11. Fleming SM, Salcedo J, Fernagut P-O, Rockenstein E, Masliah E, Levine MS, Chesselet M-F (2004) Early and progressive sensorimotor anomalies in mice overexpressing wild-type human α-synuclein. J Neurosci 24:9434–9440

    Article  PubMed  CAS  Google Scholar 

  12. Fleming SM, Salcedo J, Hutson CB, Rockenstein E, Masliah E, Levine MS, Chesselet M-F (2006) Behavioral effects of dopaminergic agonists in transgenic mice overexpressing human wildtype α-synuclein. Neuroscience 142:1245–1253

    Article  PubMed  CAS  Google Scholar 

  13. Freichel C, Neumann M, Ballard T, Müller V, Woolley M, Ozmen L, Borroni E, Kretzschmar HA, Haass C, Spooren W, Kahle PJ (2007) Age-dependent cognitive decline and amygdala pathology in α-synuclein transgenic mice. Neurobiol Aging 28:1421–1435

    Article  PubMed  CAS  Google Scholar 

  14. Fujiwara H, Hasegawa M, Dohmae N, Kawashima A, Masliah E, Goldberg MS, Shen J, Takio K, Iwatsubo T (2002) α-Synuclein is phosphorylated in synucleinopathy lesions. Nat Cell Biol 4:160–164

    Article  PubMed  CAS  Google Scholar 

  15. George JM (2002) The synucleins. Genome Biol 3:REVIEWS3002

  16. Giasson BI, Duda JE, Quinn SM, Zhang B, Trojanowski JQ, Lee VM-Y (2002) Neuronal α-synucleinopathy with severe movement disorder in mice expressing A53T human α-synuclein. Neuron 34:521–533

    Article  PubMed  CAS  Google Scholar 

  17. Giasson BI, Forman MS, Higuchi M, Golbe LI, Graves CL, Kotzbauer PT, Trojanowski JQ, Lee VM-Y (2003) Initiation and synergistic fibrillization of tau and α-synuclein. Science 300:636–640

    Article  PubMed  CAS  Google Scholar 

  18. Gispert S, Del Turco D, Garrett L, Chen A, Bernard DJ, Hamm-Clement J, Korf H-W, Deller T, Braak H, Auburger G, Nussbaum RL (2003) Transgenic mice expressing mutant A53T human α-synuclein show neuronal dysfunction in the absence of aggregate formation. Mol Cell Neurosci 24:419–429

    Article  PubMed  CAS  Google Scholar 

  19. Goedert M (2001) Alpha-synuclein and neurodegenerative diseases. Nat Rev Neurosci 2:492–501

    Article  PubMed  CAS  Google Scholar 

  20. Gomez-Isla T, Irizarry MC, Mariash A, Cheung B, Soto O, Schrump S, Sondel J, Kotilinek L, Day J, Schwarzschild MA, Cha J-HJ, Newell K, Miller DW, Uéda K, Young AB, Hyman BT, Ashe KH (2003) Motor dysfunction and gliosis with preserved dopaminergic markers in human α-synuclein A30P transgenic mice. Neurobiol Aging 24:245–258

    Article  PubMed  CAS  Google Scholar 

  21. Greenbaum EA, Graves CL, Mishizen-Eberz AJ, Lupoli MA, Lynch DR, Englander SW, Axelsen PH, Giasson BI (2005) The E46K mutation in α-synuclein increases amyloid fibril formation. J Biol Chem 280:7800–7807

    Article  PubMed  CAS  Google Scholar 

  22. Gwinn-Hardy K, Mehta ND, Farrer M, Maraganore D, Muenter M, Yen S-H, Hardy J, Dickson DW (2000) Distinctive neuropathology revealed by α-synuclein antibodies in hereditary parkinsonism and dementia linked to chromosome 4p. Acta Neuropathol 99:663–672

    Article  PubMed  CAS  Google Scholar 

  23. Hashimoto M, Rockenstein E, Mante M, Mallory M, Masliah E (2001) β-Synuclein inhibits α-synuclein aggregation. A possible role as an anti-parkinsonian factor. Neuron 32:213–223

    Article  PubMed  CAS  Google Scholar 

  24. Hishikawa N, Hashizume Y, Yoshida M, Sobue G (2001) Widespread occurrence of argyrophilic glial inclusions in Parkinson’s disease. Neuropathol Appl Neurobiol 27:362–372

    Article  PubMed  CAS  Google Scholar 

  25. Hoyer W, Cherny D, Subramaniam V, Jovin TM (2004) Impact of the acidic C-terminal region comprising amino acids 109–140 on α-synuclein aggregation in vitro. Biochemistry 43:16233–16242

    Article  PubMed  CAS  Google Scholar 

  26. Ihara M, Yamasaki N, Hagiwara A, Tanigaki A, Kitano A, Hikawa R, Tomimoto H, Noda M, Takanashi M, Mori H, Hattori N, Miyakawa T, Kinoshita M (2007) Sept4, a component of presynaptic scaffold and Lewy bodies, is required for the suppression of α-synuclein neurotoxicity. Neuron 53:519–533

    Article  PubMed  CAS  Google Scholar 

  27. Kahle PJ, Neumann M, Ozmen L, Müller V, Jacobsen H, Schindzielorz A, Okochi M, Leimer U, van der Putten H, Probst A, Kremmer E, Kretzschmar HA, Haass C (2000) Subcellular localization of wild-type and Parkinson’s disease-associated mutant α-synuclein in human and transgenic mouse brain. J Neurosci 20:6365–6373

    PubMed  CAS  Google Scholar 

  28. Kahle PJ, Neumann M, Ozmen L, Müller V, Jacobsen H, Spooren W, Fuss B, Mallon B, Macklin WB, Fujiwara H, Hasegawa M, Iwatsubo T, Kretzschmar HA, Haass C (2002) Hyperphosphorylation and insolubility of α-synuclein in transgenic mouse oligodendrocytes. EMBO Rep 3:583–588

    Article  PubMed  CAS  Google Scholar 

  29. Kahle PJ, Neumann M, Ozmen L, Müller V, Odoy S, Jacobsen H, Iwatsubo T, Trojanowski JQ, Takahashi H, Wakabayashi K, Bogdanovic N, Riederer P, Kretzschmar HA, Haass C (2001) Selective insolubility of α-synuclein in human Lewy body diseases is recapitulated in a transgenic mouse model. Am J Pathol 159:2215–2225

    PubMed  CAS  Google Scholar 

  30. Klucken J, Ingelsson M, Shin Y, Irizarry MC, Hedley-Whyte ET, Frosch MP, Growdon JH, McLean PJ, Hyman BT (2006) Clinical and biochemical correlates of insoluble α-synuclein in dementia with Lewy bodies. Acta Neuropathol 111:101–108

    Article  PubMed  CAS  Google Scholar 

  31. Krüger R, Kuhn W, Müller T, Woitalla D, Graeber M, Kösel S, Przuntek H, Epplen JT, Schöls L, Riess O (1998) Ala30Pro mutation in the gene encoding α-synuclein in Parkinson’s disease. Nat Genet 18:106–108

    Article  PubMed  Google Scholar 

  32. Krüger R, Schöls L, Del Tredici K, Seidel K, Braak H, Deller T, Rüb U (2006) Familial Parkinson’s disease: the first pathoanatomical study on a carrier of the A30P mutation in the α-synuclein gene. Movement Disord 21:S610

    Google Scholar 

  33. Lantos PL (1998) The definition of multiple system atrophy: a review of recent developments. J Neuropathol Exp Neurol 57:1099–1111

    Article  PubMed  CAS  Google Scholar 

  34. Lee MK, Stirling W, Xu Y, Xu X, Qui D, Mandir AS, Dawson TM, Copeland NG, Jenkins NA, Price DL (2002) Human α-synuclein-harboring familial Parkinson’s disease-linked Ala-53 -> Thr mutation causes neurodegenerative disease with α-synuclein aggregation in transgenic mice. Proc Natl Acad Sci USA 99:8968–8973

    Article  PubMed  CAS  Google Scholar 

  35. Li W, West N, Colla E, Pletnikova O, Troncoso JC, Marsh L, Dawson TM, Jäkälä P, Hartmann T, Price DL, Lee MK (2005) Aggregation promoting C-terminal truncation of α-synuclein is a normal cellular process and is enhanced by the familial Parkinson’s disease-linked mutations. Proc Natl Acad Sci USA 102:2162–2167

    Article  PubMed  CAS  Google Scholar 

  36. Martin LJ, Pan Y, Price AC, Sterling W, Copeland NG, Jenkins NA, Price DL, Lee MK (2006) Parkinson’s disease α-synuclein transgenic mice develop neuronal mitochondrial degeneration and cell death. J Neurosci 26:41–50

    Article  PubMed  CAS  Google Scholar 

  37. Maskri L, Zhu X, Fritzen S, Kühn K, Ullmer C, Engels P, Andriske M, Stichel CC, Lübbert H (2004) Influence of different promoters on the expression pattern of mutated human α-synuclein in transgenic mice. Neurodegener Dis 1:255–265

    Article  PubMed  CAS  Google Scholar 

  38. Masliah E, Rockenstein E, Adame A, Alford M, Crews L, Hashimoto M, Seubert P, Lee M, Goldstein J, Chilcote T, Games D, Schenk D (2005) Effects of α-synuclein immunization in a mouse model of Parkinson’s disease. Neuron 46:857–868

    Article  PubMed  CAS  Google Scholar 

  39. Masliah E, Rockenstein E, Veinbergs I, Mallory M, Hashimoto M, Takeda A, Sagara Y, Sisk A, Mucke L (2000) Dopaminergic loss and inclusion body formation in α-synuclein mice: implications for neurodegenerative disorders. Science 287:1265–1269

    Article  PubMed  CAS  Google Scholar 

  40. Masliah E, Rockenstein E, Veinbergs I, Sagara Y, Mallory M, Hashimoto M, Mucke L (2001) β-Amyloid peptides enhance α-synuclein accumulation and neuronal deficits in a transgenic mouse model linking Alzheimer’s disease and Parkinson’s disease. Proc Natl Acad Sci USA 98:12245–12250

    Article  PubMed  CAS  Google Scholar 

  41. Matsuoka Y, Vila M, Lincoln S, McCormack A, Picciano M, LaFrancois J, Yu X, Dickson D, Langston WJ, McGowan E, Farrer M, Hardy J, Duff K, Przedborski S, Di Monte DA (2001) Lack of nigral pathology in transgenic mice expressing human α-synuclein driven by the tyrosine hydroxylase promoter. Neurobiol Dis 8:535–539

    Article  PubMed  CAS  Google Scholar 

  42. Mishizen-Eberz AJ, Guttmann RP, Giasson BI, Day GA 3rd, Hodara R, Ischiropoulos H, Lee VM-Y, Trojanowski JQ, Lynch DR (2003) Distinct cleavage patterns of normal and pathologic forms of α-synuclein by calpain I in vitro. J Neurochem 86:836–847

    Article  PubMed  CAS  Google Scholar 

  43. Murray IVJ, Giasson BI, Quinn SM, Koppaka V, Axelsen PH, Ischiropoulos H, Trojanowski JQ, Lee VM-Y (2003) Role of α-synuclein carboxy-terminus on fibril formation in vitro. Biochemistry 42:8530–8540

    Article  PubMed  CAS  Google Scholar 

  44. Neumann M, Kahle PJ, Giasson BI, Ozmen L, Borroni E, Spooren W, Müller V, Odoy S, Fujiwara H, Hasegawa M, Iwatsubo T, Trojanowski JQ, Kretzschmar HA, Haass C (2002) Misfolded proteinase K-resistant hyperphosphorylated α-synuclein in aged transgenic mice with locomotor deterioration and in human α-synucleinopathies. J Clin Invest 110:1429–1439

    Article  PubMed  CAS  Google Scholar 

  45. Neumann M, Müller V, Kretzschmar HA, Haass C, Kahle PJ (2004) Regional distribution of proteinase K-resistant α-synuclein correlates with Lewy body disease stage. J Neuropathol Exp Neurol 63:1225–1235

    PubMed  CAS  Google Scholar 

  46. Nieto M, Gil-Bea FJ, Dalfó E, Cuadrado M, Cabodevilla F, Sánchez B, Catena S, Sesma T, Ribé E, Ferrer I, Ramírez MJ, Gómez-Isla T (2006) Increased sensitivity to MPTP in human α-synuclein A30P transgenic mice. Neurobiol Aging 27:848–856

    Article  PubMed  CAS  Google Scholar 

  47. Nuscher B, Kamp F, Mehnert T, Odoy S, Haass C, Kahle PJ, Beyer K (2004) α-Synuclein has a high affinity for packing defects in a bilayer membrane: a thermodynamics study. J Biol Chem 279:21966–21975

    Article  PubMed  CAS  Google Scholar 

  48. Olanow CW, Perl DP, DeMartino GN, McNaught KS (2004) Lewy-body formation is an aggresome-related process: a hypothesis. Lancet Neurol 3:496–503

    Article  PubMed  Google Scholar 

  49. Park JY, Lansbury PT Jr (2003) β-Synuclein inhibits formation of α-synuclein protofibrils: a possible therapeutic strategy against Parkinson’s disease. Biochemistry 42:3696–3700

    Article  PubMed  CAS  Google Scholar 

  50. Polymeropoulos MH, Lavedan C, Leroy E, Ide SE, Dehejia A, Dutra A, Pike B, Root H, Rubenstein J, Boyer R, Stenroos ES, Chandrasekharappa S, Athanassiadou A, Papapetropoulos T, Johnson WG, Lazzarini AM, Duvoisin RC, Di Iorio G, Golbe LI, Nussbaum RL (1997) Mutation in the α-synuclein gene identified in families with Parkinson’s disease. Science 276:2045–2047

    Article  PubMed  CAS  Google Scholar 

  51. Rathke-Hartlieb S, Kahle PJ, Neumann M, Ozmen L, Haid S, Okochi M, Haass C, Schulz JB (2001) Sensitivity to MPTP is not increased in Parkinson’s disease-associated mutant α-synuclein transgenic mice. J Neurochem 77:1181–1184

    Article  PubMed  CAS  Google Scholar 

  52. Richfield EK, Thiruchelvam MJ, Cory-Slechta DA, Wuertzer C, Gainetdinov RR, Caron MG, Di Monte DA, Federoff HJ (2002) Behavioral and neurochemical effects of wild-type and mutated human α-synuclein in transgenic mice. Exp Neurol 175:35–48

    Article  PubMed  CAS  Google Scholar 

  53. Rockenstein E, Mallory M, Hashimoto M, Song D, Shults CW, Lang I, Masliah E (2002) Differential neuropathological alterations in transgenic mice expressing α-synuclein from the platelet-derived growth factor and Thy-1 promoters. J Neurosci Res 68:568–578

    Article  PubMed  CAS  Google Scholar 

  54. Rockenstein E, Schwach G, Ingolic E, Adame A, Crews L, Mante M, Pfragner R, Schreiner E, Windisch M, Masliah E (2005) Lysosomal pathology associated with α-synuclein accumulation in transgenic models using an eGFP fusion protein. J Neurosci Res 80:247–259

    Article  PubMed  CAS  Google Scholar 

  55. Shults CW, Rockenstein E, Crews L, Adame A, Mante M, Larrea G, Hashimoto M, Song D, Iwatsubo T, Tsuboi K, Masliah E (2005) Neurological and neurodegenerative alterations in a transgenic mouse model expressing human α-synuclein under oligodendrocyte promoter: implications for multiple system atrophy. J Neurosci 25:10689–10699

    Article  PubMed  CAS  Google Scholar 

  56. Sidhu A, Wersinger C, Vernier P (2004) Does α-synuclein modulate dopaminergic synaptic content and tone at the synapse? FASEB J 18:637–647

    Article  PubMed  CAS  Google Scholar 

  57. Singleton A, Gwinn-Hardy K (2004) Parkinson’s disease and dementia with Lewy bodies: a difference in dose?. Lancet 364:1105–1107

    Article  PubMed  Google Scholar 

  58. Singleton AB, Farrer M, Johnson J, Singleton A, Hague S, Kachergus J, Hulihan M, Peuralinna T, Dutra A, Nussbaum R, Lincoln S, Crawley A, Hanson M, Maraganore D, Adler C, Cookson MR, Muenter M, Baptista M, Miller D, Blancato J, Hardy J, Gwinn-Hardy K (2003) α-Synuclein locus triplication causes Parkinson’s disease. Science 302:841

    Article  PubMed  CAS  Google Scholar 

  59. Specht CG, Schoepfer R (2004) Deletion of multimerin-1 α-synuclein-deficient mice. Genomics 83:1176–1178

    Article  PubMed  CAS  Google Scholar 

  60. Spillantini MG, Crowther RA, Jakes R, Cairns NJ, Lantos PL, Goedert M (1998) Filamentous α-synuclein inclusions link multiple system atrophy with Parkinson’s disease and dementia with Lewy bodies. Neurosci Lett 251:205–208

    Article  PubMed  CAS  Google Scholar 

  61. Spillantini MG, Schmidt ML, Lee VM-Y, Trojanowski JQ, Jakes R, Goedert M (1997) α-Synuclein in Lewy bodies. Nature 388:839–840

    Article  PubMed  CAS  Google Scholar 

  62. Spira PJ, Sharpe DM, Halliday G, Cavanagh J, Nicholson GA (2001) Clinical and pathological features of a Parkinsonian syndrome in a family with an Ala53Thr α-synuclein mutation. Ann Neurol 49:313–319

    Article  PubMed  CAS  Google Scholar 

  63. Stefanova N, Reindl M, Neumann M, Haass C, Poewe W, Kahle PJ, Wenning GK (2005) Oxidative stress in transgenic mice with oligodendroglial α-Synuclein overexpression replicates the characteristic neuropathology of multiple system atrophy. Am J Pathol 166:869–876

    PubMed  CAS  Google Scholar 

  64. Stefanova N, Reindl M, Neumann M, Kahle PJ, Poewe W, Wenning GK (2007) Microglial activation mediates neurodegeneration related to oligodendroglial α-synucleinopathy: implications for multiple system atrophy. Movement Disord. doi:10.1002/mds.21671

  65. Steidl JV, Gomez-Isla T, Mariash A, Hsiao Ashe K, Boland LM (2003) Altered short-term hippocampal synaptic plasticity in mutant α-synuclein transgenic mice. NeuroReport 14:219–223

    Article  PubMed  Google Scholar 

  66. Thiruchelvam MJ, Powers JM, Cory-Slechta DA, Richfield EK (2004) Risk factors for dopaminergic neuron loss in human α-synuclein transgenic mice. Eur J Neurosci 19:845–854

    Article  PubMed  CAS  Google Scholar 

  67. Tofaris GK, Garcia Reitböck P, Humby T, Lambourne SL, O’Connell M, Ghetti B, Gossage H, Emson PC, Wilkinson LS, Goedert M, Spillantini MG (2006) Pathological changes in dopaminergic nerve cells of the substantia nigra and olfactory bulb in mice transgenic for truncated human α-synuclein(1–120): implications for Lewy body disorders. J Neurosci 26:3942–3950

    Article  PubMed  CAS  Google Scholar 

  68. Touchman JW, Dehejia A, Chiba-Falek O, Cabin DE, Schwartz JR, Orrison BM, Polymeropoulos MH, Nussbaum RL (2001) Human and mouse α-synuclein genes: comparative genomic sequence analysis and identification of a novel gene regulatory element. Genome Res 11:78–86

    Article  PubMed  CAS  Google Scholar 

  69. Unger EL, Eve DJ, Perez XA, Reichenbach DK, Xu Y, Lee MK, Andrews AM (2006) Locomotor hyperactivity and alterations in dopamine neurotransmission are associated with overexpression of A53T mutant human α-synuclein in mice. Neurobiol Dis 21:431–443

    Article  PubMed  CAS  Google Scholar 

  70. Uversky VN, Li J, Souillac P, Millett IS, Doniach S, Jakes R, Goedert M, Fink AL (2002) Biophysical properties of the synucleins and their propensities to fibrillate: inhibition of α-synuclein assembly by β- and γ-synucleins. J Biol Chem 277:11970–11978

    Article  PubMed  CAS  Google Scholar 

  71. Uéda K, Fukushima H, Masliah E, Xia Y, Iwai A, Yoshimoto M, Otero DA, Kondo J, Ihara Y, Saitoh T (1993) Molecular cloning of cDNA encoding an unrecognized component of amyloid in Alzheimer disease. Proc Natl Acad Sci USA 90:11282–11286

    Article  PubMed  Google Scholar 

  72. van der Putten H, Wiederhold K-H, Probst A, Barbieri S, Mistl C, Danner S, Kauffmann S, Hofele K, Spooren WPJM, Ruegg MA, Lin S, Caroni P, Sommer B, Tolnay M, Bilbe G (2000) Neuropathology in mice expressing human α-synuclein. J Neurosci 20:6021–6029

    PubMed  Google Scholar 

  73. von Coelln R, Thomas B, Andrabi SA, Lim KL, Savitt JM, Saffary R, Stirling W, Bruno K, Hess EJ, Lee MK, Dawson VL, Dawson TM (2006) Inclusion body formation and neurodegeneration are parkin independent in a mouse model of α-synucleinopathy. J Neurosci 26:3685–3696

    Article  CAS  Google Scholar 

  74. Wakabayashi K, Hayashi S, Kakita A, Yamada M, Toyoshima Y, Yoshimoto M, Takahashi H (1998) Accumulation of α-synuclein/NACP is a cytopathological feature common to Lewy body disease and multiple system atrophy. Acta Neuropathol 96:445–452

    Article  PubMed  CAS  Google Scholar 

  75. Wakabayashi K, Hayashi S, Yoshimoto M, Kudo H, Takahashi H (2000) NACP/α-synuclein-positive filamentous inclusions in astrocytes and oligodendrocytes of Parkinson’s disease brains. Acta Neuropathol 99:14–20

    Article  PubMed  CAS  Google Scholar 

  76. Wakamatsu M, Ishii A, Iwata S, Sakagami J, Ukai Y, Ono M, Kanbe D, Muramatsu S, Kobayashi K, Iwatsubo T, Yoshimoto M (2006) Selective loss of nigral dopamine neurons induced by overexpression of truncated human α-synuclein in mice. Neurobiol Aging. doi:10.1016/j.neurobiolaging.2006.11.017

  77. Winner B, Lie DC, Rockenstein E, Aigner R, Aigner L, Masliah E, Kuhn HG, Winkler J (2004) Human wild-type α-synuclein impairs neurogenesis. J Neuropathol Exp Neurol 63:1155–1166

    PubMed  CAS  Google Scholar 

  78. Winner B, Rockenstein E, Lie DC, Aigner R, Mante M, Bogdahn U, Couillard-Depres S, Masliah E, Winkler J (2007) Mutant α-synuclein exacerbates age-related decrease of neurogenesis. Neurobiol Aging. doi:10.1016/j.neurobiolaging 2006.12.016

  79. Yamaguchi K, Cochran EJ, Murrell JR, Polymeropoulos MH, Shannon KM, Crowther RA, Goedert M, Ghetti B (2005) Abundant neuritic inclusions and microvacuolar changes in a case of diffuse Lewy body disease with the A53T mutation in the α-synuclein gene. Acta Neuropathol 110:298–305

    Article  PubMed  Google Scholar 

  80. Yazawa I, Giasson BI, Sasaki R, Zhang B, Joyce S, Uryu K, Trojanowski JQ, Lee VM-Y (2005) Mouse model of multiple system atrophy: α-synuclein expression in oligodendrocytes causes glial and neuronal degeneration. Neuron 45:847–859

    Article  PubMed  CAS  Google Scholar 

  81. Zarranz JJ, Alegre J, Gómez-Esteban JC, Lezcano E, Ros R, Ampuero I, Vidal L, Hoenicka J, Rodriguez O, Atarés B, Llorens V, Gomez Tortosa E, del Ser T, Muñoz DG, de Yebenes JG (2004) The new mutation, E46K, of α-synuclein causes Parkinson and Lewy body dementia. Ann Neurol 55:164–173

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgments

Own work cited here was supported by the Deutsche Forschungsgemeinschaft (grants HA 1737/4 and SFB A1) and the German National Genome Research Network (NGFN). Immunostainings were kindly provided by Manuela Neumann. I thank Wolfdieter Springer, Susanne Schöbel and Silke Nuber for their comments and suggestions.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Philipp J. Kahle.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Kahle, P.J. α-Synucleinopathy models and human neuropathology: similarities and differences. Acta Neuropathol 115, 87–95 (2008). https://doi.org/10.1007/s00401-007-0302-x

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00401-007-0302-x

Keywords

Navigation