1
|
Simon L, Constanzo J, Terraza-Aguirre C, Ibn Elfekih Z, Berthelot J, Benkhaled BT, Haute T, Pednekar K, Clark K, Emerson SJ, Atis S, Benedetti C, Langlois S, Marquant A, Prakash J, Wang A, Devoisselle JM, Montier T, Djouad F, Pouget JP, Lapinte V, Morille M. Surface modification of extracellular vesicles with polyoxazolines to enhance their plasma stability and tumor accumulation. Biomaterials 2025; 313:122748. [PMID: 39180918 DOI: 10.1016/j.biomaterials.2024.122748] [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: 04/26/2024] [Revised: 07/23/2024] [Accepted: 08/05/2024] [Indexed: 08/27/2024]
Abstract
Extracellular vesicles (EVs) are future promising therapeutics, but their instability in vivo after administration remains an important barrier to their further development. Many groups evaluated EV surface modification strategies to add a targeting group with the aim of controlling EV biodistribution. Conversely, fewer groups focused on their stabilization to obtain "stealth" allogenic EVs. Modulating their stabilization and biodistribution is an essential prerequisite for their development as nano-therapeutics. Here, we explored polyoxazolines with lipid anchors association to the EV membrane (POxylation as an alternative to PEGylation) to stabilize EVs in plasma and control their biodistribution, while preserving their native properties. We found that this modification maintained and seemed to potentiate the immunomodulatory properties of EVs derived from mesenchymal stem/stromal cells (MSC). Using a radiolabeling protocol to track EVs at a therapeutically relevant concentration in vivo, we demonstrated that POxylation is a promising option to stabilize EVs in plasma because it increased EV half-life by 6 fold at 6 h post-injection. Moreover, EV accumulation in tumors was higher after POxylation than after PEGylation.
Collapse
Affiliation(s)
- L Simon
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France
| | - J Constanzo
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut Régional Du Cancer de Montpellier (ICM), Montpellier, France
| | | | - Z Ibn Elfekih
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France
| | - J Berthelot
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France
| | - B T Benkhaled
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France
| | - T Haute
- Univ Brest, INSERM, EFS, UMR 1078, GGB, F-29200, Brest, France
| | - K Pednekar
- Department of Advanced Organ Bioengineering and Therapeutics, Engineered Therapeutics Section, Technical Medical Centre, University of Twente, 7500 AE, Enschede, the Netherlands
| | - K Clark
- Center for Surgical Bioengineering, Deparment of Surgery, University of California Davis School of Medicine, Sacramento, CA, USA; Institute for Pediatric Regenerative Medicine, Shriners Children's Northern California, Sacramento, CA, USA
| | - S J Emerson
- Center for Surgical Bioengineering, Deparment of Surgery, University of California Davis School of Medicine, Sacramento, CA, USA; Institute for Pediatric Regenerative Medicine, Shriners Children's Northern California, Sacramento, CA, USA
| | - S Atis
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut Régional Du Cancer de Montpellier (ICM), Montpellier, France
| | - C Benedetti
- Montpellier Ressources Imagerie, BioCampus, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - S Langlois
- Montpellier Ressources Imagerie, BioCampus, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - A Marquant
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France
| | - J Prakash
- Department of Advanced Organ Bioengineering and Therapeutics, Engineered Therapeutics Section, Technical Medical Centre, University of Twente, 7500 AE, Enschede, the Netherlands
| | - A Wang
- Center for Surgical Bioengineering, Deparment of Surgery, University of California Davis School of Medicine, Sacramento, CA, USA; Institute for Pediatric Regenerative Medicine, Shriners Children's Northern California, Sacramento, CA, USA
| | - J M Devoisselle
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France
| | - T Montier
- Univ Brest, INSERM, EFS, UMR 1078, GGB, F-29200, Brest, France; CHU de Brest, Service de Génétique Médicale et de Biologie de La Reproduction, Centre de Référence des Maladies Rares Maladies Neuromusculaires, 29200, Brest, France
| | - F Djouad
- IRMB, University of Montpellier, INSERM, 34295, Montpellier, France; Clinical Immunology and Osteoarticular Disease Therapeutic Unit, Department of Rheumatology, CHU Montpellier, 34095, Montpellier, France
| | - J P Pouget
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut Régional Du Cancer de Montpellier (ICM), Montpellier, France
| | - V Lapinte
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France
| | - Marie Morille
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France; Institut Universitaire de France (IUF), Paris, France.
| |
Collapse
|
2
|
Sharma N, Angori S, Sandberg A, Mermelekas G, Lehtiö J, Wiklander OPB, Görgens A, Andaloussi SE, Eriksson H, Pernemalm M. Defining the Soluble and Extracellular Vesicle Protein Compartments of Plasma Using In-Depth Mass Spectrometry-Based Proteomics. J Proteome Res 2024; 23:4114-4127. [PMID: 39141927 PMCID: PMC11385381 DOI: 10.1021/acs.jproteome.4c00490] [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] [Indexed: 08/16/2024]
Abstract
Plasma-derived extracellular vesicles (pEVs) are a potential source of diseased biomarker proteins. However, characterizing the pEV proteome is challenging due to its relatively low abundance and difficulties in enrichment. This study presents a streamlined workflow to identify EV proteins from cancer patient plasma using minimal sample input. Starting with 400 μL of plasma, we generated a comprehensive pEV proteome using size exclusion chromatography (SEC) combined with HiRIEF prefractionation-based mass spectrometry (MS). First, we compared the performance of HiRIEF and long gradient MS workflows using control pEVs, quantifying 2076 proteins with HiRIEF. In a proof-of-concept study, we applied SEC-HiRIEF-MS to a small cohort (12) of metastatic lung adenocarcinoma (LUAD) and malignant melanoma (MM) patients. We also analyzed plasma samples from the same patients to study the relationship between plasma and pEV proteomes. We identified and quantified 1583 proteins in cancer pEVs and 1468 proteins in plasma across all samples. While there was substantial overlap, the pEV proteome included several unique EV markers and cancer-related proteins. Differential analysis revealed 30 DEPs in LUAD vs the MM group, highlighting the potential of pEVs as biomarkers. This work demonstrates the utility of a prefractionation-based MS for comprehensive pEV proteomics and EV biomarker discovery. Data are available via ProteomeXchange with the identifiers PXD039338 and PXD038528.
Collapse
Affiliation(s)
- Nidhi Sharma
- Department of Oncology-Pathology, Karolinska Institute, 171 77 Stockholm, Sweden
- Science for Life Laboratory, Solna, Tomtebodavägen 23, 171 65 Solna, Sweden
| | - Silvia Angori
- Department of Oncology-Pathology, Karolinska Institute, 171 77 Stockholm, Sweden
| | - AnnSofi Sandberg
- Department of Oncology-Pathology, Karolinska Institute, 171 77 Stockholm, Sweden
- Science for Life Laboratory, Solna, Tomtebodavägen 23, 171 65 Solna, Sweden
| | - Georgios Mermelekas
- Department of Oncology-Pathology, Karolinska Institute, 171 77 Stockholm, Sweden
- Science for Life Laboratory, Solna, Tomtebodavägen 23, 171 65 Solna, Sweden
| | - Janne Lehtiö
- Department of Oncology-Pathology, Karolinska Institute, 171 77 Stockholm, Sweden
- Science for Life Laboratory, Solna, Tomtebodavägen 23, 171 65 Solna, Sweden
| | - Oscar P B Wiklander
- Theme Cancer, Skin Cancer Center, Karolinska University Hospital, 171 77 Solna, Sweden
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institute, 171 76 Solna, Sweden
| | - André Görgens
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institute, 171 76 Solna, Sweden
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, 45141 Essen, Germany
| | - Samir El Andaloussi
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institute, 171 76 Solna, Sweden
| | - Hanna Eriksson
- Department of Oncology-Pathology, Karolinska Institute, 171 77 Stockholm, Sweden
- Theme Cancer, Skin Cancer Center, Karolinska University Hospital, 171 77 Solna, Sweden
- Science for Life Laboratory, Solna, Tomtebodavägen 23, 171 65 Solna, Sweden
| | - Maria Pernemalm
- Department of Oncology-Pathology, Karolinska Institute, 171 77 Stockholm, Sweden
- Science for Life Laboratory, Solna, Tomtebodavägen 23, 171 65 Solna, Sweden
| |
Collapse
|
3
|
Senesi G, Guerricchio L, Ghelardoni M, Bertola N, Rebellato S, Grinovero N, Bartolucci M, Costa A, Raimondi A, Grange C, Bolis S, Massa V, Paladini D, Coviello D, Pandolfi A, Bussolati B, Petretto A, Fazio G, Ravera S, Barile L, Balbi C, Bollini S. Extracellular vesicles from II trimester human amniotic fluid as paracrine conveyors counteracting oxidative stress. Redox Biol 2024; 75:103241. [PMID: 38901103 PMCID: PMC11253147 DOI: 10.1016/j.redox.2024.103241] [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: 04/20/2024] [Revised: 06/07/2024] [Accepted: 06/15/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND We previously demonstrated that the human amniotic fluid (hAF) from II trimester of gestation is a feasible source of stromal progenitors (human amniotic fluid stem cells, hAFSC), with significant paracrine potential for regenerative medicine. Extracellular vesicles (EVs) separated and concentrated from hAFSC secretome can deliver pro-survival, proliferative, anti-fibrotic and cardioprotective effects in preclinical models of skeletal and cardiac muscle injury. While hAFSC-EVs isolation can be significantly influenced by in vitro cell culture, here we profiled EVs directly concentrated from hAF as an alternative option and investigated their paracrine potential against oxidative stress. METHODS II trimester hAF samples were obtained as leftover material from prenatal diagnostic amniocentesis following written informed consent. EVs were separated by size exclusion chromatography and concentrated by ultracentrifugation. hAF-EVs were assessed by nanoparticle tracking analysis, transmission electron microscopy, Western Blot, and flow cytometry; their metabolic activity was evaluated by oximetric and luminometric analyses and their cargo profiled by proteomics and RNA sequencing. hAF-EV paracrine potential was tested in preclinical in vitro models of oxidative stress and dysfunction on murine C2C12 cells and on 3D human cardiac microtissue. RESULTS Our protocol resulted in a yield of 6.31 ± 0.98 × 109 EVs particles per hAF milliliter showing round cup-shaped morphology and 209.63 ± 6.10 nm average size, with relevant expression of CD81, CD63 and CD9 tetraspanin markers. hAF-EVs were enriched in CD133/1, CD326, CD24, CD29, and SSEA4 and able to produce ATP by oxygen consumption. While oxidative stress significantly reduced C2C12 survival, hAF-EV priming resulted in significant rescue of cell viability, with notable recovery of ATP synthesis and concomitant reduction of cell damage and lipid peroxidation activity. 3D human cardiac microtissues treated with hAF-EVs and experiencing H2O2 stress and TGFβ stimulation showed improved survival with a remarkable decrease in the onset of fibrosis. CONCLUSIONS Our results suggest that leftover samples of II trimester human amniotic fluid can represent a feasible source of EVs to counteract oxidative damage on target cells, thus offering a novel candidate therapeutic option to counteract skeletal and cardiac muscle injury.
Collapse
Affiliation(s)
- Giorgia Senesi
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino and Laboratories for Traslational Research Ente Ospedaliero Cantonale, CH-6500, Bellinzona, Switzerland; Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera Italiana, CH-6900, Lugano, Switzerland
| | - Laura Guerricchio
- Department of Experimental Medicine (DIMES), University of Genova, 16132, Genova, Italy
| | | | - Nadia Bertola
- IRCCS Ospedale Policlinico San Martino, 16132, Genova, Italy
| | - Stefano Rebellato
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, 20900, Monza, Italy; School of Medicine and Surgery, University of Milano-Bicocca, 20900, Monza, Italy
| | - Nicole Grinovero
- Core Facilities - Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, 16147, Genova, Italy
| | - Martina Bartolucci
- Core Facilities - Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, 16147, Genova, Italy
| | - Ambra Costa
- IRCCS Ospedale Policlinico San Martino, 16132, Genova, Italy
| | - Andrea Raimondi
- Institute for Research in Biomedicine, Università della Svizzera Italiana, CH-6500, Bellinzona, Switzerland
| | - Cristina Grange
- VEXTRA Facility and Department of Medical Sciences, University of Turin, 10126, Turin, Italy
| | - Sara Bolis
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino and Laboratories for Traslational Research Ente Ospedaliero Cantonale, CH-6500, Bellinzona, Switzerland
| | - Valentina Massa
- Department of Health Sciences, University of Milan, 20146, Milan, Italy
| | - Dario Paladini
- Fetal Medicine and Surgery Unit, IRCCS Istituto Giannina Gaslini, 16147, Genova, Italy
| | - Domenico Coviello
- Human Genetics Laboratory, IRCCS Istituto Giannina Gaslini, 16147, Genova, Italy
| | - Assunta Pandolfi
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio" Chieti-Pescara and Center for Advanced Studies and Technology - CAST, 66100, Chieti, Italy
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126, Turin, Italy
| | - Andrea Petretto
- Core Facilities - Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, 16147, Genova, Italy
| | - Grazia Fazio
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, 20900, Monza, Italy; School of Medicine and Surgery, University of Milano-Bicocca, 20900, Monza, Italy
| | - Silvia Ravera
- Department of Experimental Medicine (DIMES), University of Genova, 16132, Genova, Italy
| | - Lucio Barile
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino and Laboratories for Traslational Research Ente Ospedaliero Cantonale, CH-6500, Bellinzona, Switzerland; Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera Italiana, CH-6900, Lugano, Switzerland.
| | - Carolina Balbi
- Center for Molecular Cardiology, University of Zurich, 8952, Schlieren, Switzerland; Department of Internal Medicine, Cantonal Hospital Baden, Baden, Switzerland.
| | - Sveva Bollini
- Department of Experimental Medicine (DIMES), University of Genova, 16132, Genova, Italy; IRCCS Ospedale Policlinico San Martino, 16132, Genova, Italy.
| |
Collapse
|
4
|
Chiabotto G, Semnani A, Ceccotti E, Guenza M, Camussi G, Bruno S. Mesenchymal Stromal Cell-Derived Extracellular Vesicles for Reversing Hepatic Fibrosis in 3D Liver Spheroids. Biomedicines 2024; 12:1849. [PMID: 39200313 PMCID: PMC11351945 DOI: 10.3390/biomedicines12081849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/08/2024] [Accepted: 08/12/2024] [Indexed: 09/02/2024] Open
Abstract
Hepatic fibrosis, arising from prolonged liver injury, entails the activation of hepatic stellate cells (HSCs) into myofibroblast-like cells expressing alpha-smooth muscle actin (α-SMA), thereby driving extracellular matrix deposition and fibrosis progression. Strategies targeting activated HSC reversal and hepatocyte regeneration show promise for fibrosis management. Previous studies suggest that extracellular vesicles (EVs) from mesenchymal stromal cells (MSCs) can suppress HSC activation, but ensuring EV purity is essential for clinical use. This study investigated the effects of MSC-derived EVs cultured in chemically defined conditions on liver spheroids and activated HSCs. Umbilical cord- and bone marrow-derived MSCs were expanded in chemically defined media, and EVs were isolated using filtration and differential ultracentrifugation. The impact of MSC-EVs was evaluated on liver spheroids generated in Sphericalplate 5D™ and on human HSCs, both activated by transforming growth factor beta 1 (TGF-β1). MSC-EVs effectively reduced the expression of profibrotic markers in liver spheroids and activated HSCs induced by TGF-β1 stimulation. These results highlight the potential of MSC-EVs collected under chemically defined conditions to mitigate the activated phenotype of HSCs and liver spheroids, suggesting MSC-EVs as a promising treatment for hepatic fibrosis.
Collapse
Affiliation(s)
| | | | | | | | | | - Stefania Bruno
- Department of Medical Sciences, University of Torino, 10126 Torino, Italy; (G.C.); (A.S.); (E.C.); (G.C.)
| |
Collapse
|
5
|
Ceccotti E, Chiabotto G, Cedrino M, Gambella A, Delsedime L, Ghigo A, Salio C, Grange C, Herrera Sanchez MB, Femminò S, Sassoè-Pognetto M, Brizzi MF, Camussi G, Bruno S. Extracellular Vesicles Derived from Human Liver Stem Cells Counteract Chronic Kidney Disease Development and Cardiac Dysfunction in Remnant Kidney Murine Model: The Possible Involvement of Proteases. Biomedicines 2024; 12:1517. [PMID: 39062090 PMCID: PMC11274379 DOI: 10.3390/biomedicines12071517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/24/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Fibrosis is a marker of chronic kidney disease (CKD) and consists of the accumulation of the extracellular matrix (ECM) components, causing the progressive deterioration of kidney function. Human liver stem cells (HLSCs) have anti-fibrotic activity, and HLSC-derived extracellular vesicles (EVs) mediate this effect. Herein, we evaluated the ability of HLSC-EVs to reverse renal and cardiac alterations in a murine model of partial nephrectomy (PNx) that mimics human CKD development. Furthermore, we investigated the contribution of extracellular matrix remodeling-related proteases to the anti-fibrotic effect of HLSC-EVs. PNx was performed by ligation of both poles of the left kidney, followed one week later by the removal of the right kidney. EV treatment started 4 weeks after the nephrectomy, when renal and cardiac alternations were already established, and mice were sacrificed at week eight. HLSC-EV treatment improved renal function and morphology, significantly decreasing interstitial fibrosis, glomerular sclerosis, and capillary rarefaction. This improvement was confirmed by the decreased expression of pro-fibrotic genes. Moreover, EV treatment improved cardiac function and reduced cardiac fibrosis. HLSC-EVs shuttled different proteases with ECM remodeling activity, and matrix metalloproteinase 1 (MMP-1) was involved in their anti-fibrotic effect on renal tissue. HLSC-EV treatment interferes with CKD development and ameliorates cardiomyopathy in PNx mice.
Collapse
Affiliation(s)
- Elena Ceccotti
- Department of Medical Sciences, University of Torino, 10126 Torino, Italy; (E.C.)
| | - Giulia Chiabotto
- Department of Medical Sciences, University of Torino, 10126 Torino, Italy; (E.C.)
| | | | - Alessandro Gambella
- Department of Medical Sciences, University of Torino, 10126 Torino, Italy; (E.C.)
| | - Luisa Delsedime
- Pathology Unit, Città delle Salute e della Scienza Hospital, 10126 Torino, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
- Molecular Biotechnology Center “Guido Tarone”, University of Torino, 10126 Torino, Italy
| | - Chiara Salio
- Department of Veterinary Sciences, University of Torino, Grugliasco, 10095 Torino, Italy
| | - Cristina Grange
- Department of Medical Sciences, University of Torino, 10126 Torino, Italy; (E.C.)
| | - Maria Beatriz Herrera Sanchez
- 2i3T, Società per la Gestione dell’Incubatore di Imprese e per il Trasferimento Tecnologico, University of Torino, 10126 Torino, Italy
| | - Saveria Femminò
- Department of Medical Sciences, University of Torino, 10126 Torino, Italy; (E.C.)
| | - Marco Sassoè-Pognetto
- Department of Neurosciences “Rita Levi Montalcini”, University of Torino, 10126 Torino, Italy
| | - Maria Felice Brizzi
- Department of Medical Sciences, University of Torino, 10126 Torino, Italy; (E.C.)
| | - Giovanni Camussi
- Department of Medical Sciences, University of Torino, 10126 Torino, Italy; (E.C.)
| | - Stefania Bruno
- Department of Medical Sciences, University of Torino, 10126 Torino, Italy; (E.C.)
| |
Collapse
|
6
|
Nguyen VVT, Welsh JA, Tertel T, Choo A, van de Wakker SI, Defourny KAY, Giebel B, Vader P, Padmanabhan J, Lim SK, Nolte‐'t Hoen ENM, Verhaar MC, Bostancioglu RB, Zickler AM, Hong JM, Jones JC, EL Andaloussi S, van Balkom BWM, Görgens A. Inter-laboratory multiplex bead-based surface protein profiling of MSC-derived EV preparations identifies MSC-EV surface marker signatures. J Extracell Vesicles 2024; 13:e12463. [PMID: 38868945 PMCID: PMC11170075 DOI: 10.1002/jev2.12463] [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: 10/20/2023] [Revised: 04/15/2024] [Accepted: 05/21/2024] [Indexed: 06/14/2024] Open
Abstract
Mesenchymal stromal cells (MSCs) are promising regenerative therapeutics that primarily exert their effects through secreted extracellular vesicles (EVs). These EVs - being small and non-living - are easier to handle and possess advantages over cellular products. Consequently, the therapeutic potential of MSC-EVs is increasingly investigated. However, due to variations in MSC-EV manufacturing strategies, MSC-EV products should be considered as highly diverse. Moreover, the diverse array of EV characterisation technologies used for MSC-EV characterisation further complicates reliable interlaboratory comparisons of published data. Consequently, this study aimed to establish a common method that can easily be used by various MSC-EV researchers to characterise MSC-EV preparations to facilitate interlaboratory comparisons. To this end, we conducted a comprehensive inter-laboratory assessment using a novel multiplex bead-based EV flow cytometry assay panel. This assessment involved 11 different MSC-EV products from five laboratories with varying MSC sources, culture conditions, and EV preparation methods. Through this assay panel covering a range of mostly MSC-related markers, we identified a set of cell surface markers consistently positive (CD44, CD73 and CD105) or negative (CD11b, CD45 and CD197) on EVs of all explored MSC-EV preparations. Hierarchical clustering analysis revealed distinct surface marker profiles associated with specific preparation processes and laboratory conditions. We propose CD73, CD105 and CD44 as robust positive markers for minimally identifying MSC-derived EVs and CD11b, CD14, CD19, CD45 and CD79 as reliable negative markers. Additionally, we highlight the influence of culture medium components, particularly human platelet lysate, on EV surface marker profiles, underscoring the influence of culture conditions on resulting EV products. This standardisable approach for MSC-EV surface marker profiling offers a tool for routine characterisation of manufactured EV products in pre-clinical and clinical research, enhances the quality control of MSC-EV preparations, and hopefully paves the way for higher consistency and reproducibility in the emerging therapeutic MSC-EV field.
Collapse
Affiliation(s)
| | - Joshua A. Welsh
- Translational Nanobiology Section, Laboratory of Pathology, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
- The Measuring Stick, LtdPeterboroughUK
- Advanced Technology GroupBecton DickinsonSan JoseCaliforniaUSA
| | - Tobias Tertel
- Institute for Transfusion MedicineUniversity Hospital EssenUniversity of Duisburg‐EssenEssenGermany
| | - Andre Choo
- Bioprocessing Technology Institute (BTI)Agency for Science, Technology and Research (A*STAR)SingaporeSingapore
| | - Simonides I. van de Wakker
- Department of Cardiology, Experimental Cardiology LaboratoryUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Kyra A. Y. Defourny
- Division of Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Bernd Giebel
- Institute for Transfusion MedicineUniversity Hospital EssenUniversity of Duisburg‐EssenEssenGermany
| | - Pieter Vader
- Department of Cardiology, Experimental Cardiology LaboratoryUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
- CDL ResearchUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Jayanthi Padmanabhan
- Bioprocessing Technology Institute (BTI)Agency for Science, Technology and Research (A*STAR)SingaporeSingapore
| | - Sai Kiang Lim
- Bioprocessing Technology Institute (BTI)Agency for Science, Technology and Research (A*STAR)SingaporeSingapore
| | - Esther N. M. Nolte‐'t Hoen
- Division of Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | | | - R. Beklem Bostancioglu
- Division of Biomolecular and Cellular Medicine, Department of Laboratory MedicineKarolinska InstitutetStockholmSweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer CenterStockholmSweden
| | - Antje M. Zickler
- Division of Biomolecular and Cellular Medicine, Department of Laboratory MedicineKarolinska InstitutetStockholmSweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer CenterStockholmSweden
- Karolinska ATMP CenterANA FuturaHuddingeSweden
| | - Jia Mei Hong
- Bioprocessing Technology Institute (BTI)Agency for Science, Technology and Research (A*STAR)SingaporeSingapore
| | - Jennifer C. Jones
- Translational Nanobiology Section, Laboratory of Pathology, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Samir EL Andaloussi
- Division of Biomolecular and Cellular Medicine, Department of Laboratory MedicineKarolinska InstitutetStockholmSweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer CenterStockholmSweden
- Karolinska ATMP CenterANA FuturaHuddingeSweden
| | | | - André Görgens
- Institute for Transfusion MedicineUniversity Hospital EssenUniversity of Duisburg‐EssenEssenGermany
- Division of Biomolecular and Cellular Medicine, Department of Laboratory MedicineKarolinska InstitutetStockholmSweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer CenterStockholmSweden
- Karolinska ATMP CenterANA FuturaHuddingeSweden
| |
Collapse
|
7
|
Wiklander OPB, Mamand DR, Mohammad DK, Zheng W, Jawad Wiklander R, Sych T, Zickler AM, Liang X, Sharma H, Lavado A, Bost J, Roudi S, Corso G, Lennaárd AJ, Abedi-Valugerdi M, Mäger I, Alici E, Sezgin E, Nordin JZ, Gupta D, Görgens A, El Andaloussi S. Antibody-displaying extracellular vesicles for targeted cancer therapy. Nat Biomed Eng 2024:10.1038/s41551-024-01214-6. [PMID: 38769158 DOI: 10.1038/s41551-024-01214-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 04/08/2024] [Indexed: 05/22/2024]
Abstract
Extracellular vesicles (EVs) function as natural delivery vectors and mediators of biological signals across tissues. Here, by leveraging these functionalities, we show that EVs decorated with an antibody-binding moiety specific for the fragment crystallizable (Fc) domain can be used as a modular delivery system for targeted cancer therapy. The Fc-EVs can be decorated with different types of immunoglobulin G antibody and thus be targeted to virtually any tissue of interest. Following optimization of the engineered EVs by screening Fc-binding and EV-sorting moieties, we show the targeting of EVs to cancer cells displaying the human epidermal receptor 2 or the programmed-death ligand 1, as well as lower tumour burden and extended survival of mice with subcutaneous melanoma tumours when systemically injected with EVs displaying an antibody for the programmed-death ligand 1 and loaded with the chemotherapeutic doxorubicin. EVs with Fc-binding domains may be adapted to display other Fc-fused proteins, bispecific antibodies and antibody-drug conjugates.
Collapse
Affiliation(s)
- Oscar P B Wiklander
- Department of Laboratory Medicine, Unit for Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden.
- Breast Center, Karolinska Comprehensive Cancer Center, Karolinska University Hospital, Stockholm, Sweden.
- Karolinska ATMP Center, ANA Futura, Huddinge, Sweden.
| | - Doste R Mamand
- Department of Laboratory Medicine, Unit for Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
- Breast Center, Karolinska Comprehensive Cancer Center, Karolinska University Hospital, Stockholm, Sweden
- Karolinska ATMP Center, ANA Futura, Huddinge, Sweden
| | - Dara K Mohammad
- Department of Medicine Huddinge, Karolinska Institute, Stockholm, Sweden
- College of Agricultural Engineering Sciences, Salahaddin University-Erbil, Erbil, Iraq
| | - Wenyi Zheng
- Department of Laboratory Medicine, Unit for Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska ATMP Center, ANA Futura, Huddinge, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
| | - Rim Jawad Wiklander
- Department of Laboratory Medicine, Unit for Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
| | - Taras Sych
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Antje M Zickler
- Department of Laboratory Medicine, Unit for Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska ATMP Center, ANA Futura, Huddinge, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
| | - Xiuming Liang
- Department of Laboratory Medicine, Unit for Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska ATMP Center, ANA Futura, Huddinge, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
| | | | | | - Jeremy Bost
- Department of Laboratory Medicine, Unit for Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
| | - Samantha Roudi
- Department of Laboratory Medicine, Unit for Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska ATMP Center, ANA Futura, Huddinge, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
| | - Giulia Corso
- Department of Laboratory Medicine, Unit for Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
| | - Angus J Lennaárd
- Department of Laboratory Medicine, Unit for Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
| | - Manuchehr Abedi-Valugerdi
- Department of Laboratory Medicine, Unit for Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
| | - Imre Mäger
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Evren Alici
- Department of Medicine Huddinge, Karolinska Institute, Stockholm, Sweden
- Hematology Center, Karolinska University Hospital, Stockholm, Sweden
| | - Erdinc Sezgin
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Joel Z Nordin
- Department of Laboratory Medicine, Unit for Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska ATMP Center, ANA Futura, Huddinge, Sweden
- Department of Clinical Immunology and Transfusion Medicine (KITM), Karolinska University Hospital, Stockholm, Sweden
| | - Dhanu Gupta
- Department of Laboratory Medicine, Unit for Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - André Görgens
- Department of Laboratory Medicine, Unit for Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska ATMP Center, ANA Futura, Huddinge, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Duisburg, Germany
| | - Samir El Andaloussi
- Department of Laboratory Medicine, Unit for Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden.
- Karolinska ATMP Center, ANA Futura, Huddinge, Sweden.
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden.
| |
Collapse
|
8
|
Wang R, Shi Y, Lv Y, Xie C, Hu Y. The novel insights of epithelial-derived exosomes in various fibrotic diseases. Biomed Pharmacother 2024; 174:116591. [PMID: 38631144 DOI: 10.1016/j.biopha.2024.116591] [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: 01/14/2024] [Revised: 04/07/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024] Open
Abstract
The characteristics of fibrosis include the abnormal accumulation of extracellular matrix proteins and abnormal tissue repair caused by injury, infection, and inflammation, leading to a significant increase in organ failure and mortality. Effective and precise treatments are urgently needed to halt and reverse the progression of fibrotic diseases. Exosomes are tiny vesicles derived from endosomes, spanning from 40 to 160 nanometers in diameter, which are expelled into the extracellular matrix environment by various cell types. They play a crucial role in facilitating cell-to-cell communication by transporting a variety of cargoes, including proteins, RNA, and DNA. Epithelial cells serve as the primary barrier against diverse external stimuli that precipitate fibrotic diseases. Numerous research suggests that exosomes from epithelial cells have a significant impact on several fibrotic diseases. An in-depth comprehension of the cellular and molecular mechanisms of epithelial cell-derived exosomes in fibrosis holds promise for advancing the exploration of novel diagnostic biomarkers and clinical drug targets. In this review, we expand upon the pathogenic mechanisms of epithelium-derived exosomes and highlight their role in the fibrotic process by inducing inflammation and activating fibroblasts. In addition, we are particularly interested in the bioactive molecules carried by epithelial-derived exosomes and their potential value in the diagnosis and treatment of fibrosis and delineate the clinical utility of exosomes as an emerging therapeutic modality, highlighting their potential application in addressing various medical conditions.
Collapse
Affiliation(s)
- Rifu Wang
- Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China
| | - Yuxin Shi
- Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China
| | - Yonglin Lv
- Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China
| | - Changqing Xie
- Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan, China.
| | - Yanjia Hu
- Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China.
| |
Collapse
|
9
|
Goldbloom-Helzner L, Bains H, Wang A. Approaches to Characterize and Quantify Extracellular Vesicle Surface Conjugation Efficiency. Life (Basel) 2024; 14:511. [PMID: 38672781 PMCID: PMC11051464 DOI: 10.3390/life14040511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/09/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Extracellular vesicles (EVs) are cell-secreted nanovesicles that play an important role in long-range cell-cell communication. Although EVs pose a promising alternative to cell-based therapy, targeted in vivo delivery still falls short. Many studies have explored the surface modification of EVs to enhance their targeting capabilities. However, to our knowledge, there are no standardized practices to confirm the successful surface modification of EVs or calculate the degree of conjugation on EV surfaces (conjugation efficiency). These pieces of information are essential in the reproducibility of targeted EV therapeutics and the determination of optimized conjugation conditions for EVs to see significant therapeutic effects in vitro and in vivo. This review will discuss the vast array of techniques adopted, technologies developed, and efficiency definitions made by studies that have calculated EV/nanoparticle surface conjugation efficiency and how differences between studies may contribute to differently reported conjugation efficiencies.
Collapse
Affiliation(s)
- Leora Goldbloom-Helzner
- Center for Surgical Bioengineering, Department of Surgery, School of Medicine, UC Davis Health, Sacramento, CA 95817, USA;
- Department of Biomedical Engineering, UC Davis, Davis, CA 95616, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, USA
| | - Harjn Bains
- Department of Biomedical Engineering, UC Davis, Davis, CA 95616, USA
| | - Aijun Wang
- Center for Surgical Bioengineering, Department of Surgery, School of Medicine, UC Davis Health, Sacramento, CA 95817, USA;
- Department of Biomedical Engineering, UC Davis, Davis, CA 95616, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, USA
| |
Collapse
|
10
|
Sych T, Schlegel J, Barriga HMG, Ojansivu M, Hanke L, Weber F, Beklem Bostancioglu R, Ezzat K, Stangl H, Plochberger B, Laurencikiene J, El Andaloussi S, Fürth D, Stevens MM, Sezgin E. High-throughput measurement of the content and properties of nano-sized bioparticles with single-particle profiler. Nat Biotechnol 2024; 42:587-590. [PMID: 37308687 PMCID: PMC11021190 DOI: 10.1038/s41587-023-01825-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 05/10/2023] [Indexed: 06/14/2023]
Abstract
We introduce a method, single-particle profiler, that provides single-particle information on the content and biophysical properties of thousands of particles in the size range 5-200 nm. We use our single-particle profiler to measure the messenger RNA encapsulation efficiency of lipid nanoparticles, the viral binding efficiencies of different nanobodies, and the biophysical heterogeneity of liposomes, lipoproteins, exosomes and viruses.
Collapse
Affiliation(s)
- Taras Sych
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Jan Schlegel
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Hanna M G Barriga
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Miina Ojansivu
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Leo Hanke
- Division of Infectious Diseases, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Florian Weber
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
- Department Medical Engineering, University of Applied Sciences Upper Austria, Linz, Austria
| | | | - Kariem Ezzat
- Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Herbert Stangl
- Medical University of Vienna, Center for Pathobiochemistry and Genetics, Institute of Medical Chemistry, Vienna, Austria
| | - Birgit Plochberger
- Department Medical Engineering, University of Applied Sciences Upper Austria, Linz, Austria
- LBG Ludwig Boltzmann Institute for Traumatology, Nanoscopy, Vienna, Austria
| | - Jurga Laurencikiene
- Lipid Laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | | | - Daniel Fürth
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Molly M Stevens
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London, UK
| | - Erdinc Sezgin
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden.
| |
Collapse
|
11
|
Groß R, Reßin H, von Maltitz P, Albers D, Schneider L, Bley H, Hoffmann M, Cortese M, Gupta D, Deniz M, Choi JY, Jansen J, Preußer C, Seehafer K, Pöhlmann S, Voelker DR, Goffinet C, Pogge-von Strandmann E, Bunz U, Bartenschlager R, El Andaloussi S, Sparrer KMJ, Herker E, Becker S, Kirchhoff F, Münch J, Müller JA. Phosphatidylserine-exposing extracellular vesicles in body fluids are an innate defence against apoptotic mimicry viral pathogens. Nat Microbiol 2024; 9:905-921. [PMID: 38528146 PMCID: PMC10994849 DOI: 10.1038/s41564-024-01637-6] [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: 03/16/2023] [Accepted: 02/14/2024] [Indexed: 03/27/2024]
Abstract
Some viruses are rarely transmitted orally or sexually despite their presence in saliva, breast milk, or semen. We previously identified that extracellular vesicles (EVs) in semen and saliva inhibit Zika virus infection. However, the antiviral spectrum and underlying mechanism remained unclear. Here we applied lipidomics and flow cytometry to show that these EVs expose phosphatidylserine (PS). By blocking PS receptors, targeted by Zika virus in the process of apoptotic mimicry, they interfere with viral attachment and entry. Consequently, physiological concentrations of EVs applied in vitro efficiently inhibited infection by apoptotic mimicry dengue, West Nile, Chikungunya, Ebola and vesicular stomatitis viruses, but not severe acute respiratory syndrome coronavirus 2, human immunodeficiency virus 1, hepatitis C virus and herpesviruses that use other entry receptors. Our results identify the role of PS-rich EVs in body fluids in innate defence against infection via viral apoptotic mimicries, explaining why these viruses are primarily transmitted via PS-EV-deficient blood or blood-ingesting arthropods rather than direct human-to-human contact.
Collapse
Affiliation(s)
- Rüdiger Groß
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Hanna Reßin
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Pascal von Maltitz
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Dan Albers
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Laura Schneider
- Institute of Virology, Philipps University Marburg, Marburg, Germany
| | - Hanna Bley
- Institute of Virology, Philipps University Marburg, Marburg, Germany
| | - Markus Hoffmann
- Infection Biology Unit, German Primate Center, Göttingen, Germany
- Georg-August University Göttingen, Göttingen, Germany
| | - Mirko Cortese
- Department of Infectious Diseases, Molecular Virology, University of Heidelberg, Heidelberg, Germany
| | - Dhanu Gupta
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Miriam Deniz
- Clinic for Gynecology and Obstetrics, Ulm University Medical Center, Ulm, Germany
| | - Jae-Yeon Choi
- Department of Medicine, National Jewish Health, Denver, CO, USA
| | - Jenny Jansen
- Institute of Virology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Christian Preußer
- Core Facility Extracellular Vesicles, Institute for Tumor Immunology, Center for Tumor Biology and Immunology, Philipps University Marburg, Marburg, Germany
| | - Kai Seehafer
- Organisch-Chemisches Institut, Ruprecht-Karls-Universität, Heidelberg, Germany
| | - Stefan Pöhlmann
- Infection Biology Unit, German Primate Center, Göttingen, Germany
- Georg-August University Göttingen, Göttingen, Germany
| | | | - Christine Goffinet
- Institute of Virology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Elke Pogge-von Strandmann
- Core Facility Extracellular Vesicles, Institute for Tumor Immunology, Center for Tumor Biology and Immunology, Philipps University Marburg, Marburg, Germany
| | - Uwe Bunz
- Organisch-Chemisches Institut, Ruprecht-Karls-Universität, Heidelberg, Germany
| | - Ralf Bartenschlager
- Department of Infectious Diseases, Molecular Virology, University of Heidelberg, Heidelberg, Germany
| | - Samir El Andaloussi
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Eva Herker
- Institute of Virology, Philipps University Marburg, Marburg, Germany
| | - Stephan Becker
- Institute of Virology, Philipps University Marburg, Marburg, Germany
| | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Janis A Müller
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany.
- Institute of Virology, Philipps University Marburg, Marburg, Germany.
| |
Collapse
|
12
|
Gençer EB, Lor YK, Abomaray F, El Andaloussi S, Pernemalm M, Sharma N, Hagey DW, Görgens A, Gustafsson MO, Le Blanc K, Asad Toonsi M, Walther-Jallow L, Götherström C. Transcriptomic and proteomic profiles of fetal versus adult mesenchymal stromal cells and mesenchymal stromal cell-derived extracellular vesicles. Stem Cell Res Ther 2024; 15:77. [PMID: 38475970 DOI: 10.1186/s13287-024-03683-7] [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: 11/08/2023] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Mesenchymal stem/stromal cells (MSCs) can regenerate tissues through engraftment and differentiation but also via paracrine signalling via extracellular vesicles (EVs). Fetal-derived MSCs (fMSCs) have been shown, both in vitro and in animal studies, to be more efficient than adult MSC (aMSCs) in generating bone and muscle but the underlying reason for this difference has not yet been clearly elucidated. In this study, we aimed to systematically investigate the differences between fetal and adult MSCs and MSC-derived EVs at the phenotypic, RNA, and protein levels. METHODS We carried out a detailed and comparative characterization of culture-expanded fetal liver derived MSCs (fMSCs) and adult bone marrow derived MSCs (aMSCs) phenotypically, and the MSCs and MSC-derived EVs were analysed using transcriptomics and proteomics approaches with RNA Sequencing and Mass Spectrometry. RESULTS Fetal MSCs were smaller, exhibited increased proliferation and colony-forming capacity, delayed onset of senescence, and demonstrated superior osteoblast differentiation capability compared to their adult counterparts. Gene Ontology analysis revealed that fMSCs displayed upregulated gene sets such as "Positive regulation of stem cell populations", "Maintenance of stemness" and "Muscle cell development/contraction/Myogenesis" in comparison to aMSCs. Conversely, aMSCs displayed upregulated gene sets such as "Complement cascade", "Adipogenesis", "Extracellular matrix glycoproteins" and "Cellular metabolism", and on the protein level, "Epithelial cell differentiation" pathways. Signalling entropy analysis suggested that fMSCs exhibit higher signalling promiscuity and hence, higher potency than aMSCs. Gene ontology comparisons revealed that fetal MSC-derived EVs (fEVs) were enriched for "Collagen fibril organization", "Protein folding", and "Response to transforming growth factor beta" compared to adult MSC-derived EVs (aEVs), whereas no significant difference in protein expression in aEVs compared to fEVs could be detected. CONCLUSIONS This study provides detailed and systematic insight into the differences between fMSCs and aMSCs, and MSC-derived EVs. The key finding across phenotypic, transcriptomic and proteomic levels is that fMSCs exhibit higher potency than aMSCs, meaning they are in a more undifferentiated state. Additionally, fMSCs and fMSC-derived EVs may possess greater bone forming capacity compared to aMSCs. Therefore, using fMSCs may lead to better treatment efficacy, especially in musculoskeletal diseases.
Collapse
Affiliation(s)
- Emine Begüm Gençer
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Yuk Kit Lor
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Fawaz Abomaray
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Samir El Andaloussi
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer Center, Stockholm, Sweden
| | - Maria Pernemalm
- Cancer Proteomics Mass Spectrometry, Science for Life Laboratory, Department of Oncology- Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Nidhi Sharma
- Cancer Proteomics Mass Spectrometry, Science for Life Laboratory, Department of Oncology- Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Daniel W Hagey
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer Center, Stockholm, Sweden
| | - André Görgens
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer Center, Stockholm, Sweden
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Manuela O Gustafsson
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer Center, Stockholm, Sweden
| | - Katarina Le Blanc
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer Center, Stockholm, Sweden
- Division of Clinical Immunology and Transfusion Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Mawaddah Asad Toonsi
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Department of Pediatrics, College of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Lilian Walther-Jallow
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Cecilia Götherström
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
13
|
Schindler CR, Hörauf JA, Weber B, Schaible I, Marzi I, Henrich D, Leppik L. Identification of novel blood-based extracellular vesicles biomarker candidates with potential specificity for traumatic brain injury in polytrauma patients. Front Immunol 2024; 15:1347767. [PMID: 38533491 PMCID: PMC10963595 DOI: 10.3389/fimmu.2024.1347767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 02/19/2024] [Indexed: 03/28/2024] Open
Abstract
Objective The goal of this study was to identify changes in extracellular vesicles (EV) surface proteins specific to traumatic brain injury (TBI), which could be used as a diagnostic and prognostic tool in polytrauma patients. Summary Background Data Known serum TBI-specific biomarkers (S100B, NSE, and GFAP), which can predict the severity and outcome of isolated TBI, lose their predictive value in the presence of additional extracranial injuries. Extracellular vesicles (EVs) are released from cells in response to various stimuli and carry specific cargo/surface molecules that could be used for tracking injury-responding cells. Methods EVs were isolated using size exclusion chromatography (SEC) from the plasma of two groups of patients (with isolated TBI, ISS≥16, AIShead≥4, n=10; and polytraumatized patients without TBI ISS≥16, AIShead=0, n=10) collected in the emergency room and 48 h after trauma. EVs' surface epitope expression was investigated using a neurospecific multiplex flow cytometry assay and compared with healthy controls (n=10). Three enrichments of EV epitopes found to be specific to TBI were validated by western blot. Results The expression of 10 EV epitopes differed significantly among the patient and control groups, and five of these epitopes (CD13, CD196, MOG, CD133, and MBP) were TBI-specific. The increased expression of CD196, CD13, and MOG-positive EVs was validated by western blot. Conclusion Our data showed that TBI is characterized by a significant increase of CD13, CD196, MOG, CD133, and MBP-positive EVs in patients' plasma. A high level of MOG-positive EVs negatively correlated with the Glasgow Coma Scale score at admission and could be an indicator of poor neurological status.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Liudmila Leppik
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe-University, Frankfurt am Main, Germany
| |
Collapse
|
14
|
Chen Z, Luo L, Ye T, Zhou J, Niu X, Yuan J, Yuan T, Fu D, Li H, Li Q, Wang Y. Identification of specific markers for human pluripotent stem cell-derived small extracellular vesicles. J Extracell Vesicles 2024; 13:e12409. [PMID: 38321535 PMCID: PMC10847391 DOI: 10.1002/jev2.12409] [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: 07/24/2023] [Revised: 12/17/2023] [Accepted: 01/12/2024] [Indexed: 02/08/2024] Open
Abstract
Pluripotent stem cell-derived small extracellular vesicles (PSC-sEVs) have demonstrated great clinical translational potential in multiple aging-related degenerative diseases. Characterizing the PSC-sEVs is crucial for their clinical applications. However, the specific marker pattern of PSC-sEVs remains unknown. Here, the sEVs derived from two typical types of PSCs including induced pluripotent stem cells (iPSC-sEVs) and embryonic stem cells (ESC-sEVs) were analysed using proteomic analysis by liquid chromatography with tandem mass spectrometry (LC-MS/MS), and surface marker phenotyping analysis by nanoparticle flow cytometry (NanoFCM). A group of pluripotency-related proteins were found to be enriched in PSC-sEVs by LC-MS/MS and then validated by Western Blot analysis. To investigate whether these proteins were specifically expressed in PSC-sEVs, sEVs derived from seven types of non-PSCs (non-PSC-sEVs) were adopted for analysis. The results showed that PODXL, OCT4, Dnmt3a, and LIN28A were specifically enriched in PSC-sEVs but not in non-PSC-sEVs. Then, commonly used surface antigens for PSC identification (SSEA4, Tra-1-60 and Tra-1-81) and PODXL were gauged at single-particle resolution by NanoFCM for surface marker identification. The results showed that the positive rates of PODXL (>50%) and SSEA4 (>70%) in PSC-sEVs were much higher than those in non-PSC-sEVs (<10%). These results were further verified with samples purified by density gradient ultracentrifugation. Taken together, this study for the first time identified a cohort of specific markers for PSC-sEVs, among which PODXL, OCT4, Dnmt3a and LIN28A can be detected with Western Blot analysis, and PODXL and SSEA4 can be detected with NanoFCM analysis. The application of these specific markers for PSC-sEVs identification may advance the clinical translation of PSCs-sEVs.
Collapse
Affiliation(s)
- Zhengsheng Chen
- Institute of Microsurgery on Extremities, Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Lei Luo
- Institute of Microsurgery on Extremities, Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
- School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Teng Ye
- Institute of Microsurgery on Extremities, Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jiacheng Zhou
- Institute of Microsurgery on Extremities, Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xin Niu
- Institute of Microsurgery on Extremities, Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Ji Yuan
- Institute of Microsurgery on Extremities, Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Ting Yuan
- Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Dehao Fu
- Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Haiyan Li
- School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
- Chemical and Environmental Engineering Department, School of EngineeringRMIT UniversityMelbourneVictoriaAustralia
| | - Qing Li
- Institute of Microsurgery on Extremities, Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yang Wang
- Institute of Microsurgery on Extremities, Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
15
|
Welsh JA, Goberdhan DCI, O'Driscoll L, Buzas EI, Blenkiron C, Bussolati B, Cai H, Di Vizio D, Driedonks TAP, Erdbrügger U, Falcon‐Perez JM, Fu Q, Hill AF, Lenassi M, Lim SK, Mahoney MG, Mohanty S, Möller A, Nieuwland R, Ochiya T, Sahoo S, Torrecilhas AC, Zheng L, Zijlstra A, Abuelreich S, Bagabas R, Bergese P, Bridges EM, Brucale M, Burger D, Carney RP, Cocucci E, Colombo F, Crescitelli R, Hanser E, Harris AL, Haughey NJ, Hendrix A, Ivanov AR, Jovanovic‐Talisman T, Kruh‐Garcia NA, Ku'ulei‐Lyn Faustino V, Kyburz D, Lässer C, Lennon KM, Lötvall J, Maddox AL, Martens‐Uzunova ES, Mizenko RR, Newman LA, Ridolfi A, Rohde E, Rojalin T, Rowland A, Saftics A, Sandau US, Saugstad JA, Shekari F, Swift S, Ter‐Ovanesyan D, Tosar JP, Useckaite Z, Valle F, Varga Z, van der Pol E, van Herwijnen MJC, Wauben MHM, Wehman AM, Williams S, Zendrini A, Zimmerman AJ, MISEV Consortium, Théry C, Witwer KW. Minimal information for studies of extracellular vesicles (MISEV2023): From basic to advanced approaches. J Extracell Vesicles 2024; 13:e12404. [PMID: 38326288 PMCID: PMC10850029 DOI: 10.1002/jev2.12404] [Citation(s) in RCA: 318] [Impact Index Per Article: 318.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 12/15/2023] [Accepted: 12/19/2023] [Indexed: 02/09/2024] Open
Abstract
Extracellular vesicles (EVs), through their complex cargo, can reflect the state of their cell of origin and change the functions and phenotypes of other cells. These features indicate strong biomarker and therapeutic potential and have generated broad interest, as evidenced by the steady year-on-year increase in the numbers of scientific publications about EVs. Important advances have been made in EV metrology and in understanding and applying EV biology. However, hurdles remain to realising the potential of EVs in domains ranging from basic biology to clinical applications due to challenges in EV nomenclature, separation from non-vesicular extracellular particles, characterisation and functional studies. To address the challenges and opportunities in this rapidly evolving field, the International Society for Extracellular Vesicles (ISEV) updates its 'Minimal Information for Studies of Extracellular Vesicles', which was first published in 2014 and then in 2018 as MISEV2014 and MISEV2018, respectively. The goal of the current document, MISEV2023, is to provide researchers with an updated snapshot of available approaches and their advantages and limitations for production, separation and characterisation of EVs from multiple sources, including cell culture, body fluids and solid tissues. In addition to presenting the latest state of the art in basic principles of EV research, this document also covers advanced techniques and approaches that are currently expanding the boundaries of the field. MISEV2023 also includes new sections on EV release and uptake and a brief discussion of in vivo approaches to study EVs. Compiling feedback from ISEV expert task forces and more than 1000 researchers, this document conveys the current state of EV research to facilitate robust scientific discoveries and move the field forward even more rapidly.
Collapse
Affiliation(s)
- Joshua A. Welsh
- Translational Nanobiology Section, Laboratory of PathologyNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Deborah C. I. Goberdhan
- Nuffield Department of Women's and Reproductive HealthUniversity of Oxford, Women's Centre, John Radcliffe HospitalOxfordUK
| | - Lorraine O'Driscoll
- School of Pharmacy and Pharmaceutical SciencesTrinity College DublinDublinIreland
- Trinity Biomedical Sciences InstituteTrinity College DublinDublinIreland
- Trinity St. James's Cancer InstituteTrinity College DublinDublinIreland
| | - Edit I. Buzas
- Department of Genetics, Cell‐ and ImmunobiologySemmelweis UniversityBudapestHungary
- HCEMM‐SU Extracellular Vesicle Research GroupSemmelweis UniversityBudapestHungary
- HUN‐REN‐SU Translational Extracellular Vesicle Research GroupSemmelweis UniversityBudapestHungary
| | - Cherie Blenkiron
- Faculty of Medical and Health SciencesThe University of AucklandAucklandNew Zealand
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health SciencesUniversity of TurinTurinItaly
| | | | - Dolores Di Vizio
- Department of Surgery, Division of Cancer Biology and TherapeuticsCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Tom A. P. Driedonks
- Department CDL ResearchUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Uta Erdbrügger
- University of Virginia Health SystemCharlottesvilleVirginiaUSA
| | - Juan M. Falcon‐Perez
- Exosomes Laboratory, Center for Cooperative Research in BiosciencesBasque Research and Technology AllianceDerioSpain
- Metabolomics Platform, Center for Cooperative Research in BiosciencesBasque Research and Technology AllianceDerioSpain
- IKERBASQUE, Basque Foundation for ScienceBilbaoSpain
| | - Qing‐Ling Fu
- Otorhinolaryngology Hospital, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Extracellular Vesicle Research and Clinical Translational CenterThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Andrew F. Hill
- Institute for Health and SportVictoria UniversityMelbourneAustralia
| | - Metka Lenassi
- Faculty of MedicineUniversity of LjubljanaLjubljanaSlovenia
| | - Sai Kiang Lim
- Institute of Molecular and Cell Biology (IMCB)Agency for Science, Technology and Research (A*STAR)SingaporeSingapore
- Paracrine Therapeutics Pte. Ltd.SingaporeSingapore
- Department of Surgery, YLL School of MedicineNational University SingaporeSingaporeSingapore
| | - Mỹ G. Mahoney
- Thomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Sujata Mohanty
- Stem Cell FacilityAll India Institute of Medical SciencesNew DelhiIndia
| | - Andreas Möller
- Chinese University of Hong KongHong KongHong Kong S.A.R.
- QIMR Berghofer Medical Research InstituteBrisbaneAustralia
| | - Rienk Nieuwland
- Laboratory of Experimental Clinical Chemistry, Amsterdam University Medical Centers, Location AMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Vesicle Center, Amsterdam University Medical Centers, Location AMCUniversity of AmsterdamAmsterdamThe Netherlands
| | | | - Susmita Sahoo
- Icahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Ana C. Torrecilhas
- Laboratório de Imunologia Celular e Bioquímica de Fungos e Protozoários, Departamento de Ciências Farmacêuticas, Instituto de Ciências Ambientais, Químicas e FarmacêuticasUniversidade Federal de São Paulo (UNIFESP) Campus DiademaDiademaBrazil
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Andries Zijlstra
- Department of PathologyVanderbilt University Medical CenterNashvilleTennesseeUSA
- GenentechSouth San FranciscoCaliforniaUSA
| | - Sarah Abuelreich
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Reem Bagabas
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Paolo Bergese
- Department of Molecular and Translational MedicineUniversity of BresciaBresciaItaly
- Center for Colloid and Surface Science (CSGI)FlorenceItaly
- National Center for Gene Therapy and Drugs based on RNA TechnologyPaduaItaly
| | - Esther M. Bridges
- Weatherall Institute of Molecular MedicineUniversity of OxfordOxfordUK
| | - Marco Brucale
- Consiglio Nazionale delle Ricerche ‐ Istituto per lo Studio dei Materiali NanostrutturatiBolognaItaly
- Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande InterfaseFlorenceItaly
| | - Dylan Burger
- Kidney Research CentreOttawa Hopsital Research InstituteOttawaCanada
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaCanada
- School of Pharmaceutical SciencesUniversity of OttawaOttawaCanada
| | - Randy P. Carney
- Department of Biomedical EngineeringUniversity of CaliforniaDavisCaliforniaUSA
| | - Emanuele Cocucci
- Division of Pharmaceutics and Pharmacology, College of PharmacyThe Ohio State UniversityColumbusOhioUSA
- Comprehensive Cancer CenterThe Ohio State UniversityColumbusOhioUSA
| | - Federico Colombo
- Division of Pharmaceutics and Pharmacology, College of PharmacyThe Ohio State UniversityColumbusOhioUSA
| | - Rossella Crescitelli
- Sahlgrenska Center for Cancer Research, Department of Surgery, Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Edveena Hanser
- Department of BiomedicineUniversity Hospital BaselBaselSwitzerland
- Department of BiomedicineUniversity of BaselBaselSwitzerland
| | | | - Norman J. Haughey
- Departments of Neurology and PsychiatryJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - An Hendrix
- Laboratory of Experimental Cancer Research, Department of Human Structure and RepairGhent UniversityGhentBelgium
- Cancer Research Institute GhentGhentBelgium
| | - Alexander R. Ivanov
- Barnett Institute of Chemical and Biological Analysis, Department of Chemistry and Chemical BiologyNortheastern UniversityBostonMassachusettsUSA
| | - Tijana Jovanovic‐Talisman
- Department of Cancer Biology and Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Nicole A. Kruh‐Garcia
- Bio‐pharmaceutical Manufacturing and Academic Resource Center (BioMARC)Infectious Disease Research Center, Colorado State UniversityFort CollinsColoradoUSA
| | - Vroniqa Ku'ulei‐Lyn Faustino
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Diego Kyburz
- Department of BiomedicineUniversity of BaselBaselSwitzerland
- Department of RheumatologyUniversity Hospital BaselBaselSwitzerland
| | - Cecilia Lässer
- Krefting Research Centre, Department of Internal Medicine and Clinical NutritionInstitute of Medicine at Sahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Kathleen M. Lennon
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Jan Lötvall
- Krefting Research Centre, Institute of Medicine at Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Adam L. Maddox
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Elena S. Martens‐Uzunova
- Erasmus MC Cancer InstituteUniversity Medical Center Rotterdam, Department of UrologyRotterdamThe Netherlands
| | - Rachel R. Mizenko
- Department of Biomedical EngineeringUniversity of CaliforniaDavisCaliforniaUSA
| | - Lauren A. Newman
- College of Medicine and Public HealthFlinders UniversityAdelaideAustralia
| | - Andrea Ridolfi
- Department of Physics and Astronomy, and LaserLaB AmsterdamVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Eva Rohde
- Department of Transfusion Medicine, University HospitalSalzburger Landeskliniken GmbH of Paracelsus Medical UniversitySalzburgAustria
- GMP Unit, Paracelsus Medical UniversitySalzburgAustria
- Transfer Centre for Extracellular Vesicle Theralytic Technologies, EV‐TTSalzburgAustria
| | - Tatu Rojalin
- Department of Biomedical EngineeringUniversity of CaliforniaDavisCaliforniaUSA
- Expansion Therapeutics, Structural Biology and BiophysicsJupiterFloridaUSA
| | - Andrew Rowland
- College of Medicine and Public HealthFlinders UniversityAdelaideAustralia
| | - Andras Saftics
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Ursula S. Sandau
- Department of Anesthesiology & Perioperative MedicineOregon Health & Science UniversityPortlandOregonUSA
| | - Julie A. Saugstad
- Department of Anesthesiology & Perioperative MedicineOregon Health & Science UniversityPortlandOregonUSA
| | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
- Celer DiagnosticsTorontoCanada
| | - Simon Swift
- Waipapa Taumata Rau University of AucklandAucklandNew Zealand
| | - Dmitry Ter‐Ovanesyan
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMassachusettsUSA
| | - Juan P. Tosar
- Universidad de la RepúblicaMontevideoUruguay
- Institut Pasteur de MontevideoMontevideoUruguay
| | - Zivile Useckaite
- College of Medicine and Public HealthFlinders UniversityAdelaideAustralia
| | - Francesco Valle
- Consiglio Nazionale delle Ricerche ‐ Istituto per lo Studio dei Materiali NanostrutturatiBolognaItaly
- Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande InterfaseFlorenceItaly
| | - Zoltan Varga
- Biological Nanochemistry Research GroupInstitute of Materials and Environmental Chemistry, Research Centre for Natural SciencesBudapestHungary
- Department of Biophysics and Radiation BiologySemmelweis UniversityBudapestHungary
| | - Edwin van der Pol
- Amsterdam Vesicle Center, Amsterdam University Medical Centers, Location AMCUniversity of AmsterdamAmsterdamThe Netherlands
- Biomedical Engineering and Physics, Amsterdam UMC, location AMCUniversity of AmsterdamAmsterdamThe Netherlands
- Laboratory of Experimental Clinical Chemistry, Amsterdam UMC, location AMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Martijn J. C. van Herwijnen
- Department of Biomolecular Health Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Marca H. M. Wauben
- Department of Biomolecular Health Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | | | | | - Andrea Zendrini
- Department of Molecular and Translational MedicineUniversity of BresciaBresciaItaly
- Center for Colloid and Surface Science (CSGI)FlorenceItaly
| | - Alan J. Zimmerman
- Barnett Institute of Chemical and Biological Analysis, Department of Chemistry and Chemical BiologyNortheastern UniversityBostonMassachusettsUSA
| | | | - Clotilde Théry
- Institut Curie, INSERM U932PSL UniversityParisFrance
- CurieCoreTech Extracellular Vesicles, Institut CurieParisFrance
| | - Kenneth W. Witwer
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- EV Core Facility “EXCEL”, Institute for Basic Biomedical SciencesJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- The Richman Family Precision Medicine Center of Excellence in Alzheimer's DiseaseJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
16
|
Wies Mancini VSB, Mattera VS, Pasquini JM, Pasquini LA, Correale JD. Microglia-derived extracellular vesicles in homeostasis and demyelination/remyelination processes. J Neurochem 2024; 168:3-25. [PMID: 38055776 DOI: 10.1111/jnc.16011] [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: 07/12/2023] [Revised: 10/10/2023] [Accepted: 11/03/2023] [Indexed: 12/08/2023]
Abstract
Microglia (MG) play a crucial role as the predominant myeloid cells in the central nervous system and are commonly activated in multiple sclerosis. They perform essential functions under normal conditions, such as actively surveying the surrounding parenchyma, facilitating synaptic remodeling, engulfing dead cells and debris, and protecting the brain against infectious pathogens and harmful self-proteins. Extracellular vesicles (EVs) are diverse structures enclosed by a lipid bilayer that originate from intracellular endocytic trafficking or the plasma membrane. They are released by cells into the extracellular space and can be found in various bodily fluids. EVs have recently emerged as a communication mechanism between cells, enabling the transfer of functional proteins, lipids, different RNA species, and even fragments of DNA from donor cells. MG act as both source and recipient of EVs. Consequently, MG-derived EVs are involved in regulating synapse development and maintaining homeostasis. These EVs also directly influence astrocytes, significantly increasing the release of inflammatory cytokines like IL-1β, IL-6, and TNF-α, resulting in a robust inflammatory response. Furthermore, EVs derived from inflammatory MG have been found to inhibit remyelination, whereas Evs produced by pro-regenerative MG effectively promote myelin repair. This review aims to provide an overview of the current understanding of MG-derived Evs, their impact on neighboring cells, and the cellular microenvironment in normal conditions and pathological states, specifically focusing on demyelination and remyelination processes.
Collapse
Affiliation(s)
- V S B Wies Mancini
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Cátedra de Química Biológica Patológica, Universidad de Buenos Aires, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Farmacia y Bioquímica, Instituto de Química y Fisicoquímica Biológicas Prof. Dr. Alejandro C. Paladini, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - V S Mattera
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Cátedra de Química Biológica Patológica, Universidad de Buenos Aires, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Farmacia y Bioquímica, Instituto de Química y Fisicoquímica Biológicas Prof. Dr. Alejandro C. Paladini, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - J M Pasquini
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Cátedra de Química Biológica Patológica, Universidad de Buenos Aires, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Farmacia y Bioquímica, Instituto de Química y Fisicoquímica Biológicas Prof. Dr. Alejandro C. Paladini, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - L A Pasquini
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Cátedra de Química Biológica Patológica, Universidad de Buenos Aires, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Farmacia y Bioquímica, Instituto de Química y Fisicoquímica Biológicas Prof. Dr. Alejandro C. Paladini, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - J D Correale
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Cátedra de Química Biológica Patológica, Universidad de Buenos Aires, Buenos Aires, Argentina
- Departamento de Neurología, Fleni, Buenos Aires, Argentina
| |
Collapse
|
17
|
Huang J, Ding M, Lu Y, Xu L, Zhang Y, Han S, Zhu X, Li Y, Chen P. MiR-1246b, a novel miRNA molecule of extracellular vesicles in bronchoalveolar lavage fluid, promotes nodule growth through FGF14 in patients with lung cancer. Cell Death Dis 2023; 14:789. [PMID: 38040694 PMCID: PMC10692082 DOI: 10.1038/s41419-023-06218-9] [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: 04/17/2023] [Revised: 09/30/2023] [Accepted: 10/13/2023] [Indexed: 12/03/2023]
Abstract
With the widespread development of chest computed tomography (CT), the detection rate of pulmonary nodules has increased; therefore, the classification of benign vs. malignant nodules has become a common problem in the clinic. MicroRNA, a potential tool, is expected to become a good choice for diagnosing and studying the occurrence and development of diseases through the vector of bronchoalveolar lavage fluid extracellular vesicles (BALF-EVs). In this study, radial endobronchial ultrasound (R-EBUS) was used to locate pulmonary nodules in patients. BALF was obtained, EVs were isolated, and small RNA sequencing was performed to screen differentially expressed miRNAs between benign and malignant pulmonary nodules. The binding targets and underlying mechanisms of the differentially expressed miRNAs were verified by in vitro and in vivo experiments. R-EBUS localization and sampling was used to obtain BALF, and EVs were successfully isolated and characterized. Differentially expressed miRNAs in BALF-EVs of patients with benign vs. malignant pulmonary nodules were screened by high-throughput small RNA sequencing. A new miRNA, miR-1246b, was identified. We found that FGF14 was the binding target of miR-1246b by luciferase assay. Subsequent mechanistic studies showed that miR-1246b inhibited the expression of FGF14 in lung cancer cells, further leading to ERK phosphorylation and epithelial-to-mesenchymal transition (EMT), which ultimately contributed to lung cancer cell proliferation, migration and invasion. In summary, our study demonstrates that the detection of miRNAs in BALF-EVs, a means of liquid biopsy, could assist in distinguishing malignant nodules from benign nodules. miR-1246b, which was extracted from BALF-EVs, targets FGF14 to promote lung cancer cell proliferation, migration and invasion.
Collapse
Affiliation(s)
- Jing Huang
- Department of Respiratory and Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Ming Ding
- Department of Respiratory and Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Yuan Lu
- Department of Respiratory and Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Lu Xu
- Department of Pathology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Yu Zhang
- Department of Pathology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Shuhua Han
- Department of Respiratory and Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Xiaoli Zhu
- Department of Respiratory and Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Yiping Li
- Department of Pathology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Pingsheng Chen
- Department of Pathology, School of Medicine, Southeast University, Nanjing, Jiangsu, China.
| |
Collapse
|
18
|
Kong F, Upadya M, Wong ASW, Dalan R, Dao M. Isolating Small Extracellular Vesicles from Small Volumes of Blood Plasma using size exclusion chromatography and density gradient ultracentrifugation: A Comparative Study. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.30.564707. [PMID: 37961562 PMCID: PMC10634961 DOI: 10.1101/2023.10.30.564707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Small extracellular vesicles (sEVs) are heterogeneous biological vesicles released by cells under both physiological and pathological conditions. Due to their potential as valuable diagnostic and prognostic biomarkers in human blood, there is a pressing need to develop effective methods for isolating high-purity sEVs from the complex milieu of blood plasma, which contains abundant plasma proteins and lipoproteins. Size exclusion chromatography (SEC) and density gradient ultracentrifugation (DGUC) are two commonly employed isolation techniques that have shown promise in addressing this challenge. In this study, we aimed to determine the optimal combination and sequence of SEC and DGUC for isolating sEVs from small plasma volumes, in order to enhance both the efficiency and purity of the resulting isolates. To achieve this, we compared sEV isolation using two combinations: SEC-DGUC and DGUC-SEC, from unit volumes of 500 μl plasma. Both protocols successfully isolated high-purity sEVs; however, the SEC-DGUC combination yielded higher sEV protein and RNA content. We further characterized the isolated sEVs obtained from the SEC-DGUC protocol using flow cytometry and mass spectrometry to assess their quality and purity. In conclusion, the optimized SEC-DGUC protocol is efficient, highly reproducible, and well-suited for isolating high-purity sEVs from small blood volumes.
Collapse
Affiliation(s)
- Fang Kong
- School of Biological Sciences, Nanyang Technological University, SINGAPORE
| | - Megha Upadya
- School of Biological Sciences, Nanyang Technological University, SINGAPORE
| | - Andrew See Weng Wong
- Facility for Analysis, Characterisation, Testing and Simulation, Nanyang Technological University, SINGAPORE
| | - Rinkoo Dalan
- Lee Kong Chian School of Medicine, Nanyang Technological University, SINGAPORE
| | - Ming Dao
- School of Biological Sciences, Nanyang Technological University, SINGAPORE
- Department of Material Science and Engineering, Massachusetts Institute of Technology, USA
| |
Collapse
|
19
|
Ono M, Zhang H, Sone H, Itonaga M. Multiplex Quantification of Exosomes via Multiple Types of Nanobeads Labeling Combined with Laser Scanning Detection. Anal Chem 2023; 95:15577-15584. [PMID: 37812687 DOI: 10.1021/acs.analchem.3c02374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/11/2023]
Abstract
In recent years, exosomes have attracted attention in many aspects from basic research to clinical application, including therapeutic reagents or biomarkers for liquid biopsy. The increasing understanding of exosome's heterogeneous properties is expected to lead to more advanced exosome research, and there is therefore a need for a multiplex system that can easily classify and analyze exosomes in complex biological samples according to their properties. In this study, we developed a simple and sensitive multiplexed exosome quantification system based on ExoCounter, an exosome quantification system utilizing optical disk technology, by introducing nanobeads made of different materials as exosome labeling substances. The refractive indices suitable for nanobead materials were analyzed by computer simulation of optical diffraction generated by nanobeads. The results showed that polymer (FG), Au, and Ag nanobeads exhibited superior discrimination capability in terms of the amplitude and polarity of detection pulses generated by each nanobead. The specificity and detection sensitivity of three types of nanobeads were confirmed by detecting HER2-positive exosomes with anti-HER2 antibody-conjugated nanobeads. Furthermore, CD147-positive, HER2-positive, and CD81-positive exosomes in 12.5 μL of serum were simultaneously quantified with high discrimination performance using the anti-CD147 antibody, anti-HER2 antibody, or anti-CD81 antibody conjugated for FG beads, Au nanobeads, or Ag nanobeads, respectively. A limit of detection was also evaluated as low as 210 exosomes/μL. This system is a promising tool for advanced exosome research because it enables multiplexed detection of heterogeneous exosomes in serum with high specificity, accuracy, and sensitivity without purification.
Collapse
Affiliation(s)
- Masayuki Ono
- Graduate School of Science and Technology, Gunma University, 1-5-1 Tenjin-cho, Kiryu, Gunma 376-8515, Japan
- Future Creation Research Laboratory, JVCKENWOOD Corporation, 58-7, Shinmei-cho, Yokosuka, Kanagawa 239-8550, Japan
| | - Hui Zhang
- Graduate School of Science and Technology, Gunma University, 1-5-1 Tenjin-cho, Kiryu, Gunma 376-8515, Japan
| | - Hayato Sone
- Graduate School of Science and Technology, Gunma University, 1-5-1 Tenjin-cho, Kiryu, Gunma 376-8515, Japan
| | - Makoto Itonaga
- Healthcare Business Division, JVCKENWOOD Corporation, 58-7 Shinmei-cho, Yokosuka, Kanagawa 239-8550, Japan
| |
Collapse
|
20
|
Brahmer A, Geiß C, Lygeraki A, Neuberger E, Tzaridis T, Nguyen TT, Luessi F, Régnier-Vigouroux A, Hartmann G, Simon P, Endres K, Bittner S, Reiners KS, Krämer-Albers EM. Assessment of technical and clinical utility of a bead-based flow cytometry platform for multiparametric phenotyping of CNS-derived extracellular vesicles. Cell Commun Signal 2023; 21:276. [PMID: 37803478 PMCID: PMC10559539 DOI: 10.1186/s12964-023-01308-9] [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: 07/17/2023] [Accepted: 09/06/2023] [Indexed: 10/08/2023] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) originating from the central nervous system (CNS) can enter the blood stream and carry molecules characteristic of disease states. Therefore, circulating CNS-derived EVs have the potential to serve as liquid-biopsy markers for early diagnosis and follow-up of neurodegenerative diseases and brain tumors. Monitoring and profiling of CNS-derived EVs using multiparametric analysis would be a major advance for biomarker as well as basic research. Here, we explored the performance of a multiplex bead-based flow-cytometry assay (EV Neuro) for semi-quantitative detection of CNS-derived EVs in body fluids. METHODS EVs were separated from culture of glioblastoma cell lines (LN18, LN229, NCH82) and primary human astrocytes and measured at different input amounts in the MACSPlex EV Kit Neuro, human. In addition, EVs were separated from blood samples of small cohorts of glioblastoma (GB), multiple sclerosis (MS) and Alzheimer's disease patients as well as healthy controls (HC) and subjected to the EV Neuro assay. To determine statistically significant differences between relative marker signal intensities, an unpaired samples t-test or Wilcoxon rank sum test were computed. Data were subjected to tSNE, heatmap clustering, and correlation analysis to further explore the relationships between disease state and EV Neuro data. RESULTS Glioblastoma cell lines and primary human astrocytes showed distinct EV profiles. Signal intensities were increasing with higher EV input. Data normalization improved identification of markers that deviate from a common profile. Overall, patient blood-derived EV marker profiles were constant, but individual EV populations were significantly increased in disease compared to healthy controls, e.g. CD36+EVs in glioblastoma and GALC+EVs in multiple sclerosis. tSNE and heatmap clustering analysis separated GB patients from HC, but not MS patients from HC. Correlation analysis revealed a potential association of CD107a+EVs with neurofilament levels in blood of MS patients and HC. CONCLUSIONS The semi-quantitative EV Neuro assay demonstrated its utility for EV profiling in complex samples. However, reliable statistical results in biomarker studies require large sample cohorts and high effect sizes. Nonetheless, this exploratory trial confirmed the feasibility of discovering EV-associated biomarkers and monitoring circulating EV profiles in CNS diseases using the EV Neuro assay. Video Abstract.
Collapse
Affiliation(s)
- Alexandra Brahmer
- Cellular Neurobiology, Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University of Mainz, Mainz, Germany.
- Department of Sports Medicine, Rehabilitation and Disease Prevention, Institute of Sports Sciences, Johannes Gutenberg University of Mainz, Mainz, Germany.
| | - Carsten Geiß
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Andriani Lygeraki
- Cellular Neurobiology, Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Elmo Neuberger
- Department of Sports Medicine, Rehabilitation and Disease Prevention, Institute of Sports Sciences, Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Theophilos Tzaridis
- Division of Clinical Neurooncology, Department of Neurology, Center of Integrated Oncology Aachen- Bonn-Cologne-Düsseldorf, Partner Site Bonn, University of Bonn, Bonn, Germany
| | - Tinh Thi Nguyen
- Department of Psychiatry and Psychotherapy, University Medical Center Mainz, Mainz, Germany
- Institute of Molecular Biology, Mainz, Germany
| | - Felix Luessi
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Anne Régnier-Vigouroux
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Gunther Hartmann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Perikles Simon
- Department of Sports Medicine, Rehabilitation and Disease Prevention, Institute of Sports Sciences, Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center Mainz, Mainz, Germany
| | - Stefan Bittner
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Katrin S Reiners
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Eva-Maria Krämer-Albers
- Cellular Neurobiology, Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University of Mainz, Mainz, Germany.
| |
Collapse
|
21
|
Hagey DW, Ojansivu M, Bostancioglu BR, Saher O, Bost JP, Gustafsson MO, Gramignoli R, Svahn M, Gupta D, Stevens MM, Görgens A, El Andaloussi S. The cellular response to extracellular vesicles is dependent on their cell source and dose. SCIENCE ADVANCES 2023; 9:eadh1168. [PMID: 37656796 DOI: 10.1126/sciadv.adh1168] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 07/31/2023] [Indexed: 09/03/2023]
Abstract
Extracellular vesicles (EVs) have been established to play important roles in cell-cell communication and shown promise as therapeutic agents. However, we still lack a basic understanding of how cells respond upon exposure to EVs from different cell sources at various doses. Thus, we treated fibroblasts with EVs from 12 different cell sources at doses between 20 and 200,000 per cell, analyzed their transcriptional effects, and functionally confirmed the findings in various cell types in vitro, and in vivo using single-cell RNA sequencing. Unbiased global analysis revealed EV dose to have a more significant effect than cell source, such that high doses down-regulated exocytosis and up-regulated lysosomal activity. However, EV cell source-specific responses were observed at low doses, and these reflected the activities of the EV's source cells. Last, we assessed EV-derived transcript abundance and found that immune cell-derived EVs were most associated with recipient cells. Together, this study provides important insights into the cellular response to EVs.
Collapse
Affiliation(s)
- Daniel W Hagey
- Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer Center, Stockholm, Sweden
| | - Miina Ojansivu
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Beklem R Bostancioglu
- Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer Center, Stockholm, Sweden
| | - Osama Saher
- Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer Center, Stockholm, Sweden
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Jeremy P Bost
- Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer Center, Stockholm, Sweden
| | - Manuela O Gustafsson
- Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer Center, Stockholm, Sweden
| | - Roberto Gramignoli
- Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | | | - Dhanu Gupta
- Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer Center, Stockholm, Sweden
- Department of Paediatrics, University of Oxford, Oxford OX3 7TY, UK
| | - Molly M Stevens
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
- Department of Materials, Department of Bioengineering, Institute of Biomedical Engineering, Imperial College London, London, UK
| | - André Görgens
- Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer Center, Stockholm, Sweden
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Samir El Andaloussi
- Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer Center, Stockholm, Sweden
| |
Collapse
|
22
|
Zheng W, Rädler J, Sork H, Niu Z, Roudi S, Bost JP, Görgens A, Zhao Y, Mamand DR, Liang X, Wiklander OPB, Lehto T, Gupta D, Nordin JZ, El Andaloussi S. Identification of scaffold proteins for improved endogenous engineering of extracellular vesicles. Nat Commun 2023; 14:4734. [PMID: 37550290 PMCID: PMC10406850 DOI: 10.1038/s41467-023-40453-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 07/27/2023] [Indexed: 08/09/2023] Open
Abstract
Extracellular vesicles (EVs) are gaining ground as next-generation drug delivery modalities. Genetic fusion of the protein of interest to a scaffold protein with high EV-sorting ability represents a robust cargo loading strategy. To address the paucity of such scaffold proteins, we leverage a simple and reliable assay that can distinguish intravesicular cargo proteins from surface- as well as non-vesicular proteins and compare the EV-sorting potential of 244 candidate proteins. We identify 24 proteins with conserved EV-sorting abilities across five types of producer cells. TSPAN2 and TSPAN3 emerge as lead candidates and outperform the well-studied CD63 scaffold. Importantly, these engineered EVs show promise as delivery vehicles in cell cultures and mice as demonstrated by efficient transfer of luminal cargo proteins as well as surface display of different functional entities. The discovery of these scaffolds provides a platform for EV-based engineering.
Collapse
Affiliation(s)
- Wenyi Zheng
- Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Julia Rädler
- Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Helena Sork
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Zheyu Niu
- Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Samantha Roudi
- Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Jeremy P Bost
- Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - André Görgens
- Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ying Zhao
- Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
- Clinical Research Center, Karolinska University Hospital, Stockholm, Sweden
| | - Doste R Mamand
- Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Xiuming Liang
- Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Oscar P B Wiklander
- Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Taavi Lehto
- Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Dhanu Gupta
- Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Joel Z Nordin
- Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Samir El Andaloussi
- Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden.
| |
Collapse
|
23
|
Li C, Sun C, Lohcharoenkal W, Ali MM, Xing P, Zheng W, Görgens A, Gustafsson MO, El Andaloussi S, Sonkoly E, Pivarcsi A. Cutaneous squamous cell carcinoma-derived extracellular vesicles exert an oncogenic role by activating cancer-associated fibroblasts. Cell Death Discov 2023; 9:260. [PMID: 37495566 PMCID: PMC10372068 DOI: 10.1038/s41420-023-01555-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 07/04/2023] [Accepted: 07/12/2023] [Indexed: 07/28/2023] Open
Abstract
Cutaneous squamous cell carcinoma (cSCC) is a fast-increasing cancer with metastatic potential. Extracellular vesicles (EVs) are small membrane-bound vesicles that play important roles in intercellular communication, particularly in the tumor microenvironment (TME). Here we report that cSCC cells secrete an increased number of EVs relative to normal human epidermal keratinocytes (NHEKs) and that interfering with the capacity of cSCC to secrete EVs inhibits tumor growth in vivo in a xenograft model of human cSCC. Transcriptome analysis of tumor xenografts by RNA-sequencing enabling the simultaneous quantification of both the human and the mouse transcripts revealed that impaired EV-production of cSCC cells prominently altered the phenotype of stromal cells, in particular genes related to extracellular matrix (ECM)-formation and epithelial-mesenchymal transition (EMT). In line with these results, co-culturing of human dermal fibroblasts (HDFs) with cSCC cells, but not with normal keratinocytes in vitro resulted in acquisition of cancer-associated fibroblast (CAF) phenotype. Interestingly, EVs derived from metastatic cSCC cells, but not primary cSCCs or NHEKs, were efficient in converting HDFs to CAFs. Multiplex bead-based flow cytometry assay and mass-spectrometry (MS)-based proteomic analyses revealed the heterogenous cargo of cSCC-derived EVs and that especially EVs derived from metastatic cSCCs carry proteins associated with EV-biogenesis, EMT, and cell migration. Mechanistically, EVs from metastatic cSCC cells result in the activation of TGFβ signaling in HDFs. Altogether, our study suggests that cSCC-derived EVs mediate cancer-stroma communication, in particular the conversion of fibroblasts to CAFs, which eventually contribute to cSCC progression.
Collapse
Affiliation(s)
- Chen Li
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- Dermatology and Venereology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Chengxi Sun
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- Department of Clinical Laboratory, Cheeloo College of Medicine, Shandong University, 250012, Jinan, Shandong, China
| | - Warangkana Lohcharoenkal
- Unit of Dermatology and Venerology, Department of Medicine, Karolinska Institutet, Stockholm, SE, 17176, Sweden
| | - Mohamad Moustafa Ali
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Pengwei Xing
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Wenyi Zheng
- Department of Laboratory Medicine, Clinical Research Center, Karolinska Institutet, Stockholm, Sweden
| | - André Görgens
- Department of Laboratory Medicine, Clinical Research Center, Karolinska Institutet, Stockholm, Sweden
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Manuela O Gustafsson
- Department of Laboratory Medicine, Clinical Research Center, Karolinska Institutet, Stockholm, Sweden
| | - Samir El Andaloussi
- Department of Laboratory Medicine, Clinical Research Center, Karolinska Institutet, Stockholm, Sweden
| | - Enikö Sonkoly
- Dermatology and Venereology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- Unit of Dermatology and Venerology, Department of Medicine, Karolinska Institutet, Stockholm, SE, 17176, Sweden
| | - Andor Pivarcsi
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden.
- Dermatology and Venereology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden.
- Unit of Dermatology and Venerology, Department of Medicine, Karolinska Institutet, Stockholm, SE, 17176, Sweden.
| |
Collapse
|
24
|
Vannini I, Rossi T, Melloni M, Valgiusti M, Urbini M, Passardi A, Bartolini G, Gallio C, Azzali I, Bandini S, Ancarani V, Montanaro L, Frassineti GL, Fabbri F, Rapposelli IG. Analysis of EVs from patients with advanced pancreatic cancer identifies antigens and miRNAs with predictive value. Mol Ther Methods Clin Dev 2023; 29:473-482. [PMID: 37273899 PMCID: PMC10238807 DOI: 10.1016/j.omtm.2023.05.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 05/09/2023] [Indexed: 06/06/2023]
Abstract
The identification of predictive factors for treatment of pancreatic cancer (PC) is an unmet clinical need. In the present work, we analyzed blood-derived extracellular vesicles (EVs) from patients with advanced PC in order to find a molecular signature predictive of response to therapy. We analyzed samples from 21 patients with advanced PC, all receiving first-line treatment with gemcitabine + nab-paclitaxel. Isolated EVs have been analyzed, and the results of laboratory have been matched with clinical data in order to investigate possible predictive factors. EV concentration and size were similar between responder and non-responder patients. Analysis of 37 EV surface epitopes showed a decreased expression of SSEA4 and CD81 in responder patients. We detected more than 450 expressed miRNAs in EVs. A comparative survey between responder and non-responder patients showed that at least 44 miRNAs were differently expressed. Some of these miRNAs have already been observed in relation to the survival and gemcitabine sensitivity of tumor cells. In conclusion, we showed the ability of our approach to identify EV-derived biomarkers with predictive value for therapy response in PC. Our findings are worthy of further investigation, including the analysis of samples from patients treated with different schedules and in different settings.
Collapse
Affiliation(s)
- Ivan Vannini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Tania Rossi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Mattia Melloni
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Martina Valgiusti
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Milena Urbini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Alessandro Passardi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Giulia Bartolini
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Chiara Gallio
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Irene Azzali
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Sara Bandini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Valentina Ancarani
- Immunotherapy-Cell Therapy and Biobank Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Lorenzo Montanaro
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum - University of Bologna, Bologna, Italy
- Departmental Program in Laboratory Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Giovanni Luca Frassineti
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Francesco Fabbri
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Ilario Giovanni Rapposelli
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| |
Collapse
|
25
|
Giovanazzi A, van Herwijnen MJC, Kleinjan M, van der Meulen GN, Wauben MHM. Surface protein profiling of milk and serum extracellular vesicles unveils body fluid-specific signatures. Sci Rep 2023; 13:8758. [PMID: 37253799 DOI: 10.1038/s41598-023-35799-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 05/24/2023] [Indexed: 06/01/2023] Open
Abstract
Cell-derived extracellular vesicles (EVs) are currently in the limelight as potential disease biomarkers. The promise of EV-based liquid biopsy resides in the identification of specific disease-associated EV signatures. Knowing the reference EV profile of a body fluid can facilitate the identification of such disease-associated EV-biomarkers. With this aim, we purified EVs from paired human milk and serum samples and used the MACSPlex bead-based flow-cytometry assay to capture EVs on bead-bound antibodies specific for a certain surface protein, followed by EV detection by the tetraspanins CD9, CD63, and CD81. Using this approach we identified body fluid-specific EV signatures, e.g. breast epithelial cell signatures in milk EVs and platelet signatures in serum EVs, as well as body fluid-specific markers associated to immune cells and stem cells. Interestingly, comparison of pan-tetraspanin detection (simultaneous CD9, CD63 and CD81 detection) and single tetraspanin detection (detection by CD9, CD63 or CD81) also unveiled body fluid-specific tetraspanin distributions on EVs. Moreover, certain EV surface proteins were associated with a specific tetraspanin distribution, which could be indicative of the biogenesis route of this EV subset. Altogether, the identified body fluid-specific EV profiles can contribute to study EV profile deviations in these fluids during disease processes.
Collapse
Affiliation(s)
- Alberta Giovanazzi
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
- TRAIN-EV Marie Skłodowska-Curie Action-ITN, Utrecht, The Netherlands
| | - Martijn J C van Herwijnen
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Marije Kleinjan
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | | | - Marca H M Wauben
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
- TRAIN-EV Marie Skłodowska-Curie Action-ITN, Utrecht, The Netherlands.
| |
Collapse
|
26
|
He Y, Xing Y, Jiang T, Wang J, Sang S, Rong H, Yu F. Fluorescence labeling of extracellular vesicles for diverse bio-applications in vitro and in vivo. Chem Commun (Camb) 2023; 59:6609-6626. [PMID: 37161668 DOI: 10.1039/d3cc00998j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Extracellular vesicles (EVs) are nanosized vesicles enclosed in a lipid membrane that are sustainably released by nearly all cell types. EVs have been deemed as valuable biomarkers for diagnostics and effective drug carriers, owing to the physiological function of transporting biomolecules for intercellular communication. To investigate their biological properties, efficient labeling strategies have been constructed for EV research, among which fluorescence labeling exerts a powerful function due to the capability of visualizing the nanovesicles with high sensitivity both in vitro and in vivo. In one aspect, with the help of functional fluorescence tags, EVs could be differentiated and categorized in vitro by various analytical techniques, which exert vital roles in disease diagnosis, prognosis, and treatment monitoring. Additionally, innovative EV reporters have been utilized for visualizing EVs, in combination with powerful microscopy techniques, which provide potential tools for investigating the dynamic events of EV release and intercellular communication in suitable animal models. In this feature article, we survey the latest advances regarding EV fluorescence labeling strategies and their application in biomedical application and in vivo biology investigation, highlighting the progresses in individual EV imaging. Finally, the challenges and future perspectives in unravelling EV physiological properties and further biomedical application are discussed.
Collapse
Affiliation(s)
- Yun He
- Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China.
| | - Yanlong Xing
- Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China.
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| | - Tongmeng Jiang
- Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China.
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| | - Juan Wang
- Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China.
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| | - Shenggang Sang
- Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China.
| | - Hong Rong
- Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China.
| | - Fabiao Yu
- Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China.
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| |
Collapse
|
27
|
Li L, Mussack V, Görgens A, Pepeldjiyska E, Hartz AS, Aslan H, Rackl E, Rank A, Schmohl J, El Andaloussi S, Pfaffl MW, Schmetzer H. The potential role of serum extracellular vesicle derived small RNAs in AML research as non-invasive biomarker. NANOSCALE ADVANCES 2023; 5:1691-1705. [PMID: 36926576 PMCID: PMC10012871 DOI: 10.1039/d2na00959e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 02/13/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Extracellular vesicles (EV) are cell-derived vesicles released by all cells in health and disease. Accordingly, EVs are also released by cells in acute myeloid leukemia (AML), a hematologic malignancy characterized by uncontrolled growth of immature myeloid cells, and these EVs likely carry markers and molecular cargo reflecting the malignant transformation occurring in diseased cells. Monitoring antileukemic or proleukemic processes during disease development and treatment is essential. Therefore, EVs and EV-derived microRNA (miRNA) from AML samples were explored as biomarkers to distinguish disease-related patterns ex vivo or in vivo. METHODOLOGY EVs were purified from serum of healthy (H) volunteers and AML patients by immunoaffinity. EV surface protein profiles were analyzed by multiplex bead-based flow cytometry (MBFCM) and total RNA was isolated from EVs prior to miRNA profiling via small RNA sequencing. RESULTS MBFCM revealed different surface protein patterns in H versus AML EVs. miRNA analysis showed individual as well as highly dysregulated patterns in H and AML samples. CONCLUSIONS In this study, we provide a proof-of-concept for the discriminative potential of EV derived miRNA profiles as biomarkers in H versus AML samples.
Collapse
Affiliation(s)
- Lin Li
- Immune-Modulation, Medical Department III, University Hospital of Munich Marchioninistraße 15 81377 Munich Germany +49 89 4400 76137 +49 89 4400 73137
| | - Veronika Mussack
- Department of Animal Physiology and Immunology, TUM School of Life Sciences Weihenstephan, Technical University of Munich Freising Germany
| | - André Görgens
- Department of Laboratory Medicine, Division of Biomolecular and Cellular Medicine, Karolinska Institutet Stockholm Sweden
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen Essen Germany
| | - Elena Pepeldjiyska
- Immune-Modulation, Medical Department III, University Hospital of Munich Marchioninistraße 15 81377 Munich Germany +49 89 4400 76137 +49 89 4400 73137
| | - Anne Sophie Hartz
- Immune-Modulation, Medical Department III, University Hospital of Munich Marchioninistraße 15 81377 Munich Germany +49 89 4400 76137 +49 89 4400 73137
| | - Hazal Aslan
- Immune-Modulation, Medical Department III, University Hospital of Munich Marchioninistraße 15 81377 Munich Germany +49 89 4400 76137 +49 89 4400 73137
| | - Elias Rackl
- Immune-Modulation, Medical Department III, University Hospital of Munich Marchioninistraße 15 81377 Munich Germany +49 89 4400 76137 +49 89 4400 73137
| | - Andreas Rank
- Department of Hematology and Oncology, University Hospital of Augsburg Augsburg Germany
| | - Jörg Schmohl
- Department of Hematology and Oncology, Hospital of Stuttgart Stuttgart Germany
| | - Samir El Andaloussi
- Department of Laboratory Medicine, Division of Biomolecular and Cellular Medicine, Karolinska Institutet Stockholm Sweden
| | - Michael W Pfaffl
- Department of Animal Physiology and Immunology, TUM School of Life Sciences Weihenstephan, Technical University of Munich Freising Germany
| | - Helga Schmetzer
- Immune-Modulation, Medical Department III, University Hospital of Munich Marchioninistraße 15 81377 Munich Germany +49 89 4400 76137 +49 89 4400 73137
| |
Collapse
|
28
|
Ransom MA, Bunn KE, Negretti NM, Jetter CS, Bressman ZJ, Sucre JMS, Pua HH. Developmental trajectory of extracellular vesicle characteristics from the lungs of preterm infants. Am J Physiol Lung Cell Mol Physiol 2023; 324:L385-L392. [PMID: 36719083 PMCID: PMC10026990 DOI: 10.1152/ajplung.00389.2022] [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: 11/23/2022] [Revised: 01/23/2023] [Accepted: 01/24/2023] [Indexed: 02/01/2023] Open
Abstract
Extracellular vesicles (EVs) are secreted lipid-enclosed particles that have emerged as potential biomarkers and therapeutic agents in lung disease, including bronchopulmonary dysplasia (BPD), a leading complication of preterm birth. Many unanswered questions remain about the content and cargo of EVs in premature infants and their role in lung development. To characterize EVs during human lung development, tracheal aspirates were collected from premature neonates between 22 and 35 wk gestational age and analyzed via nanoparticle tracking analysis, electron microscopy, and bead-based flow cytometry. EVs were detectable across late canalicular through saccular stages of lung development, demonstrating larger sizes earlier in gestation. EVs contained an abundance of the EV-enriched tetraspanins CD9, CD63, and CD81, as well as epithelial cell and immune cell markers. Increases in select surface proteins (CD24 and CD14) on EVs were associated with gestational age and with the risk of BPD. Finally, query of expression data obtained from epithelial cells in a single-cell atlas of murine lung development found that epithelial EV marker expression also changes with developmental time. Together, these data demonstrate an association between EV profile and lung development and provide a foundation for future functional classification of EVs, with the goal of determining their role in cell signaling during development and harnessing their potential as a new therapeutic target in BPD.
Collapse
Affiliation(s)
- Meaghan A Ransom
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Kaitlyn E Bunn
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Nicholas M Negretti
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Christopher S Jetter
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Zachary J Bressman
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Jennifer M S Sucre
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States
| | - Heather H Pua
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| |
Collapse
|
29
|
Chen M, Lin S, Zhou C, Cui D, Haick H, Tang N. From Conventional to Microfluidic: Progress in Extracellular Vesicle Separation and Individual Characterization. Adv Healthc Mater 2023; 12:e2202437. [PMID: 36541411 DOI: 10.1002/adhm.202202437] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 12/07/2022] [Indexed: 12/24/2022]
Abstract
Extracellular vesicles (EVs) are nanoscale membrane vesicles, which contain a wide variety of cargo such as proteins, miRNAs, and lipids. A growing body of evidence suggests that EVs are promising biomarkers for disease diagnosis and therapeutic strategies. Although the excellent clinical value, their use in personalized healthcare practice is not yet feasible due to their highly heterogeneous nature. Taking the difficulty of isolation and the small size of EVs into account, the characterization of EVs at a single-particle level is both imperative and challenging. In a bid to address this critical point, more research has been directed into a microfluidic platform because of its inherent advantages in sensitivity, specificity, and throughput. This review discusses the biogenesis and heterogeneity of EVs and takes a broad view of state-of-the-art advances in microfluidics-based EV research, including not only EV separation, but also the single EV characterization of biophysical detection and biochemical analysis. To highlight the advantages of microfluidic techniques, conventional technologies are included for comparison. The current status of artificial intelligence (AI) for single EV characterization is then presented. Furthermore, the challenges and prospects of microfluidics and its combination with AI applications in single EV characterization are also discussed. In the foreseeable future, recent breakthroughs in microfluidic platforms are expected to pave the way for single EV analysis and improve applications for precision medicine.
Collapse
Affiliation(s)
- Mingrui Chen
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Shujing Lin
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Cheng Zhou
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Daxiang Cui
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Hossam Haick
- Department of Chemical Engineering and Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Ning Tang
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| |
Collapse
|
30
|
Hagey DW, El Andaloussi S. The promise and challenges of extracellular vesicles in the diagnosis of neurodegenerative diseases. HANDBOOK OF CLINICAL NEUROLOGY 2023; 193:227-241. [PMID: 36803813 DOI: 10.1016/b978-0-323-85555-6.00014-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Extracellular vesicles (EVs) have emerged as essential means of intercommunication for all cell types, and their role in CNS physiology is increasingly appreciated. Accumulating evidence has demonstrated that EVs play important roles in neural cell maintenance, plasticity, and growth. However, EVs have also been demonstrated to spread amyloids and inflammation characteristic of neurodegenerative disease. Such dual roles suggest that EVs may be prime candidates for neurodegenerative disease biomarker analysis. This is supported by several intrinsic properties of EVs: Populations can be enriched by capturing surface proteins from their cell of origin, their diverse cargo represent the complex intracellular states of the cells they derive from, and they can pass the blood-brain barrier. Despite this promise, there are important questions outstanding in this young field that will need to be answered before it can fulfill its potential. Namely, overcoming the technical challenges of isolating rare EV populations, the difficulties inherent in detecting neurodegeneration, and the ethical considerations of diagnosing asymptomatic individuals. Although daunting, succeeding to answer these questions has the potential to provide unprecedented insight and improved treatment of neurodegenerative disease in the future.
Collapse
Affiliation(s)
- Daniel W Hagey
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.
| | | |
Collapse
|
31
|
Bauer FN, Tertel T, Stambouli O, Wang C, Dittrich R, Staubach S, Börger V, Hermann DM, Brandau S, Giebel B. CD73 activity of mesenchymal stromal cell-derived extracellular vesicle preparations is detergent-resistant and does not correlate with immunomodulatory capabilities. Cytotherapy 2023; 25:138-147. [PMID: 36244910 DOI: 10.1016/j.jcyt.2022.09.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 09/23/2022] [Accepted: 09/23/2022] [Indexed: 01/18/2023]
Abstract
BACKGROUND AIMS Extracellular vesicles (EVs) derived from human mesenchymal stromal cells (MSCs) show immunomodulatory activity in different assays both in vitro and in vivo. In previous work, the authors compared the immunomodulatory potential of independent MSC-EV preparations in a multi-donor mixed lymphocyte reaction (mdMLR) assay and an optimized steroid-refractory acute graft-versus-host disease (aGVHD) mouse model. The authors observed that only a proportion of the MSC-EV preparations showed immunomodulatory capabilities and demonstrated that only MSC-EV preparations with mdMLR immunomodulating activities were able to suppress aGVHD symptoms in vivo and vice versa. Since the mdMLR assay is complex and depends on primary human cells of different donors, the authors sought to establish an assay that is much easier to standardize and fulfills the requirements for becoming qualified as a potency assay. METHODS The bona fide MSC antigen CD73 possesses ecto-5'-nucleotidase activity that cleaves pro-inflammatory extracellular adenosine monophosphate into anti-inflammatory adenosine and free phosphate. To test whether the ecto-5'-nucleotidase activity of the MSC-EV preparations reflected their immunomodulatory potential, the authors adopted an enzymatic assay that monitors the ecto-5'-nucleotidase activity of CD73 in a quantitative manner and compared the activity of well-characterized MSC-EV preparations containing or lacking mdMLR immunomodulatory activity. RESULTS The authors showed that the ecto-5'-nucleotidase activity of the MSC-EV preparations did not correlate with their ability to modulate T-cell responses in the mdMLR assay and thus with their potency in improving disease symptomatology in the optimized mouse aGVHD model. Furthermore, the ecto-5'-nucleotidase activity was resistant to EV-destroying detergent treatment. CONCLUSIONS Ecto-5'-nucleotidase activity neither reflects the potency of the authors' MSC-EV preparations nor provides any information about the integrity of the respective EVs. Thus, ecto-5'-nucleotidase enzyme activity is not indicative for the immunomodulatory potency of the authors' MSC-EV products. The development of appropriate potency assays for MSC-EV products remains challenging.
Collapse
Affiliation(s)
- Fabiola Nardi Bauer
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Tobias Tertel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Oumaima Stambouli
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Chen Wang
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Robin Dittrich
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Simon Staubach
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Verena Börger
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Dirk M Hermann
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Sven Brandau
- Department of Otorhinolaryngology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
32
|
Welsh JA, Arkesteijn GJA, Bremer M, Cimorelli M, Dignat-George F, Giebel B, Görgens A, Hendrix A, Kuiper M, Lacroix R, Lannigan J, van Leeuwen TG, Lozano-Andrés E, Rao S, Robert S, de Rond L, Tang VA, Tertel T, Yan X, Wauben MHM, Nolan JP, Jones JC, Nieuwland R, van der Pol E. A compendium of single extracellular vesicle flow cytometry. J Extracell Vesicles 2023; 12:e12299. [PMID: 36759917 PMCID: PMC9911638 DOI: 10.1002/jev2.12299] [Citation(s) in RCA: 58] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 11/29/2022] [Accepted: 12/17/2022] [Indexed: 02/11/2023] Open
Abstract
Flow cytometry (FCM) offers a multiparametric technology capable of characterizing single extracellular vesicles (EVs). However, most flow cytometers are designed to detect cells, which are larger than EVs. Whereas cells exceed the background noise, signals originating from EVs partly overlap with the background noise, thereby making EVs more difficult to detect than cells. This technical mismatch together with complexity of EV-containing fluids causes limitations and challenges with conducting, interpreting and reproducing EV FCM experiments. To address and overcome these challenges, researchers from the International Society for Extracellular Vesicles (ISEV), International Society for Advancement of Cytometry (ISAC), and the International Society on Thrombosis and Haemostasis (ISTH) joined forces and initiated the EV FCM working group. To improve the interpretation, reporting, and reproducibility of future EV FCM data, the EV FCM working group published an ISEV position manuscript outlining a framework of minimum information that should be reported about an FCM experiment on single EVs (MIFlowCyt-EV). However, the framework contains limited background information. Therefore, the goal of this compendium is to provide the background information necessary to design and conduct reproducible EV FCM experiments. This compendium contains background information on EVs, the interaction between light and EVs, FCM hardware, experimental design and preanalytical procedures, sample preparation, assay controls, instrument data acquisition and calibration, EV characterization, and data reporting. Although this compendium focuses on EVs, many concepts and explanations could also be applied to FCM detection of other particles within the EV size range, such as bacteria, lipoprotein particles, milk fat globules, and viruses.
Collapse
Affiliation(s)
- Joshua A Welsh
- Translational Nanobiology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ger J A Arkesteijn
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Michel Bremer
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Michael Cimorelli
- Vesicle Observation Center, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Experimental Clinical Chemistry, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Department of Chemical Engineering, Drexel University, Philadelphia, Pennsylvania, USA
| | - Françoise Dignat-George
- Aix Marseille Univ, INSERM, INRAE, C2VN, UFR de Pharmacie, Marseille, France
- Hematology and Vascular Biology Department, CHU La Conception, Assistance Publique-Hôpitaux de Marseille, Marseille, France
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - André Görgens
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Clinical Research Center, Department for Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Evox Therapeutics Ltd, Oxford, UK
| | - An Hendrix
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Martine Kuiper
- Vesicle Observation Center, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Experimental Clinical Chemistry, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Biomedical Engineering & Physics, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Dutch Metrology Institute, VSL, Delft, The Netherlands
| | - Romaric Lacroix
- Aix Marseille Univ, INSERM, INRAE, C2VN, UFR de Pharmacie, Marseille, France
- Hematology and Vascular Biology Department, CHU La Conception, Assistance Publique-Hôpitaux de Marseille, Marseille, France
| | - Joanne Lannigan
- Flow Cytometry Support Services, LLC, Arlington, Virginia, USA
| | - Ton G van Leeuwen
- Vesicle Observation Center, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Biomedical Engineering & Physics, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Estefanía Lozano-Andrés
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Shoaib Rao
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Stéphane Robert
- Aix Marseille Univ, INSERM, INRAE, C2VN, UFR de Pharmacie, Marseille, France
- Hematology and Vascular Biology Department, CHU La Conception, Assistance Publique-Hôpitaux de Marseille, Marseille, France
| | - Leonie de Rond
- Vesicle Observation Center, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Experimental Clinical Chemistry, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Biomedical Engineering & Physics, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Vera A Tang
- Flow Cytometry & Virometry Core Facility, Faculty of Medicine, Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Tobias Tertel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Xiaomei Yan
- MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Marca H M Wauben
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - John P Nolan
- Scintillon Institute, San Diego, California, USA
- Cellarcus Biosciences, San Diego, California, USA
| | - Jennifer C Jones
- Translational Nanobiology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Rienk Nieuwland
- Vesicle Observation Center, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Experimental Clinical Chemistry, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, The Netherlands
| | - Edwin van der Pol
- Vesicle Observation Center, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Experimental Clinical Chemistry, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Biomedical Engineering & Physics, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| |
Collapse
|
33
|
Korchak JA, Wiest EF, Zubair AC. How do we assess batch-to-batch consistency between extracellular vesicle products? Transfusion 2023; 63:279-287. [PMID: 36329638 DOI: 10.1111/trf.17156] [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: 03/03/2022] [Revised: 09/01/2022] [Accepted: 09/29/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Extracellular vesicles (EVs) have gained interest in the field of regenerative and transfusion medicine. Specifically, current Good Manufacturing Practice (cGMP)-grade EVs produced by mesenchymal stromal cells (MSCs) are an intriguing option for cell-free therapeutics. With the development of cGMP-grade EV products, a simple and reliable method for batch-to-batch consistency is needed. STUDY DESIGN AND METHODS The objective of this study was to validate a method to semiquantitatively assess the batch-to-batch consistency of isolated EVs. A multiplex bead-based flow cytometric assay containing 37 surface markers and 2 assay control antibody-coated capture beads was validated. Detection limits (n = 10 buffer samples), repeatability (n = 9 EV samples), and intra-observer reproducibility over 2 days (n = 10 EV batches) were assessed. A Spearman correlation matrix was used to evaluate the batch-to-batch consistency of independently isolated EV products (n = 37 surface markers). Batches with a Spearman correlation coefficient ≥0.9 and p < 0.05 were considered statistically indistinguishable from previous batches. RESULTS This assay demonstrated robust repeatability as well as intra- and inter-assay reproducibility. In-house batches of EVs were significantly correlated (r ≥ 0.90; p ≤ 1×10-14 ). Compared with buffer, EV batches had correlation coefficients near zero (r ≤ -0.10; p ≥ 0.12). Commercially sourced EVs significantly correlated with in-house EV batches, but fell below the 90% correlation cutoff (r ≤ 0.71; p ≤ 0.0004). DISCUSSION This time-efficient and technically simple assay offers a robust method of quality control for assessing the batch-to-batch reproducibility of cGMP-grade EV products.
Collapse
Affiliation(s)
- Jennifer A Korchak
- Center for Regenerative Biotherapeutics, Jacksonville, Florida, USA.,Department of Laboratory Medicine and Pathology, Jacksonville, Florida, USA
| | - Elani F Wiest
- Center for Regenerative Biotherapeutics, Jacksonville, Florida, USA
| | - Abba C Zubair
- Center for Regenerative Biotherapeutics, Jacksonville, Florida, USA.,Department of Laboratory Medicine and Pathology, Jacksonville, Florida, USA.,Sheikh Shakhbout Medical City-Mayo Clinic Joint Venture, Abu Dhabi, UAE
| |
Collapse
|
34
|
Del Real Mata C, Jeanne O, Jalali M, Lu Y, Mahshid S. Nanostructured-Based Optical Readouts Interfaced with Machine Learning for Identification of Extracellular Vesicles. Adv Healthc Mater 2023; 12:e2202123. [PMID: 36443009 DOI: 10.1002/adhm.202202123] [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: 08/22/2022] [Revised: 11/14/2022] [Indexed: 11/30/2022]
Abstract
Extracellular vesicles (EVs) are shed from cancer cells into body fluids, enclosing molecular information about the underlying disease with the potential for being the target cancer biomarker in emerging diagnosis approaches such as liquid biopsy. Still, the study of EVs presents major challenges due to their heterogeneity, complexity, and scarcity. Recently, liquid biopsy platforms have allowed the study of tumor-derived materials, holding great promise for early-stage diagnosis and monitoring of cancer when interfaced with novel adaptations of optical readouts and advanced machine learning analysis. Here, recent advances in labeled and label-free optical techniques such as fluorescence, plasmonic, and chromogenic-based systems interfaced with nanostructured sensors like nanoparticles, nanoholes, and nanowires, and diverse machine learning analyses are reviewed. The adaptability of the different optical methods discussed is compared and insights are provided into prospective avenues for the translation of the technological approaches for cancer diagnosis. It is discussed that the inherent augmented properties of nanostructures enhance the sensitivity of the detection of EVs. It is concluded by reviewing recent integrations of nanostructured-based optical readouts with diverse machine learning models as novel analysis ventures that can potentially increase the capability of the methods to the point of translation into diagnostic applications.
Collapse
Affiliation(s)
| | - Olivia Jeanne
- McGill University, Department of Bioengineering, Montreal, QC, H3A 0E9, Canada
| | - Mahsa Jalali
- McGill University, Department of Bioengineering, Montreal, QC, H3A 0E9, Canada
| | - Yao Lu
- McGill University, Department of Bioengineering, Montreal, QC, H3A 0E9, Canada
| | - Sara Mahshid
- McGill University, Department of Bioengineering, Montreal, QC, H3A 0E9, Canada
| |
Collapse
|
35
|
Stem Cell-derived Extracellular Vesicles: A Promising Nano Delivery Platform to the Brain? Stem Cell Rev Rep 2023; 19:285-308. [PMID: 36173500 DOI: 10.1007/s12015-022-10455-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2022] [Indexed: 02/07/2023]
Abstract
A very important cause of the frustration with drug therapy for central nervous system (CNS) diseases is the failure of drug delivery. The blood-brain barrier (BBB) prevents most therapeutic molecules from entering the brain while maintaining CNS homeostasis. Scientists are keen to develop new brain drug delivery systems to solve this dilemma. Extracellular vesicles (EVs), as a class of naturally derived nanoscale vesicles, have been extensively studied in drug delivery due to their superior properties. This review will briefly present current brain drug delivery strategies, including invasive and non-invasive techniques that target the brain, and the application of nanocarriers developed for brain drug delivery in recent years, especially EVs. The cellular origin of EVs affects the surface protein, size, yield, luminal composition, and other properties of EVs, which are also crucial in determining whether EVs are useful as drug carriers. Stem cell-derived EVs, which inherit the properties of parental cells and avoid the drawbacks of cell therapy, have always been favored by researchers. Thus, in this review, we will focus on the application of stem cell-derived EVs for drug delivery in the CNS. Various nucleic acids, proteins, and small-molecule drugs are loaded into EVs with or without modification and undergo targeted delivery to the brain to achieve their therapeutic effects. In addition, the challenges facing the clinical application of EVs as drug carriers will also be discussed. The directions of future efforts may be to improve drug loading efficiency and precise targeting.
Collapse
|
36
|
Jakl V, Ehmele M, Winkelmann M, Ehrenberg S, Eiseler T, Friemert B, Rojewski MT, Schrezenmeier H. A novel approach for large-scale manufacturing of small extracellular vesicles from bone marrow-derived mesenchymal stromal cells using a hollow fiber bioreactor. Front Bioeng Biotechnol 2023; 11:1107055. [PMID: 36761296 PMCID: PMC9904364 DOI: 10.3389/fbioe.2023.1107055] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 01/09/2023] [Indexed: 01/25/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are promising therapeutic candidates in a variety of diseases due to having immunomodulatory and pro-regenerative properties. In recent years, MSC-derived small extracellular vesicles (sEVs) have attracted increasing interest as a possible alternative to conventional cell therapy. However, translational processes of sEVs for clinical applications are still impeded by inconsistencies regarding isolation procedures and culture conditions. We systematically compared different methods for sEV isolation from conditioned media of ex vivo expanded bone marrow-derived MSCs and demonstrated considerable variability of quantity, purity, and characteristics of sEV preparations obtained by these methods. The combination of cross flow filtration with ultracentrifugation for sEV isolation resulted in sEVs with similar properties as compared to isolation by differential centrifugation combined with ultracentrifugation, the latter is still considered as gold standard for sEV isolation. In contrast, sEV isolation by a combination of precipitation with polyethylene glycol and ultracentrifugation as well as cross flow filtration and size exclusion chromatography resulted in sEVs with different characteristics, as shown by surface antigen expression patterns. The MSC culture requires a growth-promoting supplement, such as platelet lysate, which contains sEVs itself. We demonstrated that MSC culture with EV-depleted platelet lysate does not alter MSC characteristics, and conditioned media of such MSC cultures provide sEV preparations enriched for MSC-derived sEVs. The results from the systematic stepwise evaluation of various aspects were combined with culture of MSCs in a hollow fiber bioreactor. This resulted in a strategy using cross flow filtration with subsequent ultracentrifugation for sEV isolation. In conclusion, this workflow provides a semi-automated, efficient, large-scale-applicable, and good manufacturing practice (GMP)-grade approach for the generation of sEVs for clinical use. The use of EV-depleted platelet lysate is an option to further increase the purity of MSC-derived sEVs.
Collapse
Affiliation(s)
- Viktoria Jakl
- Institute for Transfusion Medicine, University Hospital Ulm, Ulm, Germany
| | - Melanie Ehmele
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Donation Service Baden-Württemberg—Hessia and University Hospital Ulm, Ulm, Germany
| | - Martina Winkelmann
- Institute for Transfusion Medicine, University Hospital Ulm, Ulm, Germany
| | - Simon Ehrenberg
- Institute for Transfusion Medicine, University Hospital Ulm, Ulm, Germany
| | - Tim Eiseler
- Clinic of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Benedikt Friemert
- Clinic for Trauma Surgery and Orthopedics, Army Hospital Ulm, Ulm, Germany
| | - Markus Thomas Rojewski
- Institute for Transfusion Medicine, University Hospital Ulm, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Donation Service Baden-Württemberg—Hessia and University Hospital Ulm, Ulm, Germany
| | - Hubert Schrezenmeier
- Institute for Transfusion Medicine, University Hospital Ulm, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Donation Service Baden-Württemberg—Hessia and University Hospital Ulm, Ulm, Germany
| |
Collapse
|
37
|
Martel R, Shen ML, DeCorwin-Martin P, de Araujo LO, Juncker D. Extracellular Vesicle Antibody Microarray for Multiplexed Inner and Outer Protein Analysis. ACS Sens 2022; 7:3817-3828. [PMID: 36515500 PMCID: PMC9791990 DOI: 10.1021/acssensors.2c01750] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Proteins are found both outside and inside of extracellular vesicles (EVs) and govern the properties and functions of EVs, while also constituting a signature of the cell of origin and of biological function and disease. Outer proteins on EVs can be directly bound by antibodies to either enrich EVs, or probe the expression of a protein on EVs, including in a combinatorial manner. However, co-profiling of inner proteins remains challenging. Here, we present the high-throughput, multiplexed analysis of EV inner and outer proteins (EVPio). We describe the optimization of fixation and heat-induced protein epitope retrieval for EVs, along with oligo-barcoded antibodies and branched DNA signal amplification for sensitive, multiplexed, and high-throughput assays. We captured four subpopulations of EVs from colorectal cancer (CRC) cell lines HT29 and SW403 based on EpCAM, CD9, CD63, and CD81 expression, and quantified the co-expression of eight outer [integrins (ITGs) and tetraspanins] and four inner (heat shock, endosomal, and inner leaflet) proteins. The differences in co-expression patterns were consistent with the literature and known biological function. In conclusion, EVPio analysis can simultaneously detect multiple inner and outer proteins in EVs immobilized on a surface, opening the way to extensive combinatorial protein profiles for both discovery and clinical translation.
Collapse
Affiliation(s)
- Rosalie Martel
- Biomedical
Engineering Department, McGill University, Montreal, Quebec H3A 2B4, Canada,McGill
Genome Centre, McGill University, Montreal, Quebec H3A 0G1, Canada
| | - Molly L. Shen
- Biomedical
Engineering Department, McGill University, Montreal, Quebec H3A 2B4, Canada,McGill
Genome Centre, McGill University, Montreal, Quebec H3A 0G1, Canada
| | - Philippe DeCorwin-Martin
- Biomedical
Engineering Department, McGill University, Montreal, Quebec H3A 2B4, Canada,McGill
Genome Centre, McGill University, Montreal, Quebec H3A 0G1, Canada
| | - Lorenna Oliveira
Fernandes de Araujo
- Biomedical
Engineering Department, McGill University, Montreal, Quebec H3A 2B4, Canada,McGill
Genome Centre, McGill University, Montreal, Quebec H3A 0G1, Canada
| | - David Juncker
- Biomedical
Engineering Department, McGill University, Montreal, Quebec H3A 2B4, Canada,McGill
Genome Centre, McGill University, Montreal, Quebec H3A 0G1, Canada,
| |
Collapse
|
38
|
Lau H, Han DW, Park J, Lehner E, Kals C, Arzt C, Bayer E, Auer D, Schally T, Grasmann E, Fang H, Lee J, Lee HS, Han J, Gimona M, Rohde E, Bae S, Oh SW. GMP-compliant manufacturing of biologically active cell-derived vesicles produced by extrusion technology. JOURNAL OF EXTRACELLULAR BIOLOGY 2022; 1:e70. [PMID: 38938599 PMCID: PMC11080851 DOI: 10.1002/jex2.70] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/08/2022] [Accepted: 11/01/2022] [Indexed: 06/29/2024]
Abstract
Extracellular vesicles (EVs) released by a variety of cell types have been shown to act as a natural delivery system for bioactive molecules such as RNAs and proteins. EV therapy holds great promise as a safe and cell-free therapy for many immunological and degenerative diseases. However, translation to clinical application is limited by several factors, including insufficient large-scale manufacturing technologies and low yield. We have developed a novel drug delivery platform technology, BioDrone™, based on cell-derived vesicles (CDVs) produced from diverse cell sources by using a proprietary extrusion process. This extrusion technology generates nanosized vesicles in far greater numbers than naturally obtained EVs. We demonstrate that the CDVs are surrounded by a lipid bilayer membrane with a correct membrane topology. Physical, biochemical and functional characterisation results demonstrate the potential of CDVs to act as effective therapeutics. Umbilical cord mesenchymal stem cell (UCMSC)-derived CDVs exhibit a biological activity that is similar to UCMSCs or UCMSC-derived EVs. Lastly, we present the establishment of a GMP-compliant process to allow the production of a large number of UCMSC-CDVs in a reproducible manner. GMP-compliant manufacturing of CDVs will facilitate the preclinical and clinical evaluation of these emerging therapeutics in anti-inflammatory or regenerative medicine. This study also represents a crucial step in the development of this novel drug delivery platform based on CDVs.
Collapse
Affiliation(s)
| | - Dong Woo Han
- BioDrone Research InstituteMDimune Inc.SeoulKorea
| | - Jinhee Park
- BioDrone Research InstituteMDimune Inc.SeoulKorea
| | - Edwine Lehner
- GMP Unit, Spinal Cord Injury & Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical UniversitySalzburgAustria
| | - Carina Kals
- GMP Unit, Spinal Cord Injury & Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical UniversitySalzburgAustria
| | - Claudia Arzt
- Transfer Centre for Extracellular Vesicle Theralytic Technologies (EV‐TT)SalzburgAustria
| | - Elisabeth Bayer
- GMP Unit, Spinal Cord Injury & Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical UniversitySalzburgAustria
| | - Daniela Auer
- GMP Unit, Spinal Cord Injury & Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical UniversitySalzburgAustria
| | - Tanja Schally
- GMP Unit, Spinal Cord Injury & Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical UniversitySalzburgAustria
| | - Eva Grasmann
- Transfer Centre for Extracellular Vesicle Theralytic Technologies (EV‐TT)SalzburgAustria
| | - Han Fang
- Transfer Centre for Extracellular Vesicle Theralytic Technologies (EV‐TT)SalzburgAustria
| | - Jae‐Young Lee
- Department of Ophthalmology, Eunpyeong St. Mary's Hospital, College of MedicineThe Catholic University of KoreaSeoulKorea
| | - Hyun Soo Lee
- Department of Ophthalmology, Eunpyeong St. Mary's Hospital, College of MedicineThe Catholic University of KoreaSeoulKorea
| | - Jinah Han
- BioDrone Therapeutics Inc.SeattleUSA
| | - Mario Gimona
- GMP Unit, Spinal Cord Injury & Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical UniversitySalzburgAustria
- Transfer Centre for Extracellular Vesicle Theralytic Technologies (EV‐TT)SalzburgAustria
- Research Program “Nanovesicular Therapies”Paracelsus Medical UniversitySalzburgAustria
| | - Eva Rohde
- GMP Unit, Spinal Cord Injury & Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical UniversitySalzburgAustria
- Department of Transfusion Medicine, University HospitalSalzburger Landeskliniken GesmbH (SALK) and Paracelsus Medical UniversitySalzburgAustria
| | - Shingyu Bae
- BioDrone Research InstituteMDimune Inc.SeoulKorea
| | - Seung Wook Oh
- BioDrone Research InstituteMDimune Inc.SeoulKorea
- BioDrone Therapeutics Inc.SeattleUSA
| |
Collapse
|
39
|
Mecocci S, Trabalza-Marinucci M, Cappelli K. Extracellular Vesicles from Animal Milk: Great Potentialities and Critical Issues. Animals (Basel) 2022; 12:ani12233231. [PMID: 36496752 PMCID: PMC9740508 DOI: 10.3390/ani12233231] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 10/25/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022] Open
Abstract
Other than representing the main source of nutrition for newborn mammals, milk delivers a sophisticated signaling system from mother to child that promotes postnatal health. The bioactive components transferred through the milk intake are important for the development of the newborn immune system and include oligosaccharides, lactoferrin, lysozyme, α-La, and immunoglobulins. In the last 15 years, a pivotal role in this mother-to-child exchange has been attributed to extracellular vesicles (EVs). EVs are micro- and nanosized structures enclosed in a phospholipidic double-layer membrane that are produced by all cell types and released in the extracellular environment, reaching both close and distant cells. EVs mediate the intercellular cross-talk from the producing to the receiving cell through the transfer of molecules contained within them such as proteins, antigens, lipids, metabolites, RNAs, and DNA fragments. The complex cargo can induce a wide range of functional modulations in the recipient cell (i.e., anti-inflammatory, immunomodulating, angiogenetic, and pro-regenerative modulations) depending on the type of producing cells and the stimuli that these cells receive. EVs can be recovered from every biological fluid, including blood, urine, bronchoalveolar lavage fluid, saliva, bile, and milk, which is one of the most promising scalable vesicle sources. This review aimed to present the state-of-the-art of animal-milk-derived EV (mEV) studies due to the exponential growth of this field. A focus on the beneficial potentialities for human health and the issues of studying vesicles from milk, particularly for the analytical methodologies applied, is reported.
Collapse
|
40
|
Driedonks T, Jiang L, Carlson B, Han Z, Liu G, Queen SE, Shirk EN, Gololobova O, Liao Z, Nyberg LH, Lima G, Paniushkina L, Garcia‐Contreras M, Schonvisky K, Castell N, Stover M, Guerrero‐Martin S, Richardson R, Smith B, Mahairaki V, Lai CP, Izzi JM, Hutchinson EK, Pate KAM, Witwer KW. Pharmacokinetics and biodistribution of extracellular vesicles administered intravenously and intranasally to Macaca nemestrina. JOURNAL OF EXTRACELLULAR BIOLOGY 2022; 1:e59. [PMID: 36591537 PMCID: PMC9799283 DOI: 10.1002/jex2.59] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 07/23/2022] [Accepted: 08/13/2022] [Indexed: 01/05/2023]
Abstract
Extracellular vesicles (EVs) have potential in disease treatment since they can be loaded with therapeutic molecules and engineered for retention by specific tissues. However, questions remain on optimal dosing, administration, and pharmacokinetics. Previous studies have addressed biodistribution and pharmacokinetics in rodents, but little evidence is available for larger animals. Here, we investigated the pharmacokinetics and biodistribution of Expi293F-derived EVs labelled with a highly sensitive nanoluciferase reporter (palmGRET) in a non-human primate model (Macaca nemestrina), comparing intravenous (IV) and intranasal (IN) administration over a 125-fold dose range. We report that EVs administered IV had longer circulation times in plasma than previously reported in mice and were detectable in cerebrospinal fluid (CSF) after 30-60 minutes. EV association with PBMCs, especially B-cells, was observed as early as one minute post-administration. EVs were detected in liver and spleen within one hour of IV administration. However, IN delivery was minimal, suggesting that pretreatment approaches may be needed in large animals. Furthermore, EV circulation times strongly decreased after repeated IV administration, possibly due to immune responses and with clear implications for xenogeneic EV-based therapeutics. We hope that our findings from this baseline study in macaques will help to inform future research and therapeutic development of EVs.
Collapse
Affiliation(s)
- Tom Driedonks
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Linglei Jiang
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Bess Carlson
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Zheng Han
- Russell H. Morgan Department of RadiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- F.M. Kirby Research CenterKennedy Krieger InstituteBaltimoreMarylandUSA
| | - Guanshu Liu
- Russell H. Morgan Department of RadiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- F.M. Kirby Research CenterKennedy Krieger InstituteBaltimoreMarylandUSA
| | - Suzanne E. Queen
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Erin N. Shirk
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Olesia Gololobova
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Zhaohao Liao
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Lyle H. Nyberg
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Gabriela Lima
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Liliia Paniushkina
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Marta Garcia‐Contreras
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Kayla Schonvisky
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Natalie Castell
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Mitchel Stover
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Selena Guerrero‐Martin
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Division of Comparative Medicine and Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Riley Richardson
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Barbara Smith
- Department of Cell BiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Vasiliki Mahairaki
- The Richman Family Precision Medicine Center of Excellence in Alzheimer's DiseaseJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Charles P. Lai
- Institute of Atomic and Molecular SciencesAcademia SinicaTaipeiTaiwan
| | - Jessica M. Izzi
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Eric K. Hutchinson
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Kelly A. M. Pate
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Division of Comparative Medicine and Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Kenneth W. Witwer
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- The Richman Family Precision Medicine Center of Excellence in Alzheimer's DiseaseJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
41
|
Burrello J, Monticone S, Burrello A, Bolis S, Cristalli CP, Comai G, Corradetti V, Grange C, Orlando G, Bonafè M, La Manna G, Barile L, Bussolati B. Identification of a serum and urine extracellular vesicle signature predicting renal outcome after kidney transplant. Nephrol Dial Transplant 2022; 38:764-777. [PMID: 36073758 PMCID: PMC9976747 DOI: 10.1093/ndt/gfac259] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND A long-standing effort is dedicated towards the identification of biomarkers allowing the prediction of graft outcome after kidney transplant. Extracellular vesicles (EVs) circulating in body fluids represent an attractive candidate, as their cargo mirrors the originating cell and its pathophysiological status. The aim of the study was to investigate EV surface antigens as potential predictors of renal outcome after kidney transplant. METHODS We characterized 37 surface antigens by flow cytometry, in serum and urine EVs from 58 patients who were evaluated before, and at 10-14 days, 3 months and 1 year after transplant, for a total of 426 analyzed samples. The outcome was defined according to estimated glomerular filtration rate (eGFR) at 1 year. RESULTS Endothelial cells and platelets markers (CD31, CD41b, CD42a and CD62P) in serum EVs were higher at baseline in patients with persistent kidney dysfunction at 1 year, and progressively decreased after kidney transplant. Conversely, mesenchymal progenitor cell marker (CD1c, CD105, CD133, SSEEA-4) in urine EVs progressively increased after transplant in patients displaying renal recovery at follow-up. These markers correlated with eGFR, creatinine and proteinuria, associated with patient outcome at univariate analysis and were able to predict patient outcome at receiver operating characteristics curves analysis. A specific EV molecular signature obtained by supervised learning correctly classified patients according to 1-year renal outcome. CONCLUSIONS An EV-based signature, reflecting the cardiovascular profile of the recipient, and the repairing/regenerative features of the graft, could be introduced as a non-invasive tool for a tailored management of follow-up of patients undergoing kidney transplant.
Collapse
Affiliation(s)
- Jacopo Burrello
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland,Department of Medical Sciences, University of Torino, Italy
| | | | - Alessio Burrello
- Department of Electrical, Electronic and Information Engineering (DEI), University of Bologna, Italy
| | - Sara Bolis
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Carlotta Pia Cristalli
- IRCCS – Azienda Ospedaliero-Universitaria di Bologna, Nephrology, Dialysis and Renal Transplant Unit, University of Bologna, Italy
| | - Giorgia Comai
- IRCCS – Azienda Ospedaliero-Universitaria di Bologna, Nephrology, Dialysis and Renal Transplant Unit, University of Bologna, Italy
| | - Valeria Corradetti
- IRCCS – Azienda Ospedaliero-Universitaria di Bologna, Nephrology, Dialysis and Renal Transplant Unit, University of Bologna, Italy
| | | | - Giuseppe Orlando
- Department of Surgery, Section of Transplantation, Wake Forest University School of Medicine, Winston Salem, NC, USA
| | - Massimiliano Bonafè
- IRCCS – Azienda Ospedaliero-Universitaria di Bologna, Nephrology, Dialysis and Renal Transplant Unit, University of Bologna, Italy,Department of Experimental, Diagnostic and Specialty Medicine, AlmaMater Studiorum, Universitá di Bologna
| | - Gaetano La Manna
- IRCCS – Azienda Ospedaliero-Universitaria di Bologna, Nephrology, Dialysis and Renal Transplant Unit, University of Bologna, Italy
| | | | | |
Collapse
|
42
|
Franzin R, Stasi A, Sallustio F, Bruno S, Merlotti G, Quaglia M, Grandaliano G, Pontrelli P, Thurman JM, Camussi G, Stallone G, Cantaluppi V, Gesualdo L, Castellano G. Extracellular vesicles derived from patients with antibody-mediated rejection induce tubular senescence and endothelial to mesenchymal transition in renal cells. Am J Transplant 2022; 22:2139-2157. [PMID: 35583104 PMCID: PMC9546277 DOI: 10.1111/ajt.17097] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 01/25/2023]
Abstract
Extracellular vesicles (EV) are emerging mediators in several diseases. However, their role in the pathophysiology of antibody-mediated allograft rejection (AMR) has been poorly investigated. Here, we investigated the role of EV isolated from AMR patients in inducing tubular senescence and endothelial to mesenchymal transition (EndMT) and analyzed their miRNA expression profile. By multiplex bead flow cytometry, we characterized the immunophenotype of plasma AMR-derived EV and found a prevalent platelet and endothelial cell origin. In vitro, AMR-derived EV induced tubular senescence by upregulating SA-β Gal and CDKN1A mRNA. Furthermore, AMR-derived EV induced EndMT. The occurrence of tubular senescence and EndMT was confirmed by analysis of renal biopsies from the same AMR patients. Moreover, AMR-derived EV induced C3 gene upregulation and CFH downregulation in tubular epithelial cells, with C4d deposition on endothelial cells. Interestingly, RNase-mediated digestion of EV cargo completely abrogated tubular senescence and EndMT. By microarray analysis, miR-604, miR-515-3p, miR-let-7d-5p, and miR-590-3p were significantly upregulated in EV from AMR group compared with transplant controls, whereas miR-24-3p and miR-29a-3p were downregulated. Therefore, EV-associated miRNA could act as active player in AMR pathogenesis, unraveling potential mechanisms of accelerated graft senescence, complement activation and early fibrosis that might lead to new therapeutic intervention.
Collapse
Affiliation(s)
- Rossana Franzin
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ TransplantationUniversity of Bari Aldo MoroBariItaly
| | - Alessandra Stasi
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ TransplantationUniversity of Bari Aldo MoroBariItaly
| | - Fabio Sallustio
- Interdisciplinary Department of Medicine (DIM)University of Bari "Aldo Moro"BariItaly
| | - Stefania Bruno
- Department of Medical Sciences and Molecular Biotechnology CenterUniversity of TorinoTorinoItaly
| | - Guido Merlotti
- Nephrology and Kidney Transplantation Unit, Department of Translational Medicine and Center for Autoimmune and Allergic Diseases (CAAD)University of Piemonte Orientale (UPO)NovaraItaly
| | - Marco Quaglia
- Nephrology and Kidney Transplantation Unit, Department of Translational Medicine and Center for Autoimmune and Allergic Diseases (CAAD)University of Piemonte Orientale (UPO)NovaraItaly
| | - Giuseppe Grandaliano
- Department Translational Medicine and SurgeryUniversità Cattolica Sacro CuoreRomeItaly
| | - Paola Pontrelli
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ TransplantationUniversity of Bari Aldo MoroBariItaly
| | - Joshua M. Thurman
- Department of MedicineUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Giovanni Camussi
- Department of Medical Sciences and Molecular Biotechnology CenterUniversity of TorinoTorinoItaly
| | - Giovanni Stallone
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical SciencesUniversity of FoggiaFoggiaItaly
| | - Vincenzo Cantaluppi
- Nephrology and Kidney Transplantation Unit, Department of Translational Medicine and Center for Autoimmune and Allergic Diseases (CAAD)University of Piemonte Orientale (UPO)NovaraItaly
| | - Loreto Gesualdo
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ TransplantationUniversity of Bari Aldo MoroBariItaly
| | - Giuseppe Castellano
- Unit of NephrologyDialysis and Renal Transplantation ‐ Fondazione IRCCS Ca’Granda Ospedale Maggiore Policlinico di MilanoMilanItaly
| |
Collapse
|
43
|
Wu A, Wolley MJ, Fenton RA, Stowasser M. Using human urinary extracellular vesicles to study physiological and pathophysiological states and regulation of the sodium chloride cotransporter. Front Endocrinol (Lausanne) 2022; 13:981317. [PMID: 36105401 PMCID: PMC9465297 DOI: 10.3389/fendo.2022.981317] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 08/09/2022] [Indexed: 11/29/2022] Open
Abstract
The thiazide-sensitive sodium chloride cotransporter (NCC), expressed in the renal distal convoluted tubule, plays a major role in Na+, Cl- and K+ homeostasis and blood pressure as exemplified by the symptoms of patients with non-functional NCC and Gitelman syndrome. NCC activity is modulated by a variety of hormones, but is also influenced by the extracellular K+ concentration. The putative "renal-K+ switch" mechanism is a relatively cohesive model that links dietary K+ intake to NCC activity, and may offer new targets for blood pressure control. However, a remaining hurdle for full acceptance of this model is the lack of human data to confirm molecular findings from animal models. Extracellular vesicles (EVs) have attracted attention from the scientific community due to their potential roles in intercellular communication, disease pathogenesis, drug delivery and as possible reservoirs of biomarkers. Urinary EVs (uEVs) are an excellent sample source for the study of physiology and pathology of renal, urothelial and prostate tissues, but the diverse origins of uEVs and their dynamic molecular composition present both methodological and data interpretation challenges. This review provides a brief overview of the state-of-the-art, challenges and knowledge gaps in current uEV-based analyses, with a focus on the application of uEVs to study the "renal-K+ switch" and NCC regulation. We also provide recommendations regarding biospecimen handling, processing and reporting requirements to improve experimental reproducibility and interoperability towards the realisation of the potential of uEV-derived biomarkers in hypertension and clinical practice.
Collapse
Affiliation(s)
- Aihua Wu
- Endocrine Hypertension Research Centre, University of Queensland Diamantina Institute, Greenslopes and Princess Alexandra Hospitals, Brisbane, QLD, Australia
| | - Martin J. Wolley
- Endocrine Hypertension Research Centre, University of Queensland Diamantina Institute, Greenslopes and Princess Alexandra Hospitals, Brisbane, QLD, Australia
- Department of Nephrology, Royal Brisbane and Women’s Hospital, Brisbane, QLD, Australia
| | | | - Michael Stowasser
- Endocrine Hypertension Research Centre, University of Queensland Diamantina Institute, Greenslopes and Princess Alexandra Hospitals, Brisbane, QLD, Australia
| |
Collapse
|
44
|
Fortunato D, Giannoukakos S, Giménez-Capitán A, Hackenberg M, Molina-Vila MA, Zarovni N. Selective isolation of extracellular vesicles from minimally processed human plasma as a translational strategy for liquid biopsies. Biomark Res 2022; 10:57. [PMID: 35933395 PMCID: PMC9357340 DOI: 10.1186/s40364-022-00404-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 07/27/2022] [Indexed: 11/25/2022] Open
Abstract
Background Intercellular communication is mediated by extracellular vesicles (EVs), as they enclose selectively packaged biomolecules that can be horizontally transferred from donor to recipient cells. Because all cells constantly generate and recycle EVs, they provide accurate timed snapshots of individual pathophysiological status. Since blood plasma circulates through the whole body, it is often the biofluid of choice for biomarker detection in EVs. Blood collection is easy and minimally invasive, yet reproducible procedures to obtain pure EV samples from circulating biofluids are still lacking. Here, we addressed central aspects of EV immunoaffinity isolation from simple and complex matrices, such as plasma. Methods Cell-generated EV spike-in models were isolated and purified by size-exclusion chromatography, stained with cellular dyes and characterized by nano flow cytometry. Fluorescently-labelled spike-in EVs emerged as reliable, high-throughput and easily measurable readouts, which were employed to optimize our EV immunoprecipitation strategy and evaluate its performance. Plasma-derived EVs were captured and detected using this straightforward protocol, sequentially combining isolation and staining of specific surface markers, such as CD9 or CD41. Multiplexed digital transcript detection data was generated using the Nanostring nCounter platform and evaluated through a dedicated bioinformatics pipeline. Results Beads with covalently-conjugated antibodies on their surface outperformed streptavidin-conjugated beads, coated with biotinylated antibodies, in EV immunoprecipitation. Fluorescent EV spike recovery evidenced that target EV subpopulations can be efficiently retrieved from plasma, and that their enrichment is dependent not only on complex matrix composition, but also on the EV surface phenotype. Finally, mRNA profiling experiments proved that distinct EV subpopulations can be captured by directly targeting different surface markers. Furthermore, EVs isolated with anti-CD61 beads enclosed mRNA expression patterns that might be associated to early-stage lung cancer, in contrast with EVs captured through CD9, CD63 or CD81. The differential clinical value carried within each distinct EV subset highlights the advantages of selective isolation. Conclusions This EV isolation protocol facilitated the extraction of clinically useful information from plasma. Compatible with common downstream analytics, it is a readily implementable research tool, tailored to provide a truly translational solution in routine clinical workflows, fostering the inclusion of EVs in novel liquid biopsy settings. Supplementary Information The online version contains supplementary material available at 10.1186/s40364-022-00404-1.
Collapse
|
45
|
Gul B, Syed F, Khan S, Iqbal A, Ahmad I. Characterization of extracellular vesicles by flow cytometry: Challenges and promises. Micron 2022; 161:103341. [DOI: 10.1016/j.micron.2022.103341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 08/03/2022] [Accepted: 08/03/2022] [Indexed: 10/16/2022]
|
46
|
In Vitro Interactions of Amphiphilic Phosphorous Dendrons with Liposomes and Exosomes—Implications for Blood Viscosity Changes. Pharmaceutics 2022; 14:pharmaceutics14081596. [PMID: 36015222 PMCID: PMC9414926 DOI: 10.3390/pharmaceutics14081596] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/25/2022] [Accepted: 07/28/2022] [Indexed: 12/20/2022] Open
Abstract
Drug delivery by dendron-based nanoparticles is widely studied due to their ability to encapsulate or bind different ligands. For medical purposes, it is necessary (even if not sufficient) for these nanostructures to be compatible with blood. We studied the interaction of amphiphilic dendrons with blood samples from healthy volunteers using standard laboratory methods and rheological measurements. We did not observe clinically relevant abnormalities, but we found a concentration-dependent increase in whole blood viscosity, higher in males, presumably due to the formation of aggregates. To characterize the nature of the interactions among blood components and dendrons, we performed experiments on the liposomes and exosomes as models of biological membranes. Based on results obtained using diverse biophysical methods, we conclude that the interactions were of electrostatic nature. Overall, we have confirmed a concentration-dependent effect of dendrons on membrane systems, while the effect of generation was ambiguous. At higher dendron concentrations, the structure of membranes became disturbed, and membranes were prone to forming bigger aggregates, as visualized by SEM. This might have implications for blood flow disturbances when used in vivo. We propose to introduce blood viscosity measurements in early stages of investigation as they can help to optimize drug-like properties of potential drug carriers.
Collapse
|
47
|
Burrello J, Burrello A, Vacchi E, Bianco G, Caporali E, Amongero M, Airale L, Bolis S, Vassalli G, Cereda CW, Mulatero P, Bussolati B, Camici GG, Melli G, Monticone S, Barile L. Supervised and unsupervised learning to define the cardiovascular risk of patients according to an extracellular vesicle molecular signature. Transl Res 2022; 244:114-125. [PMID: 35202881 DOI: 10.1016/j.trsl.2022.02.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 01/28/2022] [Accepted: 02/16/2022] [Indexed: 12/21/2022]
Abstract
Cardiovascular (CV) disease represents the most common cause of death in developed countries. Risk assessment is highly relevant to intervene at individual level and implement prevention strategies. Circulating extracellular vesicles (EVs) are involved in the development and progression of CV diseases and are considered promising biomarkers. We aimed at identifying an EV signature to improve the stratification of patients according to CV risk and likelihood to develop fatal CV events. EVs were characterized by nanoparticle tracking analysis and flow cytometry for a standardized panel of 37 surface antigens in a cross-sectional multicenter cohort (n = 486). CV profile was defined by presence of different indicators (age, sex, body mass index, hypertension, hyperlipidemia, diabetes, coronary artery disease, cardiac heart failure, chronic kidney disease, smoking habit, organ damage) and according to the 10-year risk of fatal CV events estimated using SCORE charts of European Society of Cardiology. By combining expression levels of EV antigens using unsupervised learning, patients were classified into 3 clusters: Cluster-I (n = 288), Cluster-II (n = 83), Cluster-III (n = 30). A separate analysis was conducted on patients displaying acute CV events (n = 82). Prevalence of hypertension, diabetes, chronic heart failure, and organ damage (defined as left ventricular hypertrophy and/or microalbuminuria) increased progressively from Cluster-I to Cluster-III. Several EV antigens, including markers for platelets (CD41b-CD42a-CD62P), leukocytes (CD1c-CD2-CD3-CD4-CD8-CD14-CD19-CD20-CD25-CD40-CD45-CD69-CD86), and endothelium (CD31-CD105) were independently associated with CV risk indicators and correlated to age, blood pressure, glucometabolic profile, renal function, and SCORE risk. EV profiling, obtained from minimally invasive blood sampling, allows accurate patient stratification according to CV risk profile.
Collapse
Affiliation(s)
- Jacopo Burrello
- Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale, Bellinzona, Switzerland; Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, Italy
| | - Alessio Burrello
- Department of Electrical, Electronic and Information Engineering (DEI), University of Bologna, Italy
| | - Elena Vacchi
- Laboratory for Biomedical Neurosciences, Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland; Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Giovanni Bianco
- Neurology Clinic, Stroke Center, Neurocenter of Southern Switzerland, Lugano, Switzerland
| | - Elena Caporali
- Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Martina Amongero
- Department of Mathematical Sciences G. L. Lagrange, Polytechnic University of Torino, Italy
| | - Lorenzo Airale
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, Italy
| | - Sara Bolis
- Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Giuseppe Vassalli
- Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale, Bellinzona, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland; Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Carlo W Cereda
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland; Neurology Clinic, Stroke Center, Neurocenter of Southern Switzerland, Lugano, Switzerland
| | - Paolo Mulatero
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, Italy
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Italy
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zürich, Schlieren, Switzerland
| | - Giorgia Melli
- Laboratory for Biomedical Neurosciences, Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland; Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Silvia Monticone
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, Italy
| | - Lucio Barile
- Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale, Bellinzona, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland; Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland; Institute of Life Science, Scuola Superiore Sant'Anna, Pisa, Italy.
| |
Collapse
|
48
|
Auber M, Svenningsen P. An estimate of extracellular vesicle secretion rates of human blood cells. JOURNAL OF EXTRACELLULAR BIOLOGY 2022; 1:e46. [PMID: 38938292 PMCID: PMC11080926 DOI: 10.1002/jex2.46] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 04/26/2022] [Accepted: 05/11/2022] [Indexed: 06/29/2024]
Abstract
Extracellular vesicles (EVs) have been implicated in the intercellular transfer of RNA and proteins through cellular secretion into the extracellular space. In blood plasma, circulating EVs are mainly derived from blood cells; however, factors that control plasma EV abundance are largely unknown. Here, we estimate the EV secretion rates for blood cell types using reported values for cell-specific plasma EV abundances and their parental cell's ubiquity in healthy humans. While we found that plasma contains on average ∼2 plasma EVs/cell, the cell-specific EV-to-cell ratio spanned four orders of magnitude from 0.13 ± 0.1 erythrocyte-derived EVs/erythrocyte to (1.9 ± 1.3) × 103 monocyte-derived EVs/monocyte. The steady-state plasma EV level was maintained by an estimated plasma EV secretion rate of (1.5 ± 0.4) × 1012 EVs/min. The cell-specific secretion rate estimates were highest for monocytes (45 ± 21 EVs/cell/min) and lowest for erythrocytes ((3.2 ± 3.0) × 10-3 EVs/cell/min). The estimated basal cell-specific EV secretion rates were not significantly correlated to the cell's lifespan or size; however, we observed a highly significant correlation to cellular mitochondrial enzyme activities. Together, our analysis indicates that cell-specific mitochondrial metabolism, for example, via reactive oxygen species, affects plasma EV abundance through increased secretion rates, and the results provide a resource for understanding EV function in human health and disease.
Collapse
Affiliation(s)
- Martin Auber
- Department of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
| | - Per Svenningsen
- Department of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
| |
Collapse
|
49
|
Görgens A, Corso G, Hagey DW, Jawad Wiklander R, Gustafsson MO, Felldin U, Lee Y, Bostancioglu RB, Sork H, Liang X, Zheng W, Mohammad DK, van de Wakker SI, Vader P, Zickler AM, Mamand DR, Ma L, Holme MN, Stevens MM, Wiklander OPB, EL Andaloussi S. Identification of storage conditions stabilizing extracellular vesicles preparations. J Extracell Vesicles 2022; 11:e12238. [PMID: 35716060 PMCID: PMC9206228 DOI: 10.1002/jev2.12238] [Citation(s) in RCA: 118] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 04/23/2022] [Accepted: 05/16/2022] [Indexed: 12/31/2022] Open
Abstract
Extracellular vesicles (EVs) play a key role in many physiological and pathophysiological processes and hold great potential for therapeutic and diagnostic use. Despite significant advances within the last decade, the key issue of EV storage stability remains unresolved and under investigated. Here, we aimed to identify storage conditions stabilizing EVs and comprehensively compared the impact of various storage buffer formulations at different temperatures on EVs derived from different cellular sources for up to 2 years. EV features including concentration, diameter, surface protein profile and nucleic acid contents were assessed by complementary methods, and engineered EVs containing fluorophores or functionalized surface proteins were utilized to compare cellular uptake and ligand binding. We show that storing EVs in PBS over time leads to drastically reduced recovery particularly for pure EV samples at all temperatures tested, starting already within days. We further report that using PBS as diluent was found to result in severely reduced EV recovery rates already within minutes. Several of the tested new buffer conditions largely prevented the observed effects, the lead candidate being PBS supplemented with human albumin and trehalose (PBS-HAT). We report that PBS-HAT buffer facilitates clearly improved short-term and long-term EV preservation for samples stored at -80°C, stability throughout several freeze-thaw cycles, and drastically improved EV recovery when using a diluent for EV samples for downstream applications.
Collapse
Affiliation(s)
- André Görgens
- Department of Laboratory Medicine, Clinical Research CenterKarolinska InstitutetStockholmSweden
- Institute for Transfusion MedicineUniversity Hospital Essen, University of Duisburg‐EssenEssenGermany
- Evox Therapeutics LimitedOxfordUK
| | - Giulia Corso
- Department of Laboratory Medicine, Clinical Research CenterKarolinska InstitutetStockholmSweden
| | - Daniel W. Hagey
- Department of Laboratory Medicine, Clinical Research CenterKarolinska InstitutetStockholmSweden
| | - Rim Jawad Wiklander
- Department of Laboratory Medicine, Clinical Research CenterKarolinska InstitutetStockholmSweden
| | - Manuela O. Gustafsson
- Department of Laboratory Medicine, Clinical Research CenterKarolinska InstitutetStockholmSweden
| | - Ulrika Felldin
- Department of Laboratory Medicine, Clinical Research CenterKarolinska InstitutetStockholmSweden
| | - Yi Lee
- Department of Laboratory Medicine, Clinical Research CenterKarolinska InstitutetStockholmSweden
| | - R. Beklem Bostancioglu
- Department of Laboratory Medicine, Clinical Research CenterKarolinska InstitutetStockholmSweden
| | - Helena Sork
- Department of Laboratory Medicine, Clinical Research CenterKarolinska InstitutetStockholmSweden
- Institute of TechnologyUniversity of TartuTartuEstonia
| | - Xiuming Liang
- Department of Laboratory Medicine, Clinical Research CenterKarolinska InstitutetStockholmSweden
| | - Wenyi Zheng
- Department of Laboratory Medicine, Clinical Research CenterKarolinska InstitutetStockholmSweden
| | - Dara K. Mohammad
- Department of Laboratory Medicine, Clinical Research CenterKarolinska InstitutetStockholmSweden
- College of Agricultural Engineering SciencesSalahaddin University‐ErbilErbilKurdistan RegionIraq
| | - Simonides I. van de Wakker
- Department of CardiologyExperimental Cardiology LaboratoryUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Pieter Vader
- Department of CardiologyExperimental Cardiology LaboratoryUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
- CDL ResearchUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Antje M. Zickler
- Department of Laboratory Medicine, Clinical Research CenterKarolinska InstitutetStockholmSweden
| | - Doste R. Mamand
- Department of Laboratory Medicine, Clinical Research CenterKarolinska InstitutetStockholmSweden
| | - Li Ma
- Department of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSweden
| | - Margaret N. Holme
- Department of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSweden
| | - Molly M. Stevens
- Department of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSweden
- Department of MaterialsDepartment of Bioengineeringand Institute of Biomedical EngineeringImperial College LondonLondonUK
| | - Oscar P. B. Wiklander
- Department of Laboratory Medicine, Clinical Research CenterKarolinska InstitutetStockholmSweden
- Evox Therapeutics LimitedOxfordUK
| | - Samir EL Andaloussi
- Department of Laboratory Medicine, Clinical Research CenterKarolinska InstitutetStockholmSweden
- Evox Therapeutics LimitedOxfordUK
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| |
Collapse
|
50
|
Risk stratification of patients with SARS-CoV-2 by tissue factor expression in circulating extracellular vesicles. Vascul Pharmacol 2022; 145:106999. [PMID: 35597450 PMCID: PMC9116046 DOI: 10.1016/j.vph.2022.106999] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 04/15/2022] [Accepted: 05/13/2022] [Indexed: 01/08/2023]
Abstract
Inflammatory response following SARS-CoV-2 infection results in substantial increase of amounts of intravascular pro-coagulant extracellular vesicles (EVs) expressing tissue factor (CD142) on their surface. CD142-EV turned out to be useful as diagnostic biomarker in COVID-19 patients. Here we aimed at studying the prognostic capacity of CD142-EV in SARS-CoV-2 infection. Expression of CD142-EV was evaluated in 261 subjects admitted to hospital for pneumonia and with a positive molecular test for SARS-CoV-2. The study population consisted of a discovery cohort of selected patients (n = 60) and an independent validation cohort including unselected consecutive enrolled patients (n = 201). CD142-EV levels were correlated with post-hospitalization course of the disease and compared to the clinically available 4C Mortality Score as referral. CD142-EV showed a reliable performance to predict patient prognosis in the discovery cohort (AUC = 0.906) with an accuracy of 81.7%, that was confirmed in the validation cohort (AUC = 0.736). Kaplan-Meier curves highlighted a high discrimination power in unselected subjects with CD142-EV being able to stratify the majority of patients according to their prognosis. We obtained a comparable accuracy, being not inferior in terms of prediction of patients' prognosis and risk of mortality, with 4C Mortality Score. The expression of surface vesicular CD142 and its reliability as prognostic marker was technically validated using different immunocapture strategies and assays. The detection of CD142 on EV surface gains considerable interest as risk stratification tool to support clinical decision making in COVID-19.
Collapse
|