JoVE Logo
Faculty Resource Center

Sign In

Summary

Abstract

Introduction

Protocol

Representative Results

Discussion

Acknowledgements

Materials

References

Biochemistry

Chemical Affinity-Based Isolation of Extracellular Vesicles from Biofluids for Proteomics and Phosphoproteomics Analysis

Published: October 27th, 2023

DOI:

10.3791/65844

1Department of Biochemistry, Purdue University, 2Tymora Analytical Operations, 3Department of Chemistry, Purdue University, 4Purdue Institute for Cancer Research, Purdue University

The present protocol provides detailed descriptions for the efficient isolation of urinary extracellular vesicles utilizing functionalized magnetic beads. Moreover, it encompasses subsequent analyses, including western blotting, proteomics, and phosphoproteomics.

Extracellular vesicles (EVs) from biofluids have recently gained significant attention in the field of liquid biopsy. Released by almost every type of cell, they provide a real-time snapshot of host cells and contain a wealth of molecular information, including proteins, in particular those with post-translational modifications (PTMs) such as phosphorylation, as the main player of cellular functions and disease onset and progression. However, the isolation of EVs from biofluids remains challenging due to low yields and impurities from current EV isolation methods, making the downstream analysis of EV cargo, such as EV phosphoproteins, difficult. Here, we describe a rapid and effective EV isolation method based on functionalized magnetic beads for EV isolation from biofluids such as human urine and downstream proteomics and phosphoproteomics analysis following EV isolation. The protocol enabled a high recovery yield of urinary EVs and sensitive profiles of EV proteome and phosphoproteome. Furthermore, the versatility of this protocol and relevant technical considerations are also addressed here.

Extracellular vesicles (EVs) are membrane-encapsulated nanoparticles secreted by all types of cells and are present in biofluids such as blood, urine, saliva, etc.1,2,3,4. EVs carry a cargo of diverse bioactive molecules which reflect the physiological and pathological state of their host cells and, therefore function as crucial factors in disease progression4,5,6. Moreover, extensive studies have established that EV-based disease markers can be ide....

Log in or to access full content. Learn more about your institution’s access to JoVE content here

All urine samples were collected from healthy individuals after informed consent. The experiments were compliant with all ethical standards involving human samples and conform to the guidelines from Purdue University Human Research Protection Program.

1. Sample collection

  1. Centrifuge 12 mL of urine sample in a 15 mL conical centrifuge tube for 10 min at 2,500 x g, 4 °C to remove cell debris and large apoptotic bodies.
  2. Transfer 10 mL of the su.......

Log in or to access full content. Learn more about your institution’s access to JoVE content here

This protocol demonstrates a comprehensive workflow from the isolation of EVs to downstream proteomics and phosphoproteomics analyses (Figure 1). The triplicate urine samples were subjected to EV isolation. The isolated EVs were characterized by western blotting and subsequently processed for mass spectrometry-based proteomics sample preparation including protein extraction, enzymatic digestion, and peptide cleanup. For phosphoproteomics analysis, the phosphopeptides were further enriched ba.......

Log in or to access full content. Learn more about your institution’s access to JoVE content here

Effective EV isolation is an essential prerequisite to detecting low-abundant proteins and phosphoproteins in EVs. Despite the development of numerous methods to fulfill this need, the majority still suffer from limitations such as poor recovery or low reproducibility, which impede their utilization in large-scale studies and routine clinical settings. DUC is generally considered as the most common method for EV isolation, and the additional washing steps are normally applied to help increase the purity of target EVs

Log in or to access full content. Learn more about your institution’s access to JoVE content here

This work has been funded in part by NIH grants 3RF1AG064250 and R44CA239845.

....

Log in or to access full content. Learn more about your institution’s access to JoVE content here

NameCompanyCatalog NumberComments
1.5 mL microcentrifuge tubeLife Science ProductsM-1700C-LB
1.5 mL tube magnetic separator rackSergi Lab Supplies1005
15 mL conical centrifuge tubeCorning 352097
15 mL tube magnetic separator rackSergi Lab Supplies1002
Anti-rabbit IgG, HRP-linked AntibodyCell Signaling Technology7074P2
Benchtop incubated shakerBioerDIS-87999-3367802Bioer Thermocell Mixing Block MB-101
CD9 (D3H4P) Rabbit mAbCell Signaling Technology13403S
ChloroacetamideSigma -AldrichC0267-100GUsed for alkylation of reduced sulfide groups. Freshly prepare 400 mM in water as stock solution.
Ethyl acetate Fisher Scientific E145-4Precipitates detergents
Evosep One EvosepLiquid chromatography system
EvotipsEvosepEV2013Sample loading for Evosep One system 
EVtrapTymora AnalyticalFunctionalized magnetic beads, loading buffer, and washing buffer 
Immobilon-FL PVDF MembraneSigma -AldrichIPFL00010Blotting membrane 
NuPAGE 4-12% Bis-Tris GelInvitrogenNP0322BOXInvitrogen NuPAGE 4 to 12%, Bis-Tris, 1.0 mm, Mini Protein Gel, 12-well
NuPAGE LDS Sample Buffer (4X)InvitrogenNP0007
PBSThermoFisher10010023
Pepsep C18 15 x 75 x 1.9Bruker 1893473Separation column 
Phosphatase Inhibitor Cocktail 2Sigma -AldrichP5726-5ML100X, Phosphotase inhibitor.
Phosphatase Inhibitor Cocktail 3Sigma -AldrichP0044-1ML100X,  Phosphotase inhibitor. 
Pierce BCA Protein Assay KitThermoFisher23225
Pierce ECL Western Blotting SubstrateThermoFisher32106HRP substrate 
PolyMAC phosphopeptide enrichment kitTymora AnalyticalPolymer-based metal ion affinity capture (PolyMAC) for phosphopeptide enrichment
Sodium deoxycholate Sigma -AldrichD6750-10GDetergent for lysis buffer. Prepare 120 mM in water as stock solution.
Sodium lauroyl sarcosinate Sigma -AldrichL9150-50GDetergent for lysis buffer. Prepare 120 mM in water as stock solution.
timsTOF HTBrukerTrapped ion-mobility time-of-flight mass spectrometry
TopTip C-18 (10-200 μL) tips GlygenTT2C18.96Desalting method
TriethylamineSigma -Aldrich471283-100MLFor EV elution. 
Triethylammonium bicabonate bufferSigma -AldrichT7408-100ML1 M
Trifluoroacetic acidSigma -Aldrich302031-100ML
Tris-(2-carboxyethyl)phosphine hydrochlorideSigma -AldrichC4706Used for reducion of disulfide bonds. Prepare 200 mM in water as stock solution. Aliquot the stock solution into small volume and store it in at-20°C (avoid multiple freeze-thaw cycles).
Trypsin/Lys-C MIXThermoFisherPIA41007

  1. Abels, E. R., Breakefield, X. O. Introduction to extracellular vesicles: Biogenesis, RNA cargo selection, content, release, and uptake. Cell Mol Neurobiol. 36 (3), 301-312 (2016).
  2. Maacha, S., et al. Extracellular vesicles-mediated intercellular communication: roles in the tumor microenvironment and anti-cancer drug resistance. Mol Cancer. 18 (1), 55 (2019).
  3. van Niel, G., D’Angelo, G., Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol. 19 (4), 213-228 (2018).
  4. Becker, A., et al. extracellular vesicles in cancer: cell-to-cell mediators of metastasis. Cancer Cell. 30 (6), 836-848 (2016).
  5. 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).
  6. Urabe, F., et al. Extracellular vesicles as biomarkers and therapeutic targets for cancer. Am J Physiol Cell Physiol. 318 (1), C29-C39 (2020).
  7. Chang, W. H., Cerione, R. A., Antonyak, M. A. Extracellular Vesicles and Their Roles in Cancer Progression. Methods Mol Biol. 2174, 143-170 (2021).
  8. Chen, I. H., et al. Phosphoproteins in extracellular vesicles as candidate markers for breast cancer. Proc Natl Acad Sci U S A. 114 (12), 3175-3180 (2017).
  9. Harsha, H. C., Pandey, A. Phosphoproteomics in cancer. Mol Oncol. 4 (6), 482-495 (2010).
  10. Singh, V., et al. Phosphorylation: Implications in Cancer. Protein J. 36 (1), 1-6 (2017).
  11. Delom, F., Chevet, E. Phosphoprotein analysis: from proteins to proteomes. Proteome Sci. 4, 15 (2006).
  12. Thingholm, T. E., Jensen, O. N., Larsen, M. R. Analytical strategies for phosphoproteomics. Proteomics. 9 (6), 1451-1468 (2009).
  13. Taylor, D. D., Shah, S. Methods of isolating extracellular vesicles impact down-stream analyses of their cargoes. Methods. 87, 3-10 (2015).
  14. Witwer, K. W., et al. Standardization of sample collection, isolation and analysis methods in extracellular vesicle research. J Extracell Vesicles. 2, (2013).
  15. Zeringer, E., et al. Methods for the extraction and RNA profiling of exosomes. World J Methodol. 3 (1), 11-18 (2013).
  16. Mathivanan, S., et al. Proteomics analysis of A33 immunoaffinity-purified exosomes released from the human colon tumor cell line LIM1215 reveals a tissue-specific protein signature. Mol Cell Proteomics. 9 (2), 197-208 (2010).
  17. Enderle, D., et al. Characterization of RNA from exosomes and other extracellular vesicles isolated by a novel spin column-based method. PLoS ONE. 10 (8), e0136133 (2015).
  18. Wu, X., Li, L., Iliuk, A., Tao, W. A. Highly Efficient Phosphoproteome Capture and Analysis from Urinary Extracellular Vesicles. J Proteome Res. 17 (9), 3308-3316 (2018).
  19. Iliuk, A., et al. Plasma-derived extracellular vesicle phosphoproteomics through chemical affinity purification. J Proteome Res. 19 (7), 2563-2574 (2020).
  20. Iliuk, A. B., Martin, V. A., Alicie, B. M., Geahlen, R. L., Tao, W. A. In-depth analyses of kinase-dependent tyrosine phosphoproteomes based on metal ion-functionalized soluble nanopolymers. Mol Cell Proteomics. 9 (10), 2162-2172 (2010).
  21. Hadisurya, M., et al. Quantitative proteomics and phosphoproteomics of urinary extracellular vesicles define diagnostic and prognostic biosignatures for Parkinson’s Disease. Commun Med. 3 (1), 64 (2023).
  22. Hadisurya, M., et al. Data-independent acquisition phosphoproteomics of urinary extracellular vesicles enables renal cell carcinoma grade differentiation. Mol Cell Proteomics. 22 (5), 100536 (2023).
  23. Wu, X., Liu, Y. K., Iliuk, A. B., Tao, W. A. Mass spectrometry-based phosphoproteomics in clinical applications. Trends Analyt Chem. 163, 117066 (2023).
  24. Mathieu, M., et al. Specificities of exosome versus small ectosome secretion revealed by live intracellular tracking of CD63 and CD9. Nat Commun. 12 (1), 4389 (2021).
  25. Mahmood, T., Yang, P. C. Western blot: technique, theory, and trouble shooting. N Am J Med Sci. 4 (9), 429-434 (2012).
  26. Keerthikumar, S., et al. ExoCarta: A web-based compendium of exosomal cargo. J Mol Biol. 428 (4), 688-692 (2016).
  27. Théry, C., Amigorena, S., Raposo, G., Clayton, A. Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Curr Protoc Cell Biol. 3 (22), (2006).
  28. Livshits, M. A., et al. Isolation of exosomes by differential centrifugation: Theoretical analysis of a commonly used protocol. Sci Rep. 5, 17319 (2015).
  29. Konoshenko, M. Y., Lekchnov, E. A., Vlassov, A. V., Laktionov, P. P. Isolation of extracellular vesicles: general methodologies and latest trends. BioMed Res Int. 2018, (2018).
  30. Webber, J., Clayton, A. How pure are your vesicles. J Extracell Vesicles. 2, (2013).
  31. Erdjument-Bromage, H., Huang, F. K., Neubert, T. A. Sample preparation for relative quantitation of proteins using tandem mass tags (TMT) and mass spectrometry (MS). Methods Mol Biol. 1741, 135-149 (2018).
  32. Charles Jacob, H. K., et al. Identification of novel early pancreatic cancer biomarkers KIF5B and SFRP2 from “first contact” interactions in the tumor microenvironment. J Exp Clinl Cancer Res. 41 (1), 258 (2022).
  33. Nunez Lopez, Y. O., et al. Extracellular vesicle proteomics and phosphoproteomics identify pathways for increased risk in patients hospitalized with COVID-19 and type 2 diabetes mellitus. Diabetes Res Clin Pract. 197, 110565 (2023).
  34. Hinzman, C. P., et al. A multi-omics approach identifies pancreatic cancer cell extracellular vesicles as mediators of the unfolded protein response in normal pancreatic epithelial cells. J Extracell Vesicles. 11 (6), e12232 (2022).
  35. Kornilov, R., et al. Efficient ultrafiltration-based protocol to deplete extracellular vesicles from fetal bovine serum. J Extracell Vesicles. 7 (1), 1422674 (2018).
  36. Willms, E., et al. Cells release subpopulations of exosomes with distinct molecular and biological properties. Sci Rep. 6, 22519 (2016).
  37. Searle, B. C., et al. Generating high quality libraries for DIA MS with empirically corrected peptide predictions. Nat Commun. 11 (1), 1548 (2020).
  38. Skowronek, P., et al. Rapid and in-depth coverage of the (Phospho-)proteome with deep libraries and optimal window design for dia-PASEF. Mol Cell Proteomics. 21 (9), 100279 (2022).

This article has been published

Video Coming Soon

JoVE Logo

Privacy

Terms of Use

Policies

Research

Education

ABOUT JoVE

Copyright © 2024 MyJoVE Corporation. All rights reserved