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-mouse clearing and imaging at the cellular level with vDISCO

Abstract

Homeostatic and pathological phenomena often affect multiple organs across the whole organism. Tissue clearing methods, together with recent advances in microscopy, have made holistic examinations of biological samples feasible. Here, we report the detailed protocol for nanobody(VHH)-boosted 3D imaging of solvent-cleared organs (vDISCO), a pressure-driven, nanobody-based whole-body immunolabeling and clearing method that renders whole mice transparent in 3 weeks, consistently enhancing the signal of fluorescent proteins, stabilizing them for years. This allows the reliable detection and quantification of fluorescent signal in intact rodents enabling the analysis of an entire body at cellular resolution. Here, we show the high versatility of vDISCO applied to boost the fluorescence signal of genetically expressed reporters and clear multiple dissected organs and tissues, as well as how to image processed samples using multiple fluorescence microscopy systems. The entire protocol is accessible to laboratories with limited expertise in tissue clearing. In addition to its applications in obtaining a whole-mouse neuronal projection map, detecting single-cell metastases in whole mice and identifying previously undescribed anatomical structures, we further show the visualization of the entire mouse lymphatic system, the application for virus tracing and the visualization of all pericytes in the brain. Taken together, our vDISCO pipeline allows systematic and comprehensive studies of cellular phenomena and connectivity in whole bodies.

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 the vDISCO pipeline.
Fig. 2: vDISCO imaging of a mouse with syngeneic pancreatic cancer.
Fig. 3: Whole-body active-vDISCO setup.
Fig. 4: Whole-body active-vDISCO procedure.
Fig. 5: vDISCO processed Thy1-GFPM brain imaged with Z.1 light-sheet microscope.
Fig. 6: Passive-vDISCO on dissected organs imaged with confocal and light-sheet microscopy.
Fig. 7: Performances of different kinds of nanobodies in passive-vDISCO.
Fig. 8: Prox1-EGFP whole-body scan taken with epifluorescence, light-sheet and confocal microscopy.
Fig. 9: Virus tracing and pericytes in the brain processed by vDISCO.

Similar content being viewed by others

Data availability

The main data discussed in this protocol are available in the supporting primary research papers (https://www.nature.com/articles/s41593-018-0301-3 and https://www.cell.com/cell/fulltext/S0092-8674(19)31269-3?_returnURL=https%3A%2F%2Flinkinghub.elsevier.com%2Fretrieve%2Fpii%2FS0092867419312693%3Fshowall%3Dtrue). The raw datasets of the main data and of the additional new data shown in this work are too large to be publicly shared but are available for research purposes from the corresponding authors upon reasonable request.

References

  1. Pan, C. et al. Deep learning reveals cancer metastasis and therapeutic antibody targeting in the entire body. Cell 179, 1661–1676.e19 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Ntziachristos, V. Going deeper than microscopy: the optical imaging frontier in biology. Nat. Methods 7, 603–614 (2010).

    Article  CAS  PubMed  Google Scholar 

  3. James, M. L. & Gambhir, S. S. A molecular imaging primer: modalities, imaging agents, and applications. Physiol. Rev. 92, 897–965 (2012).

    Article  CAS  PubMed  Google Scholar 

  4. Timpson, P., McGhee, E. J. & Anderson, K. I. Imaging molecular dynamics in vivo—from cell biology to animal models. J. Cell Sci. 124, 2877–2890 (2011).

    Article  CAS  PubMed  Google Scholar 

  5. Erturk, A. et al. Three-dimensional imaging of the unsectioned adult spinal cord to assess axon regeneration and glial responses after injury. Nat. Med. 18, 166–171 (2012).

    Article  Google Scholar 

  6. Hama, H. et al. Scale: a chemical approach for fluorescence imaging and reconstruction of transparent mouse brain. Nat. Neurosci. 14, 1481–1488 (2011).

    Article  CAS  PubMed  Google Scholar 

  7. Chung, K. et al. Structural and molecular interrogation of intact biological systems. Nature 497, 332–337 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Susaki, E. A. et al. Whole-brain imaging with single-cell resolution using chemical cocktails and computational analysis. Cell 157, 726–739 (2014).

    Article  CAS  PubMed  Google Scholar 

  9. Ke, M.-T., Fujimoto, S. & Imai, T. SeeDB: a simple and morphology-preserving optical clearing agent for neuronal circuit reconstruction. Nat. Neurosci. 16, 1154–1161 (2013).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  11. 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 

  12. Belle, M. et al. Tridimensional visualization and analysis of early human development. Cell 169, 161–173.e12 (2017).

    Article  CAS  PubMed  Google Scholar 

  13. Belle, M. et al. A simple method for 3D analysis of immunolabeled axonal tracts in a transparent nervous system. Cell Rep. 9, 1191–1201 (2014).

    Article  CAS  PubMed  Google Scholar 

  14. Murray, E. et al. Simple, scalable proteomic imaging for high-dimensional profiling of intact systems. Cell 163, 1500–1514 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. 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 

  16. Pan, C. et al. Shrinkage-mediated imaging of entire organs and organisms using uDISCO. Nat. Methods 13, 859–867 (2016).

    Article  CAS  PubMed  Google Scholar 

  17. 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 

  18. Tainaka, K. et al. Whole-body imaging with single-cell resolution by tissue decolorization. Cell 159, 911–924 (2014).

    Article  CAS  PubMed  Google Scholar 

  19. Kubota, S. I. et al. Whole-body profiling of cancer metastasis with single-cell resolution. Cell Rep. 20, 236–250 (2017).

    Article  CAS  PubMed  Google Scholar 

  20. Yang, B. et al. Single-cell phenotyping within transparent intact tissue through whole-body clearing. Cell 158, 945–958 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Treweek, J. B. et al. Whole-body tissue stabilization and selective extractions via tissue–hydrogel hybrids for high-resolution intact circuit mapping and phenotyping. Nat. Protoc. 10, 1860–1896 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Jing, D. et al. Tissue clearing of both hard and soft tissue organs with the PEGASOS method. Cell Res. 28, 803–818 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Cai, R. et al. Panoptic imaging of transparent mice reveals whole-body neuronal projections and skull–meninges connections. Nat. Neurosci. 22, 317–327 (2019).

    Article  CAS  PubMed  Google Scholar 

  24. Muyldermans, S. Single domain camel antibodies: current status. Rev. Mol. Biotechnol. 74, 277–302 (2001).

    Article  CAS  Google Scholar 

  25. Muyldermans, S. Nanobodies: natural single-domain antibodies. Annu. Rev. Biochem. 82, 775–797 (2013).

    Article  CAS  PubMed  Google Scholar 

  26. Schumacher, D., Helma, J., Schneider, A. F. L., Leonhardt, H. & Hackenberger, C. P. R. Nanobodies: chemical functionalization strategies and intracellular applications. Angew. Chem. Int. Ed. 57, 2314–2333 (2018).

    Article  CAS  Google Scholar 

  27. Niess, J. H. et al. CX3CR1-mediated dendritic cell access to the intestinal lumen and bacterial clearance. Science 307, 254–258 (2005).

    Article  CAS  PubMed  Google Scholar 

  28. Gage, G. J., Kipke, D. R. & Shain, W. Whole animal perfusion fixation for rodents. J. Vis. Exp. https://doi.org/10.3791/3564 (2012).

  29. Wang, X. et al. An ocular glymphatic clearance system removes β-amyloid from the rodent eye. Sci. Transl. Med. 12, eaaw3210 (2020).

  30. Louveau, A. et al. Structural and functional features of central nervous system lymphatic vessels. Nature 523, 337–341 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  Google Scholar 

  32. Quan, T. et al. NeuroGPS-Tree: automatic reconstruction of large-scale neuronal populations with dense neurites. Nat. Methods 13, 51–54 (2016).

    Article  CAS  PubMed  Google Scholar 

  33. Li, A. et al. Micro-optical sectioning tomography to obtain a high-resolution atlas of the mouse brain. Science 330, 1404–1408 (2010).

    Article  CAS  PubMed  Google Scholar 

  34. Qi, X. et al. Fluorescence micro-optical sectioning tomography using acousto-optical deflector-based confocal scheme. Neurophotonics 2, 041406–041406 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  35. 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 

  36. Feng, G. et al. Imaging neuronal subsets in transgenic mice expressing multiple spectral variants of GFP. Neuron 28, 41–51 (2000).

    Article  CAS  PubMed  Google Scholar 

  37. Zhao, S. et al. Cellular and molecular probing of intact human organs. Cell 180, 796–812.e19 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Richardson, D. S. et al. Tissue clearing. Nat. Rev. Methods Primer 1, 1–24 (2021).

    Article  Google Scholar 

  39. Erturk, A. et al. Three-dimensional imaging of solvent-cleared organs using 3DISCO. Nat. Protoc. 7, 1983–1995 (2012).

    Article  CAS  PubMed  Google Scholar 

  40. Rothbauer, U. et al. Targeting and tracing antigens in live cells with fluorescent nanobodies. Nat. Methods 3, 887–889 (2006).

    Article  CAS  PubMed  Google Scholar 

  41. Horecker, B. L. The absorption spectra of hemoglobin and its derivatives in the visible and near infra-red regions. J. Biol. Chem. 148, 173–183 (1943).

    Article  CAS  Google Scholar 

  42. Tainaka, K., Kuno, A., Kubota, S. I., Murakami, T. & Ueda, H. R. Chemical principles in tissue clearing and staining protocols for whole-body cell profiling. Annu. Rev. Cell Dev. Biol. 32, 713–741 (2016).

    Article  CAS  PubMed  Google Scholar 

  43. Tuchin, V. V. Tissue optics and photonics: light–tissue interaction. J. Biomed. Photonics Eng. 1, 98–134 (2015).

    Article  Google Scholar 

  44. Tainaka, K. et al. Chemical landscape for tissue clearing based on hydrophilic reagents. Cell Rep. 24, 2196–2210.e9 (2018).

    Article  CAS  PubMed  Google Scholar 

  45. Kristinsson, H. G. & Hultin, H. O. Changes in trout hemoglobin conformations and solubility after exposure to acid and alkali pH. J. Agric. Food Chem. 52, 3633–3643 (2004).

    Article  CAS  PubMed  Google Scholar 

  46. Alnuami, A. A., Zeedi, B., Qadri, S. M. & Ashraf, S. S. Oxyradical-induced GFP damage and loss of fluorescence. Int. J. Biol. Macromol. 43, 182–186 (2008).

    Article  CAS  PubMed  Google Scholar 

  47. Fagan, J. M., Sleczka, B. G. & Sohar, I. Quantitation of oxidative damage to tissue proteins. Int. J. Biochem. Cell Biol. 31, 751–757 (1999).

    Article  CAS  PubMed  Google Scholar 

  48. Acar, M. et al. Deep imaging of bone marrow shows non-dividing stem cells are mainly perisinusoidal. Nature 526, 126–130 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Greenbaum, A. et al. Bone CLARITY: clearing, imaging, and computational analysis of osteoprogenitors within intact bone marrow. Sci. Transl. Med. 9, eaah6518 (2017).

    Article  PubMed  Google Scholar 

  50. Gonzalez-Chavez, S. A., Pacheco-Tena, C., Macias-Vazquez, C. E. & Luevano-Flores, E. Assessment of different decalcifying protocols on osteopontin and osteocalcin immunostaining in whole bone specimens of arthritis rat model by confocal immunofluorescence. Int. J. Clin. Exp. Pathol. 6, 1972–1983 (2013).

    PubMed  PubMed Central  Google Scholar 

  51. Xiao, X. et al. Antibody incubation at 37°C improves fluorescent immunolabeling in free-floating thick tissue sections. Biotechniques 62, 115–122 (2017).

    Article  CAS  PubMed  Google Scholar 

  52. Weiss, K. R., Voigt, F. F., Shepherd, D. P. & Huisken, J. Tutorial: practical considerations for tissue clearing and imaging. Nat. Protoc. 16, 2732–2748 (2021).

    Article  CAS  PubMed  Google Scholar 

  53. 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 

  54. Voigt, F. F. et al. The mesoSPIM initiative: open-source light-sheet microscopes for imaging cleared tissue. Nat. Methods 16, 1105–1108 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Welch, A. Technique for high-performance data compression. Computer 17, 8–19 (1984).

    Article  Google Scholar 

  56. Ma, B. et al. A fast algorithm for material image sequential stitching. Comput. Mater. Sci. 158, 1–13 (2019).

    Article  Google Scholar 

  57. Dellatorre, G. & Gadens, G. A. Wide area digital dermoscopy applied to basal cell carcinoma. An. Bras. Dermatol. 95, 379–382 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Boatright, J. H. et al. Methodologies for analysis of patterning in the mouse RPE sheet. Mol. Vis. 21, 40–60 (2015).

    PubMed  PubMed Central  Google Scholar 

  59. Hörl, D. et al. BigStitcher: reconstructing high-resolution image datasets of cleared and expanded samples. Nat. Methods 16, 870–874 (2019).

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  61. Pietzsch, T., Preibisch, S., Tomancak, P. & Saalfeld, S. ImgLib2-generic image processing in Java. Bioinformatics 28, 3009–3011 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. 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 

  63. Glaser, J. R. & Glaser, E. M. Neuron imaging with neurolucida—a PC-based system for image combining microscopy. Comput. Med. Imaging Graph. 14, 307–317 (1990).

    Article  CAS  PubMed  Google Scholar 

  64. Belthangady, C. & Royer, L. A. Applications, promises, and pitfalls of deep learning for fluorescence image reconstruction. Nat. Methods https://doi.org/10.1038/s41592-019-0458-z (2019).

  65. Moen, E. et al. Deep learning for cellular image analysis. Nat. Methods https://doi.org/10.1038/s41592-019-0403-1 (2019).

  66. Zhou, H. et al. 3D high resolution generative deep-learning network for fluorescence microscopy imaging. Opt. Lett. 45, 1695–1698 (2020).

    Article  PubMed  Google Scholar 

  67. Mano, T. et al. CUBIC-Cloud provides an integrative computational framework toward community-driven whole-mouse-brain mapping. Cell Rep. Methods 1, 100038 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Iwasato, T. et al. Cortex-restricted disruption of NMDAR1 impairs neuronal patterns in the barrel cortex. Nature 406, 726–731 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Takatoh, J. et al. New modules are added to vibrissal premotor circuitry with the emergence of exploratory whisking. Neuron 77, 346–360 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Wickersham, I. R. et al. Monosynaptic restriction of transsynaptic tracing from single, genetically targeted neurons. Neuron 53, 639–647 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Reed, H. O. et al. Lymphatic impairment leads to pulmonary tertiary lymphoid organ formation and alveolar damage. J. Clin. Invest. 129, 2514–2526 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  72. Wigle, J. T. et al. An essential role for Prox1 in the induction of the lymphatic endothelial cell phenotype. EMBO J. 21, 1505–1513 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Kivelä, R. et al. The transcription factor Prox1 is essential for satellite cell differentiation and muscle fibre-type regulation. Nat. Commun. 7, 13124 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  74. Iwano, T., Masuda, A., Kiyonari, H., Enomoto, H. & Matsuzaki, F. Prox1 postmitotically defines dentate gyrus cells by specifying granule cell identity over CA3 pyramidal cell fate in the hippocampus. Dev. Camb. Engl. 139, 3051–3062 (2012).

    CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by the Vascular Dementia Research Foundation, Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) under Germany’s Excellence Strategy within the framework of the Munich Cluster for Systems Neurology (EXC 2145 SyNergy, ID 390857198), the ERC Consolidator Grant (AE, 865323), and the Nomis Heart Atlas Project Grant (Nomis Foundation). H.M. is also supported by the China Scholarship Council (CSC) (no. 201806780034). M.M. is supported by the Turkish Ministry of Education for her PhD studies. We thank K. Sleiman, C. Veltkamp and D. Saur for providing the animal bearing pancreatic cancer; M. Voll (Zeiss Microscopy GmbH, Germany) and the Zeiss Microscopy Customer Center Europe for performing the image acquisition with the Lightsheet Z.1; J. Thomas, S. Grade and M. Götz for providing the virus traced sample; J. Shrouder and N. Plesnila for providing the PDGFRb-EGFP mice; A. Ghasemigharagoz, A. Parra-Damas and F.P. Quacquarelli for help during the initial optimization and method development; M. Bralo and B. Forstera for help during the testing of some nanobodies; F. Hellal for critical reading of the manuscript. A.E. and Z.I.K. are members of the Graduate School of Systemic Neurosciences at the Ludwig Maximilian University of Munich.

Author information

Authors and Affiliations

Authors

Contributions

A.E. and R.C. initiated the project. R.C. and C.P. developed the original vDISCO method. R.C. designed the experiments. R.C. and Z.I.K. performed most of the experiments. C.P. provided data for the cancer mouse. H.M. and S.Z. provided data for the PDGFRb-EGFP pericyte-labeled brains. F.F.V., M.M., T.-L.O. and F.H. provided mesoSPIM data. C.V. and J.A.V.G. provided the custom-made nanobodies anti-GFP. D.K. contributed to the imaging, and M.I.T helped with the stitching of the Prox1-EGFP sample. R.C. and Z.I.K. supervised the experiments. A.E. supervised the project. R.C. and Z.I.K. wrote the manuscript. All authors edited the manuscript.

Corresponding author

Correspondence to Ali Ertürk.

Ethics declarations

Competing interests

A.E., R.C., C.P. and S.Z. have filed a patent related to vDISCO.

Peer review

Peer review information

Nature Protocols thanks Nicolas Renier 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.

Related Links

Key references using this protocol

Cai, R. et al. Nat. Neurosci. 22, 317–327 (2019): https://doi.org/10.1038/s41593-018-0301-3

Pan, C. et al. Cell 179, 1661–1676.e19 (2019): https://doi.org/10.1016/j.cell.2019.11.013

Extended data

Extended Data Fig. 1 Sample preparation for whole-body active-vDISCO.

a, During the 1× PBS perfusion step of the anesthetized animal, the color of the liver (cyan dashed line) turns yellow as indication of proper drainage of the blood from the body. b, Some cuts in the gut are necessary to flush out the gut content and the feces with a syringe. c, Pictures showing the procedure of skinning the body of the animal using a blade. d, In case the animal is needed with intact skin, commercial hair removal creams may be used as illustrated. The cream should be applied to region of interest and removed with water after 3–5 min. Steps can be repeated until the desired quality of hair removal is achieved. Animal experiments followed European directive 2010/63/EU for animal research, reported according to the Animal Research: Reporting of In Vivo Experiments (ARRIVE) criteria, complied with the ‘3Rs’ measure and were approved by the ethical review board of the government of Upper Bavaria (Regierung von Oberbayern, Munich, Germany) and conformed to institutional guidelines of Klinikum der Universität München/Ludwig Maximilian University of Munich). The severity of the procedure was low.

Extended Data Fig. 2 Whole-body imaging of a sample with intact skin using a modified mesoSPIM light-sheet microscope.

a, Maximum projection of an adult Thy1-GFPM mouse with intact skin imaged with a modified mesoSPIM light-sheet microscope from the ventral side. Autofluorescence excited at 488 nm is shown in green, GFP in magenta. b, Maximum projection of the same sample imaged from the dorsal side (rotated by 180°). c, Layer 5 pyramidal neurons in the brain. d, Peripheral nerves and skin of the forepaw imaged from the lateral direction (90° rotation). e, Detail of the spinal cord and vertebrae. Images in a, b, d and e were taken at 0.9× magnification whereas image in c was imaged at 4× magnification. Animal experiments followed European directive 2010/63/EU for animal research, reported according to the Animal Research: Reporting of In Vivo Experiments (ARRIVE) criteria, complied with the ‘3Rs’ measure and were approved by the ethical review board of the government of Upper Bavaria (Regierung von Oberbayern, Munich, Germany) and conformed to institutional guidelines of Klinikum der Universität München/Ludwig Maximilian University of Munich). The severity of the procedure was low.

Extended Data Fig. 3 Performances of different kinds of nanobodies in passive-vDISCO for Thy1-GFPM line.

ae, Light-sheet microscopy images of half mouse brains from Thy1-GFPM lines showing the performances of different batches of nanobodies from different sources and companies using standard passive-vDISCO (ac) and mild passive-vDISCO (d and e). The used nanobody is indicated in the panel title. f, Illustration indicating the anatomic region of the brain (green dashed) that was displayed in ae. Note that the imaging was taken either on the right or on the left hemisphere; right hemisphere images were flipped to ease the comparison between the different nanobodies. All the results in this figure were similarly observed in at least two independent experiments for each kind of nanobody. Animal experiments followed European directive 2010/63/EU for animal research, reported according to the Animal Research: Reporting of In Vivo Experiments (ARRIVE) criteria, complied with the ‘3Rs’ measure and were approved by the ethical review board of the government of Upper Bavaria (Regierung von Oberbayern, Munich, Germany) and conformed to institutional guidelines of Klinikum der Universität München/Ludwig Maximilian University of Munich). The severity of the procedure was low.

Extended Data Fig. 4 Strategy to make spinal cord straight for passive-vDISCO.

a, Required materials: a plastic Pasteur pipette and some fine needles. The cyan arrowheads indicate the cutting points. b, The plastic Pasteur pipette is then longitudinally cut in half. c, Positioning of the needles to constrain the brain with the spinal cord inside one of the halves of the pipette. d, The whole setting is put into a container such as a 50 ml tube for passive-vDISCO protocol. Animal experiments followed European directive 2010/63/EU for animal research, reported according to the Animal Research: Reporting of In Vivo Experiments (ARRIVE) criteria, complied with the ‘3Rs’ measure and were approved by the ethical review board of the government of Upper Bavaria (Regierung von Oberbayern, Munich, Germany) and conformed to institutional guidelines of Klinikum der Universität München/Ludwig Maximilian University of Munich). The severity of the procedure was low.

Extended Data Fig. 5 Mounting of cleared samples for epifluorescence imaging and inverted confocal imaging.

ad, Mounting of different samples for AxioZoom epifluorescence imaging: different glass containers used to mount cleared organs (red dashed circle) and slices (magenta boxes) for AxioZoom epifluorescence imaging (a); epifluorescence imaging of dissected organs (red arrowhead) and slices (magenta arrowhead) with the AxioZoom microscope (b and c); epifluorescence imaging of the whole body with the AxioZoom microscope (d). eg, Mounting of different samples for inverted confocal microscope imaging: a slice (e) and a whole brain (f) are placed onto a glass-bottom dish, then the dish with the lid is positioned onto the stage of the microscope (g). Animal experiments followed European directive 2010/63/EU for animal research, reported according to the Animal Research: Reporting of In Vivo Experiments (ARRIVE) criteria, complied with the ‘3Rs’ measure and were approved by the ethical review board of the government of Upper Bavaria (Regierung von Oberbayern, Munich, Germany) and conformed to institutional guidelines of Klinikum der Universität München/Ludwig Maximilian University of Munich). The severity of the procedure was low.

Extended Data Fig. 6 Mounting of cleared samples for light-sheet imaging.

aj, Various strategies to mount different samples for LaVision-Miltenyi light-sheet Ultramicroscope II imaging. A whole brain is mounted using the screw system provided by the microscope supplier (a); a slice (d) and a whole brain (e) are mounted using Micro-Fine Ultra needles (b) attached to the sample holder (ce). To mount a whole head using a flat sample holder (f), a piece of black tape is stuck to the flat surface (g, magenta arrowhead), then by adding super glue (h) and accelerator (i) the head is stabilized onto the tape (j). ko, Mounting of a whole-body sample for light-sheet imaging using the Blaze microscope: two pieces of black tape are sticked onto the mounting region of the samples holder (k and l, magenta arrowheads), superglue is applied onto the black tapes (m, magenta arrowheads), accelerator is injected into the superglue (n) and the animal is positioned onto the sample holder at the level of the tapes (o). pr, Strategy to remove air bubbles (p and r, red arrowheads) from the samples (e.g., a whole body) by inserting a fine needle into the bubble (q, cyan arrowhead) and sucking the bubble out with the syringe (r). Animal experiments followed European directive 2010/63/EU for animal research, reported according to the Animal Research: Reporting of In Vivo Experiments (ARRIVE) criteria, complied with the ‘3Rs’ measure and were approved by the ethical review board of the government of Upper Bavaria (Regierung von Oberbayern, Munich, Germany) and conformed to institutional guidelines of Klinikum der Universität München/Ludwig Maximilian University of Munich). The severity of the procedure was low.

Extended Data Fig. 7 Whole-body imaging with a mesoSPIM: setup modifications and sample handling.

a, Overview of the modified mesoSPIM setup: an existing mesoSPIM was modified by replacing the sample XYZ translation stages with stages with larger travel range and by adding a second detection path in the front of the setup. b, Design of the modified detection path with a telecentric detection lens with fixed magnification and a camera with high pixel count. c, The sample was mounted in a custom 40 × 40 × 120 mm3 quartz cuvette. The cuvette was closed with a custom lid that includes a kinematic mount with magnets that attach to the XYZ stages. d, To stabilize the sample inside the cuvette, a 3D-printed crossbar with 40 mm length was inserted above the sample and gently pressed down. e, View of the sample between the mesoSPIM excitation lenses before the front detection path was inserted. Animal experiments followed European directive 2010/63/EU for animal research, reported according to the Animal Research: Reporting of In Vivo Experiments (ARRIVE) criteria, complied with the ‘3Rs’ measure and were approved by the ethical review board of the government of Upper Bavaria (Regierung von Oberbayern, Munich, Germany) and conformed to institutional guidelines of Klinikum der Universität München/Ludwig Maximilian University of Munich). The severity of the procedure was low.

Extended Data Fig. 8 Whole-brain imaging with Zeiss Lightsheet Z.1.

a, The cleared brain sample is glued to the Lightsheet Z.1 rotatable sample holder. b, After mounting the sample holder into the microscope and inserting the imaging chamber containing the clearing solution, the sample is positioned in front of the detection objective. The right position in X, Y, Z and the rotation angle are monitored via the door camera of the microscope. c, The focus plane is adjusted using Near-IR pseudo-bright-field illumination. Animal experiments followed European directive 2010/63/EU for animal research, reported according to the Animal Research: Reporting of In Vivo Experiments (ARRIVE) criteria, complied with the ‘3Rs’ measure and were approved by the ethical review board of the government of Upper Bavaria (Regierung von Oberbayern, Munich, Germany) and conformed to institutional guidelines of Klinikum der Universität München/Ludwig Maximilian University of Munich). The severity of the procedure was low.

Supplementary information

Supplementary Information

Supplementary Figs. 1–3.

Reporting Summary

Supplementary Video 1

Stomach incision and cleaning as a sample preparation step in vDISCO whole-body clearing protocol

Supplementary Video 2

Gut incision and cleaning as a sample preparation step in vDISCO whole-body clearing protocol

Supplementary Video 3

vDISCO clearing protocol pump set-up and demonstration of how to refresh the pumping reference tube slot

Supplementary Video 4

The vDISCO processed Prox1-eGFP mouse with details of lymphatic vessels in the whole body, neurons in the hippocampus and satellite cells in the muscle tissue

Supplementary Video 5

The vDISCO processed brain from an adult Emx1-Cre x ROGT injected with a EnvA-pseudotyped G-deleted rabies virus expressing GFP (SADB1969) that is visible in the neocortex and in the striatum

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

Cai, R., Kolabas, Z.I., Pan, C. et al. Whole-mouse clearing and imaging at the cellular level with vDISCO. Nat Protoc 18, 1197–1242 (2023). https://doi.org/10.1038/s41596-022-00788-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-022-00788-2

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