Size-Exclusion Chromatography as a Technique for the Investigation of Novel Extracellular Vesicles in Cancer
Abstract
:Simple Summary
Abstract
1. Introduction
1.1. Isolation Methods in Brief
1.2. Size-Exclusion Chromatography, an Old but New Technique
2. Methodology of Review
2.1. Objectives
2.2. Search Methods
2.3. Selection Criteria
3. Results
3.1. Comparison of Isolation Methods
3.2. EV Characterisation, Concordance with MISEV Guidelines
3.3. EV-Associated Cargo
3.3.1. EV Proteomics
EV Protein Markers Are Differentially Expressed by Cancer Cells
EV Expression Highlights Differences in Clinical Outcome and Surgical Resectability
3.3.2. EV RNA
Clinical Studies Show That EVs from SEC Are Enriched with Functionally Active Small RNAs
EVs from Cell Lines Can also Be a Useful Source of RNA-Based Biomarkers
3.3.3. EV Functionality
Functional Studies Demonstrate Tumour-Derived EV Mechanisms Involved in Immunomodulation
4. Conclusions
Supplementary Materials
Author Contributions
Funding
Conflicts of Interest
Appendix A
- exosom*.ab:ti.
- EVs.ab,ti.
- (extracellular adj vesicl*).ab,ti.
- microsom*.ab,ti.
- vesicular.ab,ti.
- microvesic*.af.
- 1 or 2 or 3 or 4 or 5 or 6
- (size adj exclusion).af.
- gel filtration.af.
- 8 or 9
- cancer*.ab,ti.
- neoplas*.ab,ti.
- carcinoma*.ab,ti.
- adenocarcinoma*.ab,ti.
- 11 or 12 or 13 or 14
- 7 and 10 and 15
References
- Taylor, U.D.; Chou, I.-N.; Black, P.H. Isolation of plasma membrane fragments from cultured murine melanoma cells. Biochem. Biophys. Res. Commun. 1983, 113, 470–476. [Google Scholar] [CrossRef]
- Kim, J.W.; Wieckowski, E.; Taylor, D.D.; E Reichert, T.; Watkins, S.; Whiteside, T.L. Fas ligand-positive membranous vesicles isolated from sera of patients with oral cancer induce apoptosis of activated T lymphocytes. Clin. Cancer Res. 2005, 11, 1010–1020. [Google Scholar] [PubMed]
- Théry, C.; Boussac, M.; Véron, P.; Ricciardi-Castagnoli, P.; Raposo, G.; Garin, J.; Amigorena, S. Proteomic Analysis of Dendritic Cell-Derived Exosomes: A Secreted Subcellular Compartment Distinct from Apoptotic Vesicles. J. Immunol. 2001, 166, 7309–7318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Théry, C.; Ostrowski, M.; Segura, E. Membrane vesicles as conveyors of immune responses. Nat. Rev. Immunol. 2009, 9, 581–593. [Google Scholar] [CrossRef] [PubMed]
- Booth, A.M.; Fang, Y.; Fallon, J.K.; Yang, J.-M.; Hildreth, J.E.; Gould, S.J. Exosomes and HIV Gag bud from endosome-like domains of the T cell plasma membrane. J. Cell Biol. 2006, 172, 923–935. [Google Scholar] [CrossRef] [PubMed]
- Valadi, H.; Ekström, K.; Bossios, A.; Sjöstrand, M.; Lee, J.J.; Tvall, J.O.L.O. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 2007, 9, 654–659. [Google Scholar] [CrossRef] [Green Version]
- Lötvall, J.; Hill, A.; Hochberg, F.; Buzás, E.I.; Di Vizio, L.; Gardiner, C.; Gho, Y.S.; Kurochkin, I.V.; Mathivanan, S.; Quesenberry, P.; et al. Minimal Experimental Requirements for Definition of Extracellular Vesicles and their Functions: A Position Statement from the International Society for Extracellular Vesicles; Taylor & Francis: Oxfordshire, UK, 2014. [Google Scholar] [CrossRef]
- Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; 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. J. Extracell. Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef] [Green Version]
- Vaswani, K.; Mitchell, M.D.; Holland, O.J.; Koh, Y.Q.; Hill, R.J.; Harb, T.; Davies, P.S.W.; Peiris, H. A Method for the Isolation of Exosomes from Human and Bovine Milk. J. Nutr. Metab. 2019, 2019, 1–6. [Google Scholar] [CrossRef]
- Lässer, C.; Alikhani, V.S.; Ekström, K.; Eldh, M.; Paredes, P.T.; Bossios, A.; Sjöstrand, M.; Gabrielsson, S.; Lötvall, J.; Valadi, H. Human saliva, plasma and breast milk exosomes contain RNA: Uptake by macrophages. J. Transl. Med. 2011, 9, 9. [Google Scholar] [CrossRef] [Green Version]
- Czystowska-Kuzmicz, M.; Sosnowska, A.; Nowis, D.; Ramji, K.; Szajnik, M.; Chlebowska-Tuz, J.; Wolinska, E.; Gaj, P.; Grazul, M.; Pilch, Z.; et al. Small extracellular vesicles containing arginase-1 suppress T-cell responses and promote tumor growth in ovarian carcinoma. Nat. Commun. 2019, 10, 3000. [Google Scholar] [CrossRef]
- Masyuk, A.I.; Huang, B.Q.; Ward, C.J.; Gradilone, S.A.; Banales, J.M.; Masyuk, T.V.; Radtke, B.; Splinter, P.L.; LaRusso, N.F. Biliary exosomes influence cholangiocyte regulatory mechanisms and proliferation through interaction with primary cilia. Am. J. Physiol. Liver Physiol. 2010, 299, G990–G999. [Google Scholar] [CrossRef] [Green Version]
- Gardiner, C.; Di Vizio, D.; Sahoo, S.; Théry, C.; Witwer, K.W.; Wauben, M.; Hill, A.F. Techniques used for the isolation and characterization of extracellular vesicles: Results of a worldwide survey. J. Extracell. Vesicles 2016, 5, 32945. [Google Scholar] [CrossRef] [PubMed]
- Van Deun, J.; Mestdagh, P.; Agostinis, P.; Akay, Ö.; Anand, S.; Anckaert, J.; Martinez, Z.A.; Baetens, T.; Beghein, E.; EV-TRACK Consortium; et al. EV-TRACK: Transparent reporting and centralizing knowledge in extracellular vesicle research. Nat. Methods 2017, 14, 228–232. [Google Scholar] [CrossRef]
- Buschmann, D.; Kirchner, B.; Hermann, S.; Märte, M.; Wurmser, C.; Brandes, F.; Kotschote, S.; Bonin, M.; Steinlein, O.K.; Pfaffl, M.W.; et al. Evaluation of serum extracellular vesicle isolation methods for profiling miRNAs by next-generation sequencing. J. Extracell. Vesicles 2018, 7, 1481321. [Google Scholar] [CrossRef] [PubMed]
- Cvjetkovic, A.; Lötvall, J.; Lässer, C. The influence of rotor type and centrifugation time on the yield and purity of extracellular vesicles. J. Extracell. Vesicles 2014, 3. [Google Scholar] [CrossRef]
- Théry, C.; Amigorena, S.; Raposo, G.; Clayton, A. Isolation and Characterization of Exosomes from Cell Culture Supernatants and Biological Fluids. Curr. Protoc. Cell Biol. 2006, 30, 3.22.1. [Google Scholar] [CrossRef] [PubMed]
- Clayton, A.; Court, J.; Navabi, H.; Adams, M.; Mason, M.D.; A Hobot, J.; Newman, G.R.; Jasani, B. Analysis of antigen presenting cell derived exosomes, based on immuno-magnetic isolation and flow cytometry. J. Immunol. Methods 2001, 247, 163–174. [Google Scholar] [CrossRef]
- Taylor, D.D.; Shah, S. Methods of isolating extracellular vesicles impact down-stream analyses of their cargoes. Methods 2015, 87, 3–10. [Google Scholar] [CrossRef] [PubMed]
- Gámez-Valero, A.; Monguió-Tortajada, M.; Carreras-Planella, L.; Franquesa, M.; Beyer, K.; Borràs, F.E. Size-Exclusion Chromatography-based isolation minimally alters Extracellular Vesicles’ characteristics compared to precipitating agents. Sci. Rep. 2016, 6, srep33641. [Google Scholar] [CrossRef] [Green Version]
- Lathe, G.H.; Ruthven, C.R. The separation of substances and estimation of their relative molecular sizes by the use of colums of starch in water. Biochem. J. 1956, 62, 665–674. [Google Scholar] [CrossRef] [Green Version]
- Eisenstein, M. Adventures in the matrix. Nat. Chem. Biol. 2006, 3, 410. [Google Scholar] [CrossRef]
- Lane, R.E.; Korbie, D.; Trau, M.; Hill, M.M. Optimizing Size Exclusion Chromatography for Extracellular Vesicle Enrichment and Proteomic Analysis from Clinically Relevant Samples. Proteom. 2019, 19, e1800156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Coumans, F.A.W.; Brisson, A.R.; Buzás, E.I.; Dignat-George, F.; Drees, E.E.E.; El Andaloussi, S.; Emanueli, C.; Gasecka, A.; Hendrix, A.; Hill, A.F.; et al. Methodological Guidelines to Study Extracellular Vesicles. Circ. Res. 2017, 120, 1632–1648. [Google Scholar] [CrossRef] [PubMed]
- Böing, A.N.; Van Der Pol, E.; Grootemaat, A.E.; Coumans, F.A.W.; Sturk, A.; Nieuwland, R. Single-step isolation of extracellular vesicles by size-exclusion chromatography. J. Extracell. Vesicles 2014, 3, 23430. [Google Scholar] [CrossRef]
- Michell, D.L.; Vickers, K.C. Lipoprotein carriers of microRNAs. Biochim. et Biophys. Acta BBA Mol. Cell Biol. Lipids 2016, 1861, 2069–2074. [Google Scholar] [CrossRef] [Green Version]
- Buzás, E.I.; Tóth, E.Á.; Sódar, B.W.; Szabó-Taylor, K.É. Molecular interactions at the surface of extracellular vesicles. Semin. Immunopathol. 2018, 40, 453–464. [Google Scholar] [CrossRef] [Green Version]
- Morales-Kastresana, A.; Telford, B.; Musich, T.A.; McKinnon, K.; Clayborne, C.; Braig, Z.; Rosner, A.; Demberg, T.; Watson, D.C.; Karpova, T.S.; et al. Labeling Extracellular Vesicles for Nanoscale Flow Cytometry. Sci. Rep. 2017, 7, 1–10. [Google Scholar] [CrossRef]
- Dhondt, B.; Geeurickx, E.; Tulkens, J.; Van Deun, J.; Vergauwen, G.; Lippens, L.; Miinalainen, I.; Rappu, P.; Heino, J.; Ost, P.; et al. Unravelling the proteomic landscape of extracellular vesicles in prostate cancer by density-based fractionation of urine. J. Extracell. Vesicles 2020, 9, 1736935. [Google Scholar] [CrossRef]
- Van Deun, J.; Hendrix, A.; On behalf of the EV-TRACK Consortium. Is your article EV-TRACKed? J. Extracell. Vesicles 2017, 6, 1379835. [Google Scholar] [CrossRef] [Green Version]
- Kawakami, K.; Fujita, Y.; Matsuda, Y.; Arai, T.; Horie, K.; Kameyama, K.; Kato, T.; Masunaga, K.; Kasuya, Y.; Tanaka, M.; et al. Gamma-glutamyltransferase activity in exosomes as a potential marker for prostate cancer. BMC Cancer 2017, 17, 1–12. [Google Scholar] [CrossRef] [Green Version]
- Muller, L.; Hong, C.-S.; Stolz, D.B.; Watkins, S.C.; Whiteside, T.L. Isolation of biologically-active exosomes from human plasma. J. Immunol. Methods 2014, 411, 55–65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Van Eijndhoven, M.A.J.; Zijlstra, J.M.; Groenewegen, N.J.; Drees, E.E.E.; Van Niele, S.; Baglio, S.R.; Koppers-Lalic, D.; Van Der Voorn, H.; Libregts, S.F.W.M.; Wauben, M.H.M.; et al. Plasma vesicle miRNAs for therapy response monitoring in Hodgkin lymphoma patients. JCI Insight 2016, 1, e89631. [Google Scholar] [CrossRef] [PubMed]
- Théry, C.; Regnault, A.; Garin, J.; Wolfers, J.; Zitvogel, L.; Ricciardi-Castagnoli, P.; Raposo, G.; Amigorena, S. Molecular Characterization of Dendritic Cell-Derived Exosomes. J. Cell Biol. 1999, 147, 599–610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Andreu, Z.; Ýañez, M. Tetraspanins in Extracellular Vesicle Formation and Function. Front. Immunol. 2014, 5, 442. [Google Scholar] [CrossRef] [Green Version]
- Taylor, D.D.; Gerçel-Taylor, C.; Lyons, K.S.; Stanson, J.; Whiteside, T.L. T-cell apoptosis and suppression of T-cell receptor/CD3-zeta by Fas ligand-containing membrane vesicles shed from ovarian tumors. Clin. Cancer Res. 2003, 9, 5113–5119. [Google Scholar]
- Raposo, G.; Nijman, H.W.; Stoorvogel, W.; Liejendekker, R.; Harding, C.V.; Melief, C.J.; Geuze, H.J. B lymphocytes secrete antigen-presenting vesicles. J. Exp. Med. 1996, 183, 1161–1172. [Google Scholar] [CrossRef]
- Baran, J.; Baj-Krzyworzeka, M.; Weglarczyk, K.; Szatanek, R.; Zembala, M.; Barbasz, J.; Czupryna, A.; Szczepanik, A.; Zembala, M. Circulating tumour-derived microvesicles in plasma of gastric cancer patients. Cancer Immunol. Immunother. 2009, 59, 841–850. [Google Scholar] [CrossRef]
- Taylor, D.D.; Black, P.H. Shedding of plasma membrane fragments. Neoplastic and developmental importance. Dev. Boil. 1986, 3, 33–57. [Google Scholar]
- Hong, C.-S.; Muller, L.; Whiteside, T.L.; Boyiadzis, M. Plasma Exosomes as Markers of Therapeutic Response in Patients with Acute Myeloid Leukemia. Front. Immunol. 2014, 5, 160. [Google Scholar] [CrossRef] [Green Version]
- Lobb, R.J.; Becker, M.; Wen, S.W.; Wong, C.S.F.; Wiegmans, A.P.; Leimgruber, A.; Möller, A. Optimized exosome isolation protocol for cell culture supernatant and human plasma. J. Extracell. Vesicles 2015, 4, 27031. [Google Scholar] [CrossRef]
- Brennan, K.; Martin, K.; Fitzgerald, S.P.; O’Sullivan, J.; Wu, Y.; Blanco, A.; Richardson, C.; Mc Gee, M.M. A comparison of methods for the isolation and separation of extracellular vesicles from protein and lipid particles in human serum. Sci. Rep. 2020, 10, 1–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Djusberg, E.; Jernberg, E.; Thysell, E.; Golovleva, I.; Lundberg, P.; Crnalic, S.; Widmark, A.; Bergh, A.; Brattsand, M.; Wikström, P. High levels of the AR-V7 Splice Variant and Co-Amplification of the Golgi Protein CodingYIPF6inARAmplified Prostate Cancer Bone Metastases. Prostate 2017, 77, 625–638. [Google Scholar] [CrossRef] [PubMed]
- Broggi, M.A.; Maillat, L.; Clement, C.C.; Bordry, N.; Corthésy, P.; Auger, A.; Matter, M.; Hamelin, R.; Potin, L.; Demurtas, D.; et al. Tumor-associated factors are enriched in lymphatic exudate compared to plasma in metastatic melanoma patients. J. Exp. Med. 2019, 216, 1091–1107. [Google Scholar] [CrossRef] [Green Version]
- Shelke, G.V.; Lässer, C.; Gho, Y.S.; Lötvall, J. Importance of exosome depletion protocols to eliminate functional and RNA-containing extracellular vesicles from fetal bovine serum. J. Extracell. Vesicles 2014, 3, 24783. [Google Scholar] [CrossRef] [Green Version]
- Theodoraki, M.; Hoffmann, T.K.; Whiteside, T.L. Separation of plasma-derived exosomes into CD3(+) and CD3(-) fractions allows for association of immune cell and tumour cell markers with disease activity in HNSCC patients. Clin. Exp. Immunol. 2018, 192, 271–283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Theodoraki, M.-N.; Yerneni, S.S.; Hoffmann, T.K.; Gooding, W.E.; Whiteside, T.L. Clinical Significance of PD-L1+Exosomes in Plasma of Head and Neck Cancer Patients. Clin. Cancer Res. 2017, 24, 896–905. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Theodoraki, M.-N.; Yerneni, S.; Gooding, W.E.; Ohr, J.; Clump, D.A.; Bauman, J.E.; Ferris, R.L.; Whiteside, T.L. Circulating exosomes measure responses to therapy in head and neck cancer patients treated with cetuximab, ipilimumab, and IMRT. OncoImmunology 2019, 8, e1593805. [Google Scholar] [CrossRef]
- Chen, G.; Huang, A.C.; Zhang, W.; Zhang, G.; Wu, M.; Xu, W.; Yu, Z.; Yang, J.; Wang, B.; Sun, H.; et al. Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nat. Cell Biol. 2018, 560, 382–386. [Google Scholar] [CrossRef]
- Daassi, D.; Mahoney, K.M.; Freeman, G.J. The importance of exosomal PDL1 in tumour immune evasion. Nat. Rev. Immunol. 2020, 20, 209–215. [Google Scholar] [CrossRef]
- Yu, X.; Harris, S.L.; Levine, A.J. The Regulation of Exosome Secretion: A Novel Function of the p53 Protein. Cancer Res. 2006, 66, 4795–4801. [Google Scholar] [CrossRef] [Green Version]
- Yu, X.; Riley, T.; Levine, A.J. The regulation of the endosomal compartment byp53the tumor suppressor gene. FEBS J. 2009, 276, 2201–2212. [Google Scholar] [CrossRef] [PubMed]
- Abramowicz, A.; Wojakowska, A.; Marczak, L.; Lysek-Gladysinska, M.; Smolarz, M.; Story, M.D.; Polanska, J.; Widlak, P.; Pietrowska, M. Ionizing radiation affects the composition of the proteome of extracellular vesicles released by head-and-neck cancer cells in vitro. J. Radiat. Res. 2019, 60, 289–297. [Google Scholar] [CrossRef] [PubMed]
- Takahashi, A.; Okada, R.; Nagao, K.; Kawamata, Y.; Hanyu, A.; Yoshimoto, S.; Takasugi, M.; Watanabe, S.; Kanemaki, M.T.; Obuse, C.; et al. Exosomes maintain cellular homeostasis by excreting harmful DNA from cells. Nat. Commun. 2017, 8, 15287. [Google Scholar] [CrossRef] [Green Version]
- Freitas, D.; Balmaña, M.; Poças, J.; Campos, D.; Osório, H.; Konstantinidi, A.; Vakhrushev, S.Y.; Magalhães, A.; Reis, C.A. Different isolation approaches lead to diverse glycosylated extracellular vesicle populations. J. Extracell. Vesicles 2019, 8, 1621131. [Google Scholar] [CrossRef] [Green Version]
- Julien, S.; Videira, P.A.; Delannoy, P. Sialyl-Tn in Cancer: (How) Did We Miss the Target? Biomolecules 2012, 2, 435–466. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ludwig, S.; Marczak, Ł.; Sharma, P.; Abramowicz, A.; Gawin, M.; Widlak, P.; Whiteside, T.L.; Pietrowska, M. Proteomes of exosomes from HPV(+) or HPV(-) head and neck cancer cells: Differential enrichment in immunoregulatory proteins. OncoImmunology 2019, 8, 1593808. [Google Scholar] [CrossRef]
- Krupar, R.; Robold, K.; Gaag, D.; Spanier, G.; Kreutz, M.; Renner, K.; Hellerbrand, C.; Hofstaedter, F.; Bosserhoff, A.K. Immunologic and metabolic characteristics of HPV-negative and HPV-positive head and neck squamous cell carcinomas are strikingly different. Virchows Archiv 2014, 465, 299–312. [Google Scholar] [CrossRef] [PubMed]
- Chandran, V.I.; Welinder, C.; Månsson, A.-S.; Offer, S.; Freyhult, E.; Pernemalm, M.; Lund, S.M.; Pedersen, S.; Lehtiö, J.; Marko-Varga, G.; et al. Ultrasensitive Immunoprofiling of Plasma Extracellular Vesicles Identifies Syndecan-1 as a Potential Tool for Minimally Invasive Diagnosis of Glioma. Clin. Cancer Res. 2019, 25, 3115–3127. [Google Scholar] [CrossRef] [Green Version]
- Baietti, M.F.; Zhang, Z.; Mortier, E.; Melchior, A.; DeGeest, G.; Geeraerts, A.; Ivarsson, Y.; Depoortere, F.; Coomans, C.; Vermeiren, E.; et al. Syndecan–syntenin–ALIX regulates the biogenesis of exosomes. Nat. Cell Biol. 2012, 14, 677–685. [Google Scholar] [CrossRef]
- Danielsson, F.; Skogs, M.; Huss, M.; Rexhepaj, E.; O’Hurley, G.; Klevebring, D.; Pontén, F.; Gad, A.K.B.; Uhlén, M.; Lundberg, E. Majority of differentially expressed genes are down-regulated during malignant transformation in a four-stage model. Proc. Natl. Acad. Sci. 2013, 110, 6853–6858. [Google Scholar] [CrossRef] [Green Version]
- Anderson, N.L.; Polanski, M.; Pieper, R.; Gatlin, T.; Tirumalai, R.S.; Conrads, T.P.; Veenstra, T.D.; Adkins, J.N.; Pounds, J.G.; Fagan, R.; et al. The Human Plasma Proteome. Mol. Cell. Proteom. 2004, 3, 311–326. [Google Scholar] [CrossRef] [Green Version]
- Sakha, S.; Muramatsu, T.; Ueda, K.; Inazawa, J. Exosomal microRNA miR-1246 induces cell motility and invasion through the regulation of DENND2D in oral squamous cell carcinoma. Sci. Rep. 2016, 6, 38750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Peacock, B.; Rigby, A.; Bradford, J.R.; Pink, R.C.; Hunter, K.D.; Lambert, D.W.; Hunt, S. Extracellular vesicle micro RNA cargo is correlated with HPV status in oropharyngeal carcinoma. J. Oral Pathol. Med. 2018, 47, 954–963. [Google Scholar] [CrossRef] [PubMed]
- Rabinowits, G.; Gerçel-Taylor, C.; Day, J.M.; Taylor, D.D.; Kloecker, G.H. Exosomal MicroRNA: A Diagnostic Marker for Lung Cancer. Clin. Lung Cancer 2009, 10, 42–46. [Google Scholar] [CrossRef] [PubMed]
- Went, P.; Vasei, M.; Bubendorf, L.; Terracciano, L.; Tornillo, L.; Riede, U.; Kononen, J.; Simon, R.; Sauter, G.; A Baeuerle, P. Frequent high-level expression of the immunotherapeutic target Ep-CAM in colon, stomach, prostate and lung cancers. Br. J. Cancer 2006, 94, 128–135. [Google Scholar] [CrossRef] [Green Version]
- Taylor, D.D.; Gercel-Taylor, C. MicroRNA signatures of tumor-derived exosomes as diagnostic biomarkers of ovarian cancer. Gynecol. Oncol. 2008, 110, 13–21. [Google Scholar] [CrossRef] [PubMed]
- Tauro, B.J.; Greening, D.W.; Mathias, R.A.; Mathivanan, S.; Ji, H.; Simpson, R.J. Two Distinct Populations of Exosomes Are Released from LIM1863 Colon Carcinoma Cell-derived Organoids. Mol. Cell. Proteom. 2012, 12, 587–598. [Google Scholar] [CrossRef] [Green Version]
- Hong, C.-S.; Funk, S.; Muller, L.; Boyiadzis, M.; Whiteside, T.L. Isolation of biologically active and morphologically intact exosomes from plasma of patients with cancer. J. Extracell. Vesicles 2016, 5, 29289. [Google Scholar] [CrossRef]
- Simonsen, J.B. Pitfalls associated with lipophilic fluorophore staining of extracellular vesicles for uptake studies. J. Extracell. Vesicles 2019, 8, 1582237. [Google Scholar] [CrossRef] [Green Version]
- Takov, K.; Yellon, D.M.; Davidson, S.M. Confounding factors in vesicle uptake studies using fluorescent lipophilic membrane dyes. J. Extracell. Vesicles 2017, 6, 1388731. [Google Scholar] [CrossRef]
- Lehrich, B.M.; Liang, Y.; Khosravi, P.; Federoff, H.J.; Fiandaca, M.S. Fetal Bovine Serum-Derived Extracellular Vesicles Persist within Vesicle-Depleted Culture Media. Int. J. Mol. Sci. 2018, 19, 3538. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Auber, M.; Fröhlich, D.; Drechsel, O.; Karaulanov, E.; Krämer-Albers, E.-M. Serum-free media supplements carry miRNAs that co-purify with extracellular vesicles. J. Extracell. Vesicles 2019, 8, 1656042. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Appaiah, H.N.; Goswami, C.P.; A Mina, L.; Badve, S.; Sledge, J.G.W.; Liu, Y.; Nakshatri, H. Persistent upregulation of U6:SNORD44 small RNA ratio in the serum of breast cancer patients. Breast Cancer Res. 2011, 13, R86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xiang, M.; Zeng, Y.; Yang, R.; Xu, H.; Chen, Z.; Zhong, J.; Xie, H.; Xu, Y.; Zeng, X. U6 is not a suitable endogenous control for the quantification of circulating microRNAs. Biochem. Biophys. Res. Commun. 2014, 454, 210–214. [Google Scholar] [CrossRef] [PubMed]
- Mestdagh, P.; Van Vlierberghe, P.; De Weer, A.; Muth, D.; Westermann, F.; Speleman, F.; Vandesompele, J. A novel and universal method for microRNA RT-qPCR data normalization. Genome Biol. 2009, 10, R64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pan, C.; Issaeva, N.; Yarbrough, W.G. HPV-driven oropharyngeal cancer: Current knowledge of molecular biology and mechanisms of carcinogenesis. Cancers Head Neck 2018, 3, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wieckowski, E.U.; Visus, C.; Szajnik, M.; Szczepanski, M.J.; Storkus, W.J.; Whiteside, T.L. Tumor-Derived Microvesicles Promote Regulatory T Cell Expansion and Induce Apoptosis in Tumor-Reactive Activated CD8+ T Lymphocytes. J. Immunol. 2009, 183, 3720–3730. [Google Scholar] [CrossRef] [Green Version]
- Antonioli, L.; Blandizzi, C.; Pacher, P.; Haskó, G. Immunity, inflammation and cancer: A leading role for adenosine. Nat. Rev. Cancer 2013, 13, 842–857. [Google Scholar] [CrossRef]
- Schuler, P.J.; Saze, Z.; Hong, C.-S.; Muller, L.; Gillespie, D.G.; Cheng, D.; Harasymczuk, M.; Mandapathil, M.; Lang, S.; Jackson, E.K.; et al. Human CD4+CD39+regulatory T cells produce adenosine upon co-expression of surface CD73 or contact with CD73+exosomes or CD73+cells. Clin. Exp. Immunol. 2014, 177, 531–543. [Google Scholar] [CrossRef]
- Ludwig, S.; Floros, T.; Theodoraki, M.-N.; Hong, C.-S.; Jackson, E.K.; Lang, S.; Whiteside, T.L. Suppression of Lymphocyte Functions by Plasma Exosomes Correlates with Disease Activity in Patients with Head and Neck Cancer. Clin. Cancer Res. 2017, 23, 4843–4854. [Google Scholar] [CrossRef] [Green Version]
- Ludwig, N.; Yerneni, S.S.; Razzo, B.M.; Whiteside, T.L. Exosomes from HNSCC Promote Angiogenesis through Reprogramming of Endothelial Cells. Mol. Cancer Res. 2018, 16, 1798–1808. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Theodoraki, M.; Yerneni, S.; Lorenz, K.; Laban, S.; Schuler, P.; Brunner, C.; Hoffmann, T.; Theodorakis, I.; Whiteside, T. PO-243 Exosomes from plasma of HNSCC patients treated with photodynamic therapy are biomarkers for epithelial-mesenchymal transition. Poster Presentation 2018, 3, A322. [Google Scholar] [CrossRef] [Green Version]
- Royo, F.; Moreno, L.; Mleczko, J.; Palomo, L.; Gonzalez, E.; Cabrera, D.; Cogolludo, A.; Vizcaino, F.P.; Van-Liempd, S.; Falcón-Pérez, J.M. Hepatocyte-secreted extracellular vesicles modify blood metabolome and endothelial function by an arginase-dependent mechanism. Sci. Rep. 2017, 7, srep42798. [Google Scholar] [CrossRef] [PubMed]
- Zhang, H.; Liu, J.; Qu, D.; Wang, L.; Wong, C.M.; Lau, C.-W.; Huang, Y.; Wang, Y.F.; Huang, H.; Xia, Y.; et al. Serum exosomes mediate delivery of arginase 1 as a novel mechanism for endothelial dysfunction in diabetes. Proc. Natl. Acad. Sci. 2018, 115, E6927–E6936. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sjöqvist, S.; Kasai, Y.; Shimura, D.; Ishikawa, T.; Ali, N.; Iwata, T.; Kanai, N. Oral keratinocyte-derived exosomes regulate proliferation of fibroblasts and epithelial cells. Biochem. Biophys. Res. Commun. 2019, 514, 706–712. [Google Scholar] [CrossRef] [PubMed]
- Arroyo, J.D.; Chevillet, J.R.; Kroh, E.M.; Ruf, I.K.; Pritchard, C.C.; Gibson, D.F.; Mitchell, P.S.; Bennett, C.F.; Pogosova-Agadjanyan, E.L.; Stirewalt, D.L.; et al. Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma. Proc. Natl. Acad. Sci. 2011, 108, 5003–5008. [Google Scholar] [CrossRef] [Green Version]
- Suárez, H.; Gámez-Valero, A.; Reyes, R.; López-Martín, S.; Rodríguez, M.J.; Carrascosa, J.L.; Cabañas, C.; Borràs, F.E.; Yáñez-Mo, M. A bead-assisted flow cytometry method for the semi-quantitative analysis of Extracellular Vesicles. Sci. Rep. 2017, 7, 11271. [Google Scholar] [CrossRef] [Green Version]
- Jeyaram, A.; Jay, S.M. Preservation and Storage Stability of Extracellular Vesicles for Therapeutic Applications. AAPS J. 2017, 20, 1–7. [Google Scholar] [CrossRef]
- Patel, G.K.; Khan, M.A.; Zubair, H.; Srivastava, S.K.; Khushman, M.; Singh, S.; Singh, A.P. Comparative analysis of exosome isolation methods using culture supernatant for optimum yield, purity and downstream applications. Sci. Rep. 2019, 9, 1–10. [Google Scholar] [CrossRef] [Green Version]
- Welton, J.L.; Webber, J.P.; Botos, L.-A.; Jones, M.; Clayton, A. Ready-made chromatography columns for extracellular vesicle isolation from plasma. J. Extracell. Vesicles 2015, 4, 27269. [Google Scholar] [CrossRef]
- Dong, X.; Chi, J.; Zheng, L.; Ma, B.; Li, Z.; Wang, S.; Zhao, C.; Liu, H. Efficient isolation and sensitive quantification of extracellular vesicles based on an integrated ExoID-Chip using photonic crystals. Lab a Chip 2019, 19, 2897–2904. [Google Scholar] [CrossRef]
- Smith, J.T.; Wunsch, B.H.; Dogra, N.; Ahsen, M.E.; Lee, K.; Yadav, K.K.; Weil, R.; Pereira, M.A.; Patel, J.V.; Duch, E.A.; et al. Integrated nanoscale deterministic lateral displacement arrays for separation of extracellular vesicles from clinically-relevant volumes of biological samples. Lab a Chip 2018, 18, 3913–3925. [Google Scholar] [CrossRef] [PubMed]
- Wunsch, B.H.; Smith, J.T.; Gifford, S.M.; Wang, C.; Brink, M.; Bruce, R.L.; Austin, R.H.; Stolovitzky, G.; Astier, B.H.W.J.T.S.S.M.G.C.W.M.B.R.L.B.G.S.Y. Nanoscale lateral displacement arrays for the separation of exosomes and colloids down to 20 nm. Nat. Nanotechnol. 2016, 11, 936–940. [Google Scholar] [CrossRef] [PubMed]
- Davies, R.T.; Kim, J.; Jang, S.C.; Choi, E.-J.; Gho, Y.S.; Park, J. Microfluidic filtration system to isolate extracellular vesicles from blood. Lab a Chip 2012, 12, 5202–5210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, H.; Lyden, D. Asymmetric-flow field-flow fractionation technology for exomere and small extracellular vesicle separation and characterization. Nat. Protoc. 2019, 14, 1027–1053. [Google Scholar] [CrossRef] [PubMed]
Method | Frequency |
---|---|
Sepharose (either 2B or 4B) | 13 |
MiniSEC (using Sepharose 2B) | 10 |
qEV column (proprietary filtration matrix) | 10 |
Sephacryl (S-400 or 500 HR) | 2 |
Microfluidic Platforms | 2 |
EVSecond (modified styrene-divinylbenzene matrix) | 1 |
Bio-Gel A (Agarose) | 1 |
Experimental Parameters Related to EV Separation | |
Density Gradient | Type and concentrations of the gradient performed, use as validation of EVs in experiments. |
EV Density | EV density should be reported if undertaken. |
Ultracentrifugation Specifics | When ultracentrifugation is used, there should be a description of the g-force, duration and rotor type of these steps. |
Protein Analysis | |
EV-Enriched Proteins | 3 or more EV-enriched proteins. |
Non EV-Enriched Protein | Assessment of one or more EV-enriched protein. |
Antibody Specifics | Antibodies used for immunoblotting should be reported to include clone/reference number and dilutions. |
Lysate Preparation | Lysis of EVs should involve reporting of buffer composition. |
Particle Analysis | |
Qualitative and Quantitative Analysis | Implementation of both qualitative (e.g., electron microscopy) and quantitative methods (e.g., nanoparticle tracking analysis, high-resolution flow cytometry). Quantitative methods should include reporting of particle concentration. |
Electron Microscopy Images | Inclusion of both wide-field and a close-up electron microscopy image. |
Study Type | Number |
---|---|
Proteomics | 13 |
Biological Function | 9 |
Feasibility of Method | 11 |
RNA | 4 |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Liu, D.S.K.; Upton, F.M.; Rees, E.; Limb, C.; Jiao, L.R.; Krell, J.; Frampton, A.E. Size-Exclusion Chromatography as a Technique for the Investigation of Novel Extracellular Vesicles in Cancer. Cancers 2020, 12, 3156. https://doi.org/10.3390/cancers12113156
Liu DSK, Upton FM, Rees E, Limb C, Jiao LR, Krell J, Frampton AE. Size-Exclusion Chromatography as a Technique for the Investigation of Novel Extracellular Vesicles in Cancer. Cancers. 2020; 12(11):3156. https://doi.org/10.3390/cancers12113156
Chicago/Turabian StyleLiu, Daniel S. K., Flora M. Upton, Eleanor Rees, Christopher Limb, Long R. Jiao, Jonathan Krell, and Adam E. Frampton. 2020. "Size-Exclusion Chromatography as a Technique for the Investigation of Novel Extracellular Vesicles in Cancer" Cancers 12, no. 11: 3156. https://doi.org/10.3390/cancers12113156
APA StyleLiu, D. S. K., Upton, F. M., Rees, E., Limb, C., Jiao, L. R., Krell, J., & Frampton, A. E. (2020). Size-Exclusion Chromatography as a Technique for the Investigation of Novel Extracellular Vesicles in Cancer. Cancers, 12(11), 3156. https://doi.org/10.3390/cancers12113156