Publications
& News

Minimal information for studies of extracellular vesicles: A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines.

1. January 2018

JOURNAL OF EXTRACELLULAR VESICLES
2018, VOL. 7, 1535750 https://doi.org/10.1080/20013078.2018.1535750
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
Clotilde Théry 103£, Kenneth W Witwer 217,218&£, Elena Aikawa19,79£, Maria Jose Alcaraz112,
Johnathon D Anderson288, Ramaroson Andriantsitohaina97, Anna Antoniou70,265, Tanina Arab257,
Fabienne Archer318, Georgia K Atkin-Smith131, D Craig Ayre15,158, Jean-Marie Bach254, Daniel Bachurski301, Hossein Baharvand195,353, Leonora Balaj143, Shawn Baldacchino321, Natalie N Bauer354, Amy A Baxter131,
Mary Bebawy357, Carla Beckham350, Apolonija Bedina Zavec165, Abderrahim Benmoussa260, Anna C Berardi179, Paolo Bergese39,111,283, Ewa Bielska282, Cherie Blenkiron277&, Sylwia Bobis-Wozowicz119, Eric Boilard260,
Wilfrid Boireau58, Antonella Bongiovanni106, Francesc E Borràs72,73,250, Steffi Bosch254, Chantal M Boulanger100,261£, Xandra Breakefield140, Andrew M Breglio92,169, Meadhbh Á Brennan82,144,258, David R Brigstock174,221,
Alain Brisson238$, Marike LD Broekman78,134,142, Jacqueline F Bromberg155,379, Paulina Bryl-Górecka136,
Shilpa Buch334, Amy H Buck305, Dylan Burger128,180,337, Sara Busatto148,283, Dominik Buschmann212,
Benedetta Bussolati360, Edit I Buzás160,201&£, James Bryan Byrd330, Giovanni Camussi359£, David RF Carter181, Sarah Caruso131, Lawrence W Chamley279, Yu-Ting Chang170, Amrita Datta Chaudhuri218, Chihchen Chen171,172, Shuai Chen133, Lesley Cheng131, Andrew R Chin25, Aled Clayton23, Stefano P Clerici239, Alex Cocks23,
Emanuele Cocucci220,222£, Robert J Coffey373, Anabela Cordeiro-da-Silva346, Yvonne Couch340,
Frank AW Coumans7$, Beth Coyle227, Rossella Crescitelli308, Miria Ferreira Criado352, Crislyn D’Souza-Schorey335, Saumya Das141, Paola de Candia116, Eliezer F De Santana Junior225, Olivier De Wever22,75,
Hernando A del Portillo101,104,117, Tanguy Demaret256, Sarah Deville262,377, Andrew Devitt12, Bert Dhondt22,74,75, Dolores Di Vizio25&£, Lothar C Dieterich49, Vincenza Dolo315, Ana Paula Dominguez Rubio243,
Massimo Dominici234,333#, Mauricio R Dourado298,338, Tom AP Driedonks369, Filipe V Duarte53,
Heather M Duncan150,152, Ramon M Eichenberger120, Karin Ekström306, Samir EL Andaloussi51,127, Celine Elie- Caille58, Uta Erdbrügger366&, Juan M Falcón-Pérez32,94&, Farah Fatima351, Jason E Fish233,362, Miguel Flores- Bellver302, András Försönits201, Annie Frelet-Barrand58, Fabia Fricke68,267, Gregor Fuhrmann86,87,197,
Susanne Gabrielsson126, Ana Gámez-Valero72,251, Chris Gardiner264&, Kathrin Gärtner85, Raphael Gaudin99,259, Yong Song Gho187£, Bernd Giebel266#, Caroline Gilbert260, Mario Gimona183, Ilaria Giusti315,
Deborah CI Goberdhan339, André Görgens51,123,266$, Sharon M Gorski16,204, David W Greening131,
Julia Christina Gross270,271, Alice Gualerzi115, Gopal N Gupta135, Dakota Gustafson362, Aase Handberg2,4,
Reka A Haraszti325, Paul Harrison281, Hargita Hegyesi201, An Hendrix22,75, Andrew F Hill131&£,
Fred H Hochberg200,293, Karl F Hoffmann6, Beth Holder95,159, Harry Holthofer263£, Baharak Hosseinkhani83, Guoku Hu334, Yiyao Huang162,217, Veronica Huber61, Stuart Hunt229, Ahmed Gamal-Eldin Ibrahim26,
Tsuneya Ikezu18, Jameel M Inal313, Mustafa Isin118, Alena Ivanova69, Hannah K Jackson227, Soren Jacobsen38,304, Steven M Jay324, Muthuvel Jayachandran145, Guido Jenster47, Lanzhou Jiang131, Suzanne M Johnson322, Jennifer C Jones166$, Ambrose Jong30,355, Tijana Jovanovic-Talisman34, Stephanie Jung71, Raghu Kalluri358&, Shin-ichi Kano219, Sukhbir Kaur167, Yumi Kawamura164,365, Evan T Keller327,331, Delaram Khamari201,
CONTACT Clotilde Théry Clotilde.Thery@curie.fr Institut Curie/INSERM U932, 26 rue d’Ulm, 75005 Paris, France; Kenneth W Witwer kwitwer1@jhmi.edu 733 North Broadway, MRB 829 Baltimore, MD 21205, USA
*These authors are co-corresponding authors and contributed equally to this work. All authors, except for the corresponding authors,are listed alphabetically.
This article was first drafted begining in late 2017 and finalized in September, 2018 with several rounds of feedback from the ISEV Executive Board and JEV Editorial Board.
&The following authors are members of the 2016-2018 and/or 2018-2020 ISEV Board of Directors: Cherie Blenkiron, Edit I Buzas, Dolores Di Vizio, Uta Erdbrügger, Juan M Falcón-Pérez, Chris Gardiner, Andrew F Hill (current President), Raghu Kalluri, Jan Lötvall (past President), Malene Møller Jørgensen, Rienk Nieuwland, Lorraine O’Driscoll, Susmita Sahoo, Carolina Soekmadji, Hidetoshi Tahara, Ana Claudia Torrecilhas, Marca HM Wauben, Alissa Weaver, Kenneth W Witwer, Hang (Hubert) Yin, Lei Zheng.
£The following authors are Editors of the Journal of Extracellular Vesicles: Editors-in-Chief: Clotilde Théry, Peter Quesenberry, Yong Song Gho; Associate Editors: Elena Aikawa, Chantal Boulanger, Edit I Buzas, Giovanni Camussi, Emanuele Cocucci, Dolores Di Vizio, Andrew F Hill, Harry Holthofer, Kwang Pyo Kim, Eva-Maria Krämer- Albers, Saara Laitinen, Cecilia Lässer, Suresh Mathivanan, Esther Nolte-’t Hoen, Takahiro Ochiya, Hector Peinado, Janusz Rak, Neta Regev-Rudzki, Raymond Schiffelers, Marca HM Wauben, Kenneth W Witwer.

Authors who are members of the International Society for Cell and Gene Therapy (ISCT) Exosomes Scientific Committee include Sai Kiang Lim (Co-Chair), Bernd Giebel (Co-Chair), Luis A Ortiz, Donald G Phinney and Daniel J Weiss (ISCT Chief Scientific Officer). Massimo Dominici is Past President of ISCT.

$Authors who are members of the joined ISEV-ISAC (International Society for Advancement of Cytometry)-ISTH (International Society on Thrombosis and Haemostasis) EV Flow Cytometry Working group are: Alain Brisson, Frank AW Coumans, André Görgens, Jennifer C Jones, Joanne Lannigan, Sten Libregts, François Mullier, Rienk Nieuwland, John P Nolan, Marca HM Wauben, Joshua A Welsh.
© 2018 The Author(s). Published by Informa UK Limited, trading as Taylor & Francis Group on behalf of The International Society for Extracellular Vesicles.
This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial License (http://creativecommons.org/licenses/by-nc/4.0/), which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

2 C. THÉRY AND K. W. WITWER
Elena Khomyakova45,56, Anastasia Khvorova325, Peter Kierulf178, Kwang Pyo Kim130£, Thomas Kislinger188,363, Mikael Klingeborn43, David J Klinke II381,382, Miroslaw Kornek66,196, Maja M Kosanović280, Árpád Ferenc Kovács201, Eva-Maria Krämer-Albers320£, Susanne Krasemann273, Mirja Krause90, Igor V Kurochkin28, Gina D Kusuma90,156, Sören Kuypers84, Saara Laitinen59£, Scott M Langevin33,300, Lucia R Languino232, Joanne Lannigan367$,
Cecilia Lässer308£, Louise C Laurent294, Gregory Lavieu103, Elisa Lázaro-Ibáñez14, Soazig Le Lay97, Myung-
Shin Lee50, Yi Xin Fiona Lee62, Debora S Lemos57, Metka Lenassi317, Aleksandra Leszczynska295, Isaac TS Li285, Ke Liao334, Sten F Libregts297$, Erzsebet Ligeti202, Rebecca Lim90,156, Sai Kiang Lim107#, Aija Linē132,
Karen Linnemannstöns270,271, Alicia Llorente177, Catherine A Lombard256, Magdalena J Lorenowicz370,
Ákos M Lörincz202, Jan Lötvall308&, Jason Lovett210, Michelle C Lowry235, Xavier Loyer100,261, Quan Lu81,
Barbara Lukomska157, Taral R Lunavat121, Sybren LN Maas371,372, Harmeet Malhi149, Antonio Marcilla252,253, Jacopo Mariani249, Javier Mariscal25, Elena S Martens-Uzunova47, Lorena Martin-Jaular103, M Carmen Martinez97, Vilma Regina Martins1, Mathilde Mathieu103, Suresh Mathivanan131£, Marco Maugeri309, Lynda K McGinnis356, Mark J McVey203,361, David G Meckes, Jr60, Katie L Meehan223, Inge Mertens276,377, Valentina R Minciacchi63, Andreas Möller189, Malene Møller Jørgensen3,52&, Aizea Morales-Kastresana166, Jess Morhayim48,
François Mullier161,255$, Maurizio Muraca342, Luca Musante366, Veronika Mussack212, Dillon C Muth217,
Kathryn H Myburgh210, Tanbir Najrana20, Muhammad Nawaz309, Irina Nazarenko67,154, Peter Nejsum5,
Christian Neri205, Tommaso Neri345, Rienk Nieuwland7&$, Leonardo Nimrichter247, John P Nolan200$,
Esther NM Nolte-’t Hoen369£, Nicole Noren Hooten168, Lorraine O’Driscoll235&, Tina O’Grady316, Ana O’Loghlen190, Takahiro Ochiya163£, Martin Olivier153, Alberto Ortiz93,206,242, Luis A Ortiz76#, Xabier Osteikoetxea13,
Ole Østergaard209,303, Matias Ostrowski286, Jaesung Park187, D. Michiel Pegtel11, Hector Peinado207£,
Francesca Perut114, Michael W Pfaffl212, Donald G Phinney224#, Bartijn CH Pieters191, Ryan C Pink181,
David S Pisetsky42,44, Elke Pogge von Strandmann184, Iva Polakovicova185,186, Ivan KH Poon131, Bonita H Powell217, Ilaria Prada37, Lynn Pulliam296,375, Peter Quesenberry231, Annalisa Radeghieri39,283, Robert L Raffai41,296,
Stefania Raimondo343, Janusz Rak151,153£, Marcel I Ramirez110,245, Graça Raposo102, Morsi S Rayyan326,
Neta Regev-Rudzki380£, Franz L Ricklefs272, Paul D Robbins332, David D Roberts167, Silvia C Rodrigues36,53,
Eva Rohde182,183,208, Sophie Rome319, Kasper MA Rouschop137, Aurelia Rughetti199, Ashley E Russell383,
Paula Saá10, Susmita Sahoo91&, Edison Salas-Huenuleo8,299, Catherine Sánchez35, Julie A Saugstad175,
Meike J Saul213, Raymond M Schiffelers275£, Raphael Schneider362,364, Tine Hiorth Schøyen217, Aaron Scott284, Eriomina Shahaj61, Shivani Sharma289,291,292, Olga Shatnyeva14, Faezeh Shekari195, Ganesh Vilas Shelke307,308, Ashok K Shetty194,214, Kiyotaka Shiba21, Pia R-M Siljander311,312, Andreia M Silva96,348,349, Agata Skowronek138, Orman L Snyder II122, Rodrigo Pedro Soares193, Barbara W Sódar201, Carolina Soekmadji189,228&, Javier Sotillo120, Philip D Stahl378, Willem Stoorvogel369, Shannon L Stott80,139, Erwin F Strasser55, Simon Swift278,
Hidetoshi Tahara88&, Muneesh Tewari327,328,329, Kate Timms323, Swasti Tiwari65,198, Rochelle Tixeira131,
Mercedes Tkach103, Wei Seong Toh173, Richard Tomasini98, Ana Claudia Torrecilhas240&, Juan Pablo Tosar105,244, Vasilis Toxavidis17, Lorena Urbanelli344, Pieter Vader275, Bas WM van Balkom274, Susanne G van der Grein369, Jan Van Deun22,75, Martijn JC van Herwijnen369, Kendall Van Keuren-Jensen215, Guillaume van Niel27,
Martin E van Royen46, Andre J van Wijnen146, M Helena Vasconcelos113,347,348, Ivan J Vechetti Jr314,
Tiago D Veit248, Laura J Vella216,226, Émilie Velot237, Frederik J Verweij27, Beate Vestad176,192,336,
Jose L Viñas128,180,337, Tamás Visnovitz201, Krisztina V Vukman201, Jessica Wahlgren310, Dionysios C Watson24,269, Marca HM Wauben369&£$, Alissa Weaver374&, Jason P Webber23, Viktoria Weber40, Ann M Wehman368,
Daniel J Weiss230#, Joshua A Welsh166$, Sebastian Wendt268, Asa M Wheelock125, Zoltán Wiener201,
Leonie Witte270,271, Joy Wolfram31,89,147, Angeliki Xagorari64, Patricia Xander246, Jing Xu16,204, Xiaomei Yan384, María Yáñez-Mó29,241, Hang Yin236&, Yuana Yuana211, Valentina Zappulli341, Jana Zarubova108,109,290,
Vytautas Žėkas376, Jian-ye Zhang77, Zezhou Zhao217, Lei Zheng162&, Alexander R Zheutlin326, Antje M Zickler124, Pascale Zimmermann9,129, Angela M Zivkovic287, Davide Zocco54 and Ewa K Zuba-Surma119
1A.C.Camargo Cancer Center, São Paulo, Brazil; 2Aalborg University Hospital, Department of Clinical Biochemistry, Aalborg, Denmark; 3Aalborg University Hospital, Department of Clinical Immunology, Aalborg, Denmark; 4Aalborg University, Clinical Institute, Aalborg, Denmark; 5Aarhus University, Department of Clinical Medicine, Aarhus, Denmark; 6Aberystwyth University, Institute of Biological, Environmental and Rural Sciences (IBERS), Aberystwyth, United Kingdom; 7Academic Medical Centre of the University of Amsterdam, Department of Clinical Chemistry and Vesicle Observation Centre, Amsterdam, The Netherlands; 8Advanced Center for Chronic Diseases, Santiago, Chile; 9Aix-Marseille Université, Institut Paoli-Calmettes, INSERM U1068, CNRS UMR7258, Centre de Recherche en Cancérologie de Marseille, Marseille, France; 10American Red Cross, Scientific Affairs, Gaithersburg, MD, USA; 11Amsterdam University Medical Centers, Department of Pathology, Amsterdam, The Netherlands; 12Aston University, School of Life & Health Sciences, Birmingham, UK; 13AstraZeneca, Discovery Sciences, IMED Biotech Unit, Cambridge, UK; 14AstraZeneca, Discovery Sciences, IMED Biotech Unit, Gothenburg, Sweden; 15Atlantic Cancer Research Institute, Moncton, Canada; 16BC Cancer, Canada’s Michael Smith Genome Sciences Centre, Vancouver, Canada; 17Beth Israel Deaconess Medical Center, Boston, MA, USA; 18Boston University School of Medicine, Boston, MA, USA; 19Brigham and Women’s Hospital, Center for Interdisciplinary Cardiovascular Sciences, Boston, MA, USA; 20Brown University, Women and Infants Hospital, Providence, RI, USA; 21Cancer Institute of JFCR, Tokyo, Japan; 22Cancer Research Institute Ghent, Ghent, Belgium; 23Cardiff University, School of Medicine, Cardiff, UK; 24Case Western Reserve University, Department of Medicine, Cleveland, OH, USA; 25Cedars-Sinai Medical Center, Los

Angeles, CA, USA; 26Cedars-Sinai Medical Center, Smidt Heart Institute, Los Angeles, CA, USA; 27Center for Psychiatry and Neuroscience, INSERM U894, Paris, France; 28Central Research Laboratories, Sysmex Co., Kobe, Japan; 29Centro de Biología Molecular Severo Ochoa, Instituto de Investigación Sanitaria la Princesa (IIS-IP), Madrid, Spain; 30Children’s Hospital of Los Angeles, Los Angeles, CA, USA; 31Chinese Academy of Sciences, Wenzhou Institute of Biomaterials and Engineering, Wenzhou, China; 32CIC bioGUNE, CIBERehd, Exosomes Laboratory & Metabolomics Platform, Derio, Spain; 33Cincinnati Cancer Center, Cincinnati, OH, USA; 34City of Hope Comprehensive Cancer Center, Beckman Research Institute, Department of Molecular Medicine, Duarte, CA, USA; 35Clínica las Condes, Extracellular Vesicles in Personalized Medicine Group, Santiago, Chile; 36CNC, Coimbra, Portugal; 37CNR Institute of Neuroscience, Milan, Italy; 38Copenhagen Lupus and Vasculitis Clinic, Section 4242 – Rigshospitalet, Copenhagen, Denmark; 39CSGI – Research Center for Colloids and Nanoscience, Florence, Italy; 40Danube University Krems, Department for Biomedical Research and Christian Doppler Laboratory for Innovative Therapy Approaches in Sepsis, Krems an der Donau, Austria; 41Department of Veterans Affairs, San Francisco, CA, USA; 42Duke University Medical Center, Departments of Medicine and Immunology, Durham, NC, USA; 43Duke University, Department of Ophthalmology, Durham, NC, USA; 44Durham VAMC, Medical Research Service, Durham, NC, USA; 45École normale supérieure, Paris, France; 46Department of Pathology, Erasmus MC, Erasmus Optical Imaging Centre, Rotterdam, The Netherlands; 47Department of Urology, Rotterdam, The Netherlands; 48Erasmus MC, Rotterdam, The Netherlands; 49ETH Zurich, Institute of Pharmaceutical Sciences, Zurich, Switzerland; 50Eulji University, School of Medicine, Daejeon, South Korea; 51Evox Therapeutics Limited, Oxford, UK; 52EVSEARCH.DK, Denmark; 53Exogenus Therapeutics, Cantanhede, Portugal; 54Exosomics Siena SpA, Siena, Italy; 55FAU Erlangen-Nuremberg, Transfusion and Haemostaseology Department, Erlangen, Germany; 56Federal Research and Clinical Center of Physical-Chemical Medicine, Moscow, Russia; 57Federal University of Paraná, Department of Genetics, Human Molecular Genetics Laboratory, Curitiba, Brazil; 58FEMTO-ST Institute, UBFC, CNRS, ENSMM, UTBM, Besançon, France; 59Finnish Red Cross Blood Service, Research and Development, Helsinki, Finland; 60Florida State University College of Medicine, Department of Biomedical Sciences, Tallahassee, FL, USA; 61Fondazione IRCCS Istituto Nazionale dei Tumori, Unit of Immunotherapy of Human Tumors, Milan, Italy; 62Genome Institute of Singapore, ASTAR, Singapore; 63Georg-Speyer-Haus Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany; 64George Papanicolaou Hospital, Public Cord Blood Bank, Department of Haematology – BMT Unit, Thessaloniki, Greece; 65Georgetown University, Department of Medicine, Washington, DC, USA; 66German Armed Forces Central Hospital, Department of General, Visceral and Thoracic Surgery, Koblenz, Germany; 67German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany; 68German Cancer Research Center (DKFZ), Clinical Cooperation Unit Applied Tumor Biology, Heidelberg, Germany; 69German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, Heidelberg, Germany; 70German Centre for Neurodegenerative Diseases (DZNE), Bonn, Germany; 71German Research Center for Environmental Health, Institute for Virology, Munich, Germany; 72Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, REMAR- IVECAT Group, Badalona, Spain; 73Germans Trias i Pujol University Hospital, Nephrology Service, Badalona, Spain; 74Ghent University Hospital, Department of Urology, Ghent, Belgium; 75Ghent University, Department of Radiation Oncology and Experimental Cancer Research, Laboratory of Experimental Cancer Research, Ghent, Belgium; 76Graduate School of Public Health at the University of Pittsburgh, Division of Occupational and Environmental Medicine, Pittsburgh, PA, USA; 77Guangzhou Medical University, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology, Guangzhou, China; 78Haaglanden Medical Center, Department of Neurosurgery, The Hague, The Netherlands; 79Harvard Medical School, Cardiovascular Medicine, Boston, MA, USA; 80Harvard Medical School, Department of Medicine, Boston, MA, USA; 81Harvard University, Harvard T.H. Chan School of Public Health, Boston, MA, USA; 82Harvard University, School of Engineering and Applied Sciences, Cambridge, MA, USA; 83Hasselt University, Biomedical Research Institute (BIOMED), Department of Medicine and Life Sciences, Hasselt, Belgium; 84Hasselt University, Biomedical Research Institute (BIOMED), Hasselt, Belgium; 85Helmholtz Center Munich German Research Center for Environmental Health, Research Unit Gene Vectors, Munich, Germany; 86Helmholtz-Centre for Infection Research, Braunschweig, Germany; 87Helmholtz-Institute for Pharmaceutical Research Saarland, Saarbrücken, Germany; 88Hiroshima University, Institute of Biomedical & Health Sciences, Department of Cellular and Molecular Biology, Hiroshima, Japan; 89Houston Methodist Research Institute, Department of Nanomedicine, Houston, TX, USA; 90Hudson Institute of Medical Research, Melbourne, Australia; 91Icahn School of Medicine at Mount Sinai, Department of Medicine, Cardiology, New York City, NY, USA; 92Icahn School of Medicine at Mount Sinai, New York City, NY, USA; 93IIS-Fundacion Jimenez Diaz-UAM, Department of Nephrology and Hypertension, Madrid, Spain; 94IKERBASQUE Research Science Foundation, Bilbao, Spain; 95Imperial College London, London, UK; 96INEB – Instituto de Engenharia Biomédica, Porto, Portugal; 97INSERM U1063, Université d’Angers, CHU d’Angers, Angers, France; 98INSERM U1068, Aix Marseille University, CNRS UMR7258, Marseille, France; 99INSERM U1110, Strasbourg, France; 100INSERM UMR-S 970, Paris Cardiovascular Research Center, Paris, France; 101Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain; 102Institut Curie, CNRS UMR144, PSL Research University, Paris, France; 103Institut Curie, INSERM U932, PSL Research University, Paris, France; 104Institut d’Investigació Germans Trias i Pujol (IGTP), PVREX group, Badalona, Spain; 105Institut Pasteur de Montevideo, Functional Genomics Unit, Montevideo, Uruguay; 106Institute of Biomedicine and Molecular Immunology (IBIM), National Research Council (CNR) of Italy, Palermo, Italy; 107Institute of Medical Biology (IMB), Agency for Science and Technology (ASTAR), Singapore; 108Institute of Physiology CAS, Department of Biomaterials and Tissue Engineering, BIOCEV, Vestec, Czech Republic; 109Institute of Physiology CAS, Department of Biomaterials and Tissue Engineering, Prague, Czech Republic; 110Instituto Oswaldo Cruz, Rio de Janeiro, Brazil; 111INSTM – National Interuniversity Consortium of Materials Science and Technology, Florence, Italy; 112Interuniversity Research Institute for Molecular Recognition and Technological Development (IDM), University of Valencia, Polytechnic University of Valencia, Valencia, Spain; 113IPATIMUP, Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal; 114IRCCS – Istituto Ortopedico Rizzoli, Laboratory for Orthopaedic Pathophysiology and Regenerative Medicine, Bologna, Italy; 115IRCCS Fondazione Don Carlo Gnocchi, Laboratory of Nanomedicine and Clinical Biophotonics (LABION), Milan, Italy; 116IRCCS MultiMedica, Milan, Italy; 117ISGlobal, Hospital Clínic – Universitat de Barcelona, PVREX Group, Barcelona, Spain; 118Istanbul University Oncology Institute, Basic Oncology Department, Istanbul, Turkey; 119Jagiellonian University, Faculty of Biochemistry, Biophysics and Biotechnology, Department of Cell Biology, Kraków, Poland; 120James Cook University, Australian Institute of Tropical Health and Medicine, Centre for Biodiscovery and Molecular Development of Therapeutics, Cairns, Australia; 121K.G. Jebsen Brain Tumor Research Centre, Department of Biomedicine, University of Bergen, Bergen, Norway; 122Kansas State University, College of Veterinary Medicine, Manhattan, KS, USA; 123Karolinska Institute, Clinical Research Center, Department of Laboratory Medicine, Stockholm, Sweden; 124Karolinska Institute, Clinical Research Center, Unit for Molecular Cell and Gene Therapy Science, Stockholm, Sweden; 125Karolinska Institute, Department of Medicine and Center for Molecular Medicine, Respiratory Medicine Unit, Stockholm, Sweden; 126Karolinska Institute, Department of Medicine Solna, Division for Immunology and Allergy, Stockholm, Sweden; 127Karolinska Institute, Stockholm, Sweden; 128Kidney Research Centre, Ottawa, Canada; 129KU Leuven (Leuven University), Department of Human Genetics, Leuven, Belgium; 130Kyung Hee University, Department of Applied Chemistry, Yongin, Korea; 131La Trobe University, La Trobe Institute for Molecular
JOURNAL OF EXTRACELLULAR VESICLES 3

4 C. THÉRY AND K. W. WITWER
Science, Department of Biochemistry and Genetics, Bundoora, Australia; 132Latvian Biomedical Research and Study Centre, Riga, Latvia; 133Leibniz Institute for Farm Animal Biology (FBN), Institute of Reproductive Biology, Dummerstorf, Germany; 134Leiden University Medical Center, Department of Neurosurgery, Leiden, The Netherlands; 135Loyola University Chicago, Department of Urology, Maywood, IL, USA; 136Lund University, Department of Cardiology, Lund, Sweden; 137Maastricht University, GROW, School for Oncology and Developmental Biology, Maastricht Radiation Oncology (MaastRO) Lab, Maastricht, The Netherlands; 138Maria Sklodowska-Curie Institute – Oncology Center, Gliwice Branch, Gliwice, Poland; 139Massachusetts General Cancer Center, Boston, MA, USA; 140Massachusetts General Hospital and Neuroscience Program, Harvard Medical School, Department of Neurology and Radiology, Boston, MA, USA; 141Massachusetts General Hospital, Boston, MA, USA; 142Massachusetts General Hospital, Department of Neurology, Boston, MA, USA; 143Massachusetts General Hospital, Department of Neurosurgery, Boston, MA, USA; 144Massachusetts General Hospital, Harvard Medical School, Department of Neurology, Boston, MA, USA; 145Mayo Clinic, College of Medicine, Department of Physiology and Biomedical Engineering, Rochester, MN, USA; 146Mayo Clinic, Department of Orthopedic Surgery, Rochester, MN, USA; 147Mayo Clinic, Department of Transplantation Medicine/ Department of Physiology and Biomedical Engineering, Jacksonville, FL, USA; 148Mayo Clinic, Department of Transplantation, Jacksonville, FL, USA; 149Mayo Clinic, Rochester, MN, USA; 150McGill University, Division of Experimental Medicine, Montreal, Canada; 151McGill University, Montreal, Canada; 152McGill University, The Research Institute of the McGill University Health Centre, Child Health and Human Development Program, Montreal, Canada; 153McGill University, The Research Institute of the McGill University Health Centre, Montreal, Canada; 154Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Institute for Infection Prevention and Hospital Epidemiology, Freiburg, Germany; 155Memorial Sloan Kettering Cancer Center, Department of Medicine, New York City, NY, USA; 156Monash University, Melbourne, Australia; 157Mossakowski Medical Research Centre, NeuroRepair Department, Warsaw, Poland; 158Mount Allison University, Department of Chemistry and Biochemistry, Sackville, Canada; 159MRC The Gambia, Fajara, The Gambia; 160MTA-SE Immuno-Proteogenomics Research Groups, Budapest, Hungary; 161Namur Thrombosis and Hemostasis Center (NTHC), NARILIS, Namur, Belgium; 162Nanfang Hospital, Southern Medical University, Department of Clinical Laboratory Medicine, Guangzhou, China; 163National Cancer Center Research Institute, Division of Molecular and Cellular Medicine, Tokyo, Japan; 164National Cancer Center Research Institute, Tokyo, Japan; 165National Institute of Chemistry, Department of Molecular Biology and Nanobiotechnology, Ljubljana, Slovenia; 166National Institutes of Health, National Cancer Institute, Center for Cancer Research, Bethesda, MD, USA; 167National Institutes of Health, National Cancer Institute, Center for Cancer Research, Laboratory of Pathology, Bethesda, MD, USA; 168National Institutes of Health, National Institute on Aging, Baltimore, MD, USA; 169National Institutes of Health, National Institute on Deafness and Other Communication Disorders, Bethesda, MD, USA; 170National Taiwan University Hospital, Department of Internal Medicine, Taipei, Taiwan; 171National Tsing Hua University, Department of Power Mechanical Engineering, Hsinchu, Taiwan; 172National Tsing Hua University, Institute of Nanoengineering and Microsystems, Hsinchu, Taiwan; 173National University of Singapore, Faculty of Dentistry, Singapore; 174Nationwide Children’s Hospital, Columbus, OH, USA; 175Oregon Health & Science University, Department of Anesthesiology & Perioperative Medicine, Portland, OR, USA; 176Oslo University Hospital Rikshospitalet, Research Institute of Internal Medicine, Oslo, Norway; 177Oslo University Hospital-The Norwegian Radium Hospital, Institute for Cancer Research, Department of Molecular Cell Biology, Oslo, Norway; 178Oslo University Hospital, Department of Medical Biochemistry, Blood Cell Research Group, Oslo, Norway; 179Ospedale Santo Spirito, Pescara, Italy; 180Ottawa Hospital Research Institute, Ottawa, Canada; 181Oxford Brookes University, Department of Biological and Medical Sciences, Oxford, UK; 182Paracelsus Medical University, Department of Transfusion Medicine, Salzburg, Austria; 183Paracelsus Medical University, GMP Unit, Salzburg, Austria; 184Philipps University Marburg, Experimental Tumor Biology, Marburg, Germany; 185Pontificia Universidad Católica de Chile, Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile; 186Pontificia Universidad Católica de Chile, Faculty of Medicine, Department of Hematology-Oncology, Santiago, Chile; 187POSTECH (Pohang University of Science and Technology), Department of Life Sciences, Pohang, South Korea; 188Princess Margaret Cancer Centre, University Health Network, Toronto, Canada; 189QIMR Berghofer Medical Research Institute, Herston, Australia; 190Queen Mary University of London, Blizard Institute, Epigenetics & Cellular Senescence Group, London, UK; 191Radboud University Medical Center, Department of Rheumatology, Nijmegen, The Netherlands; 192Regional Research Network on Extracellular Vesicles, RRNEV, Oslo, Norway; 193René Rachou Institute/FIOCRUZ, Belo Horizonte, Brazil; 194Research Service, Olin E. Teague Veterans’ Medical Center, Temple, TX, USA; 195Royan Institute for Stem Cell Biology and Technology, ACECR, Cell Science Research Center, Department of Stem Cells and Developmental Biology, Tehran, Iran; 196Saarland University Medical Center, Department of Medicine II, Homburg, Germany; 197Saarland University, Saarbrücken, Germany; 198Sanjay Gandhi Postgraduate Institute of Medical Sciences, Department of Molecular Medicine & Biotechnology, Lucknow, India; 199Sapienza University of Rome, Department of Experimental Medicine, Rome, Italy; 200Scintillon Institute, La Jolla, CA, USA; 201Semmelweis University, Department of Genetics, Cell- and Immunobiology, Budapest, Hungary; 202Semmelweis University, Department of Physiology, Budapest, Hungary; 203SickKids Hospital, Department of Anesthesia and Pain Medicine, Toronto, Canada; 204Simon Fraser University, Department of Molecular Biology and Biochemistry, Burnaby, Canada; 205Sorbonne 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; 206Spanish Kidney Research Network, REDINREN, Madrid, Spain; 207Spanish National Cancer Research Center (CNIO), Molecular Oncology Programme, Microenvironment and Metastasis Laboratory, Madrid, Spain; 208Spinal Cord Injury & Tissue Regeneration Center Salzburg (SCI- TReCS), Salzburg, Austria; 209Statens Serum Institut, Department of Autoimmunology and Biomarkers, Copenhagen, Denmark; 210Stellenbosch University, Department of Physiological Sciences, Stellenbosch, South Africa; 211Technical University Eindhoven, Faculty Biomedical Technology, Eindhoven, The Netherlands; 212Technical University of Munich, TUM School of Life Sciences Weihenstephan, Division of Animal Physiology and Immunology, Freising, Germany; 213Technische Universität Darmstadt, Department of Biology, Darmstadt, Germany; 214Texas A&M University College of Medicine, Institute for Regenerative Medicine and Department of Molecular and Cellular Medicine, College Station, TX, USA; 215TGen, Neurogenomics Division, Phoenix, AZ, USA; 216The Florey Institute of Neuroscience and Mental Health, Melbourne, Australia; 217The Johns Hopkins University School of Medicine, Department of Molecular and Comparative Pathobiology, Baltimore, MD, USA; 218The Johns Hopkins University School of Medicine, Department of Neurology, Baltimore, MD, USA; 219The Johns Hopkins University School of Medicine, Department of Psychiatry and Behavioral Sciences, Baltimore, MD, USA; 220The Ohio State University, College of Pharmacy, Division of Pharmaceutics and Pharmaceutical Chemistry, Columbus, OH, USA; 221The Ohio State University, Columbus, OH, USA; 222The Ohio State University, Comprehensive Cancer Center, Columbus, OH, USA; 223The School of Biomedical Sciences, University of Western Australia, Perth, Australia; 224The Scripps Research Institute-Scripps Florida, Department of Molecular Medicine, Jupiter, FL, USA; 225The Sociedade Beneficente Israelita Brasileira Albert Einstein, São Paulo, Brazil; 226The University of Melbourne, The Department of Medicine, Melbourne, Australia; 227The University of Nottingham, School of Medicine, Children’s Brain Tumour Research Centre, Nottingham, UK; 228The University of Queensland, Brisbane, Australia; 229The University of Sheffield, Sheffield, UK; 230The University of Vermont Medical Center, Department of Medicine, Burlington, VT, USA; 231The Warren Alpert Medical School of Brown University, Department of Medicine,

Providence, RI, USA; 232Thomas Jefferson University, Sidney Kimmel Medical School, Department of Cancer Biology, Philadelphia, PA, USA; 233Toronto General Hospital Research Institute, University Health Network, Toronto, Canada; 234TPM of Mirandola, Mirandola, Italy; 235Trinity College Dublin, School of Pharmacy and Pharmaceutical Sciences, Panoz Institute & Trinity Biomedical Sciences Institute, Dublin, Ireland; 236Tsinghua University, School of Pharmaceutical Sciences, Beijing, China; 237UMR 7365 CNRS-Université de Lorraine, Vandœuvre-lès-Nancy, France; 238UMR-CBMN, CNRS-Université de Bordeaux, Bordeaux, France; 239UNICAMP, Institute of Biology, Campinas, Brazil; 240UNIFESP, Departamento de Ciências Farmacêuticas, Diadema, Brazil; 241Universidad Autónoma de Madrid, Departamento de Biología Molecular, Madrid, Spain; 242Universidad Autónoma de Madrid, School of Medicine, Department of Medicine, Madrid, Spain; 243Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Buenos Aires, Argentina; 244Universidad de la República, Faculty of Science, Nuclear Research Center, Analytical Biochemistry Unit, Montevideo, Uruguay; 245Universidade Federal de Paraná, Paraná, Brazil; 246Universidade Federal de São Paulo Campus Diadema, Departamento de Ciências Farmacêuticas, Laboratório de Imunologia Celular e Bioquímica de Fungos e Protozoários, São Paulo, Brazil; 247Universidade Federal do Rio de Janeiro, Instituto de Microbiologia, Rio de Janeiro, Brazil; 248Universidade Federal do Rio Grande do Sul, Instituto de Ciências Básicas da Saúde, Departamento de Microbiologia, Imunologia e Parasitologia, Porto Alegre, Brazil; 249Università degli Studi di Milano, Department of Clinical Sciences and Community Health, EPIGET LAB, Milan, Italy; 250Universitat Autònoma de Barcelona, Department of Cell Biology, Physiology & Immunology, Barcelona, Spain; 251Universitat Autònoma de Barcelona, Hospital Universitari and Health Sciences Research Institute Germans Trias i Pujol, Department of Pathology, Barcelona, Spain; 252Universitat de València, Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Àrea de Parasitologia, Valencia, Spain; 253Universitat de València, Health Research Institute La Fe, Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics, Valencia, Spain; 254Université Bretagne Loire, Oniris, INRA, IECM, Nantes, France; 255Université Catholique de Louvain, CHU UCL Namur, Hematology-Hemostasis Laboratory, Yvoir, Belgium; 256Université Catholique de Louvain, Institut de Recherche Expérimentale et Clinique (IREC), Laboratory of Pediatric Hepatology and Cell Therapy, Brussels, Belgium; 257Université de Lille, INSERM, U-1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse – PRISM, Lille, France; 258Université de Nantes, INSERM UMR 1238, Bone Sarcoma and Remodeling of Calcified Tissues, PhyOS, Nantes, France; 259Université de Strasbourg, Strasbourg, France; 260Université Laval, Centre de Recherche du CHU de Québec, Department of Infectious Diseases and Immunity, Quebec City, Canada; 261Université Paris Descartes, Sorbonne Paris Cité, Paris, France; 262Universiteit Hasselt, Diepenbeek, Belgium; 263University Clinic Eppendorf, Hamburg, Germany; 264University College London, London, UK; 265University Hospital Bonn (UKB), Bonn, Germany; 266University Hospital Essen, University Duisburg-Essen, Institute for Transfusion Medicine, Essen, Germany; 267University Hospital Heidelberg, Institute of Pathology, Applied Tumor Biology, Heidelberg, Germany; 268University Hospital RWTH Aachen, Department of Thoracic and Cardiovascular Surgery, Aachen, Germany; 269University Hospitals Cleveland Medical Center, Department of Medicine, Cleveland, OH, USA; 270University Medical Center Göttingen, Developmental Biochemistry, Göttingen, Germany; 271University Medical Center Göttingen, Hematology and Oncology, Göttingen, Germany; 272University Medical Center Hamburg-Eppendorf, Department of Neurosurgery, Hamburg, Germany; 273University Medical Center Hamburg-Eppendorf, Institute of Neuropathology, Hamburg, Germany; 274University Medical Center Utrecht, Department of Nephrology and Hypertension, Utrecht, The Netherlands; 275University Medical Center Utrecht, Laboratory for Clinical Chemistry & Hematology, Utrecht, The Netherlands; 276University of Antwerp, Centre for Proteomics, Antwerp, Belgium; 277University of Auckland, Auckland, New Zealand; 278University of Auckland, Department of Molecular Medicine and Pathology, Auckland, New Zealand; 279University of Auckland, Department of Obstetrics and Gynaecology, Auckland, New Zealand; 280University of Belgrade, Institute for the Application of Nuclear Energy, INEP, Belgrade, Serbia; 281University of Birmingham, Birmingham, UK; 282University of Birmingham, Institute of Microbiology and Infection, Birmingham, UK; 283University of Brescia, Department of Molecular and Translational Medicine, Brescia, Italy; 284University of Bristol, Bristol, UK; 285University of British Columbia Okanagan, Kelowna, Canada; 286University of Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Buenos Aires, Argentina; 287University of California, Davis, Department of Nutrition, Davis, CA, USA; 288University of California, Davis, Department of Otolaryngology, Davis, CA, USA; 289University of California, Los Angeles, California NanoSystems Institute, Los Angeles, CA, USA; 290University of California, Los Angeles, Department of Bioengineering, Los Angeles, CA, USA; 291University of California, Los Angeles, Department of Pathology and Laboratory Medicine, Los Angeles, CA, USA; 292University of California, Los Angeles, Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA; 293University of California, San Diego, Department of Neurosurgery, La Jolla, CA, USA; 294University of California, San Diego, Department of Obstetrics, Gynecology, and Reproductive Sciences, La Jolla, CA, USA; 295University of California, San Diego, Department of Pediatrics, San Diego, CA, USA; 296University of California, San Francisco, CA, USA; 297University of Cambridge School of Clinical Medicine, Addenbrooke’s Hospital, Department of Medicine, Cambridge NIHR BRC Cell Phenotyping Hub, Cambridge, UK; 298University of Campinas, Piracicaba Dental School, Department of Oral Diagnosis, Piracicaba, Brazil; 299University of Chile, Faculty of Chemical and Pharmaceutical Science, Laboratory of Nanobiotechnology and Nanotoxicology, Santiago, Chile; 300University of Cincinnati College of Medicine, Cincinnati, OH, USA; 301University of Cologne, Department of Internal Medicine I, Cologne, Germany; 302University of Colorado, School of Medicine, Department of Ophthalmology, Cell Sight-Ocular Stem Cell and Regeneration Program, Aurora, CO, USA; 303University of Copenhagen, Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Protein Research, Copenhagen, Denmark; 304University of Copenhagen, Institute of Clinical Medicine, Copenhagen, Denmark; 305University of Edinburgh, Institute of Immunology & Infection Research, Edinburgh, UK; 306University of Gothenburg, Institute of Clinical Sciences at Sahlgrenska Academy, Department of Biomaterials, Gothenburg, Sweden; 307University of Gothenburg, Institute of Clinical Sciences, Department of Surgery, Sahlgrenska Cancer Center, Gothenburg, Sweden; 308University of Gothenburg, Institute of Medicine at Sahlgrenska Academy, Krefting Research Centre, Gothenburg, Sweden; 309University of Gothenburg, Sahlgrenska Academy, Department of Rheumatology and Inflammation Research, Gothenburg, Sweden; 310University of Gothenburg, The Sahlgrenska Academy, Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Mölndal, Sweden; 311University of Helsinki, EV Core Facility, Helsinki, Finland; 312University of Helsinki, Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences Research Programme, EV group, Helsinki, Finland; 313University of Hertfordshire, School of Life and Medical Sciences, Biosciences Research Group, Hatfield, UK; 314University of Kentucky, College of Medicine, Department of Physiology, Lexington, KY, USA; 315University of L’Aquila, Department of Life, Health and Environmental Sciences, L’Aquila, Italy; 316University of Liège, GIGA-R(MBD), PSI Laboratory, Liège, Belgium; 317University of Ljubljana, Faculty of Medicine, Institute of Biochemistry, Ljubljana, Slovenia; 318University of Lyon, INRA, EPHE, UMR754 Viral Infections and Comparative Pathology, Lyon, France; 319University of Lyon, Lyon-Sud Faculty of Medicine, CarMeN Laboratory (UMR INSERM 1060-INRA 1397), Pierre-Bénite, France; 320University of Mainz, Institute of Developmental Biology and Neurobiology, Mainz, Germany; 321University of Malta, Department of Pathology, Msida, Malta; 322University of Manchester, Division of Cancer Sciences, Manchester Cancer Research Centre, Manchester, UK; 323University of Manchester, Manchester, UK; 324University of Maryland, Fischell Department of Bioengineering, College Park, MD, USA; 325University of Massachusetts Medical School, RNA
JOURNAL OF EXTRACELLULAR VESICLES 5

6 C. THÉRY AND K. W. WITWER
Therapeutics Institute, Worcester, MA, USA; 326University of Michigan Medical School, Ann Arbor, MI, USA; 327University of Michigan, Biointerfaces Institute, Ann Arbor, MI, USA; 328University of Michigan, Department of Biomedical Engineering, Ann Arbor, MI, USA; 329University of Michigan, Department of Internal Medicine – Hematology/Oncology Division, Ann Arbor, MI, USA; 330University of Michigan, Department of Medicine, Ann Arbor, MI, USA; 331University of Michigan, Department of Urology, Ann Arbor, MI, USA; 332University of Minnesota Medical School, Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, Minneapolis, MN, USA; 333University of Modena and Reggio Emilia, Division of Oncology, Modena, Italy; 334University of Nebraska Medical Center, Department of Pharmacology and Experimental Neuroscience, Omaha, NE, USA; 335University of Notre Dame, Department of Biological Sciences, Notre Dame, IN, USA; 336University of Oslo, Institute of Clinical Medicine, Oslo, Norway; 337University of Ottawa, Ottawa, Canada; 338University of Oulu, Faculty of Medicine, Cancer and Translational Medicine Research Unit, Oulu, Finland; 339University of Oxford, Department of Physiology, Anatomy and Genetics, Oxford, UK; 340University of Oxford, Radcliffe Department of Medicine, Acute Stroke Programme – Investigative Medicine, Oxford, UK; 341University of Padova, Department of Comparative Biomedicine and Food Science, Padova, Italy; 342University of Padova, Department of Women’s and Children’s Health, Padova, Italy; 343University of Palermo, Department of Biopathology and Medical Biotechnologies, Palermo, Italy; 344University of Perugia, Department of Chemistry, Biology and Biotechnology, Perugia, Italy; 345University of Pisa, Centro Dipartimentale di Biologia Cellulare Cardio-Respiratoria, Pisa, Italy; 346University of Porto, Faculty of Pharmacy (FFUP), IBMC/I3S, Porto, Portugal; 347University of Porto, Faculty of Pharmacy (FFUP), Porto, Portugal; 348University of Porto, i3S- Instituto de Investigação e Inovação em Saúde, Porto, Portugal; 349University of Porto, ICBAS – Instituto de Ciências Biomédicas Abel Salazar, Porto, Portugal; 350University of Rochester, Rochester, NY, USA; 351University of São Paulo, Ribeirão Preto Medical School, Department of Pathology and Forensic Medicine, Ribeirão Preto, Brazil; 352University of São Paulo, Ribeirão Preto Medical School, Ribeirão Preto, Brazil; 353University of Science and Culture, ACECR, Department of Developmental Biology, Tehran, Iran; 354University of South Alabama, Department of Pharmacology, Center for Lung Biology, Mobile, AL, USA; 355University of Southern California Keck School of Medicine, Los Angeles, CA, USA; 356University of Southern California, Los Angeles, CA, USA; 357University of Technology Sydney, Discipline of Pharmacy, Graduate School of Health, Sydney, Australia; 358University of Texas MD Anderson Cancer Center, Department of Cancer Biology, Metastasis Research Center, Houston, TX, USA; 359University of Torino, Department of Medical Sciences, Torino, Italy; 360University of Torino, Department of Molecular Biotechnology and Health Sciences, Torino, Italy; 361University of Toronto, Department of Anesthesia, Toronto, Canada; 362University of Toronto, Department of Laboratory Medicine and Pathobiology, Toronto, Canada; 363University of Toronto, Department of Medical Biophysics, Toronto, Canada; 364University of Toronto, Department of Medicine, Division of Neurology, Toronto, Canada; 365University of Tsukuba, Tsukuba, Japan; 366University of Virginia Health System, Department of Medicine, Division of Nephrology, Charlottesville, VA, USA; 367University of Virginia, Flow Cytometry Core, School of Medicine, Charlottesville, VA, USA; 368University of Würzburg, Rudolf Virchow Center, Würzburg, Germany; 369Utrecht University, Faculty of Veterinary Medicine, Department of Biochemistry and Cell Biology, Utrecht, The Netherlands; 370Utrecht University, University Medical Center Utrecht, Center for Molecular Medicine & Regenerative Medicine Center, Utrecht, The Netherlands; 371Utrecht University, University Medical Center Utrecht, Department of Neurosurgery, Brain Center Rudolf Magnus, Institute of Neurosciences, Utrecht, The Netherlands; 372Utrecht University, University Medical Center Utrecht, Department of Pathology, Utrecht, The Netherlands; 373Vanderbilt University Medical Center, Epithelial Biology Center, Department of Medicine, Nashville, TN, USA; 374Vanderbilt University School of Medicine, Department of Cell and Developmental Biology, Nashville, TN, USA; 375Veterans Affairs Medical Center, San Francisco, CA, USA; 376Vilnius University, Institute of Biomedical Sciences, Department of Physiology, Biochemistry, Microbiology and Laboratory Medicine, Vilnius, Lithuania; 377Vlaamse Instelling voor Technologisch Onderzoek (VITO), Mol, Belgium; 378Washington University, Saint Louis, MO, USA; 379Weill Cornell Medicine, Department of Medicine, New York City, NY, USA; 380Weizmann Institute of Science, Department of Biomolecular Sciences, Rehovot, Israel; 381West Virginia University, Department of Chemical and Biomedical Engineering and WVU Cancer Institute, Morgantown, WV, USA; 382West Virginia University, Department of Microbiology Immunology and Cell Biology, Morgantown, WV, USA; 383West Virginia University, Morgantown, WV, USA; 384Xiamen University, Department of Chemical Biology, Xiamen, China
ARTICLE HISTORY
Received 20 September 2018 Accepted 25 September 2018
KEYWORDS
extracellular vesicles; exosomes; ectosomes; microvesicles; minimal information requirements; guidelines; standardization; microparticles; rigor; reproducibility
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, micropar- ticles, 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.

Table of Contents
i. Introduction ……………………………………………………………………………………………………………………………………………………… 7 ii. Note on applicability of MISEV2018: species, cells, sample types, and experimental conditions …………………… 8
1 – Nomenclature …………………………………………………………………………………………………………………………………………….. 8 2 – Collection and pre-processing: pre-analytical variables …………………………………………………………………………….. 9 2-a) Cell culture conditioned media ………………………………………………………………………………………………………… 9 2-b) Biological fluids ………………………………………………………………………………………………………………………………. 10 2-c) Tissues …………………………………………………………………………………………………………………………………………….. 11 2-d) Storage ……………………………………………………………………………………………………………………………………………. 11 3 – EV separation and concentration: How MISEV2014 evolves in 2018 …………………………………………………….. 11 Table 1: Considerations for EV separation/enrichment ………………………………………………………………………… 13 4 – EV characterization: How MISEV2014 evolves in 2018 ………………………………………………………………………….. 12 Table 2: Steps of EV characterization ……………………………………………………………………………………………………. 14 4-a) Quantification of EVs …………………………………………………………………………………………………………………….. 12 4-b) Characterization of EVs by their protein composition ………………………………………………………………….. 16 4-b-1) Selection of proteins for use as EV markers ………………………………………………………………………. 16 Table 3: Protein content-based EV characterization ……………………………………………………………………………… 19 4-b-2) Methods to assess presence of proteins in EV preparations ……………………………………………….. 18 4-b-3) Non-protein components as markers of EVs ……………………………………………………………………… 18 4-c) Single vesicle analysis ……………………………………………………………………………………………………………………… 20 4-d) New recommendation: determine the topology of EV-associated components ……………………………. 21 5 – Functional studies: How MISEV2014 evolves in 2018 …………………………………………………………………………….. 22 Table 4: EV-associated and EV-excluded biological activities ……………………………………………………………….. 22 5-a) Determine the specific versus common functions of different types of EVs ………………………………… 22 5-b) Demonstrate that the activity is observed in the absence of direct cell-cell contact …………………….. 23
5-c) Demonstrate that the activity is predominantly associated with EVs rather than
with soluble mediators ……………………………………………………………………………………………………………………. 24
5-d) Demonstrate the specific association of the activity with EVs rather than
with co-isolated components ………………………………………………………………………………………………………….. 24
5-e) Determine whether a function is specific to exosomes, as compared with other small EVs ………… 25 5-f) How to attribute particular effects mediated by EVs to specific EV components …………………………. 26 5-g) Consider whether an EV-dependent function is specific to a given EV source ……………………………. 26
6 – General considerations …………………………………………………………………………………………………………………………….. 27 6-a) Reporting requirements ………………………………………………………………………………………………………………….. 27 6-b) Exceptions to compliance with MISEV guidelines ………………………………………………………………………… 27
7 – Conclusions ………………………………………………………………………………………………………………………………………………. 27 8 – References …………………………………………………………………………………………………………………………………………………. 28 9 – MISEV2018 Quick-Reference Checklist …………………………………………………………………………………………………… 42
Introduction
In 2014, the ISEV board members published a Position Editorial detailing their recommendations, based on their own established expertise, on the “minimal experimental requirements for definition of extracellular vesicles and their functions” [1]. A list of minimal information for studies of extracellular vesicles (MISEV or MISEV2014) was provided, covering extracellular vesicle (EV) separa- tion/isolation, characterization, and functional studies. The major goal of these recommendations was to sensi- tize researchers (especially the rapidly growing numbers of scientists newly interested in EVs), as well as journal
editors and reviewers, to experimental and reporting requirements specific to the EV field. The ISEV board highlighted the need to consider these issues when mak- ing strong conclusions on the involvement of EVs, or specific populations of EVs (exosomes in particular), in any physiological or pathological situation, or when pro- posing EVs or their molecular cargo as biological mar- kers. By stimulating improved reliability and reproducibility of published EV results, the MISEV2014 authors hoped to further the promise of EVs as biomar- kers or for therapeutic applications even in the face of skepticism by some scientists outside the field.
JOURNAL OF EXTRACELLULAR VESICLES 7

8 C. THÉRY AND K. W. WITWER
As evidenced by the increasing number of EV publica- tions in high-profile journals, proposing major roles of EVs in numerous physiological pathways from aging to cancer, infectious diseases to obesity, EV science has now clearly achieved widespread interest and enthusiasm well beyond the EV research community. However, the promotion of rigorous EV science is an ongoing process; as EV experts within the ISEV community, we are still concerned to see that major conclusions in some articles are not sufficiently supported by the experiments performed or the informa- tion reported. We therefore aim to revise and renew the MISEV recommendations and to continue to work toward their wider acceptance and implementation. In this “MISEV2018” update, a much larger group of ISEV scien- tists was involved through a community outreach (the MISEV2018 Survey), striving for consensus on what is absolutely necessary, what should be done if possible, and how to cautiously interpret results if all recommendations for controls cannot be followed.
We strongly believe that most of the MISEV2014 recommendations are still valid; however, discoveries and developments within the field during the past four years necessitate certain amendments. This document explains how the 2014 recommendations evolved into MISEV2018 in Tables 1, 2 and 4; provides suggestions for protein markers to validate the presence of EVs (Table 3); and, to highlight the salient points, provides outlines of examplar approaches to address some of the most important experimental issues. Importantly, a 2-page checklist summarizing the major aspects to follow in EV science is provided at the end of this article.
The authors of MISEV2014 were careful to propose feasible experiments and controls for most experimental situations, but also to suggest alternatives for particular situations in which not all guidelines could be strictly followed, such as for limiting sample quantities. However, a recent survey of members of ISEV to lay the groundwork for MISEV2018 [2] showed that, while respondents agreed almost unanimously on the need for minimal require- ments, and a majority supported the MISEV2014 initiative and guidelines as published, almost a quarter of respon- dents found the guidelines too restrictive or too strong an imposition on the field. MISEV2018 thus provides clearer explanations of the need for each recommendation and highlights the extent of author consensus (or lack thereof) on each section. An initial draft of MISEV2018 was sub- mitted to the entire ISEV membership as a Survey asking for agreement/disagreement and comments on each sec- tion. The survey specified that, for agree/disagree ques- tions, > 20% “disagree” responses would prompt acknowledgment of major dissent in the final document, while > 40% “disagree” would prompt a focused survey or discussion of the ISEV board with selected survey
respondents to redraft the relevant section. 329 responses were received, in which there was such broad agreement on the MISEV2018 draft that the 40% threshold was not met for any section. Nevertheless, attempts were made to address as many comments as comprehensively as possible, and thus generate a semi-final version of this text. Finally, a last round of review was conducted by all previous con- tributors as well as ISEV and JEV board members and numerous additional long-standing EV experts. Although not all suggestions, references, and critiques could be included in the final product, we are confident that this document represents the views of EV scientists with broad and deep expertise.
Consensus: > 99% of MISEV2018 Survey respondents agreed with the introduction. It has been modified only slightly since the survey, mostly to convey the survey mechanism and results.
Note on applicability of MISEV2018: species, cells, sample types, and experimental conditions
Does MISEV2018 apply to all EV studies, or only to some? EVs appear to be produced by almost all organisms and cell types studied. Yet EV research to date has focused on mammalian EVs, chiefly those of human or mouse origin, and not all cell types or experimental conditions have been closely investigated. In this document, as in MISEV2014, specific examples of molecular markers (such as the mar- kers of EVs in Table 3) are based on studies of specific species, cells, and experimental conditions. Some may be broadly applicable, others less so. Nevertheless, the general principles of MISEV2018 apply to EVs produced by all organisms and all cells. The need to demonstrate presence (or enrichment) of EV markers and absence (or depletion) of putative contaminants, when contents or function of EVs are described, can be generalized to all species, cells, and conditions. We find ourselves at an exciting scientific frontier; where such markers are not yet available, we encourage their development and publication, using the principles of this document as a guide. Additional specific examples may then be incorporated into future MISEV updates.
Consensus: 93% of MISEV2018 Survey respondents agreed that the examples in this document would be based on mammalian EVs. However, applicability to non- mammalian and non-eukaryotic EVs is now addressed.
Nomenclature
ISEV endorses “extracellular vesicle” (EV) as the generic term for particles naturally released from the cell that are

delimited by a lipid bilayer and cannot replicate, i.e. do not contain a functional nucleus. Since consensus has not yet emerged on specific markers of EV subtypes, such as endo- some-origin “exosomes” and plasma membrane-derived “ectosomes” (microparticles/microvesicles) [3,4] assigning an EV to a particular biogenesis pathway remains extra- ordinarily difficult unless, e.g. the EV is caught in the act of release by live imaging techniques. Therefore, unless authors can establish specific markers of subcellular origin that are reliable within their experimental system(s), authors are urged to consider use of operational terms for EV subtypes that refer to a) physical characteristics of EVs, such as size (“small EVs” (sEVs) and “medium/large EVs” (m/lEVs), with ranges defined, for instance, respectively, < 100nm or < 200nm [small], or > 200nm [large and/or medium]) or density (low, middle, high, with each range defined); b) biochemical composition (CD63+/CD81+- EVs, Annexin A5-stained EVs, etc.); or c) descriptions of conditions or cell of origin (podocyte EVs, hypoxic EVs, large oncosomes, apoptotic bodies) in the place of terms such as exosome and microvesicle that are historically burdened by both manifold, contradictory definitions and inaccurate expectations of unique biogenesis. If it is deemed unavoidable to use these or newly coined terms, they should be defined clearly and prominently at the beginning of each publication [5]. If confirmation of EV identity cannot be achieved according to the minimal requirements of this MISEV2018 publication, other terms such as extracellular particle (EP) might be more appropriate.
Consensus: 94% of MISEV2018 Survey respondents endorsed this nomenclature recommendation. The remainder were evenly split between dissent and a preference for no nomenclature recommendation.
Collection and pre-processing: pre-analytical variables
The first step to recover EVs is to harvest an EV-containing matrix, such as fluid from tissue culture or from an orga- nismal compartment. During this pre-analytical phase, an extended constellation of factors, including characteristics of the source, how the source material is manipulated and stored, and experimental conditions, can affect EV recov- ery. Therefore, it is crucial to plan collection and experi- mental procedures to maximize the number of known, reportable parameters, and then to report as many pre- analytical parameters as are known.
Cell culture conditioned media
For EV isolation/characterization from conditioned media (an ISEV survey found that the majority of responding EV
researchers studied conditioned medium [6]), basic char- acterization of the releasing cells and culture and harvesting conditions must be performed and reported. Some precau- tions, such as regular confirmation of cellular identity (e.g. by short tandem repeat (STR) profiling or other methods) [7,8] and identification of cell lineage and provenance including mode of immortalization [9], are advisable for all cell studies. Especially important for EV studies is that the percent of dead cells at the time of EV harvest should be indicated, since even a small percentage of cell death could release cell membranes that outnumber true released EVs. Quantifying the percentage of apoptotic and necrotic cells may also be useful. (Note, however, that when cells are treated with high concentrations of EVs, cell-adherent EVs positive for apoptotic markers may skew results [10,11]). Other relevant characteristics of the cells, including state of activation, malignancy, and senescence [12,13], should be reported where applicable.
Culture and harvesting conditions such as passage number (or days in culture for suspension cells), seeding density [14], density/confluence at harvest [14], including any relevant post-confluence characteristics such as devel- opment of polarity [15–19] (in that case, were EVs collected globally or separately, from the different parts of polarized cells?), culture volume, culture vessel or bioreactor system (if used [20,21]), surface coatings, oxygen or other gas tensions (if they differ from standard cell culture) [22,23], stimulation and other treatments [24–30], and frequency and intervals of harvest [14] should be given to allow replication [31,32]. Culture conditions prior to treatment- (s), if any, should also be given. Note that EV recovery depends not only on EV release, but also on re-uptake by cells in culture, which may vary based on culture density and other conditions. Regular checks for contamination with Mycoplasma (and possibly other microbes) are needed, not only because of cellular responses to contam- ination, but also because contaminating species can release EVs [33–36]. Exact methods of medium collection should be given, as well (e.g. decanting or pipetting from flasks, centrifugation of suspension cell cultures). The suggested parameters are of course non-inclusive, and others may be necessary to report for specific types of cells and experi- ments, including co-culture systems and organoid cul- tures [37].
All culture medium composition and preparation details should be provided in methods. This should be customary for cell culture studies, and is doubly impor- tant here since supplements like glucose [38–40], anti- biotics [41], and growth factors [42] can affect EV production and/or composition. Of special note are medium components that are likely to contain EVs, such as serum. EVs are ideally obtained from culture medium conditioned by cells in the absence of fetal calf
JOURNAL OF EXTRACELLULAR VESICLES 9

10 C. THÉRY AND K. W. WITWER
serum (FCS or FBS), serum from other species, or other complex products such as platelet lysate, pituitary extract, bile salts, and more, to avoid co-isolation of exogenous EVs. When use of these supplements is unavoidable, experiments should include a non- conditioned medium control to assess the contribution of the medium itself. However, depending on down- stream use, it may not be necessary or desirable to deplete EVs [43,44]. In the case of depletion, since nutrient or EV deprivation of cells that are normally cultured in serum- or lysate-containing medium can change cellular behavior and the nature and composi- tion of released EVs [45,46], it is important to specify culture history (how and when the switch to serum- free medium occurred, including acclimatization steps). Alternatively, cells can be exposed during the EV release period to medium that has been pre- depleted of EVs. Here, too, effects on cells and EVs may be expected [47], and the methods and outcome of depletion vary greatly and should be reported. Several fairly efficient protocols are available, such as 100,000 x g ultracentrifugation of complete medium (or of serum following at least 1:4 dilution) for at least 18 hours [48], centrifugation at enhanced speeds (e.g. 200,000 x g [49]) for shorter periods of time, or tan- gential flow filtration or other forms of ultrafiltration [50]. Ultracentrifugation at around 100k x g for a few hours or without dilution will not eliminate all EVs or EV-associated RNA [51–53]. Commercial “exosome/ EV-depleted” serum and other supplements are avail- able from an increasing number of vendors. Since the method of depletion is usually not indicated, conse- quences on cell growth and EV release may not be predictable; the exact source, method, and reference of depleted supplements should be given, and the “exo- some-free” nature of the product should be checked carefully before use [54]. Additionally, vendors are encouraged to report and benchmark the methods of depletion utilized in their products, while users should report product and lot numbers as well as any pooling of biologicals. Finally, medium preparation details, including heating (heat inactivation) or filtration steps, should be reported. For example, heat inactiva- tion of additives such as serum leads to formation of protein aggregates that may co-precipitate with EVs and thus also change the growth-supporting properties of the serum.
Biological fluids
Since more than 30 types of biofluids exist in mammals, and lavages of numerous compartments add to this num- ber (despite not being true biofluids), MISEV2018 does
not provide an exhaustive review of the literature on pre- analytical variables related to all biofluids. Each biological fluid presents specific biophysical and chemical character- istics that makes it different from culture conditioned medium, and this must be taken into account when isolating EVs. For instance, plasma and serum are more viscous than conditioned medium. Plasma and serum contain numerous non-EV lipidic structures (low/very low/high density lipoproteins), milk is replete with fat- containing vesicles, urine with uromodulin (Tamm- Horsfall protein), bronchoalveolar lavage with surfactant, all of which will be co-isolated to various degrees with EVs. In each case, specific precautions to separate EVs from these components may be required. While detailed biofluid-specific reporting guidelines are beyond the scope of this MISEV, we encourage development of such guidelines under the MISEV umbrella.
For EV isolation/characterization from biofluids such as blood plasma, several previous ISEV posi- tion papers [55,56] and other publications (for just a few of many examples, see [57–63]) have listed reporting requirements that are important for stan- dardization, and these are still valid today, even if many questions remain about the effects of specific pre-analytical variables on different classes of EVs. Since many of these factors have been covered in these previous publications, we do not review them exhaustively here. To give examples of considera- tions for blood derivatives such as plasma: donor age, biological sex, current or previous pregnancy, menopause, pre/postprandial status (fasting/non- fasting), time of day of collection (Circadian varia- tions), exercise level and time of last exercise, diet, body mass index, specific infectious and non- infectious diseases, medications, and other factors may affect circulating EVs [64,65]. Similarly, tech- nical factors including fluid collection volume, first- tube discard, type of container(s), time to proces- sing, choice of anticoagulant (for blood plasma) [66–68], mixing or agitation, temperature (storage and processing), description of transport (if any), whether tube remained upright before processing, exact centrifugation or filtration procedures, degree of hemolysis, possible confirmation of platelet and lipoprotein depletion prior to storage [69–73], and other parameters should be clearly indicated. Overall, except some that are specific of plasma/ serum (such as platelet removal and coagulation), the above listed technical details of collection con- dition apply to all biofluids and must be reported. Of course, it may be that not all variables have been recorded for archived samples, and this should be acknowledged where applicable.

Tissue
As a special case of pre-analytical issues, a rapidly increasing number of groups have reported isolation of tissue EVs. These studies may involve short-term culture of tissue explants [74] such as ex vivo tumors [75], or placenta [76,77], or extraction from whole tissues [78–84]. Many of the same considerations that apply for cellular and biofluids studies also apply here, including confirmation of provenance and condition. Especially for EV extraction from tissue, it is challen- ging to ensure that recovered vesicles are truly from the extracellular space, rather than being intracellular vesi- cles or artefactual particles released from cells broken during tissue harvest, processing (e.g. mechanical dis- ruption), or storage (including freezing). This may be especially challenging in a tissue like brain, where similar procedures are used to collect synaptosomes [85]. Even apparently pure tissue-derived EVs can con- tain endosome components, which could correspond to components of intracellular vesicles including unre- leased intraluminal vesicles of late endosomes/multi- vesicular bodies (MVBs) that are released artifactually during tissue processing. The recent awareness of these challenges has led researchers to perform gentle tissue disruption (i.e. with the goal of separating EVs from cells and extracellular matrix, but not disrupting cells) and several steps of further separation (including den- sity gradients), followed by strict characterization of multiple negative markers, leading to more convincing tissue-derived EV preparations [79]. Use of genetically modified models to trace EV release from specific cells [83] is also a useful approach. More research is clearly needed and encouraged into the isolation, characteriza- tion, and function of tissue EVs, as compared with intracellular vesicles and/or non-vesicular extracellular particles (EPs).
Storage
Storage and retrieval conditions of both the matrix (e.g. biofluid, tissue, conditioned media) and isolated EVs may affect EV characteristics, including stability, num- ber of particles, aggregation, and function [57,62,63,71,86–96]. In particular, highly purified EVs may be lost upon storage by adhering to the surfaces of the storage container. How were biofluids, tissues, or media prepared and stored (type of storage container, temperature, etc.) and for how long? Were isolated EVs analyzed or used for experiments fresh, frozen/thawed, lyophilized and reconstituted, etc.? If frozen, how was freezing and thawing performed? In what buffer(s) were EVs stored? For how long? What, if any, cryo- protectant was used? How many freeze-thaw cycles did
each sample experience? If EVs were processed and stored in some other manner, details should also be provided, along with the procedure to evaluate effects of storage method and time on EV activity and other properties, where applicable.
Consensus: 96% of MISEV2018 Survey respondents endorsed the pre-analytical variables section. Since 44% of respondents also suggested at least one added literature citation or other amendment in more than 200 total comments, the section was revised to reflect as many of these suggestions as possible. However, bio- fluids-specific considerations and SOPs are beyond the current scope of MISEV; only illustrative examples are provided. The MISEV2018 Survey comments evidenced particular interest in urine and milk, consistent with the “Experts Meet” sessions during ISEV2016 and the results of a previous ISEV survey (which also highlighted cerebrospinal fluid) [6]. Beginning with the blood EV roadmap announced at the ISEV2018 annual meeting [97], development of more specific recommendations for individual biofluids and other matrices is encouraged. The overwhelming response to this section indeed demands additional research into the effects of pre- analytical variables on EV studies.
EV separation and concentration: how MISEV2014 evolves in 2018
Absolute purification, or complete isolation of EVs from other entities, is an unrealistic goal (as for many biological products). For this reason, and since the various combinations of EVs and media are colloids [98], here we use the terms separation and concentra- tion. Separation (colloquially referred to as purification or isolation) of 1) EVs from other non-EV components of the matrix (conditioned medium, biofluid, tissue) and 2) the different types of EVs from each other, are achieved to various degrees by the different techniques available. Concentration is a means to increase num- bers of EVs per unit volume, with or without separa- tion. The term “enrichment” can refer to increasing concentration, i.e. EV counts relative to volume, or to increasing EV counts/markers relative to another com- ponent. The extent of separation or concentration can be assessed by characterization, which will be detailed in the next section.
How pure should an EV preparation be? The answer depends on the experimental question and EV end use, and often segregates by basic and clinical research. Highly purified EVs are needed to attribute a function or a biomarker to vesicles as compared with other particles. Less pure EVs may be required in other cases, such as when a biomarker is useful without pre-enrichment of EVs, or in certain
JOURNAL OF EXTRACELLULAR VESICLES 11

12 C. THÉRY AND K. W. WITWER
therapeutic situations where function is paramount, not the definitive association of function with EVs. Of note, some presumed contaminants may co-isolate with EVs and may even contribute to EV function. Therefore, the choice of separation and concentration method must be informed by factors that may vary between studies such that there is no one-size-fits-all approach. More details on this issue (function and co- isolated factors) are given in section 5c-d (p.24).
At the end of 2015, according to a worldwide ISEV survey [6], differential ultracentrifugation was the most commonly used primary EV separation and concentra- tion technique, with various other techniques, such as density gradients, precipitation, filtration, size exclu- sion chromatography, and immunoisolation, used by 5–20% of respondents each. Relative success of these different methods in terms ofrecovery and specificity to EVs (as compared to non-vesicular components), or to EV subtypes, has been addressed in a previous ISEV Position Paper (see Figure 1 of [56]), and is summar- ized in Table 1 below. To achieve better specificity of EV or EV subtype separation, most researchers use one or more additional techniques following the primary step, such as washing in EV-free buffer, ultrafiltration, application of density gradients (velocity or flotation), or chromatography [6,99–102].
A variety of additional techniques or combinations of techniques have been or are currently being devel- oped, some of which may become more prominent in the coming years if they achieve better recovery or specificity than legacy methods (and this must be demonstrated as in, e.g. [103]). Such methods include tangential flow filtration and variations thereon [21,104–110], field-flow fractionation (FFF) [111], asymmetric flow field-flow fractionation (AFFF, A4F, or AF4) [112–114], field-free viscoelastic flow [115], alternating current electrophoretics [116,117], acoustics [118], variations on size exclusion chromatography (SEC) [100,119–121], ion exchange chromatography [122–124], microfiltration [125], fluorescence- activated sorting [126,127] (especially for larger EVs including large apoptotic bodies [128] and large onco- somes [129]), deterministic lateral displacement (DLD) arrays [130], novel immunoisolation or other affinity isolation technologies [131–138], including lipid affi- nity [139], novel precipitation/combination techniques [140–142], hydrostatic filtration dialysis [143], high- throughput/high-pressure methods such as fast pro- tein/high perfomance liquid chromatography (FPLC/ HPLC) that involve some form of chromatography [144] and a wide variety of microfluidics devices that harness one or more principles, including some of
those mentioned above [145–153]. Of course, combi- nations of methods will continue to be used and may outperform single-method approaches.
Table 1 summarizes the instructions given in MISEV2014 for EV isolation (left column), and their updates in MISEV2018 (right).
Consensus: 93% of MISEV2018 Survey respondents agreed with the original categorization of techniques by recovery and specificity in Section 3 and Table 1a; numerous amendments have been made in response to almost 90 comments. 98% agreed that reporting of all methods details should be mandated to allow reprodu- cibility. 97% agreed with the statement of caution on proprietary kits.
EV characterization: how MISEV2014 evolves in 2018
EV characterization by multiple, complementary techni- ques is important to assess the results of separation methods and to establish the likelihood that biomarkers or functions are associated with EVs and not other co- isolated materials. The need for guidelines for character- ization was emphasized by a consortium study led by Hendrix and colleagues [161]. These authors found that only about half of EV-related articles published within a five-year time period included positive markers of EVs, and only a small minority complemented positive with negative markers to track co-isolated non-EV com- ponents. ISEV recommends that each preparation of EVs be 1) defined by quantitative measures of the source of EVs (e.g. number of secreting cells, volume of bio- fluid, mass of tissue); 2) characterized to the extent possible to determine abundance of EVs (total particle number and/or protein or lipid content); 3) tested for presence of components associated with EV subtypes or EVs generically, depending on the specificity one wishes to achieve; and 4) tested for the presence of non- vesicular, co-isolated components.
Table 2 summarizes the instructions given in MISEV2014 for EV characterization, and their updates in MISEV2018. These recommendations apply to EVs from all sources, including non-mammalian and non- eukaryotic cells and organisms.
Quantification of EVs
Since quantifying EVs themselves remains difficult (see below), as minimal information, the total starting volume of biofluid, or, for conditioned medium, num- ber of cells or mass of tissue at the time of collecting, should be indicated for each experimental use. If the

Table 1. Considerations for EV separation/enrichment. Major recommendations of MISEV2014.
a) There is no single optimal separation method, so choose based on the downstream applications and scientific question.
Separation of non-vesicular entities from EVs is not fully achieved by common EV isolation protocols, including centrifugation protocols or commercial kits that claim EV or “‘exosome’” purification.
Validity and/or Update in 2018
Still valid. Any newly developed or applied technique for EV isolation must indicate to which of the 4 recovery/specificity options below it aims, and provide characterization information (see Table 2) to show the extent of success.
Different methods may be positioned on a recovery vs specificity grid, ranging from low to high in each dimension. Note that the degree of specificity of a particular method might vary depending on the type of biofluid from which EVs are separated.
1) high recovery, low specificity: methods that recover the highest amount of extracellular material, whatever its vesicular or non-vesicular nature, i.e. whole or near-whole concentrated secretome. Examples of protocols include but are not limited to: precipitation kits/polymer (PEG or others), low molecular weight cutoff centrifugal filters with no further separation step, and lengthy or very high speed ultracentrifugation without previous, lower-speed steps.
2) intermediate recovery, intermediate specificity: methods that recover mixed EVs along with some amount of free proteins, ribonucleoproteins, and lipoproteins, depending on the matrix. Examples of protocols: size- exclusion chromatography [154,155], high molecular weight centrifugal filters [102], differential ultracentrifugation using intermediate time/ speed with or without wash, tangential flow filtration, and membrane- affinity columns [155,156].
3) low recovery, high specificity: methods that recover a subtype (or
a few subtypes) of EVs with as few non-vesicular components as possible. Subtypes of EVs can be separated by their size (e.g. by filtration, which must be combined with another method such as SEC to eliminate non EV components), their density upon either flotation or pelleting in a density gradient, their surface protein, sugar, or lipid composition (immuno- or other affinity isolation including flow cytometry for large particles), or other biophysical properties such as surface charge. Note that the designation of “low recovery” is relative to total EVs, and that high recovery of specific subtypes may be possible using these techniques.
4) high recovery and high specificity, which may not be achievable as of this writing.
Still valid. Methods reporting is now facilitated by the EV-TRACK knowledgebase [161] (see also checklist, p.42). ISEV strongly recommends that authors deposit experimental details with EV-TRACK.
Examples for classical techniques: centrifugation (g-force, rotor, ultracentrifuge, adjusted k-factor, tube type, adaptor if relevant, time, temperature, and brake)a; gradients (materials, densities, volumes, and whether top-down (pelleting) or bottom-up (flotation) gradients were done, further processing); chromatography (matrix nature, pore size, volume; volume of loaded sample; volume and number of fractions, type of the elution buffer; further processing of fractions); immunoaffinity (antibody reference and amount per volume of fluid, particle number, or protein amount, incubation time and temperature, matrix, recovery).
Necessary technical detail reporting may have to be established for newly developed techniques.b-d
MISEV2018 additional recommendation:
Some protocols, including those associated with many commercial kits, may
result in EV populations bound to or mixed with introduced components such as antibodies, beads, polymers, and more. These materials may affect downstream profiling or functional studies and may also render the EVs unusable for therapeutic applications. Particular care in performing functional experiments must be taken (see Table 4), with controls including procedural controls and possibly with further separation of EVs.e,f
b) Report all details of the method(s) for reproducibility
c)
JOURNAL OF EXTRACELLULAR VESICLES 13
aFor ultracentrifugation, the k factor can be determined from the rotor type, tube/adapter, and centrifuge speed: the k factor represents the relative pelleting efficiency of a given centrifuge rotor at maximum rotation speed; for runs with a rotational speed lower than the maximum rotor-speed, the k factor has to be adjusted: kadj = k x (maximum rotor speed/actual rotor speed)2; we recommend that all possible parameters be reported [157,201]. See also a web calculator based on a theoretical model of centrifugation and meant for conversion of protocol parameters between rotors [158].
bFor filtration techniques, one must take care to remove cells and other large membranous structures prior to ultrafiltration; otherwise, large structures may disintegrate and re-form as small vesicles after passing through the filter [159]. Reference numbers of all filters should be specified, as filter type has been found to influence recovery profoundly [102].
cSEC: the pore size of the matrix should be taken into account. For example, if the pore size excludes EVs > 70 nm in diameter, a population of vesicles may be excluded.
dBoth size exclusion chromatography (SEC) and density gradients may co-isolate EVs and certain lipoproteins. Sequential techniques may be needed to achieve separation [160,213].
eNote, however, continuing concerns about the specificity and effects on vesicles of certain precipitation techniques [103,278].
fThis Table does not address several important considerations that are beyond the scope of these guidelines, such as ease of use, cost, and potential for
Good Manufacturing Practice (GMP) production.

14 C. THÉRY AND K. W. WITWER
Table 2. Steps of EV characterization.
Major recommendations of MISEV2014.
a) No recommendation on quantification
Validity and/or Update in 2018
New in MISEV2018:
As a rule, both the source of EVs and the EV preparation must be described quantitatively. Source: Number of cultured cells (possibly an estimate, if adherent, as well as intervals of
harvest, where applicable), total starting volume of biofluid, or weight/volume/size of tissue at the time of collection must be indicated for each experimental use. The appropriate parameters will vary by source, however; e.g. for urine, volume alone may not be meaningful, and other parameters, such as creatinine level, might be reported.
For the EV preparation, global quantification of EVs should be provided. There is no single perfect quantification method. The most commonly used are total protein amount and total particle number. Total lipid quantification could be also considered. None of these components are exclusively associated to EVs, though: proteins are also soluble, particles can be protein aggregates, and lipids are also present in lipoproteins. Thus, ratios of proteins:particles, lipids:particles or lipids:proteins should be reported along with global quantification estimates as a measure of purity and thus reliability of the quantity measure.
Still valid but has evolved with increasing knowledge of the existence of different EV types. Table 3 gives categories of proteins to consider for characterization and some examples. At least one protein of each category 1 to 3 must be evaluated in any EV preparation (at least each time pre-analytical and/or EV isolation conditions are modified). Analysis of proteins of categories 4–5 is recommended for studies that focus on one or more EV subtypes (e.g. small EVs < 200 nm, vs larger EVs: category 4), or that have identified
a functional soluble factor in EVs (category 5).

  1. All EVs bear proteins associated with the membrane or outer membrane (prokaryotic
    cells), or with plasma membrane and/or endosomes (eukaryotic cells). To demonstrate the presence of a lipid bilayer in the material analysed, at least one transmembrane or GPI-anchored extracellular protein must be shown. Examples include mammalian proteins expressed (nearly) ubiquitously in all cell types (1a), and proteins specifically expressed in some cells (1b).
  2. In all EVs, the lipid bilayer encloses cytosolic material (eukaryotic cells, Gram-positive bacteria) or periplasmic material (Gram-negative bacteria) from the secreting cell. To demonstrate that the material analysed contains more than open cell fragments, at least one cytosolic/periplasmic protein with lipid or membrane protein-binding ability must be shown (2a). Other cytosolic proteins are more promiscuously associated with EVs and other structures and thus should be only optionally used as EV markers (2b).
  3. Purity controls include proteins found in most common co-isolated contaminants of EV preparations: depending on the source of EVs, expected contaminants from category 3a (lipoproteins and serum-derived materials), or 3b (urine), should be evaluated.
  4. Proteins present in subcellular compartments other than the plasma membrane and endosomes, which may be present in certain EV subtypes (eukaryotic cells).
  5. Soluble extracellular proteins with functional activities (cytokines, growth factors, extracellular matrix) may be detected in EVs: their mode of association to EVs (via
    a specific or promiscuous receptor? Or internal?) should be determined. (See Table 2 part d), below.
    Still valid, but has evolved with a rapidly increasing number of techniques used to analyze EVs.
    i. Techniques providing images of single EVs at high resolution, such as electron microscopy and related techniques, scanning-probe microscopy (SPM) including atomic-Force microscopy (AFM), or super-resolution microscopy: these techniques are not interchangeable in the information they provide. When reporting results, both close-up and wide-field images must be provided.
    ii. Single particle analysis techniques that estimate biophysical features of EVs from other techniques than high-resolution images: size measured by resistive pulse sensing (electric field displacement), or light scattering properties [nanoparticle tracking analysis (NTA), high resolution flow cytometry, multi-angle light scattering coupled to asymmetric flow field-flow fractionation (AF4)]; or fluorescence properties [fluorescence correlation spectroscopy (FCS), high-resolution flow cytometry]. Chemical composition measured by Raman spectroscopy.
    Other techniques are being developed that may combine these two categories but have not yet been widely used (see 4c p.20).
    Whatever technique is used, all experimental details for both acquisition and analysis must be reported.
    Note that not all techniques are equally adapted to all EVs: large EVs (> 400 nm) and very small EVs (< 50 nm) are not well quantified by all NTA; small EVs are not easy to detect by most common flow cytometers. Some large EVs (and aggregates of small EVs) can be imaged by light/fluorescence microscopy. EVs smaller than the refraction limit or resolution of a microscope can still be detected by fluorescence, but no structural information can be obtained, and a single EV cannot be distinguished from a small EV cluster purely based on structural details.
    MISEV2018 additional characterization. We now recommend that the topology of EV- associated components be assessed, that is, whether a component is luminal or on/at the surface of EVs, at least for those required for a given EV-associated function. Topology may be particularly important for certain classes of biomolecules. Protease and nuclease digestions, detergent permeabilization, and antibodies to outer epitopes (should bind) or inner epitopes (should not bind) can be used to probe topology.
    b) General characterization.
    Show:
    i. At least three positive protein markers of EVs, including at
    least one -transmembrane/lipid-bound protein -cytosolic protein
    ii. At least one negative protein marker
    c) Characterization of single vesicles: use two different but complementary techniques, for example:
    i. electron or atomic force microscopy (and show both close-up and wide-field)
    ii. single particle analyzers (not electron microscope-based)
    d)

latter is not possible, for instance due to culture con- ditions (such as periodic collection in continuous bior- eactor-based cultures [162]), number of cells at initiation of culture, expected doubling time, and fre- quency of collection must be indicated. For some bio- logical fluids, like urine, the volume depends strongly on pre-analytical conditions (especially intake of liquid by the donor), thus additional means of normalization should be considered, such as urinary creatinine, as routinely done in the clinic for albumin [163].
EVs have a particulate structure and contain pro- teins, lipids, nucleic acids, and other biomolecules. Quantification of each of these components can be used as a proxy for quantification of EVs, but none of these values is necessarily perfectly correlated with EV number.
Particle number can be measured by light scattering technologies, such as nanoparticle tracking analysis (NTA); by standard flow cytometry for larger EVs [164–167] or high resolution flow cytometry for smal- ler EVs [127,168–176]; by resistive pulse sensing (RPS) for a wide range of sizes, depending on pore size [177]; by cryo-EM [174]; by a platform combining surface plasmon resonance (SPR) with AFM [178]; or by other techniques with similar capabilities. Accurate quantitation may be possible only within a certain con- centration and size range that varies by platform; where possible, this range (or the minimum and maximum diameter measured) should be reported along with concentration. The method of volume determination in flow cytometry should be reported and potential swarming/coincidence artefacts controlled for [179]; a more detailed guideline article on specifics of flow cytometry analysis of EVs is in preparation by mem- bers of ISEV, ISCT and ISAC. Some devices for particle quantification have the advantage of providing accurate sizing information amongst a complex mixture of par- ticles (see Table 2-c: single vesicle analysis). This is not the case for dynamic light scattering (DLS), which is accurate only for monodisperse particle populations [180]. Particle counting by light scatter, RPS, and simi- lar techniques typically results in overestimation of EV counts since the techniques are not specific to EVs and also register co-isolated particles including lipoproteins and protein aggregates. Possibly, ongoing development of fluorescence capabilities of NTA devices may ulti- mately allow EV-specific measurement [181], although assay sensitivity and the tendency of labeling antibodies and lipid dyes to form particles pose substantial hur- dles to such applications [127,182]. Additionally, par- ticle counting technologies may be biased towards certain particle size ranges (especially 50–150 nm [183,184]) because of pore sizes (RPS), size of
calibrator used, sensitivity (for example, smaller parti- cles scatter less light), and ability to cope with multi- dispersity (DLS versus NTA) [185]. Finally, proprietary software used for analysis of data from each device may apply unknown selection and other processing of data, resulting in differences in absolute values obtained by different software or different versions of the same software (see example in [183]).
Total protein amount can be measured by various colorimetric assays [Bradford or micro-bicinchonic acid (BCA)] or fluorimetric assays, or by global protein stain on SDS-PAGE. The EV sample concentration must be within the linear range of the reference curve. However, protein quantification can result in overestimation due to co-isolated protein contaminants (such as albumin from culture medium or plasma/serum), especially when the less specific methods of EV separation are used, or conversely can prove not sensitive enough if highly specific methods yield pure EVs. In addition, results may vary depending on the use or not of detergent to disrupt EVs and expose the entire protein content prior to performing the assay; nature and concentration of the detergent must be indicated.
Quantification of total lipids can be achieved, e.g. by sulfophosphovanilin assay [186], by measuring fluorescence of phospholipid dyes that fluoresce only when incorporated into lipid bilayers, such as DiR [187], or by total reflection Fourier-transform infrared spectroscopy [188]. However, the latter requires spe- cialized equipment, and the former two types of assays may be insufficiently sensitive for small amount of EVs. In addition, whether these techniques equally detect all EVs independent of their specific lipid com- position must still be established.
Quantification of total RNA can be performed by global RNA assays including profiles obtained by capil- lary electrophoresis instruments (see recommendations in Table 1 of [56]). Such measurements are difficult to recommend at this time for EV quantification or purity assays, though, since exRNAs associate in abundance with other circulating and potentially co-separating entities: chiefly ribonucleoproteins [189,190], but also a range of particles including exomeres [112] and lipo- proteins [191]. RNA measurements remain, however, an important parameter to report in studies of extra- cellular RNA.
Quantification of specific molecules. Other methods of EV quantification, like ELISA [192] bead-based flow cytometry [193,194], aptamer- and carbon nanotube- based colorimetric assays [195], and SPR on surfaces such as antibody-coated nanorods [178,196,197], can be used to quantify the amount of one or more specific molecules in the EV preparation. These are generally
JOURNAL OF EXTRACELLULAR VESICLES 15

16 C. THÉRY AND K. W. WITWER
proteins (usually the tetraspanins CD9, CD63 and/or CD81, but sometimes tumor-specific proteins or other molecules such as lipids [139]) and can be used to estimate the amount of EVs containing this particular component, rather than total EVs. These methods pro- vide additional information to the above methods and are in line with characterization recommended in part 4b (p.16).
Single and multiple measures and implications for purity. Quantification methods are the most informa- tive for EVs recovered by separation methods with the highest expected specificity (Table 1a-category 3), and for these preparations, one quantification method may suffice; in contrast, more than one quantification should be used for EVs recovered from low-specificity methods. Importantly, ratios of the different quantifi- cation methods may provide useful measures of purity. For example, protein:particle ratio [198,199], protein: lipid ratio [186,188,200] and RNA:particle [201] have been proposed as possible purity metrics, although their applicability across protein, lipid, RNA and par- ticle quantification methods remains to be established. Techniques that measure multiple parameters at once, such as colloidal nanoplasmonic assays or infrared (IR) spectroscopy [188,199] may be good optional methods, despite the need for specific sensors or other equipment.
Absolute EV sizing and counting methods are currently imperfect and will require further improve- ment, aided by appropriate EV reference standards that are now in development [202]. Nevertheless, current methods can provide a reasonable indication of particles per volume and particle size distributions that are best interpreted when combined with gen- eral (Table 2b) and single-particle (Table 2c) characterization.
Characterization of EVs by their protein composition
Selection of proteins for use as EV markers. Since MISEV2014, the growing recognition of the existence of many different types of EVs, of different sizes and cellular origins, has led to publication of several studies comparing the protein composition of at least two sub- types of EVs isolated from the same secreting cells. Some studies compared EVs recovered by medium speed centrifugation (called large oncosomes [203], ecto- somes [204], microvesicles [205], cell debris [206], or large [207] or medium [208] EVs), with those recovered by 100,000 x g ultracentrifugation (called exosomes in the first four studies, small EVs in the last two), and several of these applied additional separation in density gradients. Another study used differential filtration to
separate large microvesicles retained by 0.65 micron filters, and small “exosomes” passing through 0.1 micron filters [209]. Others further separated the high speed pellet to identify subpopulations of small EVs bearing different surface markers such as A33 antigen (GPA33) vs EPCAM [19], lipid moieties binding Cholera Toxin, Annexin-V or Shiga Toxin [139], or tetraspanins CD63, CD9, and/or CD81 [208]. EVs were also separated by floating at different densities within a sucrose gradient (defined as high density “HD- exo” vs low density “LD-exo”) [210] or eluting at differ- ent time points in asymmetric flow field-flow fractiona- tion (AF4) (small “exo-S” vs large “exo-L”) [112]. These studies together provide a rich source of potential EV subtype-specific markers. However, since they were per- formed with different separation approaches and with different cellular sources of EVs, it is still not possible to propose specific and universal markers of one or the other type of EVs, let alone of MVB-derived “exosomes” as compared with other small EVs.
Consequently, MISEV2018 does not propose mole- cular markers that could characterize specifically each EV subtype. Of note, although the ISEV board tried in MISEV2014 to propose general rules applying to all EVs, some suggestions of MISEV2014 were still biased by an “exosome–oriented” view of EVs. Specifically, Table 1 of MISEV2014 listed, as primary components to analyze in EVs, 2 categories of proteins present or enriched in EVs/exosomes (membrane bound and cytosolic proteins), plus another global category of proteins « not expected in EVs/exosomes » (such as mitochondria, Golgi, or nuclear proteins), and a last category of « contaminants ». In this updated version, MISEV2018, reference to exosomes and the proteins expected or not in them (the previously called “nega- tive controls” of “exosome” preparations) have been deleted, reflecting an evolving understanding of the subtypes of EVs and their associations with other enti- ties. Incorporation of any given component of the cytoplasm or other cellular compartment into an EV is determined by 1) proximity to the budding mem- brane and size of the EV (passive loading) and 2) specific association with the membrane and any energy-dependent processes (active loading). There are of course interactions between these two domains. Leaving aside the question of active loading, the larger the EV, the more likely any randomly selected mole- cular or organellar entity in the cell is to be incorpo- rated. Therefore, Golgi, endoplasmic reticulum, mitochondrial, or nuclear components may be excluded from small EVs (< 200 nm) that are presum- ably formed distant to these locations, or at least

strongly depleted relative to the cell (although we can- not exclude that particular components of such com- partments may end up in small EVs, especially in pathologic conditions that could affect incorporation). However, such proteins may be present in larger EVs, and even more so in a very large EV, ultimately the large oncosome [211], which may be as large as some cells and contain by definition any and all components except for a whole, functional nucleus. A single nega- tive control for such large EVs may thus be elusive.
Table 3 highlights three categories of markers that must be analysed in all bulk EV preparations to demonstrate the presence of EVs (Categories 1 and 2) and assess their purity from common contaminants (Category 3), but no universal “negative controls” rele- vant to a particular subtype of EVs are suggested. The three main categories are:
Category 1: Transmembrane or GPI-anchored proteins localized at the external membrane of pro- karyotic cells, and plasma membrane and/ or endosomes of eukaryotic cells represent hallmarks of any type of EVs: their pre- sence demonstrates the lipid-bilayer struc- ture specific of EVs, whether they bud directly off the plasma membrane or after
transit through the endosomal pathway; Category 2: Presence of cytosolic proteins (eukaryotic cells and Gram-positive bacteria) or peri- plasmic proteins (Gram-negative bac- teria) demonstrates that the analysed preparation displays the structure of lipid bilayers enclosing intracellular mate- rial, as expected for any EV. Proteins pre- sumably actively incorporated into EVs are those with ability to bind to mem- branes or to cytosolic sequences of trans- membrane proteins. Others, like cytosolic enzymes or cytoskeletal proteins are more
promiscuous EV components;
Category 3: Some proteins are major constituents of non-EV structures often co-isolated with EVs. Evaluation of their presence helps to assess the degree of purity of the EV pre- paration. In biofluids like blood plasma, EVs have been reported to co-isolate with other particles, including lipoproteins [212] and a variety of non-integral pro- teins, such as albumin or soluble acetyl- cholinesterase. We therefore propose apolipoproteins A1/2 and B (APOA1/2, APOB), and albumin (ALB) as the best negative markers to date [213] for
Category 4:
plasma/serum EVs, and EVs from cells cultured in the presence of bovine serum, or liver cells that secrete lipopro- teins, although it cannot be excluded that a fraction of such markers may be speci- fically associated with some EVs [214]. As another example, in urine, Tamm- Horsfall protein (uromodulin/UMOD) forms aggregates that co-precipitate with EVs unless the fluid is chemically treated [215]. Overall, however, since we cannot propose a threshold of abundance of these proteins in EV preparations below which acceptable purity is reached, we stress that it may be more appropriate to assay and report depletion than to expect a binary presence/absence of proposed negative markers.
An additional category 4) of proteins should be evaluated if authors want to claim speci- ficity of their study to the small EV subtype- (s): Proteins localized in/on intracellular compartments of eukaryotic secreting cells other than the plasma membrane and endo- somes (i.e. components of the nucleus, mitochondria, endoplasmic reticulum, Golgi apparatus, autophagosomes, peroxi- somes) are found in some types of EVs, but a priori not enriched in the smaller EVs (approximately < 200 nm diameter) of plasma membrane or endosomal origin.
Finally, category 5) covers secreted or lumi- nal proteins that can associate with EVs by binding to specific (e.g. growth factor receptors) or to promiscuous (e.g. proteo- glycan, lipid) receptors on the EV surface: their identification in EV preparations should be accompanied by exploration of the cognate EV-associated receptor(s).
Category 5:
JOURNAL OF EXTRACELLULAR VESICLES 17
Methods to assess presence of proteins in EV prepara- tions. Several methods can be used to quantify pro- teins in or on EVs. Western blotting is the most commonly used, and it should be performed by loading side-by-side EV samples and source material lysates either in specified protein amount or in cell- equivalent amounts to determine if the analyzed pro- teins are enriched in EVs as compared with their pro- ducing cells. This comparison, however, can be easily performed only for analysis of EVs from cell culture conditioned medium; it is more difficult for biofluids (in which EVs may originate from cells in the fluid, but also from cells delimiting the fluid canals, and thus are

18 C. THÉRY AND K. W. WITWER
Table 3. Protein content-based EV characterization. At least one protein of categories 1a or 1b, 2a (optionally 2b), 3a or 3b must be analysed to demonstrate the EV nature and the degree of purity of an EV preparation. Analysis of proteins of category 4 is required when claiming specific analysis of small EVs, and of category 5 to document functional activities. Examples of proteins commonly found in mammalian cell-derived EVs are provided, but other proteins that fall into the provided categories can be used, particularly for analysis of EVs from prokaryotic (bacteria) or non-mammalian eukaryotic sources (including parasites and plants). XX = human gene names. XX* or XX** used for families of multiple proteins, for example for integrins: ITGA* indicates any integrin alpha chain.
Category Use for
1- Transmembrane or GPI-anchored proteins associated to plasma membrane and/or endosomes
2- Cytosolic proteins recovered in EVs All EVs
3- Major components of non-EV co-isolated structures
4- Transmembrane, lipid- bound and soluble proteins associated to other intracellular compartments than PM/endosomes
5- Secreted proteins recovered with EVs
All EVs
All EVs as purity control
Subtypes of EVs (e.g. large oncosomes, large EVs) and/ or pathologic/atypical state
Functional component of EVs: need to determine the mode of association with EVs
1a: non-tissue specific.
Tetraspanins (CD63, CD81, CD82); other multi-pass
2a: with lipid or membrane protein-binding ability. ESCRT-I/II/III (TSG101, CHMP) and accessory 3a: lipoproteins (produced by liver, abundant in plasma, serum). 4a: nucleus. Histones (HIST1H); Lamin A/ 5a: Cytokines and growth factors. e.g. TGFB1/2; IFNG, VEGFA, FGF1/2, PDGF, EGF, interleukins (IL)… membrane proteins (CD47; heterotrimeric proteins: ALIX (PDCD6IP), VPS4A/B; ARRDC1; Flotillins-1 and 2 (FLOT1/2); caveolins (CAV); EHD; RHOA; annexins (ANXA); Heat shock proteins HSC70 (HSPA8), and HSP84 (HSP90AB1) note that both are abundant also in exomeres; ARF6; syntenin (SDCBP); microtubule-associated Tau (MAPT, neurons)
C (LMNA)
G proteins GNA) MHC class I (HLA-A/B/C, H2-K/D/Q), Integrins (ITGA/ITGB), transferrin receptor (TFR2); ApolipoproteinsA1/2 and B APOA1/2, APOB; APOB100; albumin (ALB) LAMP1/2; heparan sulfate proteoglycans including syndecans (SDC); EMMPRIN (BSG); ADAM10; GPI-anchored 5ʹnucleotidase CD73 (NT5E), complement-binding proteins CD55 and CD59; sonic hedgehog (SHH)
1b: cell/tissue specific.
Some TSPANs: TSPAN8 (epithelial cell), CD37 and
2b: promiscuous incorporation in EVs (and possibly exomeres). Heat shock protein HSP70 (HSPA1A), cytoskeleton: actin (ACT), tubulin (TUB); enzymes (GAPDH)
3b: protein and protein/ nucleic acid aggregates. Tamm-Horsfall protein (Uromodulin/UMOD) (urine); ribosomal proteins
4b: mitochondria
IMMT, cytochrome C (CYC1);
5b: adhesion and extracellular matrix proteins. Fibronectin (FN1), Collagen (COL
), MFGE8; galectin3-binding protein (LGALS3BP), CD5L; fetuin-A (AHSG)
CD53 (leukocytes), CD9 (absent from NK, B and
TOMM20
some MSC); PECAM1 (endothelial cells); ERBB2 (breast cancer); EPCAM (epithelial);
CD90 (THY1) (MSCs);
CD45 (PTPRC) (immune cells), CD41 (ITGA2B) or
CD42a (GP9) (platelets); Glycophorin A (GYPA) (RBC); CD14 (monocytes), MHC class II (HLA-DR /DP/DQ, H2-A); CD3 (T cells); Acetylcholinesterase/AChE-S (neurons), AChE-E (erythrocytes); amyloid beta A4/APP (neurons); multidrug resistance-associated protein (ABCC1)
4c: secretory pathway (endoplasmic reticulum, Golgi apparatus) calnexin (CANX); Grp94 (HSP90B1); BIP (HSPA5), GM130 (GOLGA2)
4d: others (autophagosomes, cytoskeleton…).
ATG9A, Actinin1/4 (ACTN1/4), cytokeratin 18 (KRT18)

Text Box 1. Example of a change from MISEV2014 as a paradigm for considering negative markers.
Argonaute proteins, chiefly AGO2, the most abundant family member in mammals, have been previously proposed as negative markers of EVs. In blood plasma and perhaps other fluids, most extracellular AGO is indeed found outside of EVs [189,190,216]. However, the canonical view of biogenesis of microRNAs (miRNAs) and their subsequent protection from degradation depends on AGO proteins. According to this reasoning, if a mature miRNA is detected, whether in cells, in EVs, or elsewhere, it is most likely protected by AGO (although other associations have been reported [56]). Limits of detection of proteins versus amplified nucleic acids may have contributed to the interpretation that miRNAs are present in EVs without AGO protein. It is in any case now established that AGO can be found in mammalian [217,218] and non-mammalian [219] EVs, not just outside EVs.
Text Box 2. Example of a possible change from MISEV2018 in the future as markers of non-EV co-isolated components.
The Lyden group recently reported that lipidic structures called “exomeres” contain lipids and a limited set of membrane-bound proteins, but do not feature a lipid bilayer and thus do not qualify as EVs. Exomeres were found to co-isolate with small EVs recovered from many cultured cell lines [112] but could be separated from EVs by AF4 (which unfortunately is not implementable in some laboratories). It may be necessary to evaluate this potential contaminant in all EV preparations. Proteomic analyses of exomeres identified various proteins in Table 3-category 2, above: heat shock proteins (HSPA8, HSPA1A, HSP90AB1), actin (ACT), tubulin (TUB), and GAPDH. Such proteins thus probably do not qualify as EV-specific components. In addition, a few proteins were identified exclusively in exomeres: hemoglobin (HBA1/A2), IDH1, MAT1A, transmembrane FAT4 (protocadherin, transmembrane plasma membrane), and EXT1/2 (exostosin, transmembrane, Golgi). Although this exhaustive study was performed with several cell lines, suggesting that the listed proteins are good candidates as specific markers of exomeres co-isolated with EVs, these results still have to be confirmed by other groups to reach validation and inclusion in the next update of the MISEV guidelines.
difficult to attribute in bulk to any given cell type). Flow cytometry of EVs decorating beads or of bulk EV populations (i.e. not designed to analyse single EVs) can be used, but with care to use appropriate negative controls (antibodies alone, isotype controls, etc) [48,220]. Numerous multiplexing approaches have been developed to analyse simultaneously the presence of a pre-designated set of surface protein markers on EVs. For instance, one platform uses flow cytometry after capture on an array of 37 beads, each bearing a specific antibody [100,194]. Other approaches use fluorescence scanning [221] or surface plasmon resonance [222,223] to quantify EV binding to a surface coated with antibodies to different antigens. These methods are population-level, not single-EV techniques, since the final result is an arbitrary unit of signal for a given protein marker in the global EV population. Finally, mass spectrometry has become increasingly economical and accessible for many
laboratories, allowing fingerprint-type assessment of many proteins at once. For an exhaustive review on the currently available methods of EV analysis, see [224]. Undoubtedly, new techniques and devices will become available, including commercially, in the near term. Besides the potential of such devices for diagnos- tic purposes based on amount and type of secreted EVs, one of the challenges associated with the use of such devices is whether they might allow sufficient quantities of specific EVs to be purified so that cargo (RNAs, proteins) content can be analysed on a profiling level.
Non-protein components as markers of EVs. Although proteins are emphasized in the literature and here, phospholipids present in lipid bilayers are also poten- tial positive controls for the presence of EVs [225,226], albeit non-specific as other particles (lipo- proteins) may also contain these. As an example, albeit one that may not be exposed on all EVs, outer leaflet phosphatidylserine (PS) can be evidenced indirectly by binding of fluorescently-labeled PS- binding proteins, such as Annexin V [139,167] or the C1C2 domain of lactadherin/MFGE8 [186,227]. Glycosphingolipids are similarly evidenced by binding of GM1 ganglioside [139,228]. Other lipids including cholesterol, sphingomyelin, ceramide, and phosphati- dyl-choline/ethanolamine/inositol can be detected by a variety of methods [225] including Raman spectro- scopy, which was recently used to analyze lipids in dried EVs [229]. However, in which ratio cholesterol, sphingomyelin, ceramide, and phosphatidyl-choline /ethanolamine/inositol are in EVs, and how this dif- fers from ratios found in lipoproteins is not yet estab- lished: additional comparative lipidomic studies of separated EVs and liproprotein subtypes may be informative.
Alternatively, dyes that are activated by intra- cellular components can be used to label EVs. Calcein and CFSE are examples of cell permeant, non-fluorescent pro-dyes that are cleaved by intra- cytoplasmic enzymes, resulting in an impermeant fluorescent molecule; labelling therefore theoreti- cally differentiates intact EVs from linearized mem- brane fragments, provided that the required enzymes are present in EVs [230,231]. Other tools, like a recently reported protein- and lipid- binding dye, di-8-ANEPPS [231], may be worth evaluating for sensitivity and specificity. Additionally, appropriate negative controls are necessary in all studies, such as dye only and dye plus EV-depleted matrix.
JOURNAL OF EXTRACELLULAR VESICLES 19

20 C. THÉRY AND K. W. WITWER
Concerning nucleic acids, both DNA and RNA have been detected in EVs. RNA in EV preparations can be detected by dyes [232], although some dyes may also detect non-EV-associated RNA. It is possi- ble that some nucleic acid species could serve as negative or positive markers of some EVs. For exam- ple, strictly nuclear RNAs might be identified as negative markers in the future, while RNA associated only with cytoplasmic complexes (e.g. ribosomal RNAs or mitochondrial DNA) [233,234] may be more likely to be present in certain EVs. However, several nuclear RNAs have been found in EVs, and a variety of data have been presented on specific versus non-specific incorporation of RNAs into EVs or subtypes of EVs [56,217,235–241]. More research is thus needed before specific recommendations can be made for using nucleic acids as specific markers of EVs or EV subtypes.
Single vesicle analysis
Quantification and global protein composition apply to bulk EV preparations. However, it is important to provide some information on the individual EVs pre- sent in such bulk preparations. Two different approaches provide different types of information:
i. Techniques allowing visualization of single EVs at high resolution may provide information on both the structure and the composition of EVs, when combined with antibody-mediated detec- tion of EV components. However, they may be difficult to exploit in a quantitative manner, with analysis of sufficient number of EVs to reach statistical power.
EVs with diameter larger than the diffraction limit of light (~ 200 nm) can potentially be visua- lized as single EVs by regular fluorescence micro- scopy and regular flow cytometry. For EVs smaller than this limit, confocal microscopy can detect fluorescent dots, but these dots can correspond either to very bright vesicles with diameter smaller than 200 nm [242], or to clusters of small dim vesicles, without the possibility to discriminate between the two [243].
All EVs can be analyzed by: electron microscopy or by other imaging techniques: SEM [244], TEM by contrasting and embedding in a mixture of uranyl compounds and methylcellulose to maintain the bilayered morphology, cryo-EM [174,245,246]; scan- ning-probe microscopy (SPM) including atomic force microscopy (AFM) [247]; and super-resolution
microscopy [248,249]. Note that these various techni- ques are not necessarily interchangeable or capable of providing images of comparable quality. For example, cryo-EM clearly shows the lipid-bilayer, preserves EV size better than the dehydrating conditions used to fix samples for TEM, and may be more quantitative, as all particles in a given volume can be imaged, not just those that adhere to a surface (the grid).
ii. Single particle analysis techniques that do not pro- vide high-resolution images but calculate biophysical parameters of single EVs can be used to quantify a large number of EVs with a higher statistical power than many single-EV techniques. For instance, size can be inferred from particle displacement pattern by nanoparticle tracking analysis [184,185,250]; light scattering and/or fluorescence detection in high resolution flow cytometry [251–255]; multi-angle light scattering combined with asymmetric flow field-flow fractionation (AF4-MALS) [256]; displa- cement of an electrical field in tunable resistive pulse sensing-based devices; or fluorescence correlation spectroscopy (FCS) [257–259]. Chemical composi- tion can be evaluated by Raman tweezers micro- scopy [251–253].
Other recently developed technologies aim to combine the advantages of imaging with analysis of large numbers of events. They are, however, less commonly used in the EV field and require further validation in multiple laboratories. For instance, an imaging flow cytometer that captures images of single cells going through a fluidic chan- nel can be used, with a carefully designed set of controls and settings, to image EVs of all sizes after labeling with fluorescent lipids, proteins, or antibo- dies [260,261]. Another recently designed device involves capture of biotinylated EVs on a streptavidin surface, followed by successive rounds of staining with fluorescent antibodies, ima- ging, and quenching followed by additional rounds of labeling [262]. Another example uses single- particle interferometric reflectance imaging sensing of EVs captured on antibody-coated chips [263,264], which can be accompanied by fluores- cence measurements.
Whatever technique is used, all experimental details must be reported. These include the brand and version of the instrument and the software, the settings used for acquisition (diluent buffer, camera, flow rate, threshold…) and, for analysis, the precise process for EM or fluorescence microscopy and how the imaged areas were selected, as well as controls and calibration

information where relevant. For flow cytometry, an ISTH working group has recently issued recommenda- tions [265]. Given the wide range of techniques and platforms available, many of which remain under development for EV applications, MISEV2018 cannot yet give precise protocol recommendations.
New recommendation: determine the topology of EV-associated components
Importantly, the luminal versus surface topology of var- ious EV-associated components, including nucleic acids, proteins, glycans, etc, is not entirely strictly determined. Theoretically, components localized in the cytosol of EV- secreting cells should be inside EVs, and hence protected from mild degradation by proteases or nucleases. While this protection is usually observed, some studies have unexpectedly found proteins [266], RNAs [267], and DNA [41] on the EV surface and sensitive to digestion. It is not yet clear whether this unexpected topology is due to debris from dead or dying cells, or is instead the outcome of as-yet unknown mechanisms of transport of intracellular compartments across membranes that could occur in some physio- or pathological conditions. Certainly, even a small degree of contamination with intracellular material (with the reverse topology to EVs) would complicate interpretation.
Topology may also be important for function. A luminal active component would require mem- brane fusion or two membrane transport events to achieve function in a recipient cell, whereas if it is exposed at the surface of the EV, it may affect target cells without EV-cell fusion. As a result, we advise that the actual topology of putative active compo- nents be determined by performing mild digestions, permeabilizations, or antibody studies. To give an example, one might adopt and adapt methods devel- oped to assess protein insertion within the endoplas- mic reticulum [268]. Various protocols have also already been published in the EV community [56,217,242,266,269,270].
Example of a biochemical approach for assessing surface exposure of EV components (see for instance use in [266,270]):
for a given preparation of EVs, prepare four samples:
(1) aliquot 1 is untreated;
(2) aliquot 2 is treated with a degrading enzyme
alone, which should degrade only surface-
exposed components (e.g. proteins);
(3) aliquot 3 is treated with enzyme and detergent (such as Triton X-100), which will ensure degra- dation of both surface and internal components (to verify that enzymatic treatment worked).
Note that detergents may also enhance enzy- matic digestion of certain molecules indepen- dently of permeabilization of membranes; an alternative to detergent is saponin, which per- meabilizes membranes.)
(4) aliquot 4 is treated with detergent alone, to make sure that detergent does not affect the downstream analysis.
For each sample, the cargo of interest is then ana- lyzed (after careful neutralization of the enzyme) by an appropriate method (SDS-PAGE, RT-PCR, PCR…). Extinction of the signal within the enzyme- treated, detergent-untreated aliquot indicates surface exposure of the cargo.
For RNA and DNA analysis, RNAse or DNAse must be used together with proteinase to allow access of nucleases to protein-shielded nucleic acids.
Alternatively, the topology may be determined using flow cytometry and fluorescence microscopy with anti- bodies directed towards either external or cytoplasmic epitopes on EV membranes. Single vesicle characteriza- tion by EM or AFM coupled with immunolabeling could not only provide validation of surface-accessible targets or internal targets (with permeabilization), but also aid in differentiating differences in topology among EVs of different sizes [247].
Consensus: 97% of MISEV2018 Survey respondents endorsed the structure of the characterization section. The original version of Table 3 included more proposed markers of EV subtypes. However, although the responses ranged from 69% to 93% agreement on dis- creet categories of markers, some concern was commu- nicated about the universality of subtype-specific markers based on the existing evidence. As a result, the Table was thoroughly revised to focus on classes of markers that can be applied to all EVs, not just those from certain cell types or organisms. These revisions further establish the applicability of MISEV2018 to all EVs and EV sources.
An original section on negative controls received 79% agreement and 56 comments; as a result, the approach to negative controls was substantially amended.
95% of respondents agreed that the current focus on protein markers is justified; however, lipids are now included. Although 13 respondents suggested that RNA markers could be used as generic or subtype EV mar- kers, and several mentioned post-translational modifi- cations, these comments were supported with limited or (usually) no references. Based on the limited endorse- ment of RNA markers and ongoing uncertainty about enrichment of RNAs in EVs, no specific recommenda- tions are made by MISEV2018. Instead, further research is expected and encouraged.
92% agreed with the topology recommendation (4c).
JOURNAL OF EXTRACELLULAR VESICLES 21

22 C. THÉRY AND K. W. WITWER
Functional studies: how MISEV2014 evolves in 2018
Table 4 summarizes the previous and updated recom- mendations on functional analysis of EVs. More detailed justification for these recommendations and proposed protocols follows the Table. The goals of these recom- mendations are to avoid over-interpretations or classical artefacts when analyzing functions of EVs. It is important to consider several issues when attributing a functional activity to EVs in general, or an EV subtype in particular. We describe here the controls and processes that should be included in all functional studies, unless limited amounts make it impossible to perform them. For clinical applications, for instance, after a first step of pre-clinical validation following these recommendations, systematic analysis may not be possible (see previous Position Paper on clinical applications) [95].
Determine the specific versus common functions of different types of EVs
An important point to keep in mind is that, when analyzing exclusively the function of a single type of EV (for instance either small EVs or large EVs that have been called ectosomes, microvesicles or micropar- ticles in different studies), one may miss the most active EV subtype for the particular function studied. Even if a function is found in the concentrated small EV preparation, it could also be present, and even possibly more concentrated, in other EV subtypes that had been eliminated during the small EV isolation process: keeping large EVs (e.g. “microvesicles”) and comparing their activity to that of small EVs should be
Table 4. EV-associated and EV-excluded biological activities. Major recommendations of MISEV2014.
a) Dose–response studies
b) Negative or background controls.
For conditioned medium, negative control = complete medium that has not
been conditioned by cells, but still processed in the same way as conditioned medium
c) Controls to assess influence of soluble or non-EV macromolecular components
c-i.) Density gradients or other rigorous separation method to show activity is intrinsic to EVs, not just associated
or c-ii.) EV depletion to remove activity
or c-iii.) EV/cell labelling (e.g. fluorescent labelling, with careful
interpretation)
a first step in all functional studies. In addition, when a function found in EVs may be due to soluble mole- cules that may or may not associate specifically with EVs, one must consider the possibility that the EV- associated function is only a minor fraction of the non- EV-bound soluble protein. Comparing quantitatively the effects of EV fraction(s), EV-depleted fraction(s), and also the unfractionated initial fluid, will identify the relative contributions of each to total activity.
Ideally, all functional studies of EVs recovered from any source (biofluid, conditioned medium…) could start by a crude separation of broad categories of EVs (e.g. large versus small EVs versus EV-depleted frac- tion, separated by successive centrifugation, filtration, or chromatography). Importantly, to ascribe function to different categories of EVs, each fraction should be retained for side-by-side activity analysis. However, authors who wish to analyse the function of only one subtype of EVs can justify this choice by presenting
Validity and/or Update in 2018
Still valid
In addition: quantitative comparison of the activity of conditioned medium
or biofluid 1) before, 2) after elimination of EVs, and 3) the EVs themselves, keeping in mind that the EV fraction may include co-isolated /contaminating materials.
Additional suggested control: quantitative comparison of the activity of the targeted versus the “discarded” EV subtypes (see part 1 below)
Still valid.
For biofluids, negative controls of disease-associated functions = fluids from
healthy, untreated or otherwise matched donors
Still valid.
Increasingly, it is possible to separate EVs from non-EV components by
multiple methods, e.g. density gradients and size exclusion
chromatography.
It is recommended that functional assays be performed after rigorous
separations, comparing EV and non-EV fractions to identify what proportion of activity is associated with each fraction (in case it is not EVs). If the activity is primarily associated with EVs, depleting the EVs should also deplete the activity.
The refined separation must be performed at least for a set of biological replicates, but not necessarily systematically afterwards.
Text Box 3.
As an aside, although we do not go into great detail on this point, many functional studies presume or investigate EV uptake. Time- courses and environmental determinants of EV uptake have been studied for some time [272-274], but challenges exist [275]. Detection within the cell of signal from an EV-labeling dye or other entity does not necessarily mean that the EV or its cargo has been internalized. Some labeling substances are very long-lived, can exist separate from the presumably labeled entity, and can form EV-mimicking particles that are difficult to separate from EVs. Another potential artifact is that labeling EVs with lipophilic or surface-coating fluorophores may modify physicochemical characteristics of EVs, thus altering detection mode and/or uptake by target cells. Although we cannot yet make firm recommendations, we urge researchers to be aware of these issues and to consider that each specific EV-donor/EV-recipient pair may behave in a different manner.

practical or theoretical reasons for selecting specifically this subtype for the particular experimental purpose, and further proceed with functional analyses.
Before performing functional assays, it is advisable to determine whether EVs are detected in the different fractions, in terms of total proteins, or number of particles, or ratio of total nucleic acids (RNA, DNA), or total lipids to particles (i.e. substance x-to-particle ratios): if none of these EV-associated components are detectable, the “empty” fractions may be discarded for further functional tests, once a preliminary experiment has shown lack of activity. However, any change of conditions in generation of the biofluid from which EVs are isolated (i.e. different culture conditions or treatment for the cells, different types of patients for biofluids) should be followed by re-analysis of all EV categories.
Example of an approach to compare activity of broad subcategories of EVs after separation by dif- ferential centrifugation (see example of use in [220])
From a given volume of conditioned medium:
several low speed centrifugations, transferring supernatant into a new tube each time. The pellet contains cells.
supernatant: 1x centrifugation at medium speed, to collect large EVs (if mostly live cells), large apoptotic bodies, etc., with the pellet largely devoid of cells
supernatant: 1x centrifugation at intermediate speed to collect a pellet enriched in medium size EVs and/or aggregates of small EVs (to be checked by EM)
supernatant: 1x centrifugation at high speed to col- lect a pellet enriched in small EVs.
Each pellet is resuspended once in buffer/medium and re-centrifuged at the same speed (= washed) before resuspension in a given volume of buffer/medium.
How to normalize amount of EVs used for comparative functional studies
The most appropriate normalization strategy to com- pare quantitatively the functions of different EVs will depend on the scientific question. One can choose to normalize by either characteristics of the isolated EVs, or by the source material, or by co-isolated standards. Characteristics of isolated EVs would include particle counts, total amount of a biomolecule type in the EV sample (e.g. proteins, nucleic acids, or lipids), and con- tent or activity of specific EV-associated molecules. Source characteristics include the amount of matrix from which the EVs were obtained (initial volume of biofluid, initial mass of tissue, initial number of secreting cells, time of conditioning per cell, etc). Co-isolated standards would be traceable materials added into the
matrix prior to separation [202]. Multiple normalization strategies can be pursued [120], and, as emphasized elsewhere, dose-response studies are recommended regardless of normalization method. Unfortunately, there is no clear recommendation that can be made at this point on which normalization strategy is best. Instead, the choice of normalization must be reported and justified, and relevant details of alternative strategies should be provided. For example, when studying EVs obtained from some biofluids, e.g. blood derivatives, normalization by volume may be appropriate. For other fluids, such as lavage fluids and urine, initial volume is not easily compared between donors, so another strategy might be more appropriate. As another example, for in vitro studies, normalizing by levels of an EV component (proteins, lipids, RNA), or by particle number may be appropriate, but the rationale should be provided, and information on the number of secreting cells should also be recorded and reported.
The ISEV survey comments on this section evidenced broad disagreement about normalization strategy, parti- cularly on the relative merits of protein, nucleic acid, and lipid quantitation. Additional studies of normal- ization strategy should thus be encouraged.
Demonstrate that the activity is observed in the absence of direct cell-cell contact
Theoretically, an EV-associated function, like a soluble cytokine-dependent function, should be observed between two cells that are not in direct contact with each other. Therefore, it should be obtained when the EV-donor and an EV-recipient cell are cultured in vitro at a distance, through transwell co-culture systems or more sophisticated microfluidics-based culture devices, or by incubating the recipient cells with medium con- ditioned by the donor cells.
However, the physiological relevance of optimal conditions of these co-cultures, in terms of respective numbers of EV-donor and – recipient cells, cannot be strictly determined. If such assays yield positive results, they prove that transfer of a signal occurs in a cell contact-independent manner, in which case the next necessary step (next section p24), is to distinguish EVs from soluble components. Lack of positive results at this step would suggest that cell-cell-contact is neces- sary for the exchange of signal, but it could still occur by exchange of membrane–enclosed signals by transfer of plasma membrane-derived vesicles (like trogocyto- sis), or localized release of multivesicular body-derived vesicles at the cell-cell-contact (e.g. an immune synapse). Thus, a negative result argues against signal- ing at a distance but does not conclusively disprove local involvement of EVs.
JOURNAL OF EXTRACELLULAR VESICLES 23

24 C. THÉRY AND K. W. WITWER
Demonstrate that the activity is predominantly associated with EVs rather than with soluble mediators
Typically, an EV-associated activity is explored by 1) separation and concentration of EVs from a biofluid or cell culture media, 2) application of EVs to a recipient cell or organism, and 3) observation of a readout phe- notype. However, to convincingly argue that a detected readout/function is EV-borne, it must be determined that the activity is specifically enriched in EVs (possibly with non-EV components), and not instead due to low amounts of a highly active soluble molecule remaining non-specifically in the EV preparation. This point is particularly important when the proposed or suspected active molecule on EVs is a cytokine/growth factor/ metabolite usually described as secreted in a soluble form. For this step, one must compare quantitatively the activity present in/on the EVs versus in the remain- ing EV-depleted biofluid, using the same amounts of materials in terms of initial volume of biofluid. When evaluating the relative importance of EVs and soluble mediators, it may be worth remembering that EVs and soluble mediators may have combinatorial (e.g. syner- gistic) effects on cells [275,276].
Example of an approach to determine the respec- tive contribution of EV-bound and soluble non-EV- associated factors (see example of use in [200]):
from a given volume of biofluid from which cells have been completely eliminated
split supernatant into 2 equal parts;
keep one at 4°C while processing the other with the preferred protocol allowing extensive concentration of EVs, with separation from non-EV components, but without a need to separate EV subtypes (see Table 1).
Make sure to recover the biofluid from which EVs have been separated.
Compare activity of the total biofluid, the EV- containing pellet and the EV-depleted biofluid, using material coming from the same initial volume of biofluid.
If the unprocessed supernatant must be concen- trated to display activity, the supernatant after centri- fugation must be concentrated in the same way, and the pellet can be resuspended in the same volume of concentrated fresh medium as concentrated superna- tant, before performing the functional assay.
Demonstrate the specific association of the activity with EVs rather than with co-isolated components Especially when dealing with concentrated prepara- tions enriched in small EVs, one must keep in mind that such preparations potentially contain non-EV
components (ribonucleoprotein aggregates, lipopro- teins, exomeres, etc.). The proportion of such co- isolated components differs tremendously with the type of protocol used to separate EVs, with some (like polymer-based concentration) displaying particularly abundant contaminants, and also remnants of the pre- cipitating agent that can affect cell function [277,278]. In the case of cells infected experimentally or uninten- tionally (e.g. mycoplasma) with microbes, functional microbial factors may also be co-isolated with EVs. Therefore, the functional activity of an EV preparation may be borne by EVs, or by the additional compo- nents, or by a combination of both. One must deter- mine which of these three possibilities is the case. If small amounts of working materials do not make it possible to perform these additional investigations, the authors can explain this situation and interpret their data as activity present in EV-enriched preparations, rather than EV-specific activity.
Examples of protocols to demonstrate specific association of the activity with EVs or a given EV subtype:
From a concentrated EV preparation obtained by (ultra)centrifugation, or by centrifugal concentrator:
Option 1) separate contaminants by upward flota- tion into a density gradient, where only lipid-enclosed structures float upward in the tube: analyze separately activity of each fraction of the gradient in the func- tional assay; note that density gradient medium may have to be removed, because it can interfere with some functional assays. Alternatively, a control should be performed in which the chemical used for the density gradient is mixed with unseparated EVs.
Option 2) separate soluble contaminants from EVs by SEC column, where EVs elute in the first fractions, whereas proteins and ribonucleoproteins and some lipoproteins are eluted later: analyze separately activity of each fraction (or of pools of fractions containing EVs, intermediate, or free soluble components) in the functional assay;
Option 3) compare activity of intact EVs with that of detergent-treated samples [279]. Detergent will destroy vesicular structures, without affecting other non-lipidic particles. Of course, this technique also has limitations: 1. the effect of the same amount of detergent alone in the functional assay should be tested (it could affect target cells); 2. there could be a few contaminants that are sensitive to detergents as well (liposomes and some proteins).
Option 4) immuno-isolate from half of the biofluid all EVs bearing a surface marker of interest (for instance an integrin or a tetraspanin used to

characterize the EVs, but not expected to be required for the observed activity), and use the specific EV- depleted supernatant in the functional assay, side-by- side with the other half of unprocessed biofluid, to determine if activity has been depleted (hence was associated with the EVs of interest) or not. Using the immuno-isolated EVs in the functional assay may com- plicate interpretation, since the immunoprecipitate will contain antibodies and beads used for isolation that may profoundly affect interaction of EVs with the target cells. Elimination of protein-bearing EVs by immunoisolation must be demonstrated by showing the amount of EV-associated protein in biofluid before (all) and after immuno-isolation (none), and in the immuno-isolated sample (all).
Other options may arise from ongoing evolutions of the field. See section “EV separation and concentra- tion” p11. As exemplified by AF4 [112,114], combina- tions of ultrafiltration and SEC [280], and tangential flow filtration combined with other filtration steps [106], novel EV separation processes are constantly being developed and published.
Determine whether a function is specific to exosomes, as compared with other small EVs
As highlighted here, it is now clear that different types of EVs can present functional activities that are as important to explore as those elicited by late endo- some-derived exosomes. However, in the last decade, many studies have focused exclusively on demonstrat- ing association of a given function with exosomes. This section explains the technical limitations of such stu- dies, and why they are not sufficient to conclude, as is generally done, that exosomes have specific functions compared with other EVs.
In particular, numerous approaches have been taken to inhibit or stimulate exosome secretion in loss- or gain-of-function experiments. For example, in mam- malian cells, exosome secretion has been decreased (100% inhibition is almost never achieved) by inhibit- ing neutral sphingomyelinases and ceramide genera- tion (by shRNA, genetic editing, or drugs such as GW4869, spiroepoxide, cambinol and others) [281– 284]; inducing ISGylation, which promotes lysosomal degradation of MVB proteins [285]; blocking Rab GTPases (by expressing dominant-negative mutants or silencing or knocking out Rab27, Rab11, Rab35, or others) [286–288], other small GTPases (RAL-1 [289]), SNARE proteins (YKT6 [290]), upstream regulators of protein sorting into MVBs (like SRC [291]), or cytos- keletal proteins (cortactin [292], microtubules [293]); or using other drugs (e.g. the sodium channel blocker amiloride [294]). Conversely, ionophores such as
ionomycin (calcium signaling) or monensin (sodium transport) [295,296], or drugs inhibiting endosomal acidification and/or autophagic degradation (bafilomy- cin A1 [297,298]) have been used to stimulate exosome secretion. Of note, drugs inhibiting EV uptake (e.g. heparin [299]) could lead to enhanced recovery of EVs, which could be misinterpreted as an increase of exosome release.
A few studies have also proposed ways of modulat- ing secretion of plasma membrane-derived EVs in mammals: the ARRDC1 protein uses the ESCRT machinery to induce budding of small EVs at the PM, and its blocking or depletion inhibits secretion of such EVs [300,301]. Over-activation of ARF6 has been shown to increase release of PM-derived large EVs [302]. Modulation of cytoskeletal remodelers was reported to affect PM-derived EV release [303–305], and depletion of DIAPH3 increased large oncosome release [306]. In prokaryotes, recent insights into the mechanisms of outer membrane vesicle formation [307] could suggest other molecules to test in eukar- yotic cells.
These cell treatment approaches have great potential and deserve more development; however, it is impor- tant to recognize several caveats.
(1) Small EV–containing fractions potentially con- tain EVs originating from late endosomes (“exo- somes”) and others originating from the cell surface (plasma membrane), with both classes sharing common molecular players, including the ESCRT components TSG101, VPS4, and/or Alix [308–310]. Therefore consequences of decreasing or increasing global secretion of het- erogeneous populations of small EVs should not be interpreted in terms of functional effects of exosomes, but rather of small EVs in general.
(2) Tools described until now to block or enhance exosome secretion have not been well evaluated for their possible effect on secretion of other EVs. For instance, ionophores, such as ionomy- cin, are also potent inducers of large EV and microparticle secretion [207,311]. Conversely, in one study, inhibition of neutral sphingomyeli- nases was shown to enhance secretion of larger plasma membrane-derived EVs while decreasing that of small EVs [312]. Another example is monensin, often used to stimulate EV secretion, being an inhibitor of apoptotic body formation [167]. Therefore, it is likely that putative exo- some modulators will have different conse- quences in different cells and under different conditions, and it is important to carefully
JOURNAL OF EXTRACELLULAR VESICLES 25

26 C. THÉRY AND K. W. WITWER
quantify the toxicity of each treatment in each experimental system, to exclude artefactual effects on EV recovery due to increased cell death.
(3) Some EV release modulators affect other major intracellular pathways that might indirectly affect EV secretion and modify cell functions in general (like general intracellular trafficking, secretory, or autophagy pathways). Consequently, not only EV amount, but also EV composition may be changed, together with changes in protein expression and physiol- ogy of the secreting cells. As an example, Rab27a inhibition also decreased secretion of some non- EV-bound soluble factors [313,314]. Another caveat to consider is that disrupting the secre- tion of one EV type may disrupt the production of other EV types, such that the functional EV type may be masked by the over-production of an antagonistic one, leading to an erroneous conclusion that the disrupted EV type is the functional EV. Therefore, demonstrating that only late endosome-derived exosomes bear an analyzed function remains challenging. Some previous studies managed to rescue an observed effect by re-introducing purified exosomes (or rather small EV pellets) into the functional in vitro or in vivo assays [292,313,314]. This approach is indeed recommended, with careful interpretation taking into account the degree of rescue and the required amount of EVs.
Until we achieve unambiguous identification of speci- fic, unique biogenesis machineries affecting only a given subtype of EVs, we are left with trying to isolate EV subtypes after they have left the cell. For example, if multi-tetraspanin-bearing EVs are true exosomes in a particular cell system, an EV preparation could be depleted of such EVs and the activity quantified in comparison with that of an irrelevant IgG- or mock- depleted population.
How to attribute particular effects mediated by EVs to specific EV components
Many publications include knock-out or knock-down of a certain bioactive protein or RNA in the EV donor cell, after which the effects of the modified EV on target cells are compared with the effects of non- modified EVs. If the native effect of EVs is lost, the authors conclude that EV activity was due to the spe- cifically targeted protein or RNA. However, such a conclusion may or may not be valid in the absence
of an extensive characterization of EVs released by the cells depleted for the targeted molecule. Indeed, dele- tion of the protein/RNA of interest may also lead to major alterations of the secreting cell, resulting in addi- tional changes to the quantity or molecular contents of EVs, which could also explain the changes in EV- induced effects on target cells. While a complete omics analyses of the modified EV population may be beyond the scope of many studies, there should be an awareness that other EV components may have chan- ged as well. At a minimum, a small-scale analysis of EV number or common EV-associated proteins in the modified and WT conditions must be performed. Finally, Direct EVs engineering (e.g. to deplete the particular putative active molecule) may overcome the issue of alterations in the secreting cells. However, possible loss/alteration of EV cargo due to EV manip- ulation may also occur.
Consider whether an EV-dependent function is specific to a given EV source
Finally, in all cases, one must be careful in claiming a specific function of EVs from a particular source: it is one thing to claim that the EV fraction from Cell X is potent (versus other fractions), another to claim that Cell X EVs are potent versus those from other cells. For example, do my mesenchymal stromal cell (MSC) EVs do something special, or do milk EVs, urine EVs, cancer cell EVs do the same? Of course, it will not be possible to compare EVs from all different sources, thus the final message must reflect this uncertainty.
Consensus: This functional section had broad support (> 94% average for each subsection) from MISEV2018 survey respondents and has been revised only mini- mally. The components of Table 4, on EV-associated and -excluded activities, received an average 96% endorsement. One of the least supported components of the functional section (90% agreement) was the ori- ginal normalization recommendation (which are found in the function section but can be applied to character- ization and other sections as well). This part was sub- stantially revised based on the submitted comments.
General considerations
Reporting
ISEV endorses the EV-TRACK knowledgebase as a facilitating and updatable tool for comprehensive reporting of EV experimental studies [161]. EV- TRACK invites submission of details on EV isolation and characterization via a multi-step online template, then associates each with a particular study or publica- tion and also returns an “EV-METRIC” as an aggregate

measure of the level of detail provided. ISEV strongly encourages all authors to submit their experimental protocols on EV isolation and characterization to the EV-TRACK website (evtrack.org), and to consider applying additional steps if they or reviewers/editors feel that the calculated metric is low. The important consideration is not obtaining a particular metric, which after all may vary widely between basic and clinical studies; instead, the level of detail required for approved entries in EV-TRACK ensures that the trans- parency and reproducibility of procedures can be assessed. Furthermore, the knowledgebase can be revised and expanded as technologies and techniques develop, with input and assistance from the community.1 Authors are also urged to submit EV profiling data to public databases such as those curated and maintained by the European Bioinformatics Institute, the US National Center for Biotechnology Information, and the Japanese Center for Information Biology. In addition, but not as a substitute, data may be submitted to field-specific databases such as EVpedia [315,316], Vesiclepedia [317] (formerly ExoCarta [318]), and the exRNA Atlas [319].2
Notes

  1. EV-TRACK submission and EV-METRIC calculation may assist with but do not replace appropriate peer review. Interestingly, respondents to the MISEV2018 Survey were split between advocating mandatory EV- TRACK submission and reporting and recognizing EV- TRACK as a valuable but optional tool. As a result, MISEV2018 strongly encourages but cannot mandate EV-TRACK submission, which most seem to acknowl- edge as highly valuable.
  2. The utility of field-specific databases was questioned by some respondents, who felt that data from studies ana- lysing EVs obtained through low-specificity methods renders these databases difficult to interpret in terms of specific association of a given molecule/sequence to EVs or an EV subtype. Submission to field-specific databases is thus important to update these resources, which are of value to identify different studies finding the same mole- cules, but not to demonstrate the exosomal nor EV nature of the analysed entities. It also does not substitute for deposition with publicly maintained repositories.
    Exceptions to compliance with MISEV guidelines
    Some situations may arise in which strict adherence to the MISEV guidelines is difficult. Not all biofluids, for example lacrimal fluid, are available in sufficient volume to separate EVs and perform multiple tests with each sample; also, only limited numbers of EVs may be harvested from small numbers of patient-
    derived cells, small organoids, and more. In such cases, multiple samples might be pooled to establish the reliability of the separation method(s) and charac- terize EVs before further characterization or functional studies are performed with individuals samples. If even this solution is impractical, authors should indicate the limit of detection of each applied EV characterization technology and demonstrate that the available material falls below this limit. However, applying this “escape clause” means that EVs cannot be rigorously demon- strated, requiring that authors mention (and reviewers insist on) the caveats of alternative interpretations, i.e. that EVs may contribute, but not necessarily exclu- sively, to an observed phenomenon or molecular signature.
    Consensus: Section 6a (reporting requirements) was supported by 89% of MISEV2018 Survey respondents. The comments were split between those who wanted more reliance on EV-TRACK and those who found EV- TRACK submissions to be time-consuming or otherwise cumbersome. 99% of respondents agreed with mention- ing exceptions to compliance in 6b.
    Conclusions
    Major points of MISEV2018 (see MISEV2018 quick- reference checklist, p42):
    (1) “Extracellular vesicle” is the preferred generic term for the subject of our investigations, and subtypes should be defined by physical and bio- chemical characteristics and/or conditions/ sources. When other terms are used, careful definition is required.
    (2) A growing number of separation techniques and combinations thereof are available, variously balancing recovery and specificity. In all cases, the EV separation and/or isolation procedure must be reported in detail, to allow a reliable replication.
    (3) As EV characterization evolves, protein and lipid markers continue to be highly useful to demon- strate presence of the generic structure of EVs. Markers chosen for characterization of EVs may differ based on cells of origin (including mamma- lian vs non-mammalian vs non-eukaryotic cells), and whether claims are generic to all kinds of EVs, or instead specific to subtypes of EVs (4-b p16, Table 2, Table 3). Further, newly identified EV- associated components must be validated by 1) showing their specific association with EVs or sub- types and 2) if functionally relevant: topology in/ outside EVs of the functional molecule (4-d p.21).
    JOURNAL OF EXTRACELLULAR VESICLES 27

28
C. THÉRY AND K. W. WITWER
(4)
Function. Demonstration that a function is asso- ciated specifically with EVs released from the cell (Table 4) requires demonstration that the function occurs without cell-cell contact (5-b p22) and is not present (or is present to a substantially reduced extent) in the soluble, non-EV-associated secreted factors (5-c, d p23-24). Demonstration that a function is specific to exosomes (EVs of endosomal origin), as compared with other types of small EVs, is not recommended as a major point of any EV study due to the issues elaborated in section 5-e p25. However, if authors wish to make this point, the currently proposed exosome- specific tools cannot be taken for granted, and several controls should be performed to evaluate their action on other EVs, on secretion of non-EV products, and on the general physiology of secret- ing cells. Additional techniques for separation of these vesicles and/or genetic tools specifically affecting their secretion, may become available in the future (5-e p25).
The EV-TRACK knowledgebase is endorsed by ISEV to showcase and enhance rigor and repro- ducibility in EV studies, consistent with the MISEV guidelines.
Finally, there are exceptions to every rule. MISEV2018 is meant to guide and improve the field, not stifle it. If MISEV recommendations and requirements cannot be met, authors will then need to explain their unique situation and describe their attempts to meet the guidelines and the reason for failure. These guidelines will also continue to evolve.
trademarks were avoided in the main text to reduce the likelihood of preferential treatment to specific services or products of for-profit entities.
Funding
Funding is listed only for the corresponding authors. CT’s laboratory is funded by INSERM, Institut Curie, Ministry of Education, and grants from INCa (INCA-11548), French National Research Agency (ANR-10-IDEX-0001-02 PSL* and ANR-11-LABX-0043), SIDACTION (17-1-AAE-1138), Fondation ARC (PGA1 RF20180206962, PJA 20171206453). KWW and CT receive support from NIDA (DA040385). KWW is also supported in part by NIA AG057430, NIDA DA047807 and NIMH MH118164.
Authors’ contributions
CT and KWW led the manuscript drafting team, prepared the online surveys, analyzed survey results and other input, communicated with authors and prospective authors, and prepared all revisions. All other authors contributed as spe- cified in the text and in footnotes to the author affiliations. All authors reviewed and approved the final draft of the manuscript.
ORCID
(5)
(6)
http://orcid.org/0000-0001-8294-6884 http://orcid.org/0000-0003-1664-4233
References, especially those provided to illustrate methods and approaches, are representative only, and are not meant to be a comprehensive review of the literature. Most references were derived from suggestions provided in the MISEV2018 Survey results. Each reference was checked by multiple authors. Citation implies deemed relevance of scientific content and not an endorsement by the authors or ISEV of any particular journal or editorial practice.
[1] Lotvall J, Hill AF, Hochberg F, et al. Minimal experi- mental requirements for definition of extracellular vesicles and their functions: a position statement from the international society for extracellular vesicles. J Extracell Vesicles. 2014;3:26913. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25536934
[2] Witwer KW, Soekmadji C, Hill AF, et al. Updating the MISEV minimal requirements for extracellular vesicle studies: building bridges to reproducibility. J Extracell Vesicles. 2017;6(1):1396823. Available from: https:// www.tandfonline.com/doi/full/10.1080/20013078.2017. 1396823
[3] Stein JM, Luzio JP Ectocytosis caused by sublytic auto- logous complement attack on human neutrophils. The sorting of endogenous plasma-membrane proteins and lipids into shed vesicles. Biochem J. 1991;274
Consensus: 89% of ISEV2018 Survey respondents had no major changes; 29 comments were taken into account in revisions of this section.
Acknowledgments
The authors are grateful to all respondents to the various surveys that supported the MISEV2018 effort, including those who only partially completed the surveys or completed the surveys but declined authorship. They also thank all community members who offered feedback.
Disclosure statement
No potential conflict of interest was reported by the authors. In addition, they declare that they took responsibility for avoiding any introduction of possible conflicting material into the text. For example all names of companies and
Clotilde Théry Kenneth W Witwer
References

(Pt 2):381–386. Available from: http://www.ncbi.nlm.
nih.gov/pubmed/1848755
[4] Cocucci E, Meldolesi J Ectosomes and exosomes: shed- ding the confusion between extracellular vesicles. Trends Cell Biol. 2015;25(6):364–372. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25683921
[5] Gould SJ, Raposo G As we wait: coping with an imperfect nomenclature for extracellular vesicles. J Extracell Vesicles. 2013;2. Available from: http:// www.ncbi.nlm.nih.gov/pubmed/24009890
[6] Gardiner C, Di Vizio D, Sahoo S, et al. Techniques used for the isolation and characterization of extracel- lular vesicles: results of a worldwide survey. J Extracell Vesicles. 2016;5:32945. Available from: http://www. ncbi.nlm.nih.gov/pubmed/27802845
[7] Rojas A The imperative authentication of cell lines. Antimicrob Agents Chemother. 2017;61(11):e01823– 17. Available from: http://aac.asm.org/lookup/doi/10. 1128/AAC.01823-17
[8] Reid Y, Storts D, Riss T, et al. Authentication of human cell lines by STR DNA profiling analysis [Internet]. Assay Guidance Manual. 2004. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23805434
[9] Chen TS, Arslan F, Yin Y, et al. Enabling a robust scalable manufacturing process for therapeutic exo- somes through oncogenic immortalization of human ESC-derived MSCs. J Transl Med. 2011;9(1):47. Available from: http://translational-medicine.biomed central.com/articles/10.1186/1479-5876-9-47
[10] Lima LG, Chammas R, Monteiro RQ, et al. Tumor- derived microvesicles modulate the establishment of metastatic melanoma in a phosphatidylserine-dependent manner. Cancer Lett. 2009;283(2):168–175. Available from: http://linkinghub.elsevier.com/retrieve/pii/ S0304383509002420
[11] Frey B, Gaipl US The immune functions of phosphati- dylserine in membranes of dying cells and microvesicles. Semin Immunopathol. 2011;33(5):497–516. Available from: http://link.springer.com/10.1007/s00281-010- 0228-6
[12] Roseblade A, Luk F, Ung A, et al. Targeting micro- particle biogenesis: a novel approach to the circumven- tion of cancer multidrug resistance. Curr Cancer Drug Targets. 2015;15(3):205–214. Available from: http:// www.ncbi.nlm.nih.gov/pubmed/25714701
[13] Takasugi M Emerging roles of extracellular vesicles in cellular senescence and aging. Aging Cell. 2018;17(2): e12734.
[14] Patel DB, Gray KM, Santharam Y, et al. Impact of cell culture parameters on production and vascularization bioactivity of mesenchymal stem cell-derived extracel- lular vesicles. Bioeng Transl Med. 2017;2(2):170–179.
[15] Dang VD, Jella KK, Ragheb RRT, et al. Lipidomic and proteomic analysis of exosomes from mouse cortical collecting duct cells. FASEB J. 2017;31(12):5399–5408. Available from: http://www.fasebj.org/doi/10.1096/fj. 201700417R
[16] Klingeborn M, Dismuke WM, Skiba NP, et al. Directional exosome proteomes reflect polarity-specific functions in retinal pigmented epithelium monolayers. Sci Rep. 2017;7(1):4901. Available from: http://www.nat ure.com/articles/s41598-017-05102-9
[17] Mittelbrunn M, Vicente-Manzanares M, Sánchez- Madrid F Organizing polarized delivery of exosomes at synapses. Traffic. 2015;16(4):327–337. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25614958
[18] van Niel G, Raposo G, Candalh C, et al. Intestinal epithelial cells secrete exosome-like vesicles. Gastroenterology. 2001;121(2):337–349. Available from: http://www.ncbi.nlm.nih.gov/pubmed/11487543
[19] Tauro BJ, Greening DW, Mathias RA, et al. Two dis- tinct populations of exosomes are released from LIM1863 colon carcinoma cell-derived organoids. Mol Cell Proteomics. 2013;12(3):587–598. Available from: http://www.mcponline.org/lookup/doi/10.1074/ mcp.M112.021303
[20] Yan IK, Shukla N, Borrelli DA, et al. Use of a hollow fiber bioreactor to collect extracellular vesicles from cells in culture. Methods Mol Biol. 2018;1740:35–41. Available from: http://link.springer.com/10.1007/978- 1-4939-7652-2_4
[21] Watson DC, Yung BC, Bergamaschi C, et al. Scalable, cGMP-compatible purification of extracellular vesicles carrying bioactive human heterodimeric IL-15/lactad- herin complexes. J Extracell Vesicles. 2018;7 (1):1442088. Available from: http://www.ncbi.nlm.nih. gov/pubmed/29535850
[22] Lowry MC, O’Driscoll L Can hi-jacking hypoxia inhi- bit extracellular vesicles in cancer? Drug Discov Today. 2018;23(6):1267–1273. Available from: https://linkin ghub.elsevier.com/retrieve/pii/S1359644617303252
[23] Mitchell MD, Peiris HN, Kobayashi M, et al. Placental exosomes in normal and complicated pregnancy. Am J Obstet Gynecol. 2015;213(4Suppl): S173–81. Available from: http://linkinghub.elsevier.com/ retrieve/pii/S0002937815007176
[24] de Jong OG, Verhaar MC, Chen Y, et al. Cellular stress conditions are reflected in the protein and RNA con- tent of endothelial cell-derived exosomes. J Extracell Vesicles. 2012;1(1):18396. Available from: https://www. tandfonline.com/doi/full/10.3402/jev.v1i0.18396
[25] Stratton D, Moore C, Antwi-Baffour S, et al. Microvesicles released constitutively from prostate cancer cells differ biochemically and functionally to stimulated microvesicles released through sublytic C5b-9. Biochem Biophys Res Commun. 2015;460 (3):589–595. Available from: http://linkinghub.else vier.com/retrieve/pii/S0006291X15005203
[26] Dozio V, Sanchez J-C Characterisation of extracellular vesicle-subsets derived from brain endothelial cells and analysis of their protein cargo modulation after TNF exposure. J Extracell Vesicles. 2017;6(1):1302705. Available from: https://www.tandfonline.com/doi/full/ 10.1080/20013078.2017.1302705
[27] Taylor J, Jaiswal R, Bebawy M Calcium-calpain depen- dent pathways regulate vesiculation in malignant breast cells. Curr Cancer Drug Targets. 2017;17 (5):486–494. Available from: http://www.eurekaselect. com/node/146745/article
[28] Mostefai HA, Agouni A, Carusio N, et al. Phosphatidylinositol 3-kinase and xanthine oxidase regulate nitric oxide and reactive oxygen species pro- ductions by apoptotic lymphocyte microparticles in endothelial cells. J Immunol. 2008;180(7):5028–5035.
JOURNAL OF EXTRACELLULAR VESICLES 29

30 C. THÉRY AND K. W. WITWER
Available from: http://www.ncbi.nlm.nih.gov/pubmed/
18354228
[29] Agouni A, Mostefai HA, Porro C, et al. Sonic hedge- hog carried by microparticles corrects endothelial injury through nitric oxide release. FASEB J. 2007;21 (11):2735–2741. Available from: http://www.fasebj.org/ doi/10.1096/fj.07-8079com
[30] Soekmadji C, Riches JD, Russell PJ, et al. Modulation of paracrine signaling by CD9 positive small extracel- lular vesicles mediates cellular growth of androgen deprived prostate cancer. Oncotarget. 2017;8 (32):52237–52255. Available from: http://www.oncotar get.com/fulltext/11111
[31] Saari H, Lázaro-Ibáñez E, Viitala T, et al. Microvesicle- and exosome-mediated drug delivery enhances the cytotoxicity of paclitaxel in autologous prostate cancer cells. J Control Release. 2015;220(PtB):727–737. Available from: http://linkinghub.elsevier.com/ retrieve/pii/S0168365915301322
[32] Lázaro-Ibáñez E, Neuvonen M, Takatalo M, et al. Metastatic state of parent cells influences the uptake and functionality of prostate cancer cell-derived extra- cellular vesicles. J Extracell Vesicles. 2017;6 (1):1354645. Available from: https://www.tandfonline. com/doi/full/10.1080/20013078.2017.1354645
[33] Chernov VM, Mouzykantov AA, Baranova NB, et al. Extracellular membrane vesicles secreted by myco- plasma acholeplasma laidlawii PG8 are enriched in virulence proteins. J Proteomics. 2014;110:117–128. Available from: http://linkinghub.elsevier.com/ retrieve/pii/S1874391914003819
[34] Corral-Vázquez C, Aguilar-quesada R, Catalina P, et al. Cell lines authentication and mycoplasma detec- tion as minimun quality control of cell lines in biobanking. Cell Tissue Bank. 2017;18(2):271–280. Available from: http://link.springer.com/10.1007/ s10561-017-9617-6
[35] Yang C, Chalasani G, Ng Y-H, et al. Exosomes released from mycoplasma infected tumor cells activate inhibi- tory B cells. PLoS One. 2012;7(4):e36138. Available from: http://dx.plos.org/10.1371/journal.pone.0036138
[36] Quah BJC, O’Neill HC Mycoplasma contaminants pre- sent in exosome preparations induce polyclonal B cell responses. J Leukoc Biol. 2007;82(5):1070–1082.
[37] Mathivanan S, Lim JW, Tauro BJ, et al. Proteomics analysis of A33 immunoaffinity-purified exosomes released from the human colon tumor cell line LIM1215 reveals a tissue-specific protein signature. Mol Cell Proteomics. 2010;9(2):197–208. Available from: http://www.ncbi.nlm.nih.gov/pubmed/19837982
[38] Burger D, Turner M, Xiao F, et al. High glucose increases the formation and pro-oxidative activity of endothelial microparticles. Diabetologia. 2017;60 (9):1791–1800. Available from: http://link.springer. com/10.1007/s00125-017-4331-2
[39] Thom SR, Bhopale VM, Yu K, et al. Neutrophil microparticle production and inflammasome activa- tion by hyperglycemia due to cytoskeletal instability. J Biol Chem. 2017;292(44):18312–18324. Available from: http://www.jbc.org/lookup/doi/10.1074/jbc. M117.802629
[40] Rice GE, Scholz-Romero K, Sweeney E, et al. The effect of glucose on the release and bioactivity of exosomes from first trimester trophoblast cells. J Clin Endocrinol Metab. 2015;100(10):E1280–8. Available from: https:// academic.oup.com/jcem/article-lookup/doi/10.1210/jc. 2015-2270
[41] Németh A, Orgovan N, Sódar BW, et al. Antibiotic- induced release of small extracellular vesicles (exo- somes) with surface-associated DNA. Sci Rep. 2017;7 (1):8202. Available from: http://www.nature.com/arti cles/s41598-017-08392-1
[42] Zhou X, Zhang W, Yao Q, et al. Exosome production and its regulation of EGFR during wound healing in renal tubular cells. Am J Physiol Renal Physiol. 2017;312(6):F963–70. Available from: http://www.phy siology.org/doi/10.1152/ajprenal.00078.2017
[43] Pachler K, Lener T, Streif D, et al. A good manufactur- ing practice-grade standard protocol for exclusively human mesenchymal stromal cell-derived extracellular vesicles. Cytotherapy. 2017;19(4):458–472. Available from: http://linkinghub.elsevier.com/retrieve/pii/ S1465324917300038
[44] Saury C, Lardenois A, Schleder C, et al. Human serum and platelet lysate are appropriate xeno-free alterna- tives for clinical-grade production of human MuStem cell batches. Stem Cell Res Ther. 2018;9(1):128. Available from: https://stemcellres.biomedcentral. com/articles/10.1186/s13287-018-0852-y
[45] Li J, Lee Y, Johansson HJ, et al. Serum-free culture alters the quantity and protein composition of neuroblastoma-derived extracellular vesicles. J Extracell Vesicles. 2015;4(1):26883. Available from: https://www. tandfonline.com/doi/full/10.3402/jev.v4.26883
[46] Beninson LA, Fleshner M Exosomes in fetal bovine serum dampen primary macrophage IL-1β response to lipopolysaccharide (LPS) challenge. Immunol Lett. 2015;163(2):187–192. Available from: http://www.ncbi. nlm.nih.gov/pubmed/25455591
[47] Eitan E, Zhang S, Witwer KW, et al. Extracellular vesicle-depleted fetal bovine and human sera have reduced capacity to support cell growth. J Extracell Vesicles. 2015;4:26373. Available from: http://www. ncbi.nlm.nih.gov/pubmed/25819213
[48] Théry C, Amigorena S, Raposo G, et al. Isolation and characterization of exosomes from cell culture super- natants and biological fluids. In: Current protocols in cell biology. Hoboken, NJ, USA: John Wiley & Sons, Inc.; 2006. p. Unit 3.22. Available from: http://www. ncbi.nlm.nih.gov/pubmed/18228490
[49] van Balkom BWM, de Jong OG, Smits M, et al. Endothelial cells require miR-214 to secrete exosomes that suppress senescence and induce angiogenesis in human and mouse endothelial cells. Blood. 2013;121 (19):3997–4006. Available from: http://www.ncbi.nlm. nih.gov/pubmed/23532734
[50] Kornilov R, Puhka M, Mannerström B, et al. Efficient ultrafiltration-based protocol to deplete extracellular vesicles from fetal bovine serum. J Extracell Vesicles. 2018;7(1):1422674. Available from: https://www.tandfonline.com/doi/full/10. 1080/20013078.2017.1422674

[51] Wei Z, Batagov AO, Carter DRF, et al. Fetal bovine serum RNA interferes with the cell culture derived extracellular RNA. Sci Rep. 2016;6:31175. Available from: http://www.ncbi.nlm.nih.gov/pubmed/ 27503761
[52] Shelke GV, Lässer C, Gho YS, et al. Importance of exosome depletion protocols to eliminate functional and RNA-containing extracellular vesicles from fetal bovine serum. J Extracell Vesicles. 2014;3:24783. Available from: http://www.ncbi.nlm.nih.gov/ pubmed/25317276
[53] Tosar JP, Cayota A, Eitan E, et al. Ribonucleic arte- facts: are some extracellular RNA discoveries driven by cell culture medium components? J Extracell Vesicles. 2017;6(1):1272832. Available from: http://www.ncbi. nlm.nih.gov/pubmed/28326168
[54] Kaur S, Singh SP, Elkahloun AG, et al. CD47- dependent immunomodulatory and angiogenic activ- ities of extracellular vesicles produced by T cells. Matrix Biol. 2014;37:49–59. Available from: http://lin kinghub.elsevier.com/retrieve/pii/S0945053X14000924
[55] Witwer KW, Buzas EI, Bemis LT, et al. Standardization of sample collection, isolation and analysis methods in extracellular vesicle research: an ISEV position paper. J Extracell Vesicles. 2013;2:20360.
[56] Mateescu B, Kowal EJK, van Balkom BWM, Bartel S, Bhattacharyya SN, Buzàs EI, et al. Obstacles and opportunities in the functional analysis of extracellular vesicle RNA- An ISEV Position Paper. J Extracell Vesicles. 2017;6:1286095.
[57] Bæk R, Søndergaard EKL, Varming K, et al. The impact of various preanalytical treatments on the phe- notype of small extracellular vesicles in blood analyzed by protein microarray. J Immunol Meth. 2016;438:11–20. Available from: http://linkinghub.else vier.com/retrieve/pii/S0022175916301624
[58] Barteneva NS, Fasler-Kan E, Bernimoulin M, et al. Circulating microparticles: square the circle. BMC Cell Biol. 2013;14(1):23. Available from: http://bmccell biol.biomedcentral.com/articles/10.1186/1471-2121- 14-23
[59] Mullier F, Bailly N, Chatelain C, et al. Pre-analytical issues in the measurement of circulating microparti- cles: current recommendations and pending questions. J Thromb Haemost. 2013. Available from: http://www. ncbi.nlm.nih.gov/pubmed/23410207
[60] Lacroix R, Judicone C, Poncelet P, et al. Impact of pre-analytical parameters on the measurement of cir- culating microparticles: towards standardization of protocol. J Thromb Haemost. 2012;10(3):437–446. Available from: http://www.ncbi.nlm.nih.gov/ pubmed/22212198
[61] Coumans FAW, Brisson AR, Buzas EI, et al. Methodological guidelines to study extracellular vesicles. Circ Res. 2017;120(10):1632–1648. Available from: http://circres.ahajournals.org/lookup/doi/10. 1161/CIRCRESAHA.117.309417
[62] Yuana Y, Bertina RM, Osanto S Pre-analytical and analytical issues in the analysis of blood microparticles. Thromb Haemost. 2011;105 (3):396–408. Available from: http://www.ncbi.nlm.nih. gov/pubmed/21174005
[63] Yuana Y, Böing AN, Grootemaat AE, et al. Handling and storage of human body fluids for analysis of extra- cellular vesicles. J Extracell Vesicles. 2015;4:29260.
[64] Robbins PD Extracellular vesicles and aging. Stem Cell Investig. 2017;4(12):98. Available from: http://sci. amegroups.com/article/view/17758/18069
[65] Danielson KM, Estanislau J, Tigges J, et al. Diurnal variations of circulating extracellular vesicles measured by nano flow cytometry. PLoS One. 2016;11(1): e0144678. Available from: http://www.ncbi.nlm.nih. gov/pubmed/26745887
[66] Fendl B, Weiss R, Fischer MB, et al. Characterization of extracellular vesicles in whole blood: influence of pre-analytical parameters and visualization of vesicle-cell interactions using imaging flow cytometry. Biochem Biophys Res Commun. 2016;478(1):168–173. Available from: http://linkinghub.elsevier.com/ retrieve/pii/S0006291X16311950
[67] Wisgrill L, Lamm C, Hartmann J, et al. Peripheral blood microvesicles secretion is influenced by storage time, temperature, and anticoagulants. Cytometry A. 2016;89(7):663–672.
[68] György B, Pálóczi K, Kovács A, et al. Improved circu- lating microparticle analysis in acid-citrate dextrose (ACD) anticoagulant tube. Thromb Res. 2014;133 (2):285–292. Available from: http://linkinghub.else vier.com/retrieve/pii/S004938481300546X
[69] Mitchell AJ, Gray WD, Hayek SS, et al. Platelets con- found the measurement of extracellular miRNA in archived plasma. Sci Rep. 2016;6(1):32651. Available from: http://www.ncbi.nlm.nih.gov/pubmed/27623086
[70] Cheng HH, Yi HS, Kim Y, et al. Plasma processing conditions substantially influence circulating microRNA biomarker levels. PLoS One. 2013;8(6): e64795. Available from: http://www.ncbi.nlm.nih.gov/ pubmed/23762257
[71] Muller L, Hong C-S, Stolz DB, et al. Isolation of biologically-active exosomes from human plasma. J Immunol Meth. 2014;411:55–65.
[72] Ayers L, Kohler M, Harrison P, et al. Measurement of circulating cell-derived microparticles by flow cytome- try: sources of variability within the assay. Thromb Res. 2011;127(4):370–377. Available from: http:// www.ncbi.nlm.nih.gov/pubmed/21257195
[73] Heijnen HF, Schiel AE, Fijnheer R, et al. Activated platelets release two types of membrane vesicles: microvesicles by surface shedding and exosomes derived from exocytosis of multivesicular bodies and alpha-granules. Blood. 1999;94(11):3791–3799.
[74] Mincheva-Nilsson L, Baranov V, Nagaeva O, et al. Isolation and characterization of exosomes from cul- tures of tissue explants and cell lines. Curr Protoc Immunol. 2016;115:14.42.1–14.42.21.
[75] Lunavat TR, Cheng L, Einarsdottir BO, et al. BRAFV600 inhibition alters the microRNA cargo in the vesicular secretome of malignant melanoma cells. Proc Natl Acad Sci U S A. 2017;114(29):E5930–9. Available from: http://www.pnas.org/lookup/doi/10. 1073/pnas.1705206114
[76] Gupta AK, Rusterholz C, Huppertz B, et al. A comparative study of the effect of three different syncytiotrophoblast micro-particles preparations on
JOURNAL OF EXTRACELLULAR VESICLES 31

32 C. THÉRY AND K. W. WITWER
endothelial cells. Placenta. 2005;26(1):59–66. Available from: http://linkinghub.elsevier.com/retrieve/pii/ S0143400404001080
[77] Holder BS, Tower CL, Forbes K, et al. Immune cell activation by trophoblast-derived microvesicles is mediated by syncytin 1. Immunology. 2012;136 (2):184–191.
[78] Perez-Gonzalez R, Gauthier SA, Kumar A, et al. The exosome secretory pathway transports amyloid precur- sor protein carboxyl-terminal fragments from the cell into the brain extracellular space. J Biol Chem. 2012;287(51):43108–43115. Available from: http:// www.jbc.org/lookup/doi/10.1074/jbc.M112.404467
[79] Vella LJ, Scicluna BJ, Cheng L, et al. A rigorous method to enrich for exosomes from brain tissue. J Extracell Vesicles. 2017;6(1):1348885. Available from: http://www.ncbi.nlm.nih.gov/pubmed/28804598
[80] Deng ZB, Poliakov A, Hardy RW, et al. Adipose tissue exosome-like vesicles mediate activation of macrophage-induced insulin resistance. Diabetes. 2009;58(11):2498–2505. Available from: http://www. ncbi.nlm.nih.gov/pubmed/19675137
[81] Wang GJ, Liu Y, Qin A, et al. Thymus exosomes-like particles induce regulatory T cells. J Immunol. 2008;181(8):5242–5248. Available from: http://www. ncbi.nlm.nih.gov/pubmed/18832678
[82] Kranendonk MEG, Visseren FLJ, van Balkom BWM, et al. Human adipocyte extracellular vesicles in reci- procal signaling between adipocytes and macrophages. Obesity (Silver Spring). 2014;22(5):1296–1308.
[83] Loyer X, Zlatanova I, Devue C, et al. Intra-cardiac release of extracellular vesicles shapes inflammation following myocardial infarction. Circ Res. 2018;123 (1):100–106. Available from: http://circres.ahajour nals.org/lookup/doi/10.1161/CIRCRESAHA.117. 311326
[84] Leroyer AS, Ebrahimian TG, Cochain C, et al. Microparticles from ischemic muscle promotes post- natal vasculogenesis. Circulation. 2009;119 (21):2808–2817. Available from: http://circ.ahajour nals.org/cgi/doi/10.1161/CIRCULATIONAHA.108. 816710
[85] Michaelis ML, Jiang L, Michaelis EK Isolation of synaptosomes, synaptic plasma membranes, and synaptic junctional complexes. In: Methods in mole- cular biology. Clifton, NJ. 2017. p. 107–119. Available from: http://www.ncbi.nlm.nih.gov/pubmed/27943187
[86] Zhou H, Yuen PS, Pisitkun T, et al. Collection, storage, preservation, and normalization of human urinary exosomes for biomarker discovery. Kidney Int. 2006;69(8):1471–1476. Available from: http://www. ncbi.nlm.nih.gov/pubmed/16501490
[87] Vila-Liante V, Sánchez-López V, Martínez-Sales V, et al. Impact of sample processing on the measurement of circulating microparticles: storage and centrifuga- tion parameters. Clin Chem Lab Med. 2016;54 (11):1759–1767. Available from: https://www.degruy ter.com/view/j/cclm.2016.54.issue-11/cclm-2016-0036/ cclm-2016-0036.xml
[88] Kriebardis AG, Antonelou MH, Georgatzakou HT, et al. Microparticles variability in fresh frozen plasma: preparation protocol and storage time effects. Blood
Transfus. 2016;14(2):228–237. Available from: http://
www.ncbi.nlm.nih.gov/pubmed/27136430
[89] Lőrincz ÁM, Timár CI, Marosvári KA, et al. Effect of storage on physical and functional properties of extra- cellular vesicles derived from neutrophilic granulocytes. J Extracell Vesicles. 2014;3(1):25465. Available from: https://www.tandfonline.com/doi/full/ 10.3402/jev.v3.25465
[90] Bosch S, de Beaurepaire L, Allard M, et al. Trehalose prevents aggregation of exosomes and cryodamage. Sci Rep. 2016;6(1):36162. Available from: http://www.nat ure.com/articles/srep36162
[91] Maroto R, Zhao Y, Jamaluddin M, et al. Effects of storage temperature on airway exosome integrity for diagnostic and functional analyses. J Extracell Vesicles. 2017;6(1):1359478. Available from: https://www.tand fonline.com/doi/full/10.1080/20013078.2017.1359478
[92] Jin Y, Chen K, Wang Z, et al. DNA in serum extra- cellular vesicles is stable under different storage conditions. BMC Cancer. 2016;16(1):753. Available from: http://www.ncbi.nlm.nih.gov/pubmed/ 27662833
[93] Jeyaram A, Jay SM. Preservation and storage stability of extracellular vesicles for therapeutic applications. Aaps J. 2017;20(1):1. Available from: http://link. springer.com/10.1208/s12248-017-0160-y
[94] Trummer A, De Rop C, Tiede A, et al. Recovery and composition of microparticles after snap-freezing depends on thawing temperature. Blood Coagul Fibrinolysis. 2009;20(1):52–56. Available from: https://insights.ovid. com/crossref?an=00001721-200901000-00010
[95] Lener T, Gimona M, Aigner L, et al. Applying extra- cellular vesicles based therapeutics in clinical trials – an ISEV position paper. J Extracell Vesicles. 2015;4:30087. Available from: http://www.pubmedcentral.nih.gov/ articlerender.fcgi?artid=4698466&tool=pmcentrez&ren dertype=abstract
[96] Reiner AT, Witwer KW, Van Balkom BWM, et al. Concise review: developing best-practice models for the therapeutic use of extracellular vesicles. Stem Cells Transl Med. 2017;6(8).
[97] Clayton A, Buschmann D, Byrd JB, et al. Summary of the ISEV workshop on extracellular vesicles as disease biomarkers, held in Birmingham, UK, during December 2017. J Extracell Vesicles. 2018;7 (1):1473707. Available from: https://www.tandfonline. com/doi/full/10.1080/20013078.2018.1473707
[98] Montis C, Zendrini A, Valle F, et al. Size distribution of extracellular vesicles by optical correlation techniques. Colloids Surf B Biointerfaces. 2017;158:331–338. Available from: http://linkinghub. elsevier.com/retrieve/pii/S092777651730406X
[99] Morales-Kastresana A, Telford B, Musich TA, et al. Labeling extracellular vesicles for nanoscale flow cytometry. Sci Rep. 2017;7(1):1878. Available from: http://www.nature.com/articles/s41598-017-01731-2
[100] Corso G, Mäger I, Lee Y, et al. Reproducible and scalable purification of extracellular vesicles using combined bind-elute and size exclusion chromatography. Sci Rep. 2017;7(1):11561. Available from: http://www.nature.com/articles/ s41598-017-10646-x

[101] Welton JL, Webber JP, Botos L-A, et al. Ready-made chromatography columns for extracellular vesicle iso- lation from plasma. J Extracell Vesicles. 2015;4:27269. Available from: http://www.tandfonline.com/doi/full/ 10.3402/jev.v4.27269
[102] Vergauwen G, Dhondt B, Van Deun J, et al. Confounding factors of ultrafiltration and protein ana- lysis in extracellular vesicle research. Sci Rep. 2017;7 (1):2704. Available from: http://www.nature.com/arti cles/s41598-017-02599-y
[103] Lobb RJ, Becker M, Wen SW, et al. Optimized exo- some isolation protocol for cell culture supernatant and human plasma. J Extracell Vesicles. 2015;4:27031. Available from: https://www.tandfonline.com/doi/full/ 10.3402/jev.v4.27031
[104] Tan CY, Lai RC, Wong W, et al. Mesenchymal stem cell-derived exosomes promote hepatic regeneration in drug-induced liver injury models. Stem Cell Res Ther. 2014;5(3):76. Available from: http://stemcellres.com/ content/5/3/76
[105] Jong AY, Wu C-H, Li J, et al. Large-scale isolation and cytotoxicity of extracellular vesicles derived from activated human natural killer cells. J Extracell Vesicles. 2017;6(1):1294368. Available from: https://www.tandfonline.com/doi/full/10.1080/ 20013078.2017.1294368
[106] Heinemann ML, Ilmer M, Silva LP, et al. Benchtop isolation and characterization of functional exosomes by sequential filtration. J Chromatogr A. 2014;1371:125–135. Available from: http://linkinghub. elsevier.com/retrieve/pii/S0021967314015908
[107] Heinemann ML, Vykoukal J Sequential filtration: A gentle method for the isolation of functional extra- cellular vesicles. In: Methods in molecular biology. Clifton, NJ. 2017. p. 33–41. Available from: http:// www.ncbi.nlm.nih.gov/pubmed/28828646
[108] Wei Z, Batagov AO, Schinelli S, et al. Coding and noncoding landscape of extracellular RNA released by human glioma stem cells. Nat Commun. 2017;8 (1):1145. Available from: http://www.nature.com/arti cles/s41467-017-01196-x
[109] Lamparski HG, Metha-Damani A, Yao JY, et al. Production and characterization of clinical grade exo- somes derived from dendritic cells. J Immunol Meth. 2002;270(2):211–226. Available from: http://www.ncbi. nlm.nih.gov/pubmed/12379326
[110] Escudier B, Dorval T, Chaput N, et al. Vaccination of metastatic melanoma patients with autologous dendri- tic cell (DC) derived-exosomes: results of thefirst phase I clinical trial. J Transl Med. 2005;3(1):10. Available from: http://www.ncbi.nlm.nih.gov/pubmed/15740633
[111] Roda B, Zattoni A, Reschiglian P, et al. Field-flow fractionation in bioanalysis: A review of recent trends. Anal Chim Acta. 2009;635(2):132–143. Available from: http://linkinghub.elsevier.com/ retrieve/pii/S0003267009000865
[112] Zhang H, Freitas D, Kim HS, et al. Identification of distinct nanoparticles and subsets of extracellular vesi- cles by asymmetric flow field-flow fractionation. Nat Cell Biol. 2018;20(3):332–343. Available from: http:// www.ncbi.nlm.nih.gov/pubmed/29459780
[113]
[114]
[115]
[116]
[117]
[118]
[119]
[120]
[121]
[122]
[123]
[124]
Yang JS, Lee JC, Byeon SK, et al. Size dependent lipidomic analysis of urinary exosomes from patients with prostate cancer by flow field-flow fractionation and nanoflow liquid chromatography-tandem mass spectrometry. Anal Chem. 2017;89(4):2488–2496. Available from: http://pubs.acs.org/doi/10.1021/acs.ana lchem.6b04634
Agarwal K, Saji M, Lazaroff SM, et al. Analysis of exosome release as a cellular response to MAPK path- way inhibition. Langmuir. 2015;31(19):5440–5448. Available from: http://www.ncbi.nlm.nih.gov/ pubmed/25915504
Liu C, Guo J, Tian F, et al. Field-free isolation of exosomes from extracellular vesicles by microfluidic viscoelastic flows. ACS Nano. 2017;11(7):6968–6976. Available from: http://pubs.acs.org/doi/10.1021/acs nano.7b02277
Ibsen SD, Wright J, Lewis JM, et al. Rapid isolation and detection of exosomes and associated biomarkers from plasma. ACS Nano. 2017;11(7):6641–6651. Available from: http://pubs.acs.org/doi/10.1021/acs nano.7b00549
Lewis JM, Vyas AD, Qiu Y, et al. Integrated analysis of exosomal protein biomarkers on alternating current electrokinetic chips enables rapid detection of pancrea- tic cancer in patient blood. ACS Nano. 2018;12 (4):3311–3320. Available from: http://pubs.acs.org/ doi/10.1021/acsnano.7b08199
Lee K, Shao H, Weissleder R, et al. Acoustic purifica- tion of extracellular microvesicles. ACS Nano. 2015;9 (3):2321–2327. Available from: http://pubs.acs.org/doi/ 10.1021/nn506538f
Satzer P, Wellhoefer M, Jungbauer A. Continuous separation of protein loaded nanoparticles by simu- lated moving bed chromatography. J Chromatogr A. 2014;1349:44–49. Available from: http://linkinghub. elsevier.com/retrieve/pii/S0021967314006979
Mol EA, Goumans M-J, Doevendans PA, et al. Higher functionality of extracellular vesicles isolated using size-exclusion chromatography compared to ultracentrifugation. Nanomedicine. 2017;13 (6):2061–2065. Available from: http://linkinghub.else vier.com/retrieve/pii/S1549963417300540
de Menezes-Neto A, Sáez MJF, Lozano-Ramos I, et al. Size-exclusion chromatography as a stand-alone metho- dology identifies novel markers in mass spectrometry analyses of plasma-derived vesicles from healthy individuals. J Extracell Vesicles. 2015;4:27378. Available from: http://www.ncbi.nlm.nih.gov/pubmed/26154623 Kosanović M, Milutinović B, Goč S, et al. Ion- exchange chromatography purification of extracellular vesicles. Biotechniques. 2017;63(2):65–71. Available from: https://www.future-science.com/doi/10.2144/ 000114575
Heath N, Grant L, De Oliveira TM, et al. Rapid isolation and enrichment of extracellular vesicle preparations using anion exchange chromatography. Sci Rep. 2018;8 (1):5730. Available from: http://www.ncbi.nlm.nih.gov/ pubmed/29636530
Kim D, Nishida H, An SY, et al. Chromatographically isolated CD63 + CD81 + extracellular vesicles from
JOURNAL OF EXTRACELLULAR VESICLES 33

34
C. THÉRY AND K. W. WITWER
[125]
[126]
[127]
[128]
[129]
[130]
[131]
[132]
[133]
[134]
[135]
[136]
mesenchymal stromal cells rescue cognitive impair- ments after TBI. Proc Natl Acad Sci. 2016;113 (1):170–175. Available from: http://www.ncbi.nlm.nih. gov/pubmed/26699510
Merchant ML, Powell DW, Wilkey DW, et al. Microfiltration isolation of human urinary exosomes for characterization by MS. PROTEOMICS – Clin Appl. 2010;4(1):84–96. Available from: http://www. ncbi.nlm.nih.gov/pubmed/21137018
Higginbotham JN, Zhang Q, Jeppesen DK, et al. Identification and characterization of EGF receptor in individual exosomes by fluorescence-activated vesicle sorting. J Extracell Vesicles. 2016;5:29254. Available from: http://www.ncbi.nlm.nih.gov/pubmed/27345057 Groot Kormelink T, Arkesteijn GJA, Nauwelaers FA, et al. Prerequisites for the analysis and sorting of extra- cellular vesicle subpopulations by high-resolution flow cytometry. Cytometry A. 2016;89(2):135–147. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25688721 Atkin-Smith GK, Paone S, Zanker DJ, et al. Isolation of cell type-specific apoptotic bodies by fluorescence-activated cell sorting. Sci Rep. 2017;7:39846. Available from: http:// www.nature.com/articles/srep39846
Minciacchi VR, Spinelli C, Reis-Sobreiro M, et al. MYC mediates large oncosome-induced fibroblast reprogramming in prostate cancer. Cancer Res. 2017;77(9):2306–2317. Available from: http://can cerres.aacrjournals.org/lookup/doi/10.1158/0008-5472. CAN-16-2942
Wunsch BH, Smith JT, Gifford SM, et al. Nanoscale lateral displacement arrays for the separation of exo- somes and colloids down to 20 nm. Nat Nanotechnol. 2016;11(11):936–940. Available from: http://www.nat ure.com/articles/nnano.2016.134
Echevarria J, Royo F, Pazos R, et al. Microarray-based identification of lectins for the purification of human urinary extracellular vesicles directly from urine samples. Chembiochem. 2014;15(11):1621–1626. Ghosh A, Davey M, Chute IC, et al. Rapid isolation of extracellular vesicles from cell culture and biological fluids using a synthetic peptide with specific affinity for heat shock proteins. PLoS One. 2014;9(10): e110443. Available from: http://dx.plos.org/10.1371/ journal.pone.0110443
Balaj L, Atai NA, Chen W, et al. Heparin affinity purification of extracellular vesicles. Sci Rep. 2015;5:10266. Available from: http://www.ncbi.nlm. nih.gov/pubmed/25988257
Fang X, Duan Y, Adkins GB, et al. Highly efficient exosome isolation and protein analysis by an inte- grated nanomaterial-based platform. Anal Chem. 2018;90(4):2787–2795. Available from: http://pubs.acs. org/doi/10.1021/acs.analchem.7b04861
Sharma P, Ludwig S, Muller L, et al. Immunoaffinity- based isolation of melanoma cell-derived exosomes from plasma of patients with melanoma. J Extracell Vesicles. 2018;7(1):1435138. Available from: https:// www.tandfonline.com/doi/full/10.1080/20013078.2018. 1435138
Brett SI, Lucien F, Guo C, et al. Immunoaffinity based methods are superior to kits for purification of
[137]
[138]
[139]
[140]
[141]
[142]
[143]
[144]
[145]
[146]
[147] [148]
prostate derived extracellular vesicles from plasma samples. Prostate. 2017;77(13):1335–1343.
Nakai W, Yoshida T, Diez D, et al. A novel affinity-based method for the isolation of highly pur- ified extracellular vesicles. Sci Rep. 2016;6(1):33935. Available from: http://www.nature.com/articles/ srep33935
Welton JL, Loveless S, Stone T, et al. Cerebrospinal fluid extracellular vesicle enrichment for protein bio- marker discovery in neurological disease; multiple sclerosis. J Extracell Vesicles. 2017;6(1):1369805. Available from: https://www.tandfonline.com/doi/full/ 10.1080/20013078.2017.1369805
Lai RC, Tan SS, Yeo RWY, et al. MSC secretes at least 3 EV types each with a unique permutation of mem- brane lipid, protein and RNA. J Extracell Vesicles. 2016;5(1):29828. Available from: https://www.tandfon line.com/doi/full/10.3402/jev.v5.29828
Gallart-Palau X, Serra A, Wong ASW, et al. Extracellular vesicles are rapidly purified from human plasma by PRotein Organic Solvent PRecipitation (PROSPR). Sci Rep. 2015;5(1):14664. Available from: http://www.nature.com/articles/srep14664
Shin H, Han C, Labuz JM, et al. High-yield isolation of extracellular vesicles using aqueous two-phase system. Sci Rep. 2015;5(1):13103. Available from: http://www. nature.com/articles/srep13103
Hurwitz SN, Nkosi D, Conlon MM, et al. CD63 reg- ulates epstein-barr virus LMP1 exosomal packaging, enhancement of vesicle production, and noncanonical NF-κB signaling. J Virol. 2017;91(5):e02251–16. Available from: http://jvi.asm.org/lookup/doi/10.1128/ JVI.02251-16
Musante L, Tataruch D, Gu D, et al. A simplified method to recover urinary vesicles for clinical applica- tions, and sample banking. Sci Rep. 2014;4(1):7532. Available from: http://www.nature.com/articles/ srep07532
Sedykh SE, Purvinish LV, Monogarov AS, et al. Purified horse milk exosomes contain an unpredictable small number of major proteins. Biochim Open. 2017;4:61–72. Available from: http://linkinghub.else vier.com/retrieve/pii/S2214008517300056 Contreras-Naranjo JC, Wu H-J, Ugaz VM Microfluidics for exosome isolation and analysis: enabling liquid biopsy for personalized medicine. Lab Chip. 2017;17(21):3558–3577.
Wu M, Ouyang Y, Wang Z, et al. Isolation of exo- somes from whole blood by integrating acoustics and microfluidics. Proc Natl Acad Sci U S A. 2017;114 (40):10584–10589. Available from: http://www.pnas. org/lookup/doi/10.1073/pnas.1709210114
Chen C, Skog J, Hsu CH, et al. Microfluidic isolation and transcriptome analysis of serum microvesicles. Lab Chip. 2010/02/04. 2010;10(4):505–511.
Liang L-G, Kong M-Q, Zhou S, et al. An integrated double-filtration microfluidic device for isolation, enrichment and quantification of urinary extracellular vesicles for detection of bladder cancer. Sci Rep. 2017;7:46224. Available from: http://www.nature.com/ articles/srep46224

[149]
[150]
[151]
[152] [153]
[154]
[155]
[156]
[157]
[158]
[159]
[160]
[161]
Shin S, Han D, Park MC, et al. Separation of extra- cellular nanovesicles and apoptotic bodies from cancer cell culture broth using tunable microfluidic systems. Sci Rep. 2017;7(1):9907. Available from: http://www. nature.com/articles/s41598-017-08826-w
Yasui T, Yanagida T, Ito S, et al. Unveiling massive numbers of cancer-related urinary-microRNA candi- dates via nanowires. Sci Adv. 2017;3(12):e1701133. Available from: http://advances.sciencemag.org/ lookup/doi/10.1126/sciadv.1701133
Zhao Z, Yang Y, Zeng Y, et al. A microfluidic exo- search chip for multiplexed exosome detection towards blood-based ovarian cancer diagnosis. Lab Chip. 2016;16(3):489–496.
Wang Z, Wu H, Fine D, et al. Ciliated micropillars for the microfluidic-based isolation of nanoscale lipid vesicles. Lab Chip. 2013;13(15):2879–2882.
Reátegui E, van der Vos KE, Lai CP, et al. Engineered nanointerfaces for microfluidic isolation and molecu- lar profiling of tumor-specific extracellular vesicles. Nat Commun. 2018;9(1):175. Available from: http:// www.nature.com/articles/s41467-017-02261-1
Böing AN, van der Pol E, Grootemaat AE, et al. Single- step isolation of extracellular vesicles by size-exclusion chromatography. J Extracell Vesicles. 2014;3:23430. Available from: https://www.tandfonline.com/doi/full/ 10.3402/jev.v3.23430
Stranska R, Gysbrechts L, Wouters J, et al. Comparison of membrane affinity-based method with size-exclusion chromatography for isolation of exosome-like vesicles from human plasma. J Transl Med. 2018;16(1):1. Available from: https://translational-medicine.biomed central.com/articles/10.1186/s12967-017-1374-6
Enderle D, Spiel A, Coticchia CM, et al. Characterization of RNA from exosomes and other extracellular vesicles isolated by a novel spin column-based method. PLoS One. 2015;10(8): e0136133. Available from: http://dx.plos.org/10.1371/ journal.pone.0136133
Jeppesen DK, Hvam ML, Primdahl-Bengtson B, et al. Comparative analysis of discrete exosome fractions obtained by differential centrifugation. J Extracell Vesicles. 2014;3:25011. Available from: http://www. ncbi.nlm.nih.gov/pubmed/25396408
Livshits MA, Khomyakova E, Evtushenko EG, et al. Isolation of exosomes by differential centrifugation: theoretical analysis of a commonly used protocol. Sci Rep. 2015;5(1):17319. Available from: http://www.ncbi. nlm.nih.gov/pubmed/26616523
Jang SC, Kim OY, Yoon CM, et al. Bioinspired exosome-mimetic nanovesicles for targeted delivery of chemotherapeutics to malignant tumors. ACS Nano. 2013;7(9):7698–7710. Available from: http:// pubs.acs.org/doi/10.1021/nn402232g
Li K, Wong DK, Hong KY, et al. Cushioned-density gradient ultracentrifugation (C-DGUC): a refined and high performance method for the isolation, character- ization, and use of exosomes. Methods Mol Biol. 2018;1740:69–83. Available from: http://link.springer. com/10.1007/978-1-4939-7652-2_7
Van Deun J, Mestdagh P, Agostinis P, et al. EV- TRACK: transparent reporting and centralizing
[162]
[163]
[164]
[165]
[166]
[167]
[168]
[169]
[170]
[171]
[172]
knowledge in extracellular vesicle research. Nat Methods. 2017;14(3):228–232. Available from: http:// www.ncbi.nlm.nih.gov/pubmed/28245209
Mitchell JP, Court J, Mason MD, et al. Increased exosome production from tumour cell cultures using the integra celline culture system. J Immunol Meth. 2008;335(1–2):98–105. Available from: http:// linkinghub.elsevier.com/retrieve/pii/ S0022175908000926
Ortiz A, Sanchez-Niño MD, Sanz AB The meaning of urinary creatinine concentration. Kidney Int. 2011;79 (7):791. Available from: http://linkinghub.elsevier. com/retrieve/pii/S0085253815548849
Cointe S, Judicone C, Robert S, et al. Standardization of microparticle enumeration across different flow cytometry platforms: results of a multicenter collabora- tive workshop. J Thromb Haemost. 2017;15 (1):187–193.
Krishnan SR, Luk F, Brown RD, et al. Isolation of human CD138(+) microparticles from the plasma of patients with multiple myeloma. Neoplasia. 2016;18 (1):25–32. Available from: http://linkinghub.elsevier. com/retrieve/pii/S1476558615001566
McVey MJ, Spring CM, Semple JW, et al. Microparticles as biomarkers of lung disease: enumera- tion in biological fluids using lipid bilayer microspheres. Am J Physiol Lung Cell Mol Physiol. 2016;310(9):L802–14. Available from: http://www.phy siology.org/doi/10.1152/ajplung.00369.2015 Atkin-Smith GK, Tixeira R, Paone S, et al. A novel mechanism of generating extracellular vesicles during apoptosis via a beads-on-a-string membrane structure. Nat Commun. 2015;6:7439. Available from: http:// www.nature.com/doifinder/10.1038/ncomms8439
van der Vlist EJ, Nolte-’T Hoen EN, Stoorvogel W, et al. Fluorescent labeling of nano-sized vesicles released by cells and subsequent quantitative and qua- litative analysis by high-resolution flow cytometry. Nat Protoc. 2012/06/23. 2012;7(7):1311–1326. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22722367 van der Pol E, van Gemert MJ, Sturk A, et al. Single vs. swarm detection of microparticles and exosomes by flow cytometry. J Thromb Haemost. 2012/03/08. 2012;10(5):919–930. Available from: http://www.ncbi. nlm.nih.gov/pubmed/22394434
Pospichalova V, Svoboda J, Dave Z, et al. Simplified protocol for flow cytometry analysis of fluorescently labeled exosomes and microvesicles using dedicated flow cytometer. J Extracell Vesicles. 2015;4(1):25530. Available from: https://www.tandfonline.com/doi/full/ 10.3402/jev.v4.25530
Tian Y, Ma L, Gong M, et al. Protein profiling and sizing of extracellular vesicles from colorectal cancer patients via flow cytometry. ACS Nano. 2018;12 (1):671–680. Available from: http://pubs.acs.org/doi/ 10.1021/acsnano.7b07782
McVey MJ, Spring CM, Kuebler WM. Improved resolution in extracellular vesicle populations using 405 instead of 488 nm side scatter. J Extracell Vesicles. 2018;7(1):1454776. Available from: https:// www.tandfonline.com/doi/full/10.1080/20013078. 2018.1454776
JOURNAL OF EXTRACELLULAR VESICLES 35

36
C. THÉRY AND K. W. WITWER
[173] [174]
[175]
[176]
[177]
[178]
[179]
[180]
[181]
[182]
[183]
[184]
[185]
Nolan JP, Stoner SA. A trigger channel threshold arti- fact in nanoparticle analysis. Cytometry A. 2013;83 (3):301–305.
Arraud N, Linares R, Tan S, et al. Extracellular vesicles from blood plasma: determination of their morphol- ogy, size, phenotype and concentration. J Thromb Haemost. 2014;12(5):614–627.
Arraud N, Gounou C, Linares R, et al. A simple flow cytometry method improves the detection of phosphatidylserine-exposing extracellular vesicles. J Thromb Haemost. 2015;13(2):237–247.
Maas SLN, de Vrij J, van der Vlist EJ, et al. Possibilities and limitations of current technologies for quantifica- tion of biological extracellular vesicles and synthetic mimics. J Control Release. 2015;200:87–96. Available from: http://linkinghub.elsevier.com/retrieve/pii/ S0168365914008384
de Vrij J, Maas SL, van Nispen M, et al. Quantification of nanosized extracellular membrane vesicles with scanning ion occlusion sensing. Nanomedicine (Lond). 2013. Available from: http://www.ncbi.nlm. nih.gov/pubmed/23384702
Obeid S, Ceroi A, Mourey G, et al. Development of a NanoBioAnalytical platform for on-chip qualification and quantification of platelet-derived microparticles. Biosens Bioelectron. 2017;93:250–259. Available from: https://linkinghub.elsevier.com/retrieve/pii/ S0956566316308569
Libregts SFWM, Arkesteijn GJA, Németh A, et al. Flow cytometric analysis of extracellular vesicle subsets in plasma: impact of swarm by particles of non-interest. J Thromb Haemost. 2018;16(7):1423–1436.
van der Pol E, Hoekstra AG, Sturk A, et al. Optical and non-optical methods for detection and characteriza- tion of microparticles and exosomes. J Thromb Haemost. 2010;8(12):2596–2607. Available from: http://www.ncbi.nlm.nih.gov/pubmed/20880256 Carnell-Morris P, Tannetta D, Siupa A, et al. Analysis of extracellular vesicles using fluorescence nanoparticle tracking analysis. Methods Mol Biol. 2017;1660:153–173. Available from: http://link. springer.com/10.1007/978-1-4939-7253-1_13
Takov K, Yellon DM, Davidson SM Confounding fac- tors in vesicle uptake studies using fluorescent lipophi- lic membrane dyes. J Extracell Vesicles. 2017;6 (1):1388731. Available from: http://www.ncbi.nlm.nih. gov/pubmed/29184625
van der Pol E, Coumans FAW, Grootemaat AE, et al. Particle size distribution of exosomes and microvesi- cles determined by transmission electron microscopy, flow cytometry, nanoparticle tracking analysis, and resistive pulse sensing. J Thromb Haemost. 2014;12 (7):1182–1192. Available from: http://www.ncbi.nlm. nih.gov/pubmed/24818656
Dragovic RA, Gardiner C, Brooks AS, et al. Sizing and phenotyping of cellular vesicles using nanoparticle tracking analysis. Nanomedicine. 2011;7(6):780–788. Available from: http://www.ncbi.nlm.nih.gov/ pubmed/21601655
Gardiner C, Ferreira YJ, Dragovic RA, et al. Extracellular vesicle sizing and enumeration by
[186]
[187]
[188]
[189]
[190]
[191]
[192]
[193]
[194]
[195]
[196]
[197]
nanoparticle tracking analysis. J Extracell Vesicles. 2013;2:19671. Available from:
Osteikoetxea X, Balogh A, Szabó-Taylor K, et al. Improved characterization of EV preparations based on protein to lipid ratio and lipid properties. PLoS One. 2015;10(3):e0121184. Available from: http://dx. plos.org/10.1371/journal.pone.0121184
Benmoussa A, Ly S, Shan ST, et al. A subset of extra- cellular vesicles carries the bulk of microRNAs in commercial dairy cow’s milk. J Extracell Vesicles. 2017;6(1):1401897. Available from: https://www.tand fonline.com/doi/full/10.1080/20013078.2017.1401897 Mihály J, Deák R, Szigyártó IC, et al. Characterization of extracellular vesicles by IR spectroscopy: fast and simple classification based on amide and CH stretch- ing vibrations. Biochim Biophys Acta. 2017;1859 (3):459–466. Available from: http://linkinghub.else vier.com/retrieve/pii/S000527361630390X Turchinovich A, Weiz L, Langheinz A, et al. Characterization of extracellular circulating microRNA. Nucleic Acids Res. 2011;39(16):7223–7233. Available from: http://www.ncbi.nlm.nih.gov/pubmed/21609964 Arroyo JD, Chevillet JR, Kroh EM, et al. Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma. Proc Natl Acad Sci U S A. 2011;108(12):5003–5008. Available from: http:// www.ncbi.nlm.nih.gov/pubmed/21383194
Vickers KC, Palmisano BT, Shoucri BM, et al. MicroRNAs are transported in plasma and delivered to recipient cells by high-density lipoproteins. Nat Cell Biol. 2011;13(4):423–433. Available from: http://www. ncbi.nlm.nih.gov/pubmed/21423178
Duijvesz D, Versluis CYL, van der Fels CAM, et al. Immuno-based detection of extracellular vesicles in urine as diagnostic marker for prostate cancer. Int J Cancer. 2015;137(12):2869–2878.
Suárez H, Gámez-Valero A, Reyes R, et al. A bead-assisted flow cytometry method for the semi-quantitative analysis of extracellular vesicles. Sci Rep. 2017;7(1):11271. Available from: http://www.nat ure.com/articles/s41598-017-11249-2
Koliha N, Wiencek Y, Heider U, et al. A novel multi- plex bead-based platform highlights the diversity of extracellular vesicles. J Extracell Vesicles. 2016;5:29975. Available from: http://www.ncbi.nlm. nih.gov/pubmed/26901056
Xia Y, Liu M, Wang L, et al. A visible and colorimetric aptasensor based on DNA-capped single-walled car- bon nanotubes for detection of exosomes. Biosens Bioelectron. 2017;92:8–15. Available from: http://linkin ghub.elsevier.com/retrieve/pii/S0956566317300635 Liang K, Liu F, Fan J, et al. Nanoplasmonic quantifica- tion of tumor-derived extracellular vesicles in plasma microsamples for diagnosis and treatment monitoring. Nat Biomed Eng. 2017;1(4):0021. Available from: http://www.nature.com/articles/s41551-016-0021 Rupert DLM, Lässer C, Eldh M, et al. Determination of exosome concentration in solution using surface plas- mon resonance spectroscopy. Anal Chem. 2014;86 (12):5929–5936. Available from: http://pubs.acs.org/ doi/10.1021/ac500931f

[198] Webber J, Clayton A How pure are your vesicles? J Extracell Vesicles. 2013;2:19861. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24009896
[199] Maiolo D, Paolini L, Di Noto G, et al. Colorimetric nanoplasmonic assay to determine purity and titrate extracellular vesicles. Anal Chem. 2015;87 (8):4168–4176. Available from: http://pubs.acs.org/ doi/abs/10.1021/ac504861d
[200] Lai RC, Arslan F, Lee MM, et al. Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury. Stem Cell Res. 2010;4(3):214–222.
[201] Cvjetkovic A, Lotvall J, Lasser C The influence of rotor type and centrifugation time on the yield and purity of extra- cellular vesicles. J Extracell Vesicles. 2014;3:23111. Available from: http://www.ncbi.nlm.nih.gov/pubmed/ 24678386
[202] Valkonen S, van der Pol E, Böing A, et al. Biological reference materials for extracellular vesicle studies. Eur J Pharm Sci. 2017;98:4–16. Available from: http://lin kinghub.elsevier.com/retrieve/pii/S0928098716303578
[203] Minciacchi VR, You S, Spinelli C, et al. Large oncosomes contain distinct protein cargo and represent a separate functional class of tumor-derived extracellular vesicles. Oncotarget. 2015;6(13):11327–11341. Available from: http://www.oncotarget.com/fulltext/3598
[204] Keerthikumar S, Gangoda L, Liem M, et al. Proteogenomic analysis reveals exosomes are more oncogenic than ectosomes. Oncotarget. 2015;6 (17):15375–15396. Available from: http://www.oncotar get.com/fulltext/3801
[205] Haraszti RA, Didiot M-C, Sapp E, et al. High- resolution proteomic and lipidomic analysis of exo- somes and microvesicles from different cell sources. J Extracell Vesicles. 2016;5(1):32570. Available from: https://www.tandfonline.com/doi/full/10.3402/jev.v5. 32570
[206] Clark DJ, Fondrie WE, Liao Z, et al. Redefining the breast cancer exosome proteome by tandem mass tag quantita- tive proteomics and multivariate cluster analysis. Anal Chem. 2015;87(20):10462–10469. Available from: http:// pubs.acs.org/doi/10.1021/acs.analchem.5b02586
[207] Durcin M, Fleury A, Taillebois E, et al. Characterisation of adipocyte-derived extracellular vesicle subtypes iden- tifies distinct protein and lipid signatures for large and small extracellular vesicles. J Extracell Vesicles. 2017;6 (1):1305677. Available from: https://www.tandfonline. com/doi/full/10.1080/20013078.2017.1305677
[208] Kowal J, Arras G, Colombo M, et al. Proteomic com- parison defines novel markers to characterize hetero- geneous populations of extracellular vesicle subtypes. Proc Natl Acad Sci. 2016;113(8):E968–77. Available from: http://www.pnas.org/lookup/doi/10.1073/pnas. 1521230113
[209] Xu R, Greening DW, Rai A, et al. Highly-purified exosomes and shed microvesicles isolated from the human colon cancer cell line LIM1863 by sequential centrifugal ultrafiltration are biochemically and func- tionally distinct. Methods. 2015;87:11–25. Available from: http://linkinghub.elsevier.com/retrieve/pii/ S1046202315001541
[210] Willms E, Johansson HJ, Mäger I, et al. Cells release subpopulations of exosomes with distinct molecular
[211]
[212]
[213]
[214]
[215]
[216]
[217]
[218]
[219]
[220]
[221]
[222]
[223]
and biological properties. Sci Rep. 2016;6(1):22519. Available from: http://www.nature.com/articles/ srep22519
Meehan B, Rak J, Di Vizio D Oncosomes – large and small: what are they, where they came from? J Extracell Vesicles. 2016;5:33109. Available from: http://www. ncbi.nlm.nih.gov/pubmed/27680302
Sódar BW, Kittel Á, Pálóczi K, et al. Low-density lipoprotein mimics blood plasma-derived exosomes and microvesicles during isolation and detection. Sci Rep. 2016;6:24316. Available from: http://www.ncbi. nlm.nih.gov/pubmed/27087061
Karimi N, Cvjetkovic A, Jang SC, et al. Detailed ana- lysis of the plasma extracellular vesicle proteome after separation from lipoproteins. Cell Mol Life Sci. 2018;75(15):2873–2886. Available from: http://link. springer.com/10.1007/s00018-018-2773-4
Østergaard O, Nielsen CT, Iversen LV, et al. Quantitative proteome profiling of normal human cir- culating microparticles. J Proteome Res. 2012;11 (4):2154–2163. Available from: http://pubs.acs.org/ doi/10.1021/pr200901p
Musante L, Saraswat M, Duriez E, et al. Biochemical and physical characterisation of urinary nanovesicles following CHAPS treatment. PLoS One. 2012;7(7): e37279. Available from: http://www.ncbi.nlm.nih.gov/ pubmed/22808001
Van Deun J, Mestdagh P, Sormunen R, et al. The impact of disparate isolation methods for extracellular vesicles on downstream RNA profiling. J Extracell Vesicles. 2014;3. Available from: http://www.ncbi.nlm. nih.gov/pubmed/25317274
McKenzie AJ, Hoshino D, Hong NH, et al. KRAS- MEK Signaling Controls Ago2 Sorting into Exosomes. Cell Rep. 2016;15(5):978–987. Available from: http://www.ncbi.nlm.nih.gov/pubmed/27117408 Melo SAA, Sugimoto H, O’Connell JT, et al. Cancer exosomes perform cell-independent microRNA bio- genesis and promote tumorigenesis. Cancer Cell. 2014;26(5):707–721. Available from: http://www.ncbi. nlm.nih.gov/pubmed/25446899
Buck AH, Coakley G, Simbari F, et al. Exosomes secreted by nematode parasites transfer small RNAs to mammalian cells and modulate innate immunity. Nat Commun. 2014;5(1):5488. Available from: http:// www.nature.com/articles/ncomms6488
Tkach M, Kowal J, Zucchetti AE, et al. Qualitative differences in T-cell activation by dendritic cell-derived extracellular vesicle subtypes. Embo J. 2017;36(20):3012–3028. Available from: http://emboj. embopress.org/lookup/doi/10.15252/embj.201696003 Jorgensen MM, Baek R, Varming K Potentials and capabilities of the Extracellular Vesicle (EV) Array. J Extracell Vesicles. 2015;4:26048. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25862471
Gool EL, Stojanovic I, Schasfoort RBM, et al. Surface plasmon resonance is an analytically sensitive method for antigen profiling of extracellular vesicles. Clin Chem. 2017;63(10):1633–1641. Available from: http://www.clin chem.org/lookup/doi/10.1373/clinchem.2016.271049
Zhu L, Wang K, Cui J, et al. Label-free quantitative detection of tumor-derived exosomes through surface
JOURNAL OF EXTRACELLULAR VESICLES 37

38
C. THÉRY AND K. W. WITWER
[224]
[225]
[226]
[227]
[228]
[229]
[230]
[231]
[232]
[233]
[234]
[235]
plasmon resonance imaging. Anal Chem. 2014;86 (17):8857–8864. Available from: http://pubs.acs.org/ doi/10.1021/ac5023056
Shao H, Im H, Castro CM, et al. New technologies for analysis of extracellular vesicles. Chem Rev. 2018;118 (4):1917–1950. Available from: http://pubs.acs.org/doi/ 10.1021/acs.chemrev.7b00534
Skotland T, Sandvig K, Llorente A Lipids in exosomes: current knowledge and the way forward. Prog Lipid Res. 2017;66:30–41. Available from: http://linkinghub. elsevier.com/retrieve/pii/S0163782716300492
Record M, Carayon K, Poirot M, et al. Exosomes as new vesicular lipid transporters involved in cell-cell commu- nication and various pathophysiologies. Biochim Biophys Acta. 2014;1841(1):108–120. Available from: http://linkin ghub.elsevier.com/retrieve/pii/S1388198113002199 Nielsen MH, Beck-Nielsen H, Andersen MN, et al. A flow cytometric method for characterization of circu- lating cell-derived microparticles in plasma. J Extracell Vesicles. 2014;3(1):20795. Available from: https://www. tandfonline.com/doi/full/10.3402/jev.v3.20795
de Gassart A, Geminard C, Fevrier B, et al. Lipid raft-associated protein sorting in exosomes. Blood. 2003;102(13):4336–4344. Available from: http://www. ncbi.nlm.nih.gov/pubmed/12881314
Gualerzi A, Niada S, Giannasi C, et al. Raman spectro- scopy uncovers biochemical tissue-related features of extracellular vesicles from mesenchymal stromal cells. Sci Rep. 2017;7(1):9820. Available from: http://www. nature.com/articles/s41598-017-10448-1
Neri T, Lombardi S, Faìta F, et al. Pirfenidone inhibits p38-mediated generation of procoagulant microparti- cles by human alveolar epithelial cells. Pulm Pharmacol Ther. 2016;39:1–6. Available from: http:// www.ncbi.nlm.nih.gov/pubmed/27237042
de Rond L, van der Pol E, Hau CM, et al. Comparison of generic fluorescent markers for detection of extra- cellular vesicles by flow cytometry. Clin Chem. 2018;64 (4):680–689. Available from: http://www.clinchem.org/ lookup/doi/10.1373/clinchem.2017.278978
Ullal AJ, Pisetsky DS, Reich CF. Use of SYTO 13, a fluorescent dye binding nucleic acids, for the detec- tion of microparticles in in vitro systems. Cytometry A. 2010;77(3):294–301.
Sansone P, Savini C, Kurelac I, et al. Packaging and transfer of mitochondrial DNA via exosomes regulate escape from dormancy in hormonal therapy-resistant breast cancer. Proc Natl Acad Sci U S A. 2017;114(43): E9066–75. Available from: http://www.pnas.org/ lookup/doi/10.1073/pnas.1704862114
Crescitelli R, Lässer C, Szabó TG, et al. Distinct RNA profiles in subpopulations of extracellular vesicles: apoptotic bodies, microvesicles and exosomes. J Extracell Vesicles. 2013;2(1):20677. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24223256 Nolte-’t Hoen EN, Buermans HP, Waasdorp M, et al. Deep sequencing of RNA from immune cell-derived vesicles uncovers the selective incor- poration of small non-coding RNA biotypes with potential regulatory functions. Nucleic Acids Res. 2012. Available from: http://www.ncbi.nlm.nih.gov/ pubmed/22821563
[236]
[237]
[238]
[239]
[240]
[241]
[242]
[243]
[244]
[245]
[246]
[247]
Villarroya-Beltri C, Gutierrez-Vazquez C, Sanchez- Cabo F, et al. Sumoylated hnRNPA2B1 controls the sorting of miRNAs into exosomes through binding to specific motifs. Nat Commun. 2013;4:2980. [2013/12/ 21]. Available from: http://www.ncbi.nlm.nih.gov/ pubmed/24356509
Vojtech L, Woo S, Hughes S, et al. Exosomes in human semen carry a distinctive repertoire of small non-coding RNAs with potential regulatory functions. Nucleic Acids Res. 2014;42(11):7290–7304. Available from: https://academic.oup.com/nar/article- lookup/doi/10.1093/nar/gku347
Tosar JP, Gambaro F, Sanguinetti J, et al. Assessment of small RNA sorting into different extracellular frac- tions revealed by high-throughput sequencing of breast cell lines. Nucleic Acids Res. 2015;43 (11):5601–5616. Available from: http://www.ncbi.nlm. nih.gov/pubmed/25940616
van Balkom BWM, Eisele AS, Pegtel DM, et al. Quantitative and qualitative analysis of small RNAs in human endothelial cells and exosomes provides insights into localized RNA processing, degradation and sorting. J Extracell Vesicles. 2015;4(1):26760. Available from: https://www.tandfonline.com/doi/full/ 10.3402/jev.v4.26760
Li K, Rodosthenous RS, Kashanchi F, et al. Advances, challenges, and opportunities in extracellular RNA biology: insights from the NIH exRNA strategic workshop. JCI Insight. 2018;3(7). Available from: https://insight.jci.org/articles/view/98942
Chen M, Xu R, Ji H, et al. Transcriptome and long noncoding RNA sequencing of three extracellular vesi- cle subtypes released from the human colon cancer LIM1863 cell line. Sci Rep. 2016;6(1):38397. Available from: http://www.nature.com/articles/srep38397
Lai CP, Kim EY, Badr CE., et al. Visualization and tracking of tumour extracellular vesicle delivery and RNA translation using multiplexed reporters. Nat Commun. 2015;6(May):7029.
Ter-Ovanesyan D, Kowal EJK, Regev A, et al. Imaging of isolated extracellular vesicles using fluorescence microscopy. Methods Mol Biol. 2017;1660:233–241. Available from: http://link.springer.com/10.1007/978- 1-4939-7253-1_19
Wu Y, Deng W, Klinke DJ. Exosomes: improved methods to characterize their morphology, RNA con- tent, and surface protein biomarkers. Analyst. 2015;140(19):6631–6642. Available from: http://www. ncbi.nlm.nih.gov/pubmed/26332016
Linares R, Tan S, Gounou C, et al. High-speed centri- fugation induces aggregation of extracellular vesicles. J Extracell Vesicles. 2015;4(0):29509. Available from: http://www.journalofextracellularvesicles.net/index. php/jev/article/view/29509
Höög JL, Lötvall J Diversity of extracellular vesicles in human ejaculates revealed by cryo-electron microscopy. J Extracell Vesicles. 2015;4:28680. Available from: http:// www.ncbi.nlm.nih.gov/pubmed/26563734
Sharma S, Rasool HI, Palanisamy V, et al. Structural- mechanical characterization of nanoparticle exosomes in human saliva, using correlative AFM, FESEM, and force spectroscopy. ACS Nano. 2010;4(4):1921–1926.

[248]
[249]
[250]
[251]
[252]
[253]
[254]
[255] [256]
[257]
[258]
[259] [260]
Available from: http://www.ncbi.nlm.nih.gov/pubmed/ 20218655
Treps L, Perret R, Edmond S, et al. Glioblastoma stem-like cells secrete the pro-angiogenic VEGF-A factor in extra- cellular vesicles. J Extracell Vesicles. 2017;6(1):1359479. Available from: https://www.tandfonline.com/doi/full/10. 1080/20013078.2017.1359479
Chen C, Zong S, Wang Z, et al. Imaging and intracel- lular tracking of cancer-derived exosomes using single-molecule localization-based super-resolution microscope. ACS Appl Mater Interfaces. 2016;8 (39):25825–25833. Available from: http://pubs.acs.org/ doi/10.1021/acsami.6b09442
Mehdiani A, Maier A, Pinto A, et al. An innovative method for exosome quantification and size measurement. J Vis Exp. 2015;95:50974. Available from: http://www.jove.com/video/50974/an-innovative -method-for-exosome-quantification-and-size- measurement
Tatischeff I, Larquet E, Falcón-Pérez JM, et al. Fast char- acterisation of cell-derived extracellular vesicles by nano- particles tracking analysis, cryo-electron microscopy, and Raman tweezers microspectroscopy. J Extracell Vesicles. 2012;1(1):19179. Available from: https://www.tandfon line.com/doi/full/10.3402/jev.v1i0.19179
Carney RP, Hazari S, Colquhoun M, et al. Multispectral optical tweezers for biochemical finger- printing of CD9-positive exosome subpopulations. Anal Chem. 2017;89(10):5357–5363. Available from: http://pubs.acs.org/doi/10.1021/acs.analchem.7b00017 Smith ZJ, Lee C, Rojalin T, et al. Single exosome study reveals subpopulations distributed among cell lines with variability related to membrane content. J Extracell Vesicles. 2015;4(1):28533. Available from: https://www. tandfonline.com/doi/full/10.3402/jev.v4.28533
Stoner SA, Duggan E, Condello D, et al. High sensi- tivity flow cytometry of membrane vesicles. Cytom Part A. 2016;89(2):196–206. Available from: http:// www.ncbi.nlm.nih.gov/pubmed/26484737
Nolan JP, Jones JC. Detection of platelet vesicles by flow cytometry. Platelets. 2017;28(3):256–262. Available from: http://www.ncbi.nlm.nih.gov/pubmed/28277059
Sitar S, Kejžar A, Pahovnik D, et al. Size characterization and quantification of exosomes by asymmetrical-flow field-flow fractionation. Anal Chem. 2015;87 (18):9225–9233. Available from: http://pubs.acs.org/doi/ 10.1021/acs.analchem.5b01636
Heusermann W, Hean J, Trojer D, et al. Exosomes surf on filopodia to enter cells at endocytic hot spots, traffic within endosomes, and are targeted to the ER. J Cell Biol. 2016;213(2):173–184. Available from: http://www. ncbi.nlm.nih.gov/pubmed/27114500
Wyss R, Grasso L, Wolf C, et al. Molecular and dimen- sional profiling of highly purified extracellular vesicles by fluorescence fluctuation spectroscopy. Anal Chem. 2014;86(15):7229–7233. Available from: http://pubs. acs.org/doi/10.1021/ac501801m
Baietti MF, Zhang Z, Mortier E, et al. Syndecan–syn- tenin–ALIX regulates the biogenesis of exosomes. Nat Cell Biol. 2012;14(7):677–685.
Erdbrügger U, Rudy CK, Etter ME, et al. Imaging flow cytometry elucidates limitations of microparticle
[261]
[262]
[263]
[264]
[265]
[266]
[267]
[268]
[269]
[270]
[271]
[272]
[273]
analysis by conventional flow cytometry. Cytometry A. 2014;85(9):756–770.
Headland SE, Jones HR, Asv D, et al. Cutting-edge analysis of extracellular microparticles using ImageStream(X) imaging flow cytometry. Sci Rep. 2014;4(1):5237. Available from: http://www.nature. com/articles/srep05237
Lee K, Fraser K, Ghaddar B, et al. Multiplexed profil- ing of single extracellular vesicles. ACS Nano. 2018;12 (1):494–503. Available from: http://pubs.acs.org/doi/ 10.1021/acsnano.7b07060
Daaboul GG, Lopez CA, Yurt A, et al. Label-free optical biosensors for virus detection and characterization. IEEE J Sel Top Quantum Electron. 2012;18(4):1422–1433.
Daaboul GG, Freedman DS, Scherr SM, et al. Enhanced light microscopy visualization of virus par- ticles from Zika virus to filamentous ebolaviruses. PLoS One. 2017;12(6):e0179728.
van der Pol E, Sturk A, van Leeuwen T, et al., ISTH- SSC-VB Working group. Standardization of extracel- lular vesicle measurements by flow cytometry through vesicle diameter approximation. J Thromb Haemost. 2018;16(6):1236–1245.
Cvjetkovic A, Jang SC, Konečná B, et al. Detailed analysis of protein topology of extracellular vesicles-evidence of unconventional membrane protein orientation. Sci Rep. 2016;6(1):36338. Available from: http://www.nature.com/articles/srep36338
Deregibus MC, Cantaluppi V, Calogero R, et al. Endothelial progenitor cell derived microvesicles acti- vate an angiogenic program in endothelial cells by a horizontal transfer of mRNA. Blood. 2007;110 (7):2440–2448. Available from: http://www.bloodjour nal.org/cgi/doi/10.1182/blood-2007-03-078709
Sharma A, Mariappan M, Appathurai S, et al. In vitro dissection of protein translocation into the mammalian endoplasmic reticulum. Methods Mol Biol. 2010;619:339–363. Available from: http://link. springer.com/10.1007/978-1-60327-412-8_20
Sung BH, Weaver AM. Exosome secretion promotes chemotaxis of cancer cells. Cell Adh Migr. 2017;11 (2):187–195. Available from: https://www.tandfonline. com/doi/full/10.1080/19336918.2016.1273307 Osteikoetxea X, Sódar B, Németh A, et al. Differential detergent sensitivity of extracellular vesicle subpopulations. Org Biomol Chem. 2015;13 (38):9775–9782. Available from: http://www.ncbi.nlm. nih.gov/pubmed/26264754
Parolini I, Federici C, Raggi C, et al. Microenvironmental pH is a key factor for exosome traffic in tumor cells. J Biol Chem. 2009;284 (49):34211–34222. Available from: http://www.jbc. org/lookup/doi/10.1074/jbc.M109.041152
Franzen CA, Simms PE, Van Huis AF, et al. Characterization of uptake and internalization of exo- somes by bladder cancer cells. Biomed Res Int. 2014;2014:619829. Available from: http://www.hin dawi.com/journals/bmri/2014/619829/
Christianson HC, Svensson KJ, van Kuppevelt TH, et al. Cancer cell exosomes depend on cell-surface heparan sulfate proteoglycans for their internalization
JOURNAL OF EXTRACELLULAR VESICLES 39

40
C. THÉRY AND K. W. WITWER
[274]
[275]
[276]
[277]
[278]
[279]
[280]
[281]
[282]
[283]
[284]
and functional activity. Proc Natl Acad Sci U S A. 2013;110(43):17380–17385. Available from: http:// www.pnas.org/cgi/doi/10.1073/pnas.1304266110 Mulcahy LA, Pink RC, Carter DRF. Routes and mechanisms of extracellular vesicle uptake. J Extracell Vesicles. 2014;3:24641. Available from: https://www. tandfonline.com/doi/full/10.3402/jev.v3.24641 Wahlgren J, Karlson TDL, Glader P, et al. Activated human T cells secrete exosomes that participate in IL-2 mediated immune response signaling. PLoS One. 2012;7 (11):e49723. Available from: http://dx.plos.org/10.1371/ journal.pone.0049723
Szabó GT, Tarr B, Pálóczi K, et al. Critical role of extracellular vesicles in modulating the cellular effects of cytokines. Cell Mol Life Sci. 2014;71(20):4055–4067. Available from: http://link.springer.com/10.1007/ s00018-014-1618-z
Gámez-Valero A, Monguió-Tortajada M, Carreras- Planella L, et al. Size-exclusion chromatography- based isolation minimally alters extracellular vesicles’ characteristics compared to precipitating agents. Sci Rep. 2016;6(1):33641. Available from: http://www.nat ure.com/articles/srep33641
Paolini L, Zendrini A, Di Noto G, et al. Residual matrix from different separation techniques impacts exosome biological activity. Sci Rep. 2016;6(1):23550. Available from: http://www.nature.com/articles/ srep23550
Gyorgy B, Modos K, Pallinger E, et al. Detection and isolation of cell-derived microparticles are compro- mised by protein complexes resulting from shared biophysical parameters. Blood. 2011;117(4):e39–48. Available from: http://www.ncbi.nlm.nih.gov/ pubmed/21041717
Benedikter BJ, Bouwman FG, Vajen T, et al. Ultrafiltration combined with size exclusion chroma- tography efficiently isolates extracellular vesicles from cell culture media for compositional and functional studies. Sci Rep. 2017;7(1):15297. Available from: http://www.nature.com/articles/s41598-017-15717-7 Trajkovic K, Hsu C, Chiantia S, et al. Ceramide trig- gers budding of exosome vesicles into multivesicular endosomes. Science. 2008;319(5867):1244–1247. Available from: http://www.sciencemag.org/cgi/doi/ 10.1126/science.1153124
Figuera-Losada M, Stathis M, Dorskind JM, et al. Cambinol, a novel inhibitor of neutral sphingomyeli- nase 2 shows neuroprotective properties. PLoS One. 2015;10(5):e0124481. Available from: http://dx.plos. org/10.1371/journal.pone.0124481
Dinkins MB, Enasko J, Hernandez C, et al. Neutral sphingomyelinase-2 deficiency ameliorates alzheimer’s disease pathology and improves cognition in the 5XFAD mouse. J Neurosci. 2016;36(33):8653–8667. Available from: http://www.jneurosci.org/lookup/doi/ 10.1523/JNEUROSCI.1429-16.2016
Cruz FF, Borg ZD, Goodwin M, et al. Systemic admin- istration of human bone marrow-derived mesenchy- mal stromal cell extracellular vesicles ameliorates aspergillus hyphal extract-induced allergic airway inflammation in immunocompetent mice. Stem Cells Transl Med. 2015;4(11):1302–1316.
[285]
[286]
[287]
[288]
[289]
[290]
[291]
[292]
[293]
[294]
[295]
[296]
[297]
Villarroya-Beltri C, Baixauli F, Mittelbrunn M, et al. ISGylation controls exosome secretion by promoting lysosomal degradation of MVB proteins. Nat Commun. 2016;7:13588. Available from: http://www. nature.com/doifinder/10.1038/ncomms13588
Savina A, Vidal M, Colombo MI. The exosome path- way in K562 cells is regulated by Rab11. J Cell Sci. 2002;115(Pt 12):2505–2515. Available from: http:// www.ncbi.nlm.nih.gov/pubmed/12045221
Ostrowski M, Carmo NB, Krumeich S, et al. Rab27a and Rab27b control different steps of the exosome secretion pathway. Nat Cell Biol. 2010;12(1):13–19. Available from: http://www.ncbi.nlm.nih.gov/ pubmed/19966785
Hsu C, Morohashi Y, Yoshimura S-I, et al. Regulation of exosome secretion by Rab35 and its GTPase-activating proteins TBC1D10A-C. J Cell Biol. 2010;189(2):223–232. Available from: http://www.ncbi. nlm.nih.gov/pubmed/20404108
Hyenne V, Apaydin A, Rodriguez D, et al. RAL-1 controls multivesicular body biogenesis and exosome secretion. J Cell Biol. 2015;211(1):27–37. Available from: http://www.jcb.org/lookup/doi/10.1083/jcb. 201504136
Gross JC, Chaudhary V, Bartscherer K, et al. Active Wnt proteins are secreted on exosomes. Nat Cell Biol. 2012;14(10):1036–1045. Available from: http://www. nature.com/articles/ncb2574
Imjeti NS, Menck K, Egea-Jimenez AL, et al. Syntenin mediates SRC function in exosomal cell-to-cell communication. Proc Natl Acad Sci U S A. 2017;114 (47):12495–12500. Available from: http://www.pnas. org/lookup/doi/10.1073/pnas.1713433114
Sinha S, Hoshino D, Hong NH, et al. Cortactin pro- motes exosome secretion by controlling branched actin dynamics. J Cell Biol. 2016;214(2):197–213. Available from: http://www.jcb.org/lookup/doi/10.1083/jcb. 201601025
Jackson CE, Scruggs BS, Schaffer JE, et al. Effects of inhibiting VPS4 support a general role for ESCRTs in extracellular vesicle biogenesis. Biophys J. 2017;113 (6):1342–1352. Available from: http://linkinghub.else vier.com/retrieve/pii/S0006349517305714
Chalmin F, Ladoire S, Mignot G, et al. Membrane- associated Hsp72 from tumor-derived exosomes med- iates STAT3-dependent immunosuppressive function of mouse and human myeloid-derived suppressor cells. J Clin Invest. 2010;120(2):457–471. Available from: http://www.jci.org/articles/view/40483 Montecalvo A, Larregina AT, Shufesky WJ, et al. Mechanism of transfer of functional microRNAs between mouse dendritic cells via exosomes. Blood. 2012;119(3):756–766. Available from: http://www. ncbi.nlm.nih.gov/pubmed/22031862
Savina A, Furlán M, Vidal M, et al. Exosome release is regulated by a calcium-dependent mechanism in K562 cells. J Biol Chem. 2003;278(22):20083–20090. Available from: http://www.jbc.org/lookup/doi/10. 1074/jbc.M301642200
Minakaki G, Menges S, Kittel A, et al. Autophagy inhibition promotes SNCA/alpha-synuclein release and transfer via extracellular vesicles with a hybrid

autophagosome-exosome-like phenotype. Autophagy. 2018;14(1):98–119. Available from: https://www.tand fonline.com/doi/full/10.1080/15548627.2017.1395992
[298] Edgar JR, Manna PT, Nishimura S, et al. Tetherin is an exosomal tether. Elife. 2016;5:17180. Available from: https://elifesciences.org/articles/17180
[299] Atai NA, Balaj L, van Veen H, et al. Heparin blocks transfer of extracellular vesicles between donor and reci- pient cells. J Neurooncol. 2013. Available from: http:// www.ncbi.nlm.nih.gov/pubmed/24002181
[300] Wang Q, Lu Q Plasma membrane-derived extracellular microvesicles mediate non-canonical intercellular NOTCH signaling. Nat Commun. 2017;8(1):709. Available from: http://www.nature.com/articles/ s41467-017-00767-2
[301] Nabhan JF, Hu R, Oh RS, et al. Formation and release of arrestin domain-containing protein 1-mediated microvesicles (ARMMs) at plasma membrane by recruitment of TSG101 protein. Proc Natl Acad Sci U S A. 2012;109(11):4146–4151. Available from: http://www.pnas.org/cgi/doi/10.1073/pnas.1200448109
[302] Muralidharan-Chari V, Clancy J, Plou C, et al. ARF6- regulated shedding of tumor cell-derived plasma mem- brane microvesicles. Curr Biol. 2009;19(22):1875–1885. Available from: http://linkinghub.elsevier.com/ retrieve/pii/S0960982209017722
[303] Burger D, Montezano AC, Nishigaki N, et al. Endothelial microparticle formation by angiotensin II is mediated via Ang II receptor type I/NADPH oxi- dase/Rho kinase pathways targeted to lipid rafts. Arterioscler Thromb Vasc Biol. 2011;31 (8):1898–1907. Available from: http://atvb.ahajour nals.org/cgi/doi/10.1161/ATVBAHA.110.222703
[304] Gao C, Li R, Liu Y, et al. Rho-kinase-dependent F-actin rearrangement is involved in the release of endothelial microparticles during IFN-α-induced endothelial cell apoptosis. J Trauma Acute Care Surg. 2012;73(5):1152–1160. Available from: http://content. wkhealth.com/linkback/openurl?sid=WKPTLP:landing page&an=01586154-201211000-00017
[305] Yu X, Xu J, Liu W, et al. Bubbles induce endothelial microparticle formation via a calcium-dependent path- way involving flippase inactivation and rho kinase activation. Cell Physiol Biochem. 2018;46(3):965–974. Available from: https://www.karger.com/Article/ FullText/488825
[306] Di Vizio D, Kim J, Hager MH, et al. Oncosome for- mation in prostate cancer: association with a region of frequent chromosomal deletion in metastatic disease. Cancer Res. 2009;69(13):5601–5609. Available from: http://cancerres.aacrjournals.org/cgi/doi/10.1158/0008- 5472.CAN-08-3860
[307] Schwechheimer C, Kuehn MJ. Outer-membrane vesi- cles from Gram-negative bacteria: biogenesis and functions. Nat Rev Microbiol 2015;13(10):605–619. Available from: http://www.nature.com/articles/ nrmicro3525
[308]
[309]
[310]
[311]
[312]
[313]
[314]
[315]
[316]
[317]
[318]
[319]
Colombo M, Raposo G, Théry C Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev Biol. 2014;30 (1):255–289. Available from: http://www.ncbi.nlm.nih. gov/pubmed/25288114
Romancino DP, Paterniti G, Campos Y, et al. Identification and characterization of the nano-sized vesicles released by muscle cells. FEBS Lett. 2013;587 (9):1379–1384.
Booth AM, Fang Y, Fallon JK, et al. Exosomes and HIV Gag bud from endosome-like domains of the T cell plasma membrane. J Cell Biol. 2006;172 (6):923–935. Available from: http://www.ncbi.nlm.nih. gov/pubmed/16533950
Hoang TQ, Rampon C, Freyssinet J-M, et al. A method to assess the migration properties of cell-derived microparticles within a living tissue. Biochim Biophys Acta. 2011;1810(9):863–866. Available from: http://lin kinghub.elsevier.com/retrieve/pii/S0304416511001061 Menck K, Sönmezer C, Worst TS, et al. Neutral sphin- gomyelinases control extracellular vesicles budding from the plasma membrane. J Extracell Vesicles. 2017;6(1):1378056. Available from: https://www.tand fonline.com/doi/full/10.1080/20013078.2017.1378056 Bobrie A, Colombo M, Krumeich S, et al. Diverse subpopulations of vesicles secreted by different intra- cellular mechanisms are present in exosome prepara- tions obtained by differential ultracentrifugation. J Extracell Vesicles. 2012;1:18297.
Peinado H, Alečković M, Lavotshkin S, et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med. 2012;18(6):883–891. Available from: http:// www.ncbi.nlm.nih.gov/pubmed/22635005
Kim DK, Kang B, Kim OY, et al. EVpedia: an inte- grated database of high-throughput data for systemic analyses of extracellular vesicles. J Extracell Vesicles. 2013;2. Available from: http://www.ncbi.nlm.nih.gov/ pubmed/24009897
Kim D-K, Lee J, Kim SR, et al. EVpedia: a community web portal for extracellular vesicles research. Bioinformatics. 2015;31(6):933–939. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25388151
Kalra H, Simpson RJ, Ji H, et al. Vesiclepedia: a compendium for extracellular vesicles with continu- ous community annotation. PLoS Biol. 2012;10(12): e1001450.
Mathivanan S, Simpson RJ. ExoCarta: A compendium of exosomal proteins and RNA. Proteomics. 2009;9 (21):4997–5000. Available from: http://www.ncbi.nlm. nih.gov/pubmed/19810033
Subramanian SL, Kitchen RR, Alexander R, et al. Integration of extracellular RNA profiling data using metadata, biomedical ontologies and linked data technologies. J Extracell Vesicles. 2015;4:27497. Available from: http://www.ncbi.nlm.nih.gov/ pubmed/26320941
JOURNAL OF EXTRACELLULAR VESICLES 41

42 C. THÉRY AND K. W. WITWER
MISEV2018 Checklist
Numbers refer to sections listed in the Table of contents from: C. Théry and K.W.Witwer, et al, ”Minimal Information for Studies of Extracellular Vesicles 2018 (MISEV2018): a posi- tion statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines”, J Extracell Vesicles 2018;7:1535750.
Storage and recovery (Section 2-d)
+++ Storage and recovery (e.g., thawing) of CCM, biofluid, or tissue before EV isolation (storage tempera- ture, vessel, time; method of thawing or other sample preparation)
+++ Storage and recovery of EVs after isolation (tempera- ture, vessel, time, additive(s)…)
3-EV separation and concentration
Experimental details of the method
++ Centrifugation: reference number of tube(s), rotor(s), adjusted k factor(s) of each centrifugation step (= time+ speed+ rotor, volume/density of centrifugation condi- tions), temperature, brake settings
++ Density gradient: nature of matrix, method of gen- erating gradient, reference (and size) of tubes, bottom- up (sample at bottom, high density) or top-bottom (sample on top, low density), centrifugation speed and time (with brake specified), method and volume of fraction recovery
++ Chromatography: matrix (nature, pore size,…), loaded sample volume, fraction volume, number
++ Precipitation: reference of polymer, ratio vol/vol or weight/vol polymer/fluid, time/temperature of incuba- tion, time/speed/temperature of centrifugation
++ Filtration: reference of filter type (=nature of mem- brane, pore size…), time and speed of centrifugation, volume before/after (in case of concentration)
++ Antibody-based : reference of antibodies, mass Ab/ amount of EVs, nature of Ab carrier (bead, surface) and amount of Ab/carrier surface
++ Other…: all necessary details to allow replication ++ Additional step(s) to concentrate, if any
++ Additional step(s) to wash matrix and/or sample, if any
Specify category of the chosen EV separation/concentra- tion method (Table 1):

  • High recovery, low specificity = mixed EVs and non- EV components OR
  • Intermediate recovery, intermediate specificity = mixed EVs with limited non-EV components OR
  • Low recovery, high specificity = subtype(s) of EVs with as little non-EV as possible OR
  • High recovery, high specificity = subtype(s) of EVs with as little non-EV as possible
    4-EV characterization
    Quantification (Table 2a, Section 4-a)
    +++ Volume of fluid, and/or cell number, and/or tissue mass used to isolate EVs
    +++ Global quantification by at least 2 methods: pro- tein amount, particle number, lipid amount, expressed per volume of initial fluid or number of producing cells/mass of tissue
    +++ Ratio of the 2 quantification figures
    Global characterization (Section 4-b, Table 3)
    +++ Transmembrane or GPI-anchored protein loca- lized in cells at plasma membrane or endosomes
    +++ Cytosolic protein with membrane-binding or – association capacity
    +++ Mandatory
    1-Nomenclature
    Mandatory
    ++ Mandatory if applicable
  • Encouraged
    +++ Generic term extracellular vesicle (EV): With demonstration of extracellular (no intact cells) and vesi- cular nature per these characterization (Section 4) and function (Section 5) guidelines OR
    +++ Generic term, e.g., extracellular particle (EP): no intact cells but MISEV guidelines not satisfied Encouraged (choose one)
  • Generic term extracellular vesicle (EV) + specification (size, density, other)
  • Specific term for subcellular origin: e.g., ectosome, microparticle, microvesicle (from plasma membrane), exosome (from endosomes), with demonstration of the subcellular origin
  • Other specific term: with definition of specific criteria
    2-Collection and pre-processing
    Tissue Culture Conditioned medium (CCM, Section 2-a)
    +++ General cell characterization (identity, passage, myco- plasma check…)
    +++ Medium used before and during collection (additives, serum, other)
    ++ exact protocol for depletion of EVs/EPs from addi- tives in collection medium
    +++ Nature and size of culture vessels, and volume of medium during conditioning
    ++ specific culture conditions (treatment, % O2, coat- ing, polarization…) before and during collection
    +++ Number of cells/ml or /surface area and % of live/ dead cells at time of collection (or at time of seeding with estimation at time of collection)
    +++ Frequency and interval of CM harvest
    Biofluids or Tissues (Sections 2-b and -c)
    ++ Donor status if available (age, sex, food/water intake, collection time, disease, medication, other) +++ Volume of biofluid or volume/mass of tissue sam- ple collected per donor
    ++ Total volume/mass used for EV isolation (if pooled from several donors)
    +++ All known collection conditions, including addi- tives, at time of collection
    +++ Pre-treatment to separate major fluid-specific con- taminants before EV isolation
    +++ Temperature and time of biofluid/tissue handling before and during pre-treatment
    ++ For cultured tissue explants: volume, nature of medium and time of culture before collecting condi- tioned medium
    ++ For direct tissue EV extraction: treatment of tissue to release vesicles without disrupting cells

+++ Assessment of presence/absence of expected contaminants
(At least one each of the three categories above)
++ Presence of proteins associated with compartments other than plasma membrane or endosomes
++ Presence of soluble secreted proteins and their likely transmembrane ligands

  • Topology of the relevant functional components (Section 4-d)
    Single EV characterization (Section 4-c)
    +++ Images of single EVs by wide-field and close-up: e.g. electron microscopy, scanning probe microscopy, super-resolution fluorescence microscopy
    +++ Non-image-based method analysing large numbers of single EVs: NTA, TRPS, FCS, high-resolution flow cytome- try, multi-angle light-scattering, Raman spectroscopy, etc.
    5-Functional studies
    +++ Dose-response assessment
    +++ Negative control = nonconditioned medium, bio- fluid/tissue from control donors, as applicable
    +++ Quantitative comparison of functional activity of total fluid, vs EV-depleted fluid, vs EVs (after high recovery/low specificity separation)
    +++ Quantitative comparison of functional activity of EVs vs other EPs/fractions after low recovery/high spe- cificity separation
  • Quantitative comparison of activity of EV subtypes (if subtype-specific function claimed)
  • Extent of functional activity in the absence of contact between EV donor and EV recipient
    6-Reporting
  • Submission of methodologic details to EV-TRACK (evtrack.org) with EV-TRACK number provided (strongly encouraged)
    +++ Submission of data (proteomic, sequencing, other) to relevant public, curated databases or open-access repositories
  • Data submission to EV-specific databases (e.g., EVpedia, Vesiclepedia, exRNA atlas)
    ++ Temper EV-specific claims when MISEV require- ments cannot be entirely satisfied (Section 6-b)
    JOURNAL OF EXTRACELLULAR VESICLES 43