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:

Real-time imaging of multivesicular body–plasma membrane fusion to quantify exosome release from single cells

Abstract

Exosomes are small extracellular vesicles with a diameter of 40–150 nm, and are implicated in cellular homeostasis and cell–cell communication. They can be secreted in bulk in response to cell-extrinsic and cell-intrinsic signals that cause multivesicular body (MVB) fusion with the plasma membrane (PM). However, research on the regulation of exosome release is hampered by the failure of current methods to capture the dynamics of exosome release. Here we describe how live imaging with tetraspanin-based pH-sensitive fluorescent reporters can quantify the MVB–PM fusion rate of single cells. Our approach enables identification of exogenous stimuli, signaling pathways, and fusion complexes, and can map subcellular sites of fusion events. In addition, dual-color imaging can be used to assess simultaneous release of different cargo by MVB exocytosis. This protocol describes the complete imaging experiment, consisting of transient expression of tetraspanin reporters (2 d), live-cell (dual-color) total internal reflection fluorescence microscopy (30–60 min per condition), and semiautomatic image analysis by using a newly developed ImageJ macro (±30 min per condition).

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: Model for the visualization of MVB–PM fusion with CD63-pHluorin.
Fig. 2: Simultaneous dual-color TIRF microscopy of TSPAN-pHluorin with TSPAN-pHuji reporters.
Fig. 3: Visualization of individual fusion events.
Fig. 4: Setting the TIRF angle for a CD63-pHluorin-expressing HeLa cell.
Fig. 5: Analysis of MVB–PM fusion activity with the AMvBE macro.

Similar content being viewed by others

Data availability

The data that support this study are available from the corresponding author upon reasonable request.

Code availability

The AMvBE macro, a Readme file, and example data can be found in Supplementary Software 1. The software in this protocol has been peer reviewed.

References

  1. Huotari, J. & Helenius, A. Endosome maturation. EMBO J. 30, 3481–3500 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. van Niel, G., D’Angelo, G. & Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Biol. 19, 213–228 (2018).

    CAS  PubMed  Google Scholar 

  3. Bebelman, M. P., Smit, M. J., Pegtel, D. M. & Baglio, S. R. Biogenesis and function of extracellular vesicles in cancer. Pharmacol Ther. 188, 1–11 (2018).

    CAS  PubMed  Google Scholar 

  4. Costa-Silva, B. et al. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat. Cell Biol. 17, 816–826 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Peinado, H. et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat. Med. 18, 883–891 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Bobrie, A., Colombo, M., Raposo, G. & Thery, C. Exosome secretion: molecular mechanisms and roles in immune responses. Traffic 12, 1659–1668 (2011).

    CAS  PubMed  Google Scholar 

  7. Bian, S. et al. Extracellular vesicles derived from human bone marrow mesenchymal stem cells promote angiogenesis in a rat myocardial infarction model. J. Mol. Med. 92, 387–397 (2014).

    CAS  PubMed  Google Scholar 

  8. Tkach, M. & Thery, C. Communication by extracellular vesicles: where we are and where we need to go. Cell 164, 1226–1232 (2016).

    CAS  PubMed  Google Scholar 

  9. Kowal, J. et al. Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes. Proc. Natl Acad. Sci. USA 113, E968–E977 (2016).

    CAS  PubMed  Google Scholar 

  10. van der Vlist, E. J., Nolte-‘t Hoen, E. N., Stoorvogel, W., Arkesteijn, G. J. & Wauben, M. H. Fluorescent labeling of nano-sized vesicles released by cells and subsequent quantitative and qualitative analysis by high-resolution flow cytometry. Nat. Protoc. 7, 1311–1326 (2012).

    PubMed  Google Scholar 

  11. Zhang, H. & Lyden, D. Asymmetric-flow field-flow fractionation technology for exomere and small extracellular vesicle separation and characterization. Nat. Protoc. 14, 1027–1053 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Gardiner, C. et al. Techniques used for the isolation and characterization of extracellular vesicles: results of a worldwide survey. J. Extracell. Vesicles 5, 32945 (2016).

    PubMed  Google Scholar 

  13. Pegtel, D. M. & Gould, S. J. Exosomes. Annu. Rev. Biochem 88, 487–514 (2019).

    CAS  PubMed  Google Scholar 

  14. Miesenbock, G., De Angelis, D. A. & Rothman, J. E. Visualizing secretion and synaptic transmission with pH-sensitive green fluorescent proteins. Nature 394, 192–195 (1998).

    CAS  PubMed  Google Scholar 

  15. Sankaranarayanan, S., De Angelis, D., Rothman, J. E. & Ryan, T. A. The use of pHluorins for optical measurements of presynaptic activity. Biophys. J. 79, 2199–2208 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Sung, B. H., Ketova, T., Hoshino, D., Zijlstra, A. & Weaver, A. M. Directional cell movement through tissues is controlled by exosome secretion. Nat. Commun. 6, 7164 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Verweij, F. J. et al. Quantifying exosome secretion from single cells reveals a modulatory role for GPCR signaling. J. Cell Biol. 217, 1129–1142 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Lu, A. et al. Genome-wide interrogation of extracellular vesicle biology using barcoded miRNAs. eLife 7, e41460 (2018).

    PubMed  PubMed Central  Google Scholar 

  19. Gutknecht, J. Proton/hydroxide conductance and permeability through phospholipid bilayer membranes. Proc. Natl Acad. Sci. USA 84, 6443–6446 (1987).

    CAS  PubMed  Google Scholar 

  20. Paula, S., Volkov, A. G., Van Hoek, A. N., Haines, T. H. & Deamer, D. W. Permeation of protons, potassium ions, and small polar molecules through phospholipid bilayers as a function of membrane thickness. Biophys. J. 70, 339–348 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Raven, J. A. & Beardall, J. The intrinsic permeability of biological membranes to H+: Significance for the efficiency of low rates of energy transformation. FEMS Microbiol. Lett. 10, 1–5 (1981).

    CAS  Google Scholar 

  22. Verweij, F. J. et al. LMP1 association with CD63 in endosomes and secretion via exosomes limits constitutive NF-κB activation. EMBO J. 30, 2115–2129 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Sung, B. H., Pelletier, R. & Weaver, A. M. pHluo_M153R-CD63, a bright, versatile live cell reporter of exosome secretion and uptake, reveals pathfinding behavior of migrating cells. Preprint at https://www.biorxiv.org/content/10.1101/577346v1 (2019).

  24. Mittelbrunn, M. et al. Unidirectional transfer of microRNA-loaded exosomes from T cells to antigen-presenting cells. Nat. Commun. 2, 282 (2011).

    PubMed  PubMed Central  Google Scholar 

  25. van Niel, G. et al. Intestinal epithelial cells secrete exosome-like vesicles. Gastroenterology 121, 337–349 (2001).

    PubMed  Google Scholar 

  26. Hoshino, D. et al. Exosome secretion is enhanced by invadopodia and drives invasive behavior. Cell Rep. 5, 1159–1168 (2013).

    CAS  PubMed  Google Scholar 

  27. Verweij, F. J. et al. Live tracking of inter-organ communication by endogenous exosomes in vivo. Dev. Cell 48, 573–589.e4 (2019).

    CAS  PubMed  Google Scholar 

  28. Van Deun, J. et al. EV-TRACK: transparent reporting and centralizing knowledge in extracellular vesicle research. Nat. Methods 14, 228–232 (2017).

    PubMed  Google Scholar 

  29. van der Pol, E. et al. Particle size distribution of exosomes and microvesicles determined by transmission electron microscopy, flow cytometry, nanoparticle tracking analysis, and resistive pulse sensing. J. Thromb. Haemost. 12, 1182–1192 (2014).

    PubMed  Google Scholar 

  30. Yuana, Y., Levels, J., Grootemaat, A., Sturk, A. & Nieuwland, R. Co-isolation of extracellular vesicles and high-density lipoproteins using density gradient ultracentrifugation. J. Extracell. Vesicles 3, https://doi.org/10.3402/jev.v3.23262 (2014).

    Google Scholar 

  31. Coumans, F. A. W. et al. Methodological Guidelines to Study Extracellular Vesicles. Circ. Res. 120, 1632–1648 (2017).

    CAS  PubMed  Google Scholar 

  32. Linares, R., Tan, S., Gounou, C., Arraud, N. & Brisson, A. R. High-speed centrifugation induces aggregation of extracellular vesicles. J. Extracell. Vesicles 4, 29509 (2015).

    PubMed  Google Scholar 

  33. Jeppesen, D. K. et al. Reassessment of exosome composition. Cell 177, 428–445.e18 (2019).

    CAS  PubMed  Google Scholar 

  34. Zhang, H. et al. Identification of distinct nanoparticles and subsets of extracellular vesicles by asymmetric flow field-flow fractionation. Nat. Cell Biol. 20, 332–343 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Messenger, S. W., Woo, S. S., Sun, Z. & Martin, T. F. J. A Ca2+-stimulated exosome release pathway in cancer cells is regulated by Munc13-4. J. Cell Biol. 217, 2877–2890 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Nabhan, J. F., Hu, R., Oh, R. S., Cohen, S. N. & Lu, Q. Formation and release of arrestin domain-containing protein 1-mediated microvesicles (ARMMs) at plasma membrane by recruitment of TSG101 protein. Proc. Natl Acad. Sci. USA 109, 4146–4151 (2012).

    CAS  PubMed  Google Scholar 

  37. Colombo, M., Raposo, G. & Thery, C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu. Rev. Cell Dev. Biol. 30, 255–289 (2014).

    CAS  PubMed  Google Scholar 

  38. Shen, Y., Rosendale, M., Campbell, R. E. & Perrais, D. pHuji, a pH-sensitive red fluorescent protein for imaging of exo- and endocytosis. J. Cell Biol. 207, 419–432 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. de Wit, J., Toonen, R. F. & Verhage, M. Matrix-dependent local retention of secretory vesicle cargo in cortical neurons. J. Neurosci. 29, 23–37 (2009).

    PubMed  PubMed Central  Google Scholar 

  40. Urbina, F. L., Gomez, S. M. & Gupton, S. L. Spatiotemporal organization of exocytosis emerges during neuronal shape change. J. Cell Biol. 217, 1113–1128 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Yuan, T., Lu, J., Zhang, J., Zhang, Y. & Chen, L. Spatiotemporal detection and analysis of exocytosis reveal fusion “hotspots” organized by the cytoskeleton in endocrine cells. Biophys. J. 108, 251–260 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  Google Scholar 

  43. Thevenaz, P., Ruttimann, U. E. & Unser, M. A pyramid approach to subpixel registration based on intensity. IEEE Trans. Image Process. 7, 27–41 (1998).

    CAS  PubMed  Google Scholar 

  44. Mattheyses, A. L., Simon, S. M. & Rappoport, J. Z. Imaging with total internal reflection fluorescence microscopy for the cell biologist. J. Cell Sci. 123, 3621–3628 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We acknowledge the NeurImag facility of the Institute of Psychiatry and Neuroscience of Paris where the AMvBE macro has been developed. This Protocol was funded by a Dutch Organizations for Scientific Research–Amsterdam Institute for Molecules, Medicines, and Systems STAR Graduate Program grant (022.005.031) to M.P.B., a Dutch Cancer Fund (KWF-5510) and a Cancer Center Amsterdam–VU University Medical Center grant to D.M.P., and a European Molecular Biology Organization grant (EMBO ALTF 1383-2014) and a Fondation ARC pour la Recherché sur le Cancer fellowship (PJA 20161204808) to F.J.V.

Author information

Authors and Affiliations

Authors

Contributions

M.P.B. and F.J.V. performed the experiments, prepared the figures, and wrote the paper. G.v.N., P.B., S.H., and D.M.P. provided critical feedback and helped to shape the paper. P.B. developed the AMvBE macro. S.H. helped to set up the dual-color TIRF experiments. D.M.P. conceived the study. F.J.V. and D.M.P. supervised the study.

Corresponding authors

Correspondence to D. Michiel Pegtel or Frederik J. Verweij.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Protocols thanks David Perrais and the 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 references using this protocol

Verweij, F. J. et al. J Cell Biol, 217 (3) 1129–1142 (2018): http://jcb.rupress.org/content/217/3/1129/

Supplementary information

Supplementary Software 1

AMvBE macro, Readme file and example data

Reporting Summary

Supplementary Video 1

TIRF microscopy of a CD63-pHluorin expressing HeLa cell at 8× speed. Scale bar, 20 μm

Supplementary Video 2

Dual-color TIRF microscopy of a CD63-pHluorin (green) and CD81-pHuji (red) expressing HeLa cell at 8× speed. Scale bar, 20 μm

Supplementary Video 3

Dual-color TIRF microscopy of a CD63-pHluorin (green) and CD63-mRFP (red) expressing HeLa cell at 8× speed. Scale bar, 20 μm. White arrows highlight MVBs containing CD63-pHluorin and CD63-mRFP that are visible in red before fusion

Supplementary Video 4

Dual-color TIRF microscopy of a CD63-C-term-pHluorin (green) and CD63- pHuji (red) expressing HeLa cell at 8× speed. Scale bar, 20 μm. Original source: ref. 17

Supplementary Video 5

TIRF microscopy of HeLa cells stably expressing CD63-pHluorin 6 weeks post-transduction at 8× speed. Scale bar, 20 μm

Supplementary Video 6

TIRF microscopy of a CD63-pHluorin expressing HUVEC cell at 8× speed. Scale bar, 20 μm

Supplementary Video 7

TIRF microscopy of a CD63-pHluorin expressing HEK293T cell at 8× speed. Scale bar, 20 μm

Supplementary Video 8

Example of the summary TIFF file as created by the AMvBE macro upon analysis of an MVB-PM fusion event (highlighted by a green circle) or a neutral CD63-pHluorin-positive vesicle moving in the TIRF plane (highlighted by a red circle). Scale bar, 5 μm

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bebelman, M.P., Bun, P., Huveneers, S. et al. Real-time imaging of multivesicular body–plasma membrane fusion to quantify exosome release from single cells. Nat Protoc 15, 102–121 (2020). https://doi.org/10.1038/s41596-019-0245-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-019-0245-4

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