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:

High-resolution 3D imaging of fixed and cleared organoids

Abstract

In vitro 3D organoid systems have revolutionized the modeling of organ development and diseases in a dish. Fluorescence microscopy has contributed to the characterization of the cellular composition of organoids and demonstrated organoids’ phenotypic resemblance to their original tissues. Here, we provide a detailed protocol for performing high-resolution 3D imaging of entire organoids harboring fluorescence reporters and upon immunolabeling. This method is applicable to a wide range of organoids of differing origins and of various sizes and shapes. We have successfully used it on human airway, colon, kidney, liver and breast tumor organoids, as well as on mouse mammary gland organoids. It includes a simple clearing method utilizing a homemade fructose–glycerol clearing agent that captures 3D organoids in full and enables marker quantification on a cell-by-cell basis. Sample preparation has been optimized for 3D imaging by confocal, super-resolution confocal, multiphoton and light-sheet microscopy. From organoid harvest to image analysis, the protocol takes 3 d.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Overview of the high-resolution 3D imaging protocol.
Fig. 2: Representative 3D whole-mount organoid images obtained with different light microscopy technologies.
Fig. 3: Comparison of different optical clearing agents and single- and multiphoton imaging.
Fig. 4: 3D imaging enables acquisition of accurate architectural information and 3D quantification.

Similar content being viewed by others

Data availability

All data generated or analyzed during this study are included in this published article (and its supplementary information files).

References

  1. Sato, T. & Clevers, H. SnapShot: growing organoids from stem cells. Cell 161, 1700–1700.e1 (2015).

    Article  CAS  Google Scholar 

  2. Clevers, H. Modeling development and disease with organoids. Cell 165, 1586–1597 (2016).

    Article  CAS  Google Scholar 

  3. Drost, J. & Clevers, H. Organoids in cancer research. Nat. Rev. Cancer 18, 407–418 (2018).

    Article  CAS  Google Scholar 

  4. Bartfeld, S. & Clevers, H. Stem cell-derived organoids and their application for medical research and patient treatment. J. Mol. Med. 95, 729–738 (2017).

    Article  CAS  Google Scholar 

  5. Broutier, L. et al. Human primary liver cancer-derived organoid cultures for disease modeling and drug screening. Nat. Med. 23, 1424–1435 (2017).

    Article  CAS  Google Scholar 

  6. Dekkers, J. F. et al. Characterizing responses to CFTR-modulating drugs using rectal organoids derived from subjects with cystic fibrosis. Sci. Transl. Med. 8, 344ra384 (2016).

    Article  Google Scholar 

  7. Dekkers, J. F. et al. A functional CFTR assay using primary cystic fibrosis intestinal organoids. Nat. Med. 19, 939–945 (2013).

    Article  CAS  Google Scholar 

  8. Sato, T. et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature 459, 262–265 (2009).

    Article  CAS  Google Scholar 

  9. Karthaus, W. R. et al. Identification of multipotent luminal progenitor cells in human prostate organoid cultures. Cell 159, 163–175 (2014).

    Article  CAS  Google Scholar 

  10. Lancaster, M. A. et al. Cerebral organoids model human brain development and microcephaly. Nature 501, 373–379 (2013).

    Article  CAS  Google Scholar 

  11. Hu, H. et al. Long-term expansion of functional mouse and human hepatocytes as 3D organoids. Cell 175, 1591–1606.e19 (2018).

    Article  CAS  Google Scholar 

  12. Huch, M. et al. Long-term culture of genome-stable bipotent stem cells from adult human liver. Cell 160, 299–312 (2015).

    Article  CAS  Google Scholar 

  13. Nanki, K. et al. Divergent routes toward Wnt and R-spondin niche independency during human gastric carcinogenesis. Cell 174, 856–869.e17 (2018).

    Article  CAS  Google Scholar 

  14. Jamieson, P. R. et al. Derivation of a robust mouse mammary organoid system for studying tissue dynamics. Development 144, 1065–1071 (2017).

    Article  CAS  Google Scholar 

  15. Sachs, N. et al. A living biobank of breast cancer organoids captures disease heterogeneity. Cell 172, 373–386.e10 (2018).

    Article  CAS  Google Scholar 

  16. Turco, M. Y. et al. Long-term, hormone-responsive organoid cultures of human endometrium in a chemically defined medium. Nat. Cell Biol. 19, 568–577 (2017).

    Article  CAS  Google Scholar 

  17. Maimets, M. et al. Long-term in vitro expansion of salivary gland stem cells driven by Wnt signals. Stem Cell Rep. 6, 150–162 (2016).

    Article  CAS  Google Scholar 

  18. Ren, W. et al. Single Lgr5- or Lgr6-expressing taste stem/progenitor cells generate taste bud cells ex vivo. Proc. Natl. Acad. Sci. USA 111, 16401–16406 (2014).

    Article  CAS  Google Scholar 

  19. Richardson, D. S. & Lichtman, J. W. Clarifying tissue clearing. Cell 162, 246–257 (2015).

    Article  CAS  Google Scholar 

  20. Richardson, D. S. & Lichtman, J. W. SnapShot: tissue clearing. Cell 171, 496–496.e1 (2017).

    Article  CAS  Google Scholar 

  21. Rios, A. C. et al. Essential role for a novel population of binucleated mammary epithelial cells in lactation. Nat. Commun. 7, 11400 (2016).

    Article  CAS  Google Scholar 

  22. Rios, A. C., Fu, N. Y., Lindeman, G. J. & Visvader, J. E. In situ identification of bipotent stem cells in the mammary gland. Nature 506, 322–327 (2014).

    Article  CAS  Google Scholar 

  23. Rios, A. C. & Clevers, H. Imaging organoids: a bright future ahead. Nat. Methods 15, 24–26 (2018).

    Article  CAS  Google Scholar 

  24. Sachs, N. et al. Long‐term expanding human airway organoids for disease modeling. EMBO J. 38, e100300 (2019).

    Article  Google Scholar 

  25. Shutgens, F. et al. Tubuloids derived from human adult kidney and urine for personalized disease modelling. Nat. Biotechnol. 37, 303–313 (2019).

    Article  Google Scholar 

  26. Eisenstein, M. Transparent tissues bring cells into focus for microscopy. Nature 564, 147–149 (2018).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  30. 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  Google Scholar 

  31. 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  Google Scholar 

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

    Article  CAS  Google Scholar 

  33. 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  Google Scholar 

  34. Snippert, H. J. et al. Intestinal crypt homeostasis results from neutral competition between symmetrically dividing Lgr5 stem cells. Cell 143, 134–144 (2010).

    Article  CAS  Google Scholar 

  35. Stelzer, E. H. Light-sheet fluorescence microscopy for quantitative biology. Nat. Methods 12, 23–26 (2015).

    Article  CAS  Google Scholar 

  36. Huisken, J., Swoger, J., Del Bene, F., Wittbrodt, J. & Stelzer, E. H. Optical sectioning deep inside live embryos by selective plane illumination microscopy. Science 305, 1007–1009 (2004).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We are very grateful for the technical support from the Princess Máxima Center for Pediatric Oncology and to the Hubrecht Institute and Zeiss for imaging support and collaborations. All the imaging was performed at the Princess Máxima imaging center. This work was financially supported by the Princess Máxima Center for Pediatric Oncology. J.F.D. was supported by a Marie Curie Global Fellowship and a VENI grant from the Netherlands Organisation for Scientific Research (NWO). J.E.V. was supported by the Australian National Health and Medical Research Council (NHMRC).

Author information

Authors and Affiliations

Authors

Contributions

J.F.D. designed the study, performed experiments and interpreted data; J.F.D., M.A., L.M.W., H.C.R.A. and P.R.J. performed experiments and analyzed data; J.F.D., P.R.J., A.M.V., G.D.A., H.H. and J.M.B. performed the organoid culturing. K.C.O. and H.J.G.S. provided fluorescent constructs; A.C.R. helped design the study and carried out data interpretation; J.F.D. and A.C.R. cowrote the manuscript. J.E.V., H.C. and E.J.W. helped with data interpretation, manuscript writing and corrections.

Corresponding author

Correspondence to Anne C. Rios.

Ethics declarations

Competing interests

H.C. is named as inventor on several patents related to organoid technology. J.F.D. is named as inventor on one patent related to the organoid technology.

Additional information

Journal peer review information: Nature Protocols thanks Xavier Gidrol, Melissa Skala 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 links

Key references using this protocol

Hu, H. et al. Cell 175, 1591–1606.e19 (2018): https://doi.org/10.1016/j.cell.2018.11.013

Sachs, N. et al. EMBO J. 38, e100300 (2019): https://doi.org/10.15252/embj.2018100300

Supplementary information

Supplementary Video 1

This video highlights the intricate 3D features of delicate organoid structures that can be imaged at cellular or even subcellular resolution with this sample-preparation protocol.

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dekkers, J.F., Alieva, M., Wellens, L.M. et al. High-resolution 3D imaging of fixed and cleared organoids. Nat Protoc 14, 1756–1771 (2019). https://doi.org/10.1038/s41596-019-0160-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-019-0160-8

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: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research