Summary

Evaluation of the Storage Stability of Extracellular Vesicles

Published: May 22, 2019
doi:

Summary

Here we present a readily applicable protocol to assess the storage stability of extracellular vesicles, a group of naturally occurring nanoparticles produced by cells. The vesicles are loaded with glucuronidase as a model enzyme and stored under different conditions. After storage, their physicochemical parameters and the activity of the encapsulated enzyme are evaluated.

Abstract

Extracellular vesicles (EVs) are promising targets in current research, to be used as drugs, drug-carriers, and biomarkers. For their clinical development, not only their pharmaceutical activity is important but also their production needs to be evaluated. In this context, research focuses on the isolation of EVs, their characterization, and their storage. The present manuscript aims at providing a facile procedure for the assessment of the effect of different storage conditions on EVs, without genetic manipulation or specific functional assays. This makes it possible to quickly get a first impression of the stability of EVs under a given storage condition, and EVs derived from different cell sources can be compared easily. The stability measurement is based on the physicochemical parameters of the EVs (size, particle concentration, and morphology) and the preservation of the activity of their cargo. The latter is assessed by the saponin-mediated encapsulation of the enzyme beta-glucuronidase into the EVs. Glucuronidase acts as a surrogate and allows for an easy quantification via the cleavage of a fluorescent reporter molecule. The present protocol could be a tool for researchers in the search for storage conditions that optimally retain EV properties to advance EV research toward clinical application.

Introduction

EVs are membrane-bound nanoparticles produced by nearly all cell types. For mammalian cells, EVs can be subdivided into two main groups with distinct production pathways1,2. Membrane vesicles, with a size range from roughly 100-1,000 nm, are produced by direct budding from the cell membrane. Exosomes, sized 30-200 nm, are derived from multivesicular bodies formed by inward budding into endosomes that subsequently fuse with the cell membrane to release multiple exosomes at once. The main function of these vesicles is the transport of information between cells3. For this purpose, cargos such as RNA, DNA, and proteins are actively sorted into them. EVs can convey a variety of effects on their targets, with implications for both health and disease state. On one side, they mediate positive effects such as tissue regeneration, antigen presentation, or antibiotic effects, which makes them auspicious targets for their development as therapeutics4,5. On the other side, EVs can promote tumor vascularization6, induce bystander effects in stress responses7, and might play a role in autoimmune diseases8 and inflammatory diseases9. Thus, they might be a key component to a better understanding of many pathological effects. However, the presence of altered EVs in manifold diseases, such as cancer10,11,12 and cardiovascular disorders13, and their easy accessibility in blood and urine makes them ideal biomarkers. Finally, their good biocompatibility14 and their inherent targeting ability make EVs also interesting for drug delivery15. In this manuscript, we describe a protocol for the evaluation of the storage stability of EVs derived from mammalian cells, an important property that is still little investigated.

For the clinical development of EVs, there are still many obstacles to surmount16, including the evaluation of their therapeutic effects, production, purification, and storage17. While -80 °C is widely seen as the gold standard for EV storage18, the required freezers are expensive, and maintaining the required cold chain from the production to the patient can be challenging. Moreover, some reports indicate that storage at -80 °C still not optimally preserves EVs and induces a loss in EV functionality19,20. Other methods, such as freeze-drying21,22 or spray-drying23, have been proposed as potential alternatives to the frozen storage of EVs.

The optimal way of assessing storage stability would be to test the EVs in functional assays or by the evaluation of a specific marker, for instance, their antibacterial activity19. This is possible when the desired effect of the vesicles is known and when one distinct group of EVs is to be studied. If EVs from different cell sources are to be compared (e.g., for drug encapsulation) or if there is no known functional readout, it is no longer possible to assess changes due to storage in a direct manner.

On the other hand, simply evaluating changes in their physicochemical parameters, such as size, particle recovery, and protein concentration, does not always predict changes in EV activity, as has been shown in a recent patent20.

Here, we provide a readily applicable protocol for measuring the storage stability of EVs by assessing their physicochemical parameters combined with the activity of an encapsulated beta-glucuronidase enzyme as a surrogate for the cargo of the EVs. The loading of the enzyme is done by saponin incubation, a mild method established with EVs from different sources21,24,25. Saponin forms transient pores in the EV membrane, which allows enzyme uptake into the vesicle. As enzymes are prone to lose their activity if subjected to unfavorable storage conditions, they are an ideal surrogate for the evaluation of the preservation of functional cargoes of the EVs.

We have demonstrated that the application of this protocol to EVs derived from human mesenchymal stem cells (MSCs), human umbilical vein endothelial cells (HUVECs), and human adenocarcinoma alveolar epithelial cells (A549) indeed result in great differences in storage stability between different cell lines, which should be taken into consideration when choosing the EV source21.

Protocol

1. Cell culture and the production of cell-conditioned medium Generally, cultivate cells under the individual conditions required for the respective cell line. Cultivate the cells for 24-72 h in serum-free conditions or in medium containing EV-depleted fetal bovine serum (FBS). NOTE: If EV-depleted FBS is used, employ a method proven to efficiently deplete the serum, to prevent contamination with bovine serum-derived EVs26. Collect the medium from the flasks….

Representative Results

Figure 1 displays the storage characteristics of EVs isolated from HUVECs. EVs were isolated by UC, glucuronidase was encapsulated, and after SEC, the purified EVs were evaluated for their physicochemical properties by NTA. A sample of the vesicles was subsequently subjected to AF4 purification and the glucuronidase activity was measured. The vesicles were then stored for 7 d at 4 °C or -80 °C and at 4 °C in lyophilized form, in the latter case with the addition…

Discussion

In this manuscript, we present a comprehensive protocol to study the stability of EVs under different storage conditions. With the combination of encapsulated glucuronidase as a functional readout and the evaluation of the physicochemical parameters of the EVs, the protocol allows for a straightforward storage stability evaluation of EVs and the comparison of EVs from different cell lines. SEM and TEM as complementary methods allow an insight into changes of the EVs on the single-particle level. The results presented her…

Divulgaciones

The authors have nothing to disclose.

Acknowledgements

The NanoMatFutur Junior Research program from the Federal Ministry of Education and Research, Germany (grant number 13XP5029A) supported this work. Maximilian Richter was supported by Studienstiftung des Deutschen Volkes (German Academic Scholarship Foundation) through a Ph.D. fellowship.

Materials

1,2 dimyristoyl-sn glycero-3-phospho-choline (DMPC) Sigma-Aldrich P2663-25MG
1,2-dipalmitoyl-sn-glycero-3-phospho-choline (DPPC) Sigma-Aldrich P4329-25MG
225 cm² cell culture flasks Corning 431082 Used with 25 ml of medium
30 kDa regenerated cellulose membrane Wyatt Technology Europe 1854
350 µm spacer Wyatt Technology Europe
Automated fraction collector Thermo Fisher Scientific
Beta-glucuronidase Sigma-Aldrich G7646-100KU
Chloroform Fisher scientific C/4966/17
Column oven Hitachi High-Technologies Europe
D-(+)-Trehalose dihydrate Sigma-Aldrich T9531-10G
DAWN HELEOS II, Multi-angle light scattering detector  Wyatt Technology Europe
Durapore Membrane filter, PVDF,  0,1 µm, 47 mm Merck VVLP04700 Used for the preparation of buffers for AF4
EBM-2 Lonza Verviers, S.p.r. CC-3156 Endothelial Cell Growth basal medium, used for the serum free culture of HUVEC cells
Eclipse dualtec Wyatt Technology Europe
EGM-2 Lonza Verviers, S.p.r. CC-3162 Endothelial Cell Growth medium, used for the normal culture of HUVEC cells
ELISA Plate Sealers R&D Systems DY992 used for sealing of 96-well plates for the glucuronidase assay
Ethanol Fisher scientific E/0665DF/17
Extruder Set With Holder/Heating Block Avanti Polar Lipids 610000-1EA
Filter support Avanti Polar Lipids 610014-1EA used for liposome preparation
Fluorescein di-β-D-glucoronide Thermo Fisher Scientific F2915
Gibco PBS-tablets+CA10:F36 Thermo Fisher Scientific 18912014
Hettich Universal 320 R Andreas Hettich GmbH & Co.KG Used for pelleting cells at 300 g
Hettich Rotina 420 R Andreas Hettich GmbH & Co.KG Used for pelleting larger debris at 3000 g
HUVEC cells Lonza Verviers, S.p.r. C2517A
Kimble  FlexColumn 1X30CM Kimble 420401-1030
Lyophilizer ALPHA 2-4 LSC Christ
Microcentrifuge Tubes, Polypropylene VWR international 525-0255 the tubes used for all EV-handling, found to be more favorable than comparable products from other suppliers regarding particle recovery
Nanosight LM14 equipped with a green laser Malvern Pananalytical
Nanosight-software version 3.1 Malvern Pananalytical
Nucleopore 200 nm track-etch polycarbonate membranes Whatman/GE Healthcare 110406 used for liposome preparation
PEEK Inline filter holder Wyatt Technology Europe
Phosphotungstic acid hydrate Sigma-Aldrich 79690-25G
Polycarbonate bottles for ultracentrifugation Beckman Coulter 355622
QuantiPro BCA Assay Kit Sigma-Aldrich QPBCA-1KT
Saponin Sigma-Aldrich 47036
Scanning electron microscopy Zeiss EVO HD 15 Carl Zeiss AG
Sepharose Cl-2b GE Healthcare 17014001
SEM copper grids with carbon film Plano S160-4
Small AF4 channel Wyatt Technology Europe
Sputter-coater Q150R ES Quorum Technologies
Transmission electron microscopy JEOL JEM 2011 Oxford Instruments
Type 45 Ti ultracentrifugation rotor Beckman Coulter 339160
Ultimate 3000 Dionex autosampler Thermo Fisher Scientific
Ultimate 3000 Dionex isocratic pump Thermo Fisher Scientific
Ultimate 3000 Dionex online vacuum degasser Thermo Fisher Scientific
Ultracentrifuge OptimaTM L-90 K Beckman Coulter
UV detector Thermo Fisher Scientific
Whatman 0.2 µm pore size mixed cellulose filter Whatman/GE Healthcare 10401712 Used for the filtration of all buffers used with the EVs and in SEC

Referencias

  1. Stremersch, S., De Smedt, S. C., Raemdonck, K. Therapeutic and diagnostic applications of extracellular vesicles. Journal of Controlled Release. 244, 167-183 (2016).
  2. Fuhrmann, G., Herrmann, I. K., Stevens, M. M. Cell-derived vesicles for drug therapy and diagnostics: Opportunities and challenges. Nano Today. 10 (3), 397-409 (2015).
  3. Goes, A., Fuhrmann, G. Biogenic and Biomimetic Carriers as Versatile Transporters To Treat Infections. ACS Infectious Diseases. 4 (6), 881-892 (2018).
  4. György, B., Hung, M. E., Breakefield, X. O., Leonard, J. N. Therapeutic Applications of Extracellular Vesicles: Clinical Promise and Open Questions. Annual Review of Pharmacology and Toxicology. 55, 439-464 (2015).
  5. Schulz, E., et al. Biocompatible bacteria-derived vesicles show inherent antimicrobial activity. Journal of Controlled Release. 290, 46-55 (2018).
  6. Feng, Q., et al. A class of extracellular vesicles from breast cancer cells activates VEGF receptors and tumour angiogenesis. Nature Communications. 8, 14450 (2017).
  7. Bewicke-Copley, F., et al. Extracellular vesicles released following heat stress induce bystander effect in unstressed populations. Journal of Extracellular Vesicles. 6, 1340746 (2017).
  8. Xu, Y., et al. Macrophages transfer antigens to dendritic cells by releasing exosomes containing dead-cell-associated antigens partially through a ceramide-dependent pathway to enhance CD4(+) T-cell responses. Immunology. 149 (2), 157-171 (2016).
  9. Buzas, E. I., György, B., Nagy, G., Falus, A., Gay, S. Emerging role of extracellular vesicles in inflammatory diseases. Nature Reviews Rheumatology. 10, 356 (2014).
  10. Rajappa, P., et al. Malignant Astrocytic Tumor Progression Potentiated by JAK-mediated Recruitment of Myeloid Cells. Clinical Cancer Research: An Official Journal of the American Association for Cancer Research. 23 (12), 3109-3119 (2017).
  11. Umezu, T., et al. Exosomal miR-135b shed from hypoxic multiple myeloma cells enhances angiogenesis by targeting factor-inhibiting HIF-1. Blood. 124 (25), 3748-3757 (2014).
  12. Costa-Silva, B., et al. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nature Cell Biology. 17 (6), 816-826 (2015).
  13. Boulanger, C. M., Loyer, X., Rautou, P. -. E., Amabile, N. Extracellular vesicles in coronary artery disease. Nature Reviews Cardiology. 14, 259 (2017).
  14. Zhu, X., et al. Comprehensive toxicity and immunogenicity studies reveal minimal effects in mice following sustained dosing of extracellular vesicles derived from HEK293T cells. Journal of Extracellular Vesicles. 6 (1), 1324730 (2017).
  15. Vader, P., Mol, E. A., Pasterkamp, G., Schiffelers, R. M. Extracellular vesicles for drug delivery. Advanced Drug Delivery Reviews. 106, 148-156 (2016).
  16. Ingato, D., Lee, J. U., Sim, S. J., Kwon, Y. J. Good things come in small packages: Overcoming challenges to harness extracellular vesicles for therapeutic delivery. Journal of Controlled Release. 241, 174-185 (2016).
  17. Gimona, M., Pachler, K., Laner-Plamberger, S., Schallmoser, K., Rohde, E. Manufacturing of Human Extracellular Vesicle-Based Therapeutics for Clinical Use. International Journal of Molecular Sciences. 18 (6), 1190 (2017).
  18. Jeyaram, A., Jay, S. M. Preservation and Storage Stability of Extracellular Vesicles for Therapeutic Applications. The AAPS Journal. 20 (1), 1 (2017).
  19. Lőrincz, &. #. 1. 9. 3. ;. M., et al. Effect of storage on physical and functional properties of extracellular vesicles derived from neutrophilic granulocytes. Journal of Extracellular Vesicles. 3, 25465 (2014).
  20. Kreke, M., Smith, R., Hanscome, P., Peck, K., Ibrahim, A. Processes for producing stable exosome formulations. US patent. , (2016).
  21. Frank, J., et al. Extracellular vesicles protect glucuronidase model enzymes during freeze-drying. Scientific Reports. 8 (1), 12377 (2018).
  22. Charoenviriyakul, C., Takahashi, Y., Nishikawa, M., Takakura, Y. Preservation of exosomes at room temperature using lyophilization. International Journal of Pharmaceutics. 553 (1), 1-7 (2018).
  23. Kusuma, G. D., et al. To Protect and to Preserve: Novel Preservation Strategies for Extracellular Vesicles. Frontiers in Pharmacology. 9 (1199), (2018).
  24. Haney, M. J., et al. Exosomes as drug delivery vehicles for Parkinson’s disease therapy. Journal of Controlled Release. 207, 18-30 (2015).
  25. Fuhrmann, G., Serio, A., Mazo, M., Nair, R., Stevens, M. M. Active loading into extracellular vesicles significantly improves the cellular uptake and photodynamic effect of porphyrins. Journal of Controlled Release. 205, 35-44 (2015).
  26. Théry, C., et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. Journal of Extracellular Vesicles. 7 (1), 1535750 (2018).
  27. Gardiner, C., Ferreira, Y. J., Dragovic, R. A., Redman, C. W. G., Sargent, I. L. Extracellular vesicle sizing and enumeration by nanoparticle tracking analysis. Journal of Extracellular Vesicles. 2 (1), 19671 (2013).
  28. Vestad, B., et al. Size and concentration analyses of extracellular vesicles by nanoparticle tracking analysis: a variation study. Journal of Extracellular Vesicles. 6 (1), 1344087 (2017).
  29. Bosch, S., et al. Trehalose prevents aggregation of exosomes and cryodamage. Scientific Reports. 6, 36162 (2016).
  30. Bhattacharjee, S. DLS and zeta potential – What they are and what they are not. Journal of Controlled Release. 235, 337-351 (2016).
  31. Van Deun, J., et al. The impact of disparate isolation methods for extracellular vesicles on downstream RNA profiling. Journal of Extracellular Vesicles. 3 (1), 24858 (2014).
  32. Taylor, D. D., Shah, S. Methods of isolating extracellular vesicles impact down-stream analyses of their cargoes. Methods. 87, 3-10 (2015).
  33. Patel, D. B., et al. Impact of cell culture parameters on production and vascularization bioactivity of mesenchymal stem cell-derived extracellular vesicles. Bioengineering & Translational Medicine. 2 (2), 170-179 (2017).
  34. Gardiner, C., et al. Techniques used for the isolation and characterization of extracellular vesicles: results of a worldwide survey. Journal of Extracellular Vesicles. 5 (1), 32945 (2016).
  35. Zhang, H., et al. Identification of distinct nanoparticles and subsets of extracellular vesicles by asymmetric flow field-flow fractionation. Nature Cell Biology. 20 (3), 332-343 (2018).
  36. Linares, R., Tan, S., Gounou, C., Arraud, N., Brisson, A. R. High-speed centrifugation induces aggregation of extracellular vesicles. Journal of Extracellular Vesicles. 4 (1), 29509 (2015).
  37. Lobb, R. J., et al. Optimized exosome isolation protocol for cell culture supernatant and human plasma. Journal of Extracellular Vesicles. 4, 27031 (2015).
check_url/es/59584?article_type=t

Play Video

Citar este artículo
Richter, M., Fuhrmann, K., Fuhrmann, G. Evaluation of the Storage Stability of Extracellular Vesicles. J. Vis. Exp. (147), e59584, doi:10.3791/59584 (2019).

View Video