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:

Quantifying misfolded protein oligomers as drug targets and biomarkers in Alzheimer and Parkinson diseases

Abstract

Protein misfolding and aggregation are characteristic of a wide range of neurodegenerative disorders, including Alzheimer and Parkinson diseases. A hallmark of these diseases is the aggregation of otherwise soluble and functional proteins into amyloid aggregates. Although for many decades such amyloid deposits have been thought to be responsible for disease progression, it is now increasingly recognized that the misfolded protein oligomers formed during aggregation are, instead, the main agents causing pathological processes. These oligomers are transient and heterogeneous, which makes it difficult to detect and quantify them, generating confusion about their exact role in disease. The lack of suitable methods to address these challenges has hampered efforts to investigate the molecular mechanisms of oligomer toxicity and to develop oligomer-based diagnostic and therapeutic tools to combat protein misfolding diseases. In this Review, we describe methods to quantify misfolded protein oligomers, with particular emphasis on diagnostic applications as disease biomarkers and on therapeutic applications as target biomarkers. The development of these methods is ongoing, and we discuss the challenges that remain to be addressed to establish measurement tools capable of overcoming existing limitations and to meet present needs.

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

Access options

Buy this article

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

Fig. 1: Protein oligomers are diverse and present at different concentrations and different locations in healthy and diseased individuals.
Fig. 2: Immunoassays to detect and quantify misfolded protein oligomers.
Fig. 3: Fluorescence microscopy methods to detect and quantify misfolded protein oligomers.
Fig. 4: Biosensors detect and quantify misfolded protein oligomers.
Fig. 5: Other methods to detect and quantify misfolded protein oligomers.

Similar content being viewed by others

References

  1. Alzheimer’s Disease International. World Alzheimer report 2019: attitudes to dementia. Alzheimer’s Disease International https://www.alzint.org/resource/world-alzheimer-report-2019/ (2019).

  2. Cummings, J., Lee, G., Ritter, A., Sabbagh, M. & Zhong, K. Alzheimer’s disease drug development pipeline: 2019. Alzheimers Dement. 5, 272–293 (2019).

    Google Scholar 

  3. Cummings, J. L. Translational scoring of candidate treatments for Alzheimer’s disease: a systematic approach. Dement. Geriatr. Cogn. Disord. 49, 22–37 (2020).

    CAS  PubMed  Google Scholar 

  4. Hampel, H. et al. Biomarkers for Alzheimer’s disease: academic, industry and regulatory perspectives. Nat. Rev. Drug Discov. 9, 560–574 (2010).

    CAS  PubMed  Google Scholar 

  5. Aisen, P. S. et al. The future of anti-amyloid trials. J. Prev. Alzheimers Dis. 7, 146–151 (2020).

    CAS  PubMed  Google Scholar 

  6. U.S. Department of Health and Human Services, Food and Drug Administration, Center for Drug Evaluation and Research (CDER) and Center for Biologics Evaluation and Research (CBER). Early Alzheimer’s disease: developing drugs for treatment (2018).

  7. Jack, C. R. et al. NIA-AA research framework: toward a biological definition of Alzheimer’s disease. Alzheimers Dement. 14, 535–562 (2018).

    PubMed  PubMed Central  Google Scholar 

  8. Hardy, J. & Selkoe, D. J. The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science 297, 353–356 (2002).

    CAS  PubMed  Google Scholar 

  9. Selkoe, D. J. & Hardy, J. The amyloid hypothesis of Alzheimer’s disease at 25 years. EMBO Mol. Med. 8, 595–608 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Spillantini, M. G. et al. α-Synuclein in Lewy bodies. Nature 338, 839–840 (1997).

    Google Scholar 

  11. Braak, H. & Braak, E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol. 82, 239–259 (1991).

    CAS  PubMed  Google Scholar 

  12. Prots, I. et al. α-Synuclein oligomers induce early axonal dysfunction in human iPSC-based models of synucleinopathies. Proc. Natl Acad. Sci. USA 115, 7813–7818 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Haass, C. & Selkoe, D. J. Soluble protein oligomers in neurodegeneration: lessons from the Alzheimer’s amyloid β-peptide. Nat. Rev. Mol. Cell Biol. 8, 101–112 (2007).

    CAS  PubMed  Google Scholar 

  14. 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 

  15. Flagmeier, P. et al. Ultrasensitive measurement of Ca2+ influx into lipid vesicles induced by protein aggregates. Angew. Chem. Int. Ed. 56, 7750–7754 (2017). This article reports a high-throughput oligomer quantification assay based on the measurement of Ca2+ entry into individual lipid vesicles.

    CAS  Google Scholar 

  16. Fusco, G. et al. Structural basis of membrane disruption and cellular toxicity by α-synuclein oligomers. Science 358, 1440–1443 (2017).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Tucker, S. et al. The murine version of BAN2401 (mAb158) selectively reduces amyloid-β protofibrils in brain and cerebrospinal fluid of tg-ArcSwe mice. J. Alzheimers Dis. 43, 575–588 (2015). This work describes accurate enzyme-linked immunosorbent assay (ELISA) measurements of amyloid-β protofibrils in mouse brain and cerebrospinal fluid.

    CAS  PubMed  Google Scholar 

  19. De, S. et al. Soluble aggregates present in cerebrospinal fluid change in size and mechanism of toxicity during Alzheimer’s disease progression. Acta Neuropathol. Commun. 7, 120 (2019).

    PubMed  PubMed Central  Google Scholar 

  20. Hong, W. et al. Diffusible, highly bioactive oligomers represent a critical minority of soluble Aβ in Alzheimer’s disease brain. Acta Neuropathol. 136, 19–40 (2018). This article describes characterization of the highly heterogeneous nature of misfolded protein oligomers.

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Walsh, D. M. & Selkoe, D. J. Amyloid β-protein and beyond: the path forward in Alzheimer’s disease. Curr. Opin. Neurobiol. 61, 116–124 (2020).

    CAS  PubMed  Google Scholar 

  22. Majbour, N. K. et al. Oligomeric and phosphorylated alpha-synuclein as potential CSF biomarkers for Parkinson’s disease. Mol. Neurodegener. 11, 7 (2016).

    PubMed  PubMed Central  Google Scholar 

  23. Saijo, E. et al. 4-Repeat tau seeds and templating subtypes as brain and CSF biomarkers of frontotemporal lobar degeneration. Acta Neuropathol. 139, 63–77 (2020).

    CAS  PubMed  Google Scholar 

  24. Hölttä, M. et al. Evaluating amyloid-β oligomers in cerebrospinal fluid as a biomarker for Alzheimer’s disease. PLoS ONE 8, e66381 (2013). One of the first reports on the use of amyloid-β oligomers as diagnostic markers for Alzheimer disease.

    PubMed  PubMed Central  Google Scholar 

  25. Wang-Dietrich, L. et al. The amyloid-β oligomer count in cerebrospinal fluid is a biomarker for Alzheimer’s disease. J. Alzheimers Dis. 34, 985–994 (2013).

    CAS  PubMed  Google Scholar 

  26. Yang, T. et al. Target engagement in an alzheimer trial: crenezumab lowers amyloid β oligomers in cerebrospinal fluid. Ann. Neurol. 86, 215–224 (2019). This paper demonstrates the use of an Erenna immunoassay to show target engagement of an antibody to amyloid-β aggregation in clinical trials.

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Yang, T. et al. A highly sensitive novel immunoassay specifically detects low levels of soluble Aβ oligomers in human cerebrospinal fluid. Alzheimers Res. Ther. 7, 14 (2015).

    PubMed  PubMed Central  Google Scholar 

  28. Georganopoulou, D. G. et al. Nanoparticle-based detection in cerebral spinal fluid of a soluble pathogenic biomarker for Alzheimer’s disease. Proc. Natl Acad. Sci. USA 102, 2273–2276 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Savage, M. J. et al. A sensitive Aβ oligomer assay discriminates Alzheimer’s and aged control cerebrospinal fluid. J. Neurosci. 34, 2884–2897 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Dujardin, S. et al. Tau molecular diversity contributes to clinical heterogeneity in Alzheimer’s disease. Nat. Med. 26, 1256–1263 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Wang, Z. et al. Human brain-derived Aβ oligomers bind to synapses and disrupt synaptic activity in a manner that requires APP. J. Neurosci. 37, 11947–11966 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Walsh, D. M. et al. Naturally secreted oligomers of amyloid β protein potently inhibit hippocampal long-term potentiation in vivo. Nature 416, 535–539 (2002).

    CAS  PubMed  Google Scholar 

  33. Wang, H.-W. et al. Soluble oligomers of β amyloid (1-42) inhibit long-term potentiation but not long-term depression in rat dentate gyrus. Brain Res. 924, 133–140 (2002).

    CAS  PubMed  Google Scholar 

  34. Li, S. & Selkoe, D. J. A mechanistic hypothesis for the impairment of synaptic plasticity by soluble Aβ oligomers from Alzheimer’s brain. J. Neurochem. 154, 583–597 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Lesné, S. E. et al. Brain amyloid-β oligomers in ageing and Alzheimer’s disease. Brain 136, 1383–1398 (2013).

    PubMed  PubMed Central  Google Scholar 

  36. Brinkmalm, G. et al. Identification of neurotoxic cross-linked amyloid-β dimers in the Alzheimer’s brain. Brain 142, 1441–1457 (2019).

    PubMed  PubMed Central  Google Scholar 

  37. Wang, Y. & Mandelkow, E. Tau in physiology and pathology. Nat. Rev. Neurosci. 17, 22–35 (2015).

    CAS  Google Scholar 

  38. Kopeikina, K. J., Hyman, B. J. & Spires-Jones, T. L. Soluble forms of tau are toxic in Alzheimer’s disease. Transl Neurosci. 3, 223–233 (2012).

    PubMed  Google Scholar 

  39. Kaniyappan, S., Chandupatla, R. R., Mandelkow, E. M. & Mandelkow, E. Extracellular low-n oligomers of tau cause selective synaptotoxicity without affecting cell viability. Alzheimers Dement. 13, 1270–1291 (2017).

    PubMed  Google Scholar 

  40. Hill, E., Karikari, T. K., Moffat, K. G., Richardson, M. J. E. & Wall, M. J. Introduction of tau oligomers into cortical neurons alters action potential dynamics and disrupts synaptic transmission and plasticity. eNeuro 6, 5 (2019).

    Google Scholar 

  41. Fá, M. et al. Extracellular tau oligomers produce an immediate impairment of LTP and memory. Sci. Rep. 6, 19393 (2016).

    PubMed  PubMed Central  Google Scholar 

  42. Lasagna-Reeves, C. A. et al. Alzheimer brain-derived tau oligomers propagate pathology from endogenous tau. Sci. Rep. 2, 700 (2012).

    PubMed  PubMed Central  Google Scholar 

  43. Wu, J. W. et al. Small misfolded tau species are internalized via bulk endocytosis and anterogradely and retrogradely transported in neurons. J. Biol. Chem. 288, 1856–1870 (2013).

    CAS  PubMed  Google Scholar 

  44. Das, R., Balmik, A. A. & Chinnathambi, S. Phagocytosis of full-length tau oligomers by actin-remodeling of activated microglia. J. Neuroinflammation 17, 10 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Clavaguera, F. et al. Transmission and spreading of tauopathy in transgenic mouse brain. Nat. Cell Biol. 11, 909–913 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Jiang, L. et al. TIA1 regulates the generation and response to toxic tau oligomers. Acta Neuropathol. 137, 259–277 (2019).

    CAS  PubMed  Google Scholar 

  47. Ingelsson, M. Alpha-synuclein oligomers — neurotoxic molecules in Parkinson’s disease and other Lewy body disorders. Front. Neurosci. 10, 408 (2016).

    PubMed  PubMed Central  Google Scholar 

  48. Bengoa-Vergniory, N., Roberts, R. F., Wade-Martins, R. & Alegre-Abarrategui, J. Alpha-synuclein oligomers: a new hope. Acta Neuropathol. 134, 819–838 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Alam, P., Bousset, L., Melki, R. & Otzen, D. E. α-Synuclein oligomers and fibrils: a spectrum of species, a spectrum of toxicities. J. Neurochem. 150, 522–534 (2019).

    CAS  PubMed  Google Scholar 

  50. Kayed, R., Dettmer, U. & Lesné, S. E. Soluble endogenous oligomeric α-synuclein species in neurodegenerative diseases: expression, spreading, and cross-talk. J. Parkinsons Dis. 10, 791–818 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  52. van Diggelen, F. et al. Two conformationally distinct α-synuclein oligomers share common epitopes and the ability to impair long-term potentiation. PLoS ONE 14, e0213663 (2019).

    PubMed  PubMed Central  Google Scholar 

  53. Di Maio, R. et al. α-Synuclein binds to TOM20 and inhibits mitochondrial protein import in Parkinson’s disease. Sci. Transl Med. 8, 342ra78 (2016).

    PubMed  PubMed Central  Google Scholar 

  54. Roberts, R. F., Wade-Martins, R. & Alegre-Abarrategui, J. Direct visualization of alpha-synuclein oligomers reveals previously undetected pathology in Parkinson’s disease brain. Brain 138, 1642–1657 (2015).

    PubMed  PubMed Central  Google Scholar 

  55. 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 

  56. Knowles, T. P. J. et al. An analytical solution to the kinetics of breakable filament assembly. Science 326, 1533–1537 (2009).

    CAS  PubMed  Google Scholar 

  57. Cohen, S. I. A. et al. Proliferation of amyloid-42 aggregates occurs through a secondary nucleation mechanism. Proc. Natl Acad. Sci. USA 110, 9758–9763 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Habchi, J. et al. Cholesterol catalyses Aβ42 aggregation through a heterogeneous nucleation pathway in the presence of lipid membranes. Nat. Chem. 10, 673–683 (2018).

    CAS  PubMed  Google Scholar 

  59. Michaels, T. C. T. et al. Dynamics of oligomer populations formed during the aggregation of Alzheimer’s Aβ42 peptide. Nat. Chem. 12, 445–451 (2020). This work shows that Aβ42 oligomers should undergo a structural conversion step after their initial formation in order to grow into amyloid fibrils.

    CAS  PubMed  Google Scholar 

  60. Koffie, R. M. et al. Oligomeric amyloid β associates with postsynaptic densities and correlates with excitatory synapse loss near senile plaques. Proc. Natl Acad. Sci. USA 106, 4012–4017 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Galvagnion, C. et al. Lipid vesicles trigger α-synuclein aggregation by stimulating primary nucleation. Nat. Chem. Biol. 11, 229–234 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Buell, A. K. et al. Solution conditions determine the relative importance of nucleation and growth processes in α-synuclein aggregation. Proc. Natl Acad. Sci. USA 111, 7671–7676 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Shammas, S. L. et al. A mechanistic model of tau amyloid aggregation based on direct observation of oligomers. Nat. Commun. 6, 7025 (2015).

    CAS  PubMed  Google Scholar 

  64. Dear, A. J. et al. Kinetic diversity of amyloid oligomers. Proc. Natl Acad. Sci. USA 117, 12087–12094 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Lambert, M. P. et al. Diffusible, nonfibrillar ligands derived from Aβ1–42 are potent central nervous system neurotoxins. Proc. Natl Acad. Sci. USA 95, 6448–6453 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Jackson, S. J. et al. Short fibrils constitute the major species of seed-competent tau in the brains of mice transgenic for human p301s tau. J. Neurosci. 36, 762–772 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Knowles, T. P. J., Vendruscolo, M. & Dobson, C. M. The amyloid state and its association with protein misfolding diseases. Nat. Rev. Mol. Cell Biol. 15, 384–396 (2014).

    CAS  PubMed  Google Scholar 

  68. Kayed, R. et al. Common structure of soluble amyloid oligomers implies common mechanism of pathogenesis. Science 300, 486–489 (2003).

    CAS  PubMed  Google Scholar 

  69. Lassen, L. B. et al. ELISA method to detect α-synuclein oligomers in cell and animal models. PLoS ONE 13, e0196056 (2018).

    PubMed  PubMed Central  Google Scholar 

  70. Wang, M. J. et al. Oligomeric forms of amyloid-β protein in plasma as a potential blood-based biomarker for Alzheimer’s disease. Alzheimers Res. Ther. 9, 98 (2017).

    PubMed  PubMed Central  Google Scholar 

  71. Youn, Y. C. et al. Blood amyloid-β oligomerization associated with neurodegeneration of Alzheimer’s disease. Alzheimers Res. Ther. 11, 40 (2019).

    PubMed  PubMed Central  Google Scholar 

  72. El-Agnaf, O. M. A. et al. Detection of oligomeric forms of α-synuclein protein in human plasma as a potential biomarker for Parkinson’s disease. FASEB J. 20, 419–425 (2006).

    CAS  PubMed  Google Scholar 

  73. Bruggink, K. A. et al. Amyloid-β oligomer detection by ELISA in cerebrospinal fluid and brain tissue. Anal. Biochem. 433, 112–120 (2013).

    CAS  PubMed  Google Scholar 

  74. Pryor, N. E., Moss, M. A. & Hestekin, C. N. Unraveling the early events of amyloid-β protein (Aβ) aggregation: techniques for the determination of Aβ aggregate size. Int. J. Mol. Sci. 13, 3038–3072 (2012).

    PubMed  PubMed Central  Google Scholar 

  75. 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).

    CAS  PubMed  Google Scholar 

  76. Sengupta, U. et al. Tau oligomers in cerebrospinal fluid in Alzheimer’s disease. Ann. Clin. Transl Neurol. 4, 226–235 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Kuhle, J. et al. Comparison of three analytical platforms for quantification of the neurofilament light chain in blood samples: ELISA, electrochemiluminescence immunoassay and Simoa. Clin. Chem. Lab. Med. 54, 1655–1661 (2016).

    CAS  PubMed  Google Scholar 

  78. Rissin, D. M. & Walt, D. R. Digital readout of target binding with attomole detection limits via enzyme amplification in femtoliter arrays. J. Am. Chem. Soc. 128, 6286–6287 (2006).

    CAS  PubMed  Google Scholar 

  79. Rissin, D. M. & Walt, D. R. Digital concentration readout of single enzyme molecules using femtoliter arrays and Poisson statistics. Nano Lett. 6, 520–523 (2006).

    CAS  PubMed  Google Scholar 

  80. Hwang, S. S. et al. Detection of amyloid β oligomers toward early diagnosis of Alzheimer’s disease. Anal. Biochem. 566, 40–45 (2019). This work compares the quartz crystal microbalance (QCM) and single-molecule array (SIMOA) assay for the quantification of misfolded protein oligomers.

    CAS  PubMed  Google Scholar 

  81. Li, D. & Mielke, M. M. An update on blood-based markers of Alzheimer’s disease using the SiMoA platform. Neurol. Ther. 8, 73–82 (2019).

    PubMed  PubMed Central  Google Scholar 

  82. Wu, C., Garden, P. M. & Walt, D. R. Ultrasensitive detection of attomolar protein concentrations by dropcast single molecule assays. J. Am. Chem. Soc. 142, 12314–12323 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Fulton, R. J., McDade, R. L., Smith, P. L., Kienker, L. J. & Kettman, J. R. Advanced multiplexed analysis with the FlowMetrixTM system. Clin. Chem. 43, 1749–1756 (1997).

    CAS  PubMed  Google Scholar 

  84. Herskovits, A. Z., Locascio, J. J., Peskind, E. R., Li, G. & Hyman, B. T. A Luminex assay detects amyloid β oligomers in Alzheimer’s disease cerebrospinal fluid. PLoS ONE 8, e67898 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Fukumoto, H. et al. High-molecular-weight β-amyloid oligomers are elevated in cerebrospinal fluid of Alzheimer patients. FASEB J. 24, 2716–2726 (2010).

    CAS  PubMed  Google Scholar 

  86. Esparza, T. J. et al. Amyloid-beta oligomerization in Alzheimer dementia versus high-pathology controls. Ann. Neurol. 73, 104–119 (2013).

    CAS  PubMed  Google Scholar 

  87. Park, M. C. et al. Droplet-based magnetic bead immunoassay using microchannel-connected multiwell plates (μCHAMPs) for the detection of amyloid beta oligomers. Lab Chip 16, 2245–2253 (2016).

    CAS  PubMed  Google Scholar 

  88. Takahashi, T. & Mihara, H. FRET detection of amyloid β-peptide oligomerization using a fluorescent protein probe presenting a pseudo-amyloid structure. Chem. Commun. 48, 1568–1570 (2012).

    CAS  Google Scholar 

  89. Ferreon, A. C. M., Gambin, Y., Lemke, E. A. & Deniz, A. A. Interplay of α-synuclein binding and conformational switching probed by single-molecule fluorescence. Proc. Natl Acad. Sci. USA 106, 5645–5650 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Tosatto, L. et al. Single-molecule FRET studies on alpha-synuclein oligomerization of Parkinson’s disease genetically related mutants. Sci. Rep. 5, 16696 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Nath, S., Meuvis, J., Hendrix, J., Carl, S. A. & Engelborghs, Y. Early aggregation steps in α-synuclein as measured by FCS and FRET: evidence for a contagious conformational change. Biophys. J. 98, 1302–1311 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Degorce, F. et al. HTRF: a technology tailored for drug discovery — a review of theoretical aspects and recent applications. Curr. Chem. Genomics 3, 22–32 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Clarke, E. E. & Shearman, M. S. Quantitation of amyloid-β peptides in biological milieu using a novel homogeneous time-resolved fluorescence (HTRF) assay. J. Neurosci. Methods 102, 61–68 (2000).

    CAS  PubMed  Google Scholar 

  94. Beaudet, L. et al. AlphaLISA immunoassays: the no-wash alternative to ELISAs for research and drug discovery. Nat. Methods 5, an8–an9 (2008).

    CAS  Google Scholar 

  95. Zhao, H. et al. AlphaLISA detection of alpha-synuclein in the cerebrospinal fluid and its potential application in Parkinson’s disease diagnosis. Protein Cell 8, 696–700 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Dale, N. C., Johnstone, E. K. M., White, C. W. & Pfleger, K. D. G. NanoBRET: the bright future of proximity-based assays. Front. Bioeng. Biotechnol. 7, 56 (2019).

    PubMed  PubMed Central  Google Scholar 

  97. Moriya, C. et al. PRDM14 directly interacts with heat shock proteins HSP90α and glucose-regulated protein 78. Cancer Sci. 109, 373–383 (2018).

    CAS  PubMed  Google Scholar 

  98. van Ham, T. J. et al. Towards multiparametric fluorescent imaging of amyloid formation: studies of a YFP model of α-synuclein aggregation. J. Mol. Biol. 395, 627–642 (2010).

    PubMed  Google Scholar 

  99. Chan, F. T. S., Kaminski, C. F. & Schierle, G. S. K. HomoFRET fluorescence anisotropy imaging as a tool to study molecular self-assembly in live cells. ChemPhysChem 12, 500–509 (2011).

    CAS  PubMed  Google Scholar 

  100. Kundel, F. et al. Shedding light on aberrant interactions — a review of modern tools for studying protein aggregates. FEBS J. 285, 3604–3630 (2018).

    CAS  PubMed  Google Scholar 

  101. De, S. & Klenerman, D. Imaging individual protein aggregates to follow aggregation and determine the role of aggregates in neurodegenerative disease. Biochim. Biophys. Acta Proteins Proteom. 1867, 870–878 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Horrocks, M. H. et al. Fast flow microfluidics and single-molecule fluorescence for the rapid characterization of α-synuclein oligomers. Anal. Chem. 87, 8818–8826 (2015).

    CAS  PubMed  Google Scholar 

  103. Kjaergaard, M. et al. Oligomer diversity during the aggregation of the repeat region of tau. ACS Chem. Neurosci. 9, 3060–3071 (2018).

    CAS  PubMed  Google Scholar 

  104. Anderson, V. L. & Webb, W. W. Transmission electron microscopy characterization of fluorescently labelled amyloid β 1-40 and α-synuclein aggregates. BMC Biotechnol. 11, 125 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Outeiro, T. F. et al. Formation of toxic oligomeric α-synuclein species in living cells. PLoS ONE 3, e1867 (2008).

    PubMed  PubMed Central  Google Scholar 

  106. Kiechle, M. et al. In vivo protein complementation demonstrates presynaptic α-synuclein oligomerization and age-dependent accumulation of 8–16-mer oligomer species. Cell Rep. 29, 2862–2874 (2019).

    CAS  PubMed  Google Scholar 

  107. Frey, B., AlOkda, A. & Lashuel, H. Monitoring alpha-synuclein oligomerization and inclusion formation using bimolecular fluorescence complementation assays: what you see is not always what you get. J. Neurochem. 56, 583–586 (2020).

    Google Scholar 

  108. Mannini, B. et al. Stabilization and characterization of cytotoxic Aβ40 oligomers isolated from an aggregation reaction in the presence of zinc ions. ACS Chem. Neurosci. 9, 2959–2971 (2018).

    CAS  PubMed  Google Scholar 

  109. Horrocks, M. H. et al. Single-molecule imaging of individual amyloid protein aggregates in human biofluids. ACS Chem. Neurosci. 7, 399–406 (2016).

    CAS  PubMed  Google Scholar 

  110. Cliffe, R. et al. Filamentous aggregates are fragmented by the proteasome holoenzyme. Cell Rep. 26, 2140–2149 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Needham, L.-M. et al. ThX – a next-generation probe for the early detection of amyloid aggregates. Chem. Sci. 11, 4578–4583 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Bongiovanni, M. N. et al. Multi-dimensional super-resolution imaging enables surface hydrophobicity mapping. Nat. Commun. 7, 13544 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Lee, J.-E. et al. Mapping surface hydrophobicity of α-synuclein oligomers at the nanoscale. Nano Lett. 18, 7494–7501 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Kravchenko, K. et al. Analysis of anticoagulants for blood-based quantitation of amyloid β oligomers in the sFIDA assay. Biol. Chem. 398, 465–475 (2017).

    CAS  PubMed  Google Scholar 

  115. Kühbach, K. et al. Application of an amyloid beta oligomer standard in the sFIDA assay. Front. Neurosci. 10, 8 (2016).

    PubMed  PubMed Central  Google Scholar 

  116. Kulawik, A., Heise, H., Zafiu, C., Willbold, D. & Bannach, O. Advancements of the sFIDA method for oligomer-based diagnostics of neurodegenerative diseases. FEBS Lett. 592, 516–534 (2018).

    CAS  PubMed  Google Scholar 

  117. Hulsemann, M. et al. Biofunctionalized silica nanoparticles: standards in amyloid-β oligomer-based diagnosis of Alzheimer’s disease. J. Alzheimers Dis. 54, 79–88 (2016).

    PubMed  Google Scholar 

  118. Herrmann, Y. et al. sFIDA automation yields sub-femtomolar limit of detection for Aβ aggregates in body fluids. Clin. Biochem. 50, 244–247 (2016).

    PubMed  Google Scholar 

  119. Jain, A. et al. Probing cellular protein complexes using single-molecule pull-down. Nature 473, 484–488 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Aggarwal, V. & Ha, T. Single-molecule fluorescence microscopy of native macromolecular complexes. Curr. Opin. Struct. Biol. 41, 225–232 (2016).

    CAS  PubMed  Google Scholar 

  121. Je, G. et al. Endogenous alpha-synuclein protein analysis from human brain tissues using single-molecule pull-down assay. Anal. Chem. 89, 13044–13048 (2017).

    CAS  PubMed  Google Scholar 

  122. Mitkevich, O. V. et al. DNA aptamers detecting generic amyloid epitopes. Prion 6, 400–406 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Rahimi, F. Aptamers selected for recognizing amyloid β-protein — a case for cautious optimism. Int. J. Mol. Sci. 19, 668 (2018).

    PubMed Central  Google Scholar 

  124. Schnitzbauer, J., Strauss, M. T., Schlichthaerle, T., Schueder, F. & Jungmann, R. Super-resolution microscopy with DNA-PAINT. Nat. Protoc. 12, 1198–1228 (2017).

    CAS  PubMed  Google Scholar 

  125. Whiten, D. R. et al. Nanoscopic characterisation of individual endogenous protein aggregates in human neuronal cells. ChemBioChem 19, 2033–2038 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Kundel, F. et al. Hsp70 inhibits the nucleation and elongation of tau and sequesters tau aggregates with high affinity. ACS Chem. Biol. 13, 636–646 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Stroud, J. C., Liu, C., Teng, P. K. & Eisenberg, D. Toxic fibrillar oligomers of amyloid-β have cross-β structure. Proc. Natl Acad. Sci. USA 109, 7717–7722 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  128. Ahmed, M. et al. Structural conversion of neurotoxic amyloid-β1–42 oligomers to fibrils. Nat. Struct. Mol. Biol. 17, 561–567 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Chen, S. W. et al. Structural characterization of toxic oligomers that are kinetically trapped during α-synuclein fibril formation. Proc. Natl Acad. Sci. USA 112, E1994–E2003 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Pieri, L., Madiona, K. & Melki, R. Structural and functional properties of prefibrillar α-synuclein oligomers. Sci. Rep. 6, 24526 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Karikari, T. K. et al. Preparation of stable tau oligomers for cellular and biochemical studies. Anal. Biochem. 566, 67–74 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  132. Zhang, W. et al. Heparin-induced tau filaments are polymorphic and differ from those in Alzheimer’s and Pick’s diseases. eLife 8, e43584 (2019).

    PubMed  PubMed Central  Google Scholar 

  133. Falcon, B. et al. Structures of filaments from Pick’s disease reveal a novel tau protein fold. Nature 561, 137–140 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  134. Fitzpatrick, A. W. P. et al. Cryo-EM structures of tau filaments from Alzheimer’s disease brain. Nature 547, 185–190 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  135. Guerrero-Ferreira, R. et al. Cryo-EM structure of alpha-synuclein fibrils. eLife 7, e36402 (2018).

    PubMed  PubMed Central  Google Scholar 

  136. Tian, Y., Liang, R., Kumar, A., Szwedziak, P. & Viles, J. H. 3D-visualization of amyloid-β oligomer and fibril interactions with lipid membranes by cryo-electron tomography. Preprint at bioRxiv https://doi.org/10.1101/2020.07.21.214072 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  137. Cooper, M. A. Optical biosensors: where next and how soon? Drug Discov. Today 11, 1061–1067 (2006).

    CAS  PubMed  Google Scholar 

  138. Aprile, F. A. et al. Selective targeting of primary and secondary nucleation pathways in Aβ42 aggregation using a rational antibody scanning method. Sci. Adv. 3, e1700488 (2017).

    PubMed  PubMed Central  Google Scholar 

  139. Chang, P.-T. et al. A newly designed molecule J2326 for Alzheimer’s disease disaggregates amyloid fibrils and induces neurite outgrowth. Neuropharmacology 92, 146–157 (2015).

    CAS  PubMed  Google Scholar 

  140. Mustafa, M. K. et al. Detection of β-amyloid peptide (1–16) and amyloid precursor protein (APP770) using spectroscopic ellipsometry and QCM techniques: a step forward towards Alzheimers disease diagnostics. Biosens. Bioelectron. 26, 1332–1336 (2010).

    CAS  PubMed  Google Scholar 

  141. Chauhan, N., Maekawa, T. & Kumar, D. N. S. Graphene based biosensors — accelerating medical diagnostics to new-dimensions. J. Mater. Res. 32, 2860–2882 (2017).

    CAS  Google Scholar 

  142. Jang, S. J., Lee, C. S. & Kim, T. H. α-Synuclein oligomer detection with aptamer switch on reduced graphene oxide electrode. Nanomaterials 10, 832 (2020).

    CAS  PubMed Central  Google Scholar 

  143. Peña-Bahamonde, J., Nguyen, H. N., Fanourakis, S. K. & Rodrigues, D. F. Recent advances in graphene-based biosensor technology with applications in life sciences. J. Nanobiotechnol. 16, 75 (2018).

    Google Scholar 

  144. Arter, W. E., Levin, A., Krainer, G. & Knowles, T. P. J. Microfluidic approaches for the analysis of protein–protein interactions in solution. Biophys. Rev. 12, 575–585 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  145. Yates, E. V. et al. Latent analysis of unmodified biomolecules and their complexes in solution with attomole detection sensitivity. Nat. Chem. 7, 802–809 (2015).

    CAS  PubMed  Google Scholar 

  146. Arter, W. E. et al. Combining affinity selection and specific ion mobility for microchip protein sensing. Anal. Chem. 90, 10302–10310 (2018).

    CAS  PubMed  Google Scholar 

  147. Arter, W. E. et al. Rapid fractionation and characterisation of alpha-synuclein oligomers in solution. Preprint at bioRxiv https://doi.org/10.1101/2020.03.10.985804 (2020).

    Article  Google Scholar 

  148. Aebersold, R. & Mann, M. Mass-spectrometric exploration of proteome structure and function. Nature 537, 347–355 (2016).

    CAS  PubMed  Google Scholar 

  149. Bader, J. M. et al. Proteome profiling in cerebrospinal fluid reveals novel biomarkers of Alzheimer’s disease. Mol. Syst. Biol. 16, e9356 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  150. de Souza, N. & Picotti, P. Mass spectrometry analysis of the structural proteome. Curr. Opin. Struct. Biol. 60, 57–65 (2020).

    PubMed  Google Scholar 

  151. Marx, V. A dream of single-cell proteomics. Nat. Methods 16, 809–812 (2019).

    CAS  PubMed  Google Scholar 

  152. Bernstein, S. L. et al. Amyloid-β protein oligomerization and the importance of tetramers and dodecamers in the aetiology of Alzheimer’s disease. Nat. Chem. 1, 326–331 (2009). This article introduces the use of mass spectrometry to characterize oligomer populations.

    CAS  PubMed  PubMed Central  Google Scholar 

  153. Lieblein, T. et al. Structural rearrangement of amyloid-β upon inhibitor binding suppresses formation of Alzheimer’s disease related oligomers. eLife 9, e59306 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  154. Österlund, N., Moons, R., Ilag, L. L., Sobott, F. & Gräslund, A. Native ion mobility-mass spectrometry reveals the formation of β-barrel shaped amyloid-β hexamers in a membrane-mimicking environment. J. Am. Chem. Soc. 141, 10440–10450 (2019).

    PubMed  Google Scholar 

  155. Ilitchev, A. I. et al. Hetero-oligomeric amyloid assembly and mechanism: prion fragment PrP(106–126) catalyzes the islet amyloid polypeptide β-hairpin. J. Am. Chem. Soc. 140, 9685–9695 (2018).

    CAS  PubMed  Google Scholar 

  156. Hoffmann, W. et al. NFGAIL amyloid oligomers: the onset of beta-sheet formation and the mechanism for fibril formation. J. Am. Chem. Soc. 140, 244–249 (2018).

    CAS  PubMed  Google Scholar 

  157. Smith, A. M., Jahn, T. R., Ashcroft, A. E. & Radford, S. E. Direct observation of oligomeric species formed in the early stages of amyloid fibril formation using electrospray ionisation mass spectrometry. J. Mol. Biol. 364, 9–19 (2006).

    CAS  PubMed  Google Scholar 

  158. Nakamura, A. et al. High performance plasma amyloid-β biomarkers for Alzheimer’s disease. Nature 554, 249–254 (2018).

    CAS  PubMed  Google Scholar 

  159. Chang, E. & Kuret, J. Detection and quantification of tau aggregation using a membrane filter assay. Anal. Biochem. 373, 330–336 (2008).

    CAS  PubMed  Google Scholar 

  160. Nasir, I., Linse, S. & Cabaleiro-Lago, C. Fluorescent filter-trap assay for amyloid fibril formation kinetics in complex solutions. ACS Chem. Neurosci. 6, 1436–1444 (2015).

    CAS  PubMed  Google Scholar 

  161. Ruggeri, F. S., Habchi, J., Cerreta, A. & Dietler, G. AFM-based single molecule techniques: unraveling the amyloid pathogenic species. Curr. Pharm. Des. 22, 3950–3970 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  162. Ruggeri, F. S., Šneideris, T., Vendruscolo, M. & Knowles, T. P. J. Atomic force microscopy for single molecule characterisation of protein aggregation. Arch. Biochem. Biophys. 664, 134–148 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  163. Ruggeri, F. S. et al. Microfluidic deposition for resolving single-molecule protein architecture and heterogeneity. Nat. Commun. 9, 3890 (2018).

    PubMed  PubMed Central  Google Scholar 

  164. Qamar, S. et al. FUS phase separation is modulated by a molecular chaperone and methylation of arginine cation–π interactions. Cell 173, 720–734 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  165. Ruggeri, F. S., Mannini, B., Schmid, R., Vendruscolo, M. & Knowles, T. P. J. Single molecule secondary structure determination of proteins through infrared absorption nanospectroscopy. Nat. Commun. 11, 2945 (2020). The combination of atomic force microscopy with infrared spectroscopy enables one to structurally characterize single molecules.

    CAS  PubMed  PubMed Central  Google Scholar 

  166. Yang, B., Liu, Z., Liu, H. & Nash, M. A. Next generation methods for single-molecule force spectroscopy on polyproteins and receptor–ligand complexes. Front. Mol. Biosci. 7, 85 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  167. Sonn-Segev, A. et al. Quantifying the heterogeneity of macromolecular machines by mass photometry. Nat. Commun. 11, 1772 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  168. Young, G. et al. Quantitative mass imaging of single biological macromolecules. Science 360, 423–427 (2018). This work introduces the use of interferometric scattering microscopy for single-molecule detection.

    CAS  PubMed  PubMed Central  Google Scholar 

  169. Young, G. & Kukura, P. Interferometric scattering microscopy. Annu. Rev. Phys. Chem. 19, 4827–4835 (2019).

    Google Scholar 

  170. Soltermann, F. et al. Quantifying protein–protein interactions by molecular counting with mass photometry. Angew. Chem. Int. Ed. 59, 10774–10779 (2020).

    CAS  Google Scholar 

  171. Wilham, J. M. et al. Rapid end-point quantitation of prion seeding activity with sensitivity comparable to bioassays. PLoS Pathog. 6, e1001217 (2010).

    PubMed  PubMed Central  Google Scholar 

  172. Atarashi, R. et al. Ultrasensitive human prion detection in cerebrospinal fluid by real-time quaking-induced conversion. Nat. Med. 17, 175–178 (2011).

    CAS  PubMed  Google Scholar 

  173. Saborio, G. P., Permanne, B. & Soto, C. Sensitive detection of pathological prion protein by cyclic amplification of protein misfolding. Nature 411, 810–813 (2001).

    CAS  PubMed  Google Scholar 

  174. Candelise, N. et al. Seeding variability of different alpha synuclein strains in synucleinopathies. Ann. Neurol. 85, 691–703 (2019).

    CAS  PubMed  Google Scholar 

  175. De Luca, C. M. G. et al. Efficient RT-QuIC seeding activity for α-synuclein in olfactory mucosa samples of patients with Parkinson’s disease and multiple system atrophy. Transl Neurodegener. 8, 24 (2019).

    PubMed  PubMed Central  Google Scholar 

  176. van Rumund, A. et al. α-Synuclein real-time quaking-induced conversion in the cerebrospinal fluid of uncertain cases of parkinsonism. Ann. Neurol. 85, 777–781 (2019).

    PubMed  PubMed Central  Google Scholar 

  177. Garrido, A., Fairfoul, G., Tolosa, E. S., Martí, M. J. & Green, A. α-synuclein RT-QuIC in cerebrospinal fluid of LRRK2-linked Parkinson’s disease. Ann. Clin. Transl Neurol. 6, 1024–1032 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  178. Metrick, M. A. et al. A single ultrasensitive assay for detection and discrimination of tau aggregates of Alzheimer and Pick diseases. Acta Neuropathol. Commun. 8, 22 (2020). This work illustrates the use of real-time quaking-induced conversion (RT-QuIC), a seed amplification assay, to discriminate different tauopathies.

    CAS  PubMed  PubMed Central  Google Scholar 

  179. Kraus, A. et al. Seeding selectivity and ultrasensitive detection of tau aggregate conformers of Alzheimer disease. Acta Neuropathol. 137, 585–598 (2019).

    PubMed  Google Scholar 

  180. 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 

  181. Bongianni, M. et al. α-Synuclein RT-QuIC assay in cerebrospinal fluid of patients with dementia with Lewy bodies. Ann. Clin. Transl Neurol. 6, 2120–2126 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  182. Becker, K. et al. Detecting alpha synuclein seeding activity in formaldehyde-fixed MSA patient tissue by PMCA. Mol. Neurobiol. 55, 8728–8737 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  183. Arosio, P., Cukalevski, R., Frohm, B., Knowles, T. P. J. & Linse, S. Quantification of the concentration of Aβ42 propagons during the lag phase by an amyloid chain reaction assay. J. Am. Chem. Soc. 236, 219–225 (2013).

    Google Scholar 

  184. Hooker, J. M. & Carson, R. E. Human positron emission tomography neuroimaging. Annu. Rev. Biomed. Eng. 21, 551–581 (2019).

    CAS  PubMed  Google Scholar 

  185. Sevigny, J. et al. The antibody aducanumab reduces Aβ plaques in Alzheimer’s disease. Nature 537, 50–56 (2016).

    CAS  PubMed  Google Scholar 

  186. Leuzy, A. et al. Tau PET imaging in neurodegenerative tauopathies — still a challenge. Mol. Psychiatry 24, 1112–1134 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  187. Wong, D. F. et al. Characterization of 3 novel tau radiopharmaceuticals, 11C-RO-963, 11C-RO-643, and 18F-RO-948, in healthy controls and in Alzheimer subjects. J. Nucl. Med. 59, 1869–1876 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  188. Betthauser, T. J. et al. In vivo characterization and quantification of neurofibrillary tau PET radioligand 18F-MK-6240 in humans from Alzheimer disease dementia to young controls. J. Nucl. Med. 60, 93–99 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  189. Khoury, R. & Ghossoub, E. Diagnostic biomarkers of Alzheimer’s disease: a state-of-the-art review. Biomark. Neuropsychiatry 1, 100005 (2019).

    Google Scholar 

  190. Sanchez-Catasus, C. A. et al. FDG-PET for prediction of AD dementia in mild cognitive impairment. A review of the state of the art with particular emphasis on the comparison with other neuroimaging modalities (MRI and perfusion SPECT). Curr. Alzheimer Res. 14, 127–142 (2017).

    CAS  PubMed  Google Scholar 

  191. Sehlin, D. et al. Engineered antibodies: new possibilities for brain PET? Eur. J. Nucl. Med. Mol. Imaging 46, 2848–2858 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  192. Sehlin, D. et al. Antibody-based PET imaging of amyloid beta in mouse models of Alzheimer’s disease. Nat. Commun. 7, 10759 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  193. Limbocker, R. et al. Trodusquemine enhances Aβ42 aggregation but suppresses its toxicity by displacing oligomers from cell membranes. Nat. Commun. 10, 225 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  194. De, S. et al. Different soluble aggregates of Aβ42 can give rise to cellular toxicity through different mechanisms. Nat. Commun. 10, 1541 (2019).

    PubMed  PubMed Central  Google Scholar 

  195. Kumar, S. T., Donzelli, S., Chiki, A., Syed, M. M. K. & Lashuel, H. A. A simple, versatile and robust centrifugation-based filtration protocol for the isolation and quantification of α-synuclein monomers, oligomers and fibrils: Towards improving experimental reproducibility in α-synuclein research. J. Neurochem. 153, 103–119 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  196. Lasagna-Reeves, C. A. et al. Identification of oligomers at early stages of tau aggregation in Alzheimer’s disease. FASEB J. 26, 1946–1959 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  197. Castillo-Carranza, D. L. et al. Passive immunization with tau oligomer monoclonal antibody reverses tauopathy phenotypes without affecting hyperphosphorylated neurofibrillary tangles. J. Neurosci. 34, 4260–4272 (2014).

    PubMed  PubMed Central  Google Scholar 

  198. Kayed, R. et al. Fibril specific, conformation dependent antibodies recognize a generic epitope common to amyloid fibrils and fibrillar oligomers that is absent in prefibrillar oligomers. Mol. Neurodegener. 2, 18 (2007).

    PubMed  PubMed Central  Google Scholar 

  199. Ponsel, D., Neugebauer, J., Ladetzki-Baehs, K. & Tissot, K. High affinity, developability and functional size: The holy grail of combinatorial antibody library generation. Molecules 16, 3675–3700 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  200. Winter, G., Griffiths, A. D., Hawkins, R. E. & Hoogenboom, H. R. Making antibodies by phage display technology. Annu. Rev. Immunol. 12, 433–455 (1994).

    CAS  PubMed  Google Scholar 

  201. Bradbury, A. R. M., Sidhu, S., Dübel, S. & McCafferty, J. Beyond natural antibodies: the power of in vitro display technologies. Nat. Biotechnol. 29, 245–254 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  202. Munke, A. et al. Phage display and kinetic selection of antibodies that specifically inhibit amyloid self-replication. Proc. Natl Acad. Sci. USA 114, 6444–6449 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  203. Morgado, I. et al. Molecular basis of β-amyloid oligomer recognition with a conformational antibody fragment. Proc. Natl Acad. Sci. USA 109, 12503–12508 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  204. Spencer, S., Bethea, D., Raju, T. S., Giles-Komar, J. & Feng, Y. Solubility evaluation of murine hybridoma antibodies. mAbs 4, 319–325 (2012).

    PubMed  PubMed Central  Google Scholar 

  205. Wu, S. J. et al. Structure-based engineering of a monoclonal antibody for improved solubility. Protein Eng. Des. Sel. 23, 643–651 (2010).

    CAS  PubMed  Google Scholar 

  206. Jain, T. et al. Biophysical properties of the clinical-stage antibody landscape. Proc. Natl Acad. Sci. USA 114, 944–949 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  207. Pepinsky, R. B. et al. Improving the solubility of anti-LINGO-1 monoclonal antibody Li33 by isotype switching and targeted mutagenesis. Protein Sci. 19, 954–966 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  208. Burkovitz, A., Sela-Culang, I. & Ofran, Y. Large-scale analysis of somatic hypermutations in antibodies reveals which structural regions, positions and amino acids are modified to improve affinity. FEBS J. 281, 306–319 (2014).

    CAS  PubMed  Google Scholar 

  209. Sormanni, P., Aprile, F. A. & Vendruscolo, M. Rational design of antibodies targeting specific epitopes within intrinsically disordered proteins. Proc. Natl Acad. Sci. USA 112, 9902–9907 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  210. Sormanni, P., Aprile, F. A. & Vendruscolo, M. Third generation antibody discovery methods: in silico rational design. Chem. Soc. Rev. 47, 9137–9157 (2018).

    CAS  PubMed  Google Scholar 

  211. Sormanni, P., Aprile, F. A. & Vendruscolo, M. The CamSol method of rational design of protein mutants with enhanced solubility. J. Mol. Biol. 427, 478–490 (2015).

    CAS  PubMed  Google Scholar 

  212. Sormanni, P. & Vendruscolo, M. Protein solubility predictions using the CamSol method in the study of protein homeostasis. Cold Spring Harb. Perspect. Biol. 11, a033845 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  213. Aprile, F. A. et al. Rational design of a conformation-specific antibody for the quantification of Aβ oligomers. Proc. Natl Acad. Sci. USA 117, 13509–13518 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  214. Zeng, F. et al. Versatile near-infrared fluorescent probe for in vivo detection of Aβ oligomers. Bioorg. Med. Chem. 28, 115559 (2020).

    CAS  PubMed  Google Scholar 

  215. Teoh, C. L. et al. Chemical fluorescent probe for detection of Aβ oligomers. J. Am. Chem. Soc. 137, 13503–13509 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  216. Yang, J. et al. Highly specific detection of Aβ oligomers in early Alzheimer’s disease by a near-infrared fluorescent probe with a ‘V-shaped’ spatial conformation. Chem. Commun. 56, 583–586 (2020).

    CAS  Google Scholar 

  217. Lv, G. et al. A novel near-infrared fluorescent probe for detection of early-stage Aβ protofibrils in Alzheimer’s disease. Chem. Commun. 56, 1625–1628 (2020).

    CAS  Google Scholar 

  218. McIntyre, P. G. How many drops of CSF is enough? Postgrad. Med. J. 83, 158 (2007).

    PubMed Central  Google Scholar 

  219. Lindquist, M. Neuroimaging results altered by varying analysis pipelines. Nature 582, 36–37 (2020).

    CAS  PubMed  Google Scholar 

  220. Cummings, J. The role of biomarkers in Alzheimer’s disease drug development. Adv. Exp. Med. Biol. 1118, 29–61 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  221. R&D Systems. Luminex assays & instruments. R&D Systems https://www.rndsystems.com/products/luminex-assays-and-high-performance-assays (2021).

  222. Sartorius AG. BLI technology. Sartorius https://www.fortebio.com/applications/bli-technology (2020).

  223. Arosio, P. et al. Microfluidic diffusion analysis of the sizes and interactions of proteins under native solution conditions. ACS Nano 10, 333–341 (2016).

    CAS  PubMed  Google Scholar 

  224. Gough, K. C., Rees, H. C., Ives, S. E. & Maddison, B. C. Methods for differentiating prion types in food-producing animals. Biology 4, 785–813 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  225. Rowe, C. C. et al. Standardized expression of 18F-NAV4694 and 11C-PiB β-amyloid PET results with the centiloid scale. J. Nucl. Med. 57, 1233–1237 (2016).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by the UK Medical Research Council. P.S. is supported by a Royal Society University Research Fellowship (URF\R1\201461).

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to the preparation of the manuscript.

Corresponding author

Correspondence to Michele Vendruscolo.

Ethics declarations

Competing interests

The authors declare no competing interests.

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

Kulenkampff, K., Wolf Perez, AM., Sormanni, P. et al. Quantifying misfolded protein oligomers as drug targets and biomarkers in Alzheimer and Parkinson diseases. Nat Rev Chem 5, 277–294 (2021). https://doi.org/10.1038/s41570-021-00254-9

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41570-021-00254-9

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