A novel multiplex bead-based platform highlights the diversity of extracellular vesicles

Nina Koliha, Yvonne Wiencek, Ute Heider, Christian Jüngst, Nikolay Kladt, Susanne Krauthäuser, Ian C D Johnston, Andreas Bosio, Astrid Schauss, Stefan Wild, Nina Koliha, Yvonne Wiencek, Ute Heider, Christian Jüngst, Nikolay Kladt, Susanne Krauthäuser, Ian C D Johnston, Andreas Bosio, Astrid Schauss, Stefan Wild

Abstract

The surface protein composition of extracellular vesicles (EVs) is related to the originating cell and may play a role in vesicle function. Knowledge of the protein content of individual EVs is still limited because of the technical challenges to analyse small vesicles. Here, we introduce a novel multiplex bead-based platform to investigate up to 39 different surface markers in one sample. The combination of capture antibody beads with fluorescently labelled detection antibodies allows the analysis of EVs that carry surface markers recognized by both antibodies. This new method enables an easy screening of surface markers on populations of EVs. By combining different capture and detection antibodies, additional information on relative expression levels and potential vesicle subpopulations is gained. We also established a protocol to visualize individual EVs by stimulated emission depletion (STED) microscopy. Thereby, markers on single EVs can be detected by fluorophore-conjugated antibodies. We used the multiplex platform and STED microscopy to show for the first time that NK cell-derived EVs and platelet-derived EVs are devoid of CD9 or CD81, respectively, and that EVs isolated from activated B cells comprise different EV subpopulations. We speculate that, according to our STED data, tetraspanins might not be homogenously distributed but may mostly appear as clusters on EV subpopulations. Finally, we demonstrate that EV mixtures can be separated by magnetic beads and analysed subsequently with the multiplex platform. Both the multiplex bead-based platform and STED microscopy revealed subpopulations of EVs that have been indistinguishable by most analysis tools used so far. We expect that an in-depth view on EV heterogeneity will contribute to our understanding of different EVs and functions.

Keywords: B cell; NK cell; STED; exosome; flow cytometry; magnetic isolation; platelet.

Figures

Fig. 1
Fig. 1
The multiplex bead-based platform. (a) Workflow of the multiplex platform. Isolated EVs or cell culture supernatant were incubated overnight with up to 39 different bead populations, each coupled to a different capture antibody. The different bead populations are distinguishable by flow cytometry. EVs bound to the beads were detected with single antibodies, e.g. cell-specific markers, or exosome markers such as anti-CD9-APC, anti-CD63-APC, anti-CD81-APC antibodies, or with a cocktail of the latter 3 antibodies. (b–d) Analysis example showing (b) exclusion of doublets and no bead events, (c) discrimination of differently labelled bead populations, and (d) measurement of signal intensities of the single bead populations. Positive bead populations are highlighted in colours. Black events represent beads that did not bind EVs or beads with EVs not detected by the staining cocktail.
Fig. 2
Fig. 2
Surface marker profiles of NK and platelet EVs. Background corrected APC median signal intensities after incubation of NK cell–derived EVs from 4 donors (8 µg each) or platelet-derived EVs from 11 donors (32 µg each) with 39 capture antibody bead populations, followed by staining with a cocktail of anti-CD9-, anti-CD63- and anti-CD81-APC antibodies. REA, mIgG1, mIgG2a and mIgG2b indicate isotype control-beads, i.e. beads linked to an antibody which does not bind EVs.
Fig. 3
Fig. 3
(a, b) Theoretical example of matrix profiles (a) of EVs carrying 10 molecules A and 1 molecule B on each EV or (b) of EVs carrying 10 molecules A and 50% of the EVs comprise also 10 molecules B. Every detected molecule giving a signal of 1 light unit. (c–f) Background corrected APC median signal intensities of anti-CD9-, anti-CD63-, anti-CD81-beads and isotype control-beads after incubation with (c) 16 µg NK cell–derived EVs, (d) 16 µg platelet-derived EVs, (e) a mixture of NK cell–derived EVs and platelet-derived EVs (8 µg each) or (f) 8 µg monocyte-derived EVs, followed by staining with anti-CD9-APC, anti-CD63-APC, or anti-CD81-APC antibodies or with a cocktail of these antibodies. MIgG1 indicates isotype control-beads.
Fig. 4
Fig. 4
Matrix profile of B cell EVs. Background corrected APC median signal intensity of different capture antibody bead populations after incubation with 32 µg EVs from activated B cells, followed by staining with anti-CD19-APC, anti-CD20-APC, anti-CD69-APC, anti-CD80-APC, anti-CD86-APC, anti-HLA-DQ-APC, or a cocktail of anti-CD9-APC, anti-CD63-APC and anti-CD81-APC antibodies. REA and mIgG1 indicate isotype control-beads.
Fig. 5
Fig. 5
NK cell–derived EVs bound to an anti-CD63-coated glass slide, stained with anti-CD81-STAR RED, and visualized by (a) confocal microscopy at 100-fold magnitude and (b) STED. (c) Intensity profile of the image section marked with the white line after confocal microscopy (grey) and STED microscopy (red). Scale bars represent 1 µm.
Fig. 6
Fig. 6
STED analysis of EVs bound to an anti-CD63-coated glass slides. (a) A mixture of NK cell EVs and platelet EVs stained with anti-CD9-STAR488 and anti-CD81-STAR RED and (b) the corresponding intensity profile. Scale bar represents 500 nm.
Fig. 7
Fig. 7
STED analysis of B cell EVs bound to anti-CD9/63/81-coated slides and stained with (a) anti-CD19-STAR488 and anti-CD9-STAR RED or (b) stained with anti-CD9-STAR488 and anti-CD42a-STAR RED. Scale bars represent 500 nm.
Fig. 8
Fig. 8
STED analysis of EVs bound to anti-CD63-coated glass slides. (a) Monocyte EVs stained with anti-CD81-STAR488 and anti-IgG1-STAR RED and (b) corresponding intensity profile, (c) monocyte EVs stained with anti-CD9-STAR488 and anti-CD81-STAR RED and (d) monocyte EVs stained with anti-CD81-STAR488 and anti-CD9-STAR RED. (e) Overview of the percentage of double-positive monocyte EVs from 3 images and total number of analysed spots. (f) Monocyte EVs stained with anti-CD81-STAR488 and anti-CD81-STAR RED and (g) corresponding intensity profile. Scale bars represent 500 nm.

References

    1. EL Andaloussi S, Mager I, Breakefield XO, Wood MJ. Extracellular vesicles: biology and emerging therapeutic opportunities. Nat Rev Drug Discov. 2013;12:347–57.
    1. Thery C, Ostrowski M, Segura E. Membrane vesicles as conveyors of immune responses. Nat Rev. 2009;9:581–93.
    1. Thery C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and function. Nat Rev. 2002;2:569–79.
    1. Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol. 2013;200:373–83.
    1. Caby MP, Lankar D, Vincendeau-Scherrer C, Raposo G, Bonnerot C. Exosomal-like vesicles are present in human blood plasma. Int Immunol. 2005;17:879–87.
    1. Aalberts M, van Dissel-Emiliani FM, van Adrichem NP, van Wijnen M, Wauben MH, Stout TA, et al. Identification of distinct populations of prostasomes that differentially express prostate stem cell antigen, annexin A1, and GLIPR2 in humans. Biol Reprod. 2012;86:82.
    1. Pisitkun T, Shen RF, Knepper MA. Identification and proteomic profiling of exosomes in human urine. Proc Natl Acad Sci U S A. 2004;101:13368–73.
    1. Ogawa Y, Miura Y, Harazono A, Kanai-Azuma M, Akimoto Y, Kawakami H, et al. Proteomic analysis of two types of exosomes in human whole saliva. Biol Pharmaceut Bull. 2011;34:13–23.
    1. Admyre C, Johansson SM, Qazi KR, Filen JJ, Lahesmaa R, Norman M, et al. Exosomes with immune modulatory features are present in human breast milk. J Immunol. 2007;179:1969–78.
    1. Andre F, Schartz NE, Movassagh M, Flament C, Pautier P, Morice P, et al. Malignant effusions and immunogenic tumour-derived exosomes. Lancet. 2002;360:295–305.
    1. Street JM, Barran PE, Mackay CL, Weidt S, Balmforth C, Walsh TS, et al. Identification and proteomic profiling of exosomes in human cerebrospinal fluid. J Transl Med. 2012;10:5.
    1. Lasser C, Alikhani VS, Ekstrom K, Eldh M, Paredes PT, Bossios A, et al. Human saliva, plasma and breast milk exosomes contain RNA: uptake by macrophages. J Transl Med. 2011;9:9.
    1. Mittelbrunn M, Gutierrez-Vazquez C, Villarroya-Beltri C, Gonzalez S, Sanchez-Cabo F, Gonzalez MA, et al. Unidirectional transfer of microRNA-loaded exosomes from T cells to antigen-presenting cells. Nat Comm. 2011;2:282.
    1. Valadi H, Ekstrom K, Bossios A, Sjostrand M, Lee JJ, Lotvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007;9:654–9.
    1. Simpson RJ, Lim JW, Moritz RL, Mathivanan S. Exosomes: proteomic insights and diagnostic potential. Expert Rev Proteomics. 2009;6:267–83.
    1. Levy S, Shoham T. The tetraspanin web modulates immune-signalling complexes. Nat Rev. 2005;5:136–48.
    1. Escola JM, Kleijmeer MJ, Stoorvogel W, Griffith JM, Yoshie O, Geuze HJ. Selective enrichment of tetraspan proteins on the internal vesicles of multivesicular endosomes and on exosomes secreted by human B-lymphocytes. J Biol Chem. 1998;273:20121–7.
    1. Andreu Z, Yanez-Mo M. Tetraspanins in extracellular vesicle formation and function. Front Immunol. 2014;5:442.
    1. van Niel G, Charrin S, Simoes S, Romao M, Rochin L, Saftig P, et al. The tetraspanin CD63 regulates ESCRT-independent and -dependent endosomal sorting during melanogenesis. Dev Cell. 2011;21:708–21.
    1. Chairoungdua A, Smith DL, Pochard P, Hull M, Caplan MJ. Exosome release of beta-catenin: a novel mechanism that antagonizes Wnt signaling. J Cell Biol. 2010;190:1079–91.
    1. Buschow SI, Nolte-‘t Hoen EN, van Niel G, Pols MS, ten Broeke T, Lauwen M, et al. MHC II in dendritic cells is targeted to lysosomes or T cell-induced exosomes via distinct multivesicular body pathways. Traffic. 2009;10:1528–42.
    1. van der Pol E, Coumans FA, Grootemaat AE, Gardiner C, Sargent IL, Harrison P, et al. Particle size distribution of exosomes and microvesicles determined by transmission electron microscopy, flow cytometry, nanoparticle tracking analysis, and resistive pulse sensing. J Thromb Haemost. 2014;12:1182–92.
    1. Thery C, Amigorena S, Raposo G, Clayton A. Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Curr Protoc Cell Biol. Chapter 3: Unit 3.22.
    1. Kormelink TG, Arkesteijn GJ, Nauwelaers FA, van den Engh G, Nolte-‘t Hoen EN, Wauben MH. Prerequisites for the analysis and sorting of extracellular vesicle subpopulations by high-resolution flow cytometry. Cytometry A. 2015
    1. van der Vlist EJ, Nolte-‘t Hoen EN, Stoorvogel W, Arkesteijn GJ, Wauben MH. Fluorescent labeling of nano-sized vesicles released by cells and subsequent quantitative and qualitative analysis by high-resolution flow cytometry. Nat Protocol. 2012;7:1311–26.
    1. Vicidomini G, Schonle A, Ta H, Han KY, Moneron G, Eggeling C, et al. STED nanoscopy with time-gated detection: theoretical and experimental aspects. PLoS One. 2013;8:e54421.
    1. Willig KI, Rizzoli SO, Westphal V, Jahn R, Hell SW. STED microscopy reveals that synaptotagmin remains clustered after synaptic vesicle exocytosis. Nature. 2006;440:935–9.
    1. Heijnen HF, Schiel AE, Fijnheer R, Geuze HJ, Sixma JJ. Activated platelets release two types of membrane vesicles: microvesicles by surface shedding and exosomes derived from exocytosis of multivesicular bodies and alpha-granules. Blood. 1999;94:3791–9.
    1. Leong HS, Podor TJ, Manocha B, Lewis JD. Validation of flow cytometric detection of platelet microparticles and liposomes by atomic force microscopy. J Thromb Hemostasis. 2011;9:2466–76.
    1. Mulcahy LA, Pink RC, Carter DR. Routes and mechanisms of extracellular vesicle uptake. J Extracell Vesicles. 2014;3 24641, doi: .
    1. Ida H, Anderson P. Activation-induced NK cell death triggered by CD2 stimulation. Eur J Immunol. 1998;28:1292–300.
    1. Addison EG, North J, Bakhsh I, Marden C, Haq S, Al-Sarraj S, et al. Ligation of CD8alpha on human natural killer cells prevents activation-induced apoptosis and enhances cytolytic activity. Immunology. 2005;116:354–61.
    1. Caligiuri MA. Human natural killer cells. Blood. 2008;112:461–9.
    1. Schmitz G, Rothe G, Ruf A, Barlage S, Tschope D, Clemetson KJ, et al. European working group on clinical cell analysis: consensus protocol for the flow cytometric characterisation of platelet function. Thromb Haemost. 1998;79:885–96.
    1. Robbins PD, Morelli AE. Regulation of immune responses by extracellular vesicles. Nat Rev. 2014;14:195–208.
    1. Wolfers J, Lozier A, Raposo G, Regnault A, Thery C, Masurier C, et al. Tumor-derived exosomes are a source of shared tumor rejection antigens for CTL cross-priming. Nat Med. 2001;7:297–303.
    1. Huber V, Fais S, Iero M, Lugini L, Canese P, Squarcina P, et al. Human colorectal cancer cells induce T-cell death through release of proapoptotic microvesicles: role in immune escape. Gastroenterology. 2005;128:1796–804.
    1. Klibi J, Niki T, Riedel A, Pioche-Durieu C, Souquere S, Rubinstein E, et al. Blood diffusion and Th1-suppressive effects of galectin-9-containing exosomes released by Epstein-Barr virus-infected nasopharyngeal carcinoma cells. Blood. 2009;113:1957–66.
    1. Revenfeld AL, Baek R, Nielsen MH, Stensballe A, Varming K, Jorgensen M. Diagnostic and prognostic potential of extracellular vesicles in peripheral blood. Clin Ther. 2014;36:830–46.
    1. Pant S, Hilton H, Burczynski ME. The multifaceted exosome: biogenesis, role in normal and aberrant cellular function, and frontiers for pharmacological and biomarker opportunities. Biochem Pharmacol. 2012;83:1484–94.
    1. Gould SJ, Raposo G. As we wait: coping with an imperfect nomenclature for extracellular vesicles. J Extracell Vesicles. 2013;2 20389, doi: .
    1. van der Pol E, Coumans F, Varga Z, Krumrey M, Nieuwland R. Innovation in detection of microparticles and exosomes. J Thromb Haemost. 2013;11(Suppl 1):36–45.
    1. Jorgensen M, Baek R, Pedersen S, Sondergaard EK, Kristensen SR, Varming K. Extracellular Vesicle (EV) Array: microarray capturing of exosomes and other extracellular vesicles for multiplexed phenotyping. J Extracell Vesicles. 2013;2 20920, doi: .
    1. Estelles A, Sperinde J, Roulon T, Aguilar B, Bonner C, LePecq JB, et al. Exosome nanovesicles displaying G protein-coupled receptors for drug discovery. Int J Nanomedicine. 2007;2:751–60.
    1. Higginbotham JN, Demory Beckler M, Gephart JD, Franklin JL, Bogatcheva G, Kremers GJ, et al. Amphiregulin exosomes increase cancer cell invasion. Curr Biol. 2011;21:779–86.
    1. Simpson RJ, Jensen SS, Lim JW. Proteomic profiling of exosomes: current perspectives. Proteomics. 2008;8:4083–99.
    1. Zuidscherwoude M, Gottfert F, Dunlock VM, Figdor CG, van den Bogaart G, Spriel AB. The tetraspanin web revisited by super-resolution microscopy. Sci Rep. 2015;5:12201.
    1. Kovalenko OV, Yang X, Kolesnikova TV, Hemler ME. Evidence for specific tetraspanin homodimers: inhibition of palmitoylation makes cysteine residues available for cross-linking. Biochem J. 2004;377:407–17.
    1. Min G, Wang H, Sun TT, Kong XP. Structural basis for tetraspanin functions as revealed by the cryo-EM structure of uroplakin complexes at 6-A resolution. J Cell Biol. 2006;173:975–83.
    1. Coren LV, Shatzer T, Ott DE. CD45 immunoaffinity depletion of vesicles from Jurkat T cells demonstrates that exosomes contain CD45: no evidence for a distinct exosome/HIV-1 budding pathway. Retrovirology. 2008;5:64.
    1. Clayton A, Court J, Navabi H, Adams M, Mason MD, Hobot JA, et al. Analysis of antigen presenting cell derived exosomes, based on immuno-magnetic isolation and flow cytometry. J Immunol Meth. 2001;247:163–74.
    1. Koga K, Matsumoto K, Akiyoshi T, Kubo M, Yamanaka N, Tasaki A, et al. Purification, characterization and biological significance of tumor-derived exosomes. Anticancer Res. 2005;25:3703–7.
    1. Taylor DD, Gercel-Taylor C. MicroRNA signatures of tumor-derived exosomes as diagnostic biomarkers of ovarian cancer. Gynecol Oncol. 2008;110:13–21.
    1. Tauro BJ, Greening DW, Mathias RA, Ji H, Mathivanan S, Scott AM, et al. Comparison of ultracentrifugation, density gradient separation, and immunoaffinity capture methods for isolating human colon cancer cell line LIM1863-derived exosomes. Methods. 2012;56:293–304.
    1. Gieseler F, Gamperl H, Theophil F, Stenzel I, Quecke T, Ungefroren H, et al. Using annexin V-coated magnetic beads to capture active tissue factor-bearing microparticles from body fluids. Cell Biol Int. 2014;38:277–81.
    1. Lugini L, Cecchetti S, Huber V, Luciani F, Macchia G, Spadaro F, et al. Immune surveillance properties of human NK cell-derived exosomes. J Immunol. 2012;189:2833–42.
    1. Simpson RJ, Kalra H, Mathivanan S. ExoCarta as a resource for exosomal research. J Extracell Vesicles. 2012;1 18374, doi: .
    1. Aatonen MT, Ohman T, Nyman TA, Laitinen S, Gronholm M, Siljander PR. Isolation and characterization of platelet-derived extracellular vesicles. J Extracell Vesicles. 2014;3 24692, doi: .
    1. Arita S, Baba E, Shibata Y, Niiro H, Shimoda S, Isobe T, et al. B cell activation regulates exosomal HLA production. Eur J Immunol. 2008;38:1423–34.
    1. Admyre C, Bohle B, Johansson SM, Focke-Tejkl M, Valenta R, Scheynius A, et al. B cell-derived exosomes can present allergen peptides and activate allergen-specific T cells to proliferate and produce TH2-like cytokines. J Allergy Clin Immunol. 2007;120:1418–24.
    1. Muntasell A, Berger AC, Roche PA. T cell-induced secretion of MHC class II-peptide complexes on B cell exosomes. EMBO J. 2007;26:4263–72.
    1. Oksvold MP, Kullmann A, Forfang L, Kierulf B, Li M, Brech A, et al. Expression of B-cell surface antigens in subpopulations of exosomes released from B-cell lymphoma cells. Clin Ther. 2014;36:847–62.e1.
    1. Maecker HT, Todd SC, Levy S. The tetraspanin superfamily: molecular facilitators. FASEB J. 1997;11:428–42.
    1. Rubinstein E, Le Naour F, Lagaudriere-Gesbert C, Billard M, Conjeaud H, Boucheix C. CD9, CD63, CD81, and CD82 are components of a surface tetraspan network connected to HLA-DR and VLA integrins. Eur J Immunol. 1996;26:2657–65.
    1. Parthasarathy V, Martin F, Higginbottom A, Murray H, Moseley GW, Read RC, et al. Distinct roles for tetraspanins CD9, CD63 and CD81 in the formation of multinucleated giant cells. Immunology. 2009;127:237–48.
    1. Shoham T, Rajapaksa R, Boucheix C, Rubinstein E, Poe JC, Tedder TF, et al. The tetraspanin CD81 regulates the expression of CD19 during B cell development in a postendoplasmic reticulum compartment. J Immunol. 2003;171:4062–72.
    1. Escrevente C, Keller S, Altevogt P, Costa J. Interaction and uptake of exosomes by ovarian cancer cells. BMC Canc. 2011;11:108.
    1. Hwang I, Shen X, Sprent J. Direct stimulation of naive T cells by membrane vesicles from antigen-presenting cells: distinct roles for CD54 and B7 molecules. Proc Natl Acad Sci U S A. 2003;100:6670–5.
    1. Mack M, Kleinschmidt A, Bruhl H, Klier C, Nelson PJ, Cihak J, et al. Transfer of the chemokine receptor CCR5 between cells by membrane-derived microparticles: a mechanism for cellular human immunodeficiency virus 1 infection. Nat Med. 2000;6:769–75.
    1. Al-Nedawi K, Meehan B, Micallef J, Lhotak V, May L, Guha A, et al. Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles derived from tumour cells. Nat Cell Biol. 2008;10:619–24.
    1. Peinado H, Aleckovic M, Lavotshkin S, Matei I, Costa-Silva B, Moreno-Bueno G, et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med. 2012;18:883–91.
    1. Wiklander OP, Nordin JZ, O'Loughlin A, Gustafsson Y, Corso G, Mager I, et al. Extracellular vesicle in vivo biodistribution is determined by cell source, route of administration and targeting. J Extracell Vesicles. 2015;4:26316. doi: .

Source: PubMed

3
Subscribe