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.

  • Letter
  • Published:

Matrix mechanics and water permeation regulate extracellular vesicle transport

Abstract

Cells release extracellular vesicles (EVs) to communicate over long distances, which requires EVs to traverse the extracellular matrix (ECM). However, given that the size of EVs is usually larger than the mesh size of the ECM, it is not clear how they can travel through the dense ECM. Here we show that, in contrast to synthetic nanoparticles, EVs readily transport through nanoporous ECM. Using engineered hydrogels, we demonstrate that the mechanical properties of the matrix regulate anomalous EV transport under confinement. Matrix stress relaxation allows EVs to overcome the confinement, and a higher crosslinking density facilitates a fluctuating transport motion through the polymer mesh, which leads to free diffusion and fast transport. Furthermore, water permeation through aquaporin-1 mediates the EV deformability, which further supports EV transport in hydrogels and a decellularized matrix. Our results provide evidence for the nature of EV transport within confined environments and demonstrate an unexpected dependence on matrix mechanics and water permeation.

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: EVs transport within decellularized lung tissue.
Fig. 2: Complex shear modulus and stress relaxation time regulate the bulk release of EVs from hydrogels.
Fig. 3: Individual EVs exhibit anomalous transport that is more rapid and diffusive in a stiff stress relaxing matrix.
Fig. 4: Aquaporin-1 mediates the ability of EVs to transport in engineered and decellularized matrices by increasing the EV deformability.
Fig. 5: Model for EV transport under confinement.

Similar content being viewed by others

Data availability

The data that support the plots within this paper and other findings of this study are available from the corresponding author upon reasonable request.

Code availability

The codes used to analyse the data in this study are available from the corresponding author upon reasonable request.

References

  1. Huleihel, L. et al. Matrix-bound nanovesicles within ECM bioscaffold. Sci. Adv. 6, e1600502 (2015).

    Google Scholar 

  2. Meldolesi, J. Exosomes and ectosomes in intercellular communication. Curr. Biol. 28, R435–R444 (2018).

    CAS  Google Scholar 

  3. Tomlins, P., Grant, P., Mikhalovsky, S., James, S. & Mikhalovska, L. Measurement of pore size and porosity of tissue scaffolds. J. ASTM Int. 1, 1–8 (2004).

    Google Scholar 

  4. Luker, K. E. et al. Comparative study reveals better far-red fluorescent protein for whole body imaging. Sci. Rep. 5, 10332 (2015).

    CAS  Google Scholar 

  5. Rakian, R. et al. Native extracellular matrix preserves mesenchymal stem cell ‘stemness’ and differentiation potential under serum-free culture conditions. Stem Cell Res. Ther. 6, 235 (2015).

    Google Scholar 

  6. Vining, K. H., Stafford, A. & Mooney, D. J. Sequential modes of crosslinking tune viscoelasticity of cell-instructive hydrogels. Biomaterials 188, 187–197 (2019).

    CAS  Google Scholar 

  7. Lee, K. Y. & Mooney, D. J. Alginate: properties and biomedical applications. Prog. Polym. Sci. 37, 106–126 (2012).

    CAS  Google Scholar 

  8. Chaudhuri, O. et al. Hydrogels with tunable stress relaxation regulate stem cell fate and activity. Nat. Mater. 15, 326–334 (2016).

    CAS  Google Scholar 

  9. Grant, G. T., Morris, E. R., Rees, D. A., Smith, P. J. C. & Thom, D. Biological interactions between polysaccharides and divalent cations: the egg-box model. FEBS Lett. 32, 195–198 (1973).

    CAS  Google Scholar 

  10. Armstrong, J. K., Wenby, R. B., Meiselman, H. J. & Fisher, T. C. The hydrodynamic radii of macromolecules and their effect on red blood cell aggregation. Biophys. J. 87, 4259–4270 (2004).

    CAS  Google Scholar 

  11. Skotland, T., Hessvik, N. P., Sandvig, K. & Llorente, A. Exosomal lipid composition and the role of ether lipids and phosphoinositides in exosome biology. J. Lipid Res. 60, 9–18 (2019).

    CAS  Google Scholar 

  12. Branco da Cunha, C. et al. Influence of the stiffness of three-dimensional alginate/collagen-I interpenetrating networks on fibroblast biology. Biomaterials 35, 8927–8936 (2014).

    CAS  Google Scholar 

  13. Metzler, R., Jeon, J. H., Cherstvy, A. G. & Barkai, E. Anomalous diffusion models and their properties: non-stationarity, non-ergodicity, and ageing at the centenary of single particle tracking. Phys. Chem. Chem. Phys. 16, 24128–24164 (2014).

    CAS  Google Scholar 

  14. Etoc, F. et al. Non-specific interactions govern cytosolic diffusion of nanosized objects in mammalian cells. Nat. Mater. 17, 740–746 (2018).

    CAS  Google Scholar 

  15. Schirmache, W., Ruocco, G. & Mazzone, V. Heterogeneous viscoelasticity: a combined theory of dynamic and elastic heterogeneity. Phys. Rev. Lett. 115, 015901 (2015).

    Google Scholar 

  16. Lieleg, O., Vladescu, I. & Ribbeck, K. Characterization of particle translocation through mucin hydrogels. Biophys. J. 98, 1782–1789 (2010).

    CAS  Google Scholar 

  17. Goiko, M., De Bruyn, J. R. & Heit, B. Short-lived cages restrict protein diffusion in the plasma membrane. Sci. Rep. 6, 34987 (2016).

    CAS  Google Scholar 

  18. Weigel, A. V., Simon, B., Tamkun, M. M. & Krapf, D. Ergodic and nonergodic processes coexist in the plasma membrane as observed by single-molecule tracking. Proc. Natl Acad. Sci. USA 108, 6438–6443 (2011).

    CAS  Google Scholar 

  19. Manzo, C. et al. Weak ergodicity breaking of receptor motion in living cells stemming from random diffusivity. Phys. Rev. X 5, 011021- (2015).

    Google Scholar 

  20. Parry, B. R. et al. The bacterial cytoplasm has glass-like properties and is fluidized by metabolic activity. Cell 156, 183–194 (2014).

    CAS  Google Scholar 

  21. Kusuma, G. D. et al. To protect and to preserve: novel preservation strategies for extracellular vesicles. Front. Pharmacol. 9, 01199 (2018).

    CAS  Google Scholar 

  22. Frank, J. et al. Extracellular vesicles protect glucuronidase model enzymes during freeze-drying. Sci. Rep. 8, 12377 (2018).

    Google Scholar 

  23. Stroka, K. M. et al. Water permeation drives tumor cell migration in confined microenvironments. Cell 157, 611–623 (2014).

    CAS  Google Scholar 

  24. Blanc, L. et al. The water channel aquaporin-1 partitions into exosomes during reticulocyte maturation: implication for the regulation of cell volume. Blood 114, 3928–3934 (2009).

    CAS  Google Scholar 

  25. Cai, L. H., Panyukov, S. & Rubinstein, M. Hopping diffusion of nanoparticles in polymer matrices. Macromolecules 48, 847–862 (2015).

    CAS  Google Scholar 

  26. Wong, I. Y. et al. Anomalous diffusion probes microstructure dynamics of entangled F-actin networks. Phys. Rev. Lett. 92, 178101 (2004).

    CAS  Google Scholar 

  27. Ritchie, K. et al. Detection of non-Brownian diffusion in the cell membrane in single molecule tracking. Biophys. J. 88, 2266–2277 (2005).

    CAS  Google Scholar 

  28. Mathieu, M., Martin-Jaular, L., Lavieu, G. & Théry, C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat. Cell Biol. 21, 9–17 (2019).

    CAS  Google Scholar 

  29. Manno, S., Takakuwa, Y. & Mohandas, N. Identification of a functional role for lipid asymmetry in biological membranes: phosphatidylserine-skeletal protein interactions modulate membrane stability. Proc. Natl Acad. Sci. USA 99, 1943–1948 (2002).

    CAS  Google Scholar 

  30. Guo, P. et al. Nanoparticle elasticity directs tumor uptake. Nat. Commun. 9, 130 (2018).

    Google Scholar 

  31. McGinley, L. et al. Lentiviral vector mediated modification of mesenchymal stem cells & enhanced survival in an in vitro model of ischaemia. Stem Cell Res. Ther. 2, 12 (2011).

    CAS  Google Scholar 

  32. Lobb, R. J. et al. Optimized exosome isolation protocol for cell culture supernatant and human plasm. J. Extracell. Vesicles 4, 27031 (2015).

    Google Scholar 

  33. Bonenfant, N. R. et al. The effects of storage and sterilization on de-cellularized and re-cellularized whole lung. Biomaterials 34, 3231–3245 (2013).

    CAS  Google Scholar 

  34. Pena, A. M. et al. Three-dimensional investigation and scoring of extracellular matrix remodeling during lung fibrosis using multiphoton microscopy. Microsc. Res. Tech. 70, 162–170 (2007).

    Google Scholar 

  35. Jos, RANDBLOCK (10584) (MATLAB Central File Exchange, accessed 28 May 2019); https://www.mathworks.com/matlabcentral/fileexchange/17981-randblock

  36. Desai, R. M., Koshy, S. T., Hilderbrand, S. A., Mooney, D. J. & Joshi, N. S. Versatile click alginate hydrogels crosslinked via tetrazine-norbornene chemistry. Biomaterials 50, 30–37 (2015).

    CAS  Google Scholar 

  37. Chaudhuri, O. et al. Substrate stress relaxation regulates cell spreading. Nat. Commun. 6, 6365 (2015).

    CAS  Google Scholar 

  38. Hosford, W. F. Mechanical Behavior of Materials (Cambridge Univ. Press, 2005).

  39. Carr, D. A. & Peppas, N. A. Molecular structure of physiologically-responsive hydrogels controls diffusive behavior. Macromol. Biosci. 9, 497–505 (2009).

    CAS  Google Scholar 

  40. Berger, J. et al. Structure and interactions in covalently and ionically crosslinked chitosan hydrogels for biomedical applications. Eur. J. Pharm. Biopharm. 57, 19–34 (2004).

    CAS  Google Scholar 

  41. Boontheekul, T., Kong, H. J. & Mooney, D. J. Controlling alginate gel degradation utilizing partial oxidation and bimodal molecular weight distribution. Biomaterials 26, 2455–2465 (2005).

    CAS  Google Scholar 

  42. Iza, M., Woerly, S., Danumah, C., Kaliaguine, S. & Bousmina, M. Determination of pore size distribution for mesoporous materials and polymeric gels by means of DSC measurements: thermoporometry. Polymer 41, 5885–5893 (2000).

    CAS  Google Scholar 

  43. Backlund, M. P., Joyner, R. & Moerner, W. E. Chromosomal locus tracking with proper accounting of static and dynamic errors. Phys. Rev. E. 91, 062716 (2015).

    Google Scholar 

  44. Vorselen, D. et al. The fluid membrane determines mechanics of erythrocyte extracellular vesicles and is softened in hereditary spherocytosis. Nat. Commun. 9, 4960 (2018).

    Google Scholar 

  45. Segur, J. B. & Oderstar, H. E. Viscosity of glycerol and its aqueous solutions. Ind. Eng. Chem. 43, 2117–2120 (1951).

    CAS  Google Scholar 

  46. Calò, A. et al. Force measurements on natural membrane nanovesicles reveal a composition-independent, high Young’s modulus. Nanoscale 6, 2275–2285 (2014).

    Google Scholar 

Download references

Acknowledgements

We thank B. Hoffman (Duke University) and L. Cai (University of Virginia) for critical reading of the manuscript and invaluable comments. We acknowledge P. Toth and the Core Imaging Facility at UIC, J. Li at the Department of Pharmacology at UIC, T. Foroozan at the UIC Nanotechnology Core Facility, T. Teng and J. Lee at the Department of Bioengineering at UIC, A. Song at UIC and the ANTEC facility at Northwestern University for their technical help and support. This work made use of instruments in the Fluorescence Imaging Core (Research Resources Center, UIC). This work was supported by National Institutes of Health Grant R01-HL141255 (J.-W.S.), R00-HL125884 (J.-W.S.), T32 HL07829 (S.L.) and American Heart Association Grant 19PRE34380087 (S.L.).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, S.L. and J-W.S.; data curation, S.L.; formal analysis, S.L.; funding acquisition, S.L. and J-W.S.; investigation, S.L., R.B. and G.C.; methodology, S.L. and J-W.S.; project administration, S.L. and J-W.S.; resources, J-W.S.; software, S.L.; supervision, J-W.S.; validation, S.L., R.B., G.C. and J-W.S.; visualization, S.L.; writing original draft, S.L. and J-W.S; writing the revision, S.L. and J-W.S.

Corresponding author

Correspondence to Jae-Won Shin.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Nanotechnology thanks Gregor Fuhrmann, Neill Turner 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.

Supplementary information

Supplementary Information

Supplementary Figs. 1–9, Table 1 and captions for Supplementary Videos 1–6.

Reporting Summary

Supplementary Video 1

Tracking data overlaid with imaging data for representative transport of a single EV in stiff stress relaxing matrix shown in Fig. 3A. The length scale is micrometers and the time scale is seconds.

Supplementary Video 2

Tracking data overlaid with imaging data for representative transport of a single EV in soft stress relaxing matrix shown in Fig. 3A. The length scale is micrometers and the time scale is seconds.

Supplementary Video 3

Tracking data overlaid with imaging data for representative transport of a single EV in stiff elastic matrix shown in Fig. 3A. The length scale is micrometers and the time scale is seconds.

Supplementary Video 4

Tracking data overlaid with imaging data for representative transport of multiple EVs in stiff stress relaxing matrix. The length scale is micrometers and the time scale is seconds.

Supplementary Video 5

Tracking data overlaid with imaging data for representative transport of multiple EVs in soft stress relaxing matrix. The length scale is micrometers and the time scale is seconds.

Supplementary Video 6

Tracking data overlaid with imaging data for representative transport of multiple EVs in stiff elastic matrix. The length scale is micrometers and the time scale is seconds.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lenzini, S., Bargi, R., Chung, G. et al. Matrix mechanics and water permeation regulate extracellular vesicle transport. Nat. Nanotechnol. 15, 217–223 (2020). https://doi.org/10.1038/s41565-020-0636-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41565-020-0636-2

This article is cited by

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