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:

Protein misfolding, aggregation, and conformational strains in neurodegenerative diseases

Abstract

A hallmark event in neurodegenerative diseases (NDs) is the misfolding, aggregation, and accumulation of proteins, leading to cellular dysfunction, loss of synaptic connections, and brain damage. Despite the involvement of distinct proteins in different NDs, the process of protein misfolding and aggregation is remarkably similar. A recent breakthrough in the field was the discovery that misfolded protein aggregates can self-propagate through seeding and spread the pathological abnormalities between cells and tissues in a manner akin to the behavior of infectious prions in prion diseases. This discovery has vast implications for understanding the mechanisms involved in the initiation and progression of NDs, as well as for the design of novel strategies for treatment and diagnosis. In this Review, we provide a critical discussion of the role of protein misfolding and aggregation in NDs. Commonalities and differences between distinct protein aggregates will be highlighted, in addition to evidence supporting the hypothesis that misfolded aggregates can be transmissible by the prion principle. We will also describe the molecular basis and implications for prion-like conformational strains, cross-interaction between different misfolded proteins in the brain, and how these concepts can be applied to the development of novel strategies for therapy and diagnosis.

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

Fig. 1: Protein aggregation and the prion principle of pathological transmission.
Fig. 2: Conformational strains and their implications for the spectrum of synucleinopathies and tauopathies.
Fig. 3: Cross-seeding interactions between diverse misfolded protein aggregates.
Fig. 4: Therapeutic strategies targeting the prion-like spread of misfolded proteins.
Fig. 5: Disease diagnosis by sensitive detection of misfolded seeds in biological fluids.

Similar content being viewed by others

References

  1. Ross, C. A. & Poirier, M. A. Protein aggregation and neurodegenerative disease. Nat. Med. 10(Suppl), S10–S17 (2004).

    PubMed  Google Scholar 

  2. Soto, C. Unfolding the role of protein misfolding in neurodegenerative diseases. Nat. Rev. Neurosci. 4, 49–60 (2003).

    CAS  PubMed  Google Scholar 

  3. Goedert, M. Neurodegeneration. Alzheimer’s and Parkinson’s diseases: The prion concept in relation to assembled Aβ, tau, and α-synuclein. Science 349, 1255555 (2015).

    PubMed  Google Scholar 

  4. Fitzpatrick, A. W. et al. Atomic structure and hierarchical assembly of a cross-β amyloid fibril. Proc. Natl Acad. Sci. USA 110, 5468–5473 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Rambaran, R. N. & Serpell, L. C. Amyloid fibrils: abnormal protein assembly. Prion 2, 112–117 (2008).

    PubMed  PubMed Central  Google Scholar 

  6. Caughey, B. & Lansbury, P. T. Protofibrils, pores, fibrils, and neurodegeneration: separating the responsible protein aggregates from the innocent bystanders. Annu. Rev. Neurosci. 26, 267–298 (2003).

    CAS  PubMed  Google Scholar 

  7. Gadad, B. S., Britton, G. B. & Rao, K. S. Targeting oligomers in neurodegenerative disorders: lessons from α-synuclein, tau, and amyloid-β peptide. J. Alzheimers Dis. 24(Suppl 2), 223–232 (2011).

    CAS  PubMed  Google Scholar 

  8. Glabe, C. G. Common mechanisms of amyloid oligomer pathogenesis in degenerative disease. Neurobiol. Aging 27, 570–575 (2006).

    CAS  PubMed  Google Scholar 

  9. Lesné, S. & Kotilinek, L. Amyloid plaques and amyloid-beta oligomers: an ongoing debate. J. Neurosci. 25, 9319–9320 (2005).

    PubMed  PubMed Central  Google Scholar 

  10. Benilova, I., Karran, E. & De Strooper, B. The toxic Aβ oligomer and Alzheimer’s disease: an emperor in need of clothes. Nat. Neurosci. 15, 349–357 (2012).

    CAS  PubMed  Google Scholar 

  11. Breydo, L. & Uversky, V. N. Structural, morphological, and functional diversity of amyloid oligomers. FEBS Lett. 589(19 Pt A), 2640–2648 (2015).

    CAS  PubMed  Google Scholar 

  12. Lesne, S. E. Toxic oligomer species of amyloid-β in Alzheimer’s disease, a timing issue. Swiss Med. Wkly. 144, w14021 (2014).

    PubMed  PubMed Central  Google Scholar 

  13. Jarrett, J. T. & Lansbury, P. T. Jr. Seeding “one-dimensional crystallization” of amyloid: a pathogenic mechanism in Alzheimer’s disease and scrapie? Cell 73, 1055–1058 (1993).

    CAS  PubMed  Google Scholar 

  14. Meisl, G. et al. Scaling behaviour and rate-determining steps in filamentous self-assembly. Chem. Sci. 8, 7087–7097 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Soto, C., Estrada, L. & Castilla, J. Amyloids, prions and the inherent infectious nature of misfolded protein aggregates. Trends Biochem. Sci. 31, 150–155 (2006).

    CAS  PubMed  Google Scholar 

  16. Soto, C. Transmissible proteins: expanding the prion heresy. Cell 149, 968–977 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Aguzzi, A. & Lakkaraju, A. K. Cell biology of prions and prionoids: a status report. Trends Cell Biol. 26, 40–51 (2016).

    CAS  PubMed  Google Scholar 

  18. Prusiner, S. B. Prions. Proc. Natl. Acad. Sci. USA 95, 13363–13383 (1998).

  19. Soto, C. Prion hypothesis: the end of the controversy? Trends Biochem. Sci. 36, 151–158 (2011).

    CAS  PubMed  Google Scholar 

  20. Ganowiak, K., Hultman, P., Engström, U., Gustavsson, A. & Westermark, P. Fibrils from synthetic amyloid-related peptides enhance development of experimental AA-amyloidosis in mice. Biochem. Biophys. Res. Commun. 199, 306–312 (1994). This manuscript describes one of the earliest demonstrations of prion-like transmission of a nonprion protein misfolding disease.

    CAS  PubMed  Google Scholar 

  21. Xing, Y. et al. Transmission of mouse senile amyloidosis. Lab. Invest. 81, 493–499 (2001).

    CAS  PubMed  Google Scholar 

  22. Walker, L. C. & Jucker, M. Neurodegenerative diseases: expanding the prion concept. Annu. Rev. Neurosci. 38, 87–103 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Stopschinski, B. E. & Diamond, M. I. The prion model for progression and diversity of neurodegenerative diseases. Lancet Neurol. 16, 323–332 (2017).

    CAS  PubMed  Google Scholar 

  24. Morales, R., Duran-Aniotz, C., Castilla, J., Estrada, L. D. & Soto, C. De novo induction of amyloid-β deposition in vivo. Mol. Psychiatry 17, 1347–1353 (2012).

    CAS  PubMed  Google Scholar 

  25. Luk, K. C. et al. Pathological α-synuclein transmission initiates Parkinson-like neurodegeneration in nontransgenic mice. Science 338, 949–953 (2012). This study reports the transmission of disease to wild-type, nontransgenic mice by a single intracerebral inoculation of synthetic α-Syn aggregates, leading to neurodegeneration and motor deficits.

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Guo, J. L. et al. Unique pathological tau conformers from Alzheimer’s brains transmit tau pathology in nontransgenic mice. J. Exp. Med. 213, 2635–2654 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Meyer-Luehmann, M. et al. Exogenous induction of cerebral beta-amyloidogenesis is governed by agent and host. Science 313, 1781–1784 (2006).

    CAS  PubMed  Google Scholar 

  28. Duran-Aniotz, C. et al. Aggregate-depleted brain fails to induce Aβ deposition in a mouse model of Alzheimer’s disease. PLoS One 9, e89014 (2014).

    PubMed  PubMed Central  Google Scholar 

  29. Tran, H. T. et al. Alpha-synuclein immunotherapy blocks uptake and templated propagation of misfolded α-synuclein and neurodegeneration. Cell Rep. 7, 2054–2065 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Stöhr, J. et al. Purified and synthetic Alzheimer's amyloid beta (Aβ) prions. Proc. Natl. Acad. Sci. USA 109, 11025–11030 (2012).

    PubMed  PubMed Central  Google Scholar 

  31. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Iba, M. et al. Synthetic tau fibrils mediate transmission of neurofibrillary tangles in a transgenic mouse model of Alzheimer’s-like tauopathy. J. Neurosci. 33, 1024–1037 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Supattapone, S. Elucidating the role of cofactors in mammalian prion propagation. Prion 8, 100–105 (2014).

    CAS  PubMed  Google Scholar 

  35. Morales, R., Bravo-Alegria, J., Duran-Aniotz, C. & Soto, C. Titration of biologically active amyloid-β seeds in a transgenic mouse model of Alzheimer’s disease. Sci. Rep. 5, 9349 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Fritschi, S. K. et al. Highly potent soluble amyloid-β seeds in human Alzheimer brain but not cerebrospinal fluid. Brain 137, 2909–2915 (2014).

    PubMed  Google Scholar 

  37. Eisele, Y. S. et al. Peripherally applied Abeta-containing inoculates induce cerebral beta-amyloidosis. Science 330, 980–982 (2010). This manuscript reports prion-like induction of protein aggregation by administration of seeds via peripheral routes.

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Clavaguera, F. et al. Peripheral administration of tau aggregates triggers intracerebral tauopathy in transgenic mice. Acta Neuropathol. 127, 299–301 (2014).

    PubMed  Google Scholar 

  39. Walsh, D. M. & Selkoe, D. J. A critical appraisal of the pathogenic protein spread hypothesis of neurodegeneration. Nat. Rev. Neurosci. 17, 251–260 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Irwin, D. J. et al. Evaluation of potential infectivity of Alzheimer and Parkinson disease proteins in recipients of cadaver-derived human growth hormone. JAMA Neurol. 70, 462–468 (2013).

    PubMed  PubMed Central  Google Scholar 

  41. Piccardo, P., Manson, J. C., King, D., Ghetti, B. & Barron, R. M. Accumulation of prion protein in the brain that is not associated with transmissible disease. Proc. Natl. Acad. Sci. USA 104, 4712–4717 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Moreno-Gonzalez, I. & Soto, C. Misfolded protein aggregates: mechanisms, structures and potential for disease transmission. Semin. Cell Dev. Biol. 22, 482–487 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Goedert, M., Falcon, B., Clavaguera, F. & Tolnay, M. Prion-like mechanisms in the pathogenesis of tauopathies and synucleinopathies. Curr. Neurol. Neurosci. Rep. 14, 495 (2014).

    PubMed  Google Scholar 

  44. Hayden, E. Y. & Teplow, D. B. Amyloid β-protein oligomers and Alzheimer’s disease. Alzheimers Res. Ther. 5, 60 (2013).

    PubMed  PubMed Central  Google Scholar 

  45. Eisenberg, D. & Jucker, M. The amyloid state of proteins in human diseases. Cell 148, 1188–1203 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Aguzzi, A., Heikenwalder, M. & Polymenidou, M. Insights into prion strains and neurotoxicity. Nat. Rev. Mol. Cell Biol. 8, 552–561 (2007).

    CAS  PubMed  Google Scholar 

  47. Morales, R. Prion strains in mammals: Different conformations leading to disease. PLoS Pathog. 13, e1006323 (2017).

    PubMed  PubMed Central  Google Scholar 

  48. Poggiolini, I., Saverioni, D. & Parchi, P. Prion protein misfolding, strains, and neurotoxicity: an update from studies on Mammalian prions. Int. J. Cell Biol. 2013, 910314 (2013).

    PubMed  PubMed Central  Google Scholar 

  49. Castilla, J. et al. Cell-free propagation of prion strains. EMBO J. 27, 2557–2566 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Diaz-Espinoza, R. & Soto, C. High-resolution structure of infectious prion protein: the final frontier. Nat. Struct. Mol. Biol. 19, 370–377 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Heilbronner, G. et al. Seeded strain-like transmission of β-amyloid morphotypes in APP transgenic mice. EMBO Rep. 14, 1017–1022 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Petkova, A. T. et al. Self-propagating, molecular-level polymorphism in Alzheimer’s beta-amyloid fibrils. Science 307, 262–265 (2005). This study reports the generation of different polymorphic variants of amyloid-β aggregates and their detailed structural and biochemical characterizations.

    CAS  PubMed  Google Scholar 

  53. Qiang, W., Yau, W. M., Lu, J. X., Collinge, J. & Tycko, R. Structural variation in amyloid-β fibrils from Alzheimer’s disease clinical subtypes. Nature 541, 217–221 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Watts, J. C. et al. Serial propagation of distinct strains of Aβ prions from Alzheimer's disease patients. Proc. Natl. Acad. Sci. USA 111, 10323–10328 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Kaufman, S. K. et al. Tau prion strains dictate patterns of cell pathology, progression rate, and regional vulnerability in vivo. Neuron 92, 796–812 (2016). This article describes the isolation and characterization of 18 tau strains in cells. Inoculation of transgenic mice with these strains causes strain-specific intracellular pathology in distinct cell types and brain regions.

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Sanders, D. W. et al. Distinct tau prion strains propagate in cells and mice and define different tauopathies. Neuron 82, 1271–1288 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Narasimhan, S. et al. Pathological tau strains from human brains recapitulate the diversity of tauopathies in nontransgenic mouse brain. J. Neurosci. 37, 11406–11423 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Guo, J. L. et al. Distinct α-synuclein strains differentially promote tau inclusions in neurons. Cell 154, 103–117 (2013). This study shows that different conformational strains differ in their cross-seeding activity.

    CAS  PubMed  Google Scholar 

  59. Prusiner, S. B. et al. Evidence for α-synuclein prions causing multiple system atrophy in humans with parkinsonism. Proc. Natl Acad. Sci. USA 112, E5308–E5317 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Peelaerts, W. et al. α-Synuclein strains cause distinct synucleinopathies after local and systemic administration. Nature 522, 340–344 (2015).

    CAS  PubMed  Google Scholar 

  61. Bousset, L. et al. Structural and functional characterization of two alpha-synuclein strains. Nat. Commun. 4, 2575 (2013). This article reports a complete biochemical, biological, and structural characterization of α-Syn strains generated in vitro.

    PubMed  Google Scholar 

  62. Melki, R. Role of different alpha-synuclein strains in synucleinopathies, similarities with other neurodegenerative diseases. J. Parkinsons Dis. 5, 217–227 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Williams, D. R. Tauopathies: classification and clinical update on neurodegenerative diseases associated with microtubule-associated protein tau. Intern. Med. J. 36, 652–660 (2006).

    CAS  PubMed  Google Scholar 

  64. Goedert, M., Jakes, R. & Spillantini, M. G. The synucleinopathies: twenty years on. J. Parkinsons Dis. 7(s1), S53–S71 (2017).

    Google Scholar 

  65. Melki, R. How the shapes of seeds can influence pathology. Neurobiol. Dis. 109(Pt B), 201–208 (2018).

    PubMed  Google Scholar 

  66. Fitzpatrick, A. W. P. et al. Cryo-EM structures of tau filaments from Alzheimer’s disease. Nature 547, 185–190 (2017). This study describes atomic models for tau aggregates organized in different strains.

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Condello, C. & Stöehr, J. Aβ propagation and strains: Implications for the phenotypic diversity in Alzheimer’s disease. Neurobiol. Dis. 109(Pt B), 191–200 (2018).

    PubMed  Google Scholar 

  68. Fändrich, M., Meinhardt, J. & Grigorieff, N. Structural polymorphism of Alzheimer Abeta and other amyloid fibrils. Prion 3, 89–93 (2009).

    PubMed  PubMed Central  Google Scholar 

  69. Goldsbury, C., Frey, P., Olivieri, V., Aebi, U. & Müller, S. A. Multiple assembly pathways underlie amyloid-beta fibril polymorphisms. J. Mol. Biol. 352, 282–298 (2005).

    CAS  PubMed  Google Scholar 

  70. Elkins, M. R. et al. Structural polymorphism of Alzheimer’s β-amyloid fibrils as controlled by an E22 switch: a solid-state NMR study. J. Am. Chem. Soc. 138, 9840–9852 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Lubomski, M., Rushworth, R. L., Lee, W., Bertram, K. & Williams, D. R. A cross-sectional study of clinical management, and provision of health services and their utilisation, by patients with Parkinson’s disease in urban and regional Victoria. J. Clin. Neurosci. 20, 102–106 (2013).

    PubMed  Google Scholar 

  72. O’Nuallain, B., Williams, A. D., Westermark, P. & Wetzel, R. Seeding specificity in amyloid growth induced by heterologous fibrils. J. Biol. Chem. 279, 17490–17499 (2004).

    PubMed  Google Scholar 

  73. Yan, J. et al. Cross-seeding and cross-competition in mouse apolipoprotein A-II amyloid fibrils and protein A amyloid fibrils. Am. J. Pathol. 171, 172–180 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Krebs, M. R., Morozova-Roche, L. A., Daniel, K., Robinson, C. V. & Dobson, C. M. Observation of sequence specificity in the seeding of protein amyloid fibrils. Protein Sci. 13, 1933–1938 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Ono, K., Takahashi, R., Ikeda, T. & Yamada, M. Cross-seeding effects of amyloid β-protein and α-synuclein. J. Neurochem. 122, 883–890 (2012).

    CAS  PubMed  Google Scholar 

  76. Hu, R., Zhang, M., Chen, H., Jiang, B. & Zheng, J. Cross-seeding interaction between β-amyloid and human islet amyloid polypeptide. ACS Chem. Neurosci. 6, 1759–1768 (2015).

    CAS  PubMed  Google Scholar 

  77. Moreno-Gonzalez, I. et al. Molecular interaction between type 2 diabetes and Alzheimer’s disease through cross-seeding of protein misfolding. Mol. Psychiatry 22, 1327–1334 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Morales, R. et al. Molecular cross talk between misfolded proteins in animal models of Alzheimer’s and prion diseases. J. Neurosci. 30, 4528–4535 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Giasson, B. I., Lee, V. M. & Trojanowski, J. Q. Interactions of amyloidogenic proteins. Neuromolecular Med. 4, 49–58 (2003).

    CAS  PubMed  Google Scholar 

  80. Brown, D. F. et al. Neuropathologic evidence that the Lewy body variant of Alzheimer disease represents coexistence of Alzheimer disease and idiopathic Parkinson disease. J. Neuropathol. Exp. Neurol. 57, 39–46 (1998).

    CAS  PubMed  Google Scholar 

  81. Brown, P. et al. Coexistence of Creutzfeldt-Jakob disease and Alzheimer’s disease in the same patient. Neurology 40, 226–228 (1990).

    CAS  PubMed  Google Scholar 

  82. Spires-Jones, T. L., Attems, J. & Thal, D. R. Interactions of pathological proteins in neurodegenerative diseases. Acta Neuropathol. 134, 187–205 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Clinton, L. K., Blurton-Jones, M., Myczek, K., Trojanowski, J. Q. & LaFerla, F. M. Synergistic Interactions between Abeta, tau, and alpha-synuclein: acceleration of neuropathology and cognitive decline. J. Neurosci. 30, 7281–7289 (2010). This study shows that transgenic mice expressing four different mutant genes develop both Lewy bodies and AD pathologies, exhibiting accelerated cognitive decline associated with a dramatic enhancement of Aβ, tau, and α-Syn deposition.

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Morales, R., Moreno-Gonzalez, I. & Soto, C. Cross-seeding of misfolded proteins: implications for etiology and pathogenesis of protein misfolding diseases. PLoS Pathog. 9, e1003537 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Dickson, D. W. Neuropathology of Alzheimer’s disease and other dementias. Clin. Geriatr. Med. 17, 209–228 (2001).

    CAS  PubMed  Google Scholar 

  86. Guo, J. P., Arai, T., Miklossy, J. & McGeer, P. L. Abeta and tau form soluble complexes that may promote self aggregation of both into the insoluble forms observed in Alzheimer’s disease. Proc. Natl. Acad. Sci. USA 103, 1953–1958 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Lewis, J. et al. Enhanced neurofibrillary degeneration in transgenic mice expressing mutant tau and APP. Science 293, 1487–1491 (2001).

    CAS  PubMed  Google Scholar 

  88. Götz, J., Chen, F., van Dorpe, J. & Nitsch, R. M. Formation of neurofibrillary tangles in P301l tau transgenic mice induced by Abeta 42 fibrils. Science 293, 1491–1495 (2001). This study shows that intracerebral inoculation of Aβ aggregates in tau transgenic mice enhances the formation of neurofibrillary tangles, suggesting that amyloid induces tau pathology.

    PubMed  Google Scholar 

  89. Pooler, A. M. et al. Amyloid accelerates tau propagation and toxicity in a model of early Alzheimer’s disease. Acta Neuropathol. Commun. 3, 14 (2015).

    PubMed  PubMed Central  Google Scholar 

  90. He, Z. et al. Amyloid-β plaques enhance Alzheimer’s brain tau-seeded pathologies by facilitating neuritic plaque tau aggregation. Nat. Med. 24, 29–38 (2018).

    CAS  PubMed  Google Scholar 

  91. Uchikado, H., Lin, W. L., DeLucia, M. W. & Dickson, D. W. Alzheimer disease with amygdala Lewy bodies: a distinct form of alpha-synucleinopathy. J. Neuropathol. Exp. Neurol. 65, 685–697 (2006).

    CAS  PubMed  Google Scholar 

  92. Josephs, K. A. et al. Updated TDP-43 in Alzheimer’s disease staging scheme. Acta Neuropathol. 131, 571–585 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Pigott, K. et al. Longitudinal study of normal cognition in Parkinson disease. Neurology 85, 1276–1282 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Bosboom, J. L., Stoffers, D. & Wolters, E. Ch. Cognitive dysfunction and dementia in Parkinson’s disease. J. Neural Transm. (Vienna) 111, 1303–1315 (2004).

    CAS  Google Scholar 

  95. Irwin, D. J., Lee, V. M. & Trojanowski, J. Q. Parkinson’s disease dementia: convergence of α-synuclein, tau and amyloid-β pathologies. Nat. Rev. Neurosci. 14, 626–636 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Buongiorno, M., Compta, Y. & Martí, M. J. Amyloid-β and tau biomarkers in Parkinson’s disease-dementia. J. Neurol. Sci. 310, 25–30 (2011).

    CAS  PubMed  Google Scholar 

  97. Iseki, E. Dementia with Lewy bodies: reclassification of pathological subtypes and boundary with Parkinson’s disease or Alzheimer’s disease. Neuropathology 24, 72–78 (2004).

    PubMed  Google Scholar 

  98. McKeith, I. G. et al. Diagnosis and management of dementia with Lewy bodies: fourth consensus report of the DLB Consortium. Neurology 89, 88–100 (2017).

    PubMed  PubMed Central  Google Scholar 

  99. Saxena, S. & Caroni, P. Selective neuronal vulnerability in neurodegenerative diseases: from stressor thresholds to degeneration. Neuron 71, 35–48 (2011).

    CAS  PubMed  Google Scholar 

  100. Biessels, G. J., Staekenborg, S., Brunner, E., Brayne, C. & Scheltens, P. Risk of dementia in diabetes mellitus: a systematic review. Lancet Neurol. 5, 64–74 (2006).

    PubMed  Google Scholar 

  101. Sims-Robinson, C., Kim, B., Rosko, A. & Feldman, E. L. How does diabetes accelerate Alzheimer disease pathology? Nat. Rev. Neurol. 6, 551–559 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Janson, J. et al. Increased risk of type 2 diabetes in Alzheimer disease. Diabetes 53, 474–481 (2004).

    CAS  PubMed  Google Scholar 

  103. Oskarsson, M. E. et al. In vivo seeding and cross-seeding of localized amyloidosis: a molecular link between type 2 diabetes and Alzheimer disease. Am. J. Pathol. 185, 834–846 (2015). This article describes the in vivo cross-seeding between Aβ and islet amyloid polypeptide, providing a possible molecular explanation for the link between T2D and AD.

    CAS  PubMed  Google Scholar 

  104. Friedland, R. P. & Chapman, M. R. The role of microbial amyloid in neurodegeneration. PLoS Pathog. 13, e1006654 (2017).

    PubMed  PubMed Central  Google Scholar 

  105. Pham, C. L., Kwan, A. H. & Sunde, M. Functional amyloid: widespread in Nature, diverse in purpose. Essays Biochem. 56, 207–219 (2014).

    PubMed  Google Scholar 

  106. Hammer, N. D., Wang, X., McGuffie, B. A. & Chapman, M. R. Amyloids: friend or foe? J. Alzheimers Dis. 13, 407–419 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Fowler, D. M., Koulov, A. V., Balch, W. E. & Kelly, J. W. Functional amyloid–from bacteria to humans. Trends Biochem. Sci. 32, 217–224 (2007).

    CAS  PubMed  Google Scholar 

  108. Chen, S. G. et al. Exposure to the functional bacterial amyloid protein Curli enhances alpha-synuclein aggregation in aged Fischer 344 rats and Caenorhabditis elegans. Sci. Rep. 6, 34477 (2016). This article describes the possibility that a functional bacterial amyloid may induce the aggregation of α-Syn in vivo.

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Fernández-Borges, N. et al. Infectivity versus seeding in neurodegenerative diseases sharing a prion-like mechanism. Int. J. Cell Biol. 2013, 583498 (2013).

    PubMed  PubMed Central  Google Scholar 

  110. Garske, T. & Ghani, A. C. Uncertainty in the tail of the variant Creutzfeldt-Jakob disease epidemic in the UK. PLoS One 5, e15626 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Valera, E., Spencer, B. & Masliah, E. Immunotherapeutic approaches targeting amyloid-β, α-synuclein, and tau for the treatment of neurodegenerative disorders. Neurotherapeutics 13, 179–189 (2016).

    CAS  PubMed  Google Scholar 

  112. Mohamed, N. V., Herrou, T., Plouffe, V., Piperno, N. & Leclerc, N. Spreading of tau pathology in Alzheimer’s disease by cell-to-cell transmission. Eur. J. Neurosci. 37, 1939–1948 (2013).

    PubMed  Google Scholar 

  113. Costanzo, M. & Zurzolo, C. The cell biology of prion-like spread of protein aggregates: mechanisms and implication in neurodegeneration. Biochem. J. 452, 1–17 (2013).

    CAS  PubMed  Google Scholar 

  114. Danzer, K. M. et al. Exosomal cell-to-cell transmission of alpha synuclein oligomers. Mol. Neurodegener. 7, 42 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  115. Wegrzyn, R. D., Bapat, K., Newnam, G. P., Zink, A. D. & Chernoff, Y. O. Mechanism of prion loss after Hsp104 inactivation in yeast. Mol. Cell. Biol. 21, 4656–4669 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Diaz-Espinoza, R. et al. Treatment with a non-toxic, self-replicating anti-prion delays or prevents prion disease in vivo. Mol. Psychiatry 23, 777–788 (2018). This article reports the use of the prion principle to generate a self-replication protein therapy for prion diseases.

    CAS  PubMed  Google Scholar 

  117. Li, J., Mahal, S. P., Demczyk, C. A. & Weissmann, C. Mutability of prions. EMBO Rep. 12, 1243–1250 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Telling, G. C. Nucleic acid-free mutation of prion strains. Prion 4, 252–255 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Li, J., Browning, S., Mahal, S. P., Oelschlegel, A. M. & Weissmann, C. Darwinian evolution of prions in cell culture. Science 327, 869–872 (2010). This study shows that prion strains can mutate and selectively adapt to grow under different conditions and likely constitute an ensemble of substrains.

    CAS  PubMed  Google Scholar 

  120. Oelschlegel, A. M. & Weissmann, C. Acquisition of drug resistance and dependence by prions. PLoS Pathog. 9, e1003158 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Ghaemmaghami, S. et al. Continuous quinacrine treatment results in the formation of drug-resistant prions. PLoS Pathog. 5, e1000673 (2009).

    PubMed  PubMed Central  Google Scholar 

  122. Anderson, R. M., Hadjichrysanthou, C., Evans, S. & Wong, M. M. Why do so many clinical trials of therapies for Alzheimer’s disease fail? Lancet 390, 2327–2329 (2017).

    PubMed  Google Scholar 

  123. Gómez-Río, M., Caballero, M. M., Górriz Sáez, J. M. & Mínguez-Castellanos, A. Diagnosis of neurodegenerative diseases: the clinical approach. Curr. Alzheimer Res. 13, 469–474 (2016).

    PubMed  Google Scholar 

  124. Lewczuk, P. et al. Cerebrospinal fluid and blood biomarkers for neurodegenerative dementias: an update of the consensus of the Task Force on Biological Markers in Psychiatry of the World Federation of Societies of Biological Psychiatry. World J. Biol. Psychiatry 19, 244–328 (2018).

    PubMed  Google Scholar 

  125. Schuster, J. & Funke, S. A. Methods for the specific detection and quantitation of amyloid-β oligomers in cerebrospinal fluid. J. Alzheimers Dis. 53, 53–67 (2016).

    CAS  PubMed  Google Scholar 

  126. Bateman, R. J. et al. Clinical and biomarker changes in dominantly inherited Alzheimer’s disease. N. Engl. J. Med. 367, 795–804 (2012). This article describes studies in human AD patients to model the sequence of pathological changes over decades in cerebrospinal fluid biochemical markers, brain amyloid deposition, and brain metabolism, as well as progressive cognitive impairment.

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Wegmann, S., Nicholls, S., Takeda, S., Fan, Z. & Hyman, B. T. Formation, release, and internalization of stable tau oligomers in cells. J. Neurochem. 139, 1163–1174 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  128. Chai, Y. J. et al. The secreted oligomeric form of α-synuclein affects multiple steps of membrane trafficking. FEBS Lett. 587, 452–459 (2013).

    CAS  PubMed  Google Scholar 

  129. Murakami, K. et al. Monoclonal antibody with conformational specificity for a toxic conformer of amyloid β42 and its application toward the Alzheimer’s disease diagnosis. Sci. Rep. 6, 29038 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Kamali-Moghaddam, M. et al. Sensitive detection of Aβ protofibrils by proximity ligation–relevance for Alzheimer’s disease. BMC Neurosci. 11, 124 (2010).

    PubMed  PubMed Central  Google Scholar 

  131. Hölttä, M. et al. Evaluating amyloid-β oligomers in cerebrospinal fluid as a biomarker for Alzheimer’s disease. PLoS One 8, e66381 (2013).

    PubMed  PubMed Central  Google Scholar 

  132. Pitschke, M., Prior, R., Haupt, M. & Riesner, D. Detection of single amyloid beta-protein aggregates in the cerebrospinal fluid of Alzheimer’s patients by fluorescence correlation spectroscopy. Nat. Med. 4, 832–834 (1998).

    CAS  PubMed  Google Scholar 

  133. Santos, A. N. et al. Detection of amyloid-beta oligomers in human cerebrospinal fluid by flow cytometry and fluorescence resonance energy transfer. J. Alzheimers Dis. 11, 117–125 (2007).

    CAS  PubMed  Google Scholar 

  134. Funke, S. A., Wang, L., Birkmann, E. & Willbold, D. Single-particle detection system for Abeta aggregates: adaptation of surface-fluorescence intensity distribution analysis to laser scanning microscopy. Rejuvenation Res. 13, 206–209 (2010).

    CAS  PubMed  Google Scholar 

  135. Haes, A. J., Chang, L., Klein, W. L. & Van Duyne, R. P. Detection of a biomarker for Alzheimer’s disease from synthetic and clinical samples using a nanoscale optical biosensor. J. Am. Chem. Soc. 127, 2264–2271 (2005).

    CAS  PubMed  Google Scholar 

  136. Sierks, M. R. et al. CSF levels of oligomeric alpha-synuclein and beta-amyloid as biomarkers for neurodegenerative disease. Integr. Biol. (Camb.) 3, 1188–1196 (2011).

    CAS  Google Scholar 

  137. Saborio, G. P., Permanne, B. & Soto, C. Sensitive detection of pathological prion protein by cyclic amplification of protein misfolding. Nature 411, 810–813 (2001). This article reports the use of the prion principle to develop a PCR-like methodology for highly sensitive detection of prions that can also be used for other misfolded aggregates.

    CAS  PubMed  Google Scholar 

  138. Soto, C., Saborio, G. P. & Anderes, L. Cyclic amplification of protein misfolding: application to prion-related disorders and beyond. Trends Neurosci. 25, 390–394 (2002).

    CAS  PubMed  Google Scholar 

  139. Orrù, C. D., Wilham, J. M., Vascellari, S., Hughson, A. G. & Caughey, B. New generation QuIC assays for prion seeding activity. Prion 6, 147–152 (2012).

    PubMed  PubMed Central  Google Scholar 

  140. Atarashi, R. et al. Simplified ultrasensitive prion detection by recombinant PrP conversion with shaking. Nat. Methods 5, 211–212 (2008).

    CAS  PubMed  Google Scholar 

  141. Saá, P., Castilla, J. & Soto, C. Ultra-efficient replication of infectious prions by automated protein misfolding cyclic amplification. J. Biol. Chem. 281, 35245–35252 (2006).

    PubMed  Google Scholar 

  142. Moda, F. et al. Prions in the urine of patients with variant Creutzfeldt-Jakob disease. N. Engl. J. Med. 371, 530–539 (2014).

    PubMed  PubMed Central  Google Scholar 

  143. Concha-Marambio, L. et al. Detection of prions in blood from patients with variant Creutzfeldt-Jakob disease. Sci. Transl. Med. https://doi.org/10.1126/scitranslmed.aaf6188 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  144. Bougard, D. et al. Detection of prions in the plasma of presymptomatic and symptomatic patients with variant Creutzfeldt-Jakob disease. Sci. Transl. Med. 8, 370ra182 (2016).

    PubMed  Google Scholar 

  145. Salvadores, N., Shahnawaz, M., Scarpini, E., Tagliavini, F. & Soto, C. Detection of misfolded Aβ oligomers for sensitive biochemical diagnosis of Alzheimer’s disease. Cell Rep. 7, 261–268 (2014).

    CAS  PubMed  Google Scholar 

  146. Saijo, E. et al. Ultrasensitive and selective detection of 3-repeat tau seeding activity in Pick disease brain and cerebrospinal fluid. Acta Neuropathol. 133, 751–765 (2017).

    CAS  PubMed  Google Scholar 

  147. Shahnawaz, M. et al. Development of a biochemical diagnosis of Parkinson disease by detection of α-synuclein misfolded aggregates in cerebrospinal fluid. JAMA Neurol. 74, 163–172 (2017).

    PubMed  Google Scholar 

  148. Fairfoul, G. et al. Alpha-synuclein RT-QuIC in the CSF of patients with alpha-synucleinopathies. Ann. Clin. Transl. Neurol. 3, 812–818 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank C. Mays for critical reading of the manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Claudio Soto.

Ethics declarations

Competing interests

C.S. is the inventor of the PMCA technology and is currently the Founder, Chief Scientific Officer, and major shareholder of Amprion Inc., a biotech company aiming to develop PMCA and RT-QuIC seeding amplification assays for diagnosis of neurodegenerative diseases.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Soto, C., Pritzkow, S. Protein misfolding, aggregation, and conformational strains in neurodegenerative diseases. Nat Neurosci 21, 1332–1340 (2018). https://doi.org/10.1038/s41593-018-0235-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-018-0235-9

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