Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

The many faces of α-synuclein: from structure and toxicity to therapeutic target

Key Points

  • Recent studies of the structural and functional roles of α-synuclein show that this protein participates in synaptic vesicle transport.

  • Increased expression and/or accumulation of α-synuclein owing to genetic duplication, mutations or a failure in clearance may have roles in Parkinson's disease and related disorders.

  • Different conformers of α-synuclein, including oligomers, protofibrils and fibrils, may contribute to α-synuclein-mediated toxicity.

  • Recent studies suggest that the propagation and transmission of α-synuclein participate in the pathogenesis of Parkinson's disease.

  • Reducing α-synuclein expression, aggregation or propagation, or increasing the clearance of this protein all represent viable therapeutic strategies for combating Parkinson's disease and related disorders.

Abstract

Disorders characterized by α-synuclein (α-syn) accumulation, Lewy body formation and parkinsonism (and in some cases dementia) are collectively known as Lewy body diseases. The molecular mechanism (or mechanisms) through which α-syn abnormally accumulates and contributes to neurodegeneration in these disorders remains unknown. Here, we provide an overview of current knowledge and prevailing hypotheses regarding the conformational, oligomerization and aggregation states of α-syn and their role in regulating α-syn function in health and disease. Understanding the nature of the various α-syn structures, how they are formed and their relative contributions to α-syn-mediated toxicity may inform future studies aiming to develop therapeutic prevention and intervention.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Functional properties of α-synuclein.
Figure 2: Biochemical structure of α-synuclein and its pathological distribution in Parkinson's disease, and a mouse model of Lewy body disease.
Figure 3: Cellular events controlling intracellular α-synuclein levels and possible therapeutic strategies to combat α-synuclein accumulation and transmission.
Figure 4: Mechanisms of α-synuclein aggregation and propagation.

Similar content being viewed by others

References

  1. McKeith, I. G. et al. Consensus guidelines for the clinical and pathologic diagnosis of dementia with Lewy bodies (DLB): report of the consortium on DLB international workshop. Neurology 47, 1113–1124 (1996).

    Article  CAS  PubMed  Google Scholar 

  2. Braak, H. & Braak, E. Pathoanatomy of Parkinson's disease. J. Neurol. 247, II3–II10 (2000). A detailed description of the pathoanatomy that occurs in PD.

    Article  PubMed  Google Scholar 

  3. Vekrellis, K., Xilouri, M., Emmanouilidou, E., Rideout, H. J. & Stefanis, L. Pathological roles of α-synuclein in neurological disorders. Lancet Neurol. 10, 1015–1025 (2011).

    Article  CAS  PubMed  Google Scholar 

  4. Chartier-Harlin, M. C. et al. α-synuclein locus duplication as a cause of familial Parkinson's disease. Lancet 364, 1167–1169 (2004).

    Article  CAS  PubMed  Google Scholar 

  5. Kruger, R. et al. Ala30Pro mutation in the gene encoding α-synuclein in Parkinson's disease. Nature Genet. 18, 106–108 (1998).

    Article  CAS  PubMed  Google Scholar 

  6. Polymeropoulos, M. H. et al. Mutation in the α-synuclein gene identified in families with Parkinson's disease. Science 276, 2045–2047 (1997).

    Article  CAS  PubMed  Google Scholar 

  7. Zarranz, J. J. et al. The new mutation, E46K, of α-synuclein causes Parkinson and Lewy body dementia. Ann. Neurol. 55, 164–173 (2004).

    Article  CAS  PubMed  Google Scholar 

  8. Simon-Sanchez, J. et al. Genome-wide association study reveals genetic risk underlying Parkinson's disease. Nature Genet. 41, 1308–1312 (2009).

    Article  CAS  PubMed  Google Scholar 

  9. Conway, K. A., Harper, J. D. & Lansbury, P. T. Accelerated in vitro fibril formation by a mutant α-synuclein linked to early-onset Parkinson disease. Nature Med. 4, 1318–1320 (1998).

    Article  CAS  PubMed  Google Scholar 

  10. Tsigelny, I. F. et al. Mechanisms of hybrid oligomer formation in the pathogenesis of combined Alzheimer's and Parkinson's diseases. PLoS ONE 3, e3135 (2008).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  11. Oueslati, A., Fournier, M. & Lashuel, H. A. Role of post-translational modifications in modulating the structure, function and toxicity of α-synuclein: implications for Parkinson's disease pathogenesis and therapies. Prog. Brain Res. 183, 115–145 (2010).

    Article  CAS  PubMed  Google Scholar 

  12. Taschenberger, G. et al. Aggregation of α-synuclein promotes progressive in vivo neurotoxicity in adult rat dopaminergic neurons. Acta Neuropathol. 123, 671–683 (2011).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  13. Galvin, J. E., Lee, V. M. & Trojanowski, J. Q. Synucleinopathies: clinical and pathological implications. Arch. Neurol. 58, 186–190 (2001).

    Article  CAS  PubMed  Google Scholar 

  14. Wang, S. et al. α-Synuclein disrupts stress signaling by inhibiting polo-like kinase Cdc5/Plk2. Proc. Natl Acad. Sci. USA 109, 16119–16124 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Iwai, A. et al. The precursor protein of non-Aβ component of Alzheimer's disease amyloid is a presynaptic protein of the central nervous system. Neuron 14, 467–475 (1995). This study identified NAC as a presynaptic protein.

    Article  CAS  PubMed  Google Scholar 

  16. Jakes, R., Spillantini, M. G. & Goedert, M. Identification of two distinct synucleins from human brain. FEBS Lett. 345, 27–32 (1994).

    Article  CAS  PubMed  Google Scholar 

  17. Withers, G. S., George, J. M., Banker, G. A. & Clayton, D. F. Delayed localization of synelfin (synuclein, NACP) to presynaptic terminals in cultured rat hippocampal neurons. Brain Res. Dev. Brain Res. 99, 87–94 (1997).

    Article  CAS  PubMed  Google Scholar 

  18. Kahle, P. J. et al. Subcellular localization of wild-type and Parkinson's disease-associated mutant α-synuclein in human and transgenic mouse brain. J. Neurosci. 20, 6365–6373 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Lee, S. J., Jeon, H. & Kandror, K. V. α-synuclein is localized in a subpopulation of rat brain synaptic vesicles. Acta Neurobiol. Exp. 68, 509–515 (2008).

    Google Scholar 

  20. Zhang, L. et al. Semi-quantitative analysis of α-synuclein in subcellular pools of rat brain neurons: an immunogold electron microscopic study using a C-terminal specific monoclonal antibody. Brain Res. 1244, 40–52 (2008).

    Article  CAS  PubMed  Google Scholar 

  21. Cabin, D. E. et al. Synaptic vesicle depletion correlates with attenuated synaptic responses to prolonged repetitive stimulation in mice lacking α-synuclein. J. Neurosci. 22, 8797–8807 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Abeliovich, A. et al. Mice lacking α-synuclein display functional deficits in the nigrostriatal dopamine system. Neuron. 25, 239–252 (2000).

    Article  CAS  PubMed  Google Scholar 

  23. Murphy, D. D., Rueter, S. M., Trojanowski, J. Q. & Lee, V. M. Synucleins are developmentally expressed, and α-synuclein regulates the size of the presynaptic vesicular pool in primary hippocampal neurons. J. Neurosci. 20, 3214–3220 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Yavich, L., Tanila, H., Vepsalainen, S. & Jakala, P. Role of α-synuclein in presynaptic dopamine recruitment. J. Neurosci. 24, 11165–11170 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Scott, D. A. et al. A pathologic cascade leading to synaptic dysfunction in α-synuclein-induced neurodegeneration. J. Neurosci. 30, 8083–8095 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Nemani, V. M. et al. Increased expression of α-synuclein reduces neurotransmitter release by inhibiting synaptic vesicle reclustering after endocytosis. Neuron 65, 66–79 (2010). This study found that α-syn overexpression inhibited neurotransmitter release by reducing the size of the synaptic vesicle recycling pool.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Gaugler, M. N. et al. Nigrostriatal overabundance of α-synuclein leads to decreased vesicle density and deficits in dopamine release that correlate with reduced motor activity. Acta Neuropathol. 123, 653–669 (2012).

    Article  CAS  PubMed  Google Scholar 

  28. Lundblad, M., Decressac, M., Mattsson, B. & Bjorklund, A. Impaired neurotransmission caused by overexpression of α-synuclein in nigral dopamine neurons. Proc. Natl Acad. Sci. USA 109, 3213–3219 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Larsen, K. E. et al. α-synuclein overexpression in PC12 and chromaffin cells impairs catecholamine release by interfering with a late step in exocytosis. J. Neurosci. 26, 11915–11922 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Scott, D. & Roy, S. α-synuclein inhibits intersynaptic vesicle mobility and maintains recycling-pool homeostasis. J. Neurosci. 32, 10129–10135 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Payton, J. E., Perrin, R. J., Woods, W. S. & George, J. M. Structural determinants of PLD2 inhibition by α-synuclein. J. Mol. Biol. 337, 1001–1009 (2004).

    Article  CAS  PubMed  Google Scholar 

  32. Dalfo, E. & Ferrer, I. α-synuclein binding to rab3a in multiple system atrophy. Neurosci. Lett. 380, 170–175 (2005).

    Article  CAS  PubMed  Google Scholar 

  33. Burre, J. et al. α-synuclein promotes SNARE-complex assembly in vivo and in vitro. Science 329, 1663–1667 (2010). This paper identifies α-synuclein as a non-classical chaperone that binds and promotes SNARE-complex assembly.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Chandra, S. et al. Double-knockout mice for α- and β-synucleins: effect on synaptic functions. Proc. Natl Acad. Sci. USA 101, 14966–14971 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Chandra, S., Gallardo, G., Fernandez-Chacon, R., Schluter, O. M. & Sudhof, T. C. α-synuclein cooperates with CSPα in preventing neurodegeneration. Cell 123, 383–396 (2005).

    Article  CAS  PubMed  Google Scholar 

  36. Ueda, K. et al. Molecular cloning of cDNA encoding an unrecognized component of amyloid in Alzheimer disease. Proc. Natl Acad. Sci. USA 90, 11282–11286 (1993). This is the first report of the NAC being a 140-amino-acid protein.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Ulmer, T. S., Bax, A., Cole, N. B. & Nussbaum, R. L. Structure and dynamics of micelle-bound human α-synuclein. J. Biol. Chem. 280, 9595–9603 (2005).

    Article  CAS  PubMed  Google Scholar 

  38. Eliezer, D., Kutluay, E., Bussell, R. Jr & Browne, G. Conformational properties of α-synuclein in its free and lipid-associated states. J. Mol. Biol. 307, 1061–1073 (2001).

    Article  CAS  PubMed  Google Scholar 

  39. Masliah, E., Iwai, A., Mallory, M., Ueda, K. & Saitoh, T. Altered presynaptic protein NACP is associated with plaque formation and neurodegeneration in Alzheimer's disease. Am. J. Pathol. 148, 201–210 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Spillantini, M. G. et al. α-synuclein in Lewy bodies. Nature 388, 839–840 (1997).

    Article  CAS  PubMed  Google Scholar 

  41. Hashimoto, M., Takenouchi, T., Mallory, M., Masliah, E. & Takeda, A. The role of NAC in amyloidogenesis in Alzheimer's disease. Am. J. Pathol. 156, 734–736 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. El-Agnaf, O. M., Jakes, R., Curran, M. D. & Wallace, A. Effects of the mutations Ala30 to Pro and Ala53 to Thr on the physical and morphological properties of α-synuclein protein implicated in Parkinson's disease. FEBS Lett. 440, 67–70 (1998).

    Article  CAS  PubMed  Google Scholar 

  43. Giasson, B. I., Murray, I. V., Trojanowski, J. Q. & Lee, V. M. A hydrophobic stretch of 12 amino acid residues in the middle of α-synuclein is essential for filament assembly. J. Biol. Chem. 276, 2380–2386 (2001). This study showed that the middle hydrophobic domain of α-syn is necessary and sufficient for fibrillization.

    Article  CAS  PubMed  Google Scholar 

  44. Luk, K. C. et al. Exogenous α-synuclein fibrils seed the formation of Lewy body-like intracellular inclusions in cultured cells. Proc. Natl Acad. Sci. USA 106, 20051–20056 (2009). These authors showed that α-syn seeds can recruit endogenous soluble α-syn protein to form pathological species.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Weinreb, P. H., Zhen, W., Poon, A. W., Conway, K. A. & Lansbury, P. T. Jr. NACP, a protein implicated in Alzheimer's disease and learning, is natively unfolded. Biochemistry 35, 13709–13715 (1996).

    Article  CAS  PubMed  Google Scholar 

  46. Fauvet, B. et al. α-synuclein in the central nervous system and from erythrocytes, mammalian cells and E. coli exists predominantly as a disordered monomer. J. Biol. Chem. 287, 15345–15364 (2012). This study reassessed the oligomeric state of α-syn and demonstrated that native α-syn exists predominantly as an unfolded monomer and not a tetramer.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Ramakrishnan, M., Jensen, P. H. & Marsh, D. Association of α-synuclein and mutants with lipid membranes: spin-label ESR and polarized IR. Biochemistry 45, 3386–3395 (2006).

    Article  CAS  PubMed  Google Scholar 

  48. Ullman, O., Fisher, C. K. & Stultz, C. M. Explaining the structural plasticity of α-synuclein. J. Am. Chem. Soc. 133, 19536–19546 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Hashimoto, M. et al. Oxidative stress induces amyloid-like aggregate formation of NACP/α-synuclein in vitro. Neuroreport 10, 717–721 (1999).

    Article  CAS  PubMed  Google Scholar 

  50. Andringa, G. et al. Tissue transglutaminase catalyzes the formation of α-synuclein crosslinks in Parkinson's disease. FASEB J. 18, 932–934 (2004).

    Article  CAS  PubMed  Google Scholar 

  51. Paleologou, K. E. et al. Phosphorylation at S87 is enhanced in synucleinopathies, inhibits α-synuclein oligomerization, and influences synuclein-membrane interactions. J. Neurosci. 30, 3184–3198 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Li, W. et al. 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 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Dufty, B. M. et al. Calpain-cleavage of α-synuclein: connecting proteolytic processing to disease-linked aggregation. Am. J. Pathol. 170, 1725–1738 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Perrin, R. J., Woods, W. S., Clayton, D. F. & George, J. M. Exposure to long chain polyunsaturated fatty acids triggers rapid multimerization of synucleins. J. Biol. Chem. 276, 41958–41962 (2001).

    Article  CAS  PubMed  Google Scholar 

  55. Sharon, R. et al. The formation of highly soluble oligomers of α-synuclein is regulated by fatty acids and enhanced in Parkinson's disease. Neuron 37, 583–595 (2003).

    Article  CAS  PubMed  Google Scholar 

  56. Karube, H. et al. N-terminal region of α-synuclein is essential for the fatty acid-induced oligomerization of the molecules. FEBS Lett. 582, 3693–3700 (2008).

    Article  CAS  PubMed  Google Scholar 

  57. Takeda, A. et al. Abnormal distribution of the non-Aβ component of Alzheimer's disease amyloid precursor/α-synuclein in Lewy body disease as revealed by proteinase K and formic acid pretreatment. Lab. Invest. 78, 1169–1177 (1998).

    CAS  PubMed  Google Scholar 

  58. Suh, Y. H. & Checler, F. Amyloid precursor protein, presenilins, and α-synuclein: molecular pathogenesis and pharmacological applications in Alzheimer's disease. Pharmacol. Rev. 54, 469–525 (2002).

    Article  CAS  PubMed  Google Scholar 

  59. Iwai, A. et al. The synaptic protein NACP is abnormally expressed during the progression of Alzheimer's disease. Brain Res. 720, 230–234 (1996).

    Article  CAS  PubMed  Google Scholar 

  60. Kragh, C. L., Ubhi, K., Wyss-Corey, T. & Masliah, E. Autophagy in dementias. Brain Pathol. 22, 99–109 (2012).

    Article  CAS  PubMed  Google Scholar 

  61. Seidel, K. et al. First appraisal of brain pathology owing to A30P mutant α-synuclein. Ann. Neurol. 67, 684–689 (2010).

    Article  CAS  PubMed  Google Scholar 

  62. Maraganore, D. M. et al. Collaborative analysis of α-synuclein gene promoter variability and Parkinson disease. JAMA 296, 661–670 (2006).

    Article  CAS  PubMed  Google Scholar 

  63. Iwata, A. et al. α-synuclein degradation by serine protease neurosin: implication for pathogenesis of synucleinopathies. Hum. Mol. Genet. 12, 2625–2635 (2003).

    Article  CAS  PubMed  Google Scholar 

  64. Klucken, J., Shin, Y., Masliah, E., Hyman, B. T. & McLean, P. J. Hsp70 reduces α-synuclein aggregation and toxicity. J. Biol. Chem. 279, 25497–25502 (2004).

    Article  CAS  PubMed  Google Scholar 

  65. McNaught, K. S. et al. Impairment of the ubiquitin-proteasome system causes dopaminergic cell death and inclusion body formation in ventral mesencephalic cultures. J. Neurochem. 81, 301–306 (2002).

    Article  CAS  PubMed  Google Scholar 

  66. McNaught, K. S. et al. Proteasome inhibition causes nigral degeneration with inclusion bodies in rats. Neuroreport 13, 1437–1441 (2002).

    Article  CAS  PubMed  Google Scholar 

  67. Webb, J. L., Ravikumar, B., Atkins, J., Skepper, J. N. & Rubinsztein, D. C. α-Synuclein is degraded by both autophagy and the proteasome. J. Biol. Chem. 278, 25009–25013 (2003).

    Article  CAS  PubMed  Google Scholar 

  68. Cuervo, A. M., Stefanis, L., Fredenburg, R., Lansbury, P. T. & Sulzer, D. Impaired degradation of mutant α-synuclein by chaperone-mediated autophagy. Science 305, 1292–1295 (2004). This paper identified lysosomes for degradation of wild-type α-syn by the chaperone-mediated autophagy pathway.

    Article  CAS  PubMed  Google Scholar 

  69. Desplats, P. et al. Inclusion formation and neuronal cell death through neuron-to-neuron transmission of α-synuclein. Proc. Natl Acad. Sci. USA 106, 13010–13015 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Spencer, B. et al. Beclin 1 gene transfer activates autophagy and ameliorates the neurodegenerative pathology in α-synuclein models of Parkinson's and Lewy body diseases. J. Neurosci. 29, 13578–13588 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Crews, L. et al. Selective molecular alterations in the autophagy pathway in patients with Lewy body disease and in models of α-synucleinopathy. PLoS ONE 5, e9313 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  72. Kosaka, K. Diffuse Lewy body disease in Japan. J. Neurol. 237, 197–204 (1990).

    Article  CAS  PubMed  Google Scholar 

  73. Dickson, D. W. et al. Diffuse Lewy body disease: light and electron microscopic immunocytochemistry of senile plaques. Acta Neuropathol. 78, 572–584 (1989).

    Article  CAS  PubMed  Google Scholar 

  74. Braak, H., Sastre, M. & Del Tredici, K. Development of α-synuclein immunoreactive astrocytes in the forebrain parallels stages of intraneuronal pathology in sporadic Parkinson's disease. Acta Neuropathol. 114, 231–241 (2007). Paper reporting the association of α-syn with reactive astrocytes in clinically diagnosed PD cases.

    Article  CAS  PubMed  Google Scholar 

  75. Lee, H. J. et al. Direct transfer of α-synuclein from neuron to astroglia causes inflammatory responses in synucleinopathies. J. Biol. Chem. 285, 9262–9272 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Kahle, P. J. et al. Selective insolubility of α-synuclein in human Lewy body diseases is recapitulated in a transgenic mouse model. Am. J. Pathol. 159, 2215–2225 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Baba, M. et al. Aggregation of α-synuclein in Lewy bodies of sporadic Parkinson's disease and dementia with Lewy bodies. Am. J. Pathol. 152, 879–884 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Lee, M. K. et al. 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 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Tsigelny, I. F. et al. Role of α-synuclein penetration into the membrane in the mechanisms of oligomer pore formation. FEBS J. 279, 1000–1013 (2012). Computational modelling shows that α-synuclein can form ring-like structures that penetrate the membrane.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Bellucci, A., Navarria, L., Zaltieri, M., Missale, C. & Spano, P. α-synuclein synaptic pathology and its implications in the development of novel therapeutic approaches to cure Parkinson's disease. Brain Res. 1432, 95–113 (2012).

    Article  CAS  PubMed  Google Scholar 

  81. Schulz-Schaeffer, W. J. The synaptic pathology of α-synuclein aggregation in dementia with Lewy bodies, Parkinson's disease and Parkinson's disease dementia. Acta Neuropathol. 120, 131–143 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Garcia-Reitbock, P. et al. SNARE protein redistribution and synaptic failure in a transgenic mouse model of Parkinson's disease. Brain 133, 2032–2044 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  83. Horvath, I. et al. Mechanisms of protein oligomerization: inhibitor of functional amyloids templates α-synuclein fibrillation. J. Am. Chem. Soc. 134, 3439–3444 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Conway, K. A. et al. Accelerated oligomerization by Parkinson's disease linked α-synuclein mutants. Ann. NY Acad. Sci. 920, 42–45 (2000).

    Article  CAS  PubMed  Google Scholar 

  85. Cremades, N. et al. Direct observation of the interconversion of normal and toxic forms of α-synuclein. Cell 149, 1048–1059 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Danzer, K. M. et al. Different species of α-synuclein oligomers induce calcium influx and seeding. J. Neurosci. 27, 9220–9232 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Iwatsubo, T. Pathological biochemistry of α-synucleinopathy. Neuropathology 27, 474–478 (2007).

    Article  PubMed  Google Scholar 

  88. Oueslati, A., Paleologou, K. E., Schneider, B. L., Aebischer, P. & Lashuel, H. A. Mimicking phosphorylation at serine 87 inhibits the aggregation of human α-synuclein and protects against its toxicity in a rat model of Parkinson's disease. J. Neurosci. 32, 1536–1544 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Souza, J. M., Giasson, B. I., Chen, Q., Lee, V. M. & Ischiropoulos, H. Dityrosine cross-linking promotes formation of stable α-synuclein polymers. Implication of nitrative and oxidative stress in the pathogenesis of neurodegenerative synucleinopathies. J. Biol. Chem. 275, 18344–18349 (2000).

    Article  CAS  PubMed  Google Scholar 

  90. Uversky, V. N., Li, J. & Fink, A. L. Metal-triggered structural transformations, aggregation, and fibrillation of human α-synuclein — a possible molecular link between Parkinson's disease and heavy metal exposure. J. Biol. Chem. 276, 44284–44296 (2001).

    Article  CAS  PubMed  Google Scholar 

  91. Conway, K. A., Rochet, J. C., Bieganski, R. M. & Lansbury, P. T. Jr. Kinetic stabilization of the α-synuclein protofibril by a dopamine-α-synuclein adduct. Science 294, 1346–1349 (2001).

    Article  CAS  PubMed  Google Scholar 

  92. Conway, K. A. et al. Acceleration of oligomerization, not fibrillization, is a shared property of both α-synuclein mutations linked to early-onset Parkinson's disease: implications for pathogenesis and therapy. Proc. Natl Acad. Sci. USA 97, 571–576 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Hsu, L. J. et al. α-synuclein promotes mitochondrial deficit and oxidative stress. Am. J. Pathol. 157, 401–410 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Hashimoto, M. et al. The role of α-synuclein assembly and metabolism in the pathogenesis of Lewy body disease. J. Mol. Neurosci. 24, 343–352 (2004).

    Article  CAS  PubMed  Google Scholar 

  95. Alim, M. A. et al. Demonstration of a role for α-synuclein as a functional microtubule-associated protein. J. Alzheimers Dis. 6, 435–442 (2004).

    Article  PubMed  Google Scholar 

  96. da Silveira, S. A. et al. Phosphorylation does not prompt, nor prevent, the formation of α-synuclein toxic species in a rat model of Parkinson's disease. Hum. Mol. Genet. 18, 872–887 (2009).

    Google Scholar 

  97. Gorbatyuk, O. S. et al. The phosphorylation state of Ser-129 in human α-synuclein determines neurodegeneration in a rat model of Parkinson disease. Proc. Natl Acad. Sci. USA 105, 763–768 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Winner, B. et al. In vivo demonstration that α-synuclein oligomers are toxic. Proc. Natl Acad. Sci. USA 108, 4194–4199 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Jan, A. et al. Aβ42 neurotoxicity is mediated by ongoing nucleated polymerization process rather than by discrete Aβ42 species. J. Biol. Chem. 286, 8585–8596 (2011).

    Article  CAS  PubMed  Google Scholar 

  100. Wogulis, M. et al. Nucleation-dependent polymerization is an essential component of amyloid-mediated neuronal cell death. J. Neurosci. 25, 1071–1080 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Colla, E. et al. Accumulation of toxic α-synuclein oligomer within endoplasmic reticulum occurs in α-synucleinopathy in vivo. J. Neurosci. 32, 3301–3305 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Jang, A. et al. Non-classical exocytosis of α-synuclein is sensitive to folding states and promoted under stress conditions. J. Neurochem. 113, 1263–1274 (2010).

    CAS  PubMed  Google Scholar 

  103. Danzer, K. M. et al. Heat-shock protein 70 modulates toxic extracellular α-synuclein oligomers and rescues trans-synaptic toxicity. FASEB J. 25, 326–336 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Alvarez-Erviti, L. et al. Lysosomal dysfunction increases exosome-mediated α-synuclein release and transmission. Neurobiol. Dis. 42, 360–367 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Lee, H. J. et al. Assembly-dependent endocytosis and clearance of extracellular α-synuclein. Int. J. Biochem. Cell. Biol. 40, 1835–1849 (2008).

    Article  CAS  PubMed  Google Scholar 

  106. Emmanouilidou, E. et al. Cell-produced α-synuclein is secreted in a calcium-dependent manner by exosomes and impacts neuronal survival. J. Neurosci. 30, 6838–6851 (2010). The first report that α-syn is secreted by externalized vesicles in a calcium-dependent manner.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Jao, C. C., Hegde, B. G., Chen, J., Haworth, I. S. & Langen, R. Structure of membrane-bound α-synuclein from site-directed spin labeling and computational refinement. Proc. Natl Acad. Sci. USA 105, 19666–19671 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Volpicelli-Daley, L. A. et al. Exogenous α-synuclein fibrils induce Lewy body pathology leading to synaptic dysfunction and neuron death. Neuron 72, 57–71 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Crews, L. et al. α-synuclein alters Notch-1 expression and neurogenesis in mouse embryonic stem cells and in the hippocampus of transgenic mice. J. Neurosci. 28, 4250–4260 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Letarov, A., Manival, X., Desplats, C. & Krisch, H. M. gpwac of the T4-type bacteriophages: structure, function, and evolution of a segmented coiled-coil protein that controls viral infectivity. J. Bacteriol. 187, 1055–1066 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Ekberg, H. et al. The specific monocarboxylate transporter-1 (MCT-1) inhibitor, AR-C117977, induces donor-specific suppression, reducing acute and chronic allograft rejection in the rat. Transplantation 84, 1191–1199 (2007).

    Article  CAS  PubMed  Google Scholar 

  112. Karlsson, J., Petersen, A., Gido, G., Wieloch, T. & Brundin, P. Combining neuroprotective treatment of embryonic nigral donor tissue with mild hypothermia of the graft recipient. Cell Transplant. 14, 301–309 (2005).

    Article  PubMed  Google Scholar 

  113. Karlsson, J., Emgard, M., Gido, G., Wieloch, T. & Brundin, P. Increased survival of embryonic nigral neurons when grafted to hypothermic rats. Neuroreport 11, 1665–1668 (2000).

    Article  CAS  PubMed  Google Scholar 

  114. Frodl, E. M., Duan, W. M., Sauer, H., Kupsch, A. & Brundin, P. Human embryonic dopamine neurons xenografted to the rat: effects of cryopreservation and varying regional source of donor cells on transplant survival, morphology and function. Brain Res. 647, 286–298 (1994).

    Article  CAS  PubMed  Google Scholar 

  115. Kordower, J. H., Freeman, T. B. & Olanow, C. W. Neuropathology of fetal nigral grafts in patients with Parkinson's disease. Mov. Disord. 13, 88–95 (1998).

    Article  PubMed  Google Scholar 

  116. Tang, B. et al. Forkhead box protein p1 is a transcriptional repressor of immune signaling in the CNS: implications for transcriptional dysregulation in Huntington disease. Hum. Mol. Genet. 21, 3097–3111 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Nonaka, T., Watanabe, S. T., Iwatsubo, T. & Hasegawa, M. Seeded aggregation and toxicity of α-synuclein and tau: cellular models of neurodegenerative diseases. J. Biol. Chem. 285, 34885–34898 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Luk, K. C. et al. Intracerebral inoculation of pathological α-synuclein initiates a rapidly progressive neurodegenerative α-synucleinopathy in mice. J. Exp. Med. 209, 975–986 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Ban, T. et al. Direct observation of Aβ amyloid fibril growth and inhibition. J. Mol. Biol. 344, 757–767 (2004).

    Article  CAS  PubMed  Google Scholar 

  120. Lashuel, H. A. et al. New class of inhibitors of amyloid-β fibril formation. Implications for the mechanism of pathogenesis in Alzheimer's disease. J. Biol. Chem. 277, 42881–42890 (2002).

    Article  CAS  PubMed  Google Scholar 

  121. Di Giovanni, S. et al. Entacapone and tolcapone, two catechol O-methyltransferase inhibitors, block fibril formation of α-synuclein and β-amyloid and protect against amyloid-induced toxicity. J. Biol. Chem. 285, 14941–14954 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Masuda, M. et al. Small molecule inhibitors of α-synuclein filament assembly. Biochemistry 45, 6085–6094 (2006).

    Article  CAS  PubMed  Google Scholar 

  123. Porat, Y., Abramowitz, A. & Gazit, E. Inhibition of amyloid fibril formation by polyphenols: structural similarity and aromatic interactions as a common inhibition mechanism. Chem. Biol. Drug Des. 67, 27–37 (2006).

    Article  CAS  PubMed  Google Scholar 

  124. Ayrolles-Torro, A. et al. Oligomeric-induced activity by thienyl pyrimidine compounds traps prion infectivity. J. Neurosci. 31, 14882–14892 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Hirohata, M., Ono, K., Morinaga, A. & Yamada, M. Non-steroidal anti-inflammatory drugs have potent anti-fibrillogenic and fibril-destabilizing effects for α-synuclein fibrils in vitro. Neuropharmacology 54, 620–627 (2008).

    Article  CAS  PubMed  Google Scholar 

  126. Bartels, T., Choi, J. G. & Selkoe, D. J. α-Synuclein occurs physiologically as a helically folded tetramer that resists aggregation. Nature 477, 107–110 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Wang, W. et al. A soluble α-synuclein construct forms a dynamic tetramer. Proc. Natl Acad. Sci. USA 108, 17797–17802 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Fauvet, B. et al. Semisynthesis and characterization of N-terminally acetylated α-synuclein: implications for aggregation and cellular properties. J. Biol. Chem. 287, 28243–2862 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Shaikh, S. & Nicholson, L. F. Advanced glycation end products induce in vitro cross-linking of α-synuclein and accelerate the process of intracellular inclusion body formation. J. Neurosci. Res. 86, 2071–2082 (2008).

    Article  CAS  PubMed  Google Scholar 

  130. Hashimoto, M., Takeda, A., Hsu, L. J., Takenouchi, T. & Masliah, E. Role of cytochrome c as a stimulator of α-synuclein aggregation in Lewy body disease. J. Biol. Chem. 274, 28849–28852 (1999).

    Article  CAS  PubMed  Google Scholar 

  131. Lashuel, H. A., Hartley, D., Petre, B. M., Walz, T. & Lansbury, P. T. Jr. Neurodegenerative disease: amyloid pores from pathogenic mutations. Nature 418, 291 (2002).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors acknowledge grant support from the US National Institutes of Health (grants AG5131, AG18440, AG022074 and NS044233 to E.M.), the Swiss National Science Foundation (grant #31003A_120653 to H.A.L.) and the Ecole Polytechnique Fédérale de Lausanne (European Research Council starting grant to H.A.L.).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Eliezer Masliah.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Glossary

Lewy body

An intraneuronal globular inclusion composed primarily of α-synuclein fibrils. Lewy bodies are characteristically found in Parkinson's disease, although they are also detectable in other neurodegenerative diseases, such as dementia with Lewy bodies.

Fibril

Mature fibrils are characterized by the following characteristics: a cross-β-sheet X-ray fibre diffraction pattern; β-sheet-rich circular dichroism and Fourier transform infrared spectroscopy (FTIR) spectra; binding to Congo red and Thioflavin-T/S; and a characteristic filamentous morphology (8–12nm in diameter and >1μm in length), as revealed by atomic force microscope and transmission election microscope imaging. α-synuclein can form fibrils of diverse morphologies depending on the solution conditions.

SNARE

Soluble NSF (N-ethylmaleimide-sensitive factor) attachment protein (SNAP) receptor.

Oilgomer

Oligomers comprise a number of smaller, identical units (monomers). In the context of α-synuclein oligomers, this term encompasses a wide range of species, ranging from low-molecular-weight species (including dimers, trimers and tetramers) to high-molecular-weight species (such as spherical, chain-like and annular structures.

Nuclei

Thermodynamically unstable oligomeric species that are capable of acting as sites for amyloid fibril growth.

Seed

A relatively stable entity (such as a fibril or fragmented fibril) that, when introduced into a solution containing monomeric subunits, serves as an effective nucleus and accelerates fibril formation (by eliminating the lag phase associated with nuclei formation) in a nucleation polymerization process.

Native state

The three-dimensional structure of the protein in its normal physiological milieu in the absence of any denaturing agents or conditions.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Lashuel, H., Overk, C., Oueslati, A. et al. The many faces of α-synuclein: from structure and toxicity to therapeutic target. Nat Rev Neurosci 14, 38–48 (2013). https://doi.org/10.1038/nrn3406

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrn3406

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing