101
|
Bour A, Kruglik SG, Chabanon M, Rangamani P, Puff N, Bonneau S. Lipid Unsaturation Properties Govern the Sensitivity of Membranes to Photoinduced Oxidative Stress. Biophys J 2019; 116:910-920. [PMID: 30777304 DOI: 10.1016/j.bpj.2019.01.033] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 01/28/2019] [Accepted: 01/29/2019] [Indexed: 12/11/2022] Open
Abstract
Unsaturated lipid oxidation is a fundamental process involved in different aspects of cellular bioenergetics; dysregulation of lipid oxidation is often associated with cell aging and death. To study how lipid oxidation affects membrane biophysics, we used a chlorin photosensitizer to oxidize vesicles of various lipid compositions and degrees of unsaturation in a controlled manner. We observed different shape transitions that can be interpreted as an increase in the area of the targeted membrane followed by a decrease. These area modifications induced by the chemical modification of the membrane upon oxidation were followed in situ by Raman tweezers microspectroscopy. We found that the membrane area increase corresponds to the lipids' peroxidation and is initiated by the delocalization of the targeted double bonds in the tails of the lipids. The subsequent decrease of membrane area can be explained by the formation of cleaved secondary products. As a result of these area changes, we observe vesicle permeabilization after a time lag that is characterized in relation with the level of unsaturation. The evolution of photosensitized vesicle radius was measured and yields an estimation of the mechanical changes of the membrane over oxidation time. The membrane is both weakened and permeabilized by the oxidation. Interestingly, the effect of unsaturation level on the dynamics of vesicles undergoing photooxidation is not trivial and thus carefully discussed. Our findings shed light on the fundamental dynamic mechanisms underlying the oxidation of lipid membranes and highlight the role of unsaturations on their physical and chemical properties.
Collapse
Affiliation(s)
- Aurélien Bour
- Sorbonne Université, Faculté des Sciences et Ingénierie, CNRS, Laboratoire Jean Perrin, Paris, France
| | - Sergei G Kruglik
- Sorbonne Université, Faculté des Sciences et Ingénierie, CNRS, Laboratoire Jean Perrin, Paris, France
| | - Morgan Chabanon
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California
| | - Nicolas Puff
- Sorbonne Université, Faculté des Sciences et Ingénierie, UFR 925, Paris, France; University Paris Diderot, Sorbonne Paris Cité, CNRS, Laboratoire Matière et Systèmes Complexes, UMR 7057, Paris, France
| | - Stephanie Bonneau
- Sorbonne Université, Faculté des Sciences et Ingénierie, CNRS, Laboratoire Jean Perrin, Paris, France.
| |
Collapse
|
102
|
Bruce TF, Slonecki TJ, Wang L, Huang S, Powell RR, Marcus RK. Exosome isolation and purification via hydrophobic interaction chromatography using a polyester, capillary-channeled polymer fiber phase. Electrophoresis 2019; 40:571-581. [PMID: 30548636 PMCID: PMC6881775 DOI: 10.1002/elps.201800417] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 11/21/2018] [Accepted: 11/25/2018] [Indexed: 01/27/2023]
Abstract
Extracellular vesicles, including microvesicles and exosomes, are lipidic membrane-derived vesicles that are secreted by most cell types. Exosomes, one class of these vesicles that are 30-100 nm in diameter, hold a great deal of promise in disease diagnostics, as they display the same protein biomarkers as their originating cell. For exosomes to become useful in disease diagnostics, and as burgeoning drug delivery platforms, they must be isolated efficiently and effectively without compromising their structure. Most current exosome isolation methods have practical problems including being too time-consuming and labor intensive, destructive to the exosomes, or too costly for use in clinical settings. To this end, this study examines the use of poly(ethylene terephthalate) (PET) capillary-channeled polymer (C-CP) fibers in a hydrophobic interaction chromatography (HIC) protocol to isolate exosomes from diverse matrices of practical concern. Initial results demonstrate the ability to isolate extracellular vesicles enriched in exosomes with comparable yields and size distributions on a much faster time scale when compared to traditional isolation methods. As a demonstration of the potential analytical utility of the approach, extracellular vesicle recoveries from cell culture milieu and a mock urine matrix are presented. The potential for scalable separations covering submilliliter spin-down columns to the preparative scale is anticipated.
Collapse
Affiliation(s)
- Terri F. Bruce
- Department of Bioengineering, Life Sciences Facility, Clemson University, Clemson, SC, USA
| | - Tyler J. Slonecki
- Department of Bioengineering, Life Sciences Facility, Clemson University, Clemson, SC, USA
| | - Lei Wang
- Department of Chemistry, Biosystems Research Complex, Clemson University, Clemson, SC, USA
| | - Sisi Huang
- Department of Chemistry, Biosystems Research Complex, Clemson University, Clemson, SC, USA
| | - Rhonda R. Powell
- Clemson Light Imaging Facility, Clemson University, Clemson, SC, USA
| | - R. Kenneth Marcus
- Department of Chemistry, Biosystems Research Complex, Clemson University, Clemson, SC, USA
| |
Collapse
|
103
|
Roman M, Kamińska A, Drożdż A, Platt M, Kuźniewski M, Małecki MT, Kwiatek WM, Paluszkiewicz C, Stępień EŁ. Raman spectral signatures of urinary extracellular vesicles from diabetic patients and hyperglycemic endothelial cells as potential biomarkers in diabetes. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 17:137-149. [PMID: 30703535 DOI: 10.1016/j.nano.2019.01.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 12/18/2018] [Accepted: 01/15/2019] [Indexed: 01/21/2023]
Abstract
Raman spectroscopy was applied to the measurement of urinary and in vitro endothelium-derived extracellular vesicles (EVs) isolated by hydrostatic filtration dialysis (HFD) method. Raman spectra obtained for urinary EVs (UEVs) showed distinct differences in the fingerprint region. In contrast, average Raman spectra of endothelium-derived EVs samples were almost identical. Cluster Analysis of UEVs significantly discriminated diabetic samples from control, moreover endothelium-derived EVs revealed stronger similarity between long hyperglycemia and normoglycemia samples compared to short hyperglycemia. Results obtained from Partial Least Squares analysis corresponded well with integral intensities of selected bands. Our proof-of-concept approach demonstrates the potential for Raman spectroscopy to be used both for identification of EVs molecular signatures in urine samples from patients with type 2 diabetes mellitus and good glycemic control and unsatisfactory glycemic control as well as for in vitro hyperglycemic model. This noninvasive technique may be useful in identifying new biomarkers of diabetes and renal complications.
Collapse
Affiliation(s)
- Maciej Roman
- Institute of Nuclear Physics Polish Academy of Sciences, Krakow, Poland.
| | - Agnieszka Kamińska
- Department of Medical Physics, Marian Smoluchowski Institute of Physics Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Krakow, Poland.
| | - Anna Drożdż
- Department of Medical Physics, Marian Smoluchowski Institute of Physics Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Krakow, Poland
| | - Mark Platt
- Department of Chemistry, Loughborough University, Loughborough, United Kingdom
| | - Marek Kuźniewski
- Department of Nephrology, Jagiellonian University Medical College, Krakow, Poland
| | - Maciej T Małecki
- Department of Metabolic Diseases, Jagiellonian University Medical College, Krakow, Poland
| | | | | | - Ewa Ł Stępień
- Department of Medical Physics, Marian Smoluchowski Institute of Physics Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Krakow, Poland
| |
Collapse
|
104
|
Gualerzi A, Kooijmans SAA, Niada S, Picciolini S, Brini AT, Camussi G, Bedoni M. Raman spectroscopy as a quick tool to assess purity of extracellular vesicle preparations and predict their functionality. J Extracell Vesicles 2019; 8:1568780. [PMID: 30728924 PMCID: PMC6352930 DOI: 10.1080/20013078.2019.1568780] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 12/03/2018] [Accepted: 01/09/2019] [Indexed: 12/20/2022] Open
Abstract
Extracellular vesicles (EVs) from a variety of stem cell sources are believed to harbour regenerative capacity, which may be exploited for therapeutic purposes. Because of EV interaction with other soluble secreted factors, EV activity may depend on the employed purification method, which limits cross-study comparisons and therapeutic development. Raman spectroscopy (RS) is a quick and easy method to assess EV purity and composition, giving in-depth biochemical overview on EV preparation. Hereby, we show how this method can be used to characterise EVs isolated from human liver stem cells and bone marrow mesenchymal stem/stromal cells by means of conventional ultracentrifugation (UC) and size exclusion chromatography (SEC) protocols. The obtained EV preparations were demonstrated to be characterised by different degrees of purity and a specific Raman fingerprint that represents both the cell source and the isolation procedure used. Moreover, RS provided useful hints to explore the factors underlying the functional diversity of EV preparations from the same cell source, thus representing a valuable tool to assess EV quality prior to functional assays or therapeutic application.
Collapse
Affiliation(s)
| | | | | | - Silvia Picciolini
- IRCCS Fondazione Don Carlo Gnocchi, Milano, Italy.,Nanomedicine Center NANOMIB, University of Milano-Bicocca, Monza, Italy
| | - Anna Teresa Brini
- IRCCS Istituto Ortopedico Galeazzi, Milano, Italy.,Dipartimento di Scienze Biomediche, Chirurgiche ed Odontoiatriche, Università degli Studi di Milano, Milano, Italy
| | - Giovanni Camussi
- Department of Medical Sciences, Molecular Biotechnology Center, University of Turin, Torino, Italy
| | | |
Collapse
|
105
|
Kruglik SG, Royo F, Guigner JM, Palomo L, Seksek O, Turpin PY, Tatischeff I, Falcón-Pérez JM. Raman tweezers microspectroscopy of circa 100 nm extracellular vesicles. NANOSCALE 2019; 11:1661-1679. [PMID: 30620023 DOI: 10.1039/c8nr04677h] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
The technique of Raman tweezers microspectroscopy (RTM) for the global biomolecular content characterization of a single extracellular vesicle (EV) or a small number of EVs or other nanoscale bioparticles in an aqueous dispersion in the difficult-to-access size range of near 100 nm is described in detail. The particularities and potential of RTM are demonstrated using the examples of DOPC liposomes, exosomes from human urine and rat hepatocytes, and a mixed sample of the transfection reagent FuGENE in diluted DNA solution. The approach of biomolecular component analysis for the estimation of the main biomolecular contributions (proteins, lipids, nucleic acids, carotenoids, etc.) is proposed and discussed. Direct Raman evidence for strong intra-sample biomolecular heterogeneity of individual optically trapped EVs, due to variable contributions from nucleic acids and carotenoids in some preparations, is reported. On the basis of the results obtained, we are making an attempt to convince the scientific community that RTM is a promising method of single-EV research; to our knowledge, it is the only technique available at the moment that provides unique information about the global biomolecular composition of a single vesicle or a small number of vesicles, thus being capable of unravelling the high diversity of EV subpopulations, which is one of the most significant urgent challenges to overcome. Possible RTM applications include, among others, searching for DNA biomarkers, cancer diagnosis, and discrimination between different subpopulations of EVs, lipid bodies, protein aggregates and viruses.
Collapse
Affiliation(s)
- Sergei G Kruglik
- Laboratoire Jean Perrin, Sorbonne Université, CNRS UMR 8237, 4 place Jussieu, Paris, 75005, France.
| | | | | | | | | | | | | | | |
Collapse
|
106
|
Kabe Y, Sakamoto S, Hatakeyama M, Yamaguchi Y, Suematsu M, Itonaga M, Handa H. Application of high-performance magnetic nanobeads to biological sensing devices. Anal Bioanal Chem 2019; 411:1825-1837. [PMID: 30627798 PMCID: PMC6453870 DOI: 10.1007/s00216-018-1548-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/01/2018] [Accepted: 12/12/2018] [Indexed: 02/07/2023]
Abstract
Nanomaterials have extensive applications in the life sciences and in clinical diagnosis. We have developed magnetic nanoparticles with high dispersibility and extremely low nonspecific binding to biomolecules and have demonstrated their application in chemical biology (e.g., for the screening of drug receptor proteins). Recently, the excellent properties of nanobeads have made possible the development of novel rapid immunoassay systems and high-precision technologies for exosome detection. For immunoassays, we developed a technology to encapsulate a fluorescent substance in magnetic nanobeads. The fluorescent nanobeads allow the rapid detection of a specific antigen in solution or in tissue specimens. Exosomes, which are released into the blood, are expected to become markers for several diseases, including cancer, but techniques for measuring the absolute quantity of exosomes in biological fluids are lacking. By integrating magnetic nanobead technology with an optical disc system, we developed a novel method for precisely quantifying exosomes in human serum with high sensitivity and high linearity without requiring enrichment procedures. This review focuses on the properties of our magnetic nanobeads, the development of novel biosensors using these nanobeads, and their broad practical applications. Graphical abstract ![]()
Collapse
Affiliation(s)
- Yasuaki Kabe
- Department of Biochemistry, Keio University School of Medicine, 35 Shinnanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
- Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology, Tokyo, 200-0004, Japan.
| | - Satoshi Sakamoto
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa, 226-8501, Japan
| | - Mamoru Hatakeyama
- FG Beads Development Section, Biotronics Laboratory, Tamagawa Seiki Co. Ltd, Ohyasumi, Iida, Nagano, 395-8515, Japan
| | - Yuki Yamaguchi
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa, 226-8501, Japan
| | - Makoto Suematsu
- Department of Biochemistry, Keio University School of Medicine, 35 Shinnanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Makoto Itonaga
- Healthcare Business Division, JVCKENWOOD Corporation, 3-12 Moriya-cho, Kanagawa-ku, Yokohama, Kanagawa, 221-0022, Japan
| | - Hiroshi Handa
- Department of Nanoparticle Translational Research, Tokyo Medical University, 6-2-2 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan.
| |
Collapse
|
107
|
Théry C, Witwer KW, Aikawa E, Alcaraz MJ, Anderson JD, Andriantsitohaina R, Antoniou A, Arab T, Archer F, Atkin-Smith GK, Ayre DC, Bach JM, Bachurski D, Baharvand H, Balaj L, Baldacchino S, Bauer NN, Baxter AA, Bebawy M, Beckham C, Bedina Zavec A, Benmoussa A, Berardi AC, Bergese P, Bielska E, Blenkiron C, Bobis-Wozowicz S, Boilard E, Boireau W, Bongiovanni A, Borràs FE, Bosch S, Boulanger CM, Breakefield X, Breglio AM, Brennan MÁ, Brigstock DR, Brisson A, Broekman MLD, Bromberg JF, Bryl-Górecka P, Buch S, Buck AH, Burger D, Busatto S, Buschmann D, Bussolati B, Buzás EI, Byrd JB, Camussi G, Carter DRF, Caruso S, Chamley LW, Chang YT, Chen C, Chen S, Cheng L, Chin AR, Clayton A, Clerici SP, Cocks A, Cocucci E, Coffey RJ, Cordeiro-da-Silva A, Couch Y, Coumans FAW, Coyle B, Crescitelli R, Criado MF, D’Souza-Schorey C, Das S, Datta Chaudhuri A, de Candia P, De Santana EF, De Wever O, del Portillo HA, Demaret T, Deville S, Devitt A, Dhondt B, Di Vizio D, Dieterich LC, Dolo V, Dominguez Rubio AP, Dominici M, Dourado MR, Driedonks TAP, Duarte FV, Duncan HM, Eichenberger RM, Ekström K, EL Andaloussi S, Elie-Caille C, Erdbrügger U, Falcón-Pérez JM, Fatima F, Fish JE, Flores-Bellver M, Försönits A, Frelet-Barrand A, et alThéry C, Witwer KW, Aikawa E, Alcaraz MJ, Anderson JD, Andriantsitohaina R, Antoniou A, Arab T, Archer F, Atkin-Smith GK, Ayre DC, Bach JM, Bachurski D, Baharvand H, Balaj L, Baldacchino S, Bauer NN, Baxter AA, Bebawy M, Beckham C, Bedina Zavec A, Benmoussa A, Berardi AC, Bergese P, Bielska E, Blenkiron C, Bobis-Wozowicz S, Boilard E, Boireau W, Bongiovanni A, Borràs FE, Bosch S, Boulanger CM, Breakefield X, Breglio AM, Brennan MÁ, Brigstock DR, Brisson A, Broekman MLD, Bromberg JF, Bryl-Górecka P, Buch S, Buck AH, Burger D, Busatto S, Buschmann D, Bussolati B, Buzás EI, Byrd JB, Camussi G, Carter DRF, Caruso S, Chamley LW, Chang YT, Chen C, Chen S, Cheng L, Chin AR, Clayton A, Clerici SP, Cocks A, Cocucci E, Coffey RJ, Cordeiro-da-Silva A, Couch Y, Coumans FAW, Coyle B, Crescitelli R, Criado MF, D’Souza-Schorey C, Das S, Datta Chaudhuri A, de Candia P, De Santana EF, De Wever O, del Portillo HA, Demaret T, Deville S, Devitt A, Dhondt B, Di Vizio D, Dieterich LC, Dolo V, Dominguez Rubio AP, Dominici M, Dourado MR, Driedonks TAP, Duarte FV, Duncan HM, Eichenberger RM, Ekström K, EL Andaloussi S, Elie-Caille C, Erdbrügger U, Falcón-Pérez JM, Fatima F, Fish JE, Flores-Bellver M, Försönits A, Frelet-Barrand A, Fricke F, Fuhrmann G, Gabrielsson S, Gámez-Valero A, Gardiner C, Gärtner K, Gaudin R, Gho YS, Giebel B, Gilbert C, Gimona M, Giusti I, Goberdhan DCI, Görgens A, Gorski SM, Greening DW, Gross JC, Gualerzi A, Gupta GN, Gustafson D, Handberg A, Haraszti RA, Harrison P, Hegyesi H, Hendrix A, Hill AF, Hochberg FH, Hoffmann KF, Holder B, Holthofer H, Hosseinkhani B, Hu G, Huang Y, Huber V, Hunt S, Ibrahim AGE, Ikezu T, Inal JM, Isin M, Ivanova A, Jackson HK, Jacobsen S, Jay SM, Jayachandran M, Jenster G, Jiang L, Johnson SM, Jones JC, Jong A, Jovanovic-Talisman T, Jung S, Kalluri R, Kano SI, Kaur S, Kawamura Y, Keller ET, Khamari D, Khomyakova E, Khvorova A, Kierulf P, Kim KP, Kislinger T, Klingeborn M, Klinke DJ, Kornek M, Kosanović MM, Kovács ÁF, Krämer-Albers EM, Krasemann S, Krause M, Kurochkin IV, Kusuma GD, Kuypers S, Laitinen S, Langevin SM, Languino LR, Lannigan J, Lässer C, Laurent LC, Lavieu G, Lázaro-Ibáñez E, Le Lay S, Lee MS, Lee YXF, Lemos DS, Lenassi M, Leszczynska A, Li ITS, Liao K, Libregts SF, Ligeti E, Lim R, Lim SK, Linē A, Linnemannstöns K, Llorente A, Lombard CA, Lorenowicz MJ, Lörincz ÁM, Lötvall J, Lovett J, Lowry MC, Loyer X, Lu Q, Lukomska B, Lunavat TR, Maas SLN, Malhi H, Marcilla A, Mariani J, Mariscal J, Martens-Uzunova ES, Martin-Jaular L, Martinez MC, Martins VR, Mathieu M, Mathivanan S, Maugeri M, McGinnis LK, McVey MJ, Meckes DG, Meehan KL, Mertens I, Minciacchi VR, Möller A, Møller Jørgensen M, Morales-Kastresana A, Morhayim J, Mullier F, Muraca M, Musante L, Mussack V, Muth DC, Myburgh KH, Najrana T, Nawaz M, Nazarenko I, Nejsum P, Neri C, Neri T, Nieuwland R, Nimrichter L, Nolan JP, Nolte-’t Hoen ENM, Noren Hooten N, O’Driscoll L, O’Grady T, O’Loghlen A, Ochiya T, Olivier M, Ortiz A, Ortiz LA, Osteikoetxea X, Østergaard O, Ostrowski M, Park J, Pegtel DM, Peinado H, Perut F, Pfaffl MW, Phinney DG, Pieters BCH, Pink RC, Pisetsky DS, Pogge von Strandmann E, Polakovicova I, Poon IKH, Powell BH, Prada I, Pulliam L, Quesenberry P, Radeghieri A, Raffai RL, Raimondo S, Rak J, Ramirez MI, Raposo G, Rayyan MS, Regev-Rudzki N, Ricklefs FL, Robbins PD, Roberts DD, Rodrigues SC, Rohde E, Rome S, Rouschop KMA, Rughetti A, Russell AE, Saá P, Sahoo S, Salas-Huenuleo E, Sánchez C, Saugstad JA, Saul MJ, Schiffelers RM, Schneider R, Schøyen TH, Scott A, Shahaj E, Sharma S, Shatnyeva O, Shekari F, Shelke GV, Shetty AK, Shiba K, Siljander PRM, Silva AM, Skowronek A, Snyder OL, Soares RP, Sódar BW, Soekmadji C, Sotillo J, Stahl PD, Stoorvogel W, Stott SL, Strasser EF, Swift S, Tahara H, Tewari M, Timms K, Tiwari S, Tixeira R, Tkach M, Toh WS, Tomasini R, Torrecilhas AC, Tosar JP, Toxavidis V, Urbanelli L, Vader P, van Balkom BWM, van der Grein SG, Van Deun J, van Herwijnen MJC, Van Keuren-Jensen K, van Niel G, van Royen ME, van Wijnen AJ, Vasconcelos MH, Vechetti IJ, Veit TD, Vella LJ, Velot É, Verweij FJ, Vestad B, Viñas JL, Visnovitz T, Vukman KV, Wahlgren J, Watson DC, Wauben MHM, Weaver A, Webber JP, Weber V, Wehman AM, Weiss DJ, Welsh JA, Wendt S, Wheelock AM, Wiener Z, Witte L, Wolfram J, Xagorari A, Xander P, Xu J, Yan X, Yáñez-Mó M, Yin H, Yuana Y, Zappulli V, Zarubova J, Žėkas V, Zhang JY, Zhao Z, Zheng L, Zheutlin AR, Zickler AM, Zimmermann P, Zivkovic AM, Zocco D, Zuba-Surma EK. 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. [PMID: 30637094 PMCID: PMC6322352 DOI: 10.1080/20013078.2018.1535750] [Show More Authors] [Citation(s) in RCA: 7513] [Impact Index Per Article: 1073.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 09/25/2018] [Indexed: 11/04/2022] Open
Abstract
The last decade has seen a sharp increase in the number of scientific publications describing physiological and pathological functions of extracellular vesicles (EVs), a collective term covering various subtypes of cell-released, membranous structures, called exosomes, microvesicles, microparticles, ectosomes, oncosomes, apoptotic bodies, and many other names. However, specific issues arise when working with these entities, whose size and amount often make them difficult to obtain as relatively pure preparations, and to characterize properly. The International Society for Extracellular Vesicles (ISEV) proposed Minimal Information for Studies of Extracellular Vesicles ("MISEV") guidelines for the field in 2014. We now update these "MISEV2014" guidelines based on evolution of the collective knowledge in the last four years. An important point to consider is that ascribing a specific function to EVs in general, or to subtypes of EVs, requires reporting of specific information beyond mere description of function in a crude, potentially contaminated, and heterogeneous preparation. For example, claims that exosomes are endowed with exquisite and specific activities remain difficult to support experimentally, given our still limited knowledge of their specific molecular machineries of biogenesis and release, as compared with other biophysically similar EVs. The MISEV2018 guidelines include tables and outlines of suggested protocols and steps to follow to document specific EV-associated functional activities. Finally, a checklist is provided with summaries of key points.
Collapse
Affiliation(s)
- Clotilde Théry
- Institut Curie, INSERM U932, PSL Research University, Paris, France
| | - Kenneth W Witwer
- The Johns Hopkins University School of Medicine, Department of Molecular and Comparative Pathobiology, Baltimore, MD, USA
- The Johns Hopkins University School of Medicine, Department of Neurology, Baltimore, MD, USA
| | - Elena Aikawa
- Brigham and Women’s Hospital, Center for Interdisciplinary Cardiovascular Sciences, Boston, MA, USA
- Harvard Medical School, Cardiovascular Medicine, Boston, MA, USA
| | - Maria Jose Alcaraz
- Interuniversity Research Institute for Molecular Recognition and Technological Development (IDM), University of Valencia, Polytechnic University of Valencia, Valencia, Spain
| | | | | | - Anna Antoniou
- German Centre for Neurodegenerative Diseases (DZNE), Bonn, Germany
- University Hospital Bonn (UKB), Bonn, Germany
| | - Tanina Arab
- Université de Lille, INSERM, U-1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse - PRISM, Lille, France
| | - Fabienne Archer
- University of Lyon, INRA, EPHE, UMR754 Viral Infections and Comparative Pathology, Lyon, France
| | - Georgia K Atkin-Smith
- La Trobe University, La Trobe Institute for Molecular Science, Department of Biochemistry and Genetics, Bundoora, Australia
| | - D Craig Ayre
- Atlantic Cancer Research Institute, Moncton, Canada
- Mount Allison University, Department of Chemistry and Biochemistry, Sackville, Canada
| | - Jean-Marie Bach
- Université Bretagne Loire, Oniris, INRA, IECM, Nantes, France
| | - Daniel Bachurski
- University of Cologne, Department of Internal Medicine I, Cologne, Germany
| | - Hossein Baharvand
- Royan Institute for Stem Cell Biology and Technology, ACECR, Cell Science Research Center, Department of Stem Cells and Developmental Biology, Tehran, Iran
- University of Science and Culture, ACECR, Department of Developmental Biology, Tehran, Iran
| | - Leonora Balaj
- Massachusetts General Hospital, Department of Neurosurgery, Boston, MA, USA
| | | | - Natalie N Bauer
- University of South Alabama, Department of Pharmacology, Center for Lung Biology, Mobile, AL, USA
| | - Amy A Baxter
- La Trobe University, La Trobe Institute for Molecular Science, Department of Biochemistry and Genetics, Bundoora, Australia
| | - Mary Bebawy
- University of Technology Sydney, Discipline of Pharmacy, Graduate School of Health, Sydney, Australia
| | | | - Apolonija Bedina Zavec
- National Institute of Chemistry, Department of Molecular Biology and Nanobiotechnology, Ljubljana, Slovenia
| | - Abderrahim Benmoussa
- Université Laval, Centre de Recherche du CHU de Québec, Department of Infectious Diseases and Immunity, Quebec City, Canada
| | | | - Paolo Bergese
- CSGI - Research Center for Colloids and Nanoscience, Florence, Italy
- INSTM - National Interuniversity Consortium of Materials Science and Technology, Florence, Italy
- University of Brescia, Department of Molecular and Translational Medicine, Brescia, Italy
| | - Ewa Bielska
- University of Birmingham, Institute of Microbiology and Infection, Birmingham, UK
| | | | - Sylwia Bobis-Wozowicz
- Jagiellonian University, Faculty of Biochemistry, Biophysics and Biotechnology, Department of Cell Biology, Kraków, Poland
| | - Eric Boilard
- Université Laval, Centre de Recherche du CHU de Québec, Department of Infectious Diseases and Immunity, Quebec City, Canada
| | - Wilfrid Boireau
- FEMTO-ST Institute, UBFC, CNRS, ENSMM, UTBM, Besançon, France
| | - Antonella Bongiovanni
- Institute of Biomedicine and Molecular Immunology (IBIM), National Research Council (CNR) of Italy, Palermo, Italy
| | - Francesc E Borràs
- Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, REMAR-IVECAT Group, Badalona, Spain
- Germans Trias i Pujol University Hospital, Nephrology Service, Badalona, Spain
- Universitat Autònoma de Barcelona, Department of Cell Biology, Physiology & Immunology, Barcelona, Spain
| | - Steffi Bosch
- Université Bretagne Loire, Oniris, INRA, IECM, Nantes, France
| | - Chantal M Boulanger
- INSERM UMR-S 970, Paris Cardiovascular Research Center, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Xandra Breakefield
- Massachusetts General Hospital and Neuroscience Program, Harvard Medical School, Department of Neurology and Radiology, Boston, MA, USA
| | - Andrew M Breglio
- Icahn School of Medicine at Mount Sinai, New York City, NY, USA
- National Institutes of Health, National Institute on Deafness and Other Communication Disorders, Bethesda, MD, USA
| | - Meadhbh Á Brennan
- Harvard University, School of Engineering and Applied Sciences, Cambridge, MA, USA
- Massachusetts General Hospital, Harvard Medical School, Department of Neurology, Boston, MA, USA
- Université de Nantes, INSERM UMR 1238, Bone Sarcoma and Remodeling of Calcified Tissues, PhyOS, Nantes, France
| | - David R Brigstock
- Nationwide Children’s Hospital, Columbus, OH, USA
- The Ohio State University, Columbus, OH, USA
| | - Alain Brisson
- UMR-CBMN, CNRS-Université de Bordeaux, Bordeaux, France
| | - Marike LD Broekman
- Haaglanden Medical Center, Department of Neurosurgery, The Hague, The Netherlands
- Leiden University Medical Center, Department of Neurosurgery, Leiden, The Netherlands
- Massachusetts General Hospital, Department of Neurology, Boston, MA, USA
| | - Jacqueline F Bromberg
- Memorial Sloan Kettering Cancer Center, Department of Medicine, New York City, NY, USA
- Weill Cornell Medicine, Department of Medicine, New York City, NY, USA
| | | | - Shilpa Buch
- University of Nebraska Medical Center, Department of Pharmacology and Experimental Neuroscience, Omaha, NE, USA
| | - Amy H Buck
- University of Edinburgh, Institute of Immunology & Infection Research, Edinburgh, UK
| | - Dylan Burger
- Kidney Research Centre, Ottawa, Canada
- Ottawa Hospital Research Institute, Ottawa, Canada
- University of Ottawa, Ottawa, Canada
| | - Sara Busatto
- Mayo Clinic, Department of Transplantation, Jacksonville, FL, USA
- University of Brescia, Department of Molecular and Translational Medicine, Brescia, Italy
| | - Dominik Buschmann
- Technical University of Munich, TUM School of Life Sciences Weihenstephan, Division of Animal Physiology and Immunology, Freising, Germany
| | - Benedetta Bussolati
- University of Torino, Department of Molecular Biotechnology and Health Sciences, Torino, Italy
| | - Edit I Buzás
- MTA-SE Immuno-Proteogenomics Research Groups, Budapest, Hungary
- Semmelweis University, Department of Genetics, Cell- and Immunobiology, Budapest, Hungary
| | - James Bryan Byrd
- University of Michigan, Department of Medicine, Ann Arbor, MI, USA
| | - Giovanni Camussi
- University of Torino, Department of Medical Sciences, Torino, Italy
| | - David RF Carter
- Oxford Brookes University, Department of Biological and Medical Sciences, Oxford, UK
| | - Sarah Caruso
- La Trobe University, La Trobe Institute for Molecular Science, Department of Biochemistry and Genetics, Bundoora, Australia
| | - Lawrence W Chamley
- University of Auckland, Department of Obstetrics and Gynaecology, Auckland, New Zealand
| | - Yu-Ting Chang
- National Taiwan University Hospital, Department of Internal Medicine, Taipei, Taiwan
| | - Chihchen Chen
- National Tsing Hua University, Department of Power Mechanical Engineering, Hsinchu, Taiwan
- National Tsing Hua University, Institute of Nanoengineering and Microsystems, Hsinchu, Taiwan
| | - Shuai Chen
- Leibniz Institute for Farm Animal Biology (FBN), Institute of Reproductive Biology, Dummerstorf, Germany
| | - Lesley Cheng
- La Trobe University, La Trobe Institute for Molecular Science, Department of Biochemistry and Genetics, Bundoora, Australia
| | | | - Aled Clayton
- Cardiff University, School of Medicine, Cardiff, UK
| | | | - Alex Cocks
- Cardiff University, School of Medicine, Cardiff, UK
| | - Emanuele Cocucci
- The Ohio State University, College of Pharmacy, Division of Pharmaceutics and Pharmaceutical Chemistry, Columbus, OH, USA
- The Ohio State University, Comprehensive Cancer Center, Columbus, OH, USA
| | - Robert J Coffey
- Vanderbilt University Medical Center, Epithelial Biology Center, Department of Medicine, Nashville, TN, USA
| | | | - Yvonne Couch
- University of Oxford, Radcliffe Department of Medicine, Acute Stroke Programme - Investigative Medicine, Oxford, UK
| | - Frank AW Coumans
- Academic Medical Centre of the University of Amsterdam, Department of Clinical Chemistry and Vesicle Observation Centre, Amsterdam, The Netherlands
| | - Beth Coyle
- The University of Nottingham, School of Medicine, Children’s Brain Tumour Research Centre, Nottingham, UK
| | - Rossella Crescitelli
- University of Gothenburg, Institute of Medicine at Sahlgrenska Academy, Krefting Research Centre, Gothenburg, Sweden
| | | | | | - Saumya Das
- Massachusetts General Hospital, Boston, MA, USA
| | - Amrita Datta Chaudhuri
- The Johns Hopkins University School of Medicine, Department of Neurology, Baltimore, MD, USA
| | | | - Eliezer F De Santana
- The Sociedade Beneficente Israelita Brasileira Albert Einstein, São Paulo, Brazil
| | - Olivier De Wever
- Cancer Research Institute Ghent, Ghent, Belgium
- Ghent University, Department of Radiation Oncology and Experimental Cancer Research, Laboratory of Experimental Cancer Research, Ghent, Belgium
| | - Hernando A del Portillo
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Institut d’Investigació Germans Trias i Pujol (IGTP), PVREX group, Badalona, Spain
- ISGlobal, Hospital Clínic - Universitat de Barcelona, PVREX Group, Barcelona, Spain
| | - Tanguy Demaret
- Université Catholique de Louvain, Institut de Recherche Expérimentale et Clinique (IREC), Laboratory of Pediatric Hepatology and Cell Therapy, Brussels, Belgium
| | - Sarah Deville
- Universiteit Hasselt, Diepenbeek, Belgium
- Vlaamse Instelling voor Technologisch Onderzoek (VITO), Mol, Belgium
| | - Andrew Devitt
- Aston University, School of Life & Health Sciences, Birmingham, UK
| | - Bert Dhondt
- Cancer Research Institute Ghent, Ghent, Belgium
- Ghent University Hospital, Department of Urology, Ghent, Belgium
- Ghent University, Department of Radiation Oncology and Experimental Cancer Research, Laboratory of Experimental Cancer Research, Ghent, Belgium
| | | | | | - Vincenza Dolo
- University of L’Aquila, Department of Life, Health and Environmental Sciences, L’Aquila, Italy
| | - Ana Paula Dominguez Rubio
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Buenos Aires, Argentina
| | - Massimo Dominici
- TPM of Mirandola, Mirandola, Italy
- University of Modena and Reggio Emilia, Division of Oncology, Modena, Italy
| | - Mauricio R Dourado
- University of Campinas, Piracicaba Dental School, Department of Oral Diagnosis, Piracicaba, Brazil
- University of Oulu, Faculty of Medicine, Cancer and Translational Medicine Research Unit, Oulu, Finland
| | - Tom AP Driedonks
- Utrecht University, Faculty of Veterinary Medicine, Department of Biochemistry and Cell Biology, Utrecht, The Netherlands
| | | | - Heather M Duncan
- McGill University, Division of Experimental Medicine, Montreal, Canada
- McGill University, The Research Institute of the McGill University Health Centre, Child Health and Human Development Program, Montreal, Canada
| | - Ramon M Eichenberger
- James Cook University, Australian Institute of Tropical Health and Medicine, Centre for Biodiscovery and Molecular Development of Therapeutics, Cairns, Australia
| | - Karin Ekström
- University of Gothenburg, Institute of Clinical Sciences at Sahlgrenska Academy, Department of Biomaterials, Gothenburg, Sweden
| | - Samir EL Andaloussi
- Evox Therapeutics Limited, Oxford, UK
- Karolinska Institute, Stockholm, Sweden
| | | | - Uta Erdbrügger
- University of Virginia Health System, Department of Medicine, Division of Nephrology, Charlottesville, VA, USA
| | - Juan M Falcón-Pérez
- CIC bioGUNE, CIBERehd, Exosomes Laboratory & Metabolomics Platform, Derio, Spain
- IKERBASQUE Research Science Foundation, Bilbao, Spain
| | - Farah Fatima
- University of São Paulo, Ribeirão Preto Medical School, Department of Pathology and Forensic Medicine, Ribeirão Preto, Brazil
| | - Jason E Fish
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
- University of Toronto, Department of Laboratory Medicine and Pathobiology, Toronto, Canada
| | - Miguel Flores-Bellver
- University of Colorado, School of Medicine, Department of Ophthalmology, Cell Sight-Ocular Stem Cell and Regeneration Program, Aurora, CO, USA
| | - András Försönits
- Semmelweis University, Department of Genetics, Cell- and Immunobiology, Budapest, Hungary
| | | | - Fabia Fricke
- German Cancer Research Center (DKFZ), Clinical Cooperation Unit Applied Tumor Biology, Heidelberg, Germany
- University Hospital Heidelberg, Institute of Pathology, Applied Tumor Biology, Heidelberg, Germany
| | - Gregor Fuhrmann
- Helmholtz-Centre for Infection Research, Braunschweig, Germany
- Helmholtz-Institute for Pharmaceutical Research Saarland, Saarbrücken, Germany
- Saarland University, Saarbrücken, Germany
| | - Susanne Gabrielsson
- Karolinska Institute, Department of Medicine Solna, Division for Immunology and Allergy, Stockholm, Sweden
| | - Ana Gámez-Valero
- Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, REMAR-IVECAT Group, Badalona, Spain
- Universitat Autònoma de Barcelona, Hospital Universitari and Health Sciences Research Institute Germans Trias i Pujol, Department of Pathology, Barcelona, Spain
| | | | - Kathrin Gärtner
- Helmholtz Center Munich German Research Center for Environmental Health, Research Unit Gene Vectors, Munich, Germany
| | - Raphael Gaudin
- INSERM U1110, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
| | - Yong Song Gho
- POSTECH (Pohang University of Science and Technology), Department of Life Sciences, Pohang, South Korea
| | - Bernd Giebel
- University Hospital Essen, University Duisburg-Essen, Institute for Transfusion Medicine, Essen, Germany
| | - Caroline Gilbert
- Université Laval, Centre de Recherche du CHU de Québec, Department of Infectious Diseases and Immunity, Quebec City, Canada
| | - Mario Gimona
- Paracelsus Medical University, GMP Unit, Salzburg, Austria
| | - Ilaria Giusti
- University of L’Aquila, Department of Life, Health and Environmental Sciences, L’Aquila, Italy
| | - Deborah CI Goberdhan
- University of Oxford, Department of Physiology, Anatomy and Genetics, Oxford, UK
| | - André Görgens
- Evox Therapeutics Limited, Oxford, UK
- Karolinska Institute, Clinical Research Center, Department of Laboratory Medicine, Stockholm, Sweden
- University Hospital Essen, University Duisburg-Essen, Institute for Transfusion Medicine, Essen, Germany
| | - Sharon M Gorski
- BC Cancer, Canada’s Michael Smith Genome Sciences Centre, Vancouver, Canada
- Simon Fraser University, Department of Molecular Biology and Biochemistry, Burnaby, Canada
| | - David W Greening
- La Trobe University, La Trobe Institute for Molecular Science, Department of Biochemistry and Genetics, Bundoora, Australia
| | - Julia Christina Gross
- University Medical Center Göttingen, Developmental Biochemistry, Göttingen, Germany
- University Medical Center Göttingen, Hematology and Oncology, Göttingen, Germany
| | - Alice Gualerzi
- IRCCS Fondazione Don Carlo Gnocchi, Laboratory of Nanomedicine and Clinical Biophotonics (LABION), Milan, Italy
| | - Gopal N Gupta
- Loyola University Chicago, Department of Urology, Maywood, IL, USA
| | - Dakota Gustafson
- University of Toronto, Department of Laboratory Medicine and Pathobiology, Toronto, Canada
| | - Aase Handberg
- Aalborg University Hospital, Department of Clinical Biochemistry, Aalborg, Denmark
- Aalborg University, Clinical Institute, Aalborg, Denmark
| | - Reka A Haraszti
- University of Massachusetts Medical School, RNA Therapeutics Institute, Worcester, MA, USA
| | | | - Hargita Hegyesi
- Semmelweis University, Department of Genetics, Cell- and Immunobiology, Budapest, Hungary
| | - An Hendrix
- Cancer Research Institute Ghent, Ghent, Belgium
- Ghent University, Department of Radiation Oncology and Experimental Cancer Research, Laboratory of Experimental Cancer Research, Ghent, Belgium
| | - Andrew F Hill
- La Trobe University, La Trobe Institute for Molecular Science, Department of Biochemistry and Genetics, Bundoora, Australia
| | - Fred H Hochberg
- Scintillon Institute, La Jolla, CA, USA
- University of California, San Diego, Department of Neurosurgery, La Jolla, CA, USA
| | - Karl F Hoffmann
- Aberystwyth University, Institute of Biological, Environmental and Rural Sciences (IBERS), Aberystwyth, United Kingdom
| | - Beth Holder
- Imperial College London, London, UK
- MRC The Gambia, Fajara, The Gambia
| | | | - Baharak Hosseinkhani
- Hasselt University, Biomedical Research Institute (BIOMED), Department of Medicine and Life Sciences, Hasselt, Belgium
| | - Guoku Hu
- University of Nebraska Medical Center, Department of Pharmacology and Experimental Neuroscience, Omaha, NE, USA
| | - Yiyao Huang
- Nanfang Hospital, Southern Medical University, Department of Clinical Laboratory Medicine, Guangzhou, China
- The Johns Hopkins University School of Medicine, Department of Molecular and Comparative Pathobiology, Baltimore, MD, USA
| | - Veronica Huber
- Fondazione IRCCS Istituto Nazionale dei Tumori, Unit of Immunotherapy of Human Tumors, Milan, Italy
| | | | | | - Tsuneya Ikezu
- Boston University School of Medicine, Boston, MA, USA
| | - Jameel M Inal
- University of Hertfordshire, School of Life and Medical Sciences, Biosciences Research Group, Hatfield, UK
| | - Mustafa Isin
- Istanbul University Oncology Institute, Basic Oncology Department, Istanbul, Turkey
| | - Alena Ivanova
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, Heidelberg, Germany
| | - Hannah K Jackson
- The University of Nottingham, School of Medicine, Children’s Brain Tumour Research Centre, Nottingham, UK
| | - Soren Jacobsen
- Copenhagen Lupus and Vasculitis Clinic, Section 4242 - Rigshospitalet, Copenhagen, Denmark
- University of Copenhagen, Institute of Clinical Medicine, Copenhagen, Denmark
| | - Steven M Jay
- University of Maryland, Fischell Department of Bioengineering, College Park, MD, USA
| | - Muthuvel Jayachandran
- Mayo Clinic, College of Medicine, Department of Physiology and Biomedical Engineering, Rochester, MN, USA
| | | | - Lanzhou Jiang
- La Trobe University, La Trobe Institute for Molecular Science, Department of Biochemistry and Genetics, Bundoora, Australia
| | - Suzanne M Johnson
- University of Manchester, Division of Cancer Sciences, Manchester Cancer Research Centre, Manchester, UK
| | - Jennifer C Jones
- National Institutes of Health, National Cancer Institute, Center for Cancer Research, Bethesda, MD, USA
| | - Ambrose Jong
- Children’s Hospital of Los Angeles, Los Angeles, CA, USA
- University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - Tijana Jovanovic-Talisman
- City of Hope Comprehensive Cancer Center, Beckman Research Institute, Department of Molecular Medicine, Duarte, CA, USA
| | - Stephanie Jung
- German Research Center for Environmental Health, Institute for Virology, Munich, Germany
| | - Raghu Kalluri
- University of Texas MD Anderson Cancer Center, Department of Cancer Biology, Metastasis Research Center, Houston, TX, USA
| | - Shin-ichi Kano
- The Johns Hopkins University School of Medicine, Department of Psychiatry and Behavioral Sciences, Baltimore, MD, USA
| | - Sukhbir Kaur
- National Institutes of Health, National Cancer Institute, Center for Cancer Research, Laboratory of Pathology, Bethesda, MD, USA
| | - Yumi Kawamura
- National Cancer Center Research Institute, Tokyo, Japan
- University of Tsukuba, Tsukuba, Japan
| | - Evan T Keller
- University of Michigan, Biointerfaces Institute, Ann Arbor, MI, USA
- University of Michigan, Department of Urology, Ann Arbor, MI, USA
| | - Delaram Khamari
- Semmelweis University, Department of Genetics, Cell- and Immunobiology, Budapest, Hungary
| | - Elena Khomyakova
- École normale supérieure, Paris, France
- Federal Research and Clinical Center of Physical-Chemical Medicine, Moscow, Russia
| | - Anastasia Khvorova
- University of Massachusetts Medical School, RNA Therapeutics Institute, Worcester, MA, USA
| | - Peter Kierulf
- Oslo University Hospital, Department of Medical Biochemistry, Blood Cell Research Group, Oslo, Norway
| | - Kwang Pyo Kim
- Kyung Hee University, Department of Applied Chemistry, Yongin, Korea
| | - Thomas Kislinger
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- University of Toronto, Department of Medical Biophysics, Toronto, Canada
| | | | - David J Klinke
- West Virginia University, Department of Chemical and Biomedical Engineering and WVU Cancer Institute, Morgantown, WV, USA
- West Virginia University, Department of Microbiology Immunology and Cell Biology, Morgantown, WV, USA
| | - Miroslaw Kornek
- German Armed Forces Central Hospital, Department of General, Visceral and Thoracic Surgery, Koblenz, Germany
- Saarland University Medical Center, Department of Medicine II, Homburg, Germany
| | - Maja M Kosanović
- University of Belgrade, Institute for the Application of Nuclear Energy, INEP, Belgrade, Serbia
| | - Árpád Ferenc Kovács
- Semmelweis University, Department of Genetics, Cell- and Immunobiology, Budapest, Hungary
| | | | - Susanne Krasemann
- University Medical Center Hamburg-Eppendorf, Institute of Neuropathology, Hamburg, Germany
| | - Mirja Krause
- Hudson Institute of Medical Research, Melbourne, Australia
| | | | - Gina D Kusuma
- Hudson Institute of Medical Research, Melbourne, Australia
- Monash University, Melbourne, Australia
| | - Sören Kuypers
- Hasselt University, Biomedical Research Institute (BIOMED), Hasselt, Belgium
| | - Saara Laitinen
- Finnish Red Cross Blood Service, Research and Development, Helsinki, Finland
| | - Scott M Langevin
- Cincinnati Cancer Center, Cincinnati, OH, USA
- University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Lucia R Languino
- Thomas Jefferson University, Sidney Kimmel Medical School, Department of Cancer Biology, Philadelphia, PA, USA
| | - Joanne Lannigan
- University of Virginia, Flow Cytometry Core, School of Medicine, Charlottesville, VA, USA
| | - Cecilia Lässer
- University of Gothenburg, Institute of Medicine at Sahlgrenska Academy, Krefting Research Centre, Gothenburg, Sweden
| | - Louise C Laurent
- University of California, San Diego, Department of Obstetrics, Gynecology, and Reproductive Sciences, La Jolla, CA, USA
| | - Gregory Lavieu
- Institut Curie, INSERM U932, PSL Research University, Paris, France
| | | | - Soazig Le Lay
- INSERM U1063, Université d’Angers, CHU d’Angers, Angers, France
| | - Myung-Shin Lee
- Eulji University, School of Medicine, Daejeon, South Korea
| | | | - Debora S Lemos
- Federal University of Paraná, Department of Genetics, Human Molecular Genetics Laboratory, Curitiba, Brazil
| | - Metka Lenassi
- University of Ljubljana, Faculty of Medicine, Institute of Biochemistry, Ljubljana, Slovenia
| | | | - Isaac TS Li
- University of British Columbia Okanagan, Kelowna, Canada
| | - Ke Liao
- University of Nebraska Medical Center, Department of Pharmacology and Experimental Neuroscience, Omaha, NE, USA
| | - Sten F Libregts
- University of Cambridge School of Clinical Medicine, Addenbrooke’s Hospital, Department of Medicine, Cambridge NIHR BRC Cell Phenotyping Hub, Cambridge, UK
| | - Erzsebet Ligeti
- Semmelweis University, Department of Physiology, Budapest, Hungary
| | - Rebecca Lim
- Hudson Institute of Medical Research, Melbourne, Australia
- Monash University, Melbourne, Australia
| | - Sai Kiang Lim
- Institute of Medical Biology (IMB), Agency for Science and Technology (A*STAR), Singapore
| | - Aija Linē
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Karen Linnemannstöns
- University Medical Center Göttingen, Developmental Biochemistry, Göttingen, Germany
- University Medical Center Göttingen, Hematology and Oncology, Göttingen, Germany
| | - Alicia Llorente
- Oslo University Hospital-The Norwegian Radium Hospital, Institute for Cancer Research, Department of Molecular Cell Biology, Oslo, Norway
| | - Catherine A Lombard
- Université Catholique de Louvain, Institut de Recherche Expérimentale et Clinique (IREC), Laboratory of Pediatric Hepatology and Cell Therapy, Brussels, Belgium
| | - Magdalena J Lorenowicz
- Utrecht University, University Medical Center Utrecht, Center for Molecular Medicine & Regenerative Medicine Center, Utrecht, The Netherlands
| | - Ákos M Lörincz
- Semmelweis University, Department of Physiology, Budapest, Hungary
| | - Jan Lötvall
- University of Gothenburg, Institute of Medicine at Sahlgrenska Academy, Krefting Research Centre, Gothenburg, Sweden
| | - Jason Lovett
- Stellenbosch University, Department of Physiological Sciences, Stellenbosch, South Africa
| | - Michelle C Lowry
- Trinity College Dublin, School of Pharmacy and Pharmaceutical Sciences, Panoz Institute & Trinity Biomedical Sciences Institute, Dublin, Ireland
| | - Xavier Loyer
- INSERM UMR-S 970, Paris Cardiovascular Research Center, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Quan Lu
- Harvard University, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Barbara Lukomska
- Mossakowski Medical Research Centre, NeuroRepair Department, Warsaw, Poland
| | - Taral R Lunavat
- K.G. Jebsen Brain Tumor Research Centre, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Sybren LN Maas
- Utrecht University, University Medical Center Utrecht, Department of Neurosurgery, Brain Center Rudolf Magnus, Institute of Neurosciences, Utrecht, The Netherlands
- Utrecht University, University Medical Center Utrecht, Department of Pathology, Utrecht, The Netherlands
| | | | - Antonio Marcilla
- Universitat de València, Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Àrea de Parasitologia, Valencia, Spain
- Universitat de València, Health Research Institute La Fe, Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics, Valencia, Spain
| | - Jacopo Mariani
- Università degli Studi di Milano, Department of Clinical Sciences and Community Health, EPIGET LAB, Milan, Italy
| | | | | | | | | | | | - Mathilde Mathieu
- Institut Curie, INSERM U932, PSL Research University, Paris, France
| | - Suresh Mathivanan
- La Trobe University, La Trobe Institute for Molecular Science, Department of Biochemistry and Genetics, Bundoora, Australia
| | - Marco Maugeri
- University of Gothenburg, Sahlgrenska Academy, Department of Rheumatology and Inflammation Research, Gothenburg, Sweden
| | | | - Mark J McVey
- SickKids Hospital, Department of Anesthesia and Pain Medicine, Toronto, Canada
- University of Toronto, Department of Anesthesia, Toronto, Canada
| | - David G Meckes
- Florida State University College of Medicine, Department of Biomedical Sciences, Tallahassee, FL, USA
| | - Katie L Meehan
- The School of Biomedical Sciences, University of Western Australia, Perth, Australia
| | - Inge Mertens
- University of Antwerp, Centre for Proteomics, Antwerp, Belgium
- Vlaamse Instelling voor Technologisch Onderzoek (VITO), Mol, Belgium
| | - Valentina R Minciacchi
- Georg-Speyer-Haus Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - Andreas Möller
- QIMR Berghofer Medical Research Institute, Herston, Australia
| | - Malene Møller Jørgensen
- Aalborg University Hospital, Department of Clinical Immunology, Aalborg, Denmark
- EVSEARCH.DK, Denmark
| | - Aizea Morales-Kastresana
- National Institutes of Health, National Cancer Institute, Center for Cancer Research, Bethesda, MD, USA
| | | | - François Mullier
- Namur Thrombosis and Hemostasis Center (NTHC), NARILIS, Namur, Belgium
- Université Catholique de Louvain, CHU UCL Namur, Hematology-Hemostasis Laboratory, Yvoir, Belgium
| | - Maurizio Muraca
- University of Padova, Department of Women’s and Children’s Health, Padova, Italy
| | - Luca Musante
- University of Virginia Health System, Department of Medicine, Division of Nephrology, Charlottesville, VA, USA
| | - Veronika Mussack
- Technical University of Munich, TUM School of Life Sciences Weihenstephan, Division of Animal Physiology and Immunology, Freising, Germany
| | - Dillon C Muth
- The Johns Hopkins University School of Medicine, Department of Molecular and Comparative Pathobiology, Baltimore, MD, USA
| | - Kathryn H Myburgh
- Stellenbosch University, Department of Physiological Sciences, Stellenbosch, South Africa
| | - Tanbir Najrana
- Brown University, Women and Infants Hospital, Providence, RI, USA
| | - Muhammad Nawaz
- University of Gothenburg, Sahlgrenska Academy, Department of Rheumatology and Inflammation Research, Gothenburg, Sweden
| | - Irina Nazarenko
- German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Institute for Infection Prevention and Hospital Epidemiology, Freiburg, Germany
| | - Peter Nejsum
- Aarhus University, Department of Clinical Medicine, Aarhus, Denmark
| | - Christian Neri
- Sorbonne Université, Centre National de la Recherche Scientifique, Research Unit Biology of Adaptation and Aging (B2A), Team Compensation in Neurodegenerative and Aging (Brain-C), Paris, France
| | - Tommaso Neri
- University of Pisa, Centro Dipartimentale di Biologia Cellulare Cardio-Respiratoria, Pisa, Italy
| | - Rienk Nieuwland
- Academic Medical Centre of the University of Amsterdam, Department of Clinical Chemistry and Vesicle Observation Centre, Amsterdam, The Netherlands
| | - Leonardo Nimrichter
- Universidade Federal do Rio de Janeiro, Instituto de Microbiologia, Rio de Janeiro, Brazil
| | | | - Esther NM Nolte-’t Hoen
- Utrecht University, Faculty of Veterinary Medicine, Department of Biochemistry and Cell Biology, Utrecht, The Netherlands
| | - Nicole Noren Hooten
- National Institutes of Health, National Institute on Aging, Baltimore, MD, USA
| | - Lorraine O’Driscoll
- Trinity College Dublin, School of Pharmacy and Pharmaceutical Sciences, Panoz Institute & Trinity Biomedical Sciences Institute, Dublin, Ireland
| | - Tina O’Grady
- University of Liège, GIGA-R(MBD), PSI Laboratory, Liège, Belgium
| | - Ana O’Loghlen
- Queen Mary University of London, Blizard Institute, Epigenetics & Cellular Senescence Group, London, UK
| | - Takahiro Ochiya
- National Cancer Center Research Institute, Division of Molecular and Cellular Medicine, Tokyo, Japan
| | - Martin Olivier
- McGill University, The Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Alberto Ortiz
- IIS-Fundacion Jimenez Diaz-UAM, Department of Nephrology and Hypertension, Madrid, Spain
- Spanish Kidney Research Network, REDINREN, Madrid, Spain
- Universidad Autónoma de Madrid, School of Medicine, Department of Medicine, Madrid, Spain
| | - Luis A Ortiz
- Graduate School of Public Health at the University of Pittsburgh, Division of Occupational and Environmental Medicine, Pittsburgh, PA, USA
| | | | - Ole Østergaard
- Statens Serum Institut, Department of Autoimmunology and Biomarkers, Copenhagen, Denmark
- University of Copenhagen, Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Protein Research, Copenhagen, Denmark
| | - Matias Ostrowski
- University of Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Buenos Aires, Argentina
| | - Jaesung Park
- POSTECH (Pohang University of Science and Technology), Department of Life Sciences, Pohang, South Korea
| | - D. Michiel Pegtel
- Amsterdam University Medical Centers, Department of Pathology, Amsterdam, The Netherlands
| | - Hector Peinado
- Spanish National Cancer Research Center (CNIO), Molecular Oncology Programme, Microenvironment and Metastasis Laboratory, Madrid, Spain
| | - Francesca Perut
- IRCCS - Istituto Ortopedico Rizzoli, Laboratory for Orthopaedic Pathophysiology and Regenerative Medicine, Bologna, Italy
| | - Michael W Pfaffl
- Technical University of Munich, TUM School of Life Sciences Weihenstephan, Division of Animal Physiology and Immunology, Freising, Germany
| | - Donald G Phinney
- The Scripps Research Institute-Scripps Florida, Department of Molecular Medicine, Jupiter, FL, USA
| | - Bartijn CH Pieters
- Radboud University Medical Center, Department of Rheumatology, Nijmegen, The Netherlands
| | - Ryan C Pink
- Oxford Brookes University, Department of Biological and Medical Sciences, Oxford, UK
| | - David S Pisetsky
- Duke University Medical Center, Departments of Medicine and Immunology, Durham, NC, USA
- Durham VAMC, Medical Research Service, Durham, NC, USA
| | | | - Iva Polakovicova
- Pontificia Universidad Católica de Chile, Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
- Pontificia Universidad Católica de Chile, Faculty of Medicine, Department of Hematology-Oncology, Santiago, Chile
| | - Ivan KH Poon
- La Trobe University, La Trobe Institute for Molecular Science, Department of Biochemistry and Genetics, Bundoora, Australia
| | - Bonita H Powell
- The Johns Hopkins University School of Medicine, Department of Molecular and Comparative Pathobiology, Baltimore, MD, USA
| | | | - Lynn Pulliam
- University of California, San Francisco, CA, USA
- Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Peter Quesenberry
- The Warren Alpert Medical School of Brown University, Department of Medicine, Providence, RI, USA
| | - Annalisa Radeghieri
- CSGI - Research Center for Colloids and Nanoscience, Florence, Italy
- University of Brescia, Department of Molecular and Translational Medicine, Brescia, Italy
| | - Robert L Raffai
- Department of Veterans Affairs, San Francisco, CA, USA
- University of California, San Francisco, CA, USA
| | - Stefania Raimondo
- University of Palermo, Department of Biopathology and Medical Biotechnologies, Palermo, Italy
| | - Janusz Rak
- McGill University, Montreal, Canada
- McGill University, The Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Marcel I Ramirez
- Instituto Oswaldo Cruz, Rio de Janeiro, Brazil
- Universidade Federal de Paraná, Paraná, Brazil
| | - Graça Raposo
- Institut Curie, CNRS UMR144, PSL Research University, Paris, France
| | - Morsi S Rayyan
- University of Michigan Medical School, Ann Arbor, MI, USA
| | - Neta Regev-Rudzki
- Weizmann Institute of Science, Department of Biomolecular Sciences, Rehovot, Israel
| | - Franz L Ricklefs
- University Medical Center Hamburg-Eppendorf, Department of Neurosurgery, Hamburg, Germany
| | - Paul D Robbins
- University of Minnesota Medical School, Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, Minneapolis, MN, USA
| | - David D Roberts
- National Institutes of Health, National Cancer Institute, Center for Cancer Research, Laboratory of Pathology, Bethesda, MD, USA
| | | | - Eva Rohde
- Paracelsus Medical University, Department of Transfusion Medicine, Salzburg, Austria
- Paracelsus Medical University, GMP Unit, Salzburg, Austria
- Spinal Cord Injury & Tissue Regeneration Center Salzburg (SCI-TReCS), Salzburg, Austria
| | - Sophie Rome
- University of Lyon, Lyon-Sud Faculty of Medicine, CarMeN Laboratory (UMR INSERM 1060-INRA 1397), Pierre-Bénite, France
| | - Kasper MA Rouschop
- Maastricht University, GROW, School for Oncology and Developmental Biology, Maastricht Radiation Oncology (MaastRO) Lab, Maastricht, The Netherlands
| | - Aurelia Rughetti
- Sapienza University of Rome, Department of Experimental Medicine, Rome, Italy
| | | | - Paula Saá
- American Red Cross, Scientific Affairs, Gaithersburg, MD, USA
| | - Susmita Sahoo
- Icahn School of Medicine at Mount Sinai, Department of Medicine, Cardiology, New York City, NY, USA
| | - Edison Salas-Huenuleo
- Advanced Center for Chronic Diseases, Santiago, Chile
- University of Chile, Faculty of Chemical and Pharmaceutical Science, Laboratory of Nanobiotechnology and Nanotoxicology, Santiago, Chile
| | - Catherine Sánchez
- Clínica las Condes, Extracellular Vesicles in Personalized Medicine Group, Santiago, Chile
| | - Julie A Saugstad
- Oregon Health & Science University, Department of Anesthesiology & Perioperative Medicine, Portland, OR, USA
| | - Meike J Saul
- Technische Universität Darmstadt, Department of Biology, Darmstadt, Germany
| | - Raymond M Schiffelers
- University Medical Center Utrecht, Laboratory for Clinical Chemistry & Hematology, Utrecht, The Netherlands
| | - Raphael Schneider
- University of Toronto, Department of Laboratory Medicine and Pathobiology, Toronto, Canada
- University of Toronto, Department of Medicine, Division of Neurology, Toronto, Canada
| | - Tine Hiorth Schøyen
- The Johns Hopkins University School of Medicine, Department of Molecular and Comparative Pathobiology, Baltimore, MD, USA
| | | | - Eriomina Shahaj
- Fondazione IRCCS Istituto Nazionale dei Tumori, Unit of Immunotherapy of Human Tumors, Milan, Italy
| | - Shivani Sharma
- University of California, Los Angeles, California NanoSystems Institute, Los Angeles, CA, USA
- University of California, Los Angeles, Department of Pathology and Laboratory Medicine, Los Angeles, CA, USA
- University of California, Los Angeles, Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Olga Shatnyeva
- AstraZeneca, Discovery Sciences, IMED Biotech Unit, Gothenburg, Sweden
| | - Faezeh Shekari
- Royan Institute for Stem Cell Biology and Technology, ACECR, Cell Science Research Center, Department of Stem Cells and Developmental Biology, Tehran, Iran
| | - Ganesh Vilas Shelke
- University of Gothenburg, Institute of Clinical Sciences, Department of Surgery, Sahlgrenska Cancer Center, Gothenburg, Sweden
- University of Gothenburg, Institute of Medicine at Sahlgrenska Academy, Krefting Research Centre, Gothenburg, Sweden
| | - Ashok K Shetty
- Research Service, Olin E. Teague Veterans’ Medical Center, Temple, TX, USA
- Texas A&M University College of Medicine, Institute for Regenerative Medicine and Department of Molecular and Cellular Medicine, College Station, TX, USA
| | | | - Pia R-M Siljander
- University of Helsinki, EV Core Facility, Helsinki, Finland
- University of Helsinki, Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences Research Programme, EV group, Helsinki, Finland
| | - Andreia M Silva
- INEB - Instituto de Engenharia Biomédica, Porto, Portugal
- University of Porto, i3S-Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- University of Porto, ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Porto, Portugal
| | - Agata Skowronek
- Maria Sklodowska-Curie Institute - Oncology Center, Gliwice Branch, Gliwice, Poland
| | - Orman L Snyder
- Kansas State University, College of Veterinary Medicine, Manhattan, KS, USA
| | | | - Barbara W Sódar
- Semmelweis University, Department of Genetics, Cell- and Immunobiology, Budapest, Hungary
| | - Carolina Soekmadji
- QIMR Berghofer Medical Research Institute, Herston, Australia
- The University of Queensland, Brisbane, Australia
| | - Javier Sotillo
- James Cook University, Australian Institute of Tropical Health and Medicine, Centre for Biodiscovery and Molecular Development of Therapeutics, Cairns, Australia
| | | | - Willem Stoorvogel
- Utrecht University, Faculty of Veterinary Medicine, Department of Biochemistry and Cell Biology, Utrecht, The Netherlands
| | - Shannon L Stott
- Harvard Medical School, Department of Medicine, Boston, MA, USA
- Massachusetts General Cancer Center, Boston, MA, USA
| | - Erwin F Strasser
- FAU Erlangen-Nuremberg, Transfusion and Haemostaseology Department, Erlangen, Germany
| | - Simon Swift
- University of Auckland, Department of Molecular Medicine and Pathology, Auckland, New Zealand
| | - Hidetoshi Tahara
- Hiroshima University, Institute of Biomedical & Health Sciences, Department of Cellular and Molecular Biology, Hiroshima, Japan
| | - Muneesh Tewari
- University of Michigan, Biointerfaces Institute, Ann Arbor, MI, USA
- University of Michigan, Department of Biomedical Engineering, Ann Arbor, MI, USA
- University of Michigan, Department of Internal Medicine - Hematology/Oncology Division, Ann Arbor, MI, USA
| | - Kate Timms
- University of Manchester, Manchester, UK
| | - Swasti Tiwari
- Georgetown University, Department of Medicine, Washington, DC, USA
- Sanjay Gandhi Postgraduate Institute of Medical Sciences, Department of Molecular Medicine & Biotechnology, Lucknow, India
| | - Rochelle Tixeira
- La Trobe University, La Trobe Institute for Molecular Science, Department of Biochemistry and Genetics, Bundoora, Australia
| | - Mercedes Tkach
- Institut Curie, INSERM U932, PSL Research University, Paris, France
| | - Wei Seong Toh
- National University of Singapore, Faculty of Dentistry, Singapore
| | - Richard Tomasini
- INSERM U1068, Aix Marseille University, CNRS UMR7258, Marseille, France
| | | | - Juan Pablo Tosar
- Institut Pasteur de Montevideo, Functional Genomics Unit, Montevideo, Uruguay
- Universidad de la República, Faculty of Science, Nuclear Research Center, Analytical Biochemistry Unit, Montevideo, Uruguay
| | | | - Lorena Urbanelli
- University of Perugia, Department of Chemistry, Biology and Biotechnology, Perugia, Italy
| | - Pieter Vader
- University Medical Center Utrecht, Laboratory for Clinical Chemistry & Hematology, Utrecht, The Netherlands
| | - Bas WM van Balkom
- University Medical Center Utrecht, Department of Nephrology and Hypertension, Utrecht, The Netherlands
| | - Susanne G van der Grein
- Utrecht University, Faculty of Veterinary Medicine, Department of Biochemistry and Cell Biology, Utrecht, The Netherlands
| | - Jan Van Deun
- Cancer Research Institute Ghent, Ghent, Belgium
- Ghent University, Department of Radiation Oncology and Experimental Cancer Research, Laboratory of Experimental Cancer Research, Ghent, Belgium
| | - Martijn JC van Herwijnen
- Utrecht University, Faculty of Veterinary Medicine, Department of Biochemistry and Cell Biology, Utrecht, The Netherlands
| | | | | | - Martin E van Royen
- Department of Pathology, Erasmus MC, Erasmus Optical Imaging Centre, Rotterdam, The Netherlands
| | | | - M Helena Vasconcelos
- IPATIMUP, Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
- University of Porto, Faculty of Pharmacy (FFUP), Porto, Portugal
- University of Porto, i3S-Instituto de Investigação e Inovação em Saúde, Porto, Portugal
| | - Ivan J Vechetti
- University of Kentucky, College of Medicine, Department of Physiology, Lexington, KY, USA
| | - Tiago D Veit
- Universidade Federal do Rio Grande do Sul, Instituto de Ciências Básicas da Saúde, Departamento de Microbiologia, Imunologia e Parasitologia, Porto Alegre, Brazil
| | - Laura J Vella
- The Florey Institute of Neuroscience and Mental Health, Melbourne, Australia
- The University of Melbourne, The Department of Medicine, Melbourne, Australia
| | - Émilie Velot
- UMR 7365 CNRS-Université de Lorraine, Vandœuvre-lès-Nancy, France
| | | | - Beate Vestad
- Oslo University Hospital Rikshospitalet, Research Institute of Internal Medicine, Oslo, Norway
- Regional Research Network on Extracellular Vesicles, RRNEV, Oslo, Norway
- University of Oslo, Institute of Clinical Medicine, Oslo, Norway
| | - Jose L Viñas
- Kidney Research Centre, Ottawa, Canada
- Ottawa Hospital Research Institute, Ottawa, Canada
- University of Ottawa, Ottawa, Canada
| | - Tamás Visnovitz
- Semmelweis University, Department of Genetics, Cell- and Immunobiology, Budapest, Hungary
| | - Krisztina V Vukman
- Semmelweis University, Department of Genetics, Cell- and Immunobiology, Budapest, Hungary
| | - Jessica Wahlgren
- University of Gothenburg, The Sahlgrenska Academy, Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Mölndal, Sweden
| | - Dionysios C Watson
- Case Western Reserve University, Department of Medicine, Cleveland, OH, USA
- University Hospitals Cleveland Medical Center, Department of Medicine, Cleveland, OH, USA
| | - Marca HM Wauben
- Utrecht University, Faculty of Veterinary Medicine, Department of Biochemistry and Cell Biology, Utrecht, The Netherlands
| | - Alissa Weaver
- Vanderbilt University School of Medicine, Department of Cell and Developmental Biology, Nashville, TN, USA
| | | | - Viktoria Weber
- Danube University Krems, Department for Biomedical Research and Christian Doppler Laboratory for Innovative Therapy Approaches in Sepsis, Krems an der Donau, Austria
| | - Ann M Wehman
- University of Würzburg, Rudolf Virchow Center, Würzburg, Germany
| | - Daniel J Weiss
- The University of Vermont Medical Center, Department of Medicine, Burlington, VT, USA
| | - Joshua A Welsh
- National Institutes of Health, National Cancer Institute, Center for Cancer Research, Bethesda, MD, USA
| | - Sebastian Wendt
- University Hospital RWTH Aachen, Department of Thoracic and Cardiovascular Surgery, Aachen, Germany
| | - Asa M Wheelock
- Karolinska Institute, Department of Medicine and Center for Molecular Medicine, Respiratory Medicine Unit, Stockholm, Sweden
| | - Zoltán Wiener
- Semmelweis University, Department of Genetics, Cell- and Immunobiology, Budapest, Hungary
| | - Leonie Witte
- University Medical Center Göttingen, Developmental Biochemistry, Göttingen, Germany
- University Medical Center Göttingen, Hematology and Oncology, Göttingen, Germany
| | - Joy Wolfram
- Chinese Academy of Sciences, Wenzhou Institute of Biomaterials and Engineering, Wenzhou, China
- Houston Methodist Research Institute, Department of Nanomedicine, Houston, TX, USA
- Mayo Clinic, Department of Transplantation Medicine/Department of Physiology and Biomedical Engineering, Jacksonville, FL, USA
| | - Angeliki Xagorari
- George Papanicolaou Hospital, Public Cord Blood Bank, Department of Haematology - BMT Unit, Thessaloniki, Greece
| | - Patricia Xander
- Universidade Federal de São Paulo Campus Diadema, Departamento de Ciências Farmacêuticas, Laboratório de Imunologia Celular e Bioquímica de Fungos e Protozoários, São Paulo, Brazil
| | - Jing Xu
- BC Cancer, Canada’s Michael Smith Genome Sciences Centre, Vancouver, Canada
- Simon Fraser University, Department of Molecular Biology and Biochemistry, Burnaby, Canada
| | - Xiaomei Yan
- Xiamen University, Department of Chemical Biology, Xiamen, China
| | - María Yáñez-Mó
- Centro de Biología Molecular Severo Ochoa, Instituto de Investigación Sanitaria la Princesa (IIS-IP), Madrid, Spain
- Universidad Autónoma de Madrid, Departamento de Biología Molecular, Madrid, Spain
| | - Hang Yin
- Tsinghua University, School of Pharmaceutical Sciences, Beijing, China
| | - Yuana Yuana
- Technical University Eindhoven, Faculty Biomedical Technology, Eindhoven, The Netherlands
| | - Valentina Zappulli
- University of Padova, Department of Comparative Biomedicine and Food Science, Padova, Italy
| | - Jana Zarubova
- Institute of Physiology CAS, Department of Biomaterials and Tissue Engineering, BIOCEV, Vestec, Czech Republic
- Institute of Physiology CAS, Department of Biomaterials and Tissue Engineering, Prague, Czech Republic
- University of California, Los Angeles, Department of Bioengineering, Los Angeles, CA, USA
| | - Vytautas Žėkas
- Vilnius University, Institute of Biomedical Sciences, Department of Physiology, Biochemistry, Microbiology and Laboratory Medicine, Vilnius, Lithuania
| | - Jian-ye Zhang
- Guangzhou Medical University, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology, Guangzhou, China
| | - Zezhou Zhao
- The Johns Hopkins University School of Medicine, Department of Molecular and Comparative Pathobiology, Baltimore, MD, USA
| | - Lei Zheng
- Nanfang Hospital, Southern Medical University, Department of Clinical Laboratory Medicine, Guangzhou, China
| | | | - Antje M Zickler
- Karolinska Institute, Clinical Research Center, Unit for Molecular Cell and Gene Therapy Science, Stockholm, Sweden
| | - Pascale Zimmermann
- Aix-Marseille Université, Institut Paoli-Calmettes, INSERM U1068, CNRS UMR7258, Centre de Recherche en Cancérologie de Marseille, Marseille, France
- KU Leuven (Leuven University), Department of Human Genetics, Leuven, Belgium
| | - Angela M Zivkovic
- University of California, Davis, Department of Nutrition, Davis, CA, USA
| | | | - Ewa K Zuba-Surma
- Jagiellonian University, Faculty of Biochemistry, Biophysics and Biotechnology, Department of Cell Biology, Kraków, Poland
| |
Collapse
|
108
|
Loozen GB, Caro J. On-chip optical trapping of extracellular vesicles using box-shaped composite SiO 2-Si 3N 4 waveguides. OPTICS EXPRESS 2018; 26:26985-27000. [PMID: 30469775 DOI: 10.1364/oe.26.026985] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
The application of on-chip optical trapping and Raman spectroscopy using a dual-waveguide trap has so far been limited to relatively big synthetic and biological particles (e.g., polystyrene beads and blood cells). Here, from simulations, we present the capabilities of dual-waveguide traps built from composite SiO2-Si3N4 waveguides for optical trapping of extracellular vesicles (EVs). EVs, tiny cell-derived particles of size in the range 30-1000 nm, strongly attract attention as potential biomarkers for cancer. EVs are hard to trap, because of their smallness and low index contract w.r.t. water. This poses a challenge for on-chip trapping. From finite-difference time-domain simulations we obtain the narrow beam emitted from the waveguide facet into water, for λ = 785 nm. For a pair of such beams, in a counter-propagating geometry and for facet separations of 5, 10 and 15 µm, we derive the inter-facet optical field, which has a characteristic interference pattern with hot spots for trapping, and calculate the optical force exerted on EVs of size in the range 50-1000 nm, as a function of EV position. We use two refractive index models for the EV optical properties. Integration of the force curves leads to the trapping potentials, which are well-shaped in the transverse and oscillatory in the longitudinal direction. By applying Ashkin's criterion, the conditions for stable trapping are established, the central result of this work. Very small EVs can be stably trapped with the traps by applying a power also suitable for Raman spectroscopy, down to a smallest EV diameter of 115 nm. We thus argue that this dual-waveguide trap is a promising lab-on-a-chip device with clinical relevance for diagnosis of cancer.
Collapse
|
109
|
Chiriacò MS, Bianco M, Nigro A, Primiceri E, Ferrara F, Romano A, Quattrini A, Furlan R, Arima V, Maruccio G. Lab-on-Chip for Exosomes and Microvesicles Detection and Characterization. SENSORS (BASEL, SWITZERLAND) 2018; 18:E3175. [PMID: 30241303 PMCID: PMC6210978 DOI: 10.3390/s18103175] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 09/05/2018] [Accepted: 09/16/2018] [Indexed: 12/11/2022]
Abstract
Interest in extracellular vesicles and in particular microvesicles and exosomes, which are constitutively produced by cells, is on the rise for their huge potential as biomarkers in a high number of disorders and pathologies as they are considered as carriers of information among cells, as well as being responsible for the spreading of diseases. Current methods of analysis of microvesicles and exosomes do not fulfill the requirements for their in-depth investigation and the complete exploitation of their diagnostic and prognostic value. Lab-on-chip methods have the potential and capabilities to bridge this gap and the technology is mature enough to provide all the necessary steps for a completely automated analysis of extracellular vesicles in body fluids. In this paper we provide an overview of the biological role of extracellular vesicles, standard biochemical methods of analysis and their limits, and a survey of lab-on-chip methods that are able to meet the needs of a deeper exploitation of these biological entities to drive their use in common clinical practice.
Collapse
Affiliation(s)
| | - Monica Bianco
- CNR NANOTEC Institute of Nanotechnology, via Monteroni, 73100 Lecce, Italy.
| | - Annamaria Nigro
- Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy.
| | | | - Francesco Ferrara
- CNR NANOTEC Institute of Nanotechnology, via Monteroni, 73100 Lecce, Italy.
- STMicroelectronics, Via Monteroni, I-73100 Lecce, Italy.
| | - Alessandro Romano
- Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy.
| | - Angelo Quattrini
- Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy.
| | - Roberto Furlan
- Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy.
| | - Valentina Arima
- CNR NANOTEC Institute of Nanotechnology, via Monteroni, 73100 Lecce, Italy.
| | - Giuseppe Maruccio
- CNR NANOTEC Institute of Nanotechnology, via Monteroni, 73100 Lecce, Italy.
- Department of Mathematics and Physics, University of Salento, via Monteroni, 73100 Lecce, Italy.
| |
Collapse
|
110
|
Lee W, Nanou A, Rikkert L, Coumans FAW, Otto C, Terstappen LWMM, Offerhaus HL. Label-Free Prostate Cancer Detection by Characterization of Extracellular Vesicles Using Raman Spectroscopy. Anal Chem 2018; 90:11290-11296. [PMID: 30157378 PMCID: PMC6170952 DOI: 10.1021/acs.analchem.8b01831] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
![]()
Mammalian cells release extracellular
vesicles (EVs) into their microenvironment that travel the entire
body along the stream of bodily fluids. EVs contain a wide range of
biomolecules. The transported cargo varies depending on the EV origin.
Knowledge of the origin and chemical composition of EVs can potentially
be used as a biomarker to detect, stage, and monitor diseases. In
this paper, we demonstrate the potential of EVs as a prostate cancer
biomarker. A Raman optical tweezer was employed to obtain Raman signatures
from four types of EV samples, which were red blood cell- and platelet-derived
EVs of healthy donors and the prostate cancer cell lines- (PC3 and
LNCaP) derived EVs. EVs’ Raman spectra could be clearly separated/classified
into distinct groups using principal component analysis (PCA) which
permits the discrimination of the investigated EV subtypes. These
findings may provide new methodology to detect and monitor early stage
cancer.
Collapse
Affiliation(s)
- Wooje Lee
- Optical Sciences, MESA+ Institute for Nanotechnology , University of Twente , 7500 AE , Enschede , The Netherlands
| | - Afroditi Nanou
- Department of Medical Cell BioPhysics, MIRA Institute , University of Twente , 7500 AE , Enschede , The Netherlands
| | - Linda Rikkert
- Department of Medical Cell BioPhysics, MIRA Institute , University of Twente , 7500 AE , Enschede , The Netherlands.,Laboratory of Experimental Clinical Chemistry, Academic Medical Center , University of Amsterdam , 1105 AZ , Amsterdam , The Netherlands.,Vesicle Observation Centre, Academic Medical Center , University of Amsterdam , 1105 AZ , Amsterdam , The Netherlands
| | - Frank A W Coumans
- Vesicle Observation Centre, Academic Medical Center , University of Amsterdam , 1105 AZ , Amsterdam , The Netherlands.,Department of Biomedical Engineering and Physics , Academic Medical Centre of the University of Amsterdam , 1105 AZ , Amsterdam , The Netherlands
| | - Cees Otto
- Department of Medical Cell BioPhysics, MIRA Institute , University of Twente , 7500 AE , Enschede , The Netherlands
| | - Leon W M M Terstappen
- Department of Medical Cell BioPhysics, MIRA Institute , University of Twente , 7500 AE , Enschede , The Netherlands
| | - Herman L Offerhaus
- Optical Sciences, MESA+ Institute for Nanotechnology , University of Twente , 7500 AE , Enschede , The Netherlands
| |
Collapse
|
111
|
Kabe Y, Suematsu M, Sakamoto S, Hirai M, Koike I, Hishiki T, Matsuda A, Hasegawa Y, Tsujita K, Ono M, Minegishi N, Hozawa A, Murakami Y, Kubo M, Itonaga M, Handa H. Development of a Highly Sensitive Device for Counting the Number of Disease-Specific Exosomes in Human Sera. Clin Chem 2018; 64:1463-1473. [PMID: 30021922 DOI: 10.1373/clinchem.2018.291963] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 06/12/2018] [Indexed: 01/26/2023]
Abstract
BACKGROUND Although circulating exosomes in blood play crucial roles in cancer development and progression, difficulties in quantifying exosomes hamper their application for reliable clinical testing. By combining the properties of nanobeads with optical disc technology, we have developed a novel device named the ExoCounter to determine the exact number of exosomes in the sera of patients with various types of cancer. METHOD In this system, individual exosomes were captured in the groove of an optical disc coated with antibodies against exosome surface antigens. The captured exosomes were labeled with antibody-conjugated magnetic nanobeads, and the number of the labeled exosomes was counted with an optical disc drive. RESULTS We showed that the ExoCounter could detect specific exosomes derived from cells or human serum without any enrichment procedures. The detection sensitivity and linearity with this system were higher than those with conventional detection methods such as ELISA or flow cytometry. In addition to the ubiquitous exosome markers CD9 and CD63, the cancer-related antigens CD147, carcinoembryonic antigen, and human epidermal growth factor receptor 2 (HER2) were also used to quantify cancer cell line-derived exosomes. Furthermore, analyses of a cross-sectional cohort of sera samples revealed that HER2-positive exosomes were significantly increased in patients with breast cancer or ovarian cancer compared with healthy individuals and those with noncancer diseases. CONCLUSIONS The ExoCounter system exhibits high performance in the direct detection of exosomes in cell culture and human sera. This method may enable reliable analysis of liquid biopsies.
Collapse
Affiliation(s)
- Yasuaki Kabe
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan; .,Japan Agency for Medical Research and Development (AMED), Core Research for Evolutional Science and Technology (CREST), Tokyo, Japan
| | - Makoto Suematsu
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Satoshi Sakamoto
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Miwa Hirai
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Ikko Koike
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Takako Hishiki
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Atsushi Matsuda
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Yuichi Hasegawa
- Healthcare Business Division, JVCKENWOOD Corporation, Yokosuka, Japan
| | - Koji Tsujita
- Healthcare Business Division, JVCKENWOOD Corporation, Yokosuka, Japan
| | - Masayuki Ono
- Healthcare Business Division, JVCKENWOOD Corporation, Yokosuka, Japan
| | - Naoko Minegishi
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Atsushi Hozawa
- Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Miyagi, Japan
| | - Yoshinori Murakami
- Division of Molecular Pathology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Michiaki Kubo
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Makoto Itonaga
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan; .,Healthcare Business Division, JVCKENWOOD Corporation, Yokosuka, Japan
| | - Hiroshi Handa
- Department of Nanoparticle Translational Research, Tokyo Medical University, Tokyo, Japan.
| |
Collapse
|
112
|
Lane RE, Korbie D, Hill MM, Trau M. Extracellular vesicles as circulating cancer biomarkers: opportunities and challenges. Clin Transl Med 2018; 7:14. [PMID: 29855735 PMCID: PMC5981152 DOI: 10.1186/s40169-018-0192-7] [Citation(s) in RCA: 169] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 05/23/2018] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles (EVs) are small, lipid-bound particles containing nucleic acid and protein cargo which are excreted from cells under a variety of normal and pathological conditions. EVs have garnered substantial research interest in recent years, due to their potential utility as circulating biomarkers for a variety of diseases, including numerous types of cancer. The following review will discuss the current understanding of the form and function of EVs, their specific role in cancer pathogenesis and their potential for non-invasive disease diagnosis and/or monitoring. This review will also highlight several key issues for this field, including the importance of implementing robust and reproducible sample handling protocols, and the challenge of extracting an EV-specific biomarker signal from a complex biological background.
Collapse
Affiliation(s)
- R E Lane
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD, Australia
| | - D Korbie
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD, Australia
| | - M M Hill
- The University of Queensland Diamantina Institute, Faculty of Medicine, Translational Research Institute, The University of Queensland, Woolloongabba, QLD, Australia.,QIMR-Berghofer Medical Research Institute, Herston, QLD, Australia
| | - M Trau
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD, Australia. .,School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia.
| |
Collapse
|
113
|
Sizing lipid droplets from adult and geriatric mouse liver tissue via nanoparticle tracking analysis. Anal Bioanal Chem 2018; 410:3629-3638. [PMID: 29663061 DOI: 10.1007/s00216-018-1016-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 02/23/2018] [Accepted: 03/09/2018] [Indexed: 12/24/2022]
Abstract
The significance of lipid droplets in lipid metabolism, cell signaling, and regulating longevity is increasingly recognized, yet the lipid droplet's unique properties and architecture make it difficult to size and study using conventional methods. To begin to address this issue, we demonstrate the capabilities of nanoparticle tracking analysis (NTA) for sizing of lipid droplets. NTA was found to be adequate to assess lipid droplet stability over time, indicating that lipid droplet preparations are stable for up to 24 h. NTA had the ability to compare the size distributions of lipid droplets from adult and geriatric mouse liver tissue, suggesting an age-related decrease in lipid droplet size. This is the first report on the use of NTA to size intracellular organelles. Graphical Abstract Light scattering reveals the temporal positions of individual lipid droplets, which are recorded with a camera. The two-dimensional diffusion constant of each lipid droplet is extracted from the data set, which is then used to calculate a hydrodynamic radius using the Stokes-Einstein equation.
Collapse
|
114
|
Sharma S, LeClaire M, Gimzewski JK. Ascent of atomic force microscopy as a nanoanalytical tool for exosomes and other extracellular vesicles. NANOTECHNOLOGY 2018; 29:132001. [PMID: 29376505 DOI: 10.1088/1361-6528/aaab06] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Over the last 30 years, atomic force microscopy (AFM) has made several significant contributions to the field of biology and medicine. In this review, we draw our attention to the recent applications and promise of AFM as a high-resolution imaging and force sensing technology for probing subcellular vesicles: exosomes and other extracellular vesicles. Exosomes are naturally occurring nanoparticles found in several body fluids such as blood, saliva, cerebrospinal fluid, amniotic fluid and urine. Exosomes mediate cell-cell communication, transport proteins and genetic content between distant cells, and are now known to play important roles in progression of diseases such as cancers, neurodegenerative disorders and infectious diseases. Because exosomes are smaller than 100 nm (about 30-120 nm), the structural and molecular characterization of these vesicles at the individual level has been challenging. AFM has revealed a new degree of complexity in these nanosized vesicles and generated growing interest as a nanoscale tool for characterizing the abundance, morphology, biomechanics, and biomolecular make-up of exosomes. With the recent interest in exosomes for diagnostic and therapeutic applications, AFM-based characterization promises to contribute towards improved understanding of these particles at the single vesicle and sub-vesicular levels. When coupled with complementary methods like optical super resolution STED and Raman, AFM could further unlock the potential of exosomes as disease biomarkers and as therapeutic agents.
Collapse
Affiliation(s)
- S Sharma
- California NanoSystems Institute, University of California, Los Angeles, CA 90095, United States of America
| | | | | |
Collapse
|
115
|
Manandhar S, Kothandan VK, Oh J, Yoo SH, Hwang J, Hwang SR. A pharmaceutical investigation into exosomes. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2018. [DOI: 10.1007/s40005-018-0391-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
116
|
Shao H, Im H, Castro CM, Breakefield X, Weissleder R, Lee H. New Technologies for Analysis of Extracellular Vesicles. Chem Rev 2018; 118:1917-1950. [PMID: 29384376 PMCID: PMC6029891 DOI: 10.1021/acs.chemrev.7b00534] [Citation(s) in RCA: 1114] [Impact Index Per Article: 159.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Extracellular vesicles (EVs) are diverse, nanoscale membrane vesicles actively released by cells. Similar-sized vesicles can be further classified (e.g., exosomes, microvesicles) based on their biogenesis, size, and biophysical properties. Although initially thought to be cellular debris, and thus under-appreciated, EVs are now increasingly recognized as important vehicles of intercellular communication and circulating biomarkers for disease diagnoses and prognosis. Despite their clinical potential, the lack of sensitive preparatory and analytical technologies for EVs poses a barrier to clinical translation. New analytical platforms including molecular ones are thus actively being developed to address these challenges. Recent advances in the field are expected to have far-reaching impact in both basic and translational studies. This article aims to present a comprehensive and critical overview of emerging analytical technologies for EV detection and their clinical applications.
Collapse
Affiliation(s)
- Huilin Shao
- Departments of Biomedical Engineering and Surgery, National University of Singapore
- Biomedical Institute for Global Health Research and Technology, National University of Singapore
- Institute of Molecular and Cell Biology, Agency for Science Technology and Research
| | - Hyungsoon Im
- Center for Systems Biology, Massachusetts General Hospital
- Department of Radiology, Massachusetts General Hospital
| | - Cesar M. Castro
- Center for Systems Biology, Massachusetts General Hospital
- Department of Medicine, Massachusetts General Hospital
| | - Xandra Breakefield
- Department of Radiology, Massachusetts General Hospital
- Department of Neurology, Massachusetts General Hospital
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital
- Department of Radiology, Massachusetts General Hospital
- Department of Systems Biology, Harvard Medical School
| | - Hakho Lee
- Center for Systems Biology, Massachusetts General Hospital
- Department of Radiology, Massachusetts General Hospital
| |
Collapse
|
117
|
Kailashiya J. Platelet-derived microparticles analysis: Techniques, challenges and recommendations. Anal Biochem 2018; 546:78-85. [PMID: 29408673 DOI: 10.1016/j.ab.2018.01.030] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Revised: 01/05/2018] [Accepted: 01/30/2018] [Indexed: 12/15/2022]
Abstract
Platelet-derived microparticles (PMP) are nano size fragments (100-1000 nm) released from platelets under various physiological and pathological conditions. PMP are the most abundant microparticles present in human blood. In recent past years PMP have caught attention of many clinicians as well as researchers for being associated with many diseases like cardio-vascular diseases, infections etc; and have emerged as potential biomarkers. Owing to their small size and diverse phenotype, structure and functions, microparticles including PMP render many challenges during sample handling, estimation and characterization. PMP can be analyzed for many parameters like absolute count, size distribution, functions, content, surface proteins and other phenotypic characteristics. Many techniques have been invented to analyze PMP and other extracellular vesicles for these parameters, but none of them is capable of examining all parameters alone. Apart from it, every technique has its own advantages, limitations and sets of recommendations while using it. This often leads to applying multiple techniques in combination for accurately measuring various parameters and user has to decide cautiously which technique has to be used for their selected parameter testing. This review compiles various methods, techniques, challenges during PMP analysis and recommendations based on previous studies, aimed at guiding users for selecting the most suitable techniques for their experiments with PMP.
Collapse
Affiliation(s)
- Jyotsna Kailashiya
- Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, UP, 221005, India.
| |
Collapse
|
118
|
Bimetallic nanoplasmonic gap-mode SERS substrate for lung normal and cancer-derived exosomes detection. J Taiwan Inst Chem Eng 2017. [DOI: 10.1016/j.jtice.2017.09.026] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
119
|
Raman spectroscopy uncovers biochemical tissue-related features of extracellular vesicles from mesenchymal stromal cells. Sci Rep 2017; 7:9820. [PMID: 28852131 PMCID: PMC5575260 DOI: 10.1038/s41598-017-10448-1] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 08/08/2017] [Indexed: 01/08/2023] Open
Abstract
Extracellular vesicles (EVs) from mesenchymal stromal cells (MSC) are emerging as valuable therapeutic agents for tissue regeneration and immunomodulation, but their clinical applications have so far been limited by the technical restraints of current isolation and characterisation procedures. This study shows for the first time the successful application of Raman spectroscopy as label-free, sensitive and reproducible means of carrying out the routine bulk characterisation of MSC-derived vesicles before their use in vitro or in vivo, thus promoting the translation of EV research to clinical practice. The Raman spectra of the EVs of bone marrow and adipose tissue-derived MSCs were compared with human dermal fibroblast EVs in order to demonstrate the ability of the method to distinguish the vesicles of the three cytotypes automatically with an accuracy of 93.7%. Our data attribute a Raman fingerprint to EVs from undifferentiated and differentiated cells of diverse tissue origin, and provide insights into the biochemical characteristics of EVs from different sources and into the differential contribution of sphingomyelin, gangliosides and phosphatidilcholine to the Raman spectra themselves.
Collapse
|
120
|
Coumans FAW, Brisson AR, Buzas EI, Dignat-George F, Drees EEE, El-Andaloussi S, Emanueli C, Gasecka A, Hendrix A, Hill AF, Lacroix R, Lee Y, van Leeuwen TG, Mackman N, Mäger I, Nolan JP, van der Pol E, Pegtel DM, Sahoo S, Siljander PRM, Sturk G, de Wever O, Nieuwland R. Methodological Guidelines to Study Extracellular Vesicles. Circ Res 2017; 120:1632-1648. [PMID: 28495994 DOI: 10.1161/circresaha.117.309417] [Citation(s) in RCA: 743] [Impact Index Per Article: 92.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Owing to the relationship between extracellular vesicles (EVs) and physiological and pathological conditions, the interest in EVs is exponentially growing. EVs hold high hopes for novel diagnostic and translational discoveries. This review provides an expert-based update of recent advances in the methods to study EVs and summarizes currently accepted considerations and recommendations from sample collection to isolation, detection, and characterization of EVs. Common misconceptions and methodological pitfalls are highlighted. Although EVs are found in all body fluids, in this review, we will focus on EVs from human blood, not only our most complex but also the most interesting body fluid for cardiovascular research.
Collapse
Affiliation(s)
- Frank A W Coumans
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Alain R Brisson
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Edit I Buzas
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Françoise Dignat-George
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Esther E E Drees
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Samir El-Andaloussi
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Costanza Emanueli
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Aleksandra Gasecka
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - An Hendrix
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Andrew F Hill
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Romaric Lacroix
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Yi Lee
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Ton G van Leeuwen
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Nigel Mackman
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Imre Mäger
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - John P Nolan
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Edwin van der Pol
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - D Michiel Pegtel
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Susmita Sahoo
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Pia R M Siljander
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Guus Sturk
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Olivier de Wever
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Rienk Nieuwland
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.).
| |
Collapse
|
121
|
Tang Y, Thillier Y, Liu R, Li X, Lam KS, Gao T. Single-Bead Quantification of Peptide Loading Distribution for One-Bead One-Compound Library Synthesis Using Confocal Raman Spectroscopy. Anal Chem 2017; 89:7000-7008. [PMID: 28530391 DOI: 10.1021/acs.analchem.7b00516] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
We report an analytical method to determine peptide loading of "one-bead one-compound" (OBOC) combinatorial peptide libraries at single-bead level. The quantification is based on a linear relationship between the amount of N-terminal amino groups on individual peptide beads and the intensity of Raman signal obtained from a specifically designed reporter labeled on amino groups. Confocal Raman spectroscopy was employed to characterize peptide loading of beads with defined peptide sequences and from OBOC combinatorial peptide libraries. Although amine loading of blank TentaGel beads was found to be uniform, peptide loading among beads of OBOC peptide libraries varied substantially, particularly for those libraries with long sequences. Construction of OBOC libraries can be monitored with this novel analytical technique so that synthetic conditions can be optimized for the preparation of high-quality OBOC peptide libraries. As the variability of peptide loading of individual library beads can significantly influence the screening results, quantitative information obtained by this method will allow us to gain insight into the complexity and challenge of OBOC library synthesis and screening.
Collapse
Affiliation(s)
- Yuchen Tang
- College of Chemistry, Central China Normal University , Wuhan 430079, China.,China Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, Wuhan 430079, China.,Department of Biochemistry and Molecular Medicine, University of California at Davis , Sacramento, California 95817, United States
| | - Yann Thillier
- Department of Biochemistry and Molecular Medicine, University of California at Davis , Sacramento, California 95817, United States
| | - Ruiwu Liu
- Department of Biochemistry and Molecular Medicine, University of California at Davis , Sacramento, California 95817, United States
| | - Xiaocen Li
- Department of Biochemistry and Molecular Medicine, University of California at Davis , Sacramento, California 95817, United States
| | - Kit S Lam
- Department of Biochemistry and Molecular Medicine, University of California at Davis , Sacramento, California 95817, United States
| | - Tingjuan Gao
- College of Chemistry, Central China Normal University , Wuhan 430079, China.,China Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, Wuhan 430079, China
| |
Collapse
|
122
|
The Methods of Choice for Extracellular Vesicles (EVs) Characterization. Int J Mol Sci 2017; 18:ijms18061153. [PMID: 28555055 PMCID: PMC5485977 DOI: 10.3390/ijms18061153] [Citation(s) in RCA: 367] [Impact Index Per Article: 45.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 05/23/2017] [Accepted: 05/24/2017] [Indexed: 11/24/2022] Open
Abstract
In recent years, extracellular vesicles (EVs) have become a subject of intense study. These membrane-enclosed spherical structures are secreted by almost every cell type and are engaged in the transport of cellular content (cargo) from parental to target cells. The impact of EVs transfer has been observed in many vital cellular processes including cell-to-cell communication and immune response modulation; thus, a fast and precise characterization of EVs may be relevant for both scientific and diagnostic purposes. In this review, the most popular analytical techniques used in EVs studies are presented with the emphasis on exosomes and microvesicles characterization.
Collapse
|
123
|
Pariset E, Agache V, Millet A. Extracellular Vesicles: Isolation Methods. ACTA ACUST UNITED AC 2017; 1:e1700040. [DOI: 10.1002/adbi.201700040] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 03/24/2017] [Indexed: 12/21/2022]
Affiliation(s)
- Eloise Pariset
- CEA; LETI; MINATEC Campus 38054 Grenoble France
- Université Grenoble-Alpes; 38000 Grenoble France
| | - Vincent Agache
- CEA; LETI; MINATEC Campus 38054 Grenoble France
- Université Grenoble-Alpes; 38000 Grenoble France
| | - Arnaud Millet
- ATIP/Avenir Team “Mechanobiology, Immunity and Cancer”; Inserm U1205, Brain-Tech Lab 38054 Grenoble France
- Université Grenoble-Alpes; 38000 Grenoble France
| |
Collapse
|
124
|
Procoagulant activity of extracellular vesicles as a potential biomarker for risk of thrombosis and DIC in patients with acute leukaemia. J Thromb Thrombolysis 2017; 43:224-232. [PMID: 28074413 DOI: 10.1007/s11239-016-1471-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Haemostatic complication is common for patients with hematologic malignancies. Recent studies suggest that the procoagulant activity (PCA) of extracellular vesicles (EV) may play a major role in venous thromboembolism and disseminated intravascular coagulation (DIC) in acute leukaemia. To study the impact of EVs from leukaemic patients on thrombin generation and to assess EV-PCA as a potential biomarker for thrombotic complications in patients with acute leukaemia. Blood samples from a cohort of patients with newly diagnosed acute leukaemia were obtained before treatment (D-0), 3 and 7 days after treatment (D-3 and D-7). Extracellular vesicles were isolated and concentrated by ultracentrifugation. EV-PCA was assessed by thrombin generation assay, and EV-associated tissue factor activity was measured using a commercial bio-immunoassay (Zymuphen MP-TF®). Of the 53 patients, 6 had increased EV-PCA at D-0 and 4 had a thrombotic event. Patients without thrombotic events (n = 47) had no elevated EV-PCA. One patient had increased EVs with procoagulant activity at D-3 and developed a DIC at D-5. This patient had no increased EVs-related tissue factor activity from D-0 to D-7 (<2 pg/ml). Eight patients had increased EVs with tissue factor activity (>2 pg/ml), of these, four had a thrombosis and two had haemorrhages. Procoagulant activity of extracellular vesicles could have a predictive value in excluding the risk of thrombotic events. Our findings also suggest a possible association between thrombotic events and EV-PCA.
Collapse
|
125
|
A specific spectral signature of serum and plasma-derived extracellular vesicles for cancer screening. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:835-841. [DOI: 10.1016/j.nano.2016.11.016] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 11/03/2016] [Accepted: 11/26/2016] [Indexed: 12/29/2022]
|
126
|
Mateescu B, Kowal EJK, van Balkom BWM, Bartel S, Bhattacharyya SN, Buzás EI, Buck AH, de Candia P, Chow FWN, Das S, Driedonks TAP, Fernández-Messina L, Haderk F, Hill AF, Jones JC, Van Keuren-Jensen KR, Lai CP, Lässer C, Liegro ID, Lunavat TR, Lorenowicz MJ, Maas SLN, Mäger I, Mittelbrunn M, Momma S, Mukherjee K, Nawaz M, Pegtel DM, Pfaffl MW, Schiffelers RM, Tahara H, Théry C, Tosar JP, Wauben MHM, Witwer KW, Nolte-'t Hoen ENM. Obstacles and opportunities in the functional analysis of extracellular vesicle RNA - an ISEV position paper. J Extracell Vesicles 2017; 6:1286095. [PMID: 28326170 PMCID: PMC5345583 DOI: 10.1080/20013078.2017.1286095] [Citation(s) in RCA: 548] [Impact Index Per Article: 68.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 12/25/2016] [Indexed: 02/07/2023] Open
Abstract
The release of RNA-containing extracellular vesicles (EV) into the extracellular milieu has been demonstrated in a multitude of different in vitro cell systems and in a variety of body fluids. RNA-containing EV are in the limelight for their capacity to communicate genetically encoded messages to other cells, their suitability as candidate biomarkers for diseases, and their use as therapeutic agents. Although EV-RNA has attracted enormous interest from basic researchers, clinicians, and industry, we currently have limited knowledge on which mechanisms drive and regulate RNA incorporation into EV and on how RNA-encoded messages affect signalling processes in EV-targeted cells. Moreover, EV-RNA research faces various technical challenges, such as standardisation of EV isolation methods, optimisation of methodologies to isolate and characterise minute quantities of RNA found in EV, and development of approaches to demonstrate functional transfer of EV-RNA in vivo. These topics were discussed at the 2015 EV-RNA workshop of the International Society for Extracellular Vesicles. This position paper was written by the participants of the workshop not only to give an overview of the current state of knowledge in the field, but also to clarify that our incomplete knowledge – of the nature of EV(-RNA)s and of how to effectively and reliably study them – currently prohibits the implementation of gold standards in EV-RNA research. In addition, this paper creates awareness of possibilities and limitations of currently used strategies to investigate EV-RNA and calls for caution in interpretation of the obtained data.
Collapse
Affiliation(s)
- Bogdan Mateescu
- Department of Biology, Swiss Federal Institute of Technology Zurich (ETH Zürich) , Zurich , Switzerland
| | - Emma J K Kowal
- Department of Biology, Massachusetts Institute of Technology , Cambridge , MA , USA
| | - Bas W M van Balkom
- Department of Nephrology and Hypertension, UMC Utrecht , Utrecht , the Netherlands
| | - Sabine Bartel
- Experimental Asthma Research, Priority Area Asthma & Allergy, Research Center Borstel, Leibniz-Center for Medicine and Biosciences, Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL) , Borstel , Germany
| | - Suvendra N Bhattacharyya
- Department of Science and Technology, CSIR-Indian Institute of Chemical Biology , Kolkata , India
| | - Edit I Buzás
- Department of Genetics, Cell- and Immunobiology, Semmelweis University , Budapest , Hungary
| | - Amy H Buck
- Institute of Immunology and Infection Research, Centre for Immunity, Infection and Evolution, School of Biological Sciences, University of Edinburgh , Edinburgh , UK
| | | | - Franklin W N Chow
- Institute of Immunology and Infection Research, Centre for Immunity, Infection and Evolution, School of Biological Sciences, University of Edinburgh , Edinburgh , UK
| | - Saumya Das
- Cardiovascular Research Institute, Massachusetts General Hospital , Boston , MA , USA
| | - Tom A P Driedonks
- Department of Biochemistry & Cell Biology, Faculty of Veterinary Medicine, Utrecht University , Utrecht , the Netherlands
| | | | - Franziska Haderk
- Department of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Medicine, Helen Diller Family Comprehensive Cancer Center, UC San Francisco, San Francisco, CA, USA
| | - Andrew F Hill
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University , Bundoora , Australia
| | - Jennifer C Jones
- Molecular Immunogenetics & Vaccine Research Section, Vaccine Branch, CCR, NCI , Bethesda , MD , USA
| | | | - Charles P Lai
- Institute of Biomedical Engineering, National Tsing Hua University , Hsinchu , Taiwan
| | - Cecilia Lässer
- Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA, USA; Krefting Research Centre, Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Italia di Liegro
- Department of Experimental Biomedicine and Clinical Neurosciences (BIONEC), University of Palermo , Palermo , Italy
| | - Taral R Lunavat
- Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA, USA; Krefting Research Centre, Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Magdalena J Lorenowicz
- Center for Molecular Medicine, University Medical Center Utrecht & Regenerative Medicine Center , Utrecht , the Netherlands
| | - Sybren L N Maas
- Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School , Boston , MA , USA
| | - Imre Mäger
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK; Institute of Technology, University of Tartu, Tartu, Estonia
| | - Maria Mittelbrunn
- Instituto de Investigación del Hospital 12 de Octubre , Madrid , Spain
| | - Stefan Momma
- Institute of Neurology (Edinger Institute), Frankfurt University Medical School , Frankfurt am Main , Germany
| | - Kamalika Mukherjee
- Department of Science and Technology, CSIR-Indian Institute of Chemical Biology , Kolkata , India
| | - Muhammed Nawaz
- Department of Pathology and Forensic Medicine, Ribeirão Preto School of Medicine, University of Sao Paulo , Sao Paulo , Brazil
| | - D Michiel Pegtel
- Department of Pathology, Exosomes Research Group, VU University Medical Center , Amsterdam , the Netherlands
| | - Michael W Pfaffl
- Animal Physiology and Immunology, School of Life Sciences, Technical University of Munich (TUM) Weihenstephan , Freising , Germany
| | - Raymond M Schiffelers
- Laboratory Clinical Chemistry & Haematology, University Medical Center Utrecht , Utrecht , the Netherlands
| | - Hidetoshi Tahara
- Department of Cellular and Molecular Biology, Institute of Biomedical & Health Sciences, Hiroshima University , Hiroshima , Japan
| | - Clotilde Théry
- Institut Curie, PSL Research University, INSERM U932 , Paris , France
| | - Juan Pablo Tosar
- Functional Genomics Unit, Institut Pasteur de Montevideo, Nuclear Research Center, Faculty of Science, Universidad de la República , Montevideo , Uruguay
| | - Marca H M Wauben
- Department of Biochemistry & Cell Biology, Faculty of Veterinary Medicine, Utrecht University , Utrecht , the Netherlands
| | - Kenneth W Witwer
- Department of Molecular and Comparative Pathobiology and Department of Neurology, The Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Esther N M Nolte-'t Hoen
- Department of Biochemistry & Cell Biology, Faculty of Veterinary Medicine, Utrecht University , Utrecht , the Netherlands
| |
Collapse
|
127
|
Valkonen S, van der Pol E, Böing A, Yuana Y, Yliperttula M, Nieuwland R, Laitinen S, Siljander P. Biological reference materials for extracellular vesicle studies. Eur J Pharm Sci 2017; 98:4-16. [DOI: 10.1016/j.ejps.2016.09.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 09/06/2016] [Accepted: 09/06/2016] [Indexed: 01/05/2023]
|
128
|
Abstract
Research in the field of extracellular vesicles (EVs) is challenged by the small size of the nano-sized particles. Apart from the use of transmission and scanning electron microscopy, established technical platforms to visualize, quantify, and characterize nano-sized EVs were lacking. Recently, methodologies to characterize nano-sized EVs have been developed. This chapter aims to summarize physical principles of novel and conventional technologies to be used in the EV field and to discuss advantages and limitations.
Collapse
|
129
|
Linares R, Tan S, Gounou C, Brisson AR. Imaging and Quantification of Extracellular Vesicles by Transmission Electron Microscopy. Methods Mol Biol 2017; 1545:43-54. [PMID: 27943206 DOI: 10.1007/978-1-4939-6728-5_4] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Extracellular vesicles (EVs) are cell-derived vesicles that are present in blood and other body fluids. EVs raise major interest for their diverse physiopathological roles and their potential biomedical applications. However, the characterization and quantification of EVs constitute major challenges, mainly due to their small size and the lack of methods adapted for their study. Electron microscopy has made significant contributions to the EV field since their initial discovery. Here, we describe the use of two transmission electron microscopy (TEM) techniques for imaging and quantifying EVs. Cryo-TEM combined with receptor-specific gold labeling is applied to reveal the morphology, size, and phenotype of EVs, while their enumeration is achieved after high-speed sedimentation on EM grids.
Collapse
Affiliation(s)
- Romain Linares
- Molecular Imaging and NanoBioTechnology, UMR-5248-CBMN, CNRS-University of Bordeaux-IPB, Bat. B14, Allée Geoffroy Saint-Hilaire, 33600, Pessac, France
| | - Sisareuth Tan
- Molecular Imaging and NanoBioTechnology, UMR-5248-CBMN, CNRS-University of Bordeaux-IPB, Bat. B14, Allée Geoffroy Saint-Hilaire, 33600, Pessac, France
| | - Céline Gounou
- Molecular Imaging and NanoBioTechnology, UMR-5248-CBMN, CNRS-University of Bordeaux-IPB, Bat. B14, Allée Geoffroy Saint-Hilaire, 33600, Pessac, France
| | - Alain R Brisson
- Molecular Imaging and NanoBioTechnology, UMR-5248-CBMN, CNRS-University of Bordeaux-IPB, Bat. B14, Allée Geoffroy Saint-Hilaire, 33600, Pessac, France.
| |
Collapse
|
130
|
Buzás EI, Gardiner C, Lee C, Smith ZJ. Single particle analysis: Methods for detection of platelet extracellular vesicles in suspension (excluding flow cytometry). Platelets 2016; 28:249-255. [PMID: 28033028 DOI: 10.1080/09537104.2016.1260704] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Extracellular vesicles (EVs) are small, membrane-bound particles released by all cell types, including abundant release by platelets. EVs are a topic of increasing interest in the academic and clinical community due to their increasingly recognised and diverse role in normal biology as well as in disease. However, typical analysis methods to characterise EVs released by cultured cells or isolated from whole blood or other body fluids are restricted to bulk analysis of all EVs in a sample. In this review, we discuss the motivation for analysis of individual EVs, as well as discuss three emerging methods for physical and chemical characterisation of individual EVs: nanoparticle tracking analysis, tunable resistive pulse sensing and Raman spectroscopy. We give brief descriptions of the working principles of each technique, along with a review noting the benefits and limitations of each method as applied to detection of single EVs.
Collapse
Affiliation(s)
- Edit I Buzás
- a Department of Genetics, Cell- and Immunobiology , Semmelweis University , Budapest , Hungary
| | - Chris Gardiner
- b Haemostasis Research Unit, Research Department of Hematology , University College London , London , UK
| | - Changwon Lee
- c Center for Biophotonics , University of California Davis Medical Center , Sacramento , CA , USA
| | - Zachary J Smith
- d Department of Precision Machinery and Precision Instrumentation , University of Science and Technology of China , Hefei , Anhui , 230027 , China
| |
Collapse
|
131
|
Mihály J, Deák R, Szigyártó IC, Bóta A, Beke-Somfai T, Varga Z. Characterization of extracellular vesicles by IR spectroscopy: Fast and simple classification based on amide and CH stretching vibrations. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2016; 1859:459-466. [PMID: 27989744 DOI: 10.1016/j.bbamem.2016.12.005] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 11/29/2016] [Accepted: 12/14/2016] [Indexed: 12/21/2022]
Abstract
Extracellular vesicles isolated by differential centrifugation from Jurkat T-cell line were investigated by attenuated total reflection Fourier-transform infrared spectroscopy (ATR-FTIR). Amide and CH stretching band intensity ratios calculated from IR bands, characteristic of protein and lipid components, proved to be distinctive for the different extracellular vesicle subpopulations. This proposed 'spectroscopic protein-to-lipid ratio', combined with the outlined spectrum-analysis protocol is valid also for low sample concentrations (0.15-0.05mg/ml total protein content) and can carry information about the presence of other non-vesicular formations such as aggregated proteins, lipoproteins and immune complexes. Detailed analysis of IR data reveals compositional changes of extracellular vesicles subpopulations: second derivative spectra suggest changes in protein composition from parent cell towards exosomes favoring proteins with β-turns and unordered motifs at the expense of intermolecular β-sheet structures. The IR-based protein-to-lipid assessment protocol was tested also for red blood cell derived microvesicles for which similar values were obtained. The potential applicability of this technique for fast and efficient characterization of vesicular components is high as the investigated samples require no further preparations and all the different molecular species can be determined in the same sample. The results indicate that ATR-FTIR measurements provide a simple and reproducible method for the screening of extracellular vesicle preparations. It is hoped that this sophisticated technique will have further impact in extracellular vesicle research.
Collapse
Affiliation(s)
- Judith Mihály
- Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences of the Hungarian Academy of Sciences, Magyar tudósok körútja 2, 1117 Budapest, Hungary.
| | - Róbert Deák
- Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences of the Hungarian Academy of Sciences, Magyar tudósok körútja 2, 1117 Budapest, Hungary
| | - Imola Csilla Szigyártó
- Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences of the Hungarian Academy of Sciences, Magyar tudósok körútja 2, 1117 Budapest, Hungary
| | - Attila Bóta
- Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences of the Hungarian Academy of Sciences, Magyar tudósok körútja 2, 1117 Budapest, Hungary
| | - Tamás Beke-Somfai
- Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences of the Hungarian Academy of Sciences, Magyar tudósok körútja 2, 1117 Budapest, Hungary
| | - Zoltán Varga
- Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences of the Hungarian Academy of Sciences, Magyar tudósok körútja 2, 1117 Budapest, Hungary
| |
Collapse
|
132
|
Foster BP, Balassa T, Benen TD, Dominovic M, Elmadjian GK, Florova V, Fransolet MD, Kestlerova A, Kmiecik G, Kostadinova IA, Kyvelidou C, Meggyes M, Mincheva MN, Moro L, Pastuschek J, Spoldi V, Wandernoth P, Weber M, Toth B, Markert UR. Extracellular vesicles in blood, milk and body fluids of the female and male urogenital tract and with special regard to reproduction. Crit Rev Clin Lab Sci 2016; 53:379-95. [PMID: 27191915 DOI: 10.1080/10408363.2016.1190682] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Extracellular vesicles (EVs) are released from almost all cells and tissues. They are able to transport substances (e.g. proteins, RNA or DNA) at higher concentrations than in their environment and may adhere in a receptor-controlled manner to specific cells or tissues in order to release their content into the respective target structure. Blood contains high concentrations of EVs mainly derived from platelets, and, at a smaller amount, from erythrocytes. The female and male reproductive tracts produce EVs which may be associated with fertility or infertility and are released into body fluids and mucosas of the urogenital organs. In this review, the currently relevant detection methods are presented and critically compared. During pregnancy, placenta-derived EVs are dynamically detectable in peripheral blood with changing profiles depending upon progress of pregnancy and different pregnancy-associated pathologies, such as preeclampsia. EVs offer novel non-invasive diagnostic tools which may reflect the situation of the placenta and the foetus. EVs in urine have the potential of reflecting urogenital diseases including cancers of the neighbouring organs. Several methods for detection, quantification and phenotyping of EVs have been established, which include electron microscopy, flow cytometry, ELISA-like methods, Western blotting and analyses based on Brownian motion. This review article summarises the current knowledge about EVs in blood and cord blood, in the different compartments of the male and female reproductive tracts, in trophoblast cells from normal and pre-eclamptic pregnancies, in placenta ex vivo perfusate, in the amniotic fluid, and in breast milk, as well as their potential effects on natural killer cells as possible targets.
Collapse
Affiliation(s)
- B P Foster
- a Maternal and Fetal Health Research Centre, School of Biomedicine, University of Manchester, and Manchester Academic Health Sciences Centre, University Research , Manchester , UK
| | - T Balassa
- b Department of Medical Microbiology and Immunology , Medical School, University of Pécs , Pécs , Hungary
| | - T D Benen
- c Microtrac GmbH , Krefeld , Germany
| | - M Dominovic
- d Department of Physiology and Immunology , Medical Faculty, University of Rijeka , Rijeka , Croatia
| | - G K Elmadjian
- e Repro Inova Immunology Laboratory , Sofia , Bulgaria
| | - V Florova
- f Department of Obstetrics , Gynecology and Perinatology, First Moscow State Medical University , Moscow , Russia
| | - M D Fransolet
- g Laboratory of Tumor and Development Biology , GIGA-R, University of Liège , Liège , Belgium
| | - A Kestlerova
- h Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital and First Faculty of Medicine , Charles University Prague , Czech Republic
- i Institute of Biophysics and Informatics, First Faculty of Medicine, Charles University , Prague , Czech Republic
| | - G Kmiecik
- j Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero , Brescia , Italy
| | - I A Kostadinova
- k Department of Immunoneuroendocrinology , Institute of Biology and Immunology of Reproduction , Sofia , Bulgaria
| | - C Kyvelidou
- l Department of Biology , University of Crete , Crete , Greece
| | - M Meggyes
- b Department of Medical Microbiology and Immunology , Medical School, University of Pécs , Pécs , Hungary
| | - M N Mincheva
- m Repro Inova Immunology Laboratory , Sofia , Bulgaria
| | - L Moro
- n ISGlobal, Barcelona Centre for International Health Research (CRESIB), Hospital Clínic- Universitat de Barcelona , Barcelona , Spain
- o Department of Obstetrics , Placenta-Lab, University Hospital Jena , Jena , Germany
| | - J Pastuschek
- o Department of Obstetrics , Placenta-Lab, University Hospital Jena , Jena , Germany
| | - V Spoldi
- j Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero , Brescia , Italy
| | - P Wandernoth
- p Institute of Anatomy, University Hospital, University Duisburg-Essen , Essen , Germany
| | - M Weber
- o Department of Obstetrics , Placenta-Lab, University Hospital Jena , Jena , Germany
| | - B Toth
- q Department of Gynecological Endocrinology and Fertility Disorders , Ruprecht-Karls University of Heidelberg , Heidelberg , Germany
| | - U R Markert
- o Department of Obstetrics , Placenta-Lab, University Hospital Jena , Jena , Germany
| |
Collapse
|
133
|
Xu R, Fitts A, Li X, Fernandes J, Pochampally R, Mao J, Liu YM. Quantification of Small Extracellular Vesicles by Size Exclusion Chromatography with Fluorescence Detection. Anal Chem 2016; 88:10390-10394. [PMID: 27689436 PMCID: PMC5089915 DOI: 10.1021/acs.analchem.6b03348] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Chemical analysis of small extracellular vesicles (sEVs) circulating in body fluids holds potentials in noninvasive diagnosis of diseases and evaluation of therapeutic treatments. However, quantification of sEVs remains a challenge due to lacking of cost-effective analytical protocols. Herein we report a facile method based on size exclusion chromatography with fluorescence detection (SEC-FD) for sEVs quantification. After removal of cells and cell debris, a 0.50 mL sample (e.g., cell culture medium) is incubated with CM-Dil dye to fluorescently label sEVs. The incubation solution is then separated on a SEC column packed with Sepharose CL-4B. The eluent is monitored fluorescently at Ex553 nm/Em570 nm by using a fluorometer equipped with a 50-μL flow through cuvette. Separation efficiency of the proposed SEC-FD method was evaluated by analyzing 100 nm liposomes and albumin-FITC conjugate. Liposomes were eluted out in less than 6 min, about 10 min before albumin-FITC. A separation repeatability (RSD in retention time) of 1.4% (n = 5) was obtained for liposomes. In analysis of cell culture media, linear calibration curves based on SEC-FD peak height versus sEVs concentration were obtained with r2 value of 0.996. Intraday quantification repeatability (RSD in peak height) was 3.2% (n = 5). The detection limit was estimated to be 2.9 × 107 exosome particles/mL. The proposed assay was applied to the first study of sEVs secretion from TK6 cells cultured in serum-free medium for a culturing period from 1 to 48 h.
Collapse
Affiliation(s)
- Rui Xu
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, MS 39217, USA
| | - Austin Fitts
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, MS 39217, USA
| | - Xiangtang Li
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, MS 39217, USA
| | - Joseph Fernandes
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS 39150, USA
| | - Radhika Pochampally
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS 39150, USA
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, MS 39150, USA
| | - Jinghe Mao
- Department of Biology, Tougaloo College, Tougaloo, MS 39174, USA
| | - Yi-Ming Liu
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, MS 39217, USA
| |
Collapse
|
134
|
Štukelj R, Schara K, Bedina-Zavec A, Šuštar V, Pajnič M, Pađen L, Krek JL, Kralj-Iglič V, Mrvar-Brečko A, Janša R. Effect of shear stress in the flow through the sampling needle on concentration of nanovesicles isolated from blood. Eur J Pharm Sci 2016; 98:17-29. [PMID: 27737793 DOI: 10.1016/j.ejps.2016.10.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 09/15/2016] [Accepted: 10/04/2016] [Indexed: 12/18/2022]
Abstract
During harvesting of nanovesicles (NVs) from blood, blood cells and other particles in blood are exposed to mechanical forces which may cause activation of platelets, changes of membrane properties, cell deformation and shedding of membrane fragments. We report on the effect of shear forces imposed upon blood samples during the harvesting process, on the concentration of membrane nanovesicles in isolates from blood. Mathematical models of blood flow through the needle during sampling with vacuumtubes and with free flow were constructed, starting from the Navier-Stokes formalism. Blood was modeled as a Newtonian fluid. Work of the shear stress was calculated. In experiments, nanovesicles were isolated by repeated centrifugation (up to 17,570×g) and washing, and counted by flow cytometry. It was found that the concentration of nanovesicles in the isolates positively corresponded with the work by the shear forces in the flow of the sample through the needle. We have enhanced the effect of the shear forces by shaking the samples prior to isolation with glass beads. Imaging of isolates by scanning electron microscopy revealed closed globular structures of a similar size and shape as those obtained from unshaken plasma by repetitive centrifugation and washing. Furthermore, the sizes and shapes of NVs obtained by shaking erythrocytes corresponded to those isolated from shaken platelet-rich plasma and from unshaken platelet rich plasma, and not to those induced in erythrocytes by exogenously added amphiphiles. These results are in favor of the hypothesis that a significant pool of nanovesicles in blood isolates is created during their harvesting. The identity, shape, size and composition of NVs in isolates strongly depend on the technology of their harvesting.
Collapse
Affiliation(s)
- Roman Štukelj
- Laboratory of Clinical Biophysics, Faculty of Health Sciences, University of Ljubljana, Zdravstvena pot 5, Ljubljana SI-1000, Slovenia.
| | - Karin Schara
- Laboratory of Clinical Biophysics, Chair of Orthopaedics, Faculty of Medicine, University of Ljubljana, Zaloška 9, Ljubljana SI-1000, Slovenia.
| | - Apolonija Bedina-Zavec
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, Ljubljana SI-1000, Slovenia.
| | - Vid Šuštar
- Lymphocyte Cytoskeleton Group, Institute of Biomedicine/Pathology, BioCity, University of Turku, Tykistokatu 6B, Turku SF 20520, Finland.
| | - Manca Pajnič
- Laboratory of Clinical Biophysics, Faculty of Health Sciences, University of Ljubljana, Zdravstvena pot 5, Ljubljana SI-1000, Slovenia.
| | - Ljubiša Pađen
- Laboratory of Clinical Biophysics, Faculty of Health Sciences, University of Ljubljana, Zdravstvena pot 5, Ljubljana SI-1000, Slovenia.
| | - Judita Lea Krek
- Laboratory of Clinical Biophysics, Faculty of Health Sciences, University of Ljubljana, Zdravstvena pot 5, Ljubljana SI-1000, Slovenia.
| | - Veronika Kralj-Iglič
- Laboratory of Clinical Biophysics, Faculty of Health Sciences, University of Ljubljana, Zdravstvena pot 5, Ljubljana SI-1000, Slovenia.
| | - Anita Mrvar-Brečko
- Clinical Department of Anaesthesiology and Intensive Care of Operative Branches, Ljubljana University Medical Centre, Zaloška 7, Ljubljana SI-1000, Slovenia.
| | - Rado Janša
- Clinical Department of Gastroenterology, Ljubljana University Medical Centre, Zaloška 7, Ljubljana SI-1000, Slovenia.
| |
Collapse
|
135
|
Etayash H, McGee AR, Kaur K, Thundat T. Nanomechanical sandwich assay for multiple cancer biomarkers in breast cancer cell-derived exosomes. NANOSCALE 2016; 8:15137-41. [PMID: 27492928 DOI: 10.1039/c6nr03478k] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
The use of exosomes as cancer diagnostic biomarkers is technically limited by their size, heterogeneity and the need for extensive purification and labelling. We report the use of cantilever arrays for simultaneous detection of multiple exosomal surface-antigens with high sensitivity and selectivity. Exosomes from breast cancer were selectively identified by detecting over-expressed membrane-proteins CD24, CD63, and EGFR. Excellent selectivity however, was achieved when targeting the cell-surface proteoglycan, Glypican-1 at extraordinary limits (∼200 exosomes per mL, ∼0.1 pg mL(-1)).
Collapse
Affiliation(s)
- H Etayash
- Faculty of Pharmacy and Pharmaceutical Sciences University of Alberta, Edmonton, Alberta T6G 2E1, Canada and Department of Chemical and Materials Engineering University of Alberta, Edmonton, T6G 2V4, Alberta, Canada.
| | - A R McGee
- Department of Chemical and Materials Engineering University of Alberta, Edmonton, T6G 2V4, Alberta, Canada.
| | - K Kaur
- Faculty of Pharmacy and Pharmaceutical Sciences University of Alberta, Edmonton, Alberta T6G 2E1, Canada and Chapman University School of Pharmacy (CUSP), Harry and Diane Rinker Health Science Campus, Chapman University, Irvine, California 92618-1908, USA
| | - T Thundat
- Department of Chemical and Materials Engineering University of Alberta, Edmonton, T6G 2V4, Alberta, Canada.
| |
Collapse
|
136
|
Khatun Z, Bhat A, Sharma S, Sharma A. Elucidating diversity of exosomes: biophysical and molecular characterization methods. Nanomedicine (Lond) 2016; 11:2359-77. [PMID: 27488053 DOI: 10.2217/nnm-2016-0192] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Exosomes are cell-secreted nanovesicles present in biological fluids in normal and diseased conditions. Owing to their seminal role in cell-cell communication, emerging evidences suggest that exosomes are fundamental regulators of various diseases. Due to their potential usefulness in disease diagnosis, robust isolation and characterization of exosomes is critical in developing exosome-based assays. In the last few years, different exosome characterization methods, both biophysical and molecular, have been developed to characterize these tiny vesicles. Here, in this review we summarize: first, biophysical techniques based on spectroscopy (e.g., Raman spectroscopy, dynamic light scattering) and other principles, for example, scanning electron microscopy, atomic force microscopy; second, antibody-based molecular techniques including flow cytometry, transmission electron microscopy and third, nanotechnology-dependent exosome characterization methodologies.
Collapse
Affiliation(s)
- Zamila Khatun
- ExoCan Healthcare Technologies Ltd, L4, 400 NCL Innovation Park, Pashan, Pune 411008, India
| | - Anjali Bhat
- ExoCan Healthcare Technologies Ltd, L4, 400 NCL Innovation Park, Pashan, Pune 411008, India
| | - Shivani Sharma
- California Nanosystems, University of California, Los Angeles, CA, USA
| | - Aman Sharma
- ExoCan Healthcare Technologies Ltd, L4, 400 NCL Innovation Park, Pashan, Pune 411008, India
| |
Collapse
|
137
|
Welton JL, Brennan P, Gurney M, Webber JP, Spary LK, Carton DG, Falcón-Pérez JM, Walton SP, Mason MD, Tabi Z, Clayton A. Proteomics analysis of vesicles isolated from plasma and urine of prostate cancer patients using a multiplex, aptamer-based protein array. J Extracell Vesicles 2016; 5:31209. [PMID: 27363484 PMCID: PMC4929354 DOI: 10.3402/jev.v5.31209] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 04/11/2016] [Accepted: 04/17/2016] [Indexed: 12/28/2022] Open
Abstract
Proteomics analysis of biofluid-derived vesicles holds enormous potential for discovering non-invasive disease markers. Obtaining vesicles of sufficient quality and quantity for profiling studies has, however, been a major problem, as samples are often replete with co-isolated material that can interfere with the identification of genuine low abundance, vesicle components. Here, we used a combination of ultracentrifugation and size-exclusion chromatography to isolate and analyse vesicles of plasma or urine origin. We describe a sample-handling workflow that gives reproducible, quality vesicle isolations sufficient for subsequent protein profiling. Using a semi-quantitative aptamer-based protein array, we identified around 1,000 proteins, of which almost 400 were present at comparable quantities in plasma versus urine vesicles. Significant differences were, however, apparent with elements like HSP90, integrin αVβ5 and Contactin-1 more prevalent in urinary vesicles, while hepatocyte growth factor activator, prostate-specific antigen–antichymotrypsin complex and many others were more abundant in plasma vesicles. This was also applied to a small set of specimens collected from men with metastatic prostate cancer, highlighting several proteins with the potential to indicate treatment refractory disease. The study provides a practical platform for furthering protein profiling of vesicles in prostate cancer, and, hopefully, many other disease scenarios.
Collapse
Affiliation(s)
- Joanne Louise Welton
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom.,Velindre Cancer Centre, Cardiff, United Kingdom.,Cardiff School of Health Sciences, Cardiff Metropolitan University, Cardiff, United Kingdom
| | - Paul Brennan
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Mark Gurney
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom.,Velindre Cancer Centre, Cardiff, United Kingdom
| | - Jason Paul Webber
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom.,Velindre Cancer Centre, Cardiff, United Kingdom
| | - Lisa Kate Spary
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom.,Velindre Cancer Centre, Cardiff, United Kingdom
| | - David Gil Carton
- Metabolomics Unit, CIC bioGUNE, CIBERehd, Bizkaia Technology Park, Derio, Spain
| | | | - Sean Peter Walton
- Department of Computer Science, College of Science, Swansea University, United Kingdom
| | - Malcolm David Mason
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom.,Velindre Cancer Centre, Cardiff, United Kingdom
| | - Zsuzsanna Tabi
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom.,Velindre Cancer Centre, Cardiff, United Kingdom
| | - Aled Clayton
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom.,Velindre Cancer Centre, Cardiff, United Kingdom;
| |
Collapse
|
138
|
Stremersch S, Marro M, Pinchasik BE, Baatsen P, Hendrix A, De Smedt SC, Loza-Alvarez P, Skirtach AG, Raemdonck K, Braeckmans K. Identification of Individual Exosome-Like Vesicles by Surface Enhanced Raman Spectroscopy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:3292-301. [PMID: 27171437 DOI: 10.1002/smll.201600393] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 03/11/2016] [Indexed: 05/20/2023]
Abstract
Exosome-like vesicles (ELVs) are a novel class of biomarkers that are receiving a lot of attention for the detection of cancer at an early stage. In this study the feasibility of using a surface enhanced Raman spectroscopy (SERS) based method to distinguish between ELVs derived from different cellular origins is evaluated. A gold nanoparticle based shell is deposited on the surface of ELVs derived from cancerous and healthy cells, which enhances the Raman signal while maintaining a colloidal suspension of individual vesicles. This nanocoating allows the recording of SERS spectra from single vesicles. By using partial least squares discriminant analysis on the obtained spectra, vesicles from different origin can be distinguished, even when present in the same mixture. This proof-of-concept study paves the way for noninvasive (cancer) diagnostic tools based on exosomal SERS fingerprinting in combination with multivariate statistical analysis.
Collapse
Affiliation(s)
- Stephan Stremersch
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000, Ghent, Belgium
| | - Monica Marro
- ICFO-Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, Av. Carl Friedrich Gauss 3, 08860, Castelldefels, Barcelona, Spain
| | - Bat-El Pinchasik
- Department of Interfaces, Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1 OT Golm, 14476, Potsdam, Germany
| | - Pieter Baatsen
- EM-facility EMoNe, VIB-KULeuven Bio Imaging Core and Center for Human Genetics, KULeuven, Herestraat 49, 3000, Leuven, Belgium
| | - An Hendrix
- Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University Hospital, De Pintelaan 185, 900, Ghent, Belgium
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000, Ghent, Belgium
| | - Pablo Loza-Alvarez
- ICFO-Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, Av. Carl Friedrich Gauss 3, 08860, Castelldefels, Barcelona, Spain
| | - Andre G Skirtach
- Department of Interfaces, Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1 OT Golm, 14476, Potsdam, Germany
- Department of Molecular Biotechnology, Ghent University, Coupure Links 653, 9000, Ghent, Belgium
- Centre for Nano- and Biophotonics, Ghent University, 9000, Ghent, Belgium
| | - Koen Raemdonck
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000, Ghent, Belgium
| | - Kevin Braeckmans
- Centre for Nano- and Biophotonics, Ghent University, 9000, Ghent, Belgium
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000, Ghent, Belgium
| |
Collapse
|
139
|
Osteikoetxea X, Németh A, Sódar BW, Vukman KV, Buzás EI. Extracellular vesicles in cardiovascular disease: are they Jedi or Sith? J Physiol 2016; 594:2881-94. [PMID: 26872404 DOI: 10.1113/jp271336] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 01/06/2016] [Indexed: 12/12/2022] Open
Abstract
In the recent past, extracellular vesicles have become recognized as important players in cell biology and biomedicine. Extracellular vesicles, including exosomes, microvesicles and apoptotic bodies, are phospholipid bilayer-enclosed structures found to be secreted by most if not all cells. Extracellular vesicle secretion represents a universal and highly conserved active cellular function. Importantly, increasing evidence supports that extracellular vesicles may serve as biomarkers and therapeutic targets or tools in human diseases. Cardiovascular disease undoubtedly represents one of the most intensely studied and rapidly growing areas of the extracellular vesicle field. However, in different studies related to cardiovascular disease, extracellular vesicles have been shown to exert diverse and sometimes discordant biological effects. Therefore, it might seem a puzzle whether these vesicles are in fact beneficial or detrimental to cardiovascular health. In this review we provide a general introduction to extracellular vesicles and an overview of their biological roles in cardiovascular diseases. Furthermore, we aim to untangle the various reasons for the observed discrepancy in biological effects of extracellular vesicles in cardiovascular diseases. To this end, we provide several examples that demonstrate that the observed functional diversity is in fact due to inherent differences among various types of extracellular vesicles.
Collapse
Affiliation(s)
- Xabier Osteikoetxea
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Andrea Németh
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Barbara W Sódar
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Krisztina V Vukman
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Edit Irén Buzás
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
140
|
Pocsfalvi G, Stanly C, Fiume I, Vékey K. Chromatography and its hyphenation to mass spectrometry for extracellular vesicle analysis. J Chromatogr A 2016; 1439:26-41. [PMID: 26830636 DOI: 10.1016/j.chroma.2016.01.017] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 01/05/2016] [Accepted: 01/07/2016] [Indexed: 12/14/2022]
Abstract
Extracellular vesicles (EVs), such as exosomes, microvesicles and apoptotic bodies are released by cells, both under physiological and pathological conditions. EVs can participate in a novel type of intercellular communication and deliver cargo of nucleic acids, proteins and lipids near or to distant host cells. EV research is proceeding at a fast pace; now they start to appear as promising therapeutic targets, diagnostic tools and drug delivery systems. Isolation and analysis of EVs are prerequisites for understanding their biological roles and for their clinical exploitation. In this process chromatography and mass spectrometry (MS)-based strategies are rapidly gaining importance; and are reviewed in the present communication. Isolation and purification of EVs is mostly performed by ultracentrifugation at present. Chromatography-based strategies are gaining ground, among which affinity and size exclusion chromatography (SEC) are particularly strong contenders. Their major advantages are the relative simplicity, robustness and throughput. Affinity chromatography has the added advantage of separating EV subtypes based on molecular recognition of EV surface motifs. SEC has the advantage that isolated EVs may retain their biological activity. EVs are typically isolated in small amounts, therefore high sensitivity is required for their analysis. Study of the molecular content of EVs (all compounds beside nucleic acids) is predominantly based on liquid chromatography tandem mass spectrometry (LC-MS/MS) analysis. The chromatographic separation is mostly performed by reverse phase, nanoscale, ultra high performance LC technique. The MS analysis relying typically on nano-electrospray ionization MS/MS provides high sensitivity, selectivity and resolution, so that thousand(s) of proteins can be detected/identified/quantified in a EV sample. Beside protein identification, quantitation and characterization of protein post-translational modifications (PTMs), like glycosylation and phosphorylation are becoming feasible and increasingly important. Along with conventional LC-MS/MS, other chromatographic approaches hyphenated to MS are gaining importance for EV characterization. Hydrophilic interaction LC is used to characterize PTMs; LC-inductively coupled plasma/MS to identify metal containing molecules; while gas chromatography-MS to analyze some lipids and metabolites.
Collapse
Affiliation(s)
- Gabriella Pocsfalvi
- Mass Spectrometry and Proteomics, Institute of Biosciences and BioResources, National Research Council of Italy, Naples, Italy.
| | - Christopher Stanly
- Mass Spectrometry and Proteomics, Institute of Biosciences and BioResources, National Research Council of Italy, Naples, Italy
| | - Immacolata Fiume
- Mass Spectrometry and Proteomics, Institute of Biosciences and BioResources, National Research Council of Italy, Naples, Italy
| | - Károly Vékey
- Mass Spectrometry Proteomics Group, Institute of Organic Chemistry, Research Centre for Natural Sciences of the Hungarian Academy of Sciences, Budapest, Hungary
| |
Collapse
|
141
|
van der Pol E, Böing AN, Gool EL, Nieuwland R. Recent developments in the nomenclature, presence, isolation, detection and clinical impact of extracellular vesicles. J Thromb Haemost 2016; 14:48-56. [PMID: 26564379 DOI: 10.1111/jth.13190] [Citation(s) in RCA: 241] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Indexed: 02/06/2023]
Abstract
The research field of extracellular vesicles (EVs), such as microparticles and exosomes, is growing exponentially. The goal of this review is to provide an overview of recent developments relevant to the readers of the Journal of Thrombosis and Haemostasis. We will discuss nomenclature, the presence of EVs in fluids, methods of isolation and detection, and emerging clinical implications. Although research on EVs has been performed within the ISTH for over a decade, most of the recent research on EVs has been brought together by the International Society on Extracellular Vesicles (ISEV). To achieve an overview of recent developments, the information provided in this review comes not only from publications, but also from latest meetings of the ISEV (April 2015, Washington, DC, USA), the International Society on Advancement of Cytometry (June 2015, Glasgow, UK), and the ISTH (June 2015, Toronto, Canada).
Collapse
Affiliation(s)
- E van der Pol
- Laboratory of Experimental Clinical Chemistry, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Department of Biomedical Engineering and Physics, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - A N Böing
- Laboratory of Experimental Clinical Chemistry, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - E L Gool
- Laboratory of Experimental Clinical Chemistry, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Department of Biomedical Engineering and Physics, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - R Nieuwland
- Laboratory of Experimental Clinical Chemistry, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
142
|
Greening DW, Xu R, Ji H, Tauro BJ, Simpson RJ. A protocol for exosome isolation and characterization: evaluation of ultracentrifugation, density-gradient separation, and immunoaffinity capture methods. Methods Mol Biol 2015; 1295:179-209. [PMID: 25820723 DOI: 10.1007/978-1-4939-2550-6_15] [Citation(s) in RCA: 499] [Impact Index Per Article: 49.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Exosomes are 40-150 nm extracellular vesicles that are released from a multitude of cell types, and perform diverse cellular functions including intercellular communication, antigen presentation, and transfer of tumorigenic proteins, mRNA and miRNA. Exosomes are important regulators of the cellular niche, and their altered characteristics in many diseases, such as cancer, suggest their importance for diagnostic and therapeutic applications, and as drug delivery vehicles. Exosomes have been purified from biological fluids and in vitro cell cultures using a variety of strategies and techniques. In this chapter, we reveal the protocol and key insights into the isolation, purification and characterization of exosomes, distinct from shed microvesicles and apoptotic blebs. Using the colorectal cancer cell line LIM1863 as a cell model, a comprehensive evaluation of exosome isolation methods including ultracentrifugation (UC-Exos), OptiPrep™ density-based separation (DG-Exos), and immunoaffinity capture using anti-EpCAM-coated magnetic beads (IAC-Exos) were examined. All exosome isolation methodologies contained 40-150 nm vesicles based on electron microscopy, and positive for exosome markers (Alix, TSG101, HSP70) based on immunoblotting. This protocol employed a proteomic profiling approach to characterize the protein composition of exosomes, and label-free spectral counting to evaluate the effectiveness of each method in exosome isolation. Based on the number of MS/MS spectra identified for exosome markers and proteins associated with their biogenesis, trafficking, and release, IAC-Exos was shown to be the most effective method to isolate exosomes. However, the use of density-based separation (DG-Exos) provides significant advantages for exosome isolation when the use of immunoaffinity capture is limited (due to antibody availability and suitability of exosome markers).
Collapse
Affiliation(s)
- David W Greening
- Department of Biochemistry, La Trobe Institute for Molecular Science (LIMS), LIMS1, La Trobe University, Melbourne, VIC, 3086, Australia,
| | | | | | | | | |
Collapse
|
143
|
Smith ZJ, Lee C, Rojalin T, Carney RP, Hazari S, Knudson A, Lam K, Saari H, Ibañez EL, Viitala T, Laaksonen T, Yliperttula M, Wachsmann-Hogiu S. Single exosome study reveals subpopulations distributed among cell lines with variability related to membrane content. J Extracell Vesicles 2015; 4:28533. [PMID: 26649679 PMCID: PMC4673914 DOI: 10.3402/jev.v4.28533] [Citation(s) in RCA: 226] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 10/26/2015] [Accepted: 11/06/2015] [Indexed: 12/21/2022] Open
Abstract
Current analysis of exosomes focuses primarily on bulk analysis, where exosome-to-exosome variability cannot be assessed. In this study, we used Raman spectroscopy to study the chemical composition of single exosomes. We measured spectra of individual exosomes from 8 cell lines. Cell-line-averaged spectra varied considerably, reflecting the variation in total exosomal protein, lipid, genetic, and cytosolic content. Unexpectedly, single exosomes isolated from the same cell type also exhibited high spectral variability. Subsequent spectral analysis revealed clustering of single exosomes into 4 distinct groups that were not cell-line specific. Each group contained exosomes from multiple cell lines, and most cell lines had exosomes in multiple groups. The differences between these groups are related to chemical differences primarily due to differing membrane composition. Through a principal components analysis, we identified that the major sources of spectral variation among the exosomes were in cholesterol content, relative expression of phospholipids to cholesterol, and surface protein expression. For example, exosomes derived from cancerous versus non-cancerous cell lines can be largely separated based on their relative expression of cholesterol and phospholipids. We are the first to indicate that exosome subpopulations are shared among cell types, suggesting distributed exosome functionality. The origins of these differences are likely related to the specific role of extracellular vesicle subpopulations in both normal cell function and carcinogenesis, and they may provide diagnostic potential at the single exosome level.
Collapse
Affiliation(s)
- Zachary J Smith
- Center for Biophotonics, University of California Davis, Sacramento, CA, USA.,Department of Precision Mechanics and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui, China
| | - Changwon Lee
- Center for Biophotonics, University of California Davis, Sacramento, CA, USA
| | - Tatu Rojalin
- Center for Biophotonics, University of California Davis, Sacramento, CA, USA.,Division of Pharmaceutical Biosciences, Centre for Drug Research, University of Helsinki, Helsinki, Finland
| | - Randy P Carney
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, USA
| | - Sidhartha Hazari
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, USA
| | - Alisha Knudson
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, USA
| | - Kit Lam
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, USA
| | - Heikki Saari
- Division of Pharmaceutical Biosciences, Centre for Drug Research, University of Helsinki, Helsinki, Finland
| | - Elisa Lazaro Ibañez
- Division of Pharmaceutical Biosciences, Centre for Drug Research, University of Helsinki, Helsinki, Finland
| | - Tapani Viitala
- Division of Pharmaceutical Biosciences, Centre for Drug Research, University of Helsinki, Helsinki, Finland
| | - Timo Laaksonen
- Division of Pharmaceutical Biosciences, Centre for Drug Research, University of Helsinki, Helsinki, Finland
| | - Marjo Yliperttula
- Division of Pharmaceutical Biosciences, Centre for Drug Research, University of Helsinki, Helsinki, Finland
| | - Sebastian Wachsmann-Hogiu
- Center for Biophotonics, University of California Davis, Sacramento, CA, USA.,Department of Pathology and Laboratory Medicine, University of California Davis, Sacramento, CA, USA;
| |
Collapse
|
144
|
Höög JL, Lötvall J. Diversity of extracellular vesicles in human ejaculates revealed by cryo-electron microscopy. J Extracell Vesicles 2015; 4:28680. [PMID: 26563734 PMCID: PMC4643196 DOI: 10.3402/jev.v4.28680] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 09/04/2015] [Accepted: 10/11/2015] [Indexed: 12/19/2022] Open
Abstract
Human ejaculates contain extracellular vesicles (EVs), that to a large extent are considered to originate from the prostate gland, and are often denominated “prostasomes.” These EVs are important for human fertility, for example by promoting sperm motility and by inducing immune tolerance of the female immune system to the spermatozoa. So far, the EVs present in human ejaculate have not been studied in their native state, inside the seminal fluid without prior purification and isolation procedures. Using cryo-electron microscopy and tomography, we performed a comprehensive inventory of human ejaculate EVs. The sample was neither centrifuged, fixed, filtered or sectioned, nor were heavy metals added. Approximately 1,500 extracellular structures were imaged and categorized. The extracellular environment of human ejaculate was found to be diverse, with 5 major subcategories of EVs and 6 subcategories of extracellular membrane compartments, including lamellar bodies. Furthermore, 3 morphological features, including electron density, double membrane bilayers and coated surface, are described in all subcategories. This study reveals that the extracellular environment in human ejaculate is multifaceted. Several novel morphological EV subcategories are identified and clues to their cellular origin may be found in their morphology. This inventory is therefore important for developing future experimental approaches, and to interpret previously published data to understand the role of EVs for human male fertility.
Collapse
Affiliation(s)
- Johanna L Höög
- Krefting Research Centre, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden;
| | - Jan Lötvall
- Krefting Research Centre, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
145
|
González E, Falcón-Pérez JM. Cell-derived extracellular vesicles as a platform to identify low-invasive disease biomarkers. Expert Rev Mol Diagn 2015; 15:907-23. [DOI: 10.1586/14737159.2015.1043272] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
146
|
Lee C, Carney RP, Hazari S, Smith ZJ, Knudson A, Robertson CS, Lam KS, Wachsmann-Hogiu S. 3D plasmonic nanobowl platform for the study of exosomes in solution. NANOSCALE 2015; 7:9290-7. [PMID: 25939587 PMCID: PMC11781986 DOI: 10.1039/c5nr01333j] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Thin silver film coated nanobowl Surface Enhanced Raman Spectroscopy (SERS) substrates are used to capture exosomes in solution for SERS measurements that can provide biochemical analysis of intact and ruptured exosomes. Exosomes derived via Total Exosome Isolation Reagent (TEIR) as well as ultracentrifugation (UC) from the SKOV3 cell line were analyzed. Spectra of exosomes derived via TEIR are dominated by a signal characteristic for the TEIR kit that needs to be subtracted for all measurements. Differences in SERS spectra recorded at different times during the drying of the exosome solution are statistically analyzed with Principal Component Analysis (PCA). At the beginning of the drying process, SERS spectra of exosomes exhibit peaks characteristic for both lipids and proteins. Later on during the drying process, new SERS peaks develop, suggesting that the initially intact exosome ruptures over time. This time-dependent evolution of SERS peaks enables analysis of exosomal membrane contents and the contents inside the exosomes.
Collapse
Affiliation(s)
- Changwon Lee
- Center for Biophotonics, University of California, Davis, Sacramento, CA 95817, USA.
| | | | | | | | | | | | | | | |
Collapse
|
147
|
Emanueli C, Shearn AIU, Angelini GD, Sahoo S. Exosomes and exosomal miRNAs in cardiovascular protection and repair. Vascul Pharmacol 2015; 71:24-30. [PMID: 25869502 DOI: 10.1016/j.vph.2015.02.008] [Citation(s) in RCA: 197] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 01/21/2015] [Accepted: 02/15/2015] [Indexed: 01/12/2023]
Abstract
Cell-cell communication between cardiac and vascular cells and from stem and progenitor cells to differentiated cardiovascular cells is both an important and complex process, achieved through a diversity of mechanisms that have an impact on cardiovascular biology, disease and therapeutics. In recent years, evidence has accumulated suggesting that extracellular vesicles (EVs) are a new system of intercellular communication. EVs of different sizes are produced via different biogenesis pathways and have been shown to be released and taken up by most of known cell types, including heart and vascular cells, and stem and progenitor cells. This review will focus on exosomes, the smallest EVs (up to 100nm in diameter) identified so far. Cells can package cargoes consisting of selective lipids, proteins and RNA in exosomes and such cargoes can be shipped to recipient cells, inducing expressional and functional changes. This review focuses on exosomes and microRNAs in the context of cardiovascular disease and repair. We will describe exosome biogenesis and cargo formation and discuss the available information on in vitro and in vivo exosomes-based cell-to-cell communication relevant to cardiovascular science. The methods used in exosome research will be also described. Finally, we will address the promise of exosomes as clinical biomarkers and their impact as a biomedical tool in stem cell-based cardiovascular therapeutics.
Collapse
Affiliation(s)
- Costanza Emanueli
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Bristol, England, UK; National Heart and Lung Institute, Imperial College of London, London, England, UK.
| | - Andrew I U Shearn
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Bristol, England, UK
| | - Gianni D Angelini
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Bristol, England, UK; National Heart and Lung Institute, Imperial College of London, London, England, UK
| | - Susmita Sahoo
- Cardiovascular Research Institute, Icahn School of Medicine, Mount Sinai, NY, USA
| |
Collapse
|
148
|
Size and shape characterization of hydrated and desiccated exosomes. Anal Bioanal Chem 2015; 407:3285-301. [DOI: 10.1007/s00216-015-8535-3] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 01/26/2015] [Accepted: 02/05/2015] [Indexed: 12/21/2022]
|
149
|
van der Pol E, Coumans FAW, Sturk A, Nieuwland R, van Leeuwen TG. Refractive index determination of nanoparticles in suspension using nanoparticle tracking analysis. NANO LETTERS 2014; 14:6195-201. [PMID: 25256919 DOI: 10.1021/nl503371p] [Citation(s) in RCA: 140] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The refractive index (RI) dictates interaction between light and nanoparticles and therefore is important to health, environmental, and materials sciences. Using nanoparticle tracking analysis, we have determined the RI of heterogeneous particles <500 nm in suspension. We demonstrate feasibility of distinguishing silica and polystyrene beads based on their RI. The hitherto unknown RI of extracellular vesicles from human urine was determined at 1.37 (mean). This method enables differentiation of single nanoparticles based on their RI.
Collapse
Affiliation(s)
- Edwin van der Pol
- Biomedical Engineering and Physics, ‡ Laboratory of Experimental Clinical Chemistry, Academic Medical Center, University of Amsterdam , Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
150
|
Pocsfalvi G, Stanly C, Vilasi A, Fiume I, Tatè R, Capasso G. Employing extracellular vesicles for non-invasive renal monitoring: A captivating prospect. World J Clin Urol 2014; 3:66-80. [DOI: 10.5410/wjcu.v3.i2.66] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 06/09/2014] [Accepted: 06/27/2014] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles (EVs) are fascinating nano-sized subjects extensively studied over the recent years across several disparate disciplines. EVs are endlessly secreted into the extracellular microenvironment by most cell types under physiological and pathological conditions. EVs encompass a variety of molecular constituents from their cell of origin, such as lipids, cell specific proteins and RNAs, thus constituting an informative resource for studying molecular events at the cellular level. There are three main classes of EVs classified based on their size, content, biogenesis and biological functions: exosomes, shedding microvesicles and apoptotic bodies. Besides cell culture supernatants, biological fluids have also been shown to contain different types of EVs. Amongst the various body fluids, the study of urinary extracellular vesicles (uEVs) as a source of candidate biomarkers gained much attention, since: (1) urine can be non-invasively collected in large amounts; and (2) the isolated uEVs are stable for a relatively long period of time. Here, we review the important aspects of urinary extracellular vesicles which are fast gaining attention as a promising future tool for the non-invasive monitoring of urinary tract. Recent advancements in the purification and analysis of uEVs and collection of their constituents in rapidly developing public databases, allow their better exploitation in molecular diagnostics. As a result, a growing number of studies have shown that changes in expression profile at the RNA and/or protein levels of uEVs reveal the molecular architectures of underlying key pathophysiological events of different clinically important diseases with kidney involvement.
Collapse
|