Summary

Size Exclusion Chromatography for Separating Extracellular Vesicles from Conditioned Cell Culture Media

Published: May 13, 2022
doi:

Summary

The protocol here demonstrates that extracellular vesicles can be adequately separated from conditioned cell culture media using size exclusion chromatography.

Abstract

Extracellular vesicles (EVs) are nano-sized lipid-membrane bound structures that are released from all cells, are present in all biofluids, and contain proteins, nucleic acids, and lipids that are reflective of the parent cell from which they are derived. Proper separation of EVs from other components in a sample allows for characterization of their associated cargo and lends insight into their potential as intercellular communicators and non-invasive biomarkers for numerous diseases. In the current study, oligodendrocyte derived EVs were isolated from cell culture media using a combination of state-of-the-art techniques, including ultrafiltration and size exclusion chromatography (SEC) to separate EVs from other extracellular proteins and protein complexes. Using commercially available SEC columns, EVs were separated from extracellular proteins released from human oligodendroglioma cells under both control and endoplasmic reticulum (ER) stress conditions. The canonical EV markers CD9, CD63, and CD81 were observed in fractions 1-4, but not in fractions 5-8. GM130, a protein of the Golgi apparatus, and calnexin, an integral protein of the ER, were used as negative EV markers, and were not observed in any fraction. Further, when pooling and concentrating fractions 1-4 as the EV fraction, and fractions 5-8 as the protein fraction, expression of CD63, CD81, and CD9 in the EV fraction was observed. The expression of GM130 or calnexin was not observed in either of the fraction types. The pooled fractions from both control and ER stress conditions were visualized with transmission electron microscopy and vesicles were observed in the EV fractions, but not in the protein fractions. Particles in the EV and protein fractions from both conditions were also quantified with nanoparticle tracking analysis. Together, these data demonstrate that SEC is an effective method for separating EVs from conditioned cell culture media.

Introduction

The explosion of interest in studying extracellular vesicles (EVs) has been accompanied by major advancements in the technologies and techniques used to separate and study these nano-sized, heterogeneous particles. In the time since their discovery nearly four decades ago1,2, these small membranous structures have been found to contain bioactive lipids, nucleic acids, and proteins, and play major roles in intercellular communication3,4. EVs are released from all cell types and are therefore present in all biological fluids, including blood plasma and serum, saliva, and urine. EVs within these fluids hold great promise to serve as non-invasive biomarkers for various diseases, including neuroinflammatory and neurodegenerative diseases, cancers, and autoimmune disorders5,6,7. Further, in vitro mechanistic studies can be performed through cell culture techniques by separating EVs released into the culture medium3,8,9.

To understand the role of EVs in disease pathophysiology, adequate separation from the fluid in which they are found is paramount. The gold standard for EV separation has long been differential ultracentrifugation (dUC)10, however more sophisticated techniques have arisen to achieve better separation of EVs from other extracellular components. Some of these techniques include density gradients, asymmetrical-flow field-flow fraction (A4F), flow cytometry, immunocapture, polyethylene glycol precipitation, and size exclusion chromatography (SEC)11,12,13. Each technique has its own set of advantages and disadvantages; however, SEC in particular has been shown to separate EVs from both biological fluids and cell culture supernatants quite effectively8,14,15. SEC also has the added bonus of being relatively straightforward and user-friendly.

SEC is a method that separates components of a fluid based on size. With this technique, a column of resin (either made in-house or purchased commercially) is used to fractionate a sample. Small particles in the sample get trapped between the beads within the resin, while larger particles are able to pass through the resin more freely, and thus elute earlier in the process. Because EVs are larger in size than many extracellular proteins and protein aggregates, EVs pass through the column faster and elute in earlier fractions than extracellular proteins14.

In this methods paper, the use of SEC for separation of EVs from cell culture media (CCM) from human oligodendrocytes under both control and endoplasmic reticulum (ER) stress conditions is outlined. Using this protocol, it is shown that EVs separated with this technique are found within specific fractions that can be pooled together and concentrated for downstream characterization, and that the separated EVs are derived from cells and not from an exogenous source such as fetal bovine serum (FBS) used to supplement the CCM. The presence of the canonical EV markers, CD63, CD81, and CD916,17,18,19 in the EV fractions, and their absence in the protein fractions is demonstrated with western blotting. Using transmission electron microscopy (TEM), EVs are visualized and display the expected morphology and are only observed in the EV fraction. Particles are also counted in the EV and protein fractions of both control and ER stress conditions, and a large number of particles within the expected size range of 50-200 nm in diameter are observed in the EV samples. Together, these data support the notion that SEC is an efficient and effective method for separating EVs from cell culture media.

Protocol

1. Preparation of buffers and reagents NOTE: Make cell culture reagents in cell culture hood to maintain sterility. Preparation of cell culture reagents Prepare normal high glucose DMEM by adding 50 mL of FBS and 5 mL of penicillin-streptococcus (Pen-Strep) into 500 mL of high glucose DMEM and store at 4 °C. Use this media for culturing and expanding cells. Prepare exosome-depleted high glucose DMEM by adding 50 mL of exosome-deplet…

Representative Results

Western blotting reveals adequate separation of EVs from CCM To evaluate the effectiveness of SEC for separating EVs from cell culture media, a western blot was run using each individual fraction from the control samples to probe expression of the three canonical EV markers, CD9, CD63 and CD81, as well as GM130 and calnexin18, which were used as negative controls (Figure 3). Albumin expression18 was also probed to ensure …

Discussion

SEC is a user-friendly method for adequately separating EVs from conditioned CCM. In order to specifically isolate cell derived EVs, careful consideration of the type of CCM and its supplements must be taken into account. Many cell culture medias need to be supplemented with FBS, which contains EVs derived from the animal in which the serum was harvested. These serum EVs may saturate and mask any signal produced by EVs derived from cells in culture26. Therefore, when performing experiments, EV-dep…

Disclosures

The authors have nothing to disclose.

Acknowledgements

The authors would like to thank Penn State Behrend and the Hamot Health Foundation for funding, as well as the Penn State Microscopy Facility in University Park, PA.

Materials

2-Mercaptoethanol VWR 97064-588
4X Laemmli Sample Buffer BioRad 1610747
Amicon Ultra-15 Centrifugal Filter Unit, Ultracel, 3 KDa, 15mL Sigma-Aldrich UFC900308 3 kDa cutoff
Amicon Ultra-2 Centrifugal Filter Unit with Ultracel-3 membrane Sigma-Aldrich UFC200324 3 kDa cutoff
Ammonium Persulfate Sigma-Aldrich A3678-100G
Anti rabbit IgG, HRP linked Antibody Cell Signaling Technology 7074V 1:1000 Dilution
Anti-Calnexin antibody Abcam  ab22595 1:500 Dilution
Anti-CD9 Mouse Monoclonal Antibody BioLegend 312102 1:500 Dilution
Anti-GM130 antibody [EP892Y] – cis-Golgi Marker Abcam ab52649 1:500 Dilution
Anti-mouse IgG, HRP-linked Antibody Cell Signaling Technology 7076V 1:1000 Dilution
Automatic Fraction Collector Izon Science
BCA assay Kit Bio-Rad
CCD camera Gatan Orius SC200
Cd63 Mouse anti Human BD 556019 1:1000 Dilution
CD81 Antibody Santa Cruz Biotechnology sc-23962 1:1000 Dilution
Cellstar Filter Cap Cell Culture Flasks Greiner Bio-One 660175
ChemiDoc MP Imager BioRad
Clarity Western ECL Substrate BioRad 1705061
deoxycholate Sigma-Aldrich D6750-10G
dithiothreitol Sigma 3483-12-3
DMEM/High glucose with L-glutamine; without sodium Cytiva SH300022.FS
Fetal Bovine Serum Premium grade VWR 97068-085
Fetal Bovine Serum, exosome-depleted Thermo Scientific A2720801
Glycine BioRad 1610718
Great Value Nonfat Dry Milk Amazon B076NRD2TZ
HOG Human Oligodendroglioma Cell Line Sigma-Aldrich SCC163
Izon Science Usa Ltd qev Size Exclusion Columns 5pk Izon Science
Methanol >99.8% ACS VWR BDH1135-4LP
Mini-PROTEAN Glass plates BioRad 1653310 with 0.75mm spacers
Mini-PROTEAN Short plates BioRad 1653308
NP-40 Sigma-Aldrich 492016
Penicillin-Streptomycin,Solution Sigma-Aldrich P4458-100mL
Phosphate Buffered Saline PBS Fisher Scientific BP66150
Pierce BCA Protein Assay Kits and Reagents Thermo Fisher Scientific 23227
Pierce PVDF Transfer Membranes Thermo Scientific 88518
Pierce Western Blotting Filter Paper Thermo Scientific 84783
Polyoxyethylene-20 (TWEEN 20), 500mL Bio Basic TB0560
Protease/phosphatase Inhibitor Cocktail (100X) Cell Signaling Technology 5872S
Recombinant Anti-TSG101 antibody [EPR7130(B)] ABCam ab125011 1:1000 dilution
Slodium hydroxide Sigma-Aldrich SX0603
Sodium azide Fisher Scientific BP922I-500
Sodium Chloride Sigma-Aldrich S9888-500G
Sodium dodecyl sulfate,≥99.0% (GC), dust-free pellets Sigma-Aldrich 75746-1KG
Tetramethylethylenediamine Sigma-Aldrich T9281-25ML
TGX Stain-Free FastCast Acrylamide Kit, 10% BioRad 1610183
Transmission Electron Microscope FEI Tecnai 12 Biotwin
Tris BioRad 1610716
Trypsin 0.25% protease with porcine trypsin, HBSS, EDTA; without calcium, magnesium Cytiva SH30042.01
Tunicamycin Tocris 3516
Zeta View software Analytik NTA software

References

  1. Pan, B. T., Teng, K., Wu, C., Adam, M., Johnstone, R. M. Electron microscopic evidence for externalization of the transferrin receptor in vesicular form in sheep reticulocytes. The Journal of Cell Biology. 101 (3), 942-948 (1985).
  2. Harding, C., Heuser, J., Stahl, P. Receptor-mediated endocytosis of transferrin and recycling of the transferrin receptor in rat reticulocytes. The Journal of Cell Biology. 97 (2), 329-339 (1983).
  3. Valadi, H., et al. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nature Cell Biology. 9 (6), 654-659 (2007).
  4. Raposo, G., Stoorvogel, W. Extracellular vesicles: Exosomes, microvesicles, and friends. The Journal of Cell Biology. 200 (4), 373-383 (2013).
  5. Rontogianni, S., et al. Proteomic profiling of extracellular vesicles allows for human breast cancer subtyping. Communications Biology. 2 (1), 1-13 (2019).
  6. Lane, R. E., Korbie, D., Hill, M. M., Trau, M. Extracellular vesicles as circulating cancer biomarkers: opportunities and challenges. Clinical and Translational Medicine. 7 (1), 14 (2018).
  7. Thompson, A. G., et al. Extracellular vesicles in neurodegenerative disease-pathogenesis to biomarkers. Nature Reviews Neurology. 12 (6), 346-357 (2016).
  8. O’Brien, K., Ughetto, S., Mahjoum, S., Nair, A. V., Breakefield, X. O. Uptake, functionality, and re-release of extracellular vesicle-encapsulated cargo. Cell Reports. 39 (2), 110651 (2022).
  9. Joshi, B. S., de Beer, M. A., Giepmans, B. N. G., Zuhorn, I. S. Endocytosis of extracellular vesicles and release of their cargo from endosomes. ACS Nano. 14 (4), 4444-4455 (2020).
  10. Théry, C., Amigorena, S., Raposo, G., Clayton, A. Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Current Protocols in Cell Biology. 30 (1), 3-22 (2006).
  11. Willms, E., Cabañas, C., Mäger, I., Wood, M. J. A., Vader, P. Extracellular vesicle heterogeneity: Subpopulations, isolation techniques, and diverse functions in cancer progression. Frontiers in Immunology. 9, 738 (2018).
  12. Tzaridis, T., et al. Extracellular vesicle separation techniques impact results from human blood samples: Considerations for diagnostic applications. International Journal of Molecular Sciences. 22 (17), 9211 (2021).
  13. Liangsupree, T., Multia, E., Riekkola, M. L. Modern isolation and separation techniques for extracellular vesicles. Journal of Chromatography A. 1636, 461773 (2021).
  14. Gámez-Valero, A., et al. Size-exclusion chromatography-based isolation minimally alters extracellular vesicles’ characteristics compared to precipitating agents. Scientific Reports. 6 (1), 1-9 (2016).
  15. Mol, E. A., Goumans, M. J., Doevendans, P. A., Sluijter, J. P. G., Vader, P. Higher functionality of extracellular vesicles isolated using size-exclusion chromatography compared to ultracentrifugation. Nanomedicine: Nanotechnology, Biology, and Medicine. 13 (6), 2061-2065 (2017).
  16. Campos-Silva, C., et al. High sensitivity detection of extracellular vesicles immune-captured from urine by conventional flow cytometry. Scientific Reports. 9 (1), 2042 (2019).
  17. Norman, M., et al. L1CAM is not associated with extracellular vesicles in human cerebrospinal fluid or plasma. Nature methods. 18 (6), 631-634 (2021).
  18. 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).
  19. Mathieu, M., et al. Specificities of exosome versus small ectosome secretion revealed by live intracellular tracking of CD63 and CD9. Nature Communications. 12 (1), 1-18 (2021).
  20. Guo, J., et al. Establishment of a simplified dichotomic size-exclusion chromatography for isolating extracellular vesicles toward clinical applications. Journal of Extracellular Vesicles. 10 (11), 12145 (2021).
  21. Benedikter, B. J., et al. Ultrafiltration combined with size exclusion chromatography efficiently isolates extracellular vesicles from cell culture media for compositional and functional studies. Scientific Reports. 7 (1), 1-13 (2017).
  22. Eslami, A., Lujan, J. Western blotting: sample preparation to detection. Journal of Visualized Experiments: JoVE. (44), e2359 (2010).
  23. Grassucci, R. A., Taylor, D. J., Frank, J. Preparation of macromolecular complexes for cryo-electron microscopy. Nature Protocols. 2 (12), 3239 (2007).
  24. Williams, R. L., Urbé, S. The emerging shape of the ESCRT machinery. Nature Reviews Molecular Cell Biology. 8 (5), 355-368 (2007).
  25. Willms, E., et al. Cells release subpopulations of exosomes with distinct molecular and biological properties. Scientific Reports. 6 (1), 1-12 (2016).
  26. Lehrich, B. M., Liang, Y., Fiandaca, M. S. Foetal bovine serum influence on in vitro extracellular vesicle analyses. Journal of Extracellular Vesicles. 10 (3), 12061 (2021).
  27. Kornilov, R., et al. Efficient ultrafiltration-based protocol to deplete extracellular vesicles from fetal bovine serum. Journal of Extracellular Vesicles. 7 (1), 1422674 (2018).
  28. Böing, A. N., et al. Single-step isolation of extracellular vesicles by size-exclusion chromatography. Journal of Extracellular Vesicles. 3 (1), (2014).
  29. Zhang, X., Borg, E. G. F., Liaci, A. M., Vos, H. R., Stoorvogel, W. A novel three step protocol to isolate extracellular vesicles from plasma or cell culture medium with both high yield and purity. Journal of Extracellular Vesicles. 9 (1), 1791450 (2020).
  30. Guerreiro, E. M., et al. Efficient extracellular vesicle isolation by combining cell media modifications, ultrafiltration, and size-exclusion chromatography. PLoS ONE. 13 (9), 02024276 (2018).
  31. Onódi, Z., et al. Isolation of high-purity extracellular vesicles by the combination of iodixanol density gradient ultracentrifugation and bind-elute chromatography from blood plasma. Frontiers in Physiology. 9, 1479 (2018).
  32. Yuana, Y., Levels, J., Grootemaat, A., Sturk, A., Nieuwland, R. Co-isolation of extracellular vesicles and high-density lipoproteins using density gradient ultracentrifugation. Journal of Extracellular Vesicles. 3 (1), (2014).
check_url/kr/63614?article_type=t

Play Video

Cite This Article
Jones, M. T., Manioci, S. W., Russell, A. E. Size Exclusion Chromatography for Separating Extracellular Vesicles from Conditioned Cell Culture Media. J. Vis. Exp. (183), e63614, doi:10.3791/63614 (2022).

View Video