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:

Near-infrared fluorescence lifetime imaging of amyloid-β aggregates and tau fibrils through the intact skull of mice

Abstract

Non-invasive methods for the in vivo detection of hallmarks of Alzheimer’s disease can facilitate the study of the progression of the disease in mouse models and may enable its earlier diagnosis in humans. Here we show that the zwitterionic heptamethine fluorophore ZW800-1C, which has peak excitation and emission wavelengths in the near-infrared optical window, binds in vivo and at high contrast to amyloid-β deposits and to neurofibrillary tangles, and allows for the microscopic imaging of amyloid-β and tau aggregates through the intact skull of mice. In transgenic mouse models of Alzheimer’s disease, we compare the performance of ZW800-1C with that of the two spectrally similar heptamethine fluorophores ZW800-1A and indocyanine green, and show that ZW800-1C undergoes a longer fluorescence-lifetime shift when bound to amyloid-β and tau aggregates than when circulating in blood vessels. ZW800-1C may prove advantageous for tracking the proteinic aggregates in rodent models of amyloid-β and tau pathologies.

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: Candidate heptamethine NIR fluorophores for AD imaging.
Fig. 2: Ex vivo imaging of amyloid plaques and NFTs in APP/PS1 and rTg4510 mouse brain sections.
Fig. 3: In vivo imaging of amyloid plaques and CAA with ICG, ZW800-1A and ZW800-1C in APP/PS1 mice.
Fig. 4: In vivo imaging of NFTs with ICG, ZW800-1A and ZW800-1C in rTg4510 mice.
Fig. 5: In vivo fluorescence lifetime imaging of ZW800-1C in C57BL/6J, APP/PS1 and rTg4510 mice.
Fig. 6: Two-photon microscopy and non-invasive lifetime imaging of AD pathology with ZW800-1C.

Similar content being viewed by others

Data availability

The main data supporting the results in this study are available within the paper and its Supplementary Information. The raw and analysed datasets generated during the study are available for research purposes from the corresponding authors on reasonable request. Source data are provided with this paper.

Code availability

The custom ImageJ and Matlab codes are available on request from the corresponding authors.

References

  1. Montine, T. J. et al. National Institute on Aging–Alzheimer’s Association guidelines for the neuropathologic assessment of Alzheimer’s disease: a practical approach. Acta Neuropathol. 123, 1–11 (2012).

    CAS  PubMed  Google Scholar 

  2. Mathis, C., Wang, Y. & Klunk, W. Imaging beta-amyloid plaques and neurofibrillary tangles in the aging human brain. Curr. Pharm. Des. 10, 1469–1492 (2005).

    Google Scholar 

  3. Villemagne, V. L., Doré, V., Burnham, S. C., Masters, C. L. & Rowe, C. C. Imaging tau and amyloid-β proteinopathies in Alzheimer disease and other conditions. Nat. Rev. Neurol. 14, 225–236 (2018).

    CAS  PubMed  Google Scholar 

  4. Poduslo, J. F. et al. Molecular targeting of Alzheimer’s amyloid plaques for contrast-enhanced magnetic resonance imaging. Neurobiol. Dis. 11, 315–329 (2002).

    CAS  PubMed  Google Scholar 

  5. Flaherty, D. P. et al. Polyfluorinated bis-styrylbenzene β-amyloid plaque binding ligands. J. Med. Chem. 50, 4986–4992 (2007).

    CAS  PubMed  Google Scholar 

  6. Martins, A. F. et al. PiB-conjugated, metal-based imaging probes: multimodal approaches for the visualization of β-amyloid plaques. ACS Med. Chem. Lett. 4, 436–440 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Chamberlain, R. et al. Magnetic resonance imaging of amyloid plaques in transgenic mouse models of Alzheimer’s disease. Curr. Med. Imaging Rev. 7, 3–7 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Mathis, C. A., Mason, N. S., Lopresti, B. J. & Klunk, W. E. Development of positron emission tomography β-amyloid plaque imaging agents. Semin. Nucl. Med. 42, 423–432 (2012).

    PubMed  PubMed Central  Google Scholar 

  9. Zhu, L., Ploessl, K. & Kung, H. F. PET/SPECT imaging agents for neurodegenerative diseases. Chem. Soc. Rev. 43, 6683–6691 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Krishnadas, N., Villemagne, V. L., Doré, V. & Rowe, C. C. Advances in brain amyloid imaging. Semin. Nucl. Med. 51, 241–252 (2021).

    PubMed  Google Scholar 

  11. Zhuang, Z. P. et al. Structure-activity relationship of imidazo[1,2-a]pyridines as ligands for detecting β-amyloid plaques in the brain. J. Med. Chem. 46, 237–243 (2003).

    CAS  PubMed  Google Scholar 

  12. Qu, W., Kung, M. P., Hou, C., Benedum, T. E. & Kung, H. F. Novel styrylpyridines as probes for SPECT imaging of amyloid plaques. J. Med. Chem. 50, 2157–2165 (2007).

    CAS  PubMed  Google Scholar 

  13. Valotassiou, V. et al. SPECT and PET imaging in Alzheimer’s disease. Ann. Nucl. Med. 32, 583–593 (2018).

    CAS  PubMed  Google Scholar 

  14. Tong, H., Lou, K. & Wang, W. Near-infrared fluorescent probes for imaging of amyloid plaques in Alzheimer’s disease. Acta Pharm. Sin. B 5, 25–33 (2015).

    PubMed  PubMed Central  Google Scholar 

  15. Klunk, W. E. et al. Imaging Aβ plaques in living transgenic mice with multiphoton microscopy and methoxy-X04, a systemically administered Congo red derivative. J. Neuropathol. Exp. Neurol. 61, 797–805 (2002).

    CAS  PubMed  Google Scholar 

  16. Heo, C. H. et al. A quadrupolar two-photon fluorescent probe for in vivo imaging of amyloid-β plaques. Chem. Sci. 7, 4600–4606 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. 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 

  18. Ntziachristos, V., Bremer, C. & Weissleder, R. Fluorescence imaging with near-infrared light: new technological advances that enable in vivo molecular imaging. Eur. Radiol. 13, 195–208 (2003).

    PubMed  Google Scholar 

  19. Ntziachristos, V. Fluorescence molecular imaging. Annu. Rev. Biomed. Eng. 8, 1–33 (2006).

    CAS  PubMed  Google Scholar 

  20. Frangioni, J. V. In vivo near-infrared fluorescence imaging. Curr. Opin. Chem. Biol. 7, 626–634 (2003).

    CAS  PubMed  Google Scholar 

  21. Licha, K. & Olbrich, C. Optical imaging in drug discovery and diagnostic applications. Adv. Drug Deliv. Rev. 57, 1087–1108 (2005).

    CAS  PubMed  Google Scholar 

  22. Shcherbakova, D. M., Stepanenko, O. V., Turoverov, K. K. & Verkhusha, V. V. Near-infrared fluorescent proteins: multiplexing and optogenetics across scales. Trends Biotechnol. 36, 1230–1243 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Hong, G., Antaris, A. L. & Dai, H. Near-infrared fluorophores for biomedical imaging. Nat. Biomed. Eng. 1, 0010 (2017).

    CAS  Google Scholar 

  24. Shi, L., Sordillo, L. A., Rodríguez-Contreras, A. & Alfano, R. Transmission in near-infrared optical windows for deep brain imaging. J. Biophotonics 9, 38–43 (2016).

    CAS  PubMed  Google Scholar 

  25. Kim, D. et al. Two-photon absorbing dyes with minimal autofluorescence in tissue imaging: application to in vivo imaging of amyloid-β plaques with a negligible background signal. J. Am. Chem. Soc. 137, 6781–6789 (2015).

    CAS  PubMed  Google Scholar 

  26. Yang, J. et al. Oxalate-curcumin-based probe for micro- and macroimaging of reactive oxygen species in Alzheimer’s disease. Proc. Natl Acad. Sci. USA 114, 12384–12389 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Chen, C. et al. In vivo near-infrared two-photon imaging of amyloid plaques in deep brain of Alzheimer’s disease mouse model. ACS Chem. Neurosci. 9, 3128–3136 (2018).

    CAS  PubMed  Google Scholar 

  28. Li, Y. et al. Fluoro-substituted cyanine for reliable in vivo labelling of amyloid-β oligomers and neuroprotection against amyloid-β induced toxicity. Chem. Sci. 8, 8279–8284 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Zhang, X. et al. Design and synthesis of curcumin analogues for in vivo fluorescence imaging and inhibiting copper-induced cross-linking of amyloid beta species in Alzheimer’s disease. J. Am. Chem. Soc. 135, 16397–16409 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Cui, M. et al. Smart near-infrared fluorescence probes with donor-acceptor structure for in vivo detection of β-amyloid deposits. J. Am. Chem. Soc. 136, 3388–3394 (2014).

    CAS  PubMed  Google Scholar 

  31. Zhang, X. et al. Near-infrared fluorescence molecular imaging of amyloid beta species and monitoring therapy in animal models of Alzheimer’s disease. Proc. Natl Acad. Sci. USA 112, 9734–9739 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Fu, H. et al. Amyloid-β deposits target efficient near-infrared fluorescent probes: synthesis, in vitro evaluation, and in vivo imaging. Anal. Chem. 88, 1944–1950 (2016).

    CAS  PubMed  Google Scholar 

  33. Seo, Y. et al. A smart near-infrared fluorescence probe for selective detection of tau fibrils in Alzheimer’s disease. ACS Chem. Neurosci. 7, 1474–1481 (2016).

    CAS  PubMed  Google Scholar 

  34. Park, K. S. et al. A difluoroboron β-diketonate probe shows ‘turn-on’ near-infrared fluorescence specific for tau fibrils. ACS Chem. Neurosci. 8, 2124–2131 (2017).

    CAS  PubMed  Google Scholar 

  35. Ni, R. et al. Detection of cerebral tauopathy in P301L mice using high-resolution large-field multifocal illumination fluorescence microscopy. Biomed. Opt. Express 11, 4989–5002 (2020).

  36. Tagai, K. et al. High-contrast in vivo imaging of Tau pathologies in Alzheimer’s and non-Alzheimer’s disease tauopathies. Neuron 109, 42–58.e8 (2021).

    CAS  PubMed  Google Scholar 

  37. Park, K. S. et al. A curcumin-based molecular probe for near-infrared fluorescence imaging of tau fibrils in Alzheimer’s disease. Org. Biomol. Chem. 13, 11194–11199 (2015).

    CAS  PubMed  Google Scholar 

  38. Hyun, H. et al. Central C-C bonding increases optical and chemical stability of NIR fluorophores. RSC Adv. 4, 58762–58768 (2014).

    CAS  PubMed  Google Scholar 

  39. Yang, C. et al. ZW800-PEG: a renal clearable zwitterionic near-infrared fluorophore for potential clinical translation. Angew. Chem. Int. Ed. 60, 13847–13852 (2021).

    CAS  Google Scholar 

  40. Landsman, M. L. J., Kwant, G., Mook, G. A. & Zijlstra, W. G. Light absorbing properties, stability, and spectral stabilization of indocyanine green. J. Appl. Physiol. 40, 575–583 (1976).

    CAS  PubMed  Google Scholar 

  41. Choi, H. S. et al. Synthesis and in vivo fate of zwitterionic near-infrared fluorophores. Angew. Chem. Int. Ed. 50, 6258–6263 (2011).

    CAS  Google Scholar 

  42. Raymond, S. B., Boas, D. A., Bacskai, B. J. & Kumar, A. T. N. Lifetime-based tomographic multiplexing. J. Biomed. Opt. 15, 046011 (2010).

    PubMed  PubMed Central  Google Scholar 

  43. Venugopal, V., Chen, J., Lesage, F. & Intes, X. Full-field time-resolved fluorescence tomography of small animals. Opt. Lett. 35, 3189–3191 (2010).

    PubMed  Google Scholar 

  44. Jankowsky, J. L. et al. Mutant presenilins specifically elevate the levels of the 42 residue β-amyloid peptide in vivo: evidence for augmentation of a 42-specific γ secretase. Hum. Mol. Genet. 13, 159–170 (2004).

    CAS  PubMed  Google Scholar 

  45. Santacruz, K. et al. Tau suppression in a neurodegenerative mouse model improves memory function. Science 309, 476–481 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Rajamohamedsait, H. B. & Sigurdsson, E. M. Histological staining of amyloid and pre-amyloid peptides and proteins in mouse tissue. Methods Mol. Biol. 849, 411–424 (2012).

    CAS  PubMed  Google Scholar 

  47. Kwan, A. C., Duff, K., Gouras, G. K. & Webb, W. W. Optical visualization of Alzheimer’s pathology via multiphoton-excited intrinsic fluorescence and second harmonic generation. Opt. Express 17, 3679–3689 (2009).

  48. Shirani, H. et al. A palette of fluorescent thiophene-based ligands for the identification of protein aggregates. Chemistry 21, 15133–15137 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Calvo-Rodriguez, M. et al. In vivo detection of tau fibrils and amyloid β aggregates with luminescent conjugated oligothiophenes and multiphoton microscopy. Acta Neuropathol. Commun. 7, 171 (2019).

    PubMed  PubMed Central  Google Scholar 

  50. Banks, W. A. Characteristics of compounds that cross the blood-brain barrier. BMC Neurology 9, S3 (2009).

    PubMed  PubMed Central  Google Scholar 

  51. Minogue, A. M. et al. Age-associated dysregulation of microglial activation is coupled with enhanced blood-brain barrier permeability and pathology in APP/PS1 mice. Neurobiol. Aging 35, 1442–1452 (2014).

    CAS  PubMed  Google Scholar 

  52. Ohtsuki, S. & Terasaki, T. Contribution of carrier-mediated transport systems to the blood-brain barrier as a supporting and protecting interface for the brain; importance for CNS drug discovery and development. Pharm. Res. 24, 1745–1758 (2007).

    CAS  PubMed  Google Scholar 

  53. Sanchez-Covarrubias, L., Slosky, L., Thompson, B., Davis, T. & Ronaldson, P. Transporters at CNS barrier sites: obstacles or opportunities for drug delivery? Curr. Pharm. Des. 20, 1422–1449 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Kuchibhotla, K. V. et al. Aβ plaques lead to aberrant regulation of calcium homeostasis in vivo resulting in structural and functional disruption of neuronal networks. Neuron 59, 214–225 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Wang, T. et al. Three-photon imaging of mouse brain structure and function through the intact skull. Nat. Methods 15, 789–792 (2018).

    PubMed  PubMed Central  Google Scholar 

  56. Pologruto, T. A., Sabatini, B. L. & Svoboda, K. ScanImage: flexible software for operating laser scanning microscopes. Biomed. Eng. Online 2, 13 (2003).

    PubMed  PubMed Central  Google Scholar 

  57. Choi, H. S. et al. Targeted zwitterionic near-infrared fluorophores for improved optical imaging. Nat. Biotechnol. 31, 148–153 (2013).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank K. P. R. Nilsson for providing the HS-84 probe. This study was supported by grants from the National Institutes of Health (NIA R56AG060974 to B.J.B.; NIA K01AG072046 to S.S.H.; NCI R01CA211084 to A.T.N.K.; NIBIB R01EB022230 to H.S.C.) and the Creative Materials Discovery Program through the National Research Foundation of Korea (2019M3D1A1078938 to H.S.C.). The content expressed is solely the responsibility of the authors and does not necessarily represent the official views of the NIH.

Author information

Authors and Affiliations

Authors

Contributions

S.S.H., J.H.L., B.J.B. and H.S.C. conceived the study and designed the experiments. S.S.H., J.Y., S.A. and Y.K. performed the ex vivo imaging experiments and analysed the results. S.S.H., J.Y., J.H.L., Y.K., M.C.-R. and S.K. performed the in vivo validation experiments and analysed the results. K.B. synthesized the compounds used in the study (ZW800-1A and ZW800-1C). S.S.H. and A.T.N.K. performed the fluorescence lifetime measurements and interpreted the results. S.S.H., B.J.B. and H.S.C. wrote the manuscript. B.J.B. and H.S.C. supervised the entire project. All authors approved the final version of the manuscript.

Corresponding authors

Correspondence to Brian J. Bacskai or Hak Soo Choi.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Biomedical Engineering thanks Jan Klohs and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Ex vivo labeling intensity of amyloid plaques and CAA by ZW800-1C.

Quantitative comparison of the average labeling intensity of CAA and plaques by ZW800-1C in APP/PS1 brain tissue sections. Sections were stained by incubation in 100 µM ZW800-1C solution in PBS. Significance was calculated using unpaired two-tailed t-test: ****P < 0.0001. Data are shown as mean ± s.e.m.

Extended Data Fig. 2 In vivo imaging reveals selective labeling of CAA with ICG in APP/PS1 mice.

Examples of arteries with CAA that are positively labeled with MX04 and (a) co-labeled with ICG and (b) not co-labeled with ICG. Images were acquired with confocal microscopy at 2 h post-injection.

Extended Data Fig. 3 In vivo imaging of ICG, ZW800-1A, and ZW800-1C in APP/PS1 and rTg4510 non-transgenic littermates.

Each NIR fluorophore was injected intravenously (50 nmol) and imaged 2 h post-injection with confocal microscopy. Representative images for both (a) APP/PS1 non-Tg littermates (n = 3) and (b) rTg4510 non-Tg littermates (n = 3) show low levels of periarterial labeling by all three fluorophores.

Extended Data Fig. 4 Post-mortem validation of ZW800-1C labeling in APP/PS1 and rTg4510 mice.

APP/PS1 mouse is intraperitoneally injected with MX04 and intravenously injected with ZW800-1C at 24 h and 2 h before sacrifice. Ex vivo imaging is performed on brain tissue sections, and representative images show co-stained (a) CAA and (b) amyloid plaques. (c) Tg4510 mouse is intravenously injected with HS-84 and ZW800-1C at 7 days and 2 h before sacrifice, respectively. Representative images show co-labeling of NFTs.

Extended Data Fig. 5 In vivo biodistribution study of ICG, ZW800-1A, and ZW800-1C in CD-1 mice.

25 nmol of ZW800-1C was injected in 25 g CD-1 mice 4 h prior to imaging. (a) Representative color and NIR images of ZW800-1C showing major organs in the thoracic cavity (left), abdominal cavity (middle), and after resection (right). (b) Signal-to-background ratio (SBR) of resected major organs (Or) against muscle (Mu). Abbreviations used are He, heart; Lu, lung; Li, liver; Pa, pancreas; Sp, spleen; Ki, kidney; Du, duodenum; Ga, gallbladder; BD, bile duct; Bl, bladder; In, intestine. ***P = 0.0006 and ****P < 0.0001 by two-way ANOVA with Dunnett’s multiple comparison test. (c) Comparison of the pharmacokinetic parameters of the candidate NIR fluorophores. Abbreviations used are t1/2α, distribution blood half-life; t1/2β, elimination blood half-life; Vd, the volume of distribution; AUC, area under the curve; Cl, clearance. Data are shown as mean ± s.e.m.

Extended Data Fig. 6 In vivo fluorescence lifetime imaging of ICG and ZW800-1A in C57BL/6J and APP/PS1 mice.

Histogram distribution of fluorescence lifetimes for (a) ICG and (b) ZW800-1A within blood vessels and after being bound to amyloid-β aggregates.

Extended Data Fig. 7 Two-photon imaging of ZW800-1C in APP/PS1 and rTg4510 mice.

Two-photon imaging of ZW800-1C (red) with 1,300 nm excitation. Alexa Fluor 680-70 kDa Dextran (green) was systemically injected to create a fluorescent angiogram. Representative images show (a) CAA, (b) amyloid plaques and (c) NFTs from APP/PS1 (n = 3) and rTg4510 (n = 3) mice. (d) Images of amyloid plaques taken at different depths from 7–8 months old (mo) and 22–24 mo APP/PS1 mice. (e) Comparison of the percentage volume occupied by amyloid plaques for 7–8 mo and 22–24 mo APP/PS1 mice (n = 3 mice per age group). Data are shown as mean ± s.e.m.

Supplementary information

Supplementary Information

Supplementary figures and caption for the supplementary video.

Reporting Summary

Supplementary Video

In vivo through-the-skull imaging of an APP/PS1 mouse.

Source data

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hou, S.S., Yang, J., Lee, J.H. et al. Near-infrared fluorescence lifetime imaging of amyloid-β aggregates and tau fibrils through the intact skull of mice. Nat. Biomed. Eng 7, 270–280 (2023). https://doi.org/10.1038/s41551-023-01003-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41551-023-01003-7

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