1
|
Woud WW, Pugsley HR, Bettin BA, Varga Z, van der Pol E. Size and fluorescence calibrated imaging flow cytometry: From arbitrary to standard units. Cytometry A 2024; 105:752-762. [PMID: 39238272 DOI: 10.1002/cyto.a.24895] [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: 02/26/2024] [Revised: 07/30/2024] [Accepted: 08/15/2024] [Indexed: 09/07/2024]
Abstract
Imaging flow cytometry (IFCM) is a technique that can detect, size, and phenotype extracellular vesicles (EVs) at high throughput (thousands/minute) in complex biofluids without prior EV isolation. However, the generated signals are expressed in arbitrary units, which hinders data interpretation and comparison of measurement results between instruments and institutes. While fluorescence calibration can be readily achieved, calibration of side scatter (SSC) signals presents an ongoing challenge for IFCM. Here, we present an approach to relate the SSC signals to particle size for IFCM, and perform a comparability study between three different IFCMs using a plasma EV test sample (PEVTES). SSC signals for different sizes of polystyrene (PS) and hollow organosilica beads (HOBs) were acquired with a 405 nm 120 mW laser without a notch filter before detection. Mie theory was applied to relate scatter signals to particle size. Fluorescence calibration was accomplished with 2 μm phycoerythrin (PE) and allophycocyanin (APC) MESF beads. Size and fluorescence calibration was performed for three IFCMs in two laboratories. CD235a-PE and CD61-APC stained PEVTES were used as EV-containing samples. EV concentrations were compared between instruments within a size range of 100-1000 nm and a fluorescence intensity range of 3-10,000 MESF. 81 nm PS beads could be readily discerned from background based on their SSC signals. Fitting of the obtained PS bead SSC signals with Mie theory resulted in a coefficient of determination >0.99 between theory and data for all three IFCMs. 216 nm HOBs were detected with all instruments, and confirmed the sensitivity to detect EVs by SSC. The lower limit of detection regarding EV-size for this study was determined to be ~100 nm for all instruments. Size and fluorescence calibration of IFCM data increased cross-instrument data comparability with the coefficient of variation decreasing from 33% to 21%. Here we demonstrate - for the first time - scatter calibration of an IFCM using the 405 nm laser. The quality of the scatter-to-diameter relation and scatter sensitivity of the IFCMs are similar to the most sensitive commercially available flow cytometers. This development will support the reliability of EV research with IFCM by providing robust standardization and reproducibility, which are pre-requisites for understanding the biological significance of EVs.
Collapse
Affiliation(s)
- Wouter W Woud
- Erasmus MC Transplant Institute, Department of Internal Medicine, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Haley R Pugsley
- Application Cytometry, Cytek Biosciences, Inc, Seattle, Washington, USA
| | - Britta A Bettin
- Department of Biomedical Engineering and Physics, Amsterdam University Medical Center location University of Amsterdam, Amsterdam, The Netherlands
- Department of Clinical Chemistry, Amsterdam University Medical Center location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Vesicle Center, Amsterdam University Medical Center location University of Amsterdam, Amsterdam, The Netherlands
| | - Zoltán Varga
- Biological Nanochemistry Research Group, Institute of Materials and Environmental Chemistry, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Department of Physical Chemistry and Materials Science, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Budapest, Hungary
| | - Edwin van der Pol
- Department of Biomedical Engineering and Physics, Amsterdam University Medical Center location University of Amsterdam, Amsterdam, The Netherlands
- Department of Clinical Chemistry, Amsterdam University Medical Center location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Vesicle Center, Amsterdam University Medical Center location University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
2
|
Rival C, Mandal M, Cramton K, Qiao H, Arish M, Sun J, McCann JV, Dudley AC, Solga MD, Erdbrügger U, Erickson LD. B cells secrete functional antigen-specific IgG antibodies on extracellular vesicles. Sci Rep 2024; 14:16970. [PMID: 39043800 PMCID: PMC11266516 DOI: 10.1038/s41598-024-67912-y] [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: 01/24/2024] [Accepted: 07/17/2024] [Indexed: 07/25/2024] Open
Abstract
B cells and the antibodies they produce are critical in host defense against pathogens and contribute to various immune-mediated diseases. B cells responding to activating signals in vitro release extracellular vesicles (EV) that carry surface antibodies, yet B cell production of EVs that express antibodies and their function in vivo is incompletely understood. Using transgenic mice expressing the Cre recombinase in B cells switching to IgG1 to induce expression of fusion proteins between emerald green fluorescent protein (emGFP) and the EV tetraspanin CD63 as a model, we identify emGFP expression in B cells responding to foreign antigen in vivo and characterize the emGFP+ EVs they release. Our data suggests that emGFP+ germinal center B cells undergoing immunoglobulin class switching to express IgG and their progeny memory B cells and plasma cells, also emGFP+, are sources of circulating antigen-specific IgG+ EVs. Furthermore, using a mouse model of influenza virus infection, we find that IgG+ EVs specific for the influenza hemagglutinin antigen protect against virus infection. In addition, crossing the B cell Cre driver EV reporter mice onto the Nba2 lupus-prone strain revealed increased circulating emGFP+ EVs that expressed surface IgG against nuclear antigens linked to autoimmunity. These data identify EVs loaded with antibodies as a novel route for antibody secretion in B cells that contribute to adaptive immune responses, with important implications for different functions of IgG+ EVs in infection and autoimmunity.
Collapse
Affiliation(s)
- Claudia Rival
- Beirne Carter Center for Immunology Research, University of Virginia, PO Box 801386, Charlottesville, VA, 22908, USA
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Mahua Mandal
- Beirne Carter Center for Immunology Research, University of Virginia, PO Box 801386, Charlottesville, VA, 22908, USA
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Kayla Cramton
- Beirne Carter Center for Immunology Research, University of Virginia, PO Box 801386, Charlottesville, VA, 22908, USA
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Hui Qiao
- Beirne Carter Center for Immunology Research, University of Virginia, PO Box 801386, Charlottesville, VA, 22908, USA
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Mohd Arish
- Beirne Carter Center for Immunology Research, University of Virginia, PO Box 801386, Charlottesville, VA, 22908, USA
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Jie Sun
- Beirne Carter Center for Immunology Research, University of Virginia, PO Box 801386, Charlottesville, VA, 22908, USA
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - James V McCann
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Andrew C Dudley
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, 22908, USA
- Emily Couric Cancer Center, University of Virginia, Charlottesville, VA, 22908, USA
| | - Michael D Solga
- Flow Cytometry Core, University of Virginia, Charlottesville, VA, 22908, USA
| | - Uta Erdbrügger
- Division of Nephrology, Department of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Loren D Erickson
- Beirne Carter Center for Immunology Research, University of Virginia, PO Box 801386, Charlottesville, VA, 22908, USA.
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, 22908, USA.
| |
Collapse
|
3
|
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
|
4
|
Shiju TM, Yuan A. Extracellular vesicle biomarkers in ocular fluids associated with ophthalmic diseases. Exp Eye Res 2024; 241:109831. [PMID: 38401855 DOI: 10.1016/j.exer.2024.109831] [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: 12/08/2023] [Revised: 02/06/2024] [Accepted: 02/13/2024] [Indexed: 02/26/2024]
Abstract
Extracellular vesicles (EVs) are released as highly stable lipid bilayer particles carrying proteins, lipids, glycans and miRNAs. The contents of EVs vary based on the cellular origin, biogenesis route and the functional state of the cell suggesting certain diseased conditions. A growing body of evidence show that EVs carry important molecules implicated in the development and progression of ophthalmic diseases. EVs associated with ophthalmic diseases are mainly carried by one of the three ocular biofluids which include tears, aqueous humor and vitreous humor. This review summarizes the list of EV derived biomarkers identified thus far in ocular fluids for ophthalmic disease diagnosis. Further, the methods used for sample collection, sample volume and the sample numbers used in these studies have been highlighted. Emphasis has been given to describe the EV isolation and the characterization methods used, EV size profiled and the EV concentrations analyzed by these studies, thus providing a roadmap for future EV biomarker studies in ocular fluids.
Collapse
Affiliation(s)
| | - Alex Yuan
- Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
5
|
Pozzi P, Candeo A, Paiè P, Bragheri F, Bassi A. Artificial intelligence in imaging flow cytometry. FRONTIERS IN BIOINFORMATICS 2023; 3:1229052. [PMID: 37877042 PMCID: PMC10593470 DOI: 10.3389/fbinf.2023.1229052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 09/11/2023] [Indexed: 10/26/2023] Open
Affiliation(s)
- Paolo Pozzi
- Department of Physics, Politecnico di Milano, Milano, Italy
| | - Alessia Candeo
- Department of Physics, Politecnico di Milano, Milano, Italy
| | - Petra Paiè
- Department of Physics, Politecnico di Milano, Milano, Italy
| | - Francesca Bragheri
- Institute for Photonics and Nanotechnologies, Consiglio Nazionale delle Ricerche, Milano, Italy
| | - Andrea Bassi
- Department of Physics, Politecnico di Milano, Milano, Italy
| |
Collapse
|
6
|
Brambilla M, Frigerio R, Becchetti A, Gori A, Cretich M, Conti M, Mazza A, Pengo M, Camera M. Head-to-Head Comparison of Tissue Factor-Dependent Procoagulant Potential of Small and Large Extracellular Vesicles in Healthy Subjects and in Patients with SARS-CoV-2 Infection. BIOLOGY 2023; 12:1233. [PMID: 37759632 PMCID: PMC10525820 DOI: 10.3390/biology12091233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 08/28/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023]
Abstract
The relative contribution of small (sEVs) and large extracellular vesicles (lEVs) to the total plasma procoagulant potential is not yet well defined. Thus, we compared total and TFpos-sEVs and -lEVs isolated from healthy subjects and COVID-19 patients during the acute phase of the infection and after symptom remission in terms of (1) vesicle enumeration using nanoparticle tracking assay, imaging flow cytometry, and TF immunofluorescence localization in a single-vesicle analysis using microarrays; (2) cellular origin; and (3) TF-dependent Xa generation capacity, as well as assessing the contribution of the TF inhibitor, TFPI. In healthy subjects, the plasma concentration of CD9/CD63/CD81pos sEVs was 30 times greater than that of calceinpos lEVs, and both were mainly released by platelets. Compared to lEVs, the levels of TFpos-sEVs were 2-fold higher. The TF-dependent Xa generation capacity of lEVs was three times greater than that of sEVs, with the latter being hindered by TFPI. Compared to HSs, the amounts of total and TFpos-sEVs and -lEVs were significantly greater in acute COVID-19 patients, which reverted to the physiological values at the 6-month follow-up. Interestingly, the FXa generation of lEVs only significantly increased during acute infection, with that of sEV being similar to that of HSs. Thus, in both healthy subjects and COVID-19 patients, the TF-dependent procoagulant potential is mostly sustained by large vesicles.
Collapse
Affiliation(s)
- Marta Brambilla
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.B.)
| | - Roberto Frigerio
- National Research Council of Italy (SCITEC-CNR), 20133 Milan, Italy
| | | | - Alessandro Gori
- National Research Council of Italy (SCITEC-CNR), 20133 Milan, Italy
| | - Marina Cretich
- National Research Council of Italy (SCITEC-CNR), 20133 Milan, Italy
| | - Maria Conti
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.B.)
| | - Antonella Mazza
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.B.)
| | - Martino Pengo
- Istituto Auxologico Italiano IRCCS, 20149 Milan, Italy
| | - Marina Camera
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.B.)
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| |
Collapse
|
7
|
Haddad M, Frickenstein A, Wilhelm S. High-Throughput Single-Cell Analysis of Nanoparticle-Cell Interactions. Trends Analyt Chem 2023; 166:117172. [PMID: 37520860 PMCID: PMC10373476 DOI: 10.1016/j.trac.2023.117172] [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] [Indexed: 08/01/2023]
Abstract
Understanding nanoparticle-cell interactions at single-nanoparticle and single-cell resolutions is crucial to improving the design of next-generation nanoparticles for safer, more effective, and more efficient applications in nanomedicine. This review focuses on recent advances in the continuous high-throughput analysis of nanoparticle-cell interactions at the single-cell level. We highlight and discuss the current trends in continual flow high-throughput methods for analyzing single cells, such as advanced flow cytometry techniques and inductively coupled plasma mass spectrometry methods, as well as their intersection in the form of mass cytometry. This review further discusses the challenges and opportunities with current single-cell analysis approaches and provides proposed directions for innovation in the high-throughput analysis of nanoparticle-cell interactions.
Collapse
Affiliation(s)
- Majood Haddad
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Alex Frickenstein
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Stefan Wilhelm
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, USA
- Institute for Biomedical Engineering, Science, and Technology (IBEST), University of Oklahoma, Norman, Oklahoma, 73019, USA
| |
Collapse
|
8
|
Franco C, Ghirardello A, Bertazza L, Gasparotto M, Zanatta E, Iaccarino L, Valadi H, Doria A, Gatto M. Size-Exclusion Chromatography Combined with Ultrafiltration Efficiently Isolates Extracellular Vesicles from Human Blood Samples in Health and Disease. Int J Mol Sci 2023; 24:ijms24043663. [PMID: 36835073 PMCID: PMC9963337 DOI: 10.3390/ijms24043663] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/22/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023] Open
Abstract
There is still a need for an efficient method for the isolation of extracellular vesicles (EVs) from human blood that provides a reliable yield with acceptable purity. Blood is a source of circulating EVs, but soluble proteins and lipoproteins hamper their concentration, isolation, and detection. This study aims to investigate the efficiency of EV isolation and characterization methods not defined as "gold standard". EVs were isolated from human platelet-free plasma (PFP) of patients and healthy donors through size-exclusion chromatography (SEC) combined with ultrafiltration (UF). Then, EVs were characterized using transmission electron microscopy (TEM), imaging flow cytometry (IFC), and nanoparticle tracking analysis (NTA). TEM images showed intact and roundish nanoparticles in pure samples. IFC analysis detected a prevalence of CD63+ EVs compared to CD9+, CD81+, and CD11c+ EVs. NTA confirmed the presence of small EVs with a concentration of ~1010 EVs/mL that were comparable when stratifying the subjects by baseline demographics; conversely, concentration differed according to the health status across healthy donors and patients affected with autoimmune diseases (130 subjects in total, with 65 healthy donors and 65 idiopathic inflammatory myopathy (IIM) patients). Altogether, our data show that a combined EV isolation method, i.e., SEC followed by UF, is a reliable approach to isolate intact EVs with a significant yield from complex fluids, which might characterize disease conditions early.
Collapse
Affiliation(s)
- Chiara Franco
- Unit of Rheumatology, Department of Medicine (DIMED), University of Padova, 35128 Padova, Italy
| | - Anna Ghirardello
- Unit of Rheumatology, Department of Medicine (DIMED), University of Padova, 35128 Padova, Italy
| | - Loris Bertazza
- Unit of Endocrinology, Department of Medicine (DIMED), University of Padova, 35128 Padova, Italy
| | - Michela Gasparotto
- Unit of Rheumatology, Department of Medicine (DIMED), University of Padova, 35128 Padova, Italy
| | - Elisabetta Zanatta
- Unit of Rheumatology, Department of Medicine (DIMED), University of Padova, 35128 Padova, Italy
| | - Luca Iaccarino
- Unit of Rheumatology, Department of Medicine (DIMED), University of Padova, 35128 Padova, Italy
| | - Hadi Valadi
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Guldhedsgatan 10A, 413 46 Gothenburg, Sweden
| | - Andrea Doria
- Unit of Rheumatology, Department of Medicine (DIMED), University of Padova, 35128 Padova, Italy
- Correspondence: ; Tel.: +39-0498212190
| | - Mariele Gatto
- Unit of Rheumatology, Department of Medicine (DIMED), University of Padova, 35128 Padova, Italy
| |
Collapse
|
9
|
Isolation-free measurement of single urinary extracellular vesicles by imaging flow cytometry. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 48:102638. [PMID: 36549551 DOI: 10.1016/j.nano.2022.102638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/01/2022] [Accepted: 12/02/2022] [Indexed: 12/23/2022]
Abstract
Urinary extracellular vesicles (uEVs) are promising biomarkers for various diseases. However, many tools measuring uEVs rely on time-consuming uEV isolation methods, which could induce sample bias. This study demonstrates the detection of single uEVs without isolation using imaging flow cytometry (IFCM). Unstained urine samples contained auto-fluorescent (A-F) particles when characterized with IFCM. Centrifugation successfully removed A-F particles from the unprocessed urine. Based on the disappearance of A-F particles, a gate was defined to distinguish uEVs from A-F particles. The final readouts of IFCM were verified as single EVs based on detergent treatment and serial dilutions. When developing this protocol to measure urine samples with abnormally high protein levels, 25 mg/mL dithiothreitol (DTT) showed improved uEV recovery over 200 mg/mL DTT. This study provides an isolation-free protocol using IFCM to quantify and phenotype single uEVs, eliminating the hindrance and influence of A-F particles, protein aggregates, and coincidence events.
Collapse
|
10
|
De Sousa KP, Rossi I, Abdullahi M, Ramirez MI, Stratton D, Inal JM. Isolation and characterization of extracellular vesicles and future directions in diagnosis and therapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1835. [PMID: 35898167 PMCID: PMC10078256 DOI: 10.1002/wnan.1835] [Citation(s) in RCA: 53] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 06/23/2022] [Accepted: 06/30/2022] [Indexed: 01/31/2023]
Abstract
Extracellular vesicles (EVs) are a unique and heterogeneous class of lipid bilayer nanoparticles secreted by most cells. EVs are regarded as important mediators of intercellular communication in both prokaryotic and eukaryotic cells due to their ability to transfer proteins, lipids and nucleic acids to recipient cells. In addition to their physiological role, EVs are recognized as modulators in pathological processes such as cancer, infectious diseases, and neurodegenerative disorders, providing new potential targets for diagnosis and therapeutic intervention. For a complete understanding of EVs as a universal cellular biological system and its translational applications, optimal techniques for their isolation and characterization are required. Here, we review recent progress in those techniques, from isolation methods to characterization techniques. With interest in therapeutic applications of EVs growing, we address fundamental points of EV-related cell biology, such as cellular uptake mechanisms and their biodistribution in tissues as well as challenges to their application as drug carriers or biomarkers for less invasive diagnosis or as immunogens. This article is categorized under: Diagnostic Tools > Biosensing Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease.
Collapse
Affiliation(s)
- Karina P. De Sousa
- Bioscience Research Group, School of Life and Medical SciencesUniversity of HertfordshireHertfordshireUK
| | - Izadora Rossi
- School of Human SciencesLondon Metropolitan UniversityLondonUK
- Federal University of ParanáCuritibaBrazil
| | | | - Marcel Ivan Ramirez
- Federal University of ParanáCuritibaBrazil
- Carlos Chagas Institute (ICC)CuritibaBrazil
| | - Dan Stratton
- Open UniversityThe School of Life, Health and Chemical SciencesMilton KeynesUK
| | - Jameel Malhador Inal
- Bioscience Research Group, School of Life and Medical SciencesUniversity of HertfordshireHertfordshireUK
- School of Human SciencesLondon Metropolitan UniversityLondonUK
| |
Collapse
|
11
|
The unperturbed picture: Label-free real-time optical monitoring of cells and extracellular vesicles for therapy. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2022. [DOI: 10.1016/j.cobme.2022.100414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
12
|
Bauer KM, Nelson MC, Tang WW, Chiaro TR, Brown DG, Ghazaryan A, Lee SH, Weis AM, Hill JH, Klag KA, Tran VB, Thompson JW, Ramstead AG, Monts JK, Marvin JE, Alexander M, Voth WP, Stephens WZ, Ward DM, Petrey AC, Round JL, O'Connell RM. CD11c+ myeloid cell exosomes reduce intestinal inflammation during colitis. JCI Insight 2022; 7:159469. [PMID: 36214220 PMCID: PMC9675566 DOI: 10.1172/jci.insight.159469] [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: 02/17/2022] [Accepted: 08/19/2022] [Indexed: 01/25/2023] Open
Abstract
Intercellular communication is critical for homeostasis in mammalian systems, including the gastrointestinal (GI) tract. Exosomes are nanoscale lipid extracellular vesicles that mediate communication between many cell types. Notably, the roles of immune cell exosomes in regulating GI homeostasis and inflammation are largely uncharacterized. By generating mouse strains deficient in cell-specific exosome production, we demonstrate deletion of the small GTPase Rab27A in CD11c+ cells exacerbated murine colitis, which was reversible through administration of DC-derived exosomes. Profiling RNAs within colon exosomes revealed a distinct subset of miRNAs carried by colon- and DC-derived exosomes. Among antiinflammatory exosomal miRNAs, miR-146a was transferred from gut immune cells to myeloid and T cells through a Rab27-dependent mechanism, targeting Traf6, IRAK-1, and NLRP3 in macrophages. Further, we have identified a potentially novel mode of exosome-mediated DC and macrophage crosstalk that is capable of skewing gut macrophages toward an antiinflammatory phenotype. Assessing clinical samples, RAB27A, select miRNAs, and RNA-binding proteins that load exosomal miRNAs were dysregulated in ulcerative colitis patient samples, consistent with our preclinical mouse model findings. Together, our work reveals an exosome-mediated regulatory mechanism underlying gut inflammation and paves the way for potential use of miRNA-containing exosomes as a novel therapeutic for inflammatory bowel disease.
Collapse
Affiliation(s)
- Kaylyn M Bauer
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, Utah, USA
| | - Morgan C Nelson
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, Utah, USA
| | - William W Tang
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, Utah, USA
| | - Tyson R Chiaro
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, Utah, USA
| | - D Garrett Brown
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, Utah, USA
| | - Arevik Ghazaryan
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, Utah, USA
| | - Soh-Hyun Lee
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, Utah, USA
| | - Allison M Weis
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, Utah, USA
| | - Jennifer H Hill
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, Utah, USA
| | - Kendra A Klag
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, Utah, USA
| | - Van B Tran
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, Utah, USA
| | - Jacob W Thompson
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, Utah, USA
| | - Andrew G Ramstead
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, Utah, USA
| | - Josh K Monts
- University of Utah Flow Cytometry Core, Salt Lake City, Utah, USA
| | - James E Marvin
- University of Utah Flow Cytometry Core, Salt Lake City, Utah, USA
| | - Margaret Alexander
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, Utah, USA
| | - Warren P Voth
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, Utah, USA
| | - W Zac Stephens
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, Utah, USA
| | - Diane M Ward
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, Utah, USA
| | - Aaron C Petrey
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, Utah, USA.,Department of Internal Medicine, Division of Gastroenterology, and
| | - June L Round
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, Utah, USA.,Hunstman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Ryan M O'Connell
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, Utah, USA.,Hunstman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
13
|
Visan KS, Lobb RJ, Ham S, Lima LG, Palma C, Edna CPZ, Wu L, Gowda H, Datta KK, Hartel G, Salomon C, Möller A. Comparative analysis of tangential flow filtration and ultracentrifugation, both combined with subsequent size exclusion chromatography, for the isolation of small extracellular vesicles. J Extracell Vesicles 2022; 11:e12266. [PMID: 36124834 PMCID: PMC9486818 DOI: 10.1002/jev2.12266] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 07/15/2022] [Accepted: 09/05/2022] [Indexed: 11/07/2022] Open
Abstract
Small extracellular vesicles (sEVs) provide major promise for advances in cancer diagnostics, prognostics, and therapeutics, ascribed to their distinctive cargo reflective of pathophysiological status, active involvement in intercellular communication, as well as their ubiquity and stability in bodily fluids. As a result, the field of sEV research has expanded exponentially. Nevertheless, there is a lack of standardisation in methods for sEV isolation from cells grown in serum-containing media. The majority of researchers use serum-containing media for sEV harvest and employ ultracentrifugation as the primary isolation method. Ultracentrifugation is inefficient as it is devoid of the capacity to isolate high sEV yields without contamination of non-sEV materials or disruption of sEV integrity. We comprehensively evaluated a protocol using tangential flow filtration and size exclusion chromatography to isolate sEVs from a variety of human and murine cancer cell lines, including HeLa, MDA-MB-231, EO771 and B16F10. We directly compared the performance of traditional ultracentrifugation and tangential flow filtration methods, that had undergone further purification by size exclusion chromatography, in their capacity to separate sEVs, and rigorously characterised sEV properties using multiple quantification devices, protein analyses and both image and nano-flow cytometry. Ultracentrifugation and tangential flow filtration both enrich consistent sEV populations, with similar size distributions of particles ranging up to 200 nm. However, tangential flow filtration exceeds ultracentrifugation in isolating significantly higher yields of sEVs, making it more suitable for large-scale research applications. Our results demonstrate that tangential flow filtration is a reliable and robust sEV isolation approach that surpasses ultracentrifugation in yield, reproducibility, time, costs and scalability. These advantages allow for implementation in comprehensive research applications and downstream investigations.
Collapse
Affiliation(s)
- Kekoolani S. Visan
- Tumour Microenvironment LaboratoryQIMR Berghofer Medical Research InstituteHerstonQLDAustralia
| | - Richard J. Lobb
- Tumour Microenvironment LaboratoryQIMR Berghofer Medical Research InstituteHerstonQLDAustralia
- Centre for Personalized NanomedicineAustralian Institute for Bioengineering and Nanotechnology (AIBN)The University of QueenslandBrisbaneQLDAustralia
| | - Sunyoung Ham
- Tumour Microenvironment LaboratoryQIMR Berghofer Medical Research InstituteHerstonQLDAustralia
| | - Luize G. Lima
- Tumour Microenvironment LaboratoryQIMR Berghofer Medical Research InstituteHerstonQLDAustralia
| | - Carlos Palma
- Exosome Biology LaboratoryFaculty of Medicine and Biomedical SciencesCentre for Clinical DiagnosticsUniversity of Queensland Centre for Clinical ResearchRoyal Brisbane and Women's HospitalThe University of QueenslandBrisbaneQLDAustralia
| | - Chai Pei Zhi Edna
- Tumour Microenvironment LaboratoryQIMR Berghofer Medical Research InstituteHerstonQLDAustralia
| | - Li‐Ying Wu
- Tumour Microenvironment LaboratoryQIMR Berghofer Medical Research InstituteHerstonQLDAustralia
- School of Biomedical Sciences, Faculty of HealthQueensland University of TechnologyBrisbaneQLD4059Australia
| | - Harsha Gowda
- Cancer Precision Medicine LaboratoryQIMR Berghofer Medical Research InstituteHerstonQLDAustralia
| | - Keshava K. Datta
- Cancer Precision Medicine LaboratoryQIMR Berghofer Medical Research InstituteHerstonQLDAustralia
- Proteomics and Metabolomics PlatformLa Trobe UniversityBundooraVICAustralia
| | - Gunter Hartel
- Statistics UnitQIMR Berghofer Medical Research InstituteHerstonQLDAustralia
| | - Carlos Salomon
- Exosome Biology LaboratoryFaculty of Medicine and Biomedical SciencesCentre for Clinical DiagnosticsUniversity of Queensland Centre for Clinical ResearchRoyal Brisbane and Women's HospitalThe University of QueenslandBrisbaneQLDAustralia
- Departamento de InvestigaciónPostgrado y Educación Continua (DIPEC)Facultad de Ciencias de la SaludUniversidad del AlbaSantiagoChile
| | - Andreas Möller
- Tumour Microenvironment LaboratoryQIMR Berghofer Medical Research InstituteHerstonQLDAustralia
| |
Collapse
|
14
|
Zhang DX, Dang XTT, Vu LT, Lim CMH, Yeo EYM, Lam BWS, Leong SM, Omar N, Putti TC, Yeh YC, Ma V, Luo JY, Cho WC, Chen G, Lee VKM, Grimson A, Le MTN. αvβ1 integrin is enriched in extracellular vesicles of metastatic breast cancer cells: A mechanism mediated by galectin-3. J Extracell Vesicles 2022; 11:e12234. [PMID: 35923105 PMCID: PMC9451529 DOI: 10.1002/jev2.12234] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 05/13/2022] [Accepted: 05/19/2022] [Indexed: 12/12/2022] Open
Abstract
Breast cancer cells release a large quantity of biocargo-bearing extracellular vesicles (EVs), which mediate intercellular communication within the tumour microenvironment and promote metastasis. To identify EV-bound proteins related to metastasis, we used mass spectrometry to profile EVs from highly and poorly metastatic breast cancer lines of human and mouse origins. Comparative mass spectrometry indicated that integrins, including αv and β1 subunits, are preferentially enriched in EVs of highly metastatic origin over those of poorly metastatic origin. These results are consistent with our histopathological findings, which show that integrin αv is associated with disease progression in breast cancer patients. Integrin αv colocalizes with the multivesicular-body marker CD63 at a higher frequency in the tumour and is enriched in circulating EVs of breast cancer patients at late stages when compared with circulating EVs from early-stage patients. With a magnetic bead-based flow cytometry assay, we confirmed that integrins αv and β1 are enriched in the CD63+ subsets of EVs from both human and mouse highly metastatic cells. By analysing the level of integrin αv on circulating EVs, this assay could predict the metastatic potential of a xenografted mouse model. To explore the export mechanism of integrins into EVs, we performed immunoprecipitation mass spectrometry and identified members of the galectin family as potential shuttlers of integrin αvβ1 into EVs. In particular, knockdown of galectin-3, but not galectin-1, causes a reduction in the levels of cell surface integrins β1 and αv, and decreases the colocalization of these integrins with CD63. Importantly, knockdown of galectin-3 leads to a decrease of integrin αvβ1 export into the EVs concomitant with a decrease in the metastatic potential of breast cancer cells. Moreover, inhibition of the integrin αvβ1 complex leads to a reduction in the binding of EVs to fibronectin, suggesting that integrin αvβ1 is important for EV retention in the extracellular matrix. EVs retained in the extracellular matrix are taken up by fibroblasts, which differentiate into cancer associated fibroblasts. In summary, our data indicate an important link between EV-bound integrin αvβ1 with breast cancer metastasis and provide additional insights into the export of integrin αvβ1 into EVs in the context of metastasis.
Collapse
Affiliation(s)
- Daniel Xin Zhang
- Department of Pharmacology and Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, Singapore.,Department of Surgery, Cancer Program, Immunology Program, and Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, Singapore.,Department of Biomedical Sciences, Jocky Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong SAR.,Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, USA
| | - Xuan T T Dang
- Department of Pharmacology and Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, Singapore.,Department of Surgery, Cancer Program, Immunology Program, and Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, Singapore
| | - Luyen Tien Vu
- Department of Pharmacology and Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, Singapore.,Department of Surgery, Cancer Program, Immunology Program, and Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, Singapore.,Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, USA
| | - Claudine Ming Hui Lim
- Department of Pharmacology and Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, Singapore.,Department of Surgery, Cancer Program, Immunology Program, and Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, Singapore
| | - Eric Yew Meng Yeo
- Department of Pharmacology and Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, Singapore.,Department of Surgery, Cancer Program, Immunology Program, and Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, Singapore.,Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, Singapore
| | - Brenda Wan Shing Lam
- Department of Pharmacology and Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, Singapore.,Department of Surgery, Cancer Program, Immunology Program, and Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, Singapore
| | - Sai Mun Leong
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, Singapore
| | - Noorjehan Omar
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, Singapore
| | - Thomas Choudary Putti
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, Singapore
| | - Yu Chen Yeh
- Department of Biomedical Sciences, Jocky Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong SAR
| | - Victor Ma
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong SAR
| | - Jia-Yuan Luo
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong SAR
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Victor Kwan Min Lee
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, Singapore
| | - Andrew Grimson
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, USA
| | - Minh T N Le
- Department of Pharmacology and Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, Singapore.,Department of Surgery, Cancer Program, Immunology Program, and Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, Singapore
| |
Collapse
|
15
|
Woud WW, van der Pol E, Mul E, Hoogduijn MJ, Baan CC, Boer K, Merino A. An imaging flow cytometry-based methodology for the analysis of single extracellular vesicles in unprocessed human plasma. Commun Biol 2022; 5:633. [PMID: 35768629 PMCID: PMC9243126 DOI: 10.1038/s42003-022-03569-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 05/18/2022] [Indexed: 01/02/2023] Open
Abstract
Extracellular vesicles (EVs) are tissue-specific particles released by cells containing valuable diagnostic information in the form of various biomolecules. To rule out selection bias or introduction of artefacts caused by EV isolation techniques, we present a clinically feasible, imaging flow cytometry (IFCM)-based methodology to phenotype and determine the concentration of EVs with a diameter ≤400 nm in human platelet-poor plasma (PPP) without prior isolation of EVs. Instrument calibration (both size and fluorescence) were performed with commercial polystyrene beads. Detergent treatment of EVs was performed to discriminate true vesicular events from artefacts. Using a combination of markers (CFSE & Tetraspanins, or CD9 & CD31) we found that >90% of double-positive fluorescent events represented single EVs. Through this work, we provide a framework that will allow the application of IFCM for EV analysis in peripheral blood plasma in a plethora of experimental and potentially diagnostic settings. Additionally, this direct approach for EV analysis will enable researchers to explore corners of EVs as cellular messengers in healthy and pathological conditions.
Collapse
Affiliation(s)
- Wouter W Woud
- Erasmus MC Transplant Institute, Department of Internal Medicine, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| | - Edwin van der Pol
- Biomedical Engineering & Physics, Laboratory Experimental Clinical Chemistry, Vesicle Observation Center, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Erik Mul
- Department Central Cell Analysis Facility, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Martin J Hoogduijn
- Erasmus MC Transplant Institute, Department of Internal Medicine, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Carla C Baan
- Erasmus MC Transplant Institute, Department of Internal Medicine, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Karin Boer
- Erasmus MC Transplant Institute, Department of Internal Medicine, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Ana Merino
- Erasmus MC Transplant Institute, Department of Internal Medicine, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
16
|
Helbig C, Menzen T, Wuchner K, Hawe A. Imaging Flow Cytometry for Sizing and Counting of Subvisible Particles in Biotherapeutics. J Pharm Sci 2022; 111:2458-2470. [PMID: 35777484 DOI: 10.1016/j.xphs.2022.06.019] [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: 03/01/2022] [Revised: 06/21/2022] [Accepted: 06/21/2022] [Indexed: 10/17/2022]
Abstract
Imaging flow cytometry (IFC), a technique originally designed for cellular imaging, is featured by the parallel acquisition in brightfield (BF), fluorescence (FL), and side scattering channels. Introduced to the field of subvisible particle analysis in biopharmaceuticals roughly ten years ago, it has the potential to yield additional information, e.g., on particle origin. Here, we present an extensive, systematic development of masks for IFC image analysis to optimize the accuracy of size determination of polystyrene beads and pharmaceutically relevant particles (protein, silicone oil) in BF and FL channels. Based on the developed masks, particle sizing and counting by IFC are compared to flow imaging microscopy (FIM). Mask verification based on fluorescent polystyrene particles revealed good agreement between sizes obtained from IFC and FIM. In the evaluation of counting accuracy, IFC reported lower concentrations for polystyrene particle standards than FIM. For the analysis of fluorescently stained silicone oil and protein particles however, IFC FL imaging reported higher particle concentrations in the low micrometer size range. Overall, we identified IFC as suitable tool to generate supportive data for particle characterization purposes or trouble shooting activities, but not as routine quantitative technique, e.g., for subvisible particle analysis during drug product development or quality control.
Collapse
Affiliation(s)
- C Helbig
- Coriolis Pharma Research, Fraunhoferstr. 18 b, 82152 Martinsried, Germany.
| | - T Menzen
- Coriolis Pharma Research, Fraunhoferstr. 18 b, 82152 Martinsried, Germany
| | - K Wuchner
- Janssen Research and Development, DPD&S Biotherapeutics Development, Hochstr. 201, 8200 Schaffhausen, Switzerland
| | - A Hawe
- Coriolis Pharma Research, Fraunhoferstr. 18 b, 82152 Martinsried, Germany
| |
Collapse
|
17
|
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
|
18
|
Bragina VA, Khomyakova E, Orlov AV, Znoyko SL, Mochalova EN, Paniushkina L, Shender VO, Erbes T, Evtushenko EG, Bagrov DV, Lavrenova VN, Nazarenko I, Nikitin PI. Highly Sensitive Nanomagnetic Quantification of Extracellular Vesicles by Immunochromatographic Strips: A Tool for Liquid Biopsy. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:1579. [PMID: 35564289 PMCID: PMC9101557 DOI: 10.3390/nano12091579] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/18/2022] [Accepted: 05/02/2022] [Indexed: 01/27/2023]
Abstract
Extracellular vesicles (EVs) are promising agents for liquid biopsy-a non-invasive approach for the diagnosis of cancer and evaluation of therapy response. However, EV potential is limited by the lack of sufficiently sensitive, time-, and cost-efficient methods for their registration. This research aimed at developing a highly sensitive and easy-to-use immunochromatographic tool based on magnetic nanoparticles for EV quantification. The tool is demonstrated by detection of EVs isolated from cell culture supernatants and various body fluids using characteristic biomarkers, CD9 and CD81, and a tumor-associated marker-epithelial cell adhesion molecules. The detection limit of 3.7 × 105 EV/µL is one to two orders better than the most sensitive traditional lateral flow system and commercial ELISA kits. The detection specificity is ensured by an isotype control line on the test strip. The tool's advantages are due to the spatial quantification of EV-bound magnetic nanolabels within the strip volume by an original electronic technique. The inexpensive tool, promising for liquid biopsy in daily clinical routines, can be extended to other relevant biomarkers.
Collapse
Affiliation(s)
- Vera A. Bragina
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov St., 119991 Moscow, Russia; (V.A.B.); (E.K.); (A.V.O.); (S.L.Z.); (E.N.M.)
| | - Elena Khomyakova
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov St., 119991 Moscow, Russia; (V.A.B.); (E.K.); (A.V.O.); (S.L.Z.); (E.N.M.)
| | - Alexey V. Orlov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov St., 119991 Moscow, Russia; (V.A.B.); (E.K.); (A.V.O.); (S.L.Z.); (E.N.M.)
- Moscow Institute of Physics and Technology, 9 Institutskii per., 141700 Dolgoprudny, Russia
| | - Sergey L. Znoyko
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov St., 119991 Moscow, Russia; (V.A.B.); (E.K.); (A.V.O.); (S.L.Z.); (E.N.M.)
| | - Elizaveta N. Mochalova
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov St., 119991 Moscow, Russia; (V.A.B.); (E.K.); (A.V.O.); (S.L.Z.); (E.N.M.)
- Sirius University of Science and Technology, 1 Olympic Ave., 354340 Sochi, Russia
| | - Liliia Paniushkina
- Institute for Infection Prevention and Hospital Epidemiology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (L.P.); (I.N.)
| | - Victoria O. Shender
- Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, 1a Malaya Pirogovskaya St., 119992 Moscow, Russia; (V.O.S.); (V.N.L.)
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., 117997 Moscow, Russia
| | - Thalia Erbes
- Department of Obstetrics and Gynecology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany;
| | - Evgeniy G. Evtushenko
- Lomonosov Moscow State University, 1 Leninskie Gory, 119991 Moscow, Russia; (E.G.E.); (D.V.B.)
| | - Dmitry V. Bagrov
- Lomonosov Moscow State University, 1 Leninskie Gory, 119991 Moscow, Russia; (E.G.E.); (D.V.B.)
| | - Victoria N. Lavrenova
- Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, 1a Malaya Pirogovskaya St., 119992 Moscow, Russia; (V.O.S.); (V.N.L.)
- Lomonosov Moscow State University, 1 Leninskie Gory, 119991 Moscow, Russia; (E.G.E.); (D.V.B.)
| | - Irina Nazarenko
- Institute for Infection Prevention and Hospital Epidemiology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (L.P.); (I.N.)
- German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Petr I. Nikitin
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov St., 119991 Moscow, Russia; (V.A.B.); (E.K.); (A.V.O.); (S.L.Z.); (E.N.M.)
- National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), 31 Kashirskoe Shosse, 115409 Moscow, Russia
| |
Collapse
|
19
|
Conkright WR, Beckner ME, Sahu A, Mi Q, Clemens ZJ, Lovalekar M, Flanagan SD, Martin BJ, Ferrarelli F, Ambrosio F, Nindl BC. Men and women display distinct extracellular vesicle biomarker signatures in response to military operational stress. J Appl Physiol (1985) 2022; 132:1125-1136. [PMID: 35297690 PMCID: PMC9054257 DOI: 10.1152/japplphysiol.00664.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 03/08/2022] [Accepted: 03/09/2022] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles (EVs) are mediators of physiological changes that occur during physical exertion. This study examined the effects of physical exertion with and without sleep and caloric restriction on EV size, concentration, and surface proteins in men and women. Twenty participants (10 men) completed a 5-day simulated military operational stress protocol with daily physical exertion. Blood was drawn before and immediately after exertion at baseline (D1) and following 48-h of sleep and caloric restriction (D3). EV size and concentration were assessed using nanoparticle tracking analysis. EVs were identified with markers associated with exosomes (CD63), microvesicles (VAMP3), apoptotic bodies (THSD1), and skeletal muscle-derived EVs (SGCA) and quantified using imaging flow cytometry. Interactive and main effects of sex, day, and time on EVs were assessed using three-way ANOVAs. EV concentration declined pre to postexertion in women on D1 and D3 but was stable in men. EV size increased from pre to postexertion and from D1 to D3 in men and women. Physical exertion following sleep and caloric restriction increased CD63+ EV concentration, proportion of total EVs, and CD63 surface protein expression regardless of sex. The proportion of SGCA+ EVs increased in men and women following exertion and from D1 to D3 but was higher in women than in men. No differences were observed in VAMP3+ and THSD1+ EVs. This study identified sexually dimorphic EV profiles in response to various stressors. Further investigations are necessary to determine if dimorphic EV responses affect health and performance outcomes during stress.NEW & NOTEWORTHY Sex is understudied in EV research, and most studies limit EV analysis to single stress conditions such as exercise. Multistress conditions consisting of physical exertion and sleep and caloric restriction are common in real-world settings. We demonstrate that physical exertion results in sex-specific EV signatures and that EV profiles vary according to single versus multistress conditions. Our data highlight important biological and ecological characteristics that should be considered in EV research.
Collapse
Affiliation(s)
- William R Conkright
- Neuromuscular Research Lab/Warrior Human Performance Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Meaghan E Beckner
- Neuromuscular Research Lab/Warrior Human Performance Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Amrita Sahu
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Qi Mi
- Neuromuscular Research Lab/Warrior Human Performance Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Zachary J Clemens
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mita Lovalekar
- Neuromuscular Research Lab/Warrior Human Performance Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Shawn D Flanagan
- Neuromuscular Research Lab/Warrior Human Performance Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Brian J Martin
- Neuromuscular Research Lab/Warrior Human Performance Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Fabio Ferrarelli
- School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Fabrisia Ambrosio
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Bradley C Nindl
- Neuromuscular Research Lab/Warrior Human Performance Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
20
|
Yuan HX, Liang KF, Chen C, Li YQ, Liu XJ, Chen YT, Jian YP, Liu JS, Xu YQ, Ou ZJ, Li Y, Ou JS. Size Distribution of Microparticles: A New Parameter to Predict Acute Lung Injury After Cardiac Surgery With Cardiopulmonary Bypass. Front Cardiovasc Med 2022; 9:893609. [PMID: 35571221 PMCID: PMC9098995 DOI: 10.3389/fcvm.2022.893609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 03/30/2022] [Indexed: 11/13/2022] Open
Abstract
Background Acute lung injury (ALI) is a common complication after cardiac surgery with cardiopulmonary bypass (CPB). No precise way, however, is currently available to predict its occurrence. We and others have demonstrated that microparticles (MPs) can induce ALI and were increased in patients with ALI. However, whether MPs can be used to predict ALI after cardiac surgery with CPB remains unknown. Methods In this prospective study, 103 patients undergoing cardiac surgery with CPB and 53 healthy subjects were enrolled. MPs were isolated from the plasma before, 12 h after, and 3 d after surgery. The size distributions of MPs were measured by the LitesizerTM 500 Particle Analyzer. The patients were divided into two subgroups (ALI and non-ALI) according to the diagnosis of ALI. Descriptive and correlational analyzes were conducted between the size distribution of MPs and clinical data. Results Compared to the non-ALI group, the size at peak and interquartile range (IQR) of MPs in patients with ALI were smaller, but the peak intensity of MPs is higher. Multivariate logistic regression analysis indicated that the size at peak of MPs at postoperative 12 h was an independent risk factor for ALI. The area under the curve (AUC) of peak diameter at postoperative 12 h was 0.803. The best cutoff value of peak diameter to diagnose ALI was 223.05 nm with a sensitivity of 88.0% and a negative predictive value of 94.5%. The AUC of IQR at postoperative 12 h was 0.717. The best cutoff value of IQR to diagnose ALI was 132.65 nm with a sensitivity of 88.0% and a negative predictive value of 92.5%. Combining these two parameters, the sensitivity reached 92% and the negative predictive value was 96%. Conclusions Our findings suggested that the size distribution of MPs could be a novel biomarker to predict and exclude ALI after cardiac surgery with CPB.
Collapse
Affiliation(s)
- Hao-Xiang Yuan
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
- NHC key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
| | - Kai-Feng Liang
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
- NHC key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
| | - Chao Chen
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
- NHC key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
| | - Yu-Quan Li
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
- NHC key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
| | - Xiao-Jun Liu
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
- NHC key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
| | - Ya-Ting Chen
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
- NHC key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
| | - Yu-Peng Jian
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
- NHC key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
| | - Jia-Sheng Liu
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
- NHC key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
| | - Ying-Qi Xu
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
- NHC key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
| | - Zhi-Jun Ou
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
- NHC key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
- Division of Hypertension and Vascular Diseases, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Zhi-Jun Ou
| | - Yan Li
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
- NHC key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
- Yan Li
| | - Jing-Song Ou
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
- NHC key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, China
- Jing-Song Ou ;
| |
Collapse
|
21
|
Beckner ME, Conkright WR, Sahu A, Mi Q, Clemens ZJ, Martin BJ, Flanagan SD, Ferrarelli F, Ambrosio F, Nindl BC. Utility of extracellular vesicles as a potential biological indicator of physiological resilience during military operational stress. Physiol Rep 2022; 10:e15219. [PMID: 35373929 PMCID: PMC8978596 DOI: 10.14814/phy2.15219] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 12/01/2022] Open
Abstract
Extracellular vesicles (EVs) transport biological content between cells to mediate physiological processes. The association between EVs and resilience, the ability to cope with stress, is unknown. Using unbiased machine learning approaches, we aimed to identify a biological profile of resilience. Twenty servicemen (27.8 ± 5.9 years) completed the Connor Davidson Resilience (CD-RISC) questionnaire and were exposed to daily physical and cognitive exertion with 48-hr sleep and caloric restriction. Blood samples from baseline and the second day of stress were analyzed for neuroendocrine biomarkers impacted by military stress. EVs were isolated from plasma and stained with antibodies associated with exosomes (CD63), microvesicles (VAMP3), and apoptotic bodies (THSD1). Individuals were separated into high (n = 10, CD-RISC > 90) and low (n = 10, CD-RISC < 79) resilience. EV features were stratified by size, then down-selected using regression trees and compared between groups. Diagnostic accuracy was assessed using receiver operating characteristic curves. Compared to low resilience, high resilience demonstrated a greater increase in variability of THSD1 local bright spot intensities among large-sized EVs in response to stress (p = 0.002, Hedges' g = 1.59). Among medium-sized EVs, high resilience exhibited a greater decrease in side scatter intensity (p = 0.014, Hedges' g = 1.17). Both features demonstrated high to moderate diagnostic accuracy for high resilience (AUC = 0.90 and 0.79). In contrast, neuroendocrine biomarker concentrations were similar between groups. The increase in variability among THSD1 + EVs in high, but not low, resilient individuals following stress may suggest high resilience is accompanied by stress-triggered apoptotic adaptations to the environment that are not detected in neuroendocrine biomarkers.
Collapse
Affiliation(s)
- Meaghan E Beckner
- Neuromuscular Research Laboratory/Warrior Human Performance Research Center, Department of Sports Medicine and Nutrition, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - William R Conkright
- Neuromuscular Research Laboratory/Warrior Human Performance Research Center, Department of Sports Medicine and Nutrition, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Amrita Sahu
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Qi Mi
- Neuromuscular Research Laboratory/Warrior Human Performance Research Center, Department of Sports Medicine and Nutrition, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Zachary J Clemens
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Brian J Martin
- Neuromuscular Research Laboratory/Warrior Human Performance Research Center, Department of Sports Medicine and Nutrition, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Shawn D Flanagan
- Neuromuscular Research Laboratory/Warrior Human Performance Research Center, Department of Sports Medicine and Nutrition, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Fabio Ferrarelli
- School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Fabrisia Ambrosio
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Bradley C Nindl
- Neuromuscular Research Laboratory/Warrior Human Performance Research Center, Department of Sports Medicine and Nutrition, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
22
|
Analysis of the Interaction of Human Neuroblastoma Cell-Derived Cytochalasin B Induced Membrane Vesicles with Mesenchymal Stem Cells Using Imaging Flow Cytometry. BIONANOSCIENCE 2022; 12:293-301. [PMID: 35261871 PMCID: PMC8894839 DOI: 10.1007/s12668-021-00931-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2021] [Indexed: 11/24/2022]
Abstract
At present, there is an increasing interest in the potential role of extracellular vesicles (EVs), acting as multi-signal messengers of the tumor stroma, in the development and progression of tumor. Tumor cell-derived EVs are considered a potential vector for the targeted delivery of antitumor agents due to the ability to fuse with parental cells through endocytosis and release their contents into the cytoplasm of the recipient cell. Tumor cell-derived EVs could be also used for priming immune cells and therapeutic vaccine development. It is also known that mesenchymal stem cells (MSCs) have a tropism toward tumor niches. It is believed that MSC migration to the tumor is due to its inflammatory signaling. Presumably, with the accumulation of MSCs at tumor sites, these cells differentiate into pericytes or tumor-associated fibroblasts, thereby forming a supporting tumor growth microenvironment. However, besides the ability to promote tumor progression, MSCs can also suppress its growth by inhibiting proliferation and cell cycle progression, and angiogenesis. Thus, the further studies of the MSC role in TME and MSC interaction with other cells of the tumor stroma, including through EVs, are of particular interest. To increase the yield of vesicles the isolation method based on pharmacological disorganization of the actin cytoskeleton induced by treating with cytochalasin B was used in this study. In this investigation the interaction of SH-SY5Y neuroblastoma cell-derived membrane vesicles, obtained using cytochalasin B (CIMVs), with human bone marrow-derived MSCs was analyzed using imaging flow cytometry. Using transmission electron microscopy, it was shown that CIMVs have a size similar to that of natural microvesicles, which is 100–1000 nm. Using imaging flow cytometry, it was shown that after 24 h of co-cultivation 6% of the MSCs contained a large number of CIMVs, and 42% of the MSCs contained a small amount of CIMVs. Cultivation of MSCs with SH-SY5Y cell-derived CIMVs also induced dose-dependent decrease in the expression of CD markers typical for MSCs. Thus, the internalization of SH-SY5Y cell-derived CIMVs within MSCs and the ability of the CIMVs to modulate immunophenotype of the recipient cells were shown. However, further studies are required to determine the effect of CIMVs on pro- or antioncogenic phenotype and function of MSCs.
Collapse
|
23
|
Alfandari D, Ben Ami Pilo H, Abou Karam P, Dagan O, Joubran C, Rotkopf R, Regev-Rudzki N, Porat Z. Monitoring Distribution Dynamics of EV RNA Cargo Within Recipient Monocytes and Macrophages. Front Cell Infect Microbiol 2022; 11:739628. [PMID: 35155269 PMCID: PMC8825493 DOI: 10.3389/fcimb.2021.739628] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 12/31/2021] [Indexed: 12/19/2022] Open
Abstract
Extracellular vesicles (EVs) are produced by across almost all the living kingdoms and play a crucial role in cell-cell communication processes. EVs are especially important for pathogens, as Plasmodium falciparum (Pf) parasite, the leading causing species in human malaria. Malaria parasites are able to modulate the host immune response from a distance via delivering diverse cargo components inside the EVs, such as proteins and nucleic acids. We have previously shown that imaging flow cytometry (IFC) can be effectively used to monitor the uptake of different cargo components of malaria derived EVs by host human monocytes. Here, we take this approach one step further and demonstrate that we can directly investigate the dynamics of the cargo distribution pattern over time by monitoring its distribution within two different recipient cells of the immune system, monocytes vs macrophages. By staining the RNA cargo of the vesicles and monitor the signal we were able to evaluate the kinetics of its delivery and measure different parameters of the cargo’s distribution post internalization. Interestingly, we found that while the level of the EV uptake is similar, the pattern of the signal for RNA cargo distribution is significantly different between these two recipient immune cells. Our results demonstrate that this method can be applied to study the distribution dynamics of the vesicle cargo post uptake to different types of cells. This can benefit significantly to our understanding of the fate of cargo components post vesicle internalization in the complex interface between pathogen-derived vesicles and their host recipient cells.
Collapse
Affiliation(s)
- Daniel Alfandari
- Department of Biomolecular Sciences, Faculty of Biochemistry, Weizmann Institute of Science, Rehovot, Israel
| | - Hila Ben Ami Pilo
- Department of Biomolecular Sciences, Faculty of Biochemistry, Weizmann Institute of Science, Rehovot, Israel
| | - Paula Abou Karam
- Department of Biomolecular Sciences, Faculty of Biochemistry, Weizmann Institute of Science, Rehovot, Israel
| | - Osnat Dagan
- Department of Biomolecular Sciences, Faculty of Biochemistry, Weizmann Institute of Science, Rehovot, Israel
| | - Carine Joubran
- Department of Biomolecular Sciences, Faculty of Biochemistry, Weizmann Institute of Science, Rehovot, Israel
| | - Ron Rotkopf
- Bioinformatics Unit, Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Neta Regev-Rudzki
- Department of Biomolecular Sciences, Faculty of Biochemistry, Weizmann Institute of Science, Rehovot, Israel
- *Correspondence: Neta Regev-Rudzki, ; Ziv Porat,
| | - Ziv Porat
- Flow Cytometry Unit, Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
- *Correspondence: Neta Regev-Rudzki, ; Ziv Porat,
| |
Collapse
|
24
|
In-Cell Labeling Coupled to Direct Analysis of Extracellular Vesicles in the Conditioned Medium to Study Extracellular Vesicles Secretion with Minimum Sample Processing and Particle Loss. Cells 2022; 11:cells11030351. [PMID: 35159161 PMCID: PMC8833937 DOI: 10.3390/cells11030351] [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: 12/22/2021] [Revised: 01/06/2022] [Accepted: 01/12/2022] [Indexed: 02/04/2023] Open
Abstract
Extracellular vesicles (EVs) are involved in a multitude of physiological functions and play important roles in health and disease. The largest proportion of studies on EVs is based on the analysis and characterization of EVs secreted in the cell culture medium. These studies remain challenging due to the small size of the EV particles, a lack of universal EV markers, and sample loss or technical artifacts that are often associated with EV labeling for single particle tracking and/or separation techniques. To address these problems, we characterized and validated a method for in-cell EV labeling with fluorescent lipids coupled with direct analysis of lipid-labeled EVs in the conditioned medium by imaging flow cytometry (IFC). This approach significantly reduces sample processing and loss compared to established methods for EV separation and labeling in vitro, resulting in improved detection of quantitative changes in EV secretion and subpopulations compared to protocols that rely on EV separation by size-exclusion chromatography and ultracentrifugation. Our optimized protocol for in-cell EV labeling and analysis of the conditioned medium reduces EV sample processing and loss, and is well-suited for cell biology studies that focus on modulation of EV secretion by cells in culture.
Collapse
|
25
|
Mugoni V, Ciani Y, Nardella C, Demichelis F. Circulating RNAs in prostate cancer patients. Cancer Lett 2022; 524:57-69. [PMID: 34656688 DOI: 10.1016/j.canlet.2021.10.011] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 10/06/2021] [Accepted: 10/09/2021] [Indexed: 12/12/2022]
Abstract
Growing bodies of evidence have demonstrated that the identification of prostate cancer (PCa) biomarkers in the patients' blood and urine may remarkably improve PCa diagnosis and progression monitoring. Among diverse cancer-derived circulating materials, extracellular RNA molecules (exRNAs) represent a compelling component to investigate cancer-related alterations. Once outside the intracellular environment, exRNAs circulate in biofluids either in association with protein complexes or encapsulated inside extracellular vesicles (EVs). Notably, EV-associated RNAs (EV-RNAs) were used for the development of several assays (such as the FDA-approved Progensa Prostate Cancer Antigen 3 (PCA3 test) aiming at improving early PCa detection. EV-RNAs encompass a mixture of species, including small non-coding RNAs (e.g. miRNA and circRNA), lncRNAs and mRNAs. Several methods have been proposed to isolate EVs and relevant RNAs, and to perform RNA-Seq studies to identify potential cancer biomarkers. However, EVs in the circulation of a cancer patient include a multitude of diverse populations that are released by both cancer and normal cells from different tissues, thereby leading to a heterogeneous EV-RNA-associated transcriptional signal. Decrypting the complexity of such a composite signal is nowadays the major challenge faced in the identification of specific tumor-associated RNAs. Multiple deconvolution algorithms have been proposed so far to infer the enrichment of cancer-specific signals from gene expression data. However, novel strategies for EVs sorting and sequencing of RNA associated to single EVs populations will remarkably facilitate the identification of cancer-related molecules. Altogether, the studies summarized here demonstrate the high potential of using EV-RNA biomarkers in PCa and highlight the urgent need of improving technologies and computational approaches to characterize specific EVs populations and their relevant RNA cargo.
Collapse
Affiliation(s)
- Vera Mugoni
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Yari Ciani
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Caterina Nardella
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Francesca Demichelis
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy.
| |
Collapse
|
26
|
Highton PJ, March DS, Churchward DR, Grantham CE, Young HML, Graham-Brown MPM, Estruel S, Martin N, Brunskill NJ, Smith AC, Burton JO, Bishop NC. Intradialytic cycling does not exacerbate microparticles or circulating markers of systemic inflammation in haemodialysis patients. Eur J Appl Physiol 2021; 122:599-609. [PMID: 34854982 PMCID: PMC8854296 DOI: 10.1007/s00421-021-04846-7] [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: 09/08/2021] [Accepted: 11/09/2021] [Indexed: 11/24/2022]
Abstract
Purpose Patients receiving haemodialysis (HD) display elevated circulating microparticle (MP) concentration, tissue factor (TF) expression and markers of systemic inflammation, though regular intradialytic cycling (IDC) may have a therapeutic effect. This study investigated the impact of regular, moderate-intensity IDC on circulating MPs and inflammatory markers in unit-based HD patients. Methods Patients were cluster-randomised to intervention (n = 20, age: 51.4 ± 18.1 years, body mass: 77.6 ± 18.3 kg, mean ± SD) or no-exercise control (n = 20, 56.8 ± 14.0 years, 80.5 ± 26.5 kg). Intervention participants completed 30 min of moderate intensity (rating of perceived exertion [RPE] of 12–14) IDC, thrice weekly for 6 months. Pre-dialysis venous blood samples were obtained at 0, 3 and 6 months. Circulating MP phenotypes, cytokines, chemokine and MP TF expression were quantified using flow cytometry and cytometric bead array assays. Results Despite high exercise compliance (82%), no IDC-dependent effects were observed for any MP, cytokine or chemokine measure (p ≥ 0.051, ηρ2 ≤ 0.399) other than TNF-α (p = 0.001, ηρ2 = 0.186), though no significance was revealed upon post hoc analysis. Conclusion Six months of regular, moderate-intensity IDC had no effect on MPs, cytokines or chemokines. This suggests that the exercise did not exacerbate thrombotic or inflammatory status, though further functional assays are required to confirm this. Trial registration ISRCTN1129707, prospectively registered on 05/03/2015.
Collapse
Affiliation(s)
- Patrick J Highton
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.,NIHR Applied Research Collaboration East Midlands, Leicester Diabetes Centre of Research, University of Leicester, Leicester, UK
| | - Daniel S March
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK.,John Walls Renal Unit, Leicester General Hospital, University Hospitals of Leicester, Leicester, UK
| | - Darren R Churchward
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | | | - Hannah M L Young
- John Walls Renal Unit, Leicester General Hospital, University Hospitals of Leicester, Leicester, UK.,Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Matthew P M Graham-Brown
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK.,John Walls Renal Unit, Leicester General Hospital, University Hospitals of Leicester, Leicester, UK
| | - Seila Estruel
- Department of Physiological Sciences, University of Barcelona, Barcelona, Spain
| | - Naomi Martin
- Leicester School of Allied Health Sciences, Faculty of Health and Life Sciences, De Montfort University, Leicester, UK
| | - Nigel J Brunskill
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK.,John Walls Renal Unit, Leicester General Hospital, University Hospitals of Leicester, Leicester, UK
| | - Alice C Smith
- John Walls Renal Unit, Leicester General Hospital, University Hospitals of Leicester, Leicester, UK.,Department of Health Sciences, University of Leicester, Leicester, UK
| | - James O Burton
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.,Department of Cardiovascular Sciences, University of Leicester, Leicester, UK.,John Walls Renal Unit, Leicester General Hospital, University Hospitals of Leicester, Leicester, UK
| | - Nicolette C Bishop
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.
| |
Collapse
|
27
|
Zhao M, Zhang F, Zarnowski R, Barns K, Jones R, Fossen J, Sanchez H, Rajski SR, Audhya A, Bugni TS, Andes DR. Turbinmicin inhibits Candida biofilm growth by disrupting fungal vesicle-mediated trafficking. J Clin Invest 2021; 131:145123. [PMID: 33373326 DOI: 10.1172/jci145123] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 12/16/2020] [Indexed: 12/12/2022] Open
Abstract
The emergence of drug-resistant fungi has prompted an urgent threat alert from the US Centers for Disease Control (CDC). Biofilm assembly by these pathogens further impairs effective therapy. We recently identified an antifungal, turbinmicin, that inhibits the fungal vesicle-mediated trafficking pathway and demonstrates broad-spectrum activity against planktonically growing fungi. During biofilm growth, vesicles with unique features play a critical role in the delivery of biofilm extracellular matrix components. As these components are largely responsible for the drug resistance associated with biofilm growth, we explored the utility of turbinmicin in the biofilm setting. We found that turbinmicin disrupted extracellular vesicle (EV) delivery during biofilm growth and that this impaired the subsequent assembly of the biofilm matrix. We demonstrated that elimination of the extracellular matrix rendered the drug-resistant biofilm communities susceptible to fungal killing by turbinmicin. Furthermore, the addition of turbinmicin to otherwise ineffective antifungal therapy potentiated the activity of these drugs. The underlying role of vesicles explains this dramatic activity and was supported by phenotype reversal with the addition of exogenous biofilm EVs. This striking capacity to cripple biofilm assembly mechanisms reveals a new approach to eradicating biofilms and sheds light on turbinmicin as a promising anti-biofilm drug.
Collapse
Affiliation(s)
- Miao Zhao
- Department of Medicine, Medical Microbiology and Immunology
| | - Fan Zhang
- Pharmaceutical Sciences Division, and
| | | | | | - Ryley Jones
- Department of Medicine, Medical Microbiology and Immunology
| | - Jen Fossen
- Department of Medicine, Medical Microbiology and Immunology
| | - Hiram Sanchez
- Department of Medicine, Medical Microbiology and Immunology
| | | | - Anjon Audhya
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, Wisconsin, USA
| | | | - David R Andes
- Department of Medicine, Medical Microbiology and Immunology
| |
Collapse
|
28
|
Scott A, Sueiro Ballesteros L, Bradshaw M, Tsuji C, Power A, Lorriman J, Love J, Paul D, Herman A, Emanueli C, Richardson RJ. In Vivo Characterization of Endogenous Cardiovascular Extracellular Vesicles in Larval and Adult Zebrafish. Arterioscler Thromb Vasc Biol 2021; 41:2454-2468. [PMID: 34261327 PMCID: PMC8384253 DOI: 10.1161/atvbaha.121.316539] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 06/30/2021] [Indexed: 01/08/2023]
Abstract
Objective Extracellular vesicles (EVs) facilitate molecular transport across extracellular space, allowing local and systemic signaling during homeostasis and in disease. Extensive studies have described functional roles for EV populations, including during cardiovascular disease, but the in vivo characterization of endogenously produced EVs is still in its infancy. Because of their genetic tractability and live imaging amenability, zebrafish represent an ideal but under-used model to investigate endogenous EVs. We aimed to establish a transgenic zebrafish model to allow the in vivo identification, tracking, and extraction of endogenous EVs produced by different cell types. Approach and Results Using a membrane-tethered fluorophore reporter system, we show that EVs can be fluorescently labeled in larval and adult zebrafish and demonstrate that multiple cell types including endothelial cells and cardiomyocytes actively produce EVs in vivo. Cell-type specific EVs can be tracked by high spatiotemporal resolution light-sheet live imaging and modified flow cytometry methods allow these EVs to be further evaluated. Additionally, cryo electron microscopy reveals the full morphological diversity of larval and adult EVs. Importantly, we demonstrate the utility of this model by showing that different cell types exchange EVs in the adult heart and that ischemic injury models dynamically alter EV production. Conclusions We describe a powerful in vivo zebrafish model for the investigation of endogenous EVs in all aspects of cardiovascular biology and pathology. A cell membrane fluorophore labeling approach allows cell-type specific tracing of EV origin without bias toward the expression of individual protein markers and will allow detailed future examination of their function.
Collapse
Affiliation(s)
- Aaron Scott
- School of Physiology, Pharmacology & Neuroscience, Faculty of Biomedical Sciences (A.S., M.B., C.T., J.L., D.P., R.J.R.)
| | - Lorena Sueiro Ballesteros
- Flow Cytometry Facility, Faculty of Biomedical Sciences (L.S.B., A.H.)
- Now with Charles River Laboratories, Discovery House, Quays Office Park, Conference Avenue, Portishead, Bristol, United Kingdom (L.S.B.)
| | - Marston Bradshaw
- School of Physiology, Pharmacology & Neuroscience, Faculty of Biomedical Sciences (A.S., M.B., C.T., J.L., D.P., R.J.R.)
| | - Chisato Tsuji
- School of Physiology, Pharmacology & Neuroscience, Faculty of Biomedical Sciences (A.S., M.B., C.T., J.L., D.P., R.J.R.)
| | - Ann Power
- BioEconomy Centre, The Henry Wellcome Building for Biocatalysis, Biosciences, University of Exeter, United Kingdom (A.P., J.L.)
| | - James Lorriman
- School of Physiology, Pharmacology & Neuroscience, Faculty of Biomedical Sciences (A.S., M.B., C.T., J.L., D.P., R.J.R.)
| | - John Love
- BioEconomy Centre, The Henry Wellcome Building for Biocatalysis, Biosciences, University of Exeter, United Kingdom (A.P., J.L.)
| | - Danielle Paul
- School of Physiology, Pharmacology & Neuroscience, Faculty of Biomedical Sciences (A.S., M.B., C.T., J.L., D.P., R.J.R.)
| | - Andrew Herman
- Flow Cytometry Facility, Faculty of Biomedical Sciences (L.S.B., A.H.)
| | - Costanza Emanueli
- Bristol Heart Institute, School of Clinical Science (C.E.), University of Bristol, United Kingdom
- National Heart and Lung Institute, Imperial College London, United Kingdom (C.E.)
| | - Rebecca J. Richardson
- School of Physiology, Pharmacology & Neuroscience, Faculty of Biomedical Sciences (A.S., M.B., C.T., J.L., D.P., R.J.R.)
| |
Collapse
|
29
|
Zhao Z, Wijerathne H, Godwin AK, Soper SA. Isolation and analysis methods of extracellular vesicles (EVs). EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2021; 2:80-103. [PMID: 34414401 PMCID: PMC8372011 DOI: 10.20517/evcna.2021.07] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Extracellular vesicles (EVs) have been recognized as an evolving biomarker within the liquid biopsy family. While carrying both host cell proteins and different types of RNAs, EVs are also present in sufficient quantities in biological samples to be tested using many molecular analysis platforms to interrogate their content. However, because EVs in biological samples are comprised of both disease and non-disease related EVs, enrichment is often required to remove potential interferences from the downstream molecular assay. Most benchtop isolation/enrichment methods require > milliliter levels of sample and can cause varying degrees of damage to the EVs. In addition, some of the common EV benchtop isolation methods do not sort the diseased from the non-diseased related EVs. Simultaneously, the detection of the overall concentration and size distribution of the EVs is highly dependent on techniques such as electron microscopy and Nanoparticle Tracking Analysis, which can include unexpected variations and biases as well as complexity in the analysis. This review discusses the importance of EVs as a biomarker secured from a liquid biopsy and covers some of the traditional and non-traditional, including microfluidics and resistive pulse sensing, technologies for EV isolation and detection, respectively.
Collapse
Affiliation(s)
- Zheng Zhao
- Bioengineering Program, University of Kansas, Lawrence, KS 66045, USA.,Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA
| | - Harshani Wijerathne
- Department of Mechanical Engineering, Temple University, Philadelphia, PA 19122, USA
| | - Andrew K Godwin
- KU Cancer Center, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Steven A Soper
- Bioengineering Program, University of Kansas, Lawrence, KS 66045, USA.,Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA.,Department of Chemistry, University of Kansas, Lawrence, KS 66045, USA.,Department of Mechanical Engineering, University of Kansas, Lawrence, KS 66045, USA.,KU Cancer Center, University of Kansas Medical Center, Kansas City, KS 66160, USA.,Ulsan National Institute of Science & Technology, Ulju-gun, Ulsan, 44919, South Korea
| |
Collapse
|
30
|
RoŽanc J, Finšgar M, Maver U. Progressive use of multispectral imaging flow cytometry in various research areas. Analyst 2021; 146:4985-5007. [PMID: 34337638 DOI: 10.1039/d1an00788b] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Multi-spectral imaging flow cytometry (MIFC) has become one of the most powerful technologies for investigating general analytics, molecular and cell biology, biotechnology, medicine, and related fields. It combines the capabilities of the morphometric and photometric analysis of single cells and micrometer-sized particles in flux with regard to thousands of events. It has become the tool of choice for a wide range of research and clinical applications. By combining the features of flow cytometry and fluorescence microscopy, it offers researchers the ability to couple the spatial resolution of multicolour images of cells and organelles with the simultaneous analysis of a large number of events in a single system. This provides the opportunity to visually confirm findings and collect novel data that would otherwise be more difficult to obtain. This has led many researchers to design innovative assays to gain new insight into important research questions. To date, it has been successfully used to study cell morphology, surface and nuclear protein co-localization, protein-protein interactions, cell signaling, cell cycle, cell death, and cytotoxicity, intracellular calcium, drug uptake, pathogen internalization, and other applications. Herein we describe some of the recent advances in the field of multiparametric imaging flow cytometry methods in various research areas.
Collapse
Affiliation(s)
- Jan RoŽanc
- University of Maribor, Faculty of Medicine, Institute of Biomedical Sciences, Taborska ulica 8, SI-2000 Maribor, Slovenia.
| | | | | |
Collapse
|
31
|
Jiao Y, Lu W, Xu P, Shi H, Chen D, Chen Y, Shi H, Ma Y. Hepatocyte-derived exosome may be as a biomarker of liver regeneration and prognostic valuation in patients with acute-on-chronic liver failure. Hepatol Int 2021; 15:957-969. [PMID: 34232468 DOI: 10.1007/s12072-021-10217-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 05/31/2021] [Indexed: 01/20/2023]
Abstract
BACKGROUND The assessment of liver regeneration is particularly critical for patients with acute-on-chronic liver failure (ACLF). Exosome has both the advantages of specificity of liver biopsy and noninvasion of peripheral blood, which may be the potential biomarker of liver disease. METHODS The patients with chronic hepatitis B (CHB) and ACLF were enrolled from outpatients and inpatients in Beijing Youan Hospital, Capital Medical University. The exosomes in plasma were extracted by ultracentrifuge using Optima XPN-100 Ultracentrifuge. Exosomes were dyed with fluorescent direct-labeled antibody and the expression profile was assayed using ImageStream® X MKII Imaging Flow Cytometer. RESULTS The percentage of exosomes with ALB and CD63 was significant higher in ACLF than that in CHB. The percentage of exosomes with ALB and CD63 and VEGF increased in CHB, but decreased in ACLF. The exosomes with ALB, CD63, and VEGF were significant more in survival group than that in dead group in patients with ACLF. The sensitivity and specificity of exosomes with CD63, ALB, and VEGF were significantly higher than the other markers of liver regeneration and prognostic valuation in patients with ACLF including AFP. The hepatocyte-derived exosomes expression profile had no difference in different stages and different AFP levels of patients with ACLF. CONCLUSION The exosomes profile with ALB and VEGF may be a more accurate and specific biomarker of liver regeneration and prognostic valuation than AFP in patients with ACLF. In addition, the exosomes profile with CD63 and ALB may be an early-warning marker in patients with ACLF.
Collapse
Affiliation(s)
- Yan Jiao
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
- Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing, 100069, China
| | - Wang Lu
- Fourth Department of Liver Disease, Beijing Youan Hospital, Capital Medical University, Beijing Municipal Key Laboratory of Liver Failure and Artificial Liver Treatment Research, Beijing, 100069, China
| | - Ping Xu
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
- Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing, 100069, China
| | - Honglin Shi
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
- Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing, 100069, China
| | - Dexi Chen
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
- Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing, 100069, China
| | - Yu Chen
- Fourth Department of Liver Disease, Beijing Youan Hospital, Capital Medical University, Beijing Municipal Key Laboratory of Liver Failure and Artificial Liver Treatment Research, Beijing, 100069, China.
| | - Hongbo Shi
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China.
- Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing, 100069, China.
| | - Yingmin Ma
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China.
- Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing, 100069, China.
| |
Collapse
|
32
|
Pekárková K, Soukup J, Kostelanská M, Širc J, Straňák Z, Holada K. Cord Blood Extracellular Vesicles Analyzed by Flow Cytometry with Thresholding Using 405 nm or 488 nm Laser Leads to Concurrent Results. Diagnostics (Basel) 2021; 11:1320. [PMID: 34441255 PMCID: PMC8392526 DOI: 10.3390/diagnostics11081320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/15/2021] [Accepted: 07/20/2021] [Indexed: 11/17/2022] Open
Abstract
Extracellular vesicles (EVs) from liquid biopsies are extensively analyzed by flow cytometry, a technology that is continuously evolving. Thresholding utilizing a violet 405 nm laser side scatter (VSSC) has recently been implemented. Here, we collected set of large EV (lEV) samples from cord blood, which we analyzed using a standard flow cytometer improved via a 405 nm laser side scatter. Samples were analyzed using two distinct thresholding methods-one based on VSSC, and one based on VSSC combined with fluorescence thresholding on stained phosphatidylserine. Through these thresholding methods, we compared lEVs from pre-term births and control cord blood. Double-labeled lEVs with platelet CD36+/CD41+, activated platelet CD41+/CD62P+ and endothelial CD31+/CD105+ antibodies were used. Apart from comparing the two groups together, we also correlated measured lEVs with the thresholding methods. We also correlated the results of this study with data analyzed in our previous study in which we used a conventional 488 nm laser SSC. We did not find any difference between the two cord blood groups. However, we found highly concurrent data via our correlation of the thresholding methods, with correlation coefficients ranging from 0.80 to 0.96 even though the numbers of detected lEVs differed between thresholding methods. In conclusion, our approaches to thresholding provided concurrent data and it seems that improving the cytometer with the use of a VSSC increases its sensitivity, despite not being particularly critical to the validity of flow cytometric studies that compare pathological and physiological conditions in liquid biopsies.
Collapse
Affiliation(s)
- Kristýna Pekárková
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University, 128 00 Prague, Czech Republic; (K.P.); (M.K.); (K.H.)
| | - Jakub Soukup
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University, 128 00 Prague, Czech Republic; (K.P.); (M.K.); (K.H.)
- Department of Genetics and Microbiology, Faculty of Science, Charles University, 128 44 Prague, Czech Republic
| | - Marie Kostelanská
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University, 128 00 Prague, Czech Republic; (K.P.); (M.K.); (K.H.)
| | - Jan Širc
- The Institute for the Care of Mother and Child, 147 00 Prague, Czech Republic; (J.Š.); (Z.S.)
| | - Zbyněk Straňák
- The Institute for the Care of Mother and Child, 147 00 Prague, Czech Republic; (J.Š.); (Z.S.)
| | - Karel Holada
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University, 128 00 Prague, Czech Republic; (K.P.); (M.K.); (K.H.)
| |
Collapse
|
33
|
Mastoridis S, Londoño MC, Kurt A, Kodela E, Crespo E, Mason J, Bestard O, Martínez-Llordella M, Sánchez-Fueyo A. Impact of donor extracellular vesicle release on recipient cell "cross-dressing" following clinical liver and kidney transplantation. Am J Transplant 2021; 21:2387-2398. [PMID: 32515541 DOI: 10.1111/ajt.16123] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 05/26/2020] [Accepted: 05/30/2020] [Indexed: 01/25/2023]
Abstract
In several murine models of transplantation, the "cross-dressing" of recipient antigen presenting cells (APCs) with intact donor major histocompatibility complex (MHC) derived from allograft-released small extracellular vesicles (sEVs) has been recently described as a key mechanism in eliciting and sustaining alloimmune responses. Investigation of these processes in clinical organ transplantation has, however, been hampered by the lack of sensitivity of conventional instruments and assays. We have employed advanced imaging flow cytometry (iFCM) to explore the kinetics of allograft sEV release and the extent to which donor sEVs might induce cross-dressing following liver and kidney transplantation. We report for the first time that recipient APC cross-dressing can be transiently detected in the circulation shortly after liver, but not kidney, transplantation in association with the release of HLA-bearing allograft-derived sEVs. In liver transplant recipients the majority of circulating cells exhibiting donor HLA are indeed cross-dressed cells and not passenger leukocytes. In keeping with experimental animal data, the downstream functional consequences of the transfer of circulating sEVs harvested from human transplant recipients varies depending on the type of transplant and time posttransplant. sEVs released shortly after liver, but not kidney, transplantation exhibit immunoinhibitory effects that could influence liver allograft immunogenicity.
Collapse
Affiliation(s)
- Sotiris Mastoridis
- Medical Research Council (MRC) Centre for Transplantation, Institute of Liver Studies, King's College London, London, United Kingdom
| | - María-Carlota Londoño
- Medical Research Council (MRC) Centre for Transplantation, Institute of Liver Studies, King's College London, London, United Kingdom.,Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, Barcelona, Spain
| | - Ada Kurt
- Medical Research Council (MRC) Centre for Transplantation, Institute of Liver Studies, King's College London, London, United Kingdom
| | - Elisavet Kodela
- Medical Research Council (MRC) Centre for Transplantation, Institute of Liver Studies, King's College London, London, United Kingdom
| | - Elena Crespo
- Kidney Transplant Unit, Bellvitge University Hospital, Barcelona, Spain
| | - John Mason
- Department of Physiology, Anatomy & Genetics, University of Oxford, United Kingdom
| | - Oriol Bestard
- Kidney Transplant Unit, Bellvitge University Hospital, Barcelona, Spain
| | - Marc Martínez-Llordella
- Medical Research Council (MRC) Centre for Transplantation, Institute of Liver Studies, King's College London, London, United Kingdom
| | - Alberto Sánchez-Fueyo
- Medical Research Council (MRC) Centre for Transplantation, Institute of Liver Studies, King's College London, London, United Kingdom
| |
Collapse
|
34
|
Rodrigues MA, Probst CE, Zayats A, Davidson B, Riedel M, Li Y, Venkatachalam V. The in vitro micronucleus assay using imaging flow cytometry and deep learning. NPJ Syst Biol Appl 2021; 7:20. [PMID: 34006858 PMCID: PMC8131758 DOI: 10.1038/s41540-021-00179-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 02/16/2021] [Indexed: 02/07/2023] Open
Abstract
The in vitro micronucleus (MN) assay is a well-established assay for quantification of DNA damage, and is required by regulatory bodies worldwide to screen chemicals for genetic toxicity. The MN assay is performed in two variations: scoring MN in cytokinesis-blocked binucleated cells or directly in unblocked mononucleated cells. Several methods have been developed to score the MN assay, including manual and automated microscopy, and conventional flow cytometry, each with advantages and limitations. Previously, we applied imaging flow cytometry (IFC) using the ImageStream® to develop a rapid and automated MN assay based on high throughput image capture and feature-based image analysis in the IDEAS® software. However, the analysis strategy required rigorous optimization across chemicals and cell lines. To overcome the complexity and rigidity of feature-based image analysis, in this study we used the Amnis® AI software to develop a deep-learning method based on convolutional neural networks to score IFC data in both the cytokinesis-blocked and unblocked versions of the MN assay. We show that the use of the Amnis AI software to score imagery acquired using the ImageStream® compares well to manual microscopy and outperforms IDEAS® feature-based analysis, facilitating full automation of the MN assay.
Collapse
Affiliation(s)
| | | | - Artiom Zayats
- Amnis Flow Cytometry, Luminex Corporation, Seattle, WA, USA
| | - Bryan Davidson
- Amnis Flow Cytometry, Luminex Corporation, Seattle, WA, USA
| | - Michael Riedel
- Amnis Flow Cytometry, Luminex Corporation, Seattle, WA, USA
| | - Yang Li
- Amnis Flow Cytometry, Luminex Corporation, Seattle, WA, USA
| | | |
Collapse
|
35
|
Liang Y, Lehrich BM, Zheng S, Lu M. Emerging methods in biomarker identification for extracellular vesicle-based liquid biopsy. J Extracell Vesicles 2021; 10:e12090. [PMID: 34012517 PMCID: PMC8114032 DOI: 10.1002/jev2.12090] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 03/17/2021] [Accepted: 04/13/2021] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles (EVs) are released by many cell types and distributed within various biofluids. EVs have a lipid membrane-confined structure that allows for carrying unique molecular information originating from their parent cells. The species and quantity of EV cargo molecules, including nucleic acids, proteins, lipids, and metabolites, may vary largely owing to their parent cell types and the pathophysiologic status. Such heterogeneity in EV populations provides immense challenges to researchers, yet allows for the possibility to prognosticate the pathogenesis of a particular tissue from unique molecular signatures of dispersing EVs within biofluids. However, the inherent nature of EV's small size requires advanced methods for EV purification and evaluation from the complex biofluid. Recently, the interdisciplinary significance of EV research has attracted growing interests, and the EV analytical platforms for their diagnostic prospect have markedly progressed. This review summarizes the recent advances in these EV detection techniques and methods with the intention of translating an EV-based liquid biopsy into clinical practice. This article aims to present an overview of current EV assessment techniques, with a focus on their progress and limitations, as well as an outlook on the clinical translation of an EV-based liquid biopsy that may augment current paradigms for the diagnosis, prognosis, and monitoring the response to therapy in a variety of disease settings.
Collapse
Affiliation(s)
- Yaxuan Liang
- Center for Biological Science and Technology, Advanced Institute of Natural SciencesBeijing Normal University at ZhuhaiZhuhaiChina
| | - Brandon M. Lehrich
- Medical Scientist Training ProgramUniversity of Pittsburgh School of Medicine and Carnegie Mellon UniversityPittsburghPennsylvaniaUSA
| | - Siyang Zheng
- Department Biomedical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
- Department of Electrical and Computer EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
| | - Mengrou Lu
- Department Biomedical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
| |
Collapse
|
36
|
Sahoo S, Adamiak M, Mathiyalagan P, Kenneweg F, Kafert-Kasting S, Thum T. Therapeutic and Diagnostic Translation of Extracellular Vesicles in Cardiovascular Diseases: Roadmap to the Clinic. Circulation 2021; 143:1426-1449. [PMID: 33819075 PMCID: PMC8021236 DOI: 10.1161/circulationaha.120.049254] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Exosomes are small membrane-bound vesicles of endocytic origin that are actively secreted. The potential of exosomes as effective communicators of biological signaling in myocardial function has previously been investigated, and a recent explosion in exosome research not only underscores their significance in cardiac physiology and pathology, but also draws attention to methodological limitations of studying these extracellular vesicles. In this review, we discuss recent advances and challenges in exosome research with an emphasis on scientific innovations in isolation, identification, and characterization methodologies, and we provide a comprehensive summary of web-based resources available in the field. Importantly, we focus on the biology and function of exosomes, highlighting their fundamental role in cardiovascular pathophysiology to further support potential applications of exosomes as biomarkers and therapeutics for cardiovascular diseases.
Collapse
Affiliation(s)
- Susmita Sahoo
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York (S.S., M.A., P.M.)
| | - Marta Adamiak
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York (S.S., M.A., P.M.)
| | - Prabhu Mathiyalagan
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York (S.S., M.A., P.M.)
| | - Franziska Kenneweg
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS) (F.K., S.K-K., T.T.), Hannover Medical School, Germany
| | - Sabine Kafert-Kasting
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS) (F.K., S.K-K., T.T.), Hannover Medical School, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany (S.K-K., T.T.)
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS) (F.K., S.K-K., T.T.), Hannover Medical School, Germany
- REBIRTH Center for Translational Regenerative Medicine (T.T.), Hannover Medical School, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany (S.K-K., T.T.)
| |
Collapse
|
37
|
Ricci G, Minsker K, Kapish A, Osborn J, Ha S, Davide J, Califano JP, Sehlin D, Rustandi RR, Dick LW, Vlasak J, Culp TD, Baudy A, Bell E, Mukherjee M. Flow virometry for process monitoring of live virus vaccines-lessons learned from ERVEBO. Sci Rep 2021; 11:7432. [PMID: 33795759 PMCID: PMC8016999 DOI: 10.1038/s41598-021-86688-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 03/04/2021] [Indexed: 12/19/2022] Open
Abstract
Direct at line monitoring of live virus particles in commercial manufacturing of vaccines is challenging due to their small size. Detection of malformed or damaged virions with reduced potency is rate-limited by release potency assays with long turnaround times. Thus, preempting batch failures caused by out of specification potency results is almost impossible. Much needed are in-process tools that can monitor and detect compromised viral particles in live-virus vaccines (LVVs) manufacturing based on changes in their biophysical properties to provide timely measures to rectify process stresses leading to such damage. Using ERVEBO, MSD's Ebola virus vaccine as an example, here we describe a flow virometry assay that can quickly detect damaged virus particles and provide mechanistic insight into process parameters contributing to the damage. Furthermore, we describe a 24-h high throughput infectivity assay that can be used to correlate damaged particles directly to loss in viral infectivity (potency) in-process. Collectively, we provide a set of innovative tools to enable rapid process development, process monitoring, and control strategy implementation in large scale LVV manufacturing.
Collapse
Affiliation(s)
- Geoffri Ricci
- Vaccines Process Development and Commercialization, Merck & Co., Inc., 770 Sumneytown Pike, WP 42-3, West Point, PA, 19486, USA
| | - Kevin Minsker
- Vaccines Process Development and Commercialization, Merck & Co., Inc., 770 Sumneytown Pike, WP 42-3, West Point, PA, 19486, USA
| | - Austin Kapish
- Vaccines Process Development and Commercialization, Merck & Co., Inc., 770 Sumneytown Pike, WP 42-3, West Point, PA, 19486, USA
| | - James Osborn
- Vaccines Process Development and Commercialization, Merck & Co., Inc., 770 Sumneytown Pike, WP 42-3, West Point, PA, 19486, USA
| | - Sha Ha
- Vaccines Process Development and Commercialization, Merck & Co., Inc., 770 Sumneytown Pike, WP 42-3, West Point, PA, 19486, USA
| | - Joseph Davide
- Vaccines Process Development and Commercialization, Merck & Co., Inc., 770 Sumneytown Pike, WP 42-3, West Point, PA, 19486, USA
| | - Joseph P Califano
- Vaccines Process Development and Commercialization, Merck & Co., Inc., 770 Sumneytown Pike, WP 42-3, West Point, PA, 19486, USA
| | - Darrell Sehlin
- Vaccines Process Development and Commercialization, Merck & Co., Inc., 770 Sumneytown Pike, WP 42-3, West Point, PA, 19486, USA
| | - Richard R Rustandi
- Vaccines Analytical Research and Development, Merck & Co., Inc., West Point, PA, USA
| | - Lawrence W Dick
- Vaccines Analytical Research and Development, Merck & Co., Inc., West Point, PA, USA
| | - Josef Vlasak
- Vaccines Analytical Research and Development, Merck & Co., Inc., West Point, PA, USA
| | - Timothy D Culp
- Vaccines Process Development, Merck & Co., Inc., West Point, PA, USA
| | - Andreas Baudy
- Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., West Point, PA, USA
| | - Edward Bell
- Vaccines Process Development and Commercialization, Merck & Co., Inc., 770 Sumneytown Pike, WP 42-3, West Point, PA, 19486, USA
| | - Malini Mukherjee
- Vaccines Process Development and Commercialization, Merck & Co., Inc., 770 Sumneytown Pike, WP 42-3, West Point, PA, 19486, USA.
| |
Collapse
|
38
|
Extracellular Vesicles in Allergic Rhinitis and Asthma and Laboratory Possibilities for Their Assessment. Int J Mol Sci 2021; 22:ijms22052273. [PMID: 33668821 PMCID: PMC7956366 DOI: 10.3390/ijms22052273] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 12/12/2022] Open
Abstract
Currently, extracellular vesicles (EVs) have been implicated in the etiopathogenesis of many diseases, including lung disorders, with the possibility of diagnostic and therapeutic applications. The analysis of EV in respiratory tract diseases faces many obstacles, including material collection from airways, standardization of isolation techniques, detection methods, the analysis of their content, etc. This review focuses on the role of extracellular vesicles in the pathogenesis of atopic respiratory diseases, especially asthma, with a special focus on their clinical applicability as a diagnostic tool. We also summarize available laboratory techniques that enable the detection of EVs in various biological materials, with particular emphasis on flow cytometry. The opportunities and limitations of detecting EV in bronchoalveolar lavage fluid (BALF) were also described.
Collapse
|
39
|
Yamamoto M, Robinson JP. Quantum approach for nanoparticle fluorescence by sub-ns photon detection. Cytometry A 2021; 99:145-151. [PMID: 33476076 DOI: 10.1002/cyto.a.24310] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 12/23/2020] [Accepted: 01/10/2021] [Indexed: 11/10/2022]
Abstract
Well defined detection and analysis of nanoparticle-sized samples such as extracellular vesicles or viruses may be important for potential disease diagnostics. However, using conventional flow-cytometry optical methods to evaluate such small particles is quite challenging. The reason is that the particle is smaller than the diffraction limit, making detection difficult. An alternative approach is fluorescence detection via conjugated fluorochromes attached to the nanoparticles; the challenge in this case is the limitation imposed upon detection of a very small number of emitted photons buried in high background photon counts. Emitted fluorescence is described by the well-known equation kf = σa I Q, which describes the emitted fluorescence rate (kf) (photons/s) as the multiplication of molecular absorption cross section(σa), excitation intensity (I), and quantum yield (Q). In addition, the excitation rate is equal to 1/t, which is the inverse of the lifetime of several ns representing the most typical conjugated fluorescent molecules used in flow cytometry. We recently developed a sub-ns photon sensor that is faster than most fluorescence lifetimes, since sub-ns speed is a critically important parameter for the separation of individual emitted photons. Based on our observation of fluorescence and background levels on typical commercial flow cytometers it is evident that a significant component of the background is induced by water-molecular vibrations. Therefore, understanding what constitutes all the components that contribute to the signals we measure in flow cytometry would help in defining what we currently call "background signals." We attempted to define a theoretical model to try to unravel these issues. This model was based on use of a reflective dry surface in the absence of water molecules. Our objective was to determine if it is possible to minimize background and enhance signal, and to provide valuable information on the contributing components of the signals collected. In order to test this model, we tested a single dried particle 50 nm in diameter on a reflective surface with minimum background. While this is clearly not a standard biological system, our results suggest that this quantum approach closely follows established photon base theory. Our goal was to define the parameters for practical nanoparticle-fluorescence analysis while enhancing our knowledge of the contribution of background properties.
Collapse
Affiliation(s)
- Masanobu Yamamoto
- Miftek Corporation, West Lafayette, Indiana, USA.,Basic Medical Sciences, Purdue University, West Lafayette, Indiana, USA
| | - J Paul Robinson
- Miftek Corporation, West Lafayette, Indiana, USA.,Basic Medical Sciences, Purdue University, West Lafayette, Indiana, USA.,Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
40
|
Botha J, Pugsley HR, Handberg A. Conventional, High-Resolution and Imaging Flow Cytometry: Benchmarking Performance in Characterisation of Extracellular Vesicles. Biomedicines 2021; 9:biomedicines9020124. [PMID: 33513846 PMCID: PMC7911094 DOI: 10.3390/biomedicines9020124] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/20/2021] [Accepted: 01/25/2021] [Indexed: 12/14/2022] Open
Abstract
Flow cytometry remains a commonly used methodology due to its ability to characterise multiple parameters on single particles in a high-throughput manner. In order to address limitations with lacking sensitivity of conventional flow cytometry to characterise extracellular vesicles (EVs), novel, highly sensitive platforms, such as high-resolution and imaging flow cytometers, have been developed. We provided comparative benchmarks of a conventional FACS Aria III, a high-resolution Apogee A60 Micro-PLUS and the ImageStream X Mk II imaging flow cytometry platform. Nanospheres were used to systematically characterise the abilities of each platform to detect and quantify populations with different sizes, refractive indices and fluorescence properties, and the repeatability in concentration determinations was reported for each population. We evaluated the ability of the three platforms to detect different EV phenotypes in blood plasma and the intra-day, inter-day and global variabilities in determining EV concentrations. By applying this or similar methodology to characterise methods, researchers would be able to make informed decisions on choice of platforms and thereby be able to match suitable flow cytometry platforms with projects based on the needs of each individual project. This would greatly contribute to improving the robustness and reproducibility of EV studies.
Collapse
Affiliation(s)
- Jaco Botha
- Department of Clinical Biochemistry, Aalborg University Hospital, North Denmark Region, DK-9000 Aalborg, Denmark;
- Department of Clinical Medicine, Aalborg University, DK-9000 Aalborg, Denmark
- Correspondence:
| | | | - Aase Handberg
- Department of Clinical Biochemistry, Aalborg University Hospital, North Denmark Region, DK-9000 Aalborg, Denmark;
- Department of Clinical Medicine, Aalborg University, DK-9000 Aalborg, Denmark
| |
Collapse
|
41
|
Brahmer A, Neuberger EWI, Simon P, Krämer-Albers EM. Considerations for the Analysis of Small Extracellular Vesicles in Physical Exercise. Front Physiol 2020; 11:576150. [PMID: 33343383 PMCID: PMC7744614 DOI: 10.3389/fphys.2020.576150] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 11/18/2020] [Indexed: 12/16/2022] Open
Abstract
Physical exercise induces acute physiological changes leading to enhanced tissue cross-talk and a liberation of extracellular vesicles (EVs) into the circulation. EVs are cell-derived membranous entities which carry bioactive material, such as proteins and RNA species, and are important mediators of cell-cell-communication. Different types of physical exercise interventions trigger the release of diverse EV subpopulations, which are hypothesized to be involved in physiological adaptation processes leading to health benefits and longevity. Large EVs (“microvesicles” and “microparticles”) are studied frequently in the context of physical exercise using straight forward flow cytometry approaches. However, the analysis of small EVs (sEVs) including exosomes is hampered by the complex composition of blood, confounding the methodology of EV isolation and characterization. This mini review presents a concise overview of the current state of research on sEVs released upon physical exercise (ExerVs), highlighting the technical limits of ExerV analysis. The purity of EV preparations is highly influenced by the co-isolation of non-EV structures in the size range or density of EVs, such as lipoproteins and protein aggregates. Technical constraints associated with EV purification challenge the quantification of distinct ExerV populations, the identification of their cargo, and the investigation of their biological functions. Here, we offer recommendations for the isolation and characterization of ExerVs to minimize the effects of these drawbacks. Technological advances in the ExerV research field will improve understanding of the inter-cellular cross-talk induced by physical exercise leading to health benefits.
Collapse
Affiliation(s)
- Alexandra Brahmer
- Extracellular Vesicles Research Group, Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University of Mainz, Mainz, Germany.,Department of Sports Medicine, Rehabilitation and Disease Prevention, Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Elmo W I Neuberger
- Department of Sports Medicine, Rehabilitation and Disease Prevention, Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Perikles Simon
- Department of Sports Medicine, Rehabilitation and Disease Prevention, Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Eva-Maria Krämer-Albers
- Extracellular Vesicles Research Group, Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University of Mainz, Mainz, Germany
| |
Collapse
|
42
|
Mastoridis S, Martinez-Llordella M, Sanchez-Fueyo A. Extracellular vesicles as mediators of alloimmunity and their therapeutic potential in liver transplantation. World J Transplant 2020; 10:330-344. [PMID: 33312894 PMCID: PMC7708876 DOI: 10.5500/wjt.v10.i11.330] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/06/2020] [Accepted: 11/17/2020] [Indexed: 02/05/2023] Open
Abstract
Extracellular vesicles (EVs) are a heterogenous group of nanosized, membrane-bound particles which are released by most cell types. They are known to play an essential role in cellular communication by way of their varied cargo which includes selectively enriched proteins, lipids, and nucleic acids. In the last two decades, wide-ranging evidence has established the involvement of EVs in the regulation of immunity, with EVs released by immune and non-immune cells shown to be capable of mediating immune stimulation or suppression and to drive inflammatory, autoimmune, and infectious disease pathology. More recently, studies have demonstrated the involvement of allograft-derived EVs in alloimmune responses following transplantation, with EVs shown to be capable of eliciting allograft rejection as well as promoting tolerance. These insights are necessitating the reassessment of standard paradigms of T cell alloimmunity. In this article, we explore the latest understanding of the impact of EVs on alloresponses following transplantation and we highlight the recent technological advances which have enabled the study of EVs in clinical transplantation. Furthermore, we discuss the rapid progress afoot in the development of EVs as novel therapeutic vehicles in clinical transplantation with particular focus on liver transplantation.
Collapse
Affiliation(s)
- Sotiris Mastoridis
- Department ofSurgery, Oxford University Hospitals, Oxford OX37LE, United Kingdom
| | - Marc Martinez-Llordella
- Institute of Liver Studies, King's College Hospital, Medical Research Council (MRC) Centre for Transplantation, London SE59NU, United Kingdom
| | - Alberto Sanchez-Fueyo
- Department of Liver Sciences, King's College Hospital, Medical Research Council (MRC) Centre for Transplantation, London SE59NU, United Kingdom
| |
Collapse
|
43
|
Large Extracellular Vesicles Can be Characterised by Multiplex Labelling Using Imaging Flow Cytometry. Int J Mol Sci 2020; 21:ijms21228723. [PMID: 33218198 PMCID: PMC7699300 DOI: 10.3390/ijms21228723] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 11/13/2020] [Accepted: 11/16/2020] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles (EVs) are heterogeneous in size (30 nm–10 µm), content (lipid, RNA, DNA, protein), and potential function(s). Many isolation techniques routinely discard the large EVs at the early stages of small EV or exosome isolation protocols. We describe here a standardised method to isolate large EVs from medulloblastoma cells and examine EV marker expression and diameter using imaging flow cytometry. Our approach permits the characterisation of each large EVs as an individual event, decorated with multiple fluorescently conjugated markers with the added advantage of visualising each event to ensure robust gating strategies are applied. Methods: We describe step-wise isolation and characterisation of a subset of large EVs from the medulloblastoma cell line UW228-2 assessed by fluorescent light microscopy, transmission electron microscopy (TEM) and tunable resistance pulse sensing (TRPS). Viability of parent cells was assessed by Annexin V exposure by flow cytometry. Imaging flow cytometry (Imagestream Mark II) identified EVs by direct fluorescent membrane labelling with Cell Mask Orange (CMO) in conjunction with EV markers. A stringent gating algorithm based on side scatter and fluorescence intensity was applied and expression of EV markers CD63, CD9 and LAMP 1 assessed. Results: UW228-2 cells prolifically release EVs of up to 6 µm. We show that the Imagestream Mark II imaging flow cytometer allows robust and reproducible analysis of large EVs, including assessment of diameter. We also demonstrate a correlation between increasing EV size and co-expression of markers screened. Conclusions: We have developed a labelling and stringent gating strategy which is able to explore EV marker expression (CD63, CD9, and LAMP1) on individual EVs within a widely heterogeneous population. Taken together, data presented here strongly support the value of exploring large EVs in clinical samples for potential biomarkers, useful in diagnostic screening and disease monitoring.
Collapse
|
44
|
La Salvia S, Musante L, Lannigan J, Gigliotti JC, Le TH, Erdbrügger U. T cell-derived extracellular vesicles are elevated in essential HTN. Am J Physiol Renal Physiol 2020; 319:F868-F875. [PMID: 33017187 PMCID: PMC7789979 DOI: 10.1152/ajprenal.00433.2020] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/23/2020] [Accepted: 10/01/2020] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles (EVs) are novel mediators of cell-to-cell communication and appear to mediate the pathogenesis of hypertension (HTN). However, the mechanisms underlying the involvement of EVs in HTN remain unclear. The adaptive and innate immune systems play an important role affecting the kidney and vasculature in animal models of HTN. Evolving evidence shows that immune cell-derived EVs can modulate the immune system in a paracrine fashion and therefore may mediate the effects of inflammation in the pathogenesis of HTN. Therefore, we aimed to understand if specific subtypes of leukocyte/immune cell-derived EVs are altered in essential HTN using an in vivo model of angiotensin II (ANG II)-induced HTN. After 4 wk of ANG II treatment, EVs were isolated from the blood and kidney. EV origin and counts were characterized with Imaging Flow Cytometry, antibody panels targeting platelets, endothelial cells, and leukocytes including B and T cells, monocytes, and neutrophils. Leukocyte-derived EVs (CD45+) were elevated in the circulation and kidney tissue in ANG II-induced HTN. Subgroup analysis depicted T cell-derived EVs (CD3+) to be significantly elevated in ANG II-induced HTN (3.50e+5 particles/mL) compared with control groups (9.16e+4 particles/mL, P = 0.0106). T cell-derived EVs also significantly correlated with systolic blood pressure levels (r2 = 0.898, P = 0.0012). In summary, leukocyte-derived EVs, and more specifically T cell-derived EVs (CD3+), are elevated in ANG II-induced HTN in the circulation and kidney tissue and correlate well with blood pressure severity. EVs from the circulation and kidney may be sensitive biomarkers for HTN and end-organ damage and may lead to new mechanistic insights in this silent disease.
Collapse
Affiliation(s)
- Sabrina La Salvia
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia
| | - Luca Musante
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia
| | - Joanne Lannigan
- School of Medicine, Flow Cytometry Core, University of Virginia, Charlottesville, Virginia
| | - Joseph Christopher Gigliotti
- Department of Integrated Physiology and Pharmacology, Liberty University College of Osteopathic Medicine, Lynchburg, Virginia
| | - Thu H Le
- Division of Nephrology, Department of Medicine, University of Rochester Medical Center, Rochester, New York
| | - Uta Erdbrügger
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
45
|
Assessment of extracellular vesicles using IFC for application in transfusion medicine. Transfus Apher Sci 2020; 59:102942. [PMID: 32943325 DOI: 10.1016/j.transci.2020.102942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Extracellular vesicles (EVs) have been shown to be involved in various physiological and pathophysiological processes. With respect to Transfusion Medicine, the accumulation of EVs in blood products during hypothermic storage is an indicator of the storage lesion and reportedly correlates with adverse effects after transfusion, including but not limited to immunomodulation, activation of coagulation, endothelial activation, and others. To optimally reduce such an impact on blood product quality degradation and improve post-transfusion outcomes, better methods for detection, enumeration, characterisation by size and phenotype, and functional involvement of EVs in different pathophysiological and physiological processes are required. Currently, Imaging Flow Cytometry (IFC) technology provides the most comprehensive assessment of EV subsets in different body fluids. The unique ability of IFC to detect EVs of 20 nm size by registration of a single pixel of fluorescence signal makes this approach highly promising for comprehensive studies of EVs. In this review, we will focus on the recent breakthrough and advantages of using the ImageStreamX MKII IFC platform for the detection and characterisation of EVs and its future prospects for routine application of IFC in Transfusion Medicine.
Collapse
|
46
|
La Salvia S, Gunasekaran PM, Byrd JB, Erdbrügger U. Extracellular Vesicles in Essential Hypertension: Hidden Messengers. Curr Hypertens Rep 2020; 22:76. [PMID: 32880744 DOI: 10.1007/s11906-020-01084-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW Hypertension affects about half of all Americans, yet in the vast majority of cases, the factors causing the hypertension cannot be clearly delineated. Developing a more precise understanding of the molecular pathogenesis of HTN and its various phenotypes is therefore a pressing priority. Circulating and urinary extracellular vesicles (EVs) are potential novel candidates as biomarkers and bioactivators in HTN. EVs are a heterogeneous population of small membrane fragments shed from various cell types into various body fluids. As EVs carry protein, RNA, and lipids, they also play a role as effectors and novel cell-to-cell communicators. In this review, we discuss the diagnostic, functional, and regenerative role of EVs in essential HTN and focus on EV protein and RNA cargo as the most extensively studied EV cargo. RECENT FINDINGS The field of EVs in HTN is still a young one and earlier studies have not used the novel EV detection tools currently available. More rigor and transparency in EV research are needed. Current data suggest that EVs represent potential novel biomarkers in HTN. EVs correlate with HTN severity and possibly end-organ damage. However, it has yet to be discerned which specific subtype(s) of EV reflects best HTN pathophysiology. Evolving studies are also showing that EVs might be novel regulators in vascular and renal tubular function and also be therapeutic. RNA in EVs has been studied in the context of hypertension, largely in the form of studies of miRNA, which are reviewed herein. Beyond miRNAs, mRNA in urinary EVs changed in response to sodium loading in humans. EVs represent promising novel biomarkers and bioactivators in essential HTN. Novel tools are being developed to apply more rigor in EV research including more in vivo models and translation to humans.
Collapse
Affiliation(s)
- Sabrina La Salvia
- Department of Internal Medicine, Division of Nephrology, University of Virginia Health System, 1300 Jefferson Park Avenue, Charlottesville, VA, 22908-0133, USA.
| | - Pradeep Moon Gunasekaran
- Department of Internal Medicine, Division of Cardiovascular Medicine, Medical School, University of Michigan Medical School, 5570C MSRB II, 1150 W. Medical Center Dr, Ann Arbor, MI, 48109, USA
| | - James Brian Byrd
- Department of Internal Medicine, Division of Cardiovascular Medicine, Medical School, University of Michigan Medical School, 5570C MSRB II, 1150 W. Medical Center Dr, Ann Arbor, MI, 48109, USA
| | - Uta Erdbrügger
- Department of Internal Medicine, Division of Nephrology, University of Virginia Health System, 1300 Jefferson Park Avenue, Charlottesville, VA, 22908-0133, USA
| |
Collapse
|
47
|
Morani M, Mai TD, Krupova Z, Defrenaix P, Multia E, Riekkola ML, Taverna M. Electrokinetic characterization of extracellular vesicles with capillary electrophoresis: A new tool for their identification and quantification. Anal Chim Acta 2020; 1128:42-51. [DOI: 10.1016/j.aca.2020.06.073] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 06/29/2020] [Indexed: 01/08/2023]
|
48
|
Jurgielewicz BJ, Yao Y, Stice SL. Kinetics and Specificity of HEK293T Extracellular Vesicle Uptake using Imaging Flow Cytometry. NANOSCALE RESEARCH LETTERS 2020; 15:170. [PMID: 32833066 PMCID: PMC7445225 DOI: 10.1186/s11671-020-03399-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 08/11/2020] [Indexed: 05/31/2023]
Abstract
Extracellular vesicles (EVs) are nanosized lipid bilayer-bound vesicles that are naturally secreted from most cell types as a communication mechanism to deliver proteins, lipids, and genetic material. Despite the therapeutic potential of EVs, there is limited information on EV uptake kinetics and specificity. Here, we optimized an imaging flow cytometry (IFC)-based platform to quantitatively assess dose, time, and recipient cell specificity effects on human embryonic kidney cell (HEK293T) EV internalization in a high-throughput manner. We found that HEK293T EV uptake is an active process that is dose and time dependent. Further, the selectivity of EV uptake was quantified in vitro, and we found that HEK293T EVs were internalized at higher quantities by cells of the same origin. Lastly, neural stem cells internalized significantly more HEK293T EVs relative to mature neurons, suggesting that stem cells or progenitors, which are more metabolically active than terminally differentiated cells, may have higher rates of active EV internalization. The characterization of EV uptake, notably specificity, dose and time dependence, and kinetic assays will help inform and develop targeted and efficient EV-based therapeutics.
Collapse
Affiliation(s)
- Brian J Jurgielewicz
- Regenerative Bioscience Center, University of Georgia, Athens, GA, 30602, USA
- Department of Animal and Dairy Science, University of Georgia, Athens, GA, 30602, USA
| | - Yao Yao
- Regenerative Bioscience Center, University of Georgia, Athens, GA, 30602, USA.
- Department of Animal and Dairy Science, University of Georgia, Athens, GA, 30602, USA.
| | - Steven L Stice
- Regenerative Bioscience Center, University of Georgia, Athens, GA, 30602, USA.
- Department of Animal and Dairy Science, University of Georgia, Athens, GA, 30602, USA.
- ArunA Bio, Athens, GA, 30602, USA.
| |
Collapse
|
49
|
Shen M, Di K, He H, Xia Y, Xie H, Huang R, Liu C, Yang M, Zheng S, He N, Li Z. Progress in exosome associated tumor markers and their detection methods. MOLECULAR BIOMEDICINE 2020; 1:3. [PMID: 35006428 PMCID: PMC8603992 DOI: 10.1186/s43556-020-00002-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 07/15/2020] [Indexed: 02/08/2023] Open
Abstract
Exosomes are secreted by cells and are widely present in body fluids. Exosomes contain various molecular constituents of their cells of origin such as proteins, mRNA, miRNAs, DNA, lipid and glycans which are very similar as the content in tumor cells. These contents play an important role in various stages of tumor development, and make the tumor-derived exosome as a hot and emerging biomarker for various cancers diagnosis and management in non-invasive manner. The present problems of exosome isolation and detection hinder the application of exosomes. With the development of exosome isolation and detection technology, the contents of exosomes can be exploited for early cancer diagnosis. This review summarizes the recent progress on exosome-associated tumor biomarkers and some new technologies for exosome isolation and detection. Furthermore, we have also discussed the future development direction in exosome analysis methods.
Collapse
Affiliation(s)
- Mengjiao Shen
- Department of Clinical Laboratory, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
- Shanghai Health Development Research Center, Shanghai, China
| | - Kaili Di
- Department of Clinical Laboratory, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Hongzhang He
- Captis Diagnostics Inc, Pittsburgh, PA, 15213, USA
| | - Yanyan Xia
- Department of Clinical Laboratory, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Hui Xie
- Department of Clinical Laboratory, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Rongrong Huang
- Department of Clinical Laboratory, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Chang Liu
- Department of Clinical Laboratory, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Mo Yang
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hunghom, Kowloon, Hong Kong, People's Republic of China.
| | - Siyang Zheng
- Department of Biomedical Engineering and Electrical & Computer Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Scott Hall 4N211, Pittsburgh, PA, 15213, USA.
| | - Nongyue He
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| | - Zhiyang Li
- Department of Clinical Laboratory, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China.
| |
Collapse
|
50
|
Dehghani M, Gulvin SM, Flax J, Gaborski TR. Systematic Evaluation of PKH Labelling on Extracellular Vesicle Size by Nanoparticle Tracking Analysis. Sci Rep 2020; 10:9533. [PMID: 32533028 PMCID: PMC7293335 DOI: 10.1038/s41598-020-66434-7] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 05/19/2020] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EVs) are membrane vesicles secreted by cells and can modulate biological activities by transferring their content following uptake into recipient cells. Labelling of EVs is a commonly used technique for understanding their cellular targeting and biodistribution. A reliable fluorescent technique needs to preserve the size of EVs since changes in size may alter their uptake and biodistribution. Lipophilic fluorescent dye molecules such as the PKH family have been widely used for EV labelling. Here, the effect of PKH labelling on the size of EVs was systematically evaluated using nanoparticle tracking analysis (NTA), which is a widely used technique for determining the size and concentration of nanoparticles. NTA analysis showed a size increase in all the PKH labelling conditions tested. As opposed to lipophilic dye molecules, no significant shift in the size of labelled EVs was detected with luminal binding dye molecules such as 5-(and-6)-carboxyfluorescein diacetate succinimidyl ester (CFDA-SE, hereinafter CFSE). This finding suggests that PKH labelling may not be a reliable technique for the tracking of EVs.
Collapse
Affiliation(s)
- Mehdi Dehghani
- Department of Microsystems Engineering, Rochester Institute of Technology, Rochester, NY, United States
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, United States
| | - Shannon M Gulvin
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, United States
| | - Jonathan Flax
- Department of Urology, University of Rochester Medical Center, Rochester, NY, United States
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Thomas R Gaborski
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, United States.
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States.
| |
Collapse
|