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.

  • Protocol
  • Published:

Whole-brain block-face serial microscopy tomography at subcellular resolution using FAST

Abstract

Here, we describe an optimized and detailed protocol for block-face serial microscopy tomography (FAST). FAST enables high-speed serial section fluorescence imaging of fixed brains at an axonal spatial resolution and subsequent image data processing. It renders brain-wide anatomical and functional analyses, including structural profiling of nuclear-stained brain at the single-cell level, cell-type-specific mapping with reporter animal brains and neuronal tracing with anterograde/retrograde labeling. Light-sheet fluorescence microscopy of cleared brains is advantageous in regard to imaging speed, but its spatial resolution is generally limited, whereas the opposite is true for conventional confocal microscopy. FAST offers a solution to overcome these technical limitations. This protocol describes detailed procedures for assembling the FAST hardware, sample preparation, imaging and image processing. A single imaging session takes as little as 2.4 h per mouse brain, and sample preparation requires 1 to several days, depending on pretreatments; however, multiple samples can be prepared simultaneously. We anticipate that FAST will contribute to unbiased and hypothesis-free approaches for a better understanding of brain systems.

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: Overview of FAST imaging analyses.
Fig. 2: Assembling FAST hardware.
Fig. 3: Design of the sample chamber.
Fig. 4: FAST imaging procedures.
Fig. 5: Whole-brain imaging of nuclear-stained brain at a voxel resolution of 2.5 µm3.
Fig. 6: Chemical staining of a whole mouse brain and multicolor imaging.
Fig. 7: Whole-brain imaging of a fluorescent-reporter animal brain: Arc-dVenus mouse.
Fig. 8: Whole-brain imaging of axonal projections from the anterior cingulate cortex.
Fig. 9: Multicolor whole-brain imaging with two different fluorescent proteins whose expression is driven by the cell-type-specific promoter and enhancer.

Similar content being viewed by others

References

  1. Ecker, J. R. et al. The BRAIN initiative cell census consortium: lessons learned toward generating a comprehensive brain cell atlas. Neuron 96, 542–557 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Seiriki, K. et al. High-speed and scalable whole-brain imaging in rodents and primates. Neuron 94, 1085–1100.e6 (2017).

    Article  CAS  PubMed  Google Scholar 

  3. Renier, N. et al. Mapping of brain activity by automated volume analysis of immediate early genes. Cell 165, 1789–1802 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Ye, L. et al. Wiring and molecular features of prefrontal ensembles representing distinct experiences. Cell 165, 1776–1788 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Eguchi, M. & Yamaguchi, S. In vivo and in vitro visualization of gene expression dynamics over extensive areas of the brain. Neuroimage 44, 1274–1283 (2009).

    Article  PubMed  Google Scholar 

  6. Reijmers, L. G., Perkins, B. L., Matsuo, N. & Mayford, M. Localization of a stable neural correlate of associative memory. Science 317, 1230–1233 (2007).

    Article  CAS  PubMed  Google Scholar 

  7. Vousden, D. A. et al. Whole-brain mapping of behaviourally induced neural activation in mice. Brain Struct. Funct. 220, 2043–2057 (2014).

    Article  PubMed  Google Scholar 

  8. Kim, Y. et al. Mapping social behavior-induced brain activation at cellular resolution in the mouse. Cell Rep. 10, 292–305 (2015).

    Article  CAS  PubMed  Google Scholar 

  9. Tatsuki, F. et al. Involvement of Ca(2+)-dependent hyperpolarization in sleep duration in mammals. Neuron 90, 70–85 (2016).

    Article  CAS  PubMed  Google Scholar 

  10. Seiriki, K. et al. Critical involvement of the orbitofrontal cortex in hyperlocomotion induced by NMDA receptor blockade in mice. Biochem. Biophys. Res. Commun. 480, 558–563 (2016).

    Article  CAS  PubMed  Google Scholar 

  11. Kim, Y. et al. Whole-brain mapping of neuronal activity in the learned helplessness model of depression. Front. Neural Circuits 10, 3 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Dodt, H. U. et al. Ultramicroscopy: three-dimensional visualization of neuronal networks in the whole mouse brain. Nat. Methods 4, 331–336 (2007).

    Article  CAS  PubMed  Google Scholar 

  13. Ragan, T. et al. Serial two-photon tomography for automated ex vivo mouse brain imaging. Nat. Methods 9, 255–258 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Gong, H. et al. High-throughput dual-colour precision imaging for brain-wide connectome with cytoarchitectonic landmarks at the cellular level. Nat. Commun. 7, 12142 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Tomer, R., Ye, L., Hsueh, B. & Deisseroth, K. Advanced CLARITY for rapid and high-resolution imaging of intact tissues. Nat. Protoc. 9, 1682–1697 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Susaki, E. A. et al. Advanced CUBIC protocols for whole-brain and whole-body clearing and imaging. Nat. Protoc. 10, 1709–1727 (2015).

    Article  CAS  PubMed  Google Scholar 

  17. Hama, H. et al. ScaleS: an optical clearing palette for biological imaging. Nat. Neurosci. 18, 1518–1529 (2015).

    Article  CAS  PubMed  Google Scholar 

  18. Silvestri, L., Bria, A., Sacconi, L., Iannello, G. & Pavone, F. S. Confocal light sheet microscopy: micron-scale neuroanatomy of the entire mouse brain. Opt. Express 20, 20582–20598 (2012).

    Article  CAS  PubMed  Google Scholar 

  19. Chen, F., Tillberg, P. W. & Boyden, E. S. Optical imaging. Expansion microscopy. Science 347, 543–548 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Murakami, T. C. et al. A three-dimensional single-cell-resolution whole-brain atlas using CUBIC-X expansion microscopy and tissue clearing. Nat. Neurosci. 21, 625–637 (2018).

    Article  CAS  PubMed  Google Scholar 

  21. Economo, M. N. et al. A platform for brain-wide imaging and reconstruction of individual neurons. Elife 5, e10566 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Economo, M. N. et al. Distinct descending motor cortex pathways and their roles in movement. Nature 563, 79–84 (2018).

    Article  CAS  PubMed  Google Scholar 

  23. Ellegood, J. et al. Clustering autism: using neuroanatomical differences in 26 mouse models to gain insight into the heterogeneity. Mol. Psychiatry 20, 118–125 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Renier, N. et al. iDISCO: a simple, rapid method to immunolabel large tissue samples for volume imaging. Cell 159, 896–910 (2014).

    Article  CAS  PubMed  Google Scholar 

  25. Susaki, E. A. & Ueda, H. R. Whole-body and whole-organ clearing and imaging techniques with single-cell resolution: toward organism-level systems biology in mammals. Cell Chem. Biol. 23, 137–157 (2016).

    Article  CAS  PubMed  Google Scholar 

  26. Kais, B. et al. DMSO modifies the permeability of the zebrafish (Danio rerio) chorion-implications for the fish embryo test (FET). Aquat. Toxicol. 140-141, 229–238 (2013).

    Article  CAS  PubMed  Google Scholar 

  27. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 (2012).

    Article  CAS  PubMed  Google Scholar 

  28. Bria, A. & Iannello, G. TeraStitcher—a tool for fast automatic 3D-stitching of teravoxel-sized microscopy images. BMC Bioinformatics 13, 316 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Niedworok, C. J. et al. aMAP is a validated pipeline for registration and segmentation of high-resolution mouse brain data. Nat. Commun. 7, 11879 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Fürth, D. et al. An interactive framework for whole-brain maps at cellular resolution. Nat. Neurosci. 21, 139–149 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Dimidschstein, J. et al. A viral strategy for targeting and manipulating interneurons across vertebrate species. Nat. Neurosci. 19, 1743–1749 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Chan, K. Y. et al. Engineered AAVs for efficient noninvasive gene delivery to the central and peripheral nervous systems. Nat. Neurosci. 20, 1172–1179 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Dittgen, T. et al. Lentivirus-based genetic manipulations of cortical neurons and their optical and electrophysiological monitoring in vivo. Proc. Natl. Acad. Sci. USA 101, 18206–18211 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We are grateful to S. Yamaguchi (Gifu University) for providing Arc-dVenus-reporter mice; to K.-I. Inoue (Kyoto University) and M. Takada (Kyoto University) for providing and supporting the AAV-CMV-tdTomato vectors and the plasmid vector to express the AAV-PHP.eB capsid; to A. Yamanaka (Nagoya University) for providing the AAV plasmid vector, including the mouse alpha-CaMKII promoter; to G. Fishell (Harvard Medical School) for providing the pAAV-mDlx-GFP-Fishell-1 plasmid vector (AAV-mDLX-EGFP in Fig. 9) (Addgene, plasmid no. 83900)31; to K. Fujita (Osaka University) and T. Nagai (Osaka University) for helpful suggestions on the setup of the FAST apparatus; and to T. Hashimoto (Shizuoka University) for valuable support in image data processing. We also thank T. Funato (Nikon Instech), M. Sato (COMS), H. Tanaka (Yokogawa Electric), O. Kunitaki (Andor Technology) and S. Kameishi (Dosaka EM) for their valuable suggestions and support. This work was supported in part by JSPS KAKENHI, grant nos. JP17H06842 (K.S.), JP18K19498 (K.S.), JP17H05054 (A.K.) and JP17H03989 (H.H.); the JSPS Research Fellowships for Young Scientists, grant no. JP18J10350 (M.N.); MEXT KAKENHI, grant nos. JP18H05416 (H.H.) and JP18H05132 (A.K.); AMED, grant nos. JP18dm0107122h (H.H.), JP18dm0207061h (H.H.) and JP18am0101084; and grants from the Takeda Science Foundation, Japan (A.K.).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, K.S., A.K. and H.H.; methodology, K.S., A.K., T.N. and H.H.; sample preparation and imaging, K.S., A.K., M.N., Y.N., M.T., H.I. and K.Y.; investigation, K.S., A.K., M.N., A.H.-T., Y.A. and H.H.; writing, K.S., A.K., T.N. and H.H.; funding acquisition, K.S., A.K., M.N. and H.H.

Corresponding authors

Correspondence to Atsushi Kasai or Hitoshi Hashimoto.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Journal peer review information: Nature Protocols thanks Pavel Osten and 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.

Related link

Key reference using this protocol

Seiriki, K. et al. Neuron 94, 1085–1100.e6 (2017): https://doi.org/10.1016/j.neuron.2017.05.017

Integrated supplementary information

Supplementary Figure 1 Example of imaging software settings for whole-brain imaging.

An example of the imaging protocol in Andor iQ software. Trigger out signal and reloading protocol must be included for the subsequent sectioning procedure and automatic repetition using the CP-700 program. In addition to this protocol setup, the external start mode is required to receive the start signal from the CP-700 program after sectioning. The mechanical laser shutter must be kept open for high-speed imaging; ‘561 ShutterKeepOpen’ is a user-designated protocol to keep the shutter of the 561-nm laser open.

Supplementary information

Supplementary Text and Figures

Supplementary Figure 1, Supplementary Tables 1 and 2

Reporting Summary

Supplementary Data

FASTitcher scripts, along with a user guide and a small sample dataset.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Seiriki, K., Kasai, A., Nakazawa, T. et al. Whole-brain block-face serial microscopy tomography at subcellular resolution using FAST. Nat Protoc 14, 1509–1529 (2019). https://doi.org/10.1038/s41596-019-0148-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-019-0148-4

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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