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.

  • Article
  • Published:

Towards non-invasive diagnostic imaging of early-stage Alzheimer's disease

Subjects

Abstract

One way to image the molecular pathology in Alzheimer's disease is by positron emission tomography using probes that target amyloid fibrils. However, these fibrils are not closely linked to the development of the disease. It is now thought that early-stage biomarkers that instigate memory loss are composed of Aβ oligomers. Here, we report a sensitive molecular magnetic resonance imaging contrast probe that is specific for Aβ oligomers. We attach oligomer-specific antibodies onto magnetic nanostructures and show that the complex is stable and binds to Aβ oligomers on cells and brain tissues to give a magnetic resonance imaging signal. When intranasally administered to an Alzheimer's disease mouse model, the probe readily reached hippocampal Aβ oligomers. In isolated samples of human brain tissue, we observed a magnetic resonance imaging signal that distinguished Alzheimer's disease from controls. Such nanostructures that target neurotoxic Aβ oligomers are potentially useful for evaluating the efficacy of new drugs and ultimately for early-stage Alzheimer's disease diagnosis and disease management.

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

Figure 1: Aβ oligomers (AβOs) bind to neuronal surfaces in a saturable, receptor-mediated manner and are distinct from amyloid plaques.
Figure 2: Individual components of the NU4MNS probe.
Figure 3: NU4MNS attachment to hippocampal neurons is specific to AβOs.
Figure 4: NU4 and NU4MNS discriminate Alzheimer's disease human frontal cortex sections from aged controls.
Figure 5: NU4 antibody detects dendrite-bound AβOs in fixed tissue and binds its target within 4 h following intranasal inoculation.
Figure 6: NU4MNS labelling of human and Tg mouse brain slices gives a pronounced, Alzheimer's disease-dependent MRI signal that is confirmed in vivo.

Similar content being viewed by others

References

  1. 2014 Alzheimer's disease facts and figures. Alzheimers Dement. 10, e47–e92 (2014).

  2. 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).

    Article  CAS  Google Scholar 

  3. Toledo, J. B., Xie, S. X., Trojanowski, J. Q. & Shaw, L. M. Longitudinal change in CSF τ and Aβ biomarkers for up to 48 months in ADNI. Acta Neuropathol. 126, 659–670 (2013).

    Article  CAS  Google Scholar 

  4. Slemmon, J. R. et al. Measurement of Aβ1–42 in cerebrospinal fluid is influenced by matrix effects. J. Neurochem. 120, 325–333 (2012).

    Article  CAS  Google Scholar 

  5. Klunk, W. E. et al. Imaging brain amyloid in Alzheimer's disease with Pittsburgh compound-B. Annals Neurol. 55, 306–319 (2004).

    Article  CAS  Google Scholar 

  6. Johnson, K. A. et al. Appropriate use criteria for amyloid PET: a report of the Amyloid Imaging Task Force, the Society of Nuclear Medicine and Molecular Imaging, and the Alzheimer's Association. Alzheimers Dement. 9, e1–e16 (2013).

    Article  Google Scholar 

  7. Terry, R. D. et al. Physical basis of cognitive alterations in Alzheimer's disease: synapse loss is the major correlate of cognitive impairment. Ann. Neurol. 30, 572–580 (1991).

    Article  CAS  Google Scholar 

  8. Nyborg, A. C. et al. In vivo and ex vivo imaging of amyloid-beta cascade aggregates with a pronucleon peptide. J. Alzheimer's Dis. 34, 957–967 (2013).

    Article  CAS  Google Scholar 

  9. Klein, W. L., Krafft, G. A. & Finch, C. E. Targeting small Aβ oligomers: the solution to an Alzheimer's disease conundrum?. Trends Neurosci. 24, 219–224 (2001).

    Article  CAS  Google Scholar 

  10. Lublin, A. L. & Gandy, S. Amyloid-β oligomers: possible roles as key neurotoxins in Alzheimer's disease. Mount Sinai J. Med. New York 77, 43–49 (2010).

    Article  Google Scholar 

  11. Ferreira, S. T. & Klein, W. L. The Aβ oligomer hypothesis for synapse failure and memory loss in Alzheimer's disease. Neurobiol. Learn. Mem. 96, 529–543 (2011).

    Article  CAS  Google Scholar 

  12. Schnabel, J. Amyloid little proteins, big clues. Nature 475, S12–S14 (2011).

    Article  CAS  Google Scholar 

  13. Mucke, L. & Selkoe, D. J. Neurotoxicity of amyloid β-protein: synaptic and network dysfunction. Cold Spring Harbor Persp. Med. 2, a006338 (2012).

    Google Scholar 

  14. Lacor, P. N. et al. Synaptic targeting by Alzheimer's-related amyloid β oligomers. J. Neurosci. 24, 10191–10200 (2004).

    Article  CAS  Google Scholar 

  15. De, M., Chou, S. S., Joshi, H. M. & Dravid, V. P. Hybrid magnetic nanostructures (MNS) for magnetic resonance imaging applications. Adv. Drug Deliv. Rev. 63, 1282–1299 (2011).

    Article  CAS  Google Scholar 

  16. Corot, C. et al. in Molecular and Cellular MR Imaging (eds Modo, M. M. J. & Bulte, J. W. M.) Ch. 4, 59–84 (CRC Press, 2007).

  17. Liu, S. et al. A novel type of dual-modality molecular probe for MR and nuclear imaging of tumor: preparation, characterization and in vivo application. Mol. Pharmaceut. 6, 1074–1082 (2009).

    Article  CAS  Google Scholar 

  18. Serres, S. et al. Molecular MRI enables early and sensitive detection of brain metastases. Proc. Natl Acad. Sci. USA 109, 6674–6679 (2012).

    Article  CAS  Google Scholar 

  19. Lambert, M. P. et al. Monoclonal antibodies that target pathological assemblies of Aβ. J. Neurochem. 100, 23–35 (2007).

    Article  CAS  Google Scholar 

  20. Lambert, M. P., Velasco, P. T., Viola, K. L. & Klein, W. L. Targeting generation of antibodies specific to conformational epitopes of amyloid β-derived neurotoxins. CNS Neurol. Disord. Drug Targets 8, 65–81 (2009).

    Article  CAS  Google Scholar 

  21. Acton, P. et al. Anti-ADDL antibodies and uses thereof. US patent 7,811,563 (2010).

  22. Xiao, C. et al. Brain transit and ameliorative effects of intranasally delivered anti-amyloid-β oligomer antibody in 5xFAD mice. J. Alzheimer's Dis. 35, 777–788 (2013).

    Article  Google Scholar 

  23. Gong, Y. et al. Alzheimer's disease-affected brain: presence of oligomeric Aβ ligands (ADDLs) suggests a molecular basis for reversible memory loss. Proc. Natl Acad. Sci. USA 100, 10417–10422 (2003).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  25. Chang, L. et al. Femtomole immunodetection of synthetic and endogenous amyloid-β oligomers and its application to Alzheimer's disease drug candidate screening. J. Mol. Neurosci. 20, 305–313 (2003).

    Article  CAS  Google Scholar 

  26. Lesne, S. et al. A specific amyloid-β protein assembly in the brain impairs memory. Nature 440, 352–357 (2006).

    Article  CAS  Google Scholar 

  27. Ohno, M. et al. Temporal memory deficits in Alzheimer's mouse models: rescue by genetic deletion of BACE1. Eur. J. Neurosci. 23, 251–260 (2006).

    Article  Google Scholar 

  28. Mucke, L. et al. High-level neuronal expression of Aβ1–42 in wild-type human amyloid protein precursor transgenic mice: synaptotoxicity without plaque formation. J. Neurosci. 20, 4050–4058 (2000).

    Article  CAS  Google Scholar 

  29. Balducci, C. et al. Cognitive deficits associated with alteration of synaptic metaplasticity precede plaque deposition in AβPP23 transgenic mice. J. Alzheimer's Dis. 21, 1367–1381 (2010).

    Article  CAS  Google Scholar 

  30. Ferretti, M. T. et al. Transgenic mice as a model of pre-clinical Alzheimer's disease. Curr. Alzheimer Res. 8, 4–23 (2011).

    Article  CAS  Google Scholar 

  31. Takamura, A. et al. Extracellular and intraneuronal HMW-AβOs represent a molecular basis of memory loss in Alzheimer's disease model mouse. Mol. Neurodegen. 6, 20 (2011).

    Article  CAS  Google Scholar 

  32. Velasco, P. T. et al. Synapse-binding subpopulations of Aβ oligomers sensitive to peptide assembly blockers and scFv antibodies. ACS Chem. Neurosci. 3, 972–981 (2012).

    Article  CAS  Google Scholar 

  33. Oakley, H. et al. Intraneuronal β-amyloid aggregates, neurodegeneration, and neuron loss in transgenic mice with five familial Alzheimer's disease mutations: potential factors in amyloid plaque formation. J. Neurosci. 26, 10129–10140 (2006).

    Article  CAS  Google Scholar 

  34. Barick, K. C. et al. Nanoscale assembly of amine-functionalized colloidal iron oxide. J. Magn. Magn. Mater. 321, 1529–1532 (2009).

    Article  CAS  Google Scholar 

  35. Na, H. B., Song, I. C. & Hyeon, T. Inorganic nanoparticles for MRI contrast agents. Adv. Mater. 21, 2133–2148 (2009).

    Article  CAS  Google Scholar 

  36. Pinho, S. L. C. et al. Fine tuning of the relaxometry of γ-Fe2O3@SiO2 nanoparticles by tweaking the silica coating thickness. ACS Nano. 4, 5339–5349 (2010).

    Article  CAS  Google Scholar 

  37. Prakash, A. et al. Bilayers as phase transfer agents for nanocrystals prepared in nonpolar solvents. ACS Nano. 3, 2139–2146 (2009).

    Article  CAS  Google Scholar 

  38. Amstad, E., Textor, M. & Reimhult, E. Stabilization and functionalization of iron oxide nanoparticles for biomedical applications. Nanoscale 3, 2819–2843 (2011).

    Article  CAS  Google Scholar 

  39. Tong, S. et al. Coating optimization of superparamagnetic iron oxide nanoparticles for high T2 relaxivity. Nano Lett. 10, 4607–4613 (2010).

    Article  CAS  Google Scholar 

  40. Amstad, E. et al. Ultrastable iron oxide nanoparticle colloidal suspensions using dispersants with catechol-derived anchor groups. Nano Lett. 9, 4042–4048 (2009).

    Article  CAS  Google Scholar 

  41. Palumbo, A., Napolitano, A., Barone, P. & d'Ischia, M. Nitrite- and peroxide-dependent oxidation pathways of dopamine: 6-nitrodopamine and 6-hydroxydopamine formation as potential contributory mechanisms of oxidative stress- and nitric oxide induced neurotoxicity in neuronal degeneration. Chem. Res. Toxicol. 12, 1213–1222 (1999).

    Article  CAS  Google Scholar 

  42. Robinson, R. MRI probe for amyloid-β oligomers offers potential advantages for detecting Alzheimer's disease. Neurol. Today 12, 20–22 (2012).

    Article  Google Scholar 

  43. Park, J. et al. Ultra-large-scale syntheses of monodisperse nanocrystals. Nature Mater. 3, 891–895 (2004).

    Article  CAS  Google Scholar 

  44. Mundt, A. P. et al. Targeting activated microglia in Alzheimer's pathology by intraventricular delivery of a phagocytosable MRI contrast agent in APP23 transgenic mice. NeuroImage 46, 367–372 (2009).

    Article  Google Scholar 

Download references

Acknowledgements

The authors thank the staff of the Biological Imaging Facility (BIF), the Center for Advanced Molecular Imaging (CAMI) and Northwestern University's Atomic and Nanoscale Characterization Experimental Center (NUANCE) at Northwestern University, and the Department of Radiology at NorthShore University HealthSystems. The authors also thank the Northwestern Brain Bank at the Cognitive Neurology and Alzheimer's Disease Center (CNADC) for providing the human Alzheimer's disease and control brain tissue. The authors acknowledge support from their funding agencies and grants. This work was funded by the National Institutes of Health (AG022547, AG029460 and AG045637 to W.L.K.), by Baxter Healthcare (to W.L.K.) and partially by the National Institutes of Health–Centers of Cancer Nanotechnology Excellence through the Nanoconstruct Core (award U54CA119341 to V.P.D., H.J. and M.D.)

Author information

Authors and Affiliations

Authors

Contributions

The study concept and design was provided by K.L.V., J.S., R.S., M.D., V.P.D. and W.L.K. The acquisition of data was performed by M.D., H.J., J.S. and S.V. (MNS development, production, and characterization), K.L.V., J.S., R.S., M.A.B., J.W., S.V., S.S., S.W., C.L. and J.P. (antibody conjugation, cell experiments, tissue experiments and immunoprecipitation experiments), K.L.V. and M.A.B. (animal experiments), K.M. (inductively coupled plasma mass spectrometry experiments) and E.A.W. and P.P (magnetic resonance image acquisition). All authors discussed the results and contributed to the analysis of the data. Critical revision of the article for intellectual content was conducted by K.L.V., J.S., R.S., M.D., P.T.V., V.P.D. and W.L.K. Funding was obtained and studies supervised by W.L.K. and V.P.D.

Corresponding authors

Correspondence to Vinayak P. Dravid or William L. Klein.

Ethics declarations

Competing interests

This work was funded, in part, by Baxter Healthcare. Northwestern University holds the rights to two US and several international patents concerning antibodies that target Aβ oligomer assemblies. Acumen Pharmaceuticals holds the licensing rights to develop anti-Aβ oligomer antibodies for therapeutic use.

Supplementary information

Supplementary information

Supplementary Information (PDF 6608 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Viola, K., Sbarboro, J., Sureka, R. et al. Towards non-invasive diagnostic imaging of early-stage Alzheimer's disease. Nature Nanotech 10, 91–98 (2015). https://doi.org/10.1038/nnano.2014.254

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nnano.2014.254

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