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:

Super-resolution microscopy: a closer look at synaptic dysfunction in Alzheimer disease

Abstract

The synapse has emerged as a critical neuronal structure in the degenerative process of Alzheimer disease (AD), in which the pathogenic signals of two key players — amyloid-β (Aβ) and tau — converge, thereby causing synaptic dysfunction and cognitive deficits. The synapse presents a dynamic, confined microenvironment in which to explore how key molecules travel, localize, interact and assume different levels of organizational complexity, thereby affecting neuronal function. However, owing to their small size and the diffraction-limited resolution of conventional light microscopic approaches, investigating synaptic structure and dynamics has been challenging. Super-resolution microscopy (SRM) techniques have overcome the resolution barrier and are revolutionizing our quantitative understanding of biological systems in unprecedented spatio-temporal detail. Here we review critical new insights provided by SRM into the molecular architecture and dynamic organization of the synapse and, in particular, the interactions between Aβ and tau in this compartment. We further highlight how SRM can transform our understanding of the molecular pathological mechanisms that underlie AD. The application of SRM for understanding the roles of synapses in AD pathology will provide a stepping stone towards a broader understanding of dysfunction in other subcellular compartments and at cellular and circuit levels in this disease.

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: The spatio-temporal dynamics of the healthy synapse.
Fig. 2: Examples of super-resolution imaging of molecules implicated in Alzheimer disease.
Fig. 3: Overview of outstanding questions that super-resolution microscopy can address.
Fig. 4: Towards a quantitative understanding of pathomechanisms in Alzheimer disease.

Similar content being viewed by others

References

  1. DeTure, M. A. & Dickson, D. W. The neuropathological diagnosis of Alzheimer’s disease. Mol. Neurodegener. 14, 32 (2019).

    PubMed  PubMed Central  Google Scholar 

  2. Brookmeyer, R., Gray, S. & Kawas, C. Projections of Alzheimer’s disease in the United States and the public health impact of delaying disease onset. Am. J. Public Health 88, 1337–1342 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Hou, Y. et al. Ageing as a risk factor for neurodegenerative disease. Nat. Rev. Neurol. 15, 565–581 (2019).

    PubMed  Google Scholar 

  4. Kohler, C. Granulovacuolar degeneration: a neurodegenerative change that accompanies Tau pathology. Acta Neuropathol. 132, 339–359 (2016).

    PubMed  Google Scholar 

  5. Jack, C. R. Jr. et al. Prediction of AD with MRI-based hippocampal volume in mild cognitive impairment. Neurology 52, 1397–1403 (1999).

    PubMed  Google Scholar 

  6. Forner, S., Baglietto-Vargas, D., Martini, A. C., Trujillo-Estrada, L. & LaFerla, F. M. Synaptic impairment in Alzheimer’s disease: a dysregulated symphony. Trends Neurosci. 40, 347–357 (2017).

    CAS  PubMed  Google Scholar 

  7. Ittner, L. M. et al. Dendritic function of Tau mediates amyloid-beta toxicity in Alzheimer’s disease mouse models. Cell 142, 387–397 (2010). This study describes the crosstalk between Aβ and tau mediated by FYN.

    CAS  PubMed  Google Scholar 

  8. Li, C. & Götz, J. Somatodendritic accumulation of Tau in Alzheimer’s disease is promoted by Fyn-mediated local protein translation. EMBO J. 36, 3120–3138 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Roberson, E. D. et al. Reducing endogenous tau ameliorates amyloid beta-induced deficits in an Alzheimer’s disease mouse model. Science 316, 750–754 (2007).

    CAS  PubMed  Google Scholar 

  10. Selkoe, D. J. Alzheimer’s disease is a synaptic failure. Science 298, 789–791 (2002).

    CAS  PubMed  Google Scholar 

  11. Polanco, J. C. et al. Amyloid-beta and Tau complexity - towards improved biomarkers and targeted therapies. Nat. Rev. Neurol. 14, 22–39 (2018).

    CAS  PubMed  Google Scholar 

  12. Spires-Jones, T. L. & Hyman, B. T. The intersection of amyloid beta and Tau at synapses in Alzheimer’s disease. Neuron 82, 756–771 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Ittner, A. & Ittner, L. M. Dendritic Tau in Alzheimer’s disease. Neuron 99, 13–27 (2018).

    CAS  PubMed  Google Scholar 

  14. Kanaan, N. M. & Grabinski, T. Neuronal and glial distribution of tau protein in the adult rat and monkey. Front. Mol. Neurosci. 14, 607303 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Xu, J., Kurup, P., Foscue, E. & Lombroso, P. J. Striatal-enriched protein tyrosine phosphatase regulates the PTPα/Fyn signaling pathway. J. Neurochem. 134, 629–641 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Boehm, J. A ‘danse macabre’: tau and Fyn in STEP with amyloid beta to facilitate induction of synaptic depression and excitotoxicity. Eur. J. Neurosci. 37, 1925–1930 (2013).

    PubMed  Google Scholar 

  17. Yamada, K. Extracellular Tau and its potential role in the propagation of Tau pathology. Front. Neurosci. 11, 667 (2017).

    PubMed  PubMed Central  Google Scholar 

  18. Kneynsberg, A., Combs, B., Christensen, K., Morfini, G. & Kanaan, N. M. Axonal degeneration in tauopathies: disease relevance and underlying mechanisms. Front. Neurosci. 11, 572 (2017).

    PubMed  PubMed Central  Google Scholar 

  19. 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 

  20. Puzzo, D. et al. LTP and memory impairment caused by extracellular Abeta and Tau oligomers is APP-dependent. eLife 6, e26991 (2017).

    PubMed  PubMed Central  Google Scholar 

  21. Fa, M. et al. Extracellular Tau oligomers produce an immediate impairment of LTP and memory. Sci. Rep. 6, 19393 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Um, J. W. et al. Alzheimer amyloid-beta oligomer bound to postsynaptic prion protein activates Fyn to impair neurons. Nat. Neurosci. 15, 1227–1235 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Larson, M. et al. The complex PrP(c)-Fyn couples human oligomeric Abeta with pathological Tau changes in Alzheimer’s disease. J. Neurosci. 32, 16857–16871a (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Lee, S., Salazar, S. V., Cox, T. O. & Strittmatter, S. M. Pyk2 signaling through Graf1 and RhoA GTPase is required for amyloid-beta oligomer-triggered synapse loss. J. Neurosci. 39, 1910–1929 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Um, J. W. et al. Metabotropic glutamate receptor 5 is a coreceptor for Alzheimer Aβ oligomer bound to cellular prion protein. Neuron 79, 887–902 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Snyder, E. M. et al. Regulation of NMDA receptor trafficking by amyloid-beta. Nat. Neurosci. 8, 1051–1058 (2005).

    CAS  PubMed  Google Scholar 

  27. Kurup, P. et al. Abeta-mediated NMDA receptor endocytosis in Alzheimer’s disease involves ubiquitination of the tyrosine phosphatase STEP61. J. Neurosci. 30, 5948–5957 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Amar, F. et al. The amyloid-beta oligomer Abeta*56 induces specific alterations in neuronal signaling that lead to tau phosphorylation and aggregation. Sci. Signal. 10, eaal2021 (2017).

    PubMed  PubMed Central  Google Scholar 

  29. Mairet-Coello, G. et al. The CAMKK2-AMPK kinase pathway mediates the synaptotoxic effects of Abeta oligomers through Tau phosphorylation. Neuron 78, 94–108 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Ittner, A. et al. Site-specific phosphorylation of Tau inhibits amyloid-beta toxicity in Alzheimer’s mice. Science 354, 904–908 (2016).

    CAS  PubMed  Google Scholar 

  31. Cisse, M. et al. Reversing EphB2 depletion rescues cognitive functions in Alzheimer model. Nature 469, 47–52 (2011).

    CAS  PubMed  Google Scholar 

  32. Vargas, L. M. et al. EphA4 activation of c-Abl mediates synaptic loss and LTP blockade caused by amyloid-beta oligomers. PLoS ONE 9, e92309 (2014).

    PubMed  Google Scholar 

  33. Kim, T. et al. Human LilrB2 is a beta-amyloid receptor and its murine homolog PirB regulates synaptic plasticity in an Alzheimer’s model. Science 341, 1399–1404 (2013).

    CAS  PubMed  Google Scholar 

  34. Shankar, G. M. et al. Natural oligomers of the Alzheimer amyloid-beta protein induce reversible synapse loss by modulating an NMDA-type glutamate receptor-dependent signaling pathway. J. Neurosci. 27, 2866–2875 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Opazo, P. et al. CaMKII metaplasticity drives Abeta oligomer-mediated synaptotoxicity. Cell Rep. 23, 3137–3145 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Abbott, L. F. & Regehr, W. G. Synaptic computation. Nature 431, 796–803 (2004).

    CAS  PubMed  Google Scholar 

  37. Sudhof, T. C. The presynaptic active zone. Neuron 75, 11–25 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Biederer, T., Kaeser, P. S. & Blanpied, T. A. Transcellular nanoalignment of synaptic function. Neuron 96, 680–696 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Engert, F. & Bonhoeffer, T. Dendritic spine changes associated with hippocampal long-term synaptic plasticity. Nature 399, 66–70 (1999).

    CAS  PubMed  Google Scholar 

  40. Zucker, R. S. & Regehr, W. G. Short-term synaptic plasticity. Annu. Rev. Physiol. 64, 355–405 (2002).

    CAS  PubMed  Google Scholar 

  41. Motanis, H., Seay, M. J. & Buonomano, D. V. Short-term synaptic plasticity as a mechanism for sensory timing. Trends Neurosci. 41, 701–711 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Lynch, M. A. Long-term potentiation and memory. Physiol. Rev. 84, 87–136 (2004).

    CAS  PubMed  Google Scholar 

  43. Matsuzaki, M., Honkura, N., Ellis-Davies, G. C. & Kasai, H. Structural basis of long-term potentiation in single dendritic spines. Nature 429, 761–766 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Zhou, Q., Homma, K. J. & Poo, M. M. Shrinkage of dendritic spines associated with long-term depression of hippocampal synapses. Neuron 44, 749–757 (2004).

    CAS  PubMed  Google Scholar 

  45. Nishiyama, J. & Yasuda, R. Biochemical computation for spine structural plasticity. Neuron 87, 63–75 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Evans, R. C. & Blackwell, K. T. Calcium: amplitude, duration, or location? Biol. Bull. 228, 75–83 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Lüscher, C. & Malenka, R. C. NMDA receptor-dependent long-term potentiation and long-term depression (LTP/LTD). Cold Spring Harb. Perspect. Biol. 4, a005710 (2012).

    PubMed  PubMed Central  Google Scholar 

  48. Becker, N., Wierenga, C. J., Fonseca, R., Bonhoeffer, T. & Nägerl, U. V. LTD induction causes morphological changes of presynaptic boutons and reduces their contacts with spines. Neuron 60, 590–597 (2008).

    CAS  PubMed  Google Scholar 

  49. Borgdorff, A. J. & Choquet, D. Regulation of AMPA receptor lateral movements. Nature 417, 649–653 (2002).

    CAS  PubMed  Google Scholar 

  50. Dahan, M. et al. Diffusion dynamics of glycine receptors revealed by single-quantum dot tracking. Science 302, 442–445 (2003).

    CAS  PubMed  Google Scholar 

  51. Meier, J., Vannier, C., Serge, A., Triller, A. & Choquet, D. Fast and reversible trapping of surface glycine receptors by gephyrin. Nat. Neurosci. 4, 253–260 (2001).

    CAS  PubMed  Google Scholar 

  52. Tardin, C., Cognet, L., Bats, C., Lounis, B. & Choquet, D. Direct imaging of lateral movements of AMPA receptors inside synapses. EMBO J. 22, 4656–4665 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Penn, A. C. et al. Hippocampal LTP and contextual learning require surface diffusion of AMPA receptors. Nature 549, 384–388 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Heck, J. et al. Transient confinement of CaV2.1 Ca2+-channel splice variants shapes synaptic short-term plasticity. Neuron 103, 66–79 e12 (2019).

    CAS  PubMed  Google Scholar 

  55. Padmanabhan, P. et al. Need for speed: super-resolving the dynamic nanoclustering of syntaxin-1 at exocytic fusion sites. Neuropharmacology 169, 107554 (2020).

    CAS  PubMed  Google Scholar 

  56. Bademosi, A. T. et al. In vivo single-molecule imaging of syntaxin1A reveals polyphosphoinositide- and activity-dependent trapping in presynaptic nanoclusters. Nat. Commun. 8, 13660 (2017).

    CAS  PubMed  Google Scholar 

  57. Sakamoto, H. et al. Synaptic weight set by Munc13-1 supramolecular assemblies. Nat. Neurosci. 21, 41–49 (2018).

    CAS  PubMed  Google Scholar 

  58. Nair, D. et al. Super-resolution imaging reveals that AMPA receptors inside synapses are dynamically organized in nanodomains regulated by PSD95. J. Neurosci. 33, 13204–13224 (2013). This study uses several super-resolution techniques to provide insight into the dynamics of AMPAR nanodomains.

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Kellermayer, B. et al. Differential nanoscale topography and functional role of GluN2-NMDA receptor subtypes at glutamatergic synapses. Neuron 100, 106–119 e107 (2018).

    CAS  PubMed  Google Scholar 

  60. Padmanabhan, P., Martinez-Marmol, R., Xia, D., Götz, J. & Meunier, F. A. Frontotemporal dementia mutant Tau promotes aberrant Fyn nanoclustering in hippocampal dendritic spines. eLife 8, e45040 (2019). This study uses single-particle tracking techniques to reveal that frontotemporal dementia-linked P301L mutant tau immobilizes Fyn and increases Fyn clustering in dendritic spines.

    CAS  PubMed  PubMed Central  Google Scholar 

  61. MacGillavry, H. D., Song, Y., Raghavachari, S. & Blanpied, T. A. Nanoscale scaffolding domains within the postsynaptic density concentrate synaptic AMPA receptors. Neuron 78, 615–622 (2013). This single-molecule imaging study provides evidence for nanodomains of PSD scaffold proteins.

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Trotter, J. H. et al. Synaptic neurexin-1 assembles into dynamically regulated active zone nanoclusters. J. Cell Biol. 218, 2677–2698 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Heine, M. & Holcman, D. Asymmetry between pre- and postsynaptic transient nanodomains shapes neuronal communication. Trends Neurosci. 43, 182–196 (2020).

    CAS  PubMed  Google Scholar 

  64. Sieber, J. J. et al. Anatomy and dynamics of a supramolecular membrane protein cluster. Science 317, 1072–1076 (2007).

    CAS  PubMed  Google Scholar 

  65. Merklinger, E. et al. The packing density of a supramolecular membrane protein cluster is controlled by cytoplasmic interactions. eLife 6, e20705 (2017).

    PubMed  PubMed Central  Google Scholar 

  66. van den Bogaart, G. et al. Membrane protein sequestering by ionic protein-lipid interactions. Nature 479, 552–555 (2011).

    PubMed  PubMed Central  Google Scholar 

  67. Milovanovic, D. et al. Hydrophobic mismatch sorts SNARE proteins into distinct membrane domains. Nat. Commun. 6, 5984 (2015).

    CAS  PubMed  Google Scholar 

  68. Rebola, N. et al. Distinct nanoscale calcium channel and synaptic vesicle topographies contribute to the diversity of synaptic function. Neuron 104, 693–710 e699 (2019).

    CAS  PubMed  Google Scholar 

  69. Benda, A., Aitken, H., Davies, D. S., Whan, R. & Goldsbury, C. STED imaging of Tau filaments in Alzheimer’s disease cortical grey matter. J. Struct. Biol. 195, 345–352 (2016).

    CAS  PubMed  Google Scholar 

  70. Haas, K. T. et al. Pre-post synaptic alignment through neuroligin-1 tunes synaptic transmission efficiency. eLife 7, e31755 (2018).

    PubMed  PubMed Central  Google Scholar 

  71. Tarusawa, E. et al. Input-specific intrasynaptic arrangements of ionotropic glutamate receptors and their impact on postsynaptic responses. J. Neurosci. 29, 12896–12908 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Franks, K. M., Stevens, C. F. & Sejnowski, T. J. Independent sources of quantal variability at single glutamatergic synapses. J. Neurosci. 23, 3186–3195 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Trachtenberg, J. T. et al. Long-term in vivo imaging of experience-dependent synaptic plasticity in adult cortex. Nature 420, 788–794 (2002).

    CAS  PubMed  Google Scholar 

  74. Grutzendler, J., Kasthuri, N. & Gan, W. B. Long-term dendritic spine stability in the adult cortex. Nature 420, 812–816 (2002).

    CAS  PubMed  Google Scholar 

  75. Berry, K. P. & Nedivi, E. Spine dynamics: are they all the same? Neuron 96, 43–55 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Adrian, M., Kusters, R., Storm, C., Hoogenraad, C. C. & Kapitein, L. C. Probing the interplay between dendritic spine morphology and membrane-bound diffusion. Biophys. J. 113, 2261–2270 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Arellano, J. I., Benavides-Piccione, R., Defelipe, J. & Yuste, R. Ultrastructure of dendritic spines: correlation between synaptic and spine morphologies. Front. Neurosci. 1, 131–143 (2007).

    PubMed  PubMed Central  Google Scholar 

  78. Harris, K. M. & Stevens, J. K. Dendritic spines of CA 1 pyramidal cells in the rat hippocampus: serial electron microscopy with reference to their biophysical characteristics. J. Neurosci. 9, 2982–2997 (1989).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Dent, E. W. Of microtubules and memory: implications for microtubule dynamics in dendrites and spines. Mol. Biol. Cell 28, 1–8 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Chazeau, A. et al. Nanoscale segregation of actin nucleation and elongation factors determines dendritic spine protrusion. EMBO J. 33, 2745–2764 (2014). This single-molecule imaging study reveals the spatio-temporal dynamics of F-actin regulators in spines.

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Tarrant, S. B. & Routtenberg, A. The synaptic spinule in the dendritic spine: electron microscopic study of the hippocampal dentate gyrus. Tissue Cell 9, 461–473 (1977).

    CAS  PubMed  Google Scholar 

  82. Spacek, J. & Harris, K. M. Trans-endocytosis via spinules in adult rat hippocampus. J. Neurosci. 24, 4233–4241 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Westrum, L. E. & Blackstad, T. W. An electron microscopic study of the stratum radiatum of the rat hippocampus (regio superior, CA 1) with particular emphasis on synaptology. J. Comp. Neurol. 119, 281–309 (1962).

    CAS  PubMed  Google Scholar 

  84. Tatavarty, V., Kim, E. J., Rodionov, V. & Yu, J. Investigating sub-spine actin dynamics in rat hippocampal neurons with super-resolution optical imaging. PLoS ONE 4, e7724 (2009).

    PubMed  PubMed Central  Google Scholar 

  85. Frost, N. A., Shroff, H., Kong, H., Betzig, E. & Blanpied, T. A. Single-molecule discrimination of discrete perisynaptic and distributed sites of actin filament assembly within dendritic spines. Neuron 67, 86–99 (2010). This work reveals heterogeneous actin dynamics within individual spines.

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Schatzle, P. et al. Activity-dependent actin remodeling at the base of dendritic spines promotes microtubule entry. Curr. Biol. 28, 2081–2093 e2086 (2018).

    CAS  PubMed  Google Scholar 

  87. Xu, K., Zhong, G. & Zhuang, X. Actin, spectrin, and associated proteins form a periodic cytoskeletal structure in axons. Science 339, 452–456 (2013).

    CAS  PubMed  Google Scholar 

  88. Zhong, G. et al. Developmental mechanism of the periodic membrane skeleton in axons. eLife 3, e04581 (2014).

    PubMed Central  Google Scholar 

  89. D’Este, E., Kamin, D., Gottfert, F., El-Hady, A. & Hell, S. W. STED nanoscopy reveals the ubiquity of subcortical cytoskeleton periodicity in living neurons. Cell Rep. 10, 1246–1251 (2015).

    PubMed  Google Scholar 

  90. Sidenstein, S. C. et al. Multicolour multilevel STED nanoscopy of actin/spectrin organization at synapses. Sci. Rep. 6, 26725 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Bar, J., Kobler, O., van Bommel, B. & Mikhaylova, M. Periodic F-actin structures shape the neck of dendritic spines. Sci. Rep. 6, 37136 (2016).

    PubMed  PubMed Central  Google Scholar 

  92. Tonnesen, J., Katona, G., Rozsa, B. & Nagerl, U. V. Spine neck plasticity regulates compartmentalization of synapses. Nat. Neurosci. 17, 678–685 (2014). This study uses stimulated emission depletion microscopy to show that the dendritic spine neck shortens and widens after LTP.

    CAS  PubMed  Google Scholar 

  93. Hruska, M., Henderson, N., Le Marchand, S. J., Jafri, H. & Dalva, M. B. Synaptic nanomodules underlie the organization and plasticity of spine synapses. Nat. Neurosci. 21, 671–682 (2018). This study uses multicolour stimulated emission depletion microscopy to reveal that the number and mobility of aligned modules of presynaptic and postsynaptic proteins increase with chemically induced LTP.

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Pfeiffer, T. et al. Chronic 2P-STED imaging reveals high turnover of dendritic spines in the hippocampus in vivo. eLife 7, e34700 (2018). This super-resolution study provides a more accurate measurement of spine density and turnover.

    PubMed  PubMed Central  Google Scholar 

  95. Attardo, A., Fitzgerald, J. E. & Schnitzer, M. J. Impermanence of dendritic spines in live adult CA1 hippocampus. Nature 523, 592–596 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Gu, L. et al. Long-term in vivo imaging of dendritic spines in the hippocampus reveals structural plasticity. J. Neurosci. 34, 13948–13953 (2014).

    PubMed  PubMed Central  Google Scholar 

  97. Marsh, J. & Alifragis, P. Synaptic dysfunction in Alzheimer’s disease: the effects of amyloid beta on synaptic vesicle dynamics as a novel target for therapeutic intervention. Neural Regen. Res. 13, 616–623 (2018).

    PubMed  PubMed Central  Google Scholar 

  98. Fagiani, F., Lanni, C., Racchi, M., Pascale, A. & Govoni, S. Amyloid-beta and synaptic vesicle dynamics: a cacophonic orchestra. J. Alzheimers Dis. 72, 1–14 (2019).

    CAS  PubMed  Google Scholar 

  99. Barthet, G. & Mulle, C. Presynaptic failure in Alzheimer’s disease. Prog. Neurobiol. 194, 101801 (2020).

    CAS  PubMed  Google Scholar 

  100. de Wilde, M. C., Overk, C. R., Sijben, J. W. & Masliah, E. Meta-analysis of synaptic pathology in Alzheimer’s disease reveals selective molecular vesicular machinery vulnerability. Alzheimers Dement. 12, 633–644 (2016).

    PubMed  PubMed Central  Google Scholar 

  101. Haytural, H. et al. The proteome of the dentate terminal zone of the perforant path indicates presynaptic impairment in Alzheimer disease. Mol. Cell Proteom. 19, 128–141 (2020).

    CAS  Google Scholar 

  102. Kokubo, H., Kayed, R., Glabe, C. G. & Yamaguchi, H. Soluble Abeta oligomers ultrastructurally localize to cell processes and might be related to synaptic dysfunction in Alzheimer’s disease brain. Brain Res. 1031, 222–228 (2005).

    CAS  PubMed  Google Scholar 

  103. Sokolow, S. et al. Preferential accumulation of amyloid-beta in presynaptic glutamatergic terminals (VGluT1 and VGluT2) in Alzheimer’s disease cortex. Neurobiol. Dis. 45, 381–387 (2012).

    CAS  PubMed  Google Scholar 

  104. Fein, J. A. et al. Co-localization of amyloid beta and Tau pathology in Alzheimer’s disease synaptosomes. Am. J. Pathol. 172, 1683–1692 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Sokolow, S. et al. Pre-synaptic C-terminal truncated Tau is released from cortical synapses in Alzheimer’s disease. J. Neurochem. 133, 368–379 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Tai, H. C. et al. Frequent and symmetric deposition of misfolded Tau oligomers within presynaptic and postsynaptic terminals in Alzheimer’s disease. Acta Neuropathol. Commun. 2, 146 (2014).

    PubMed  PubMed Central  Google Scholar 

  107. McInnes, J. et al. Synaptogyrin-3 mediates presynaptic dysfunction induced by Tau. Neuron 97, 823–835 e828 (2018).

    CAS  PubMed  Google Scholar 

  108. Russell, C. L. et al. Amyloid-beta acts as a regulator of neurotransmitter release disrupting the interaction between synaptophysin and VAMP2. PLoS ONE 7, e43201 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Zhou, L. et al. Tau association with synaptic vesicles causes presynaptic dysfunction. Nat. Commun. 8, 15295 (2017).

    PubMed  PubMed Central  Google Scholar 

  110. Abramov, E. et al. Amyloid-beta as a positive endogenous regulator of release probability at hippocampal synapses. Nat. Neurosci. 12, 1567–1576 (2009).

    CAS  PubMed  Google Scholar 

  111. Parodi, J. et al. Beta-amyloid causes depletion of synaptic vesicles leading to neurotransmission failure. J. Biol. Chem. 285, 2506–2514 (2010).

    CAS  PubMed  Google Scholar 

  112. Yang, Y. et al. Amyloid-beta oligomers may impair SNARE-mediated exocytosis by direct binding to syntaxin-1a. Cell Rep. 12, 1244–1251 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. He, Y. et al. Amyloid beta oligomers suppress excitatory transmitter release via presynaptic depletion of phosphatidylinositol-4,5-bisphosphate. Nat. Commun. 10, 1193 (2019).

    PubMed  PubMed Central  Google Scholar 

  114. Kelly, B. L. & Ferreira, A. -Amyloid-induced dynamin-1 degradation is mediated by N-methyl-D-aspartate receptors in hippocampal neurons. J. Biol. Chem. 281, 28079–28089 (2006).

    CAS  PubMed  Google Scholar 

  115. Kelly, B. L., Vassar, R. & Ferreira, A. Beta-amyloid-induced dynamin-1 depletion in hippocampal neurons. A potential mechanism for early cognitive decline in Alzheimer disease. J. Biol. Chem. 280, 31746–31753 (2005).

    CAS  PubMed  Google Scholar 

  116. Park, D. et al. Activation of CaMKIV by soluble amyloid-beta1-42 impedes trafficking of axonal vesicles and impairs activity-dependent synaptogenesis. Sci Signal 10, eaam8661 (2017).

    PubMed  Google Scholar 

  117. Marsh, J., Bagol, S. H., Williams, R. S. B., Dickson, G. & Alifragis, P. Synapsin I phosphorylation is dysregulated by beta-amyloid oligomers and restored by valproic acid. Neurobiol. Dis. 106, 63–75 (2017).

    CAS  PubMed  Google Scholar 

  118. Decker, J. M. et al. Pro-aggregant Tau impairs mossy fiber plasticity due to structural changes and Ca++ dysregulation. Acta Neuropathol. Commun. 3, 23 (2015).

    PubMed  PubMed Central  Google Scholar 

  119. Hatch, R. J., Wei, Y., Xia, D. & Götz, J. Hyperphosphorylated Tau causes reduced hippocampal CA1 excitability by relocating the axon initial segment. Acta Neuropathol. 133, 717–730 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Sohn, P. D. et al. Pathogenic Tau impairs axon initial segment plasticity and excitability homeostasis. Neuron 104, 458–470 e455 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Gramlich, M. W. & Klyachko, V. A. Nanoscale organization of vesicle release at central synapses. Trends Neurosci. 42, 425–437 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Jahn, R. & Fasshauer, D. Molecular machines governing exocytosis of synaptic vesicles. Nature 490, 201–207 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Ribrault, C. et al. Syntaxin1A lateral diffusion reveals transient and local SNARE interactions. J. Neurosci. 31, 17590–17602 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Knowles, M. K. et al. Single secretory granules of live cells recruit syntaxin-1 and synaptosomal associated protein 25 (SNAP-25) in large copy numbers. Proc. Natl Acad. Sci. USA 107, 20810–20815 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Tang, A. H. et al. A trans-synaptic nanocolumn aligns neurotransmitter release to receptors. Nature 536, 210–214 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Joensuu, M. et al. Subdiffractional tracking of internalized molecules reveals heterogeneous motion states of synaptic vesicles. J. Cell Biol. 215, 277–292 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Joensuu, M. et al. Visualizing endocytic recycling and trafficking in live neurons by subdiffractional tracking of internalized molecules. Nat. Protoc. 12, 2590–2622 (2017).

    CAS  PubMed  Google Scholar 

  128. Querol-Vilaseca, M. et al. Nanoscale structure of amyloid-beta plaques in Alzheimer’s disease. Sci. Rep. 9, 5181 (2019).

    PubMed  PubMed Central  Google Scholar 

  129. Codron, P. et al. STochastic Optical Reconstruction Microscopy (STORM) reveals the nanoscale organization of pathological aggregates in human brain. Neuropathol. Appl. Neurobiol. 47, 127–142 (2020).

    PubMed  PubMed Central  Google Scholar 

  130. Westphal, V. et al. Video-rate far-field optical nanoscopy dissects synaptic vesicle movement. Science 320, 246–249 (2008).

    CAS  PubMed  Google Scholar 

  131. Gramlich, M. W. & Klyachko, V. A. Actin/myosin-V- and activity-dependent inter-synaptic vesicle exchange in central neurons. Cell Rep. 18, 2096–2104 (2017).

    CAS  PubMed  Google Scholar 

  132. Forte, L. A., Gramlich, M. W. & Klyachko, V. A. Activity-dependence of synaptic vesicle dynamics. J. Neurosci. 37, 10597–10610 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Maschi, D. & Klyachko, V. A. Spatiotemporal dynamics of multi-vesicular release is determined by heterogeneity of release sites within central synapses. eLife 9, e55210 (2020).

    PubMed  PubMed Central  Google Scholar 

  134. Maschi, D. & Klyachko, V. A. Spatiotemporal regulation of synaptic vesicle fusion sites in central synapses. Neuron 94, 65–73 e63 (2017).

    CAS  PubMed  Google Scholar 

  135. Lu, H. E., MacGillavry, H. D., Frost, N. A. & Blanpied, T. A. Multiple spatial and kinetic subpopulations of CaMKII in spines and dendrites as resolved by single-molecule tracking PALM. J. Neurosci. 34, 7600–7610 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Mehidi, A. et al. Transient activations of Rac1 at the lamellipodium tip trigger membrane protrusion. Curr. Biol. 29, 2852–2866 e2855 (2019).

    CAS  PubMed  Google Scholar 

  137. Garcia-Parajo, M. F., Cambi, A., Torreno-Pina, J. A., Thompson, N. & Jacobson, K. Nanoclustering as a dominant feature of plasma membrane organization. J. Cell Sci. 127, 4995–5005 (2014).

    PubMed  PubMed Central  Google Scholar 

  138. Harding, A. S. & Hancock, J. F. Using plasma membrane nanoclusters to build better signaling circuits. Trends Cell Biol. 18, 364–371 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  139. Xia, D., Li, C. & Götz, J. Pseudophosphorylation of Tau at distinct epitopes or the presence of the P301L mutation targets the microtubule-associated protein Tau to dendritic spines. Biochim. Biophys. Acta 1852, 913–924 (2015).

    CAS  PubMed  Google Scholar 

  140. Bhaskar, K., Yen, S. H. & Lee, G. Disease-related modifications in Tau affect the interaction between Fyn and Tau. J. Biol. Chem. 280, 35119–35125 (2005).

    CAS  PubMed  Google Scholar 

  141. Viana da Silva, S. et al. Early synaptic deficits in the APP/PS1 mouse model of Alzheimer’s disease involve neuronal adenosine A2A receptors. Nat. Commun. 7, 11915 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Siskova, Z. et al. Dendritic structural degeneration is functionally linked to cellular hyperexcitability in a mouse model of Alzheimer’s disease. Neuron 84, 1023–1033 (2014).

    CAS  PubMed  Google Scholar 

  143. Kommaddi, R. P. et al. Abeta mediates F-actin disassembly in dendritic spines leading to cognitive deficits in Alzheimer’s disease. J. Neurosci. 38, 1085–1099 (2018). This study provides insight into Aβ-mediated changes in actin organization in spines.

    CAS  PubMed  PubMed Central  Google Scholar 

  144. Nanguneri, S. et al. Characterization of nanoscale organization of f-actin in morphologically distinct dendritic spines in vitro using supervised learning. eNeuro https://doi.org/10.1523/ENEURO.0425-18.2019 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  145. Kommaddi, R. P. et al. Glutaredoxin1 diminishes amyloid beta-mediated oxidation of F-actin and reverses cognitive deficits in an Alzheimer’s disease mouse model. Antioxid. Redox Signal. 31, 1321–1338 (2019).

    CAS  PubMed  Google Scholar 

  146. Dani, A., Huang, B., Bergan, J., Dulac, C. & Zhuang, X. Superresolution imaging of chemical synapses in the brain. Neuron 68, 843–856 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  147. Ferreira, J. S. et al. Distance-dependent regulation of NMDAR nanoscale organization along hippocampal neuron dendrites. Proc. Natl Acad. Sci. USA 117, 24526–24533 (2020). This study shows that the proximal and distal dendritic segments have different NMDAR nanoscale organizations.

    CAS  PubMed  PubMed Central  Google Scholar 

  148. Jarosz-Griffiths, H. H., Noble, E., Rushworth, J. V. & Hooper, N. M. Amyloid-beta receptors: the good, the bad, and the prion protein. J. Biol. Chem. 291, 3174–3183 (2016).

    CAS  PubMed  Google Scholar 

  149. Gomez-Ramos, A., Diaz-Hernandez, M., Rubio, A., Miras-Portugal, M. T. & Avila, J. Extracellular Tau promotes intracellular calcium increase through M1 and M3 muscarinic receptors in neuronal cells. Mol. Cell Neurosci. 37, 673–681 (2008).

    CAS  PubMed  Google Scholar 

  150. Rauch, J. N. et al. LRP1 is a master regulator of Tau uptake and spread. Nature 580, 381–385 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  151. Shrivastava, A. N. et al. Clustering of Tau fibrils impairs the synaptic composition of alpha3-Na+/K+-ATPase and AMPA receptors. EMBO J. 38, e99871 (2019). This study demonstrates the lateral diffusion and clustering of extracellular tau fibrils on the plasma membrane.

    PubMed  PubMed Central  Google Scholar 

  152. Renner, M. et al. Deleterious effects of amyloid beta oligomers acting as an extracellular scaffold for mGluR5. Neuron 66, 739–754 (2010). This publication shows the clustering of extracellular Aβos on the plasma membrane accompanied by a concomitant increase in mGluR5 clustering.

    CAS  PubMed  PubMed Central  Google Scholar 

  153. Calamai, M. & Pavone, F. S. Single molecule tracking analysis reveals that the surface mobility of amyloid oligomers is driven by their conformational structure. J. Am. Chem. Soc. 133, 12001–12008 (2011). This work reveals that the mobility patterns of prefibrillar and fibrillar forms of Aβos are different.

    CAS  PubMed  Google Scholar 

  154. Calamai, M. et al. Single molecule experiments emphasize GM1 as a key player of the different cytotoxicity of structurally distinct Abeta1-42 oligomers. Biochim. Biophys. Acta 1858, 386–392 (2016).

    CAS  PubMed  Google Scholar 

  155. Hafner, A. S., Donlin-Asp, P. G., Leitch, B., Herzog, E. & Schuman, E. M. Local protein synthesis is a ubiquitous feature of neuronal pre- and postsynaptic compartments. Science 364, eaau3644 (2019). This study uses expansion microscopy and provides evidence that both presynaptic and postsynaptic compartments contain mRNA molecules and translational machinery necessary for protein synthesis.

    CAS  PubMed  Google Scholar 

  156. Younts, T. J. et al. Presynaptic protein synthesis Is required for long-term plasticity of GABA release. Neuron 92, 479–492 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Scarnati, M. S., Kataria, R., Biswas, M. & Paradiso, K. G. Active presynaptic ribosomes in the mammalian brain, and altered transmitter release after protein synthesis inhibition. eLife 7, e36697 (2018).

    PubMed  PubMed Central  Google Scholar 

  158. Donlin-Asp, P. G., Polisseni, C., Klimek, R., Heckel, A. & Schuman, E. M. Differential regulation of local mRNA dynamics and translation following long-term potentiation and depression. Proc. Natl Acad. Sci. USA 118, e2017578118 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  159. Rangaraju, V., Lauterbach, M. & Schuman, E. M. Spatially stable mitochondrial compartments fuel local translation during plasticity. Cell 176, 73–84 e15 (2019). This study describes stable dendritic mitochondrial compartments providing energy for synaptic translation.

    CAS  PubMed  Google Scholar 

  160. Evans, H. T., Benetatos, J., van Roijen, M., Bodea, L. G. & Götz, J. Decreased synthesis of ribosomal proteins in tauopathy revealed by non-canonical amino acid labelling. EMBO J. 38, e101174 (2019).

    PubMed  PubMed Central  Google Scholar 

  161. Haass, C., Kaether, C., Thinakaran, G. & Sisodia, S. Trafficking and proteolytic processing of APP. Cold Spring Harb. Perspect. Med. 2, a006270 (2012).

    PubMed  PubMed Central  Google Scholar 

  162. Lazarov, O., Lee, M., Peterson, D. A. & Sisodia, S. S. Evidence that synaptically released beta-amyloid accumulates as extracellular deposits in the hippocampus of transgenic mice. J. Neurosci. 22, 9785–9793 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  163. Wei, W. et al. Amyloid beta from axons and dendrites reduces local spine number and plasticity. Nat. Neurosci. 13, 190–196 (2010).

    CAS  PubMed  Google Scholar 

  164. Bero, A. W. et al. Neuronal activity regulates the regional vulnerability to amyloid-beta deposition. Nat. Neurosci. 14, 750–756 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  165. Cirrito, J. R. et al. Synaptic activity regulates interstitial fluid amyloid-beta levels in vivo. Neuron 48, 913–922 (2005).

    CAS  PubMed  Google Scholar 

  166. Kamenetz, F. et al. APP processing and synaptic function. Neuron 37, 925–937 (2003).

    CAS  PubMed  Google Scholar 

  167. Cirrito, J. R. et al. Endocytosis is required for synaptic activity-dependent release of amyloid-beta in vivo. Neuron 58, 42–51 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  168. Buckner, R. L. et al. Molecular, structural, and functional characterization of Alzheimer’s disease: evidence for a relationship between default activity, amyloid, and memory. J. Neurosci. 25, 7709–7717 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  169. Escamilla-Ayala, A. A. et al. Super-resolution microscopy reveals majorly mono- and dimeric presenilin1/γ-secretase at the cell surface. eLife 9, e56679 (2020). This work describes the single-molecule dynamics of PSEN1/γ-secretase at the plasma membrane.

    CAS  PubMed  PubMed Central  Google Scholar 

  170. Schedin-Weiss, S., Caesar, I., Winblad, B., Blom, H. & Tjernberg, L. O. Super-resolution microscopy reveals gamma-secretase at both sides of the neuronal synapse. Acta Neuropathol. Commun. 4, 29 (2016). This stimulated emission depletion microscopy study indicates the presence of γ-secretase at both the presynapse and the postsynapse.

    PubMed  PubMed Central  Google Scholar 

  171. Yu, Y., Jans, D. C., Winblad, B., Tjernberg, L. O. & Schedin-Weiss, S. Neuronal Abeta42 is enriched in small vesicles at the presynaptic side of synapses. Life Sci. Alliance 1, e201800028 (2018).

    PubMed  PubMed Central  Google Scholar 

  172. de Coninck, D., Schmidt, T. H., Schloetel, J. G. & Lang, T. Packing density of the amyloid precursor protein in the cell membrane. Biophys. J. 114, 1128–1141 (2018).

    PubMed  PubMed Central  Google Scholar 

  173. Kedia, S. et al. Alteration in synaptic nanoscale organization dictates amyloidogenic processing in Alzheimer’s disease. iScience 24, 101924 (2021).

    CAS  PubMed  Google Scholar 

  174. Kedia, S. et al. Real-time nanoscale organization of amyloid precursor protein. Nanoscale 12, 8200–8215 (2020). This study reveals the organization of APP into nanodomains at the postsynapse.

    CAS  PubMed  Google Scholar 

  175. Rice, H. C. et al. Secreted amyloid-β precursor protein functions as a GABABR1a ligand to modulate synaptic transmission. Science 363, eaao4827 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  176. Polanco, J. C., Li, C., Durisic, N., Sullivan, R. & Götz, J. Exosomes taken up by neurons hijack the endosomal pathway to spread to interconnected neurons. Acta Neuropathol. Commun. 6, 10 (2018).

    PubMed  PubMed Central  Google Scholar 

  177. Wang, Y. et al. The release and trans-synaptic transmission of Tau via exosomes. Mol. Neurodegener. 12, 5 (2017).

    PubMed  PubMed Central  Google Scholar 

  178. Dujardin, S. et al. Ectosomes: a new mechanism for non-exosomal secretion of tau protein. PLoS ONE 9, e100760 (2014).

    PubMed  PubMed Central  Google Scholar 

  179. Abounit, S., Wu, J. W., Duff, K., Victoria, G. S. & Zurzolo, C. Tunneling nanotubes: a possible highway in the spreading of tau and other prion-like proteins in neurodegenerative diseases. Prion 10, 344–351 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  180. Merezhko, M. et al. Secretion of Tau via an unconventional non-vesicular mechanism. Cell Rep. 25, 2027–2035 e2024 (2018).

    CAS  PubMed  Google Scholar 

  181. Katsinelos, T. et al. Unconventional secretion mediates the trans-cellular spreading of Tau. Cell Rep. 23, 2039–2055 (2018).

    CAS  PubMed  Google Scholar 

  182. Calafate, S. et al. Synaptic contacts enhance cell-to-cell tau pathology propagation. Cell Rep. 11, 1176–1183 (2015).

    CAS  PubMed  Google Scholar 

  183. de Calignon, A. et al. Propagation of tau pathology in a model of early Alzheimer’s disease. Neuron 73, 685–697 (2012).

    PubMed  PubMed Central  Google Scholar 

  184. Brunello, C. A., Merezhko, M., Uronen, R. L. & Huttunen, H. J. Mechanisms of secretion and spreading of pathological Tau protein. Cell Mol. Life Sci. 77, 1721–1744 (2020).

    CAS  PubMed  Google Scholar 

  185. Chai, X., Dage, J. L. & Citron, M. Constitutive secretion of Tau protein by an unconventional mechanism. Neurobiol. Dis. 48, 356–366 (2012).

    CAS  PubMed  Google Scholar 

  186. Pooler, A. M., Phillips, E. C., Lau, D. H., Noble, W. & Hanger, D. P. Physiological release of endogenous Tau is stimulated by neuronal activity. EMBO Rep. 14, 389–394 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  187. Michel, C. H. et al. Extracellular monomeric Tau protein is sufficient to initiate the spread of Tau protein pathology. J. Biol. Chem. 289, 956–967 (2014).

    CAS  PubMed  Google Scholar 

  188. Polanco, J. C., Hand, G. R., Briner, A., Li, C. & Götz, J. Exosomes induce endolysosomal permeabilization as a gateway by which exosomal tau seeds escape into the cytosol. Acta Neuropathol. 141, 235–256 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  189. Groc, L. & Choquet, D. Linking glutamate receptor movements and synapse function. Science 368, eaay4631 (2020). This article provides an excellent overview of receptor movement and trafficking in synapses.

    CAS  PubMed  Google Scholar 

  190. Willems, J. et al. ORANGE: a CRISPR/Cas9-based genome editing toolbox for epitope tagging of endogenous proteins in neurons. PLoS Biol. 18, e3000665 (2020).

    PubMed  PubMed Central  Google Scholar 

  191. Inavalli, V. et al. A super-resolution platform for correlative live single-molecule imaging and STED microscopy. Nat. Methods 16, 1263–1268 (2019). This study introduces a powerful imaging platform that combines complementary super-resolution imaging techniques.

    CAS  PubMed  Google Scholar 

  192. Yasui, M., Hiroshima, M., Kozuka, J., Sako, Y. & Ueda, M. Automated single-molecule imaging in living cells. Nat. Commun. 9, 3061 (2018).

    PubMed  PubMed Central  Google Scholar 

  193. Beghin, A. et al. Localization-based super-resolution imaging meets high-content screening. Nat. Methods 14, 1184–1190 (2017).

    CAS  PubMed  Google Scholar 

  194. Schneider, R. et al. Mobility of calcium channels in the presynaptic membrane. Neuron 86, 672–679 (2015).

    CAS  PubMed  Google Scholar 

  195. Park, H., Li, Y. & Tsien, R. W. Influence of synaptic vesicle position on release probability and exocytotic fusion mode. Science 335, 1362–1366 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  196. Padmanabhan, P., Garaigorta, U. & Dixit, N. M. Emergent properties of the interferon-signalling network may underlie the success of hepatitis C treatment. Nat. Commun. 5, 3872 (2014).

    CAS  PubMed  Google Scholar 

  197. Richter, M. C. et al. Distinct in vivo roles of secreted APP ectodomain variants APPsα and APPsβ in regulation of spine density, synaptic plasticity, and cognition. EMBO J. 37 (2018).

  198. Zhang, Z. et al. δ-Secretase-cleaved Tau stimulates Aβ production via upregulating STAT1-BACE1 signaling in Alzheimer’s disease. Mol. Psychiatry 26, 586–603 (2021).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  200. Kollmer, M. et al. Cryo-EM structure and polymorphism of Aβ amyloid fibrils purified from Alzheimer’s brain tissue. Nat. Commun. 10, 4760 (2019).

    PubMed  PubMed Central  Google Scholar 

  201. 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 

  202. Latif-Hernandez, A. et al. The two faces of synaptic failure in AppNL-G-F knock-in mice. Alzheimers Res. Ther. 12, 100 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  203. Götz, J., Bodea, L. G. & Goedert, M. Rodent models for Alzheimer disease. Nat. Rev. Neurosci. 19, 583–598 (2018).

    PubMed  Google Scholar 

  204. Berry, B. J., Smith, A. S. T., Young, J. E. & Mack, D. L. Advances and current challenges associated with the use of human induced pluripotent stem cells in modeling neurodegenerative disease. Cell Tissues Organs 205, 331–349 (2018).

    CAS  Google Scholar 

  205. Wegel, E. et al. Imaging cellular structures in super-resolution with SIM, STED and localisation microscopy: a practical comparison. Sci. Rep. 6, 27290 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  206. Schermelleh, L. et al. Super-resolution microscopy demystified. Nat. Cell Biol. 21, 72–84 (2019).

    CAS  PubMed  Google Scholar 

  207. Lu, M. et al. Structural progression of amyloid-beta Arctic mutant aggregation in cells revealed by multiparametric imaging. J. Biol. Chem. 294, 1478–1487 (2019).

    CAS  PubMed  Google Scholar 

  208. Ball, G., Parton, R. M., Hamilton, R. S. & Davis, I. A cell biologist’s guide to high resolution imaging. Methods Enzymol. 504, 29–55 (2012).

    CAS  PubMed  Google Scholar 

  209. Yuan, P. et al. TREM2 haplodeficiency in mice and humans impairs the microglia barrier function leading to decreased amyloid compaction and severe axonal dystrophy. Neuron 92, 252–264 (2016). This study uses stochastic optical reconstruction microscopy and provides structural insights into Aβ plaques.

    CAS  PubMed  Google Scholar 

  210. Khater, I. M., Nabi, I. R. & Hamarneh, G. A review of super-resolution single-molecule localization microscopy cluster analysis and quantification methods. Patterns 1, 100038 (2020).

    PubMed  PubMed Central  Google Scholar 

  211. Maglione, M. & Sigrist, S. J. Seeing the forest tree by tree: super-resolution light microscopy meets the neurosciences. Nat. Neurosci. 16, 790–797 (2013).

    CAS  PubMed  Google Scholar 

  212. Sydor, A. M., Czymmek, K. J., Puchner, E. M. & Mennella, V. Super-resolution microscopy: from single molecules to supramolecular assemblies. Trends Cell Biol. 25, 730–748 (2015).

    CAS  PubMed  Google Scholar 

  213. Nicovich, P. R., Owen, D. M. & Gaus, K. Turning single-molecule localization microscopy into a quantitative bioanalytical tool. Nat. Protoc. 12, 453–460 (2017).

    CAS  PubMed  Google Scholar 

  214. Spehar, K. et al. Super-resolution imaging of amyloid structures over extended times by using transient binding of single thioflavin T molecules. Chembiochem 19, 1944–1948 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  215. Cosentino, M., Canale, C., Bianchini, P. & Diaspro, A. AFM-STED correlative nanoscopy reveals a dark side in fluorescence microscopy imaging. Sci. Adv. 5, eaav8062 (2019).

    PubMed  PubMed Central  Google Scholar 

  216. Xu, S. C. S. et al. Sequence-independent recognition of the amyloid structural motif by GFP protein family. Proc. Natl Acad. Sci. USA 117, 22122–22127 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  217. Valades Cruz, C. A. et al. Quantitative nanoscale imaging of orientational order in biological filaments by polarized superresolution microscopy. Proc. Natl Acad. Sci. USA 113, E820–E828 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  218. Shaban, H. A., Valades-Cruz, C. A., Savatier, J. & Brasselet, S. Polarized super-resolution structural imaging inside amyloid fibrils using thioflavine T. Sci. Rep. 7, 12482 (2017).

    PubMed  PubMed Central  Google Scholar 

  219. Kaminski Schierle, G. S. et al. In situ measurements of the formation and morphology of intracellular beta-amyloid fibrils by super-resolution fluorescence imaging. J. Am. Chem. Soc. 133, 12902–12905 (2011).

    CAS  PubMed  Google Scholar 

  220. Esbjorner, E. K. et al. Direct observations of amyloid beta self-assembly in live cells provide insights into differences in the kinetics of Abeta(1-40) and Abeta(1-42) aggregation. Chem. Biol. 21, 732–742 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  221. Mlodzianoski, M. J. et al. Active PSF shaping and adaptive optics enable volumetric localization microscopy through brain sections. Nat. Methods 15, 583–586 (2018). This super-resolution study reconstructs the three-dimensional structure of amyloid plaques in a mouse model of AD.

    CAS  PubMed  PubMed Central  Google Scholar 

  222. Jonsson, T. et al. Variant of TREM2 associated with the risk of Alzheimer’s disease. N. Engl. J. Med. 368, 107–116 (2013).

    CAS  PubMed  Google Scholar 

  223. Zhang, W. I. et al. Super-resolution microscopy of cerebrospinal fluid biomarkers as a tool for Alzheimer’s disease diagnostics. J. Alzheimers Dis. 46, 1007–1020 (2015).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank S. Lesné and V. Anggono for helpful feedback on Supplementary Fig. 1, N. M. Dixit for critical feedback on Fig. 4, R. Amor for helpful suggestions for the figure in Box 2, N. Valmas for the graphical artwork in Fig. 1a and R. Tweedale and A. Briner for critical reading of the manuscript. The authors acknowledge support by the Estate of Clem Jones, the State Government of Queensland, the Federal Government of Australia, the Australian Research Council (DP190102789) and the National Health and Medical Research Council of Australia (GNT1176326 and GNT1147569) to J.G.

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to all aspects of the article

Corresponding authors

Correspondence to Pranesh Padmanabhan or Jürgen Götz.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Neuroscience thanks M. Fuhrmann, who co-reviewed with S. Poll, D. Nair and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Glossary

Alzheimer disease

(AD). A progressive neurodegenerative disease that is characterized by the loss of memory and other cognitive functions.

Amyloid plaques

Major hallmark brain lesions of patients with Alzheimer disease composed of oligomeric and fibrillar forms of the amyloid-β peptide.

Neurofibrillary tangles

A critical hallmark of Alzheimer disease comprising aggregated forms of the microtubule-associated protein tau.

Super-resolution microscopy

(SRM). A collection of techniques that has overcome the diffraction limit of light and allows quantitative understanding of molecular and cellular processes with unprecedented spatio-temporal detail.

Dendritic spine

A micron-sized subcellular structure protruding from dendritic branches of neurons that houses most of the excitatory synapses in the mammalian CNS.

Synaptic plasticity

A neural activity-dependent process leading to strengthening or weakening of synaptic connections.

Presynapse

A subcompartment of axons or at the distal end of axons where synaptic vesicles fuse to release neurotransmitters.

Nanodomains

Discrete, nanometre-sized regions where diffusing molecules are trapped and concentrated.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Padmanabhan, P., Kneynsberg, A. & Götz, J. Super-resolution microscopy: a closer look at synaptic dysfunction in Alzheimer disease. Nat Rev Neurosci 22, 723–740 (2021). https://doi.org/10.1038/s41583-021-00531-y

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41583-021-00531-y

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