1
|
Bebesi T, Pálmai M, Szigyártó IC, Gaál A, Wacha A, Bóta A, Varga Z, Mihály J. Surface-enhanced infrared spectroscopic study of extracellular vesicles using plasmonic gold nanoparticles. Colloids Surf B Biointerfaces 2025; 246:114366. [PMID: 39531836 DOI: 10.1016/j.colsurfb.2024.114366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/30/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
Extracellular vesicles (EVs), sub-micrometer lipid-bound particles released by most cells, are considered a novel area in both biology and medicine. Among characterization methods, infrared (IR) spectroscopy, especially attenuated total reflection (ATR), is a rapidly emerging label-free tool for molecular characterization of EVs. The relatively low number of vesicles in biological fluids (∼1010 particle/mL), however, and the complex content of the EVs' milieu (protein aggregates, lipoproteins, buffer molecules) might result in poor signal-to-noise ratio in the IR analysis of EVs. Exploiting the increment of the electromagnetic field at the surface of plasmonic nanomaterials, surface-enhanced infrared spectroscopy (SEIRS) provides an amplification of characteristic IR signals of EV samples. Negatively charged citrate-capped and positively charged cysteamine-capped gold nanoparticles with around 10 nm diameter were synthesized and tested with blood-derived EVs. Both types of gold nanoparticles contributed to an enhancement of the EVs' IR spectroscopic signature. Joint evaluation of UV-Vis and IR spectroscopic results, supported by FF-TEM images, revealed that proper interaction of gold nanoparticles with EVs is crucial, and an aggregation or clustering of gold nanoparticles is necessary to obtain the SEIRS effect. Positively charged gold nanoparticles resulted in higher enhancement, probably due to electrostatic interaction with EVs, commonly negatively charged.
Collapse
Affiliation(s)
- Tímea Bebesi
- Institute of Materials and Environmental Chemistry, HUN-REN Research Centre for Natural Sciences (RCNS), Magyar tudósok körútja 2, Budapest 1117, Hungary; Hevesy György PhD School of Chemistry, Eötvös Lóránd University, Pázmány Péter sétány 1/A, Budapest 1117, Hungary
| | - Marcell Pálmai
- Institute of Materials and Environmental Chemistry, HUN-REN Research Centre for Natural Sciences (RCNS), Magyar tudósok körútja 2, Budapest 1117, Hungary
| | - Imola Csilla Szigyártó
- Institute of Materials and Environmental Chemistry, HUN-REN Research Centre for Natural Sciences (RCNS), Magyar tudósok körútja 2, Budapest 1117, Hungary
| | - Anikó Gaál
- Institute of Materials and Environmental Chemistry, HUN-REN Research Centre for Natural Sciences (RCNS), Magyar tudósok körútja 2, Budapest 1117, Hungary
| | - András Wacha
- Institute of Materials and Environmental Chemistry, HUN-REN Research Centre for Natural Sciences (RCNS), Magyar tudósok körútja 2, Budapest 1117, Hungary
| | - Attila Bóta
- Institute of Materials and Environmental Chemistry, HUN-REN Research Centre for Natural Sciences (RCNS), Magyar tudósok körútja 2, Budapest 1117, Hungary
| | - Zoltán Varga
- Institute of Materials and Environmental Chemistry, HUN-REN Research Centre for Natural Sciences (RCNS), Magyar tudósok körútja 2, Budapest 1117, Hungary; Department of Physical Chemistry and Materials Science, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3, Budapest 1111, Hungary
| | - Judith Mihály
- Institute of Materials and Environmental Chemistry, HUN-REN Research Centre for Natural Sciences (RCNS), Magyar tudósok körútja 2, Budapest 1117, Hungary; Department of Chemistry, Eszterházy Károly Catholic University, Leányka u. 6, Eger 3300, Hungary.
| |
Collapse
|
2
|
Loria F, Picciotto S, Adamo G, Zendrini A, Raccosta S, Manno M, Bergese P, Liguori GL, Bongiovanni A, Zarovni N. A decision-making tool for navigating extracellular vesicle research and product development. J Extracell Vesicles 2024; 13:e70021. [PMID: 39670350 PMCID: PMC11638734 DOI: 10.1002/jev2.70021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 10/31/2024] [Accepted: 11/26/2024] [Indexed: 12/14/2024] Open
Abstract
Due to their intercellular communication properties and involvement in a wide range of biological processes, extracellular vesicles (EVs) are increasingly being studied and exploited for different applications. Nevertheless, their complex nature and heterogeneity, as well as the challenges related to their purification and characterization procedures, require a cautious assessment of the qualitative and quantitative parameters that need to be monitored. This translates into a multitude of choices and putative solutions that any EV researcher must confront in both research and translational environments. In this respect, decision-making tools may help assess various options, weigh pros and cons, and ultimately arrive at a thought-out decision that considers both the best fit-to-source and fit-to-scope EV application(s). Here, we present a multi-criteria EV decision-making grid (EV-DMG) as a novel, efficient, customizable, and easy-to-use tool to support EV research and innovation. By identifying and weighing key assessment criteria for comparing distinct EV-based preparations and related processes, our EV-DMG may assist any EV community member in making informed, traceable, and reproducible decisions regarding the management of EV sources or samples. Ultimately, this EV-DMG may guide the adoption of the most suitable EV production and analytical pipelines for targeting a defined aim or application.
Collapse
Affiliation(s)
- Francesca Loria
- HansaBioMed Life Sciences LtdTallinnEstonia
- Department of Chemistry and BiotechnologyTallinn University of TechnologyTallinnEstonia
| | - Sabrina Picciotto
- Institute of Biomedical Research and Innovation (IRIB)National Research Council of ItalyPalermoItaly
| | - Giorgia Adamo
- Institute of Biomedical Research and Innovation (IRIB)National Research Council of ItalyPalermoItaly
| | - Andrea Zendrini
- Department of Molecular and Translational MedicineUniversity of BresciaBresciaItaly
- CSGIItalian Center for Colloid and Interface ScienceFlorenceItaly
| | - Samuele Raccosta
- Institute of BiophysicsNational Research Council of ItalyPalermoItaly
| | - Mauro Manno
- Institute of BiophysicsNational Research Council of ItalyPalermoItaly
- EVEBiofactory SrlPalermoItaly
| | - Paolo Bergese
- Department of Molecular and Translational MedicineUniversity of BresciaBresciaItaly
- CSGIItalian Center for Colloid and Interface ScienceFlorenceItaly
| | - Giovanna L. Liguori
- Institute of Genetics and Biophysics (IGB)National Research Council of ItalyNaplesItaly
| | - Antonella Bongiovanni
- Institute of Biomedical Research and Innovation (IRIB)National Research Council of ItalyPalermoItaly
- EVEBiofactory SrlPalermoItaly
| | | |
Collapse
|
3
|
Salvi V, Gaudenzi C, Mariotti B, Giongrandi G, Alacqua S, Gianello V, Schioppa T, Tiberio L, Ceribelli A, Selmi C, Bergese P, Calza S, Del Prete A, Sozzani S, Bazzoni F, Bosisio D. Cell damage shifts the microRNA content of small extracellular vesicles into a Toll-like receptor 7-activating cargo capable to propagate inflammation and immunity. Cell Commun Signal 2024; 22:536. [PMID: 39516877 PMCID: PMC11545887 DOI: 10.1186/s12964-024-01924-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND The physiological relevance of cell-to-cell communication mediated by small extracellular vesicle-encapsulated microRNAs (sEV-miRNAs) remains debated because of the limiting representativity of specific miRNAs within the extracellular pool. We hypothesize that sEV-miRNA non-canonical function consisting of the stimulation of Toll-like receptor 7 (TLR7) may rely on a global shift of the sEV cargo rather than on the induction of one or few specific miRNAs. Psoriasis represents an ideal model to test such hypothesis as it is driven by overt activation of TLR7-expressing plasmacytoid dendritic cells (pDCs) following keratinocyte damage. METHODS To mimic the onset of psoriasis, keratinocytes were treated with a cocktail of psoriatic cytokines or UV-irradiated. SmallRNA sequencing was performed on sEVs released by healthy and UV-treated keratinocytes. sEV-miRNAs were analyzed for nucleotide composition as well as for the presence of putative TLR7-binding triplets. Primary human pDCs where stimulated with sEVs +/- inhibitors of TLR7 (Enpatoran), of sEV release (GW4869 + manumycin) and of TLR7-mediated pDC activation (anti-BDCA-2 antibody). Secretion of type I IFNs and activation of CD8+T cells were used as readouts. qPCR on psoriatic and healthy skin biopsies was conducted to identify induced miRNAs. RESULTS sEV-miRNAs released by damaged keratinocytes revealed a significantly higher content of TLR7-activating sequences than healthy cells. As expected, differential expression analysis confirmed the presence of miRNAs upregulated in psoriatic skin, including miR203a. More importantly, 76.5% of induced miRNAs possessed TLR7-binding features and among these we could detect several previously demonstrated TLR7 ligands. In accordance with this in silico analysis, sEVs from damaged keratinocytes recapitulated key events of psoriatic pathogenesis by triggering pDCs to release type I interferon and activate cytotoxic CD8+T cells in a TLR7- and sEV-dependent manner. DISCUSSION Our results demonstrate that miR203a is just one paradigmatic TLR7-activating miRNA among the hundreds released by UV-irradiated keratinocytes, which altogether trigger pDC activation in psoriatic conditions. This represents the first evidence that cell damage shifts the miRNA content of sEVs towards a TLR7-activating cargo capable to propagate inflammation and immunity, offering strong support to the physiological role of systemic miRNA-based cell-to-cell communication.
Collapse
Affiliation(s)
- Valentina Salvi
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, Brescia, 25123, Italy
| | - Carolina Gaudenzi
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, Brescia, 25123, Italy
| | | | - Gaia Giongrandi
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, Brescia, 25123, Italy
| | - Silvia Alacqua
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, Brescia, 25123, Italy
| | - Veronica Gianello
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, Brescia, 25123, Italy
| | - Tiziana Schioppa
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, Brescia, 25123, Italy
- IRCCS Humanitas Research Hospital, Milan, Italy
| | - Laura Tiberio
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, Brescia, 25123, Italy
| | - Angela Ceribelli
- Department of Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Carlo Selmi
- Department of Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Paolo Bergese
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, Brescia, 25123, Italy
| | - Stefano Calza
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, Brescia, 25123, Italy
| | - Annalisa Del Prete
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, Brescia, 25123, Italy
- IRCCS Humanitas Research Hospital, Milan, Italy
| | - Silvano Sozzani
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Institute Pasteur-Italia, Rome, Italy
| | - Flavia Bazzoni
- Department of Medicine, University of Verona, Verona, Italy
| | - Daniela Bosisio
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, Brescia, 25123, Italy.
| |
Collapse
|
4
|
Mateos H, Mallardi A, Oliver M, Dell'Aglio M, Giannone P, Palazzo G. Cooperative aggregation of gold nanoparticles on phospholipid vesicles is electrostatically driven. Phys Chem Chem Phys 2024; 26:23103-23115. [PMID: 39177151 DOI: 10.1039/d4cp02060j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
Gold nanoparticles (AuNP) are known to aggregate on the surface of lipid vesicles, yet the molecular mechanism behind this phenomenom remains unclear. In this work, we have investigated the binding behaviour of AuNPs, synthesized with pulsed laser ablation, to phospholipid vesicles under varying conditions of ionic strength (KCl concentration) and NP to vesicle ratios. Our observations reveal a strong influence of electrolyte concentration on AuNP aggregation mediated by vesicles. Notably, cluster formation is observed even at less than one AuNP per vesicle ratio at low enough ionic strengths. These results evidence a binding mechanism governed by electrostatic attraction with a distinct cooperative behaviour at very low salt concentrations, resulting in a significant increase in nanoparticle clustering. This behaviour is quantitatively analysed through a model that incorporates the Derjaguin-Landau-Verwey-Overbeek (DLVO) theory, considering the electrical double layer attraction between dissimilar, non-oppositely charged objects. This study not only provides insight into the fundamental understanding of nanoparticle-vesicle interactions but also suggests potential strategies for controlling nanoparticle assembly in biological and synthetic systems by tuning the ionic strength.
Collapse
Affiliation(s)
- Helena Mateos
- Dipartimento di Chimica and CSGI (Center for Colloid and Surface Science), Università degli Studi di Bari "Aldo Moro", via Orabona n. 4, 70125 Bari, Italy.
| | - Antonia Mallardi
- CNR-IPCF, Institute for Chemical-Physical Processes, c/o Chemistry Department, University of Bari, Via Orabona 4, 70125 Bari, Italy
| | - Miquel Oliver
- FI-TRACE Group, Department of Chemistry, University of the Balearic Islands, Carretera de Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain
| | - Marcella Dell'Aglio
- CNR-IFN (National Research Council - Institute for Photonics and Nanotechnologies), c/o Physics Department, University of Bari, Via Amendola 173, 70126 Bari, Italy
| | - Pamela Giannone
- Dipartimento di Chimica and CSGI (Center for Colloid and Surface Science), Università degli Studi di Bari "Aldo Moro", via Orabona n. 4, 70125 Bari, Italy.
| | - Gerardo Palazzo
- Dipartimento di Chimica and CSGI (Center for Colloid and Surface Science), Università degli Studi di Bari "Aldo Moro", via Orabona n. 4, 70125 Bari, Italy.
| |
Collapse
|
5
|
Poupardin R, Wolf M, Maeding N, Paniushkina L, Geissler S, Bergese P, Witwer KW, Schallmoser K, Fuhrmann G, Strunk D. Advances in Extracellular Vesicle Research Over the Past Decade: Source and Isolation Method are Connected with Cargo and Function. Adv Healthc Mater 2024; 13:e2303941. [PMID: 38270559 DOI: 10.1002/adhm.202303941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/23/2023] [Indexed: 01/26/2024]
Abstract
The evolution of extracellular vesicle (EV) research has introduced nanotechnology into biomedical cell communication science while recognizing what is formerly considered cell "dust" as constituting an entirely new universe of cell signaling particles. To display the global EV research landscape, a systematic review of 20 364 original research articles selected from all 40 684 EV-related records identified in PubMed 2013-2022 is performed. Machine-learning is used to categorize the high-dimensional data and further dissected significant associations between EV source, isolation method, cargo, and function. Unexpected correlations between these four categories indicate prevalent experimental strategies based on cargo connectivity with function of interest being associated with certain EV sources or isolation strategies. Conceptually relevant association of size-based EV isolation with protein cargo and uptake function will guide strategic conclusions enhancing future EV research and product development. Based on this study, an open-source database is built to facilitate further analysis with conventional or AI tools to identify additional causative associations of interest.
Collapse
Affiliation(s)
- Rodolphe Poupardin
- Cell Therapy Institute, Paracelsus Medical University, Salzburg, 5020, Austria
| | - Martin Wolf
- Cell Therapy Institute, Paracelsus Medical University, Salzburg, 5020, Austria
| | - Nicole Maeding
- Cell Therapy Institute, Paracelsus Medical University, Salzburg, 5020, Austria
| | - Liliia Paniushkina
- Cell Therapy Institute, Paracelsus Medical University, Salzburg, 5020, Austria
- Departments of Molecular and Comparative Pathobiology and Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Sven Geissler
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 10178, Berlin, Germany
| | - Paolo Bergese
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, 25121, Italy
- INSTM - National Interuniversity Consortium of Materials Science and Technology, Firenze, 50121, Italy
- National Center for Gene Therapy and Drugs based on RNA Technology - CN3, Padova, 35122, Italy
| | - Kenneth W Witwer
- Departments of Molecular and Comparative Pathobiology and Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Katharina Schallmoser
- Institute of Transfusion Medicine, Paracelsus Medical University, Salzburg, 5020, Austria
| | - Gregor Fuhrmann
- Department of Biology, Friedrich-Alexander-University Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Dirk Strunk
- Cell Therapy Institute, Paracelsus Medical University, Salzburg, 5020, Austria
- Institute of Transfusion Medicine, Paracelsus Medical University, Salzburg, 5020, Austria
| |
Collapse
|
6
|
Gorgzadeh A, Nazari A, Ali Ehsan Ismaeel A, Safarzadeh D, Hassan JAK, Mohammadzadehsaliani S, Kheradjoo H, Yasamineh P, Yasamineh S. A state-of-the-art review of the recent advances in exosome isolation and detection methods in viral infection. Virol J 2024; 21:34. [PMID: 38291452 PMCID: PMC10829349 DOI: 10.1186/s12985-024-02301-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 01/22/2024] [Indexed: 02/01/2024] Open
Abstract
Proteins, RNA, DNA, lipids, and carbohydrates are only some of the molecular components found in exosomes released by tumor cells. They play an essential role in healthy and diseased cells as messengers of short- and long-distance intercellular communication. However, since exosomes are released by every kind of cell and may be found in blood and other bodily fluids, they may one day serve as biomarkers for a wide range of disorders. In many pathological conditions, including cancer, inflammation, and infection, they play a role. It has been shown that the biogenesis of exosomes is analogous to that of viruses and that the exosomal cargo plays an essential role in the propagation, dissemination, and infection of several viruses. Bidirectional modulation of the immune response is achieved by the ability of exosomes associated with viruses to facilitate immunological escape and stimulate the body's antiviral immune response. Recently, exosomes have received a lot of interest due to their potential therapeutic use as biomarkers for viral infections such as human immunodeficiency virus (HIV), Hepatitis B virus (HBV), Hepatitis C virus (HCV), Epstein-Barr virus (EBV), and SARS-CoV-2. This article discusses the purification procedures and detection techniques for exosomes and examines the research on exosomes as a biomarker of viral infection.
Collapse
Affiliation(s)
| | - Ahmad Nazari
- Tehran University of Medical Sciences, Tehran, Iran
| | | | - Diba Safarzadeh
- Vocational School of Health Service, Near East University, Nicosia, Cyprus
| | - Jawad A K Hassan
- National University of Science and Technology, Nasiriyah, Dhi Qar, Iraq
| | | | | | - Pooneh Yasamineh
- Young Researchers and Elite Club, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Saman Yasamineh
- Young Researchers and Elite Club, Tabriz Branch, Islamic Azad University, Tabriz, Iran.
| |
Collapse
|
7
|
Cardellini J, Balestri A, Comparini L, Lonetti B, Brucale M, Valle F, Berti D, Montis C. Controlling plasmonic suprastructures through self-assembly of gold nanoparticles with hybrid copolymer-lipid vesicles. J Colloid Interface Sci 2024; 654:848-858. [PMID: 37898069 DOI: 10.1016/j.jcis.2023.10.082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/30/2023]
Abstract
Hybrid lipid membranes incorporating amphiphilic copolymers have gained significant attention due to their potential applications in various fields, including drug delivery and sensing. By combining the properties of copolymers and lipid membranes, such as enhanced chemical tunability and stability, environmental responsiveness, and multidomain nature, novel membrane architectures have been proposed. In this study, we investigated the potentialities of hybrid membranes made of two distinct components: the rigid fully saturated phospholipid 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) and the soft copolymer poly(butadiene-b-ethyleneoxide) (PBD-b-PEO). The objective was to explore the interaction of citrate-coated gold nanoparticles (AuNPs) and the hybrid membrane, aiming at constructing AuNPs-hybrid vesicles suprastructures with controlled and adjustable plasmonic properties. A series of experimental techniques were employed to investigate hybrid free-standing and supported membranes. The results revealed that the incorporation of the copolymer into the lipid membrane promotes AuNPs clustering, demonstrating a distinctive aggregative phenomenon of citrate-coated AuNPs on multidomain membranes. Importantly, we show that the size and morphology of AuNPs clusters can be precisely controlled in non-homogeneous membranes, enabling the formation of hybrid suprastructures with controlled patch properties. These results highlight the potential of lipid-copolymer hybrid membranes for designing functional materials with tailored plasmonic properties, with potential applications in nanomedicine and sensing.
Collapse
Affiliation(s)
- Jacopo Cardellini
- Department of Chemistry, University of Florence, and CSGI, Florence, Italy
| | - Arianna Balestri
- Department of Chemistry, University of Florence, and CSGI, Florence, Italy
| | - Luca Comparini
- Department of Chemistry, University of Florence, and CSGI, Florence, Italy
| | - Barbara Lonetti
- Laboratoire des IMRCP, Université de Toulouse, CNRS, Toulouse 31062, France
| | | | | | - Debora Berti
- Department of Chemistry, University of Florence, and CSGI, Florence, Italy
| | - Costanza Montis
- Department of Chemistry, University of Florence, and CSGI, Florence, Italy.
| |
Collapse
|
8
|
Hanai H, Hart DA, Jacob G, Shimomura K, Ando W, Yoshioka Y, Ochiya T, Nakagawa S, Nakamura M, Okada S, Nakamura N. Small extracellular vesicles derived from human adipose-derived mesenchymal stromal cells cultured in a new chemically-defined contaminate-free media exhibit enhanced biological and therapeutic effects on human chondrocytes in vitro and in a mouse osteoarthritis model. J Extracell Vesicles 2023; 12:e12337. [PMID: 37367299 PMCID: PMC10295161 DOI: 10.1002/jev2.12337] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 06/13/2023] [Indexed: 06/28/2023] Open
Abstract
Human small extracellular vesicles (sEVs) derived from adipose-derived mesenchymal stromal cells (ASC) have been reported to suppress the progression of osteoarthritis (OA) in animal studies and subsequently, translation of this potential to assess their clinical efficacy is anticipated. However, fabrication protocols for sEVs to eliminate potential contamination by culture medium-derived components need to be established prior to their clinical use. The purpose of the present studies was to elucidate the influence of medium-derived contaminants on the biological effects of sEVs, and to establish isolation methods for sEVs using a new clinical grade chemically-defined media (CDM). The quantity and purity of ASC-derived sEVs cultured in four different CDMs (CDM1, 2, 3 and 4) were evaluated. The concentrates of the four media incubated without cells were used as the background (BG) control for each set of sEVs. The biological effect of sEVs fabricated in the four different CDMs on normal human articular chondrocytes (hACs) were evaluated in vitro using a variety of methodological assessments. Finally, the sEVs with the highest purity were tested for their ability to suppress the progression of knee OA mouse model. Analysis of the BG controls revealed that CDM1-3 contained detectable particles, while there was no visible contamination of culture media-derived components detected with CDM4. Accordingly, the sEVs fabricated with CDM4 (CDM4-sEVs) exhibited the highest purity and yield. Notably, the CDM4-sEVs were the most efficient in promoting the cellular proliferation, migration, chondrogenic differentiation, and anti-apoptotic activity of hACs. Furthermore, CDM4-sEVs significantly suppressed the osteochondral degeneration in vivo model. Small EVs derived from ASCs cultured in a CDM without detectable contaminants demonstrated enhanced biological effects on hACs and the progression of OA. Thus, sEVs isolated with CDM4 most optimally meet the requirements of efficacy and safety for assessment in their future clinical applications.
Collapse
Affiliation(s)
- Hiroto Hanai
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - David A Hart
- Department of Surgery and the McCaig Institute for Bone & Joint Health, University of Calgary, Calgary, Canada
| | - George Jacob
- Department of Orthopaedics, VPS Lakeshore Hospital, Kerala, India
| | - Kazunori Shimomura
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Wataru Ando
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Orthopaedic Surgery, Kansai Rosai Hospital, Hyogo, Japan
| | - Yusuke Yoshioka
- Department of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Takahiro Ochiya
- Department of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Shinicihi Nakagawa
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masato Nakamura
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Seiji Okada
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Norimasa Nakamura
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
- Institute for Medical Science in Sports, Osaka Health Science University, Osaka, Japan
- Global Center for Medical Engineering and Informatics, Osaka University, Osaka, Japan
| |
Collapse
|
9
|
Brezgin S, Parodi A, Kostyusheva A, Ponomareva N, Lukashev A, Sokolova D, Pokrovsky VS, Slatinskaya O, Maksimov G, Zamyatnin AA, Chulanov V, Kostyushev D. Technological aspects of manufacturing and analytical control of biological nanoparticles. Biotechnol Adv 2023; 64:108122. [PMID: 36813011 DOI: 10.1016/j.biotechadv.2023.108122] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 01/19/2023] [Accepted: 02/09/2023] [Indexed: 02/22/2023]
Abstract
Extracellular vesicles (EVs) are cell-derived biological nanoparticles that gained great interest for drug delivery. EVs have numerous advantages compared to synthetic nanoparticles, such as ideal biocompatibility, safety, ability to cross biological barriers and surface modification via genetic or chemical methods. On the other hand, the translation and the study of these carriers resulted difficult, mostly because of significant issues in up-scaling, synthesis and impractical methods of quality control. However, current manufacturing advances enable EV packaging with any therapeutic cargo, including DNA, RNA (for RNA vaccines and RNA therapeutics), proteins, peptides, RNA-protein complexes (including gene-editing complexes) and small molecules drugs. To date, an array of new and upgraded technologies have been introduced, substantially improving EV production, isolation, characterization and standardization. The used-to-be "gold standards" of EV manufacturing are now outdated, and the state-of-art requires extensive revision. This review re-evaluates the pipeline for EV industrial production and provides a critical overview of the modern technologies required for their synthesis and characterization.
Collapse
Affiliation(s)
- Sergey Brezgin
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow 119048, Russia; Sirius University of Science and Technology, Sochi 354340, Russia
| | | | - Anastasiya Kostyusheva
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow 119048, Russia
| | - Natalia Ponomareva
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow 119048, Russia; Sirius University of Science and Technology, Sochi 354340, Russia
| | - Alexander Lukashev
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow 119048, Russia
| | - Darina Sokolova
- Sirius University of Science and Technology, Sochi 354340, Russia; Blokhin National Medical Research Center of Oncology, Moscow 115478, Russia; People's Friendship University, Moscow 117198, Russia
| | - Vadim S Pokrovsky
- Sirius University of Science and Technology, Sochi 354340, Russia; Blokhin National Medical Research Center of Oncology, Moscow 115478, Russia; People's Friendship University, Moscow 117198, Russia
| | - Olga Slatinskaya
- Lomonosov Moscow State University, Faculty of Biology, Moscow 119991, Russia
| | - Georgy Maksimov
- Lomonosov Moscow State University, Faculty of Biology, Moscow 119991, Russia
| | - Andrey A Zamyatnin
- Sirius University of Science and Technology, Sochi 354340, Russia; Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia; Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia; Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7X, UK
| | - Vladimir Chulanov
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow 119048, Russia; Sirius University of Science and Technology, Sochi 354340, Russia; Department of Infectious Diseases, Sechenov University, Moscow 119048, Russia; National Medical Research Center for Tuberculosis and Infectious Diseases, Moscow 127994, Russia
| | - Dmitry Kostyushev
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow 119048, Russia; Sirius University of Science and Technology, Sochi 354340, Russia.
| |
Collapse
|
10
|
Farzanehpour M, Miri A, Ghorbani Alvanegh A, Esmaeili Gouvarchinghaleh H. Viral Vectors, Exosomes, and Vexosomes: Potential Armamentarium for Delivering CRISPR/Cas to Cancer Cells. Biochem Pharmacol 2023; 212:115555. [PMID: 37075815 DOI: 10.1016/j.bcp.2023.115555] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 04/10/2023] [Accepted: 04/11/2023] [Indexed: 04/21/2023]
Abstract
The underlying cause of cancer is genetic disruption, so gene editing technologies, particularly CRISPR/Cas systems can be used to go against cancer. The field of gene therapy has undergone many transitions over its 40-year history. Despite its many successes, it has also suffered many failures in the battle against malignancies, causing really adverse effects instead of therapeutic outcomes. At the tip of this double-edged sword are viral and non-viral-based vectors, which have profoundly transformed the way scientists and clinicians develop therapeutic platforms. Viruses such as lentivirus, adenovirus, and adeno-associated viruses are the most common viral vectors used for delivering the CRISPR/Cas system into human cells. In addition, among non-viral vectors, exosomes, especially tumor-derived exosomes (TDEs), have proven to be quite effective at delivering this gene editing tool. The combined use of viral vectors and exosomes, called vexosomes, seems to be a solution to overcoming the obstacles of both delivery systems.
Collapse
Affiliation(s)
- Mahdieh Farzanehpour
- Applied Virology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ali Miri
- Human Genetics Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | | |
Collapse
|
11
|
Cardellini J, Ridolfi A, Donati M, Giampietro V, Severi M, Brucale M, Valle F, Bergese P, Montis C, Caselli L, Berti D. Probing the coverage of nanoparticles by biomimetic membranes through nanoplasmonics. J Colloid Interface Sci 2023; 640:100-109. [PMID: 36842416 DOI: 10.1016/j.jcis.2023.02.073] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/29/2023] [Accepted: 02/13/2023] [Indexed: 02/24/2023]
Abstract
Although promising for biomedicine, the clinical translation of inorganic nanoparticles (NPs) is limited by low biocompatibility and stability in biological fluids. A common strategy to circumvent this drawback consists in disguising the active inorganic core with a lipid bilayer coating, reminiscent of the structure of the cell membrane to redefine the chemical and biological identity of NPs. While recent reports introduced membrane-coating procedures for NPs, a robust and accessible method to quantify the integrity of the bilayer coverage is not yet available. To fill this gap, we prepared SiO2 nanoparticles (SiO2NPs) with different membrane coverage degrees and monitored their interaction with AuNPs by combining microscopic, scattering, and optical techniques. The membrane-coating on SiO2NPs induces spontaneous clustering of AuNPs, whose extent depends on the coating integrity. Remarkably, we discovered a linear correlation between the membrane coverage and a spectral descriptor for the AuNPs' plasmonic resonance, spanning a wide range of coating yields. These results provide a fast and cost-effective assay to monitor the compatibilization of NPs with biological environments, essential for bench tests and scale-up. In addition, we introduce a robust and scalable method to prepare SiO2NPs/AuNPs hybrids through spontaneous self-assembly, with a high-fidelity structural control mediated by a lipid bilayer.
Collapse
Affiliation(s)
- Jacopo Cardellini
- Department of Chemistry "Ugo Schiff", University of Florence, Florence, Italy; CSGI, Consorzio Sistemi a Grande Interfase, University of Florence, Sesto Fiorentino, Italy
| | - Andrea Ridolfi
- Department of Chemistry "Ugo Schiff", University of Florence, Florence, Italy; CSGI, Consorzio Sistemi a Grande Interfase, University of Florence, Sesto Fiorentino, Italy; Istituto per lo Studio dei Materiali Nanostrutturati, Consiglio Nazionale delle Ricerche, 40129 Bologna, Italy; Department of Physics and Astronomy and LaserLaB Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands(1)
| | - Melissa Donati
- Department of Chemistry "Ugo Schiff", University of Florence, Florence, Italy
| | | | - Mirko Severi
- Department of Chemistry "Ugo Schiff", University of Florence, Florence, Italy
| | - Marco Brucale
- CSGI, Consorzio Sistemi a Grande Interfase, University of Florence, Sesto Fiorentino, Italy; Istituto per lo Studio dei Materiali Nanostrutturati, Consiglio Nazionale delle Ricerche, 40129 Bologna, Italy
| | - Francesco Valle
- CSGI, Consorzio Sistemi a Grande Interfase, University of Florence, Sesto Fiorentino, Italy; Istituto per lo Studio dei Materiali Nanostrutturati, Consiglio Nazionale delle Ricerche, 40129 Bologna, Italy
| | - Paolo Bergese
- CSGI, Consorzio Sistemi a Grande Interfase, University of Florence, Sesto Fiorentino, Italy; Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; Consorzio Interuniversitario Nazionale per la Scienza e la Tecnologia dei Materiali, Florence, Italy
| | - Costanza Montis
- Department of Chemistry "Ugo Schiff", University of Florence, Florence, Italy; CSGI, Consorzio Sistemi a Grande Interfase, University of Florence, Sesto Fiorentino, Italy
| | - Lucrezia Caselli
- Department of Chemistry "Ugo Schiff", University of Florence, Florence, Italy; CSGI, Consorzio Sistemi a Grande Interfase, University of Florence, Sesto Fiorentino, Italy; Department of Physical Chemistry 1, University of Lund, SE-22100 Lund, Sweden(1).
| | - Debora Berti
- Department of Chemistry "Ugo Schiff", University of Florence, Florence, Italy; CSGI, Consorzio Sistemi a Grande Interfase, University of Florence, Sesto Fiorentino, Italy.
| |
Collapse
|
12
|
Mangolini V, Gualerzi A, Picciolini S, Rodà F, Del Prete A, Forleo L, Rossetto RA, Bedoni M. Biochemical Characterization of Human Salivary Extracellular Vesicles as a Valuable Source of Biomarkers. BIOLOGY 2023; 12:227. [PMID: 36829504 PMCID: PMC9953587 DOI: 10.3390/biology12020227] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 01/27/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023]
Abstract
Extracellular vesicles (EVs) are natural nanoparticles secreted under physiological and pathological conditions. Thanks to their diagnostic potential, EVs are increasingly being studied as biomarkers of a variety of diseases, including neurological disorders. To date, most studies on EV biomarkers use blood as the source, despite different disadvantages that may cause an impure isolation of the EVs. In the present article, we propose the use of saliva as a valuable source of EVs that could be studied as biomarkers in an easily accessible biofluid. Using a comparable protocol for the isolation of EVs from both liquid biopsies, salivary EVs showed greater purity in terms of co-isolates (evaluated by nanoparticle tracking analysis and Conan test). In addition, Raman spectroscopy was used for the identification of the overall biochemical composition of EVs coming from the two different biofluids. Even considering the limited amount of EVs that can be isolated from saliva, the use of Raman spectroscopy was not hampered, and it was able to provide a comprehensive characterization of EVs in a high throughput and repeatable manner. Raman spectroscopy can thus represent a turning point in the application of salivary EVs in clinics, taking advantage of the simple method of collection of the liquid biopsy and of the quick, sensitive and label-free biophotonics-based approach.
Collapse
Affiliation(s)
- Valentina Mangolini
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, 20148 Milano, Italy
- Dipartimento di Medicina Molecolare e Traslazionale, Università degli Studi di Brescia, 25122 Brescia, Italy
| | - Alice Gualerzi
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, 20148 Milano, Italy
| | | | - Francesca Rodà
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, 20148 Milano, Italy
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, 42100 Modena, Italy
| | | | - Luana Forleo
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, 20148 Milano, Italy
| | | | - Marzia Bedoni
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, 20148 Milano, Italy
| |
Collapse
|
13
|
Practical Considerations for Translating Mesenchymal Stromal Cell-Derived Extracellular Vesicles from Bench to Bed. Pharmaceutics 2022; 14:pharmaceutics14081684. [PMID: 36015310 PMCID: PMC9414392 DOI: 10.3390/pharmaceutics14081684] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 11/16/2022] Open
Abstract
Extracellular vesicles (EVs) derived from mesenchymal stromal cells (MSCs) have shown potential for the treatment of tendon and ligament injuries. This approach can eliminate the need to transplant live cells to the human body, thereby reducing issues related to the maintenance of cell viability and stability and potential erroneous differentiation of transplanted cells to bone or tumor. Despite these advantages, there are practical issues that need to be considered for successful clinical application of MSC-EV-based products in the treatment of tendon and ligament injuries. This review aims to discuss the general and tissue-specific considerations for manufacturing MSC-EVs for clinical translation. Specifically, we will discuss Good Manufacturing Practice (GMP)-compliant manufacturing and quality control (parent cell source, culture conditions, concentration method, quantity, identity, purity and impurities, sterility, potency, reproducibility, storage and formulation), as well as safety and efficacy issues. Special considerations for applying MSC-EVs, such as their compatibility with arthroscopy for the treatment of tendon and ligament injuries, are also highlighted.
Collapse
|
14
|
Ceccarelli L, Marchetti L, Rizzo M, Moscardini A, Cappello V, Da Pozzo E, Romano M, Giacomelli C, Bergese P, Martini C. Human Microglia Extracellular Vesicles Derived from Different Microglia Cell Lines: Similarities and Differences. ACS OMEGA 2022; 7:23127-23137. [PMID: 35847267 PMCID: PMC9280972 DOI: 10.1021/acsomega.2c00816] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Microglial cells are a component of the innate immune system in the brain that support cell-to-cell communication via secreted molecules and extracellular vesicles (EVs). EVs can be divided into two major populations: large (LEVs) and small (SEVs) EVs, carrying different mediators, such as proteins, lipids, and miRNAs. The microglia EVs cargo crucially reflects the status of parental cells and can lead to both beneficial and detrimental effects in many physiopathological states. Herein, a workflow for the extraction and characterization of SEVs and LEVs from human C20 and HMC3 microglia cell lines derived, respectively, from adult and embryonic microglia is reported. EVs were gathered from the culture media of the two cell lines by sequential ultracentrifugation steps and their biochemical and biophysical properties were analyzed by Western blot, transmission electron microscopy, and dynamic light scattering. Although the C20- and HMC3-derived EVs shared several common features, C20-derived EVs were slightly lower in number and more polydispersed. Interestingly, C20- but not HMC3-SEVs were able to interfere with the proliferation of U87 glioblastoma cells. This correlated with the different relative levels of eight miRNAs involved in neuroinflammation and tumor progression in the C20- and HMC3-derived EVs, which in turn reflected a different basal activation state of the two cell types. Our data fill a gap in the community of microglia EVs, in which the preparations from human cells have been poorly characterized so far. Furthermore, these results shed light on both the differences and similarities of EVs extracted from different human microglia cell models, underlining the need to better characterize the features and biological effects of EVs for therein useful and correct application.
Collapse
Affiliation(s)
- Lorenzo Ceccarelli
- Department
of Pharmacy, University of Pisa, Pisa 56126, Italy
- Department
of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Laura Marchetti
- Department
of Pharmacy, University of Pisa, Pisa 56126, Italy
| | - Milena Rizzo
- Institute
of Clinical Physiology (IFC), CNR, Pisa 56124, Italy
| | - Aldo Moscardini
- SNS
(Scuola Normale Superiore, NEST laboratories), Pisa 56127, Italy
| | - Valentina Cappello
- Center
for Materials Interfaces, Electron Crystallography, Istituto Italiano di Tecnologia, Pontedera 56025, Italy
| | | | - Miriam Romano
- Department
of Molecular and Translational Medicine, University of Brescia, Brescia 25121, Italy
- Center
for Colloid and Surface Science (CSGI), Firenze 50019, Italy
| | | | - Paolo Bergese
- Department
of Molecular and Translational Medicine, University of Brescia, Brescia 25121, Italy
- Center
for Colloid and Surface Science (CSGI), Firenze 50019, Italy
- Institute
for Research and Biomedical Innovation- IRIB, Consiglio Nazionale delle Ricerche—CNR, Palermo 900146, Italy
| | - Claudia Martini
- Department
of Pharmacy, University of Pisa, Pisa 56126, Italy
| |
Collapse
|
15
|
Nguyen PH, Le AH, Pek JSQ, Pham TT, Jayasinghe MK, Do DV, Phung CD, Le MT. Extracellular vesicles and lipoproteins - Smart messengers of blood cells in the circulation. JOURNAL OF EXTRACELLULAR BIOLOGY 2022; 1:e49. [PMID: 38938581 PMCID: PMC11080875 DOI: 10.1002/jex2.49] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 06/12/2022] [Accepted: 06/19/2022] [Indexed: 06/29/2024]
Abstract
Blood cell-derived extracellular vesicles (BCEVs) and lipoproteins are the major circulating nanoparticles in blood that play an important role in intercellular communication. They have attracted significant interest for clinical applications, given their endogenous characteristics which make them stable, biocompatible, well tolerated, and capable of permeating biological barriers efficiently. In this review, we describe the basic characteristics of BCEVs and lipoproteins and summarize their implications in both physiological and pathological processes. We also outline well accepted workflows for the isolation and characterization of these circulating nanoparticles. Importantly, we highlight the latest progress and challenges associated with the use of circulating nanoparticles as diagnostic biomarkers and therapeutic interventions in multiple diseases. We spotlight novel engineering approaches and designs to facilitate the development of these nanoparticles by enhancing their stability, targeting capability, and delivery efficiency. Therefore, the present work provides a comprehensive overview of composition, biogenesis, functions, and clinical translation of circulating nanoparticles from the bench to the bedside.
Collapse
Affiliation(s)
- Phuong H.D. Nguyen
- Department of Pharmacology and Institute for Digital MedicineYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Anh Hong Le
- Department of Pharmacology and Institute for Digital MedicineYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Jonetta Shi Qi Pek
- Department of Pharmacology and Institute for Digital MedicineYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Thach Tuan Pham
- Department of Pharmacology and Institute for Digital MedicineYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Migara Kavishka Jayasinghe
- Department of Pharmacology and Institute for Digital MedicineYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Immunology ProgrammeCancer Programme and Nanomedicine Translational ProgrammeYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of SurgeryYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Dang Vinh Do
- Department of Pharmacology and Institute for Digital MedicineYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Cao Dai Phung
- Department of Pharmacology and Institute for Digital MedicineYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Minh T.N. Le
- Department of Pharmacology and Institute for Digital MedicineYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Immunology ProgrammeCancer Programme and Nanomedicine Translational ProgrammeYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of SurgeryYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| |
Collapse
|
16
|
Borup A, Boysen AT, Ridolfi A, Brucale M, Valle F, Paolini L, Bergese P, Nejsum P. Comparison of separation methods for immunomodulatory extracellular vesicles from helminths. JOURNAL OF EXTRACELLULAR BIOLOGY 2022; 1:e41. [PMID: 38939526 PMCID: PMC11080882 DOI: 10.1002/jex2.41] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/03/2022] [Accepted: 04/04/2022] [Indexed: 06/29/2024]
Abstract
Helminths survive within their host by secreting immunomodulatory compounds, which hold therapeutic potential for inflammatory conditions. Helminth-derived extracellular vesicles (EVs) are one such component proposed to possess immunomodulatory activities. Due to the recent discovery of helminth EVs, standardised protocols for EV separation are lacking. Excretory/secretory products of the porcine helminth, Ascaris suum, were used to compare three EV separation methods: Size exclusion chromatography (SEC), ultracentrifugation (UC) and a combination of the two. Their performance was evaluated by EV yield, sample purity and the ability of EVs to suppress lipopolysaccharide (LPS)-induced inflammation in vitro. We found that all three separation methods successfully separated helminth EVs with a similar EV yield. Functional studies showed that EVs from all three methods reduced LPS-induced levels of tumour necrosis factor (TNF-α) in a dose-dependent manner. Overall, the three separation methods showed similar performance, however, the combination of UC+SEC presented with slightly higher purity than either method alone.
Collapse
Affiliation(s)
- Anne Borup
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | | | - Andrea Ridolfi
- Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande Interfase (CSGI)University of FlorenceFlorenceItaly
- Consiglio Nazionale delle Ricerche (CNR)Istituto per lo Studio dei Materiali Nanostrutturati (ISMN)University of BolognaBolognaItaly
- Department of ChemistryUniversity of FlorenceFlorenceItaly
| | - Marco Brucale
- Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande Interfase (CSGI)University of FlorenceFlorenceItaly
- Consiglio Nazionale delle Ricerche (CNR)Istituto per lo Studio dei Materiali Nanostrutturati (ISMN)University of BolognaBolognaItaly
| | - Francesco Valle
- Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande Interfase (CSGI)University of FlorenceFlorenceItaly
- Consiglio Nazionale delle Ricerche (CNR)Istituto per lo Studio dei Materiali Nanostrutturati (ISMN)University of BolognaBolognaItaly
| | - Lucia Paolini
- Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande Interfase (CSGI)University of FlorenceFlorenceItaly
- Department of Molecular and Translational MedicineUniversity of BresciaBresciaItaly
| | - Paolo Bergese
- Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande Interfase (CSGI)University of FlorenceFlorenceItaly
- Department of Molecular and Translational MedicineUniversity of BresciaBresciaItaly
- Consiglio Nazionale delle Ricerche (CNR)Institute for Research and Biomedical Innovation (IRIB)University of PalermoPalermoItaly
| | - Peter Nejsum
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
| |
Collapse
|
17
|
Baidya S, Hassan AM. SARS-CoV-2 Detection using Colorimetric Plasmonic Sensors: A Proof-of-Concept Computational Study. IEEE Trans Nanobioscience 2022; 22:71-77. [PMID: 35235520 PMCID: PMC9983695 DOI: 10.1109/tnb.2022.3156077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Traditional molecular techniques for SARS-CoV-2 viral detection are time-consuming and can exhibit a high probability of false negatives. In this work, we present a computational study of SARS-CoV-2 detection using plasmonic gold nanoparticles. The resonance wavelength of a SARS-CoV-2 virus was recently estimated to be in the near-infrared region. By engineering gold nanospheres to specifically bind with the outer surface of the SARS-CoV-2 virus, the resonance frequency can be shifted to the visible range (380 nm - 700 nm). Moreover, we show that broadband absorption will emerge in the visible spectrum when the virus is partially covered with gold nanoparticles at a specific coverage percentage. This broadband absorption can be used to guide the development of an efficient and accurate colorimetric plasmon sensor for COVID-19 detection. Our observation also suggests that this technique is unaffected by the number of protein spikes present on the virus outer surface, hence can pave a potential path for a label-free COVID-19 diagnostic tool independent of the number of protein spikes.
Collapse
Affiliation(s)
- Somen Baidya
- Department of Computer Science and Electrical Engineering, University of Missouri–Kansas City, Kansas City, MO, USA
| | - Ahmed M. Hassan
- Department of Computer Science and Electrical Engineering, University of Missouri–Kansas City, Kansas City, MO, USA
| |
Collapse
|
18
|
Serratì S, Palazzo A, Lapenna A, Mateos H, Mallardi A, Marsano RM, Quarta A, Del Rosso M, Azzariti A. Salting-Out Approach Is Worthy of Comparison with Ultracentrifugation for Extracellular Vesicle Isolation from Tumor and Healthy Models. Biomolecules 2021; 11:biom11121857. [PMID: 34944501 PMCID: PMC8699204 DOI: 10.3390/biom11121857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/29/2021] [Accepted: 12/08/2021] [Indexed: 11/20/2022] Open
Abstract
The role of extracellular vesicles (EVs) has been completely re-evaluated in the recent decades, and EVs are currently considered to be among the main players in intercellular communication. Beyond their functional aspects, there is strong interest in the development of faster and less expensive isolation protocols that are as reliable for post-isolation characterisations as already-established methods. Therefore, the identification of easy and accessible EV isolation techniques with a low price/performance ratio is of paramount importance. We isolated EVs from a wide spectrum of samples of biological and clinical interest by choosing two isolation techniques, based on their wide use and affordability: ultracentrifugation and salting-out. We collected EVs from human cancer and healthy cell culture media, yeast, bacteria and Drosophila culture media and human fluids (plasma, urine and saliva). The size distribution and concentration of EVs were measured by nanoparticle tracking analysis and dynamic light scattering, and protein depletion was measured by a colorimetric nanoplasmonic assay. Finally, the EVs were characterised by flow cytometry. Our results showed that the salting-out method had a good efficiency in EV separation and was more efficient in protein depletion than ultracentrifugation. Thus, salting-out may represent a good alternative to ultracentrifugation.
Collapse
Affiliation(s)
- Simona Serratì
- Nanotecnology Laboratory, IRCCS Istituto Tumori Giovanni Paolo II, Viale Orazio Flacco 65, 70124 Bari, Italy;
- Correspondence: (S.S.); (A.P.)
| | - Antonio Palazzo
- Nanotecnology Laboratory, IRCCS Istituto Tumori Giovanni Paolo II, Viale Orazio Flacco 65, 70124 Bari, Italy;
- Correspondence: (S.S.); (A.P.)
| | - Annamaria Lapenna
- Department of Chemistry, University of Bari and CSGI (Center for Colloid and Surface Science), Via Orabona 4, 70125 Bari, Italy; (A.L.); (H.M.)
| | - Helena Mateos
- Department of Chemistry, University of Bari and CSGI (Center for Colloid and Surface Science), Via Orabona 4, 70125 Bari, Italy; (A.L.); (H.M.)
| | - Antonia Mallardi
- Istituto per i Processi Chimico Fisici, National Research Council (IPCF-CNR), c/o ChemistryDepartment, Via Orabona 4, 70125 Bari, Italy;
| | | | - Alessandra Quarta
- CNR NANOTEC—Istituto di Nanotecnologia, National Research Council (CNR), Via Monteroni, 73100 Lecce, Italy;
| | - Mario Del Rosso
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale G.B. Morgagni 50, 50134 Florence, Italy;
| | - Amalia Azzariti
- Nanotecnology Laboratory, IRCCS Istituto Tumori Giovanni Paolo II, Viale Orazio Flacco 65, 70124 Bari, Italy;
- Laboratory of Experimental Pharmacology, IRCCS Istituto Tumori Giovanni Paolo II, Viale Orazio Flacco 65, 70124 Bari, Italy
| |
Collapse
|
19
|
Ma X, Hao Y, Liu L. Progress in Nanomaterials-Based Optical and Electrochemical Methods for the Assays of Exosomes. Int J Nanomedicine 2021; 16:7575-7608. [PMID: 34803380 PMCID: PMC8599324 DOI: 10.2147/ijn.s333969] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 10/28/2021] [Indexed: 12/11/2022] Open
Abstract
Exosomes with diameters of 30-150 nm are small membrane-bound vesicles secreted by a variety of cells. They play an important role in many biological processes, such as tumor-related immune response and intercellular signal transduction. Exosomes have been considered as emerging and noninvasive biomarkers for cancer diagnosis. Recently, a large number of optical and electrochemical biosensors have been proposed for sensitive detection of exosomes. To meet the increasing demands for ultrasensitive detection, nanomaterials have been integrated with various techniques as powerful components. Because of their intrinsic merits of biological compatibility, excellent physicochemical features and unique catalytic ability, nanomaterials have significantly improved the analytical performances of exosome biosensors. In this review, we summarized the recent progress in nanomaterials-based biosensors for the detection of cancer-derived exosomes, including fluorescence, colorimetry, surface plasmon resonance spectroscopy, surface enhanced Raman scattering spectroscopy, electrochemistry, electrochemiluminescence and so on.
Collapse
Affiliation(s)
- Xiaohua Ma
- Henan Key Laboratory of Biomolecular Recognition and Sensing, Shangqiu Normal University, Shangqiu, Henan, 476000, People’s Republic of China
| | - Yuanqiang Hao
- Henan Key Laboratory of Biomolecular Recognition and Sensing, Shangqiu Normal University, Shangqiu, Henan, 476000, People’s Republic of China
| | - Lin Liu
- Henan Key Laboratory of Biomolecular Recognition and Sensing, Shangqiu Normal University, Shangqiu, Henan, 476000, People’s Republic of China
- College of Chemistry and Chemical Engineering, Anyang Normal University, Anyang, Henan, 455000, People’s Republic of China
| |
Collapse
|
20
|
Elgamal S, Cocucci E, Sass EJ, Mo XM, Blissett AR, Calomeni EP, Rogers KA, Woyach JA, Bhat SA, Muthusamy N, Johnson AJ, Larkin KT, Byrd JC. Optimizing extracellular vesicles' isolation from chronic lymphocytic leukemia patient plasma and cell line supernatant. JCI Insight 2021; 6:e137937. [PMID: 34369387 PMCID: PMC8410027 DOI: 10.1172/jci.insight.137937] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
In chronic lymphocytic leukemia (CLL) and very likely all cancer types, extracellular vesicles (EVs) are a common mechanism by which intercellular messages are communicated between normal, diseased, and transformed cells. Studies of EVs in CLL and other cancers have great variability and often lack reproducibility. For CLL patient plasma and cell lines, we sought to characterize current approaches used in isolating EV products and understand whether cell culture-conditioned media or complex biological fluids confound results. Utilizing nanoparticle tracking analysis, protein quantification, and electron microscopy, we show that ultracentrifugation with an OptiPrep cushion can effectively minimize contaminants from starting materials including plasma and conditioned media of CLL cell lines grown in EV-depleted complete RPMI media but not grown in the serum-free media AIM V commonly used in CLL experimental work. Moreover, we confirm the benefit of including 25 mM trehalose in PBS during EV isolation steps to reduce EV aggregation, to preserve function for downstream applications and characterization. Furthermore, we report the highest particles/μg EVs were obtained from our CLL cell lines utilizing the CELLine bioreactor flask. Finally, we optimized a proliferation assay that offers a functional evaluation of our EVs with minimal sample requirements.
Collapse
Affiliation(s)
- Sara Elgamal
- Division of Hematology, Department of Internal Medicine, College of Medicine.,Comprehensive Cancer Center
| | - Emanuele Cocucci
- Comprehensive Cancer Center.,Division of Pharmaceutics, College of Pharmacy
| | - Ellen J Sass
- Division of Hematology, Department of Internal Medicine, College of Medicine.,Comprehensive Cancer Center
| | - Xiaokui M Mo
- Comprehensive Cancer Center.,Department of Biomedical Informatics, College of Medicine
| | | | | | - Kerry A Rogers
- Division of Hematology, Department of Internal Medicine, College of Medicine.,Comprehensive Cancer Center
| | - Jennifer A Woyach
- Division of Hematology, Department of Internal Medicine, College of Medicine.,Comprehensive Cancer Center
| | - Seema A Bhat
- Division of Hematology, Department of Internal Medicine, College of Medicine.,Comprehensive Cancer Center
| | - Natarajan Muthusamy
- Division of Hematology, Department of Internal Medicine, College of Medicine.,Comprehensive Cancer Center.,College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Amy J Johnson
- Division of Hematology, Department of Internal Medicine, College of Medicine.,Comprehensive Cancer Center.,Division of Pharmaceutics, College of Pharmacy
| | - Karilyn T Larkin
- Division of Hematology, Department of Internal Medicine, College of Medicine.,Comprehensive Cancer Center
| | - John C Byrd
- Division of Hematology, Department of Internal Medicine, College of Medicine.,Comprehensive Cancer Center.,Division of Pharmaceutics, College of Pharmacy.,College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
21
|
Trapani M, Scala A, Mineo PG, Pistone A, Díaz-Moscoso A, Fragoso A, Monsù Scolaro L, Mazzaglia A. Thiolated amphiphilic β-cyclodextrin-decorated gold colloids: Synthesis, supramolecular nanoassemblies and controlled release of dopamine. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2021.116880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
22
|
Caselli L, Ridolfi A, Cardellini J, Sharpnack L, Paolini L, Brucale M, Valle F, Montis C, Bergese P, Berti D. A plasmon-based nanoruler to probe the mechanical properties of synthetic and biogenic nanosized lipid vesicles. NANOSCALE HORIZONS 2021; 6:543-550. [PMID: 33870976 DOI: 10.1039/d1nh00012h] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Nanosized lipid vesicles are ubiquitous in living systems (e.g. cellular compartments or extracellular vesicles, EVs) and in formulations for nanomedicine (e.g. liposomes for RNA vaccine formulations). The mechanical properties of such vesicles are crucial in several physicochemical and biological processes, ranging from cellular uptake to stability in aerosols. However, their accurate determination remains challenging and requires sophisticated instruments and data analysis. Here we report the first evidence that the surface plasmon resonance (SPR) of citrated gold nanoparticles (AuNPs) adsorbed on synthetic vesicles is finely sensitive to the vesicles' mechanical properties. We then leverage this finding to show that the SPR tracking provides quantitative access to the stiffness of vesicles of synthetic and natural origin, such as EVs. The demonstration of this plasmon-based "stiffness nanoruler" paves the way for developing a facile, cost-effective and high-throughput method to assay the mechanical properties of dispersions of vesicles of nanometric size and unknown composition at a collective level.
Collapse
Affiliation(s)
- Lucrezia Caselli
- Department of Chemistry, University of Florence, Via della Lastruccia 3, Sesto Fiorentino, Florence 50019, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Sun Z, Yang J, Li H, Wang C, Fletcher C, Li J, Zhan Y, Du L, Wang F, Jiang Y. Progress in the research of nanomaterial-based exosome bioanalysis and exosome-based nanomaterials tumor therapy. Biomaterials 2021; 274:120873. [PMID: 33989972 DOI: 10.1016/j.biomaterials.2021.120873] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 04/13/2021] [Accepted: 05/02/2021] [Indexed: 12/18/2022]
Abstract
Exosomes and their internal components have been proven to play critical roles in cell-cell interactions and intrinsic cellular regulations, showing promising prospects in both biomedical and clinical fields. Although conventional methods have so far been utilized to great effect, accurate bioanalysis remains a major challenge. In recent years, the fast-paced development of nanomaterials with unique physiochemical properties has led to a boom in the potential bioapplications of such materials. In particular, the application of nanomaterials in exosome bioanalysis provides a great opportunity to overcome the current challenges and limitations of conventional methods. A timely review of the research progress in this field is thus of great significance to the continued development of new methods. This review outlines the properties and potential uses of exosomes, and discusses the conventional methods currently used for their analysis. We then focus on exploring the current state of the art regarding the use of nanomaterials for the isolation, detection and even the subsequent profiling of exosomes. The main methods are based on principles including fluorescence, surface-enhanced Raman spectroscopy, colorimetry, electrochemistry, and surface plasmon resonance. Additionally, research on exosome-based nanomaterials tumor therapy is also promising from a clinical perspective, so the research progress in this branch is also summarized. Finally, we look at ways in which the field might develop in the future.
Collapse
Affiliation(s)
- Zhiwei Sun
- Key Laboratory for Liquid-Solid Structural Evolution and Processing of Materials, Ministry of Education, Shandong University, Jinan, China
| | - Jingjing Yang
- Key Laboratory for Liquid-Solid Structural Evolution and Processing of Materials, Ministry of Education, Shandong University, Jinan, China
| | - Hui Li
- Key Laboratory for Liquid-Solid Structural Evolution and Processing of Materials, Ministry of Education, Shandong University, Jinan, China
| | - Chuanxin Wang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, China; Tumor Marker Detection Engineering Technology Research Center of Shandong Province, Jinan, China; Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, China; Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, China
| | - Cameron Fletcher
- School of Chemical Engineering, University of New South Wales, Sydney, Australia
| | - Juan Li
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, China; Tumor Marker Detection Engineering Technology Research Center of Shandong Province, Jinan, China; Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, China; Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, China
| | - Yao Zhan
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, China; Tumor Marker Detection Engineering Technology Research Center of Shandong Province, Jinan, China; Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, China; Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, China
| | - Lutao Du
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, China; Tumor Marker Detection Engineering Technology Research Center of Shandong Province, Jinan, China; Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, China; Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, China.
| | - Fenglong Wang
- Key Laboratory for Liquid-Solid Structural Evolution and Processing of Materials, Ministry of Education, Shandong University, Jinan, China.
| | - Yanyan Jiang
- Key Laboratory for Liquid-Solid Structural Evolution and Processing of Materials, Ministry of Education, Shandong University, Jinan, China.
| |
Collapse
|
24
|
Zivko C, Fuhrmann G, Luciani P. Liver-derived extracellular vesicles: A cell by cell overview to isolation and characterization practices. Biochim Biophys Acta Gen Subj 2021; 1865:129559. [DOI: 10.1016/j.bbagen.2020.129559] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/16/2020] [Accepted: 02/11/2020] [Indexed: 02/08/2023]
|
25
|
Fang X, Chen C, Liu B, Ma Z, Hu F, Li H, Gu H, Xu H. A magnetic bead-mediated selective adsorption strategy for extracellular vesicle separation and purification. Acta Biomater 2021; 124:336-347. [PMID: 33578055 DOI: 10.1016/j.actbio.2021.02.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 01/19/2021] [Accepted: 02/02/2021] [Indexed: 11/20/2022]
Abstract
Extracellular vesicles (EVs) are membrane-encapsulated particles with critical biomedical functions, including mediating intercellular communication, assisting tumor metastasis, and carrying protein and microRNA biomarkers. The downstream applications of EVs are greatly influenced by the quality of the isolated EVs. However, almost none of the separation methods can simultaneously achieve both high yield and high purity of the isolated EVs, thus making the isolation of EVs an essential challenge in EV research. Here, we developed a magnetic bead-mediated selective adsorption strategy (MagExo) for easy-to-operate EV isolation. Benefited from the presence of an adsorption window between EVs and proteins under the effect of a hydrophilic polymer, EVs tend to adsorb on the surface of magnetic beads selectively and can be separated from biological fluids with high purity by simple magnetic separation. The proposed method was used for EV isolation from plasma and cell culture media (CCM), with two times higher yield and comparable purity of the harvested EVs to that obtained by ultracentrifugation (UC). Downstream applications in proteomics analysis showed 86.6% (plasma) and 86.5% (CCM) of the analyzed proteins were matched with the ExoCarta database, which indicates MagExo indeed enriches EVs efficiently. Furthermore, we found the target RNA amount of the isolated EVs by MagExo were almost dozens and hundred times higher than the gold standard DG-UC and ultracentrifugation (UC) methods, respectively. All the results show that MagExo is a reliable, easy, and efficient approach to harvest EVs for a wide variety of downstream applications with minimized sample usage. STATEMENT OF SIGNIFICANCE: Extracellular vesicles (EVs) are presently attracting increasing interest among clinical and scientific researchers. Although the downstream applications of EVs are recognized to be greatly affected by the quality of the isolated EVs, almost none of the separation methods can simultaneously achieve high yield and high purity of the isolated EVs; this makes the isolation of EVs an essential challenge in EV research. In the present work, we proposed a simple and easy-to-operate method (MagExo) for the separation and purification of EVs based on the phenomenon that EVs can be selectively adsorbed on the surface of magnetic microspheres in the presence of a hydrophilic polymer. The performance of MagExo was comparable to or even better than that of gold standard methods and commercial kits, with two times higher yield and comparable purity of the harvested EVs to that achieved with ultracentrifugation (UC); this could meet the requirements of various EV-associated downstream applications. In addition, MagExo can be easily automated by commercial liquid workstations, thus significantly improving the isolation throughput and paving a new way in clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Xiaoxia Fang
- School of Biomedical Engineering /Med-X Research Institute, Shanghai Jiaotong University, Shanghai, PR China
| | - Cang Chen
- School of Biomedical Engineering /Med-X Research Institute, Shanghai Jiaotong University, Shanghai, PR China
| | - Bing Liu
- School of Biomedical Engineering /Med-X Research Institute, Shanghai Jiaotong University, Shanghai, PR China
| | - Zhijie Ma
- School of Biomedical Engineering /Med-X Research Institute, Shanghai Jiaotong University, Shanghai, PR China
| | - Fenglin Hu
- School of Biomedical Engineering /Med-X Research Institute, Shanghai Jiaotong University, Shanghai, PR China
| | - Haiyan Li
- School of Biomedical Engineering /Med-X Research Institute, Shanghai Jiaotong University, Shanghai, PR China
| | - Hongchen Gu
- School of Biomedical Engineering /Med-X Research Institute, Shanghai Jiaotong University, Shanghai, PR China.
| | - Hong Xu
- School of Biomedical Engineering /Med-X Research Institute, Shanghai Jiaotong University, Shanghai, PR China.
| |
Collapse
|
26
|
Wang X, Yuan X, Fu K, Liu C, Bai L, Wang X, Tan X, Zhang Y. Colorimetric analysis of extracellular vesicle surface proteins based on controlled growth of Au aptasensors. Analyst 2021; 146:2019-2028. [PMID: 33528468 DOI: 10.1039/d0an02080j] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Protein profiling of extracellular vesicles (EVs) provides important information in both clinical cancer diagnosis and relevant biological research studies. Although a variety of bioanalytical techniques have been investigated for EV characterization, limitations such as time-consuming operations, the requirement of large sample volume and demand for specialized instruments hinder their practical applications. Here, we report a simple and wash-free homogeneous colorimetric assay for sensitive detection of surface proteins on EVs. Au nanoparticles were modified with thiolated aptamers to fabricate aptasensors and incubated with EVs. Upon addition of a Au growth reagent, the solution color changed from light red to blue in the presence of target proteins and became deep red when the targets were absent. Expression of CD63, epithelial cell adhesion molecules (EpCAM), and mucin1 in EVs derived from two breast cancer cell lines (MCF-7 and MDA-MB-231) were compared, showing results consistent with western blotting results. The colorimetric assay achieves a limit of detection (LOD) down to 0.7 ng μL-1 against MCF-7 EVs based on the assessment of EpCAM expression, suggesting its potential to be applied in clinical breast cancer diagnosis.
Collapse
Affiliation(s)
- Xiaojie Wang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Kanno S, Hirano S, Sakamoto T, Furuyama A, Takase H, Kato H, Fukuta M, Aoki Y. Scavenger receptor MARCO contributes to cellular internalization of exosomes by dynamin-dependent endocytosis and macropinocytosis. Sci Rep 2020; 10:21795. [PMID: 33311558 PMCID: PMC7733512 DOI: 10.1038/s41598-020-78464-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/25/2020] [Indexed: 12/14/2022] Open
Abstract
Macrophage receptor with collagenous structure (MARCO) is a scavenger receptor class-A protein that is expressed on the cell surface of macrophages. MARCO mediates binding and ingestion of unopsonized environmental particles, including nano-sized materials. Exosomes are cell-derived, nano-sized vesicles (40–150 nm) that can contain lipids, RNA, DNA, and various proteins. Exosomes play an essential role in cell-to-cell communication via body fluids. However, mechanisms for the recognition and internalization of exosomes by recipient cells remain poorly characterized. In this study, cellular association of serum-derived fluorescent exosomes and 20-nm fluorescent nanoparticles (positive control) was compared between MARCO-expressing (CHO-MARCO) and control (CHO-CT) CHO-K1 cells to examine whether MARCO expression by recipient cells mediates the cellular uptake of exosomes and environmental nanoparticles. Fluorescence microscopic studies and quantitative analyses revealed that the cellular associations of both exosomes and 20-nm nanoparticles were greater in CHO-MARCO cells than in CHO-CT cells. Exosomes and nanoparticles colocalized with green fluorescent protein (GFP)-MARCO in cells transfected with GFP-MARCO-encoding constructs . Furthermore, inhibitory studies showed that actin reorganization and dynamin are involved in the MARCO-mediated cellular internalization of exosomes. These results indicated that MARCO plays a role in the uptake of exosomes.
Collapse
Affiliation(s)
- Sanae Kanno
- Department of Forensic Medicine, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan.
| | - Seishiro Hirano
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba, Ibaraki, 305-8506, Japan
| | - Tsubasa Sakamoto
- Department of Forensic Medicine, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Akiko Furuyama
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba, Ibaraki, 305-8506, Japan
| | - Hiroshi Takase
- Core Laboratory, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Hideaki Kato
- Department of Forensic Medicine, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Mamiko Fukuta
- Department of Forensic Medicine, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Yasuhiro Aoki
- Department of Forensic Medicine, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| |
Collapse
|
28
|
Grossi I, Radeghieri A, Paolini L, Porrini V, Pilotto A, Padovani A, Marengoni A, Barbon A, Bellucci A, Pizzi M, Salvi A, De Petro G. MicroRNA‑34a‑5p expression in the plasma and in its extracellular vesicle fractions in subjects with Parkinson's disease: An exploratory study. Int J Mol Med 2020; 47:533-546. [PMID: 33416118 PMCID: PMC7797475 DOI: 10.3892/ijmm.2020.4806] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 11/09/2020] [Indexed: 02/07/2023] Open
Abstract
Parkinson's disease (PD) is an important disabling age-related disorder and is the second most common neuro-degenerative disease. Currently, no established molecular biomarkers exist for the early diagnosis of PD. Circulating microRNAs (miRNAs), either vesicle-free or encapsulated in extracellular vesicles (EVs), have emerged as potential blood-based biomarkers also for neurodegenerative diseases. In this exploratory study, we focused on miR-34a-5p because of its well-documented involvement in neurobiology. To explore a differential profile of circulating miR-34a-5p in PD, PD patients and age-matched control subjects were enrolled. Serial ultracentrifugation steps and density gradient were used to separate EV subpopulations from plasma according to their different sedimentation properties (Large, Medium, Small EVs). Characterization of EV types was performed using western blotting and atomic force microscopy (AFM); purity from protein contaminants was checked with the colorimetric nanoplasmonic assay. Circulating miR-34a-5p levels were evaluated using qPCR in plasma and in each EV type. miR-34a-5p was significantly up-regulated in small EVs devoid of exogenous protein contaminants (pure SEVs) from PD patients and ROC analysis indicated a good diagnostic performance in discriminating patients from controls (AUC=0.74, P<0.05). Moreover, miR-34a-5p levels in pure SEVs were associated with disease duration, Hoehn and Yahr and Beck Depression Inventory scores. These results under-line the necessity to examine the miRNA content of each EV subpopulation to identify miRNA candidates with potential diagnostic value and lay the basis for future studies to validate the overexpression of circulating miR-34a-5p in PD via the use of pure SEVs.
Collapse
Affiliation(s)
- Ilaria Grossi
- Department of Molecular and Translational Medicine, University of Brescia, I‑25123 Brescia, Italy
| | - Annalisa Radeghieri
- Department of Molecular and Translational Medicine, University of Brescia, I‑25123 Brescia, Italy
| | - Lucia Paolini
- Department of Molecular and Translational Medicine, University of Brescia, I‑25123 Brescia, Italy
| | - Vanessa Porrini
- Department of Molecular and Translational Medicine, University of Brescia, I‑25123 Brescia, Italy
| | - Andrea Pilotto
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, I‑25123 Brescia, Italy
| | - Alessandro Padovani
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, I‑25123 Brescia, Italy
| | - Alessandra Marengoni
- General Medicine and Geriatrics Unit, Department of Clinical and Experimental Sciences, University of Brescia, I‑25123 Brescia, Italy
| | - Alessandro Barbon
- Department of Molecular and Translational Medicine, University of Brescia, I‑25123 Brescia, Italy
| | - Arianna Bellucci
- Department of Molecular and Translational Medicine, University of Brescia, I‑25123 Brescia, Italy
| | - Marina Pizzi
- Department of Molecular and Translational Medicine, University of Brescia, I‑25123 Brescia, Italy
| | - Alessandro Salvi
- Department of Molecular and Translational Medicine, University of Brescia, I‑25123 Brescia, Italy
| | - Giuseppina De Petro
- Department of Molecular and Translational Medicine, University of Brescia, I‑25123 Brescia, Italy
| |
Collapse
|
29
|
Recent advances in nanomaterial-based biosensors for the detection of exosomes. Anal Bioanal Chem 2020; 413:83-102. [PMID: 33164151 DOI: 10.1007/s00216-020-03000-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 10/04/2020] [Accepted: 10/09/2020] [Indexed: 12/24/2022]
Abstract
Exosomes are a type of extracellular vesicle actively secreted by almost all eukaryotic cells. They are ideal candidates for reliable next-generation biomarkers in the early diagnosis and therapeutic response evaluation of cancer. Thus, the quantification of exosomes is crucial in facilitating clinical research and application. Compared with traditional materials, nanomaterials have better optical, magnetic, electrical, and catalytic properties due to their small size, high specific surface area, and variable structure. The incorporation of nanomaterials into sensing systems is an attractive approach towards improving sensitivity and can provide improved sensor selectivity and stability. In this paper, we summarize the progress in nanomaterial-based exosome detection methods, including electrochemical biosensors, photoelectrochemical biosensors, colorimetric biosensors, fluorescence biosensors, chemiluminescence biosensors, electrochemiluminescence biosensors, surface plasmon resonance biosensors, and surface-enhanced Raman spectroscopy biosensors. Moreover, future research directions and challenges in exosome detection methods are discussed. We hope that this article will offer an overview of nanomaterial-based exosome detection techniques and open new avenues in disease research.Graphical abstract.
Collapse
|
30
|
Maia J, Batista S, Couto N, Gregório AC, Bodo C, Elzanowska J, Strano Moraes MC, Costa-Silva B. Employing Flow Cytometry to Extracellular Vesicles Sample Microvolume Analysis and Quality Control. Front Cell Dev Biol 2020; 8:593750. [PMID: 33195266 PMCID: PMC7661467 DOI: 10.3389/fcell.2020.593750] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 09/30/2020] [Indexed: 12/20/2022] Open
Abstract
Extracellular Vesicles (EVs), membrane vesicles released by all cells, are emerging mediators of cell-cell communication. By carrying biomolecules from tissues to biofluids, EVs have attracted attention as non-invasive sources of clinical biomarkers in liquid biopsies. EVs-based liquid biopsies usually require EVs isolation before content analysis, which frequently increases sample volume requirements. We here present a Flow Cytometry (FC) strategy that does not require isolation or concentration of EVs prior to staining. By doing so, it enables population analysis of EVs in samples characterized by challenging small volumes, while reducing overall sample processing time. To illustrate its application, we performed longitudinal non-lethal population analysis of EVs in mouse plasma and in single-animal collections of murine vitreous humor. By quantifying the proportion of vesicular particles in purified and non-purified biological samples, this method also serves as a precious tool to quality control isolates of EVs purified by different protocols. Our FC strategy has an unexplored clinical potential to analyze EVs in biofluids with intrinsically limited volumes and to multiply the number of different analytes in EVs that can be studied from a single collection of biofluid.
Collapse
Affiliation(s)
- Joana Maia
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal.,Graduate Program in Areas of Basic and Applied Biology, University of Porto, Porto, Portugal
| | - Silvia Batista
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Nuno Couto
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal.,Digestive Unit, Champalimaud Clinical Centre, Lisbon, Portugal
| | - Ana C Gregório
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Cristian Bodo
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Julia Elzanowska
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | | | - Bruno Costa-Silva
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal
| |
Collapse
|
31
|
Yon M, Pibourret C, Marty JD, Ciuculescu-Pradines D. Easy colorimetric detection of gadolinium ions based on gold nanoparticles: key role of phosphine-sulfonate ligands. NANOSCALE ADVANCES 2020; 2:4671-4681. [PMID: 36132884 PMCID: PMC9417556 DOI: 10.1039/d0na00374c] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 09/05/2020] [Indexed: 05/10/2023]
Abstract
The possibility to easily and rapidly assess the presence of Gd3+ ions in solution is of paramount importance in many domains like magnetic resonance imaging. In that context, the use of easy to implement colorimetric sensing probes based on gold nanoparticles (AuNPs) is of special interest. Herein, AuNPs functionalized with a commercial bis(p-sulfonatophenyl)phenyl phosphine ligand (BSPP) (AuNP@BSPP), bearing negatively charged sulfonate groups are used as a colorimetric sensing probe. The addition of Gd3+ ions onto these NPs was studied through UV-visible absorbance measurements, Quartz Crystal Microbalance with Dissipation monitoring (QCM-D) and transmission electron microscopy and compared with citrate covered AuNPs. We evidenced interactions between the Gd3+ ions and their water rich coordination sphere and sulfonate groups on the surface of AuNP@BSPP via electrostatic interactions and hydrogen bonding. These interactions induce the reversible aggregation of AuNP@BSPP in the presence of concentrations of Gd3+ ions at a μM level. We took advantage of this phenomenon to develop a simple and fast bench colorimetric assay for the detection of free Gd3+ ions, based on the determination of a flocculation parameter thanks to UV-visible measurements. Limits of detection and quantification were found equal to 0.74 μM and 4.76 μM of Gd3+ ions, respectively, with a high sensitivity that competes with conventional methods used for lanthanide detection.
Collapse
Affiliation(s)
- Marjorie Yon
- Laboratoire IMRCP, CNRS UMR 5623, Paul Sabatier University 118 route de Narbonne 31062 Toulouse France
| | - Claire Pibourret
- Laboratoire IMRCP, CNRS UMR 5623, Paul Sabatier University 118 route de Narbonne 31062 Toulouse France
| | - Jean-Daniel Marty
- Laboratoire IMRCP, CNRS UMR 5623, Paul Sabatier University 118 route de Narbonne 31062 Toulouse France
| | - Diana Ciuculescu-Pradines
- Laboratoire IMRCP, CNRS UMR 5623, Paul Sabatier University 118 route de Narbonne 31062 Toulouse France
| |
Collapse
|
32
|
Soekmadji C, Li B, Huang Y, Wang H, An T, Liu C, Pan W, Chen J, Cheung L, Falcon-Perez JM, Gho YS, Holthofer HB, Le MTN, Marcilla A, O'Driscoll L, Shekari F, Shen TL, Torrecilhas AC, Yan X, Yang F, Yin H, Xiao Y, Zhao Z, Zou X, Wang Q, Zheng L. The future of Extracellular Vesicles as Theranostics - an ISEV meeting report. J Extracell Vesicles 2020; 9:1809766. [PMID: 33144926 PMCID: PMC7580849 DOI: 10.1080/20013078.2020.1809766] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The utilization of extracellular vesicles (EVs) in clinical theranostics has rapidly advanced in the past decade. In November 2018, the International Society for Extracellular Vesicles (ISEV) held a workshop on “EVs in Clinical Theranostic”. Here, we report the conclusions of roundtable discussions on the current advancement in the analysis technologies and we provide some guidelines to researchers in the field to consider the use of EVs in clinical application. The main challenges and the requirements for EV separation and characterization strategies, quality control and clinical investigation were discussed to promote the application of EVs in future clinical studies.
Collapse
Affiliation(s)
- Carolina Soekmadji
- Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Bo Li
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yiyao Huang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Haifang Wang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Taixue An
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Chunchen Liu
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Weilun Pan
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jing Chen
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Lesley Cheung
- The Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia
| | - Juan Manuel Falcon-Perez
- Exosomes Laboratory and Metabolomics Platform, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain.,Centro De Investigación Biomédica En Red De Enfermedades Hepáticas Y Digestivas (Ciberehd), Madrid, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Yong Song Gho
- Laboratory of Intercellular Communication, Department of Life Science, POSTECH, South Korea
| | - Harry B Holthofer
- Medical Department, University Medical Center Hamburg-Eppendorf, Germany
| | - Minh T N Le
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Antonio Marcilla
- Àrea De Parasitologia, Departament De Farmàcia I Tecnologia Farmacèutica I Parasitologia, Universitat De València, Burjassot, Valencia, Spain.,Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics, Health Research Institute La Fe-Universitat De Valencia, Valencia, Spain
| | - Lorraine O'Driscoll
- School of Pharmacy and Pharmaceutical Sciences, Panoz Institute & Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Trinity St. James's Cancer Institute (TSJCI), Trinity College Dublin, Dublin, Ireland
| | - Faezeh Shekari
- Department of Stem Cells and Developmental BiologyCell Science, Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Tang Long Shen
- Department of Plant Pathology and Microbiology & Center for Biotechnology, National Taiwan University, Taipei, Taiwan
| | | | - Xiaomei Yan
- Department of Chemical Biology, the MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, the Key Laboratory for Chemical Biology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian, China
| | - Fuquan Yang
- Laboratory of Proteomics, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Hang Yin
- School of Pharmaceutical Sciences, Tsinghua University-Peking University Joint Center for Life Sciences, Tsinghua University, Beijing, China
| | - Yu Xiao
- Laboratory of Medicine Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zezhou Zhao
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Xue Zou
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
| | - Qian Wang
- Laboratory of Medicine Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
33
|
Montis C, Caselli L, Valle F, Zendrini A, Carlà F, Schweins R, Maccarini M, Bergese P, Berti D. Shedding light on membrane-templated clustering of gold nanoparticles. J Colloid Interface Sci 2020; 573:204-214. [DOI: 10.1016/j.jcis.2020.03.123] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 03/30/2020] [Accepted: 03/31/2020] [Indexed: 12/30/2022]
|
34
|
Effect of the Surface Chemical Composition and of Added Metal Cation Concentration on the Stability of Metal Nanoparticles Synthesized by Pulsed Laser Ablation in Water. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10124169] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Metal nanoparticles (NPs) made of gold, silver, and platinum have been synthesized by means of pulsed laser ablation in liquid aqueous solution. Independently from the metal nature, all NPs have an average diameter of 10 ± 5 nm. The ζ-potential values are: −62 ± 7 mV for gold, −44 ± 2 mV for silver and −58 ± 3 for platinum. XPS analysis demonstrates the absence of metal oxides in the case of gold and silver NPs. In the case of platinum NPs, 22% of the particle surface is ascribed to platinum oxidized species. This points to a marginal role of the metal oxides in building the negative charge that stabilizes these colloidal suspensions. The investigation of the colloidal stability of gold NPs in the presence of metal cations shows these NPs can be destabilized by trace amounts of selected metal ions. The case of Ag+ is paradigmatic since it is able to reduce the NP ζ-potential and to induce coagulation at concentrations as low as 3 μM, while in the case of K+ the critical coagulation concentration is around 8 mM. It is proposed that such a huge difference in destabilization power between monovalent cations can be accounted for by the difference in the reduction potential.
Collapse
|
35
|
Kluszczyńska K, Czernek L, Cypryk W, Pęczek Ł, Düchler M. Methods for the Determination of the Purity of Exosomes. Curr Pharm Des 2020; 25:4464-4485. [PMID: 31808383 DOI: 10.2174/1381612825666191206162712] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 12/03/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Exosomes open exciting new opportunities for advanced drug transport and targeted release. Furthermore, exosomes may be used for vaccination, immunosuppression or wound healing. To fully utilize their potential as drug carriers or immune-modulatory agents, the optimal purity of exosome preparations is of crucial importance. METHODS Articles describing the isolation and purification of exosomes were retrieved from the PubMed database. RESULTS Exosomes are often separated from biological fluids containing high concentrations of proteins, lipids and other molecules that keep vesicle purification challenging. A great number of purification protocols have been published, however, their outcome is difficult to compare because the assessment of purity has not been standardized. In this review, we first give an overview of the generation and composition of exosomes, as well as their multifaceted biological functions that stimulated various medical applications. Finally, we describe various methods that have been used to purify small vesicles and to assess the purity of exosome preparations and critically compare the quality of these evaluation protocols. CONCLUSION Combinations of various techniques have to be applied to reach the required purity and quality control of exosome preparations.
Collapse
Affiliation(s)
- Katarzyna Kluszczyńska
- Department of Bioorganic Chemistry, Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, 112 Sienkiewicza Street, 90-363 Lodz, Poland
| | - Liliana Czernek
- Department of Bioorganic Chemistry, Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, 112 Sienkiewicza Street, 90-363 Lodz, Poland
| | - Wojciech Cypryk
- Department of Bioorganic Chemistry, Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, 112 Sienkiewicza Street, 90-363 Lodz, Poland
| | - Łukasz Pęczek
- Department of Bioorganic Chemistry, Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, 112 Sienkiewicza Street, 90-363 Lodz, Poland
| | - Markus Düchler
- Department of Bioorganic Chemistry, Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, 112 Sienkiewicza Street, 90-363 Lodz, Poland
| |
Collapse
|
36
|
Rodriguez-Quijada C, Dahl JB. Non-contact microfluidic mechanical property measurements of single apoptotic bodies. Biochim Biophys Acta Gen Subj 2020; 1865:129657. [PMID: 32512171 DOI: 10.1016/j.bbagen.2020.129657] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 03/18/2020] [Accepted: 06/02/2020] [Indexed: 01/29/2023]
Abstract
BACKGROUND Cells exchange information by secreting micro- and nanosized extracellular vesicles (EVs), ranging from exosomes (30-100 nm) to apoptotic bodies (ABs, 1-5 μm). There is still much to understand about fundamental EV biological, physical, and chemical properties before clinical applications can be developed. EV mechanical properties have only been measured with atomic force microscopy (AFM) with its problematic adhesion and hard substrate effects. To understand EV mechanical behavior in less extreme mechanical conditions relevant to blood flow and many soft tissue environments, a non-contact measurement technique is needed. METHODS We measured the mechanical properties of single microscale ABs derived from human blood plasma using non-contact microfluidics. EVs were gently stretched in extensional flow, similar to a traditional tensile test, and a linear mechanical model was applied to estimate mechanical stiffnesses from the observed stretching. RESULTS The effective shear elastic modulus of ABs in non-contact flow conditions is approximately 5.6 ± 0.5 Pa, 7 orders of magnitude lower than previously reported AFM-measured biological exosome stiffnesses and 200 times smaller than suspended cells. CONCLUSIONS Apoptotic bodies are very soft in fluid environments and exhibit lower effective stiffnesses than suspended cells. By measuring ABs in a natural fluid environment and low-force regime without hard probes and surfaces, we achieved closer agreement with linear mechanical theory and therefore more accurate stiffness measurements. GENERAL SIGNIFICANCE AFM manufacturers and users should consider implementing new mechanical models to interpret AFM force indentation curves so that accurate extracellular vesicle mechanical properties can be extracted.
Collapse
Affiliation(s)
| | - Joanna B Dahl
- Engineering Department, University of Massachusetts Boston, Boston, MA 02125, United States of America.
| |
Collapse
|
37
|
Serrano-Pertierra E, Oliveira-Rodríguez M, Matos M, Gutiérrez G, Moyano A, Salvador M, Rivas M, Blanco-López MC. Extracellular Vesicles: Current Analytical Techniques for Detection and Quantification. Biomolecules 2020; 10:E824. [PMID: 32481493 PMCID: PMC7357140 DOI: 10.3390/biom10060824] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/24/2020] [Accepted: 05/25/2020] [Indexed: 02/06/2023] Open
Abstract
Since their first observation, understanding the biology of extracellular vesicles (EV) has been an important and challenging field of study. They play a key role in the intercellular communication and are involved in important physiological and pathological functions. Therefore, EV are considered as potential biomarkers for diagnosis, prognosis, and monitoring the response to treatment in some diseases. In addition, due to their properties, EV may be used for therapeutic purposes. In the study of EV, three major points have to be addressed: 1. How to isolate EV from cell culture supernatant/biological fluids, 2. how to detect them, and 3. how to characterize and quantify. In this review, we focus on the last two questions and provide the main analytical techniques up-to-date for detection and profiling of EV. We critically analyze the advantages and disadvantages of each one, aimed to be of relevance for all researchers working on EV biology and their potential applications.
Collapse
Affiliation(s)
- Esther Serrano-Pertierra
- Department of Physical and Analytical Chemistry, University of Oviedo, 33006 Oviedo, Spain; (E.S.-P.); (M.O.-R.); (A.M.)
- Instituto Universitario de Biotecnología de Asturias, University of Oviedo, 33006 Oviedo, Spain; (M.M.); (G.G.)
| | - Myriam Oliveira-Rodríguez
- Department of Physical and Analytical Chemistry, University of Oviedo, 33006 Oviedo, Spain; (E.S.-P.); (M.O.-R.); (A.M.)
| | - María Matos
- Instituto Universitario de Biotecnología de Asturias, University of Oviedo, 33006 Oviedo, Spain; (M.M.); (G.G.)
- Department of Chemical and Enviromental Engineering, University of Oviedo, 33006 Oviedo, Spain
| | - Gemma Gutiérrez
- Instituto Universitario de Biotecnología de Asturias, University of Oviedo, 33006 Oviedo, Spain; (M.M.); (G.G.)
- Department of Chemical and Enviromental Engineering, University of Oviedo, 33006 Oviedo, Spain
| | - Amanda Moyano
- Department of Physical and Analytical Chemistry, University of Oviedo, 33006 Oviedo, Spain; (E.S.-P.); (M.O.-R.); (A.M.)
- Instituto Universitario de Biotecnología de Asturias, University of Oviedo, 33006 Oviedo, Spain; (M.M.); (G.G.)
| | - María Salvador
- Department of Physics & IUTA, University of Oviedo, Campus de Viesques, 33204 Gijón, Spain; (M.S.); (M.R.)
| | - Montserrat Rivas
- Department of Physics & IUTA, University of Oviedo, Campus de Viesques, 33204 Gijón, Spain; (M.S.); (M.R.)
| | - María Carmen Blanco-López
- Department of Physical and Analytical Chemistry, University of Oviedo, 33006 Oviedo, Spain; (E.S.-P.); (M.O.-R.); (A.M.)
- Instituto Universitario de Biotecnología de Asturias, University of Oviedo, 33006 Oviedo, Spain; (M.M.); (G.G.)
| |
Collapse
|
38
|
Xu L, Shoaie N, Jahanpeyma F, Zhao J, Azimzadeh M, Al Jamal KT. Optical, electrochemical and electrical (nano)biosensors for detection of exosomes: A comprehensive overview. Biosens Bioelectron 2020; 161:112222. [PMID: 32365010 DOI: 10.1016/j.bios.2020.112222] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 12/13/2022]
Abstract
Exosomes are small extracellular vesicles involved in many physiological activities of cells in the human body. Exosomes from cancer cells have great potential to be applied in clinical diagnosis, early cancer detection and target identification for molecular therapy. While this field is gaining increasing interests from both academia and industry, barriers such as supersensitive detection techniques and highly-efficient isolation methods remain. In the clinical settings, there is an urgent need for rapid analysis, reliable detection and point-of-care testing (POCT). With these challenges to be addressed, this article aims to review recent developments and technical breakthroughs including optical, electrochemical and electrical biosensors for exosomes detection in the field of cancer and other diseases and demonstrate how nanobiosensors could enhance the performance of conventional sensors. Working strategies, limit of detections, advantages and shortcomings of the studies are summarized. New trends, challenges and future perspectives of exosome-driven POCT in liquid biopsy have been discussed.
Collapse
Affiliation(s)
- Lizhou Xu
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, United Kingdom
| | - Nahid Shoaie
- Department of Biotechnology, Tarbiat Modares University of Medical Science, Tehran, Iran
| | - Fatemeh Jahanpeyma
- Department of Biotechnology, Tarbiat Modares University of Medical Science, Tehran, Iran
| | - Junjie Zhao
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, United Kingdom
| | - Mostafa Azimzadeh
- Medical Nanotechnology & Tissue Engineering Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, 89195-999, Yazd, Iran; Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, 89195-999, Yazd, Iran; Department of Advanced Medical Sciences and Technologies, School of Paramedicine, Shahid Sadoughi University of Medical Sciences, 8916188635, Yazd, Iran.
| | - Khuloud T Al Jamal
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, United Kingdom.
| |
Collapse
|
39
|
Busatto S, Zendrini A, Radeghieri A, Paolini L, Romano M, Presta M, Bergese P. The nanostructured secretome. Biomater Sci 2020; 8:39-63. [PMID: 31799977 DOI: 10.1039/c9bm01007f] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The term secretome, which traditionally strictly refers to single proteins, should be expanded to also include the great variety of nanoparticles secreted by cells (secNPs) into the extracellular space, which ranges from high-density lipoproteins of a few nanometers to extracellular vesicles and fat globules of hundreds of nanometers. Widening the definition is urged by the ever-increasing understanding of the role of secNPs as regulators/mediators of key physiological and pathological processes, which also puts them in the running as breakthrough cell-free therapeutics and diagnostics. "Made by cells for cells", secNPs are envisioned as a sweeping paradigm shift in nanomedicine, promising to overcome the limitations of synthetic nanoparticles by unsurpassed circulation and targeting abilities, precision and sustainability. From a longer/wider perspective, advanced manipulation would possibly make secNPs available as building blocks for future "biogenic" nanotechnology. However, the current knowledge is fragmented and sectorial (the majority of the studies being focused on a specific biological and/or medical aspect of a given secNP class or subclass), the understanding of the nanoscale and interfacial properties is limited and the development of bioprocesses and regulatory initiatives is in the early days. We believe that new multidisciplinary competencies and synergistic efforts need to be attracted and augmented to move forward. This review will contribute to the effort by attempting for the first time to rationally gather and elaborate secNPs and their traits into a unique concise framework - from biogenesis to colloidal properties, engineering and clinical translation - disclosing the overall view and easing comparative analysis and future exploitation.
Collapse
Affiliation(s)
- S Busatto
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
| | | | | | | | | | | | | |
Collapse
|
40
|
Paolini L, Federici S, Consoli G, Arceri D, Radeghieri A, Alessandri I, Bergese P. Fourier-transform Infrared (FT-IR) spectroscopy fingerprints subpopulations of extracellular vesicles of different sizes and cellular origin. J Extracell Vesicles 2020; 9:1741174. [PMID: 32341767 PMCID: PMC7170381 DOI: 10.1080/20013078.2020.1741174] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 02/20/2020] [Accepted: 02/28/2020] [Indexed: 12/13/2022] Open
Abstract
Identification of extracellular vesicle (EV) subpopulations remains an open challenge. To date, the common strategy is based on searching and probing set of molecular components and physical properties intended to be univocally characteristics of the target subpopulation. Pitfalls include the risk to opt for an unsuitable marker set - which may either not represent the subpopulation or also cover other unintended subpopulations - and the need to use different characterization techniques and equipment. This approach focused on specific markers may result inadequate to routinely deal with EV subpopulations that have an intrinsic high level of heterogeneity. In this paper, we show that Fourier-transform Infrared (FT-IR) spectroscopy can provide a collective fingerprint of EV subpopulations in one single experiment. FT-IR measurements were performed on large (LEVs, ~600 nm), medium (MEVs, ~200 nm) and small (SEVs ~60 nm) EVs enriched from two different cell lines medium: murine prostate cancer (TRAMP-C2) and skin melanoma (B16). Spectral regions between 3100-2800 cm-1 and 1880-900 cm-1, corresponding to functional groups mainly ascribed to lipid and protein contributions, were acquired and processed by Principal Component Analysis (PCA). LEVs, MEVs and SEVs were separately grouped for both the considered cell lines. Moreover, subpopulations of the same size but from different sources were assigned (with different degrees of accuracy) to two different groups. These findings demonstrate that FT-IR has the potential to quickly fingerprint EV subpopulations as a whole, suggesting an appealing complement/alternative for their characterization and grading, extendable to healthy and pathological EVs and fully artificial nanovesicles.
Collapse
Affiliation(s)
- Lucia Paolini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Consorzio Sistemi a Grande Interfase (CSGI), Department of Chemistry, University of Florence, Sesto Fiorentino (FI), Italy
| | - Stefania Federici
- Department of Mechanical and Industrial Engineering, University of Brescia, Brescia, Italy
- Consorzio Interuniversitario Nazionale per la Scienza e la Tecnologia dei Materiali (INSTM), Florence, Italy
| | - Giovanni Consoli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Diletta Arceri
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Annalisa Radeghieri
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Consorzio Sistemi a Grande Interfase (CSGI), Department of Chemistry, University of Florence, Sesto Fiorentino (FI), Italy
| | - Ivano Alessandri
- Consorzio Interuniversitario Nazionale per la Scienza e la Tecnologia dei Materiali (INSTM), Florence, Italy
- Department of Information Engineering, University of Brescia, Brescia, Italy
- National Institute of Optics, National Research Council of Italy (CNR-INO), Unit of Brescia, Italy
| | - Paolo Bergese
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Consorzio Sistemi a Grande Interfase (CSGI), Department of Chemistry, University of Florence, Sesto Fiorentino (FI), Italy
- Consorzio Interuniversitario Nazionale per la Scienza e la Tecnologia dei Materiali (INSTM), Florence, Italy
- Institute for Biomedical Research and Innovation, National Research Council of Italy, Palermo, Italy
| |
Collapse
|
41
|
Gissi C, Radeghieri A, Antonetti Lamorgese Passeri C, Gallorini M, Calciano L, Oliva F, Veronesi F, Zendrini A, Cataldi A, Bergese P, Maffulli N, Berardi AC. Extracellular vesicles from rat-bone-marrow mesenchymal stromal/stem cells improve tendon repair in rat Achilles tendon injury model in dose-dependent manner: A pilot study. PLoS One 2020; 15:e0229914. [PMID: 32163452 PMCID: PMC7067391 DOI: 10.1371/journal.pone.0229914] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 02/17/2020] [Indexed: 01/18/2023] Open
Abstract
Mesenchymal stromal/stem cells (MSCs) are increasingly employed for tissue regeneration, largely mediated through paracrine actions. Currently, extracellular vesicles (EVs) released by MSCs are major mediators of these paracrine effects. We evaluated whether rat-bone-marrow-MSC-derived EVs (rBMSCs-EVs) can ameliorate tendon injury in an in vivo rat model. Pro-collagen1A2 and MMP14 protein are expressed in rBMSC-EVs, and are important factors for extracellular-matrix tendon-remodeling. In addition, we found pro-collagen1A2 in rBMSC-EV surface-membranes by dot blot. In vitro on cells isolated from Achilles tendons, utilized as rBMSC -EVs recipient cells, EVs at both low and high doses induce migration of tenocytes; at higher concentration, they induce proliferation and increase expression of Collagen type I in tenocytes. Pretreatment with trypsin abrogate the effect of EVs on cell proliferation and migration, and the expression of collagen I. When either low- or high-dose rBMSCs-EVs were injected into a rat-Achilles tendon injury-model (immediately after damage), at 30 days, rBMSC-EVs were found to have accelerated the remodeling stage of tendon repair in a dose-dependent manner. At histology and histomorphology evaluation, high doses of rBMSCs-EVs produced better restoration of tendon architecture, with optimal tendon-fiber alignment and lower vascularity. Higher EV-concentrations demonstrated greater expression of collagen type I and lower expression of collagen type III. BMSC-EVs hold promise as a novel cell-free modality for the management of tendon injuries.
Collapse
Affiliation(s)
- Clarissa Gissi
- Laboratory of Stem Cells, U.O.C. of Immunohaematology and Transfusion Medicine, Santo Spirito Hospital, Pescara, Italy
| | - Annalisa Radeghieri
- Department of Molecular and Translational Medicine and CSGI, University of Brescia, Brescia, Italy
| | | | | | - Lucia Calciano
- Dipartimento di Sanità Pubblica e Medicina di Comunità, Sezione di Epidemiologia e Statistica Medica, Università di Verona, Verona, Italy
| | - Francesco Oliva
- Department of Musculoskeletal Disorders, School of Medicine and Surgery, University of Salerno, Salerno, Italy
| | - Francesca Veronesi
- Laboratory of Preclinical and Surgical Studies, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Andrea Zendrini
- Department of Molecular and Translational Medicine and CSGI, University of Brescia, Brescia, Italy
| | - Amelia Cataldi
- Department of Pharmacy, University G. d’Annunzio, Chieti, Italy
| | - Paolo Bergese
- Department of Molecular and Translational Medicine and CSGI, University of Brescia, Brescia, Italy
| | - Nicola Maffulli
- Department of Orthopaedics and Traumatology, Azienda Ospedaliera San Giovanni di Dio e Ruggi d'Aragona, University of Salerno School of Medicine, Surgery and Dentistry, Salerno, Italy
- School of Pharmacy and Bioengineering, Faculty of Medicine, Keele University, Stoke on Trent, Keele, England, United Kingdom
- Centre for Sports and Exercise Medicine, Queen Mary University of London, Barts and the London School of Medicine and Dentistry, Mile End Hospital, London, England, United Kingdom
| | - Anna Concetta Berardi
- Laboratory of Stem Cells, U.O.C. of Immunohaematology and Transfusion Medicine, Santo Spirito Hospital, Pescara, Italy
- * E-mail: ,
| |
Collapse
|
42
|
Zendrini A, Paolini L, Busatto S, Radeghieri A, Romano M, Wauben MHM, van Herwijnen MJC, Nejsum P, Borup A, Ridolfi A, Montis C, Bergese P. Augmented COlorimetric NANoplasmonic (CONAN) Method for Grading Purity and Determine Concentration of EV Microliter Volume Solutions. Front Bioeng Biotechnol 2020; 7:452. [PMID: 32117903 PMCID: PMC7028770 DOI: 10.3389/fbioe.2019.00452] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 12/18/2019] [Indexed: 12/15/2022] Open
Abstract
This protocol paper describes how to assign a purity grade and to subsequently titrate extracellular vesicle (EV) solutions of a few microliters in volume by microplate COlorimetric NANoplasmonic (CONAN) assay. The CONAN assay consists of a solution of gold nanoparticles (AuNPs) into which the EV preparation is added. The solution turns blue if the EV preparation is pure, whereas it stays red if soluble exogenous single and aggregated proteins (SAPs; often referred to as protein contaminants) are present. The color change is visible by the naked eye or can be quantified by UV-Vis spectroscopy, providing an index of purity (a unique peculiarity to date). The assay specifically targets SAPs, and not the EV-related proteins, with a detection limit <50 ng/μl (an order of magnitude higher resolution than that of the Bradford protein assay). For pure solutions, the assay also allows for determining the EV number, as the color shift is linearly dependent on the AuNP/EV molar ratio. Instead, it automatically reports if the solution bears SAP contaminants, thus avoiding counting artifacts. The CONAN assay proves to be robust and reliable and displays very interesting performances in terms of cost (inexpensive reagents, run by standard microplate readers), working volumes (1–2 μl of sample required), and time (full procedure takes <1 h). The assay is applicable to all classes of natural and artificial lipid microvesicles and nanovesicles.
Collapse
Affiliation(s)
- Andrea Zendrini
- Department of Animal Science, Food and Nutrition-Università Cattolica del Sacro Cuore, Piacenza, Italy.,Consorzio Interuniversitario Nazionale per la Scienza e la Tecnologia dei Materiali, Florence, Italy
| | - Lucia Paolini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.,Consorzio Sistemi a Grande Interfase, Department of Chemistry, University of Florence, Sesto Fiorentino, Italy
| | - Sara Busatto
- Consorzio Sistemi a Grande Interfase, Department of Chemistry, University of Florence, Sesto Fiorentino, Italy.,Department of Transplantation, Mayo Clinic, Jacksonville, FL, United States
| | - Annalisa Radeghieri
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.,Consorzio Sistemi a Grande Interfase, Department of Chemistry, University of Florence, Sesto Fiorentino, Italy
| | - Miriam Romano
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.,Consorzio Sistemi a Grande Interfase, Department of Chemistry, University of Florence, Sesto Fiorentino, Italy
| | - Marca H M Wauben
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Martijn J C van Herwijnen
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Peter Nejsum
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Anne Borup
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Andrea Ridolfi
- Consorzio Sistemi a Grande Interfase, Department of Chemistry, University of Florence, Sesto Fiorentino, Italy.,Istituto per lo Studio dei Materiali Nanostrutturati (CNR-ISMN), Bologna, Italy.,Department of Chemistry, University of Florence, Sesto Fiorentino, Italy
| | - Costanza Montis
- Consorzio Sistemi a Grande Interfase, Department of Chemistry, University of Florence, Sesto Fiorentino, Italy.,Department of Chemistry, University of Florence, Sesto Fiorentino, Italy
| | - Paolo Bergese
- Consorzio Interuniversitario Nazionale per la Scienza e la Tecnologia dei Materiali, Florence, Italy.,Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.,Consorzio Sistemi a Grande Interfase, Department of Chemistry, University of Florence, Sesto Fiorentino, Italy
| |
Collapse
|
43
|
Isolation and analysis of extracellular vesicles in a Morpho butterfly wing-integrated microvortex biochip. Biosens Bioelectron 2020; 154:112073. [PMID: 32056968 DOI: 10.1016/j.bios.2020.112073] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 01/30/2020] [Accepted: 02/03/2020] [Indexed: 12/17/2022]
Abstract
With the function of mediating intercellular communication between cells, extracellular vesicles (EVs) have been intently studied for their physiopathology and clinical application values. However, efficient EV isolation from biological fluids remains a significant challenge. To address this, this work constructs a new microvortex chip that can isolate EVs efficiently by integrating the lipid nanoprobe modified Morpho Menelaus (M. Menelaus) butterfly wing into microfluidic chip. M. Menelaus wing is well known for its orderly arranged periodic nanostructures and can generate microvortex when liquid passes through it, leading to increased interaction between EVs and M. Menelaus wing. In addition, the nanoprobe containing lipid tails can be inserted into EVs through their lipid bilayer membrane structure. Based on the described properties, high-throughput enrichment of EVs with over 70% isolation efficiency was realized. Moreover, it was demonstrated that the nanoprobe system based on M. Menelaus wing enabled downstream biological analysis of nucleic acids and proteins in EVs. Microvortex chips showed potential application value in efficient EV isolation for biomedical research and cancer diagnosis.
Collapse
|
44
|
Mendozza M, Caselli L, Salvatore A, Montis C, Berti D. Nanoparticles and organized lipid assemblies: from interaction to design of hybrid soft devices. SOFT MATTER 2019; 15:8951-8970. [PMID: 31680131 DOI: 10.1039/c9sm01601e] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
This contribution reviews the state of art on hybrid soft matter assemblies composed of inorganic nanoparticles (NP) and lamellar or non-lamellar lipid bilayers. After a short outline of the relevant energetic contributions, we address the interaction of NPs with synthetic lamellar bilayers, meant as cell membrane mimics. We then review the design of hybrid nanostructured materials composed of lipid bilayers and some classes of inorganic NPs, with particular emphasis on the effects on the amphiphilic phase diagram and on the additional properties contributed by the NPs. Then, we present the latest developments on the use of lipid bilayers as coating agents for inorganic NPs. Finally, we remark on the main achievements of the last years and our vision for the development of the field.
Collapse
Affiliation(s)
- Marco Mendozza
- Department of Chemistry "Ugo Schiff", University of Florence, and CSGI (Italian Center for Colloid and Surface Science, Via della Lastruccia 3, Sesto Fiorentino, 50019 Firenze, Italy.
| | - Lucrezia Caselli
- Department of Chemistry "Ugo Schiff", University of Florence, and CSGI (Italian Center for Colloid and Surface Science, Via della Lastruccia 3, Sesto Fiorentino, 50019 Firenze, Italy.
| | - Annalisa Salvatore
- Department of Chemistry "Ugo Schiff", University of Florence, and CSGI (Italian Center for Colloid and Surface Science, Via della Lastruccia 3, Sesto Fiorentino, 50019 Firenze, Italy.
| | - Costanza Montis
- Department of Chemistry "Ugo Schiff", University of Florence, and CSGI (Italian Center for Colloid and Surface Science, Via della Lastruccia 3, Sesto Fiorentino, 50019 Firenze, Italy.
| | - Debora Berti
- Department of Chemistry "Ugo Schiff", University of Florence, and CSGI (Italian Center for Colloid and Surface Science, Via della Lastruccia 3, Sesto Fiorentino, 50019 Firenze, Italy.
| |
Collapse
|
45
|
Russell AE, Sneider A, Witwer KW, Bergese P, Bhattacharyya SN, Cocks A, Cocucci E, Erdbrügger U, Falcon-Perez JM, Freeman DW, Gallagher TM, Hu S, Huang Y, Jay SM, Kano SI, Lavieu G, Leszczynska A, Llorente AM, Lu Q, Mahairaki V, Muth DC, Noren Hooten N, Ostrowski M, Prada I, Sahoo S, Schøyen TH, Sheng L, Tesch D, Van Niel G, Vandenbroucke RE, Verweij FJ, Villar AV, Wauben M, Wehman AM, Yin H, Carter DRF, Vader P. Biological membranes in EV biogenesis, stability, uptake, and cargo transfer: an ISEV position paper arising from the ISEV membranes and EVs workshop. J Extracell Vesicles 2019; 8:1684862. [PMID: 31762963 PMCID: PMC6853251 DOI: 10.1080/20013078.2019.1684862] [Citation(s) in RCA: 176] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 08/23/2019] [Accepted: 10/04/2019] [Indexed: 02/07/2023] Open
Abstract
Paracrine and endocrine roles have increasingly been ascribed to extracellular vesicles (EVs) generated by multicellular organisms. Central to the biogenesis, content, and function of EVs are their delimiting lipid bilayer membranes. To evaluate research progress on membranes and EVs, the International Society for Extracellular Vesicles (ISEV) conducted a workshop in March 2018 in Baltimore, Maryland, USA, bringing together key opinion leaders and hands-on researchers who were selected on the basis of submitted applications. The workshop was accompanied by two scientific surveys and covered four broad topics: EV biogenesis and release; EV uptake and fusion; technologies and strategies used to study EV membranes; and EV transfer and functional assays. In this ISEV position paper, we synthesize the results of the workshop and the related surveys to outline important outstanding questions about EV membranes and describe areas of consensus. The workshop discussions and survey responses reveal that while much progress has been made in the field, there are still several concepts that divide opinion. Good consensus exists in some areas, including particular aspects of EV biogenesis, uptake and downstream signalling. Areas with little to no consensus include EV storage and stability, as well as whether and how EVs fuse with target cells. Further research is needed in these key areas, as a better understanding of membrane biology will contribute substantially towards advancing the field of extracellular vesicles.
Collapse
Affiliation(s)
- Ashley E. Russell
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alexandra Sneider
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Kenneth W. Witwer
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Paolo Bergese
- Department of Molecular and Translational Medicine, Università degli Studi di Brescia, CSGI and INSTM, Brescia, Italy
| | | | | | - Emanuele Cocucci
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, Columbus, OH, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | | | - Juan M. Falcon-Perez
- Exosomes laboratory and Metabolomics Platform, CIC bioGUNE, CIBERehd, Bizkaia, Spain
- IKERBASQUE, Basque Foundation for Science, Bizkaia, Spain
| | - David W. Freeman
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Thomas M. Gallagher
- Department of Microbiology and Immunology, Loyola University Chicago, Chicago, IL, USA
| | - Shuaishuai Hu
- School of Biological and Healthy Sciences, Technological University Dublin, Dublin, Ireland
| | - Yiyao Huang
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Clinical Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Steven M. Jay
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Shin-ichi Kano
- Department of Psychiatry and Behavioral Neurobiology, The University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
| | - Gregory Lavieu
- INSERM U932, Institut Curie, PSL Research University, France
| | | | - Alicia M. Llorente
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Quan Lu
- Program in Molecular and Integrative Physiological Sciences Departments of Environmental Health, Genetics & Complex Diseases Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Vasiliki Mahairaki
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Dillon C. Muth
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Matias Ostrowski
- INBIRS Institute, UBA-CONICET School of Medicine University of Buenos Aires, Buenos Aires, Argentina
| | | | - Susmita Sahoo
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tine Hiorth Schøyen
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- K. G. Jebsen - Thrombosis Research and Expertise Center (TREC), Department of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Lifu Sheng
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Deanna Tesch
- Department of Chemistry, Shaw University, Raleigh, NC, USA
| | - Guillaume Van Niel
- Institute for Psychiatry and Neuroscience of Paris, INSERM U1266, Hopital Saint-Anne, Université Descartes, Paris, France
| | - Roosmarijn E. Vandenbroucke
- VIB Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Frederik J. Verweij
- Institute for Psychiatry and Neuroscience of Paris, INSERM U1266, Hopital Saint-Anne, Université Descartes, Paris, France
| | - Ana V. Villar
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC) CSIC-Universidad de Cantabria and Departamento de Fisiología y Farmacología, Universidad de Cantabria, Santander, Spain
| | - Marca Wauben
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Ann M. Wehman
- Rudolf Virchow Center, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Hang Yin
- School of Pharmaceutical Sciences, Tsinghua University-Peking University Joint Center for Life Sciences, Tsinghua University, Beijing, China
| | | | - Pieter Vader
- Laboratory of Clinical Chemistry and Haematology & Department of Experimental Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
46
|
Rojalin T, Phong B, Koster HJ, Carney RP. Nanoplasmonic Approaches for Sensitive Detection and Molecular Characterization of Extracellular Vesicles. Front Chem 2019; 7:279. [PMID: 31134179 PMCID: PMC6514246 DOI: 10.3389/fchem.2019.00279] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 04/04/2019] [Indexed: 12/19/2022] Open
Abstract
All cells release a multitude of nanoscale extracellular vesicles (nEVs) into circulation, offering immense potential for new diagnostic strategies. Yet, clinical translation for nEVs remains a challenge due to their vast heterogeneity, our insufficient ability to isolate subpopulations, and the low frequency of disease-associated nEVs in biofluids. The growing field of nanoplasmonics is poised to address many of these challenges. Innovative materials engineering approaches based on exploiting nanoplasmonic phenomena, i.e., the unique interaction of light with nanoscale metallic materials, can achieve unrivaled sensitivity, offering real-time analysis and new modes of medical and biological imaging. We begin with an introduction into the basic structure and function of nEVs before critically reviewing recent studies utilizing nanoplasmonic platforms to detect and characterize nEVs. For the major techniques considered, surface plasmon resonance (SPR), localized SPR, and surface enhanced Raman spectroscopy (SERS), we introduce and summarize the background theory before reviewing the studies applied to nEVs. Along the way, we consider notable aspects, limitations, and considerations needed to apply plasmonic technologies to nEV detection and analysis.
Collapse
Affiliation(s)
- Tatu Rojalin
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Davis, CA, United States
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States
| | - Brian Phong
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States
| | - Hanna J. Koster
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States
| | - Randy P. Carney
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States
| |
Collapse
|
47
|
Gualerzi A, Kooijmans SAA, Niada S, Picciolini S, Brini AT, Camussi G, Bedoni M. Raman spectroscopy as a quick tool to assess purity of extracellular vesicle preparations and predict their functionality. J Extracell Vesicles 2019; 8:1568780. [PMID: 30728924 PMCID: PMC6352930 DOI: 10.1080/20013078.2019.1568780] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 12/03/2018] [Accepted: 01/09/2019] [Indexed: 12/20/2022] Open
Abstract
Extracellular vesicles (EVs) from a variety of stem cell sources are believed to harbour regenerative capacity, which may be exploited for therapeutic purposes. Because of EV interaction with other soluble secreted factors, EV activity may depend on the employed purification method, which limits cross-study comparisons and therapeutic development. Raman spectroscopy (RS) is a quick and easy method to assess EV purity and composition, giving in-depth biochemical overview on EV preparation. Hereby, we show how this method can be used to characterise EVs isolated from human liver stem cells and bone marrow mesenchymal stem/stromal cells by means of conventional ultracentrifugation (UC) and size exclusion chromatography (SEC) protocols. The obtained EV preparations were demonstrated to be characterised by different degrees of purity and a specific Raman fingerprint that represents both the cell source and the isolation procedure used. Moreover, RS provided useful hints to explore the factors underlying the functional diversity of EV preparations from the same cell source, thus representing a valuable tool to assess EV quality prior to functional assays or therapeutic application.
Collapse
Affiliation(s)
| | | | | | - Silvia Picciolini
- IRCCS Fondazione Don Carlo Gnocchi, Milano, Italy.,Nanomedicine Center NANOMIB, University of Milano-Bicocca, Monza, Italy
| | - Anna Teresa Brini
- IRCCS Istituto Ortopedico Galeazzi, Milano, Italy.,Dipartimento di Scienze Biomediche, Chirurgiche ed Odontoiatriche, Università degli Studi di Milano, Milano, Italy
| | - Giovanni Camussi
- Department of Medical Sciences, Molecular Biotechnology Center, University of Turin, Torino, Italy
| | | |
Collapse
|
48
|
Théry C, Witwer KW, Aikawa E, Alcaraz MJ, Anderson JD, Andriantsitohaina R, Antoniou A, Arab T, Archer F, Atkin-Smith GK, Ayre DC, Bach JM, Bachurski D, Baharvand H, Balaj L, Baldacchino S, Bauer NN, Baxter AA, Bebawy M, Beckham C, Bedina Zavec A, Benmoussa A, Berardi AC, Bergese P, Bielska E, Blenkiron C, Bobis-Wozowicz S, Boilard E, Boireau W, Bongiovanni A, Borràs FE, Bosch S, Boulanger CM, Breakefield X, Breglio AM, Brennan MÁ, Brigstock DR, Brisson A, Broekman MLD, Bromberg JF, Bryl-Górecka P, Buch S, Buck AH, Burger D, Busatto S, Buschmann D, Bussolati B, Buzás EI, Byrd JB, Camussi G, Carter DRF, Caruso S, Chamley LW, Chang YT, Chen C, Chen S, Cheng L, Chin AR, Clayton A, Clerici SP, Cocks A, Cocucci E, Coffey RJ, Cordeiro-da-Silva A, Couch Y, Coumans FAW, Coyle B, Crescitelli R, Criado MF, D’Souza-Schorey C, Das S, Datta Chaudhuri A, de Candia P, De Santana EF, De Wever O, del Portillo HA, Demaret T, Deville S, Devitt A, Dhondt B, Di Vizio D, Dieterich LC, Dolo V, Dominguez Rubio AP, Dominici M, Dourado MR, Driedonks TAP, Duarte FV, Duncan HM, Eichenberger RM, Ekström K, EL Andaloussi S, Elie-Caille C, Erdbrügger U, Falcón-Pérez JM, Fatima F, Fish JE, Flores-Bellver M, Försönits A, Frelet-Barrand A, Fricke F, Fuhrmann G, Gabrielsson S, Gámez-Valero A, Gardiner C, Gärtner K, Gaudin R, Gho YS, Giebel B, Gilbert C, Gimona M, Giusti I, Goberdhan DCI, Görgens A, Gorski SM, Greening DW, Gross JC, Gualerzi A, Gupta GN, Gustafson D, Handberg A, Haraszti RA, Harrison P, Hegyesi H, Hendrix A, Hill AF, Hochberg FH, Hoffmann KF, Holder B, Holthofer H, Hosseinkhani B, Hu G, Huang Y, Huber V, Hunt S, Ibrahim AGE, Ikezu T, Inal JM, Isin M, Ivanova A, Jackson HK, Jacobsen S, Jay SM, Jayachandran M, Jenster G, Jiang L, Johnson SM, Jones JC, Jong A, Jovanovic-Talisman T, Jung S, Kalluri R, Kano SI, Kaur S, Kawamura Y, Keller ET, Khamari D, Khomyakova E, Khvorova A, Kierulf P, Kim KP, Kislinger T, Klingeborn M, Klinke DJ, Kornek M, Kosanović MM, Kovács ÁF, Krämer-Albers EM, Krasemann S, Krause M, Kurochkin IV, Kusuma GD, Kuypers S, Laitinen S, Langevin SM, Languino LR, Lannigan J, Lässer C, Laurent LC, Lavieu G, Lázaro-Ibáñez E, Le Lay S, Lee MS, Lee YXF, Lemos DS, Lenassi M, Leszczynska A, Li ITS, Liao K, Libregts SF, Ligeti E, Lim R, Lim SK, Linē A, Linnemannstöns K, Llorente A, Lombard CA, Lorenowicz MJ, Lörincz ÁM, Lötvall J, Lovett J, Lowry MC, Loyer X, Lu Q, Lukomska B, Lunavat TR, Maas SLN, Malhi H, Marcilla A, Mariani J, Mariscal J, Martens-Uzunova ES, Martin-Jaular L, Martinez MC, Martins VR, Mathieu M, Mathivanan S, Maugeri M, McGinnis LK, McVey MJ, Meckes DG, Meehan KL, Mertens I, Minciacchi VR, Möller A, Møller Jørgensen M, Morales-Kastresana A, Morhayim J, Mullier F, Muraca M, Musante L, Mussack V, Muth DC, Myburgh KH, Najrana T, Nawaz M, Nazarenko I, Nejsum P, Neri C, Neri T, Nieuwland R, Nimrichter L, Nolan JP, Nolte-’t Hoen ENM, Noren Hooten N, O’Driscoll L, O’Grady T, O’Loghlen A, Ochiya T, Olivier M, Ortiz A, Ortiz LA, Osteikoetxea X, Østergaard O, Ostrowski M, Park J, Pegtel DM, Peinado H, Perut F, Pfaffl MW, Phinney DG, Pieters BCH, Pink RC, Pisetsky DS, Pogge von Strandmann E, Polakovicova I, Poon IKH, Powell BH, Prada I, Pulliam L, Quesenberry P, Radeghieri A, Raffai RL, Raimondo S, Rak J, Ramirez MI, Raposo G, Rayyan MS, Regev-Rudzki N, Ricklefs FL, Robbins PD, Roberts DD, Rodrigues SC, Rohde E, Rome S, Rouschop KMA, Rughetti A, Russell AE, Saá P, Sahoo S, Salas-Huenuleo E, Sánchez C, Saugstad JA, Saul MJ, Schiffelers RM, Schneider R, Schøyen TH, Scott A, Shahaj E, Sharma S, Shatnyeva O, Shekari F, Shelke GV, Shetty AK, Shiba K, Siljander PRM, Silva AM, Skowronek A, Snyder OL, Soares RP, Sódar BW, Soekmadji C, Sotillo J, Stahl PD, Stoorvogel W, Stott SL, Strasser EF, Swift S, Tahara H, Tewari M, Timms K, Tiwari S, Tixeira R, Tkach M, Toh WS, Tomasini R, Torrecilhas AC, Tosar JP, Toxavidis V, Urbanelli L, Vader P, van Balkom BWM, van der Grein SG, Van Deun J, van Herwijnen MJC, Van Keuren-Jensen K, van Niel G, van Royen ME, van Wijnen AJ, Vasconcelos MH, Vechetti IJ, Veit TD, Vella LJ, Velot É, Verweij FJ, Vestad B, Viñas JL, Visnovitz T, Vukman KV, Wahlgren J, Watson DC, Wauben MHM, Weaver A, Webber JP, Weber V, Wehman AM, Weiss DJ, Welsh JA, Wendt S, Wheelock AM, Wiener Z, Witte L, Wolfram J, Xagorari A, Xander P, Xu J, Yan X, Yáñez-Mó M, Yin H, Yuana Y, Zappulli V, Zarubova J, Žėkas V, Zhang JY, Zhao Z, Zheng L, Zheutlin AR, Zickler AM, Zimmermann P, Zivkovic AM, Zocco D, Zuba-Surma EK. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles 2018; 7:1535750. [PMID: 30637094 PMCID: PMC6322352 DOI: 10.1080/20013078.2018.1535750] [Citation(s) in RCA: 7374] [Impact Index Per Article: 1053.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 09/25/2018] [Indexed: 11/04/2022] Open
Abstract
The last decade has seen a sharp increase in the number of scientific publications describing physiological and pathological functions of extracellular vesicles (EVs), a collective term covering various subtypes of cell-released, membranous structures, called exosomes, microvesicles, microparticles, ectosomes, oncosomes, apoptotic bodies, and many other names. However, specific issues arise when working with these entities, whose size and amount often make them difficult to obtain as relatively pure preparations, and to characterize properly. The International Society for Extracellular Vesicles (ISEV) proposed Minimal Information for Studies of Extracellular Vesicles ("MISEV") guidelines for the field in 2014. We now update these "MISEV2014" guidelines based on evolution of the collective knowledge in the last four years. An important point to consider is that ascribing a specific function to EVs in general, or to subtypes of EVs, requires reporting of specific information beyond mere description of function in a crude, potentially contaminated, and heterogeneous preparation. For example, claims that exosomes are endowed with exquisite and specific activities remain difficult to support experimentally, given our still limited knowledge of their specific molecular machineries of biogenesis and release, as compared with other biophysically similar EVs. The MISEV2018 guidelines include tables and outlines of suggested protocols and steps to follow to document specific EV-associated functional activities. Finally, a checklist is provided with summaries of key points.
Collapse
Affiliation(s)
- Clotilde Théry
- Institut Curie, INSERM U932, PSL Research University, Paris, France
| | - Kenneth W Witwer
- The Johns Hopkins University School of Medicine, Department of Molecular and Comparative Pathobiology, Baltimore, MD, USA
- The Johns Hopkins University School of Medicine, Department of Neurology, Baltimore, MD, USA
| | - Elena Aikawa
- Brigham and Women’s Hospital, Center for Interdisciplinary Cardiovascular Sciences, Boston, MA, USA
- Harvard Medical School, Cardiovascular Medicine, Boston, MA, USA
| | - Maria Jose Alcaraz
- Interuniversity Research Institute for Molecular Recognition and Technological Development (IDM), University of Valencia, Polytechnic University of Valencia, Valencia, Spain
| | | | | | - Anna Antoniou
- German Centre for Neurodegenerative Diseases (DZNE), Bonn, Germany
- University Hospital Bonn (UKB), Bonn, Germany
| | - Tanina Arab
- Université de Lille, INSERM, U-1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse - PRISM, Lille, France
| | - Fabienne Archer
- University of Lyon, INRA, EPHE, UMR754 Viral Infections and Comparative Pathology, Lyon, France
| | - Georgia K Atkin-Smith
- La Trobe University, La Trobe Institute for Molecular Science, Department of Biochemistry and Genetics, Bundoora, Australia
| | - D Craig Ayre
- Atlantic Cancer Research Institute, Moncton, Canada
- Mount Allison University, Department of Chemistry and Biochemistry, Sackville, Canada
| | - Jean-Marie Bach
- Université Bretagne Loire, Oniris, INRA, IECM, Nantes, France
| | - Daniel Bachurski
- University of Cologne, Department of Internal Medicine I, Cologne, Germany
| | - Hossein Baharvand
- Royan Institute for Stem Cell Biology and Technology, ACECR, Cell Science Research Center, Department of Stem Cells and Developmental Biology, Tehran, Iran
- University of Science and Culture, ACECR, Department of Developmental Biology, Tehran, Iran
| | - Leonora Balaj
- Massachusetts General Hospital, Department of Neurosurgery, Boston, MA, USA
| | | | - Natalie N Bauer
- University of South Alabama, Department of Pharmacology, Center for Lung Biology, Mobile, AL, USA
| | - Amy A Baxter
- La Trobe University, La Trobe Institute for Molecular Science, Department of Biochemistry and Genetics, Bundoora, Australia
| | - Mary Bebawy
- University of Technology Sydney, Discipline of Pharmacy, Graduate School of Health, Sydney, Australia
| | | | - Apolonija Bedina Zavec
- National Institute of Chemistry, Department of Molecular Biology and Nanobiotechnology, Ljubljana, Slovenia
| | - Abderrahim Benmoussa
- Université Laval, Centre de Recherche du CHU de Québec, Department of Infectious Diseases and Immunity, Quebec City, Canada
| | | | - Paolo Bergese
- CSGI - Research Center for Colloids and Nanoscience, Florence, Italy
- INSTM - National Interuniversity Consortium of Materials Science and Technology, Florence, Italy
- University of Brescia, Department of Molecular and Translational Medicine, Brescia, Italy
| | - Ewa Bielska
- University of Birmingham, Institute of Microbiology and Infection, Birmingham, UK
| | | | - Sylwia Bobis-Wozowicz
- Jagiellonian University, Faculty of Biochemistry, Biophysics and Biotechnology, Department of Cell Biology, Kraków, Poland
| | - Eric Boilard
- Université Laval, Centre de Recherche du CHU de Québec, Department of Infectious Diseases and Immunity, Quebec City, Canada
| | - Wilfrid Boireau
- FEMTO-ST Institute, UBFC, CNRS, ENSMM, UTBM, Besançon, France
| | - Antonella Bongiovanni
- Institute of Biomedicine and Molecular Immunology (IBIM), National Research Council (CNR) of Italy, Palermo, Italy
| | - Francesc E Borràs
- Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, REMAR-IVECAT Group, Badalona, Spain
- Germans Trias i Pujol University Hospital, Nephrology Service, Badalona, Spain
- Universitat Autònoma de Barcelona, Department of Cell Biology, Physiology & Immunology, Barcelona, Spain
| | - Steffi Bosch
- Université Bretagne Loire, Oniris, INRA, IECM, Nantes, France
| | - Chantal M Boulanger
- INSERM UMR-S 970, Paris Cardiovascular Research Center, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Xandra Breakefield
- Massachusetts General Hospital and Neuroscience Program, Harvard Medical School, Department of Neurology and Radiology, Boston, MA, USA
| | - Andrew M Breglio
- Icahn School of Medicine at Mount Sinai, New York City, NY, USA
- National Institutes of Health, National Institute on Deafness and Other Communication Disorders, Bethesda, MD, USA
| | - Meadhbh Á Brennan
- Harvard University, School of Engineering and Applied Sciences, Cambridge, MA, USA
- Massachusetts General Hospital, Harvard Medical School, Department of Neurology, Boston, MA, USA
- Université de Nantes, INSERM UMR 1238, Bone Sarcoma and Remodeling of Calcified Tissues, PhyOS, Nantes, France
| | - David R Brigstock
- Nationwide Children’s Hospital, Columbus, OH, USA
- The Ohio State University, Columbus, OH, USA
| | - Alain Brisson
- UMR-CBMN, CNRS-Université de Bordeaux, Bordeaux, France
| | - Marike LD Broekman
- Haaglanden Medical Center, Department of Neurosurgery, The Hague, The Netherlands
- Leiden University Medical Center, Department of Neurosurgery, Leiden, The Netherlands
- Massachusetts General Hospital, Department of Neurology, Boston, MA, USA
| | - Jacqueline F Bromberg
- Memorial Sloan Kettering Cancer Center, Department of Medicine, New York City, NY, USA
- Weill Cornell Medicine, Department of Medicine, New York City, NY, USA
| | | | - Shilpa Buch
- University of Nebraska Medical Center, Department of Pharmacology and Experimental Neuroscience, Omaha, NE, USA
| | - Amy H Buck
- University of Edinburgh, Institute of Immunology & Infection Research, Edinburgh, UK
| | - Dylan Burger
- Kidney Research Centre, Ottawa, Canada
- Ottawa Hospital Research Institute, Ottawa, Canada
- University of Ottawa, Ottawa, Canada
| | - Sara Busatto
- Mayo Clinic, Department of Transplantation, Jacksonville, FL, USA
- University of Brescia, Department of Molecular and Translational Medicine, Brescia, Italy
| | - Dominik Buschmann
- Technical University of Munich, TUM School of Life Sciences Weihenstephan, Division of Animal Physiology and Immunology, Freising, Germany
| | - Benedetta Bussolati
- University of Torino, Department of Molecular Biotechnology and Health Sciences, Torino, Italy
| | - Edit I Buzás
- MTA-SE Immuno-Proteogenomics Research Groups, Budapest, Hungary
- Semmelweis University, Department of Genetics, Cell- and Immunobiology, Budapest, Hungary
| | - James Bryan Byrd
- University of Michigan, Department of Medicine, Ann Arbor, MI, USA
| | - Giovanni Camussi
- University of Torino, Department of Medical Sciences, Torino, Italy
| | - David RF Carter
- Oxford Brookes University, Department of Biological and Medical Sciences, Oxford, UK
| | - Sarah Caruso
- La Trobe University, La Trobe Institute for Molecular Science, Department of Biochemistry and Genetics, Bundoora, Australia
| | - Lawrence W Chamley
- University of Auckland, Department of Obstetrics and Gynaecology, Auckland, New Zealand
| | - Yu-Ting Chang
- National Taiwan University Hospital, Department of Internal Medicine, Taipei, Taiwan
| | - Chihchen Chen
- National Tsing Hua University, Department of Power Mechanical Engineering, Hsinchu, Taiwan
- National Tsing Hua University, Institute of Nanoengineering and Microsystems, Hsinchu, Taiwan
| | - Shuai Chen
- Leibniz Institute for Farm Animal Biology (FBN), Institute of Reproductive Biology, Dummerstorf, Germany
| | - Lesley Cheng
- La Trobe University, La Trobe Institute for Molecular Science, Department of Biochemistry and Genetics, Bundoora, Australia
| | | | - Aled Clayton
- Cardiff University, School of Medicine, Cardiff, UK
| | | | - Alex Cocks
- Cardiff University, School of Medicine, Cardiff, UK
| | - Emanuele Cocucci
- The Ohio State University, College of Pharmacy, Division of Pharmaceutics and Pharmaceutical Chemistry, Columbus, OH, USA
- The Ohio State University, Comprehensive Cancer Center, Columbus, OH, USA
| | - Robert J Coffey
- Vanderbilt University Medical Center, Epithelial Biology Center, Department of Medicine, Nashville, TN, USA
| | | | - Yvonne Couch
- University of Oxford, Radcliffe Department of Medicine, Acute Stroke Programme - Investigative Medicine, Oxford, UK
| | - Frank AW Coumans
- Academic Medical Centre of the University of Amsterdam, Department of Clinical Chemistry and Vesicle Observation Centre, Amsterdam, The Netherlands
| | - Beth Coyle
- The University of Nottingham, School of Medicine, Children’s Brain Tumour Research Centre, Nottingham, UK
| | - Rossella Crescitelli
- University of Gothenburg, Institute of Medicine at Sahlgrenska Academy, Krefting Research Centre, Gothenburg, Sweden
| | | | | | - Saumya Das
- Massachusetts General Hospital, Boston, MA, USA
| | - Amrita Datta Chaudhuri
- The Johns Hopkins University School of Medicine, Department of Neurology, Baltimore, MD, USA
| | | | - Eliezer F De Santana
- The Sociedade Beneficente Israelita Brasileira Albert Einstein, São Paulo, Brazil
| | - Olivier De Wever
- Cancer Research Institute Ghent, Ghent, Belgium
- Ghent University, Department of Radiation Oncology and Experimental Cancer Research, Laboratory of Experimental Cancer Research, Ghent, Belgium
| | - Hernando A del Portillo
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Institut d’Investigació Germans Trias i Pujol (IGTP), PVREX group, Badalona, Spain
- ISGlobal, Hospital Clínic - Universitat de Barcelona, PVREX Group, Barcelona, Spain
| | - Tanguy Demaret
- Université Catholique de Louvain, Institut de Recherche Expérimentale et Clinique (IREC), Laboratory of Pediatric Hepatology and Cell Therapy, Brussels, Belgium
| | - Sarah Deville
- Universiteit Hasselt, Diepenbeek, Belgium
- Vlaamse Instelling voor Technologisch Onderzoek (VITO), Mol, Belgium
| | - Andrew Devitt
- Aston University, School of Life & Health Sciences, Birmingham, UK
| | - Bert Dhondt
- Cancer Research Institute Ghent, Ghent, Belgium
- Ghent University Hospital, Department of Urology, Ghent, Belgium
- Ghent University, Department of Radiation Oncology and Experimental Cancer Research, Laboratory of Experimental Cancer Research, Ghent, Belgium
| | | | | | - Vincenza Dolo
- University of L’Aquila, Department of Life, Health and Environmental Sciences, L’Aquila, Italy
| | - Ana Paula Dominguez Rubio
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Buenos Aires, Argentina
| | - Massimo Dominici
- TPM of Mirandola, Mirandola, Italy
- University of Modena and Reggio Emilia, Division of Oncology, Modena, Italy
| | - Mauricio R Dourado
- University of Campinas, Piracicaba Dental School, Department of Oral Diagnosis, Piracicaba, Brazil
- University of Oulu, Faculty of Medicine, Cancer and Translational Medicine Research Unit, Oulu, Finland
| | - Tom AP Driedonks
- Utrecht University, Faculty of Veterinary Medicine, Department of Biochemistry and Cell Biology, Utrecht, The Netherlands
| | | | - Heather M Duncan
- McGill University, Division of Experimental Medicine, Montreal, Canada
- McGill University, The Research Institute of the McGill University Health Centre, Child Health and Human Development Program, Montreal, Canada
| | - Ramon M Eichenberger
- James Cook University, Australian Institute of Tropical Health and Medicine, Centre for Biodiscovery and Molecular Development of Therapeutics, Cairns, Australia
| | - Karin Ekström
- University of Gothenburg, Institute of Clinical Sciences at Sahlgrenska Academy, Department of Biomaterials, Gothenburg, Sweden
| | - Samir EL Andaloussi
- Evox Therapeutics Limited, Oxford, UK
- Karolinska Institute, Stockholm, Sweden
| | | | - Uta Erdbrügger
- University of Virginia Health System, Department of Medicine, Division of Nephrology, Charlottesville, VA, USA
| | - Juan M Falcón-Pérez
- CIC bioGUNE, CIBERehd, Exosomes Laboratory & Metabolomics Platform, Derio, Spain
- IKERBASQUE Research Science Foundation, Bilbao, Spain
| | - Farah Fatima
- University of São Paulo, Ribeirão Preto Medical School, Department of Pathology and Forensic Medicine, Ribeirão Preto, Brazil
| | - Jason E Fish
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
- University of Toronto, Department of Laboratory Medicine and Pathobiology, Toronto, Canada
| | - Miguel Flores-Bellver
- University of Colorado, School of Medicine, Department of Ophthalmology, Cell Sight-Ocular Stem Cell and Regeneration Program, Aurora, CO, USA
| | - András Försönits
- Semmelweis University, Department of Genetics, Cell- and Immunobiology, Budapest, Hungary
| | | | - Fabia Fricke
- German Cancer Research Center (DKFZ), Clinical Cooperation Unit Applied Tumor Biology, Heidelberg, Germany
- University Hospital Heidelberg, Institute of Pathology, Applied Tumor Biology, Heidelberg, Germany
| | - Gregor Fuhrmann
- Helmholtz-Centre for Infection Research, Braunschweig, Germany
- Helmholtz-Institute for Pharmaceutical Research Saarland, Saarbrücken, Germany
- Saarland University, Saarbrücken, Germany
| | - Susanne Gabrielsson
- Karolinska Institute, Department of Medicine Solna, Division for Immunology and Allergy, Stockholm, Sweden
| | - Ana Gámez-Valero
- Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, REMAR-IVECAT Group, Badalona, Spain
- Universitat Autònoma de Barcelona, Hospital Universitari and Health Sciences Research Institute Germans Trias i Pujol, Department of Pathology, Barcelona, Spain
| | | | - Kathrin Gärtner
- Helmholtz Center Munich German Research Center for Environmental Health, Research Unit Gene Vectors, Munich, Germany
| | - Raphael Gaudin
- INSERM U1110, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
| | - Yong Song Gho
- POSTECH (Pohang University of Science and Technology), Department of Life Sciences, Pohang, South Korea
| | - Bernd Giebel
- University Hospital Essen, University Duisburg-Essen, Institute for Transfusion Medicine, Essen, Germany
| | - Caroline Gilbert
- Université Laval, Centre de Recherche du CHU de Québec, Department of Infectious Diseases and Immunity, Quebec City, Canada
| | - Mario Gimona
- Paracelsus Medical University, GMP Unit, Salzburg, Austria
| | - Ilaria Giusti
- University of L’Aquila, Department of Life, Health and Environmental Sciences, L’Aquila, Italy
| | - Deborah CI Goberdhan
- University of Oxford, Department of Physiology, Anatomy and Genetics, Oxford, UK
| | - André Görgens
- Evox Therapeutics Limited, Oxford, UK
- Karolinska Institute, Clinical Research Center, Department of Laboratory Medicine, Stockholm, Sweden
- University Hospital Essen, University Duisburg-Essen, Institute for Transfusion Medicine, Essen, Germany
| | - Sharon M Gorski
- BC Cancer, Canada’s Michael Smith Genome Sciences Centre, Vancouver, Canada
- Simon Fraser University, Department of Molecular Biology and Biochemistry, Burnaby, Canada
| | - David W Greening
- La Trobe University, La Trobe Institute for Molecular Science, Department of Biochemistry and Genetics, Bundoora, Australia
| | - Julia Christina Gross
- University Medical Center Göttingen, Developmental Biochemistry, Göttingen, Germany
- University Medical Center Göttingen, Hematology and Oncology, Göttingen, Germany
| | - Alice Gualerzi
- IRCCS Fondazione Don Carlo Gnocchi, Laboratory of Nanomedicine and Clinical Biophotonics (LABION), Milan, Italy
| | - Gopal N Gupta
- Loyola University Chicago, Department of Urology, Maywood, IL, USA
| | - Dakota Gustafson
- University of Toronto, Department of Laboratory Medicine and Pathobiology, Toronto, Canada
| | - Aase Handberg
- Aalborg University Hospital, Department of Clinical Biochemistry, Aalborg, Denmark
- Aalborg University, Clinical Institute, Aalborg, Denmark
| | - Reka A Haraszti
- University of Massachusetts Medical School, RNA Therapeutics Institute, Worcester, MA, USA
| | | | - Hargita Hegyesi
- Semmelweis University, Department of Genetics, Cell- and Immunobiology, Budapest, Hungary
| | - An Hendrix
- Cancer Research Institute Ghent, Ghent, Belgium
- Ghent University, Department of Radiation Oncology and Experimental Cancer Research, Laboratory of Experimental Cancer Research, Ghent, Belgium
| | - Andrew F Hill
- La Trobe University, La Trobe Institute for Molecular Science, Department of Biochemistry and Genetics, Bundoora, Australia
| | - Fred H Hochberg
- Scintillon Institute, La Jolla, CA, USA
- University of California, San Diego, Department of Neurosurgery, La Jolla, CA, USA
| | - Karl F Hoffmann
- Aberystwyth University, Institute of Biological, Environmental and Rural Sciences (IBERS), Aberystwyth, United Kingdom
| | - Beth Holder
- Imperial College London, London, UK
- MRC The Gambia, Fajara, The Gambia
| | | | - Baharak Hosseinkhani
- Hasselt University, Biomedical Research Institute (BIOMED), Department of Medicine and Life Sciences, Hasselt, Belgium
| | - Guoku Hu
- University of Nebraska Medical Center, Department of Pharmacology and Experimental Neuroscience, Omaha, NE, USA
| | - Yiyao Huang
- Nanfang Hospital, Southern Medical University, Department of Clinical Laboratory Medicine, Guangzhou, China
- The Johns Hopkins University School of Medicine, Department of Molecular and Comparative Pathobiology, Baltimore, MD, USA
| | - Veronica Huber
- Fondazione IRCCS Istituto Nazionale dei Tumori, Unit of Immunotherapy of Human Tumors, Milan, Italy
| | | | | | - Tsuneya Ikezu
- Boston University School of Medicine, Boston, MA, USA
| | - Jameel M Inal
- University of Hertfordshire, School of Life and Medical Sciences, Biosciences Research Group, Hatfield, UK
| | - Mustafa Isin
- Istanbul University Oncology Institute, Basic Oncology Department, Istanbul, Turkey
| | - Alena Ivanova
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, Heidelberg, Germany
| | - Hannah K Jackson
- The University of Nottingham, School of Medicine, Children’s Brain Tumour Research Centre, Nottingham, UK
| | - Soren Jacobsen
- Copenhagen Lupus and Vasculitis Clinic, Section 4242 - Rigshospitalet, Copenhagen, Denmark
- University of Copenhagen, Institute of Clinical Medicine, Copenhagen, Denmark
| | - Steven M Jay
- University of Maryland, Fischell Department of Bioengineering, College Park, MD, USA
| | - Muthuvel Jayachandran
- Mayo Clinic, College of Medicine, Department of Physiology and Biomedical Engineering, Rochester, MN, USA
| | | | - Lanzhou Jiang
- La Trobe University, La Trobe Institute for Molecular Science, Department of Biochemistry and Genetics, Bundoora, Australia
| | - Suzanne M Johnson
- University of Manchester, Division of Cancer Sciences, Manchester Cancer Research Centre, Manchester, UK
| | - Jennifer C Jones
- National Institutes of Health, National Cancer Institute, Center for Cancer Research, Bethesda, MD, USA
| | - Ambrose Jong
- Children’s Hospital of Los Angeles, Los Angeles, CA, USA
- University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - Tijana Jovanovic-Talisman
- City of Hope Comprehensive Cancer Center, Beckman Research Institute, Department of Molecular Medicine, Duarte, CA, USA
| | - Stephanie Jung
- German Research Center for Environmental Health, Institute for Virology, Munich, Germany
| | - Raghu Kalluri
- University of Texas MD Anderson Cancer Center, Department of Cancer Biology, Metastasis Research Center, Houston, TX, USA
| | - Shin-ichi Kano
- The Johns Hopkins University School of Medicine, Department of Psychiatry and Behavioral Sciences, Baltimore, MD, USA
| | - Sukhbir Kaur
- National Institutes of Health, National Cancer Institute, Center for Cancer Research, Laboratory of Pathology, Bethesda, MD, USA
| | - Yumi Kawamura
- National Cancer Center Research Institute, Tokyo, Japan
- University of Tsukuba, Tsukuba, Japan
| | - Evan T Keller
- University of Michigan, Biointerfaces Institute, Ann Arbor, MI, USA
- University of Michigan, Department of Urology, Ann Arbor, MI, USA
| | - Delaram Khamari
- Semmelweis University, Department of Genetics, Cell- and Immunobiology, Budapest, Hungary
| | - Elena Khomyakova
- École normale supérieure, Paris, France
- Federal Research and Clinical Center of Physical-Chemical Medicine, Moscow, Russia
| | - Anastasia Khvorova
- University of Massachusetts Medical School, RNA Therapeutics Institute, Worcester, MA, USA
| | - Peter Kierulf
- Oslo University Hospital, Department of Medical Biochemistry, Blood Cell Research Group, Oslo, Norway
| | - Kwang Pyo Kim
- Kyung Hee University, Department of Applied Chemistry, Yongin, Korea
| | - Thomas Kislinger
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- University of Toronto, Department of Medical Biophysics, Toronto, Canada
| | | | - David J Klinke
- West Virginia University, Department of Chemical and Biomedical Engineering and WVU Cancer Institute, Morgantown, WV, USA
- West Virginia University, Department of Microbiology Immunology and Cell Biology, Morgantown, WV, USA
| | - Miroslaw Kornek
- German Armed Forces Central Hospital, Department of General, Visceral and Thoracic Surgery, Koblenz, Germany
- Saarland University Medical Center, Department of Medicine II, Homburg, Germany
| | - Maja M Kosanović
- University of Belgrade, Institute for the Application of Nuclear Energy, INEP, Belgrade, Serbia
| | - Árpád Ferenc Kovács
- Semmelweis University, Department of Genetics, Cell- and Immunobiology, Budapest, Hungary
| | | | - Susanne Krasemann
- University Medical Center Hamburg-Eppendorf, Institute of Neuropathology, Hamburg, Germany
| | - Mirja Krause
- Hudson Institute of Medical Research, Melbourne, Australia
| | | | - Gina D Kusuma
- Hudson Institute of Medical Research, Melbourne, Australia
- Monash University, Melbourne, Australia
| | - Sören Kuypers
- Hasselt University, Biomedical Research Institute (BIOMED), Hasselt, Belgium
| | - Saara Laitinen
- Finnish Red Cross Blood Service, Research and Development, Helsinki, Finland
| | - Scott M Langevin
- Cincinnati Cancer Center, Cincinnati, OH, USA
- University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Lucia R Languino
- Thomas Jefferson University, Sidney Kimmel Medical School, Department of Cancer Biology, Philadelphia, PA, USA
| | - Joanne Lannigan
- University of Virginia, Flow Cytometry Core, School of Medicine, Charlottesville, VA, USA
| | - Cecilia Lässer
- University of Gothenburg, Institute of Medicine at Sahlgrenska Academy, Krefting Research Centre, Gothenburg, Sweden
| | - Louise C Laurent
- University of California, San Diego, Department of Obstetrics, Gynecology, and Reproductive Sciences, La Jolla, CA, USA
| | - Gregory Lavieu
- Institut Curie, INSERM U932, PSL Research University, Paris, France
| | | | - Soazig Le Lay
- INSERM U1063, Université d’Angers, CHU d’Angers, Angers, France
| | - Myung-Shin Lee
- Eulji University, School of Medicine, Daejeon, South Korea
| | | | - Debora S Lemos
- Federal University of Paraná, Department of Genetics, Human Molecular Genetics Laboratory, Curitiba, Brazil
| | - Metka Lenassi
- University of Ljubljana, Faculty of Medicine, Institute of Biochemistry, Ljubljana, Slovenia
| | | | - Isaac TS Li
- University of British Columbia Okanagan, Kelowna, Canada
| | - Ke Liao
- University of Nebraska Medical Center, Department of Pharmacology and Experimental Neuroscience, Omaha, NE, USA
| | - Sten F Libregts
- University of Cambridge School of Clinical Medicine, Addenbrooke’s Hospital, Department of Medicine, Cambridge NIHR BRC Cell Phenotyping Hub, Cambridge, UK
| | - Erzsebet Ligeti
- Semmelweis University, Department of Physiology, Budapest, Hungary
| | - Rebecca Lim
- Hudson Institute of Medical Research, Melbourne, Australia
- Monash University, Melbourne, Australia
| | - Sai Kiang Lim
- Institute of Medical Biology (IMB), Agency for Science and Technology (A*STAR), Singapore
| | - Aija Linē
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Karen Linnemannstöns
- University Medical Center Göttingen, Developmental Biochemistry, Göttingen, Germany
- University Medical Center Göttingen, Hematology and Oncology, Göttingen, Germany
| | - Alicia Llorente
- Oslo University Hospital-The Norwegian Radium Hospital, Institute for Cancer Research, Department of Molecular Cell Biology, Oslo, Norway
| | - Catherine A Lombard
- Université Catholique de Louvain, Institut de Recherche Expérimentale et Clinique (IREC), Laboratory of Pediatric Hepatology and Cell Therapy, Brussels, Belgium
| | - Magdalena J Lorenowicz
- Utrecht University, University Medical Center Utrecht, Center for Molecular Medicine & Regenerative Medicine Center, Utrecht, The Netherlands
| | - Ákos M Lörincz
- Semmelweis University, Department of Physiology, Budapest, Hungary
| | - Jan Lötvall
- University of Gothenburg, Institute of Medicine at Sahlgrenska Academy, Krefting Research Centre, Gothenburg, Sweden
| | - Jason Lovett
- Stellenbosch University, Department of Physiological Sciences, Stellenbosch, South Africa
| | - Michelle C Lowry
- Trinity College Dublin, School of Pharmacy and Pharmaceutical Sciences, Panoz Institute & Trinity Biomedical Sciences Institute, Dublin, Ireland
| | - Xavier Loyer
- INSERM UMR-S 970, Paris Cardiovascular Research Center, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Quan Lu
- Harvard University, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Barbara Lukomska
- Mossakowski Medical Research Centre, NeuroRepair Department, Warsaw, Poland
| | - Taral R Lunavat
- K.G. Jebsen Brain Tumor Research Centre, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Sybren LN Maas
- Utrecht University, University Medical Center Utrecht, Department of Neurosurgery, Brain Center Rudolf Magnus, Institute of Neurosciences, Utrecht, The Netherlands
- Utrecht University, University Medical Center Utrecht, Department of Pathology, Utrecht, The Netherlands
| | | | - Antonio Marcilla
- Universitat de València, Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Àrea de Parasitologia, Valencia, Spain
- Universitat de València, Health Research Institute La Fe, Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics, Valencia, Spain
| | - Jacopo Mariani
- Università degli Studi di Milano, Department of Clinical Sciences and Community Health, EPIGET LAB, Milan, Italy
| | | | | | | | | | | | - Mathilde Mathieu
- Institut Curie, INSERM U932, PSL Research University, Paris, France
| | - Suresh Mathivanan
- La Trobe University, La Trobe Institute for Molecular Science, Department of Biochemistry and Genetics, Bundoora, Australia
| | - Marco Maugeri
- University of Gothenburg, Sahlgrenska Academy, Department of Rheumatology and Inflammation Research, Gothenburg, Sweden
| | | | - Mark J McVey
- SickKids Hospital, Department of Anesthesia and Pain Medicine, Toronto, Canada
- University of Toronto, Department of Anesthesia, Toronto, Canada
| | - David G Meckes
- Florida State University College of Medicine, Department of Biomedical Sciences, Tallahassee, FL, USA
| | - Katie L Meehan
- The School of Biomedical Sciences, University of Western Australia, Perth, Australia
| | - Inge Mertens
- University of Antwerp, Centre for Proteomics, Antwerp, Belgium
- Vlaamse Instelling voor Technologisch Onderzoek (VITO), Mol, Belgium
| | - Valentina R Minciacchi
- Georg-Speyer-Haus Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - Andreas Möller
- QIMR Berghofer Medical Research Institute, Herston, Australia
| | - Malene Møller Jørgensen
- Aalborg University Hospital, Department of Clinical Immunology, Aalborg, Denmark
- EVSEARCH.DK, Denmark
| | - Aizea Morales-Kastresana
- National Institutes of Health, National Cancer Institute, Center for Cancer Research, Bethesda, MD, USA
| | | | - François Mullier
- Namur Thrombosis and Hemostasis Center (NTHC), NARILIS, Namur, Belgium
- Université Catholique de Louvain, CHU UCL Namur, Hematology-Hemostasis Laboratory, Yvoir, Belgium
| | - Maurizio Muraca
- University of Padova, Department of Women’s and Children’s Health, Padova, Italy
| | - Luca Musante
- University of Virginia Health System, Department of Medicine, Division of Nephrology, Charlottesville, VA, USA
| | - Veronika Mussack
- Technical University of Munich, TUM School of Life Sciences Weihenstephan, Division of Animal Physiology and Immunology, Freising, Germany
| | - Dillon C Muth
- The Johns Hopkins University School of Medicine, Department of Molecular and Comparative Pathobiology, Baltimore, MD, USA
| | - Kathryn H Myburgh
- Stellenbosch University, Department of Physiological Sciences, Stellenbosch, South Africa
| | - Tanbir Najrana
- Brown University, Women and Infants Hospital, Providence, RI, USA
| | - Muhammad Nawaz
- University of Gothenburg, Sahlgrenska Academy, Department of Rheumatology and Inflammation Research, Gothenburg, Sweden
| | - Irina Nazarenko
- German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Institute for Infection Prevention and Hospital Epidemiology, Freiburg, Germany
| | - Peter Nejsum
- Aarhus University, Department of Clinical Medicine, Aarhus, Denmark
| | - Christian Neri
- Sorbonne Université, Centre National de la Recherche Scientifique, Research Unit Biology of Adaptation and Aging (B2A), Team Compensation in Neurodegenerative and Aging (Brain-C), Paris, France
| | - Tommaso Neri
- University of Pisa, Centro Dipartimentale di Biologia Cellulare Cardio-Respiratoria, Pisa, Italy
| | - Rienk Nieuwland
- Academic Medical Centre of the University of Amsterdam, Department of Clinical Chemistry and Vesicle Observation Centre, Amsterdam, The Netherlands
| | - Leonardo Nimrichter
- Universidade Federal do Rio de Janeiro, Instituto de Microbiologia, Rio de Janeiro, Brazil
| | | | - Esther NM Nolte-’t Hoen
- Utrecht University, Faculty of Veterinary Medicine, Department of Biochemistry and Cell Biology, Utrecht, The Netherlands
| | - Nicole Noren Hooten
- National Institutes of Health, National Institute on Aging, Baltimore, MD, USA
| | - Lorraine O’Driscoll
- Trinity College Dublin, School of Pharmacy and Pharmaceutical Sciences, Panoz Institute & Trinity Biomedical Sciences Institute, Dublin, Ireland
| | - Tina O’Grady
- University of Liège, GIGA-R(MBD), PSI Laboratory, Liège, Belgium
| | - Ana O’Loghlen
- Queen Mary University of London, Blizard Institute, Epigenetics & Cellular Senescence Group, London, UK
| | - Takahiro Ochiya
- National Cancer Center Research Institute, Division of Molecular and Cellular Medicine, Tokyo, Japan
| | - Martin Olivier
- McGill University, The Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Alberto Ortiz
- IIS-Fundacion Jimenez Diaz-UAM, Department of Nephrology and Hypertension, Madrid, Spain
- Spanish Kidney Research Network, REDINREN, Madrid, Spain
- Universidad Autónoma de Madrid, School of Medicine, Department of Medicine, Madrid, Spain
| | - Luis A Ortiz
- Graduate School of Public Health at the University of Pittsburgh, Division of Occupational and Environmental Medicine, Pittsburgh, PA, USA
| | | | - Ole Østergaard
- Statens Serum Institut, Department of Autoimmunology and Biomarkers, Copenhagen, Denmark
- University of Copenhagen, Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Protein Research, Copenhagen, Denmark
| | - Matias Ostrowski
- University of Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Buenos Aires, Argentina
| | - Jaesung Park
- POSTECH (Pohang University of Science and Technology), Department of Life Sciences, Pohang, South Korea
| | - D. Michiel Pegtel
- Amsterdam University Medical Centers, Department of Pathology, Amsterdam, The Netherlands
| | - Hector Peinado
- Spanish National Cancer Research Center (CNIO), Molecular Oncology Programme, Microenvironment and Metastasis Laboratory, Madrid, Spain
| | - Francesca Perut
- IRCCS - Istituto Ortopedico Rizzoli, Laboratory for Orthopaedic Pathophysiology and Regenerative Medicine, Bologna, Italy
| | - Michael W Pfaffl
- Technical University of Munich, TUM School of Life Sciences Weihenstephan, Division of Animal Physiology and Immunology, Freising, Germany
| | - Donald G Phinney
- The Scripps Research Institute-Scripps Florida, Department of Molecular Medicine, Jupiter, FL, USA
| | - Bartijn CH Pieters
- Radboud University Medical Center, Department of Rheumatology, Nijmegen, The Netherlands
| | - Ryan C Pink
- Oxford Brookes University, Department of Biological and Medical Sciences, Oxford, UK
| | - David S Pisetsky
- Duke University Medical Center, Departments of Medicine and Immunology, Durham, NC, USA
- Durham VAMC, Medical Research Service, Durham, NC, USA
| | | | - Iva Polakovicova
- Pontificia Universidad Católica de Chile, Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
- Pontificia Universidad Católica de Chile, Faculty of Medicine, Department of Hematology-Oncology, Santiago, Chile
| | - Ivan KH Poon
- La Trobe University, La Trobe Institute for Molecular Science, Department of Biochemistry and Genetics, Bundoora, Australia
| | - Bonita H Powell
- The Johns Hopkins University School of Medicine, Department of Molecular and Comparative Pathobiology, Baltimore, MD, USA
| | | | - Lynn Pulliam
- University of California, San Francisco, CA, USA
- Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Peter Quesenberry
- The Warren Alpert Medical School of Brown University, Department of Medicine, Providence, RI, USA
| | - Annalisa Radeghieri
- CSGI - Research Center for Colloids and Nanoscience, Florence, Italy
- University of Brescia, Department of Molecular and Translational Medicine, Brescia, Italy
| | - Robert L Raffai
- Department of Veterans Affairs, San Francisco, CA, USA
- University of California, San Francisco, CA, USA
| | - Stefania Raimondo
- University of Palermo, Department of Biopathology and Medical Biotechnologies, Palermo, Italy
| | - Janusz Rak
- McGill University, Montreal, Canada
- McGill University, The Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Marcel I Ramirez
- Instituto Oswaldo Cruz, Rio de Janeiro, Brazil
- Universidade Federal de Paraná, Paraná, Brazil
| | - Graça Raposo
- Institut Curie, CNRS UMR144, PSL Research University, Paris, France
| | - Morsi S Rayyan
- University of Michigan Medical School, Ann Arbor, MI, USA
| | - Neta Regev-Rudzki
- Weizmann Institute of Science, Department of Biomolecular Sciences, Rehovot, Israel
| | - Franz L Ricklefs
- University Medical Center Hamburg-Eppendorf, Department of Neurosurgery, Hamburg, Germany
| | - Paul D Robbins
- University of Minnesota Medical School, Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, Minneapolis, MN, USA
| | - David D Roberts
- National Institutes of Health, National Cancer Institute, Center for Cancer Research, Laboratory of Pathology, Bethesda, MD, USA
| | | | - Eva Rohde
- Paracelsus Medical University, Department of Transfusion Medicine, Salzburg, Austria
- Paracelsus Medical University, GMP Unit, Salzburg, Austria
- Spinal Cord Injury & Tissue Regeneration Center Salzburg (SCI-TReCS), Salzburg, Austria
| | - Sophie Rome
- University of Lyon, Lyon-Sud Faculty of Medicine, CarMeN Laboratory (UMR INSERM 1060-INRA 1397), Pierre-Bénite, France
| | - Kasper MA Rouschop
- Maastricht University, GROW, School for Oncology and Developmental Biology, Maastricht Radiation Oncology (MaastRO) Lab, Maastricht, The Netherlands
| | - Aurelia Rughetti
- Sapienza University of Rome, Department of Experimental Medicine, Rome, Italy
| | | | - Paula Saá
- American Red Cross, Scientific Affairs, Gaithersburg, MD, USA
| | - Susmita Sahoo
- Icahn School of Medicine at Mount Sinai, Department of Medicine, Cardiology, New York City, NY, USA
| | - Edison Salas-Huenuleo
- Advanced Center for Chronic Diseases, Santiago, Chile
- University of Chile, Faculty of Chemical and Pharmaceutical Science, Laboratory of Nanobiotechnology and Nanotoxicology, Santiago, Chile
| | - Catherine Sánchez
- Clínica las Condes, Extracellular Vesicles in Personalized Medicine Group, Santiago, Chile
| | - Julie A Saugstad
- Oregon Health & Science University, Department of Anesthesiology & Perioperative Medicine, Portland, OR, USA
| | - Meike J Saul
- Technische Universität Darmstadt, Department of Biology, Darmstadt, Germany
| | - Raymond M Schiffelers
- University Medical Center Utrecht, Laboratory for Clinical Chemistry & Hematology, Utrecht, The Netherlands
| | - Raphael Schneider
- University of Toronto, Department of Laboratory Medicine and Pathobiology, Toronto, Canada
- University of Toronto, Department of Medicine, Division of Neurology, Toronto, Canada
| | - Tine Hiorth Schøyen
- The Johns Hopkins University School of Medicine, Department of Molecular and Comparative Pathobiology, Baltimore, MD, USA
| | | | - Eriomina Shahaj
- Fondazione IRCCS Istituto Nazionale dei Tumori, Unit of Immunotherapy of Human Tumors, Milan, Italy
| | - Shivani Sharma
- University of California, Los Angeles, California NanoSystems Institute, Los Angeles, CA, USA
- University of California, Los Angeles, Department of Pathology and Laboratory Medicine, Los Angeles, CA, USA
- University of California, Los Angeles, Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Olga Shatnyeva
- AstraZeneca, Discovery Sciences, IMED Biotech Unit, Gothenburg, Sweden
| | - Faezeh Shekari
- Royan Institute for Stem Cell Biology and Technology, ACECR, Cell Science Research Center, Department of Stem Cells and Developmental Biology, Tehran, Iran
| | - Ganesh Vilas Shelke
- University of Gothenburg, Institute of Clinical Sciences, Department of Surgery, Sahlgrenska Cancer Center, Gothenburg, Sweden
- University of Gothenburg, Institute of Medicine at Sahlgrenska Academy, Krefting Research Centre, Gothenburg, Sweden
| | - Ashok K Shetty
- Research Service, Olin E. Teague Veterans’ Medical Center, Temple, TX, USA
- Texas A&M University College of Medicine, Institute for Regenerative Medicine and Department of Molecular and Cellular Medicine, College Station, TX, USA
| | | | - Pia R-M Siljander
- University of Helsinki, EV Core Facility, Helsinki, Finland
- University of Helsinki, Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences Research Programme, EV group, Helsinki, Finland
| | - Andreia M Silva
- INEB - Instituto de Engenharia Biomédica, Porto, Portugal
- University of Porto, i3S-Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- University of Porto, ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Porto, Portugal
| | - Agata Skowronek
- Maria Sklodowska-Curie Institute - Oncology Center, Gliwice Branch, Gliwice, Poland
| | - Orman L Snyder
- Kansas State University, College of Veterinary Medicine, Manhattan, KS, USA
| | | | - Barbara W Sódar
- Semmelweis University, Department of Genetics, Cell- and Immunobiology, Budapest, Hungary
| | - Carolina Soekmadji
- QIMR Berghofer Medical Research Institute, Herston, Australia
- The University of Queensland, Brisbane, Australia
| | - Javier Sotillo
- James Cook University, Australian Institute of Tropical Health and Medicine, Centre for Biodiscovery and Molecular Development of Therapeutics, Cairns, Australia
| | | | - Willem Stoorvogel
- Utrecht University, Faculty of Veterinary Medicine, Department of Biochemistry and Cell Biology, Utrecht, The Netherlands
| | - Shannon L Stott
- Harvard Medical School, Department of Medicine, Boston, MA, USA
- Massachusetts General Cancer Center, Boston, MA, USA
| | - Erwin F Strasser
- FAU Erlangen-Nuremberg, Transfusion and Haemostaseology Department, Erlangen, Germany
| | - Simon Swift
- University of Auckland, Department of Molecular Medicine and Pathology, Auckland, New Zealand
| | - Hidetoshi Tahara
- Hiroshima University, Institute of Biomedical & Health Sciences, Department of Cellular and Molecular Biology, Hiroshima, Japan
| | - Muneesh Tewari
- University of Michigan, Biointerfaces Institute, Ann Arbor, MI, USA
- University of Michigan, Department of Biomedical Engineering, Ann Arbor, MI, USA
- University of Michigan, Department of Internal Medicine - Hematology/Oncology Division, Ann Arbor, MI, USA
| | - Kate Timms
- University of Manchester, Manchester, UK
| | - Swasti Tiwari
- Georgetown University, Department of Medicine, Washington, DC, USA
- Sanjay Gandhi Postgraduate Institute of Medical Sciences, Department of Molecular Medicine & Biotechnology, Lucknow, India
| | - Rochelle Tixeira
- La Trobe University, La Trobe Institute for Molecular Science, Department of Biochemistry and Genetics, Bundoora, Australia
| | - Mercedes Tkach
- Institut Curie, INSERM U932, PSL Research University, Paris, France
| | - Wei Seong Toh
- National University of Singapore, Faculty of Dentistry, Singapore
| | - Richard Tomasini
- INSERM U1068, Aix Marseille University, CNRS UMR7258, Marseille, France
| | | | - Juan Pablo Tosar
- Institut Pasteur de Montevideo, Functional Genomics Unit, Montevideo, Uruguay
- Universidad de la República, Faculty of Science, Nuclear Research Center, Analytical Biochemistry Unit, Montevideo, Uruguay
| | | | - Lorena Urbanelli
- University of Perugia, Department of Chemistry, Biology and Biotechnology, Perugia, Italy
| | - Pieter Vader
- University Medical Center Utrecht, Laboratory for Clinical Chemistry & Hematology, Utrecht, The Netherlands
| | - Bas WM van Balkom
- University Medical Center Utrecht, Department of Nephrology and Hypertension, Utrecht, The Netherlands
| | - Susanne G van der Grein
- Utrecht University, Faculty of Veterinary Medicine, Department of Biochemistry and Cell Biology, Utrecht, The Netherlands
| | - Jan Van Deun
- Cancer Research Institute Ghent, Ghent, Belgium
- Ghent University, Department of Radiation Oncology and Experimental Cancer Research, Laboratory of Experimental Cancer Research, Ghent, Belgium
| | - Martijn JC van Herwijnen
- Utrecht University, Faculty of Veterinary Medicine, Department of Biochemistry and Cell Biology, Utrecht, The Netherlands
| | | | | | - Martin E van Royen
- Department of Pathology, Erasmus MC, Erasmus Optical Imaging Centre, Rotterdam, The Netherlands
| | | | - M Helena Vasconcelos
- IPATIMUP, Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
- University of Porto, Faculty of Pharmacy (FFUP), Porto, Portugal
- University of Porto, i3S-Instituto de Investigação e Inovação em Saúde, Porto, Portugal
| | - Ivan J Vechetti
- University of Kentucky, College of Medicine, Department of Physiology, Lexington, KY, USA
| | - Tiago D Veit
- Universidade Federal do Rio Grande do Sul, Instituto de Ciências Básicas da Saúde, Departamento de Microbiologia, Imunologia e Parasitologia, Porto Alegre, Brazil
| | - Laura J Vella
- The Florey Institute of Neuroscience and Mental Health, Melbourne, Australia
- The University of Melbourne, The Department of Medicine, Melbourne, Australia
| | - Émilie Velot
- UMR 7365 CNRS-Université de Lorraine, Vandœuvre-lès-Nancy, France
| | | | - Beate Vestad
- Oslo University Hospital Rikshospitalet, Research Institute of Internal Medicine, Oslo, Norway
- Regional Research Network on Extracellular Vesicles, RRNEV, Oslo, Norway
- University of Oslo, Institute of Clinical Medicine, Oslo, Norway
| | - Jose L Viñas
- Kidney Research Centre, Ottawa, Canada
- Ottawa Hospital Research Institute, Ottawa, Canada
- University of Ottawa, Ottawa, Canada
| | - Tamás Visnovitz
- Semmelweis University, Department of Genetics, Cell- and Immunobiology, Budapest, Hungary
| | - Krisztina V Vukman
- Semmelweis University, Department of Genetics, Cell- and Immunobiology, Budapest, Hungary
| | - Jessica Wahlgren
- University of Gothenburg, The Sahlgrenska Academy, Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Mölndal, Sweden
| | - Dionysios C Watson
- Case Western Reserve University, Department of Medicine, Cleveland, OH, USA
- University Hospitals Cleveland Medical Center, Department of Medicine, Cleveland, OH, USA
| | - Marca HM Wauben
- Utrecht University, Faculty of Veterinary Medicine, Department of Biochemistry and Cell Biology, Utrecht, The Netherlands
| | - Alissa Weaver
- Vanderbilt University School of Medicine, Department of Cell and Developmental Biology, Nashville, TN, USA
| | | | - Viktoria Weber
- Danube University Krems, Department for Biomedical Research and Christian Doppler Laboratory for Innovative Therapy Approaches in Sepsis, Krems an der Donau, Austria
| | - Ann M Wehman
- University of Würzburg, Rudolf Virchow Center, Würzburg, Germany
| | - Daniel J Weiss
- The University of Vermont Medical Center, Department of Medicine, Burlington, VT, USA
| | - Joshua A Welsh
- National Institutes of Health, National Cancer Institute, Center for Cancer Research, Bethesda, MD, USA
| | - Sebastian Wendt
- University Hospital RWTH Aachen, Department of Thoracic and Cardiovascular Surgery, Aachen, Germany
| | - Asa M Wheelock
- Karolinska Institute, Department of Medicine and Center for Molecular Medicine, Respiratory Medicine Unit, Stockholm, Sweden
| | - Zoltán Wiener
- Semmelweis University, Department of Genetics, Cell- and Immunobiology, Budapest, Hungary
| | - Leonie Witte
- University Medical Center Göttingen, Developmental Biochemistry, Göttingen, Germany
- University Medical Center Göttingen, Hematology and Oncology, Göttingen, Germany
| | - Joy Wolfram
- Chinese Academy of Sciences, Wenzhou Institute of Biomaterials and Engineering, Wenzhou, China
- Houston Methodist Research Institute, Department of Nanomedicine, Houston, TX, USA
- Mayo Clinic, Department of Transplantation Medicine/Department of Physiology and Biomedical Engineering, Jacksonville, FL, USA
| | - Angeliki Xagorari
- George Papanicolaou Hospital, Public Cord Blood Bank, Department of Haematology - BMT Unit, Thessaloniki, Greece
| | - Patricia Xander
- Universidade Federal de São Paulo Campus Diadema, Departamento de Ciências Farmacêuticas, Laboratório de Imunologia Celular e Bioquímica de Fungos e Protozoários, São Paulo, Brazil
| | - Jing Xu
- BC Cancer, Canada’s Michael Smith Genome Sciences Centre, Vancouver, Canada
- Simon Fraser University, Department of Molecular Biology and Biochemistry, Burnaby, Canada
| | - Xiaomei Yan
- Xiamen University, Department of Chemical Biology, Xiamen, China
| | - María Yáñez-Mó
- Centro de Biología Molecular Severo Ochoa, Instituto de Investigación Sanitaria la Princesa (IIS-IP), Madrid, Spain
- Universidad Autónoma de Madrid, Departamento de Biología Molecular, Madrid, Spain
| | - Hang Yin
- Tsinghua University, School of Pharmaceutical Sciences, Beijing, China
| | - Yuana Yuana
- Technical University Eindhoven, Faculty Biomedical Technology, Eindhoven, The Netherlands
| | - Valentina Zappulli
- University of Padova, Department of Comparative Biomedicine and Food Science, Padova, Italy
| | - Jana Zarubova
- Institute of Physiology CAS, Department of Biomaterials and Tissue Engineering, BIOCEV, Vestec, Czech Republic
- Institute of Physiology CAS, Department of Biomaterials and Tissue Engineering, Prague, Czech Republic
- University of California, Los Angeles, Department of Bioengineering, Los Angeles, CA, USA
| | - Vytautas Žėkas
- Vilnius University, Institute of Biomedical Sciences, Department of Physiology, Biochemistry, Microbiology and Laboratory Medicine, Vilnius, Lithuania
| | - Jian-ye Zhang
- Guangzhou Medical University, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology, Guangzhou, China
| | - Zezhou Zhao
- The Johns Hopkins University School of Medicine, Department of Molecular and Comparative Pathobiology, Baltimore, MD, USA
| | - Lei Zheng
- Nanfang Hospital, Southern Medical University, Department of Clinical Laboratory Medicine, Guangzhou, China
| | | | - Antje M Zickler
- Karolinska Institute, Clinical Research Center, Unit for Molecular Cell and Gene Therapy Science, Stockholm, Sweden
| | - Pascale Zimmermann
- Aix-Marseille Université, Institut Paoli-Calmettes, INSERM U1068, CNRS UMR7258, Centre de Recherche en Cancérologie de Marseille, Marseille, France
- KU Leuven (Leuven University), Department of Human Genetics, Leuven, Belgium
| | - Angela M Zivkovic
- University of California, Davis, Department of Nutrition, Davis, CA, USA
| | | | - Ewa K Zuba-Surma
- Jagiellonian University, Faculty of Biochemistry, Biophysics and Biotechnology, Department of Cell Biology, Kraków, Poland
| |
Collapse
|
49
|
Wang W, Luo J, Wang S. Recent Progress in Isolation and Detection of Extracellular Vesicles for Cancer Diagnostics. Adv Healthc Mater 2018; 7:e1800484. [PMID: 30009550 DOI: 10.1002/adhm.201800484] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 06/20/2018] [Indexed: 12/21/2022]
Abstract
Extracellular vesicles (EVs) are emerging as one of the many new and promising biomarkers for liquid biopsy of cancer due to their loading capability of some specific proteins and nucleic acids that are closely associated with cancer states. As such, the isolation and detection of cancer-derived EVs offer important information in noninvasive diagnosis of early-stage cancer and real-time monitoring of cancer development. In light of the importance of EVs, over the last decade, researchers have made remarkable innovations to advance the development of EV isolation and detection methods by taking advantage of microfluidics, biomolecule probes, nanomaterials, surface plasmon, optics, and so on. This review introduces the basic properties of EVs and common cancer-derived EV ingredients, and provides a comprehensive overview of EV isolation and detection strategies, with emphasis on liquid biopsies of EVs for cancer diagnostics.
Collapse
Affiliation(s)
- Wenshuo Wang
- CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science; Technical Institute of Physics and Chemistry; Chinese Academy of Sciences; Beijing 100190 P. R. China
- University of Chinese Academy of Sciences; Beijing 100049 P. R. China
| | - Jing Luo
- CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science; Technical Institute of Physics and Chemistry; Chinese Academy of Sciences; Beijing 100190 P. R. China
- University of Chinese Academy of Sciences; Beijing 100049 P. R. China
| | - Shutao Wang
- CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science; Technical Institute of Physics and Chemistry; Chinese Academy of Sciences; Beijing 100190 P. R. China
- University of Chinese Academy of Sciences; Beijing 100049 P. R. China
| |
Collapse
|
50
|
High degradation and no bioavailability of artichoke miRNAs assessed using an in vitro digestion/Caco-2 cell model. Nutr Res 2018; 60:68-76. [PMID: 30527261 DOI: 10.1016/j.nutres.2018.08.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 08/22/2018] [Accepted: 08/31/2018] [Indexed: 01/21/2023]
Abstract
Although the cross-kingdom transfer of vegetable miRNAs (miRNAs) in mammalian species, including humans, is still controversial, recent studies have rejected this theory. Based on these recent studies, we hypothesized that artichoke-derived miRNAs (cca-miRNAs) are not adsorbed into human intestinal cells after cooking and in vitro digestion. In order to test this hypothesis, we evaluated miRNA (cca-miRNAs) in the edible part of globe artichokes (head portion), after cooking and digestion by an in vitro digestion system. The cca-miRNA levels were analyzed by real-time PCR (RT-qPCR), and those that withstood cooking and digestion conditions were further analyzed for their bioavailability using an in vitro system (Caco-2/TC7 cell clone). We detected 20 cca-miRNAs after cooking, 5 of which were statistically down-regulated in comparison with uncooked samples. Only 4 cca-miRNAs were found after in vitro digestion. By using scanning electron microscopy (SEM), we also evaluated the extracellular vesicles (EVs) in homogenized artichoke as possible miRNA transporters. However, approximately 81% were degraded after cooking, while the remaining EVs had changed shape from round to elliptical. Finally, we detected no cell-free cca-miRNAs, miRNAs bound to protein complex, and no cca-miRNAs encapsulated in EVs inside Caco-2 cells or in basolateral medium after bioavailability experiments. In conclusion, the data from the present study agrees with recent findings that the human small intestine does not uptake dietary miRNAs from raw or cooked artichoke heads.
Collapse
|