Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Jun 4.
Published in final edited form as: Expert Rev Proteomics. 2014 Jan 29;11(3):251–253. doi: 10.1586/14789450.2014.874290

Biology and proteomics of extracellular vesicles: harnessing their clinical potential

Crislyn D’Souza-Schorey 1, Dolores Di Vizio 2
PMCID: PMC4455922  NIHMSID: NIHMS604945  PMID: 24476411

Abstract

Extracellular membrane vesicles have recently emerged as versatile mediators of intercellular communication, pathogenesis, drug and gene delivery and as potentially rich reservoirs of clinical biomarkers. Channeling their properties toward patient care is dependent on technological progress in approaches used for their analysis and molecular profiling.

Keywords: disease biomarkers, exosomes, microvesicles


Eukaryotic cells release membrane vesicles under physiological conditions, although aberrant shedding of vesicles into the extracellular environment can arise in disease states, as has been well documented in cancer [1,2]. These shed membrane vesicles vary with respect to morphological and functional characteristics. They may originate either at the plasma membrane (ectosomes) or are generated in intracellular multivesicular bodies and released upon fusion of the limiting membrane of multivesicular body with the cell surface (exosomes) [3]. Shed vesicles that originate at the cell surface are larger than exosomes and can range from 200 nm to a few microns in diameter. Vesicles that fit these latter criteria and derived from tumor cells have been referred to as tumor-derived micro-vesicles and oncosomes [46]. All of the aforementioned membrane-enclosed sacs, collectively referred to as extracellular vesicles (EVs), can impact a variety of cellular processes, including inflammatory responses and cell invasion [5,7,8]. They contain complex sets of cargo depending on the physiological conditions in which they are generated and released. EVs may indeed act as mediators of paracrine signaling because of their ability to transfer bioactive molecules, including lipids, proteins and functional nucleic acids to recipient cells in the extracellular microenvironment [1,2,5,7]. This transfer of functional molecules may result from direct membrane fusion or the internalization of the EVs into the target cell [2]. Moreover, their release into body fluids such as blood, saliva and urine, points to their potential as circulating biomarkers in the surveillance of disease progression [9]. EVs have been detected in the circulation of patients with several types of cancers such as glioblastoma, breast, lung, ovary, prostate, colorectal and gastric cancers [9]. Mass spectrometry (MS)-based proteomic profiling of EVs in the past several years have allowed their characterization from different types of cell lines, body fluids and various animal and human organ models of disease, generating a plethora of data stored in openly accessible databases such as ExoCarta [10] and Vesiclepedia [11].

Protein profiling of EVs released from human colon and prostate cancer cell lines shows that these vesicles contain signaling molecules that could potentially affect processes such as angiogenesis, cell metabolism, mRNA processing, cell invasion and growth, in addition to molecules that are likely required for vesicle biogenesis [12]. In early MS-based investigations, EV proteins were first separated by gel electrophoresis allowing identification of proteins enriched in EVs [1315]. In more recent work, EVs purified by ultracentrifugation and more sophisticated fractionation methods, in addition to affinity binding have been used in high-throughput proteomic studies increasing the number of EV-associated proteins [16,17]. These and other studies [2] have suggested that inclusion of protein cargo into shed vesicles is not based on protein abundance or random blebbing but rather on selective sorting of cargo into shedding EV populations. Given the complexity of cargo, both proteins and nucleic acids, it is likely that more than one intracellular transport route engages in cargo delivery to sites of EV biogenesis. Co-sorting of cytoplasmic proteins via protein–protein interactions have also been proposed to play a role [17]. A recent study that analyzed 78 qualified high-throughput data sets on EV protein profiles, found that 797 common EV proteins were those originating from the cytoskeleton, cytosol, plasma membrane and intracellular vesicles [16]. Annotation of these proteins to biological processes revealed association with cell motility, metabolism, apoptosis regulation, small GTPase-linked signaling and vesicular transport. Furthermore, protein interaction network analyses reveal that these 797 proteins were extensively connected via physical interactions, whereas functional connections displayed modules for cytoskeleton remodeling and vesicular transport.

As stated earlier, given the heterogeneity of EV populations, it is likely that multiple shed vesicle populations were examined in the majority of investigations described to date. Particularly advantageous to the enrichment of EV antigens for use as diagnostic or prognostic indicators would be the ability to effectively concentrate and capture individual populations of EVs from body fluids. This approach, which is distinct from the analysis of whole tissue or unfractionated body fluid or even circulating tumor cells, would be especially significant if individual vesicle populations indeed concentrate molecular changes that occur in the parental tissue, as it would increase the sensitivity of detecting critical markers of disease progression. In this regard, molecular markers that would aid in the identification and/or capture of individual vesicle populations would be particularly beneficial. In a recent study utilizing immunobead-based capture, exosomes from colon cancer cells with protein marker A33 could be separated from those that contained epithelial cell adhesion molecule [18]. Proteomic profiling revealed significant differences in the two exosome subpopulations.

A largely unexplored hypothesis is that the proteins in individual EV populations might bear characteristic post-translational modifications. Unique glycosylation patterns of growth factors and signaling proteins, as they traffic through the endoplasmic reticulum and Golgi, might be one such modification. Similarly, investigations of the phospho-proteomic profile of individual shed vesicle populations could reveal unique modifications of cargo and will also shed light on current understanding of EV biogenesis. Lipid-based post-translational modifications in EVs are also yet unexplored. Palmitoylation of several signaling proteins, including Src family kinases and Ras, modulate protein localization in specific plasma membrane microdomains [19]. Such lipid anchors could promote stabilization of tetraspanin microdomains or even facilitate protein sorting to EVs, although this has yet to be explored. Importantly, post-translational modification could also potentially provide insight into physiological cellular changes.

EVs could also offer markers that are tissue specific. Panels of EV markers in combination with tissue-specific markers would potentially constitute unique and identifiable biosignatures for individual cancers or other types of disease. Furthermore, as the genome and proteome of tumors undergo changes with disease progression or in response to therapy, EVs might also be useful in disease staging as well as assessing the efficacy of response to therapy. This tenet is especially pertinent for tissues such as the ovary, prostate or pancreas, where repeat biopsies of these organs are difficult or unrealistic. Besides blood, sampling of other body fluids (e.g., saliva, urine, ascites or pleural effusions) could enable efficient disease detection. Complicating the utilization of EVs in cancer detection strategies is the presence of shed vesicles from other non-tumor cell types that likely might also be present in body fluids. Thus, it is equally important to identify markers that would allow selective capture of individual subpopulations of tumor-derived EVs, away from other shed vesicle populations in fluids. The methods currently used to discriminate between various shed vesicle populations found in peripheral fluids have been discussed [2,20], but a refinement of these procedures and additional approaches is needed.

Molecular profiling of EVs could form the basis of personalized therapeutics, especially as more reliable and rapid profiling technologies become available. For example, patients presenting increased expression of the breast cancer-associated oncogenic receptor, HER-2/neu, in subsets of gastric and ovarian cancers would benefit from existing breast cancer treatment strategies that target the HER-2 receptor [21]. Indeed, HER-2 has been detected in serum microvesicles derived from gastric tumors [22]. This type of information can be vital, as therapeutic treatments are coupled to ‘oncogenic’ mutations and cellular changes. The detection of such targets in EVs could pave the way for potential diagnostic and therapeutic strategies that preclude the need for major surgical intervention.

Recent work has explored the potential of EVs as novel therapeutic vehicles in cancer immunotherapy, as well as delivery of RNA-interference reagents and drugs [23,24]. EVs are poorly immunogenic and therefore protect its cargoes from rapid degradation in vivo, a distinct advantage over the packaging of similar cargo in liposomes [25]. Further investigation into the circulating levels and clearance dynamics in vivo is warranted to better define therapeutic effectiveness of EVs as delivery vehicles in clinical applications.

In conclusion, proteomic profiling studies represent a valuable, emerging strategy to gain better insight into the biology and clinical potential of circulating EVs in patients. Shotgun proteomic approaches, especially LC/LC-MS/MS-based techniques such as multidimensional protein identification technology that allow rapid and global protein identification and quantification [26], as well as multiplexed multiple reaction monitoring wherein peptides can be quantitated in a single analysis at high flow rates [27], coupled with sensitive immunodetection methods, would be especially helpful in this regard. Further, high-resolution MS, as has been recently reported to detect secreted proteins with picogram sensitivity [28], would be especially well suited for proteomic profiling of EVs in body fluids. However, resolving of EV heterogeneity and improvement in and standardization of isolation protocols are warranted to fully unleash the clinical potential of circulating EVs.

Acknowledgments

Due to constraints in length and number of citations, the authors were not able to cite many original research articles on this topic. The authors gratefully acknowledge support on related research in their laboratories from the National Cancer Institute.

Footnotes

Financial & competing interests disclosure

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending or royalties.

No writing assistance was utilized in the production of this manuscript.

Contributor Information

Crislyn D’Souza-Schorey, Email: cdsouzas@nd.edu, Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA.

Dolores Di Vizio, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA.

References

  • 1.Cocucci E, Racchetti G, Meldolesi J. Shedding microvesicles: artefacts no more. Trends Cell Biol. 2009;19:43–51. doi: 10.1016/j.tcb.2008.11.003. [DOI] [PubMed] [Google Scholar]
  • 2.D’Souza-Schorey C, Clancy JW. Tumor-derived microvesicles: shedding light on novel microenvironment modulators and prospective cancer biomarkers. Genes Dev. 2012;26:1287–99. doi: 10.1101/gad.192351.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Harding CV, Heuser JE, Stahl PD. Exosomes: looking back three decades and into the future. J Cell Biol. 2013;200:367–71. doi: 10.1083/jcb.201212113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Di Vizio D, Morello M, Dudley AC, et al. Large oncosomes in human prostate cancer tissues and in the circulation of mice with metastatic disease. Am J Pathol. 2012;181:1573–84. doi: 10.1016/j.ajpath.2012.07.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Muralidharan-Chari V, Clancy J, Plou C, et al. ARF6-regulated shedding of tumor cell-derived plasma membrane microvesicles. Curr Biol. 2009;19:1875–85. doi: 10.1016/j.cub.2009.09.059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Di Vizio D, Kim J, Hager MH, et al. Oncosome formation in prostate cancer: association with a region of frequent chromosomal deletion in metastatic disease. Cancer Res. 2009;69:5601–9. doi: 10.1158/0008-5472.CAN-08-3860. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Lee TH, D’Asti E, Magnus N, et al. Microvesicles as mediators of intercellular communication in cancer – the emerging science of cellular ‘debris’. Semin Immunopathol. 2011;33:455–67. doi: 10.1007/s00281-011-0250-3. [DOI] [PubMed] [Google Scholar]
  • 8.Cossetti C, Smith JA, Iraci N, et al. Extracellular membrane vesicles and immune regulation in the brain. Front Physiol. 2012;3:117. doi: 10.3389/fphys.2012.00117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.van Doormaal FF, Kleinjan A, Di Nisio M, et al. Cell-derived microvesicles and cancer. Neth J Med. 2009;67:266–73. [PubMed] [Google Scholar]
  • 10.ExoCarta. Exosome protein, lipid and lipid database. Available from: www.exocarta.org/
  • 11.Vesiclepedia. A community compendium of extracellular vesicles. Available from: http://microvesicles.org/
  • 12.Mathivanan S, Ji H, Simpson RJ. Exosomes: extracellular organelles important in intercellular communication. J Proteomics. 2010;73:1907–20. doi: 10.1016/j.jprot.2010.06.006. [DOI] [PubMed] [Google Scholar]
  • 13.van Niel G, Raposo G, Candalh C, et al. Intestinal epithelial cells secrete exosome-like vesicles. Gastroenterology. 2001;121:337–49. doi: 10.1053/gast.2001.26263. [DOI] [PubMed] [Google Scholar]
  • 14.Skokos D, Le Panse S, Villa I, et al. Mast cell-dependent B and T lymphocyte activation is mediated by the secretion of immunologically active exosomes. J Immunol. 2001;166:868–76. doi: 10.4049/jimmunol.166.2.868. [DOI] [PubMed] [Google Scholar]
  • 15.Thery C, Boussac M, Veron P, et al. Proteomic analysis of dendritic cell-derived exosomes: a secreted subcellular compartment distinct from apoptotic vesicles. J Immunol. 2001;166:7309–18. doi: 10.4049/jimmunol.166.12.7309. [DOI] [PubMed] [Google Scholar]
  • 16.Choi DS, Kim DK, Kim YK, et al. Proteomics, transcriptomics and lipidomics of exosomes and ectosomes. Proteomics. 2013;13:1554–71. doi: 10.1002/pmic.201200329. [DOI] [PubMed] [Google Scholar]
  • 17.Choi DS, Yang JS, Choi EJ, et al. The protein interaction network of extracellular vesicles derived from human colorectal cancer cells. J Proteome Res. 2012;11:1144–51. doi: 10.1021/pr200842h. [DOI] [PubMed] [Google Scholar]
  • 18.Tauro BJ, Greening DW, Mathias RA, et al. Two distinct populations of exosomes are released from LIM1863 colon carcinoma cell-derived organoids. Mol Cell Proteomics. 2013;12:587–98. doi: 10.1074/mcp.M112.021303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Yang W, Di Vizio D, Kirchner M, et al. Proteome scale characterization of human S-acylated proteins in lipid raft-enriched and non-raft membranes. Mol Cell Proteomics. 2010;9:54–70. doi: 10.1074/mcp.M800448-MCP200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Momen-Heravi F, Balaj L, Alian S, et al. Current methods for the isolation of extracellular vesicles. Biol Chem. 2013;394:1253–62. doi: 10.1515/hsz-2013-0141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Hung MC, Lau YK. Basic science of HER-2/neu: a review. Semin Oncol. 1999;26:51–9. [PubMed] [Google Scholar]
  • 22.Baran J, Baj-Krzyworzeka M, Weglarczyk K, et al. Circulating tumour-derived microvesicles in plasma of gastric cancer patients. Cancer Immunol Immunother. 2010;59:841–50. doi: 10.1007/s00262-009-0808-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Lakhal S, Wood MJ. Exosome nanotechnology: an emerging paradigm shift in drug delivery: exploitation of exosome nanovesicles for systemic in vivo delivery of RNAi heralds new horizons for drug delivery across biological barriers. Bioessays. 2011;33:737–41. doi: 10.1002/bies.201100076. [DOI] [PubMed] [Google Scholar]
  • 24.Alvarez-Erviti L, Seow Y, Yin H, et al. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat Biotechnol. 2011;29:341–5. doi: 10.1038/nbt.1807. [DOI] [PubMed] [Google Scholar]
  • 25.Noerholm M, Balaj L, Limperg T, et al. RNA expression patterns in serum microvesicles from patients with glioblastoma multiforme and controls. BMC Cancer. 2012;12:22. doi: 10.1186/1471-2407-12-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Wu CC, MacCoss MJ. Shotgun proteomics: tools for the analysis of complex biological systems. Curr Opin Mol Ther. 2002;4:242–50. [PubMed] [Google Scholar]
  • 27.Mani DR, Abbatiello SE, Carr SA. Statistical characterization of multiple-reaction monitoring mass spectrometry (MRM-MS) assays for quantitative proteomics. BMC Bioinformatics. 2012;13:S9. doi: 10.1186/1471-2105-13-S16-S9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Meissner F, Scheltema RA, Mollenkopf HJ, et al. Direct proteomic quantification of the secretome of activated immune cells. Science. 2013;340:475–8. doi: 10.1126/science.1232578. [DOI] [PubMed] [Google Scholar]

RESOURCES