451
|
Gerlach JQ, Maguire CM, Krüger A, Joshi L, Prina-Mello A, Griffin MD. Urinary nanovesicles captured by lectins or antibodies demonstrate variations in size and surface glycosylation profile. Nanomedicine (Lond) 2017; 12:1217-1229. [PMID: 28520506 DOI: 10.2217/nnm-2017-0016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
AIM The use of carbohydrate-binding proteins (lectins) to isolate urinary extracellular vesicles (uEVs) was investigated and the captured subpopulations were characterized. METHODS Pooled uEVs from multiple healthy donors were exposed to lectin-conjugated or antibody-conjugated beads. Recovered uEVs were evaluated by protein estimation, transmission electron microscopy, nanoparticle tracking analysis and lectin microarray profiling. RESULTS uEVs isolated by lectin- and antibody-based affinity capture exhibited distinct variations in size and surface content. Transmission electron microscopy confirmed similar EV diameters to those established by nanoparticle tracking analysis, but total particle counts did not correlate closely with protein-based quantification. Lectin microarray profiling demonstrated capture-dependent differences in surface glycosylation. CONCLUSION Selective, carbohydrate-mediated EV isolation by lectin affinity approaches may prove immediately useful for research and find eventual use in clinical applications.
Collapse
Affiliation(s)
- Jared Q Gerlach
- Advanced Glycoscience Research Cluster (AGRC), National Centre for Biomedical Engineering Science (NCBES), National University of Ireland, Galway, Ireland
| | - Ciaran M Maguire
- Laboratory for Biological Characterization of Advanced Materials (LBCAM), School of Medicine, Trinity College Dublin, Ireland
| | - Anja Krüger
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, Ireland
| | - Lokesh Joshi
- Advanced Glycoscience Research Cluster (AGRC), National Centre for Biomedical Engineering Science (NCBES), National University of Ireland, Galway, Ireland
| | - Adriele Prina-Mello
- Laboratory for Biological Characterization of Advanced Materials (LBCAM), School of Medicine, Trinity College Dublin, Ireland
| | - Matthew D Griffin
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, Ireland.,School of Medicine, College of Medicine, Nursing & Health Sciences, National University of Ireland Galway, Ireland
| |
Collapse
|
452
|
Carney RP, Hazari S, Rojalin T, Knudson A, Gao T, Tang Y, Liu R, Viitala T, Yliperttula M, Lam KS. Targeting Tumor-Associated Exosomes with Integrin-Binding Peptides. ADVANCED BIOSYSTEMS 2017; 1:1600038. [PMID: 29911169 PMCID: PMC6001286 DOI: 10.1002/adbi.201600038] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
All cells expel a variety of nano-sized extracellular vesicles (EVs), including exosomes, with composition reflecting the cells' biological state. Cancer pathology is dramatically mediated by EV trafficking via key proteins, lipids, metabolites, and microRNAs. Recent proteomics evidence suggests that tumor-associated exosomes exhibit distinct expression of certain membrane proteins, rendering those proteins as attractive targets for diagnostic or therapeutic application. Yet, it is not currently feasible to distinguish circulating EVs in complex biofluids according to their tissue of origin or state of disease. Here we demonstrate peptide binding to tumor-associated EVs via overexpressed membrane protein. We find that SKOV-3 ovarian tumor cells and their released EVs express α3β1 integrin, which can be targeted by our in-house cyclic nonapeptide, LXY30. After measuring bulk SKOV-3 EV association with LXY30 by flow cytometry, Raman spectral analysis of laser-trapped single exosomes with LXY30-dialkyne conjugate enabled us to differentiate cancer-associated exosomes from non-cancer exosomes. Furthermore, we introduce the foundation for a highly specific detection platform for tumor-EVs in solution with biosensor surface-immobilized LXY30. LXY30 not only exhibits high specificity and affinity to α3β1 integrin-expressing EVs, but also reduces EV uptake into SKOV-3 parent cells, demonstrating the possibility for therapeutic application.
Collapse
Affiliation(s)
- Randy P Carney
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Sidhartha Hazari
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Tatu Rojalin
- Division of Pharmaceutical Biosciences, Centre for Drug Research, University of Helsinki, Helsinki, Finland. Center for Biophotonics, University of California Davis, Sacramento, CA, 95817 USA
| | - Alisha Knudson
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Tingjuan Gao
- College of Chemistry, Central China Normal University, Wuhan 430079, China. Center for Biophotonics, University of California Davis, Sacramento, CA, 95817 USA
| | - Yuchen Tang
- College of Chemistry, Central China Normal University, Wuhan 430079, China. Center for Biophotonics, University of California Davis, Sacramento, CA, 95817 USA
| | - Ruiwu Liu
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Tapani Viitala
- Division of Pharmaceutical Biosciences, Centre for Drug Research, University of Helsinki, Helsinki, Finland
| | - Marjo Yliperttula
- Division of Pharmaceutical Biosciences, Centre for Drug Research, University of Helsinki, Helsinki, Finland. Department of Pharmaceutical Sciences, University of Padova, Italy
| | - Kit S Lam
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA 95817, USA. University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA
| |
Collapse
|
453
|
Carney RP, Hazari S, Colquhoun M, Tran D, Hwang B, Mulligan MS, Bryers JD, Girda E, Leiserowitz GS, Smith ZJ, Lam KS. Multispectral Optical Tweezers for Biochemical Fingerprinting of CD9-Positive Exosome Subpopulations. Anal Chem 2017; 89:5357-5363. [PMID: 28345878 DOI: 10.1021/acs.analchem.7b00017] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Extracellular vesicles (EVs), including exosomes, are circulating nanoscale particles heavily implicated in cell signaling and can be isolated in vast numbers from human biofluids. Study of their molecular profiling and materials properties is currently underway for purposes of describing a variety of biological functions and diseases. However, the large, and as yet largely unquantified, variety of EV subpopulations differing in composition, size, and likely function necessitates characterization schemes capable of measuring single vesicles. Here we describe the first application of multispectral optical tweezers (MS-OTs) to single vesicles for molecular fingerprinting of EV subpopulations. This versatile imaging platform allows for sensitive measurement of Raman chemical composition (e.g., variation in protein, lipid, cholesterol, nucleic acids), coupled with discrimination by fluorescence markers. For exosomes isolated by ultracentrifugation, we use MS-OTs to interrogate the CD9-positive subpopulations via antibody fluorescence labeling and Raman spectra measurement. We report that the CD9-positive exosome subset exhibits reduced component concentration per vesicle and reduced chemical heterogeneity compared to the total purified EV population. We observed that specific vesicle subpopulations are present across exosomes isolated from cell culture supernatant of several clonal varieties of mesenchymal stromal cells and also from plasma and ascites isolated from human ovarian cancer patients.
Collapse
Affiliation(s)
- Randy P Carney
- Department of Biochemistry and Molecular Medicine, University of California Davis , Sacramento, California 95817, United States
| | - Sidhartha Hazari
- Department of Biochemistry and Molecular Medicine, University of California Davis , Sacramento, California 95817, United States
| | - Macalistair Colquhoun
- Department of Biochemistry and Molecular Medicine, University of California Davis , Sacramento, California 95817, United States
| | - Di Tran
- Department of Biochemistry and Molecular Medicine, University of California Davis , Sacramento, California 95817, United States
| | - Billanna Hwang
- Department of Surgery, University of Washington , 850 Republican Street, Seattle, Washington 98195-0005, United States
| | - Michael S Mulligan
- Department of Surgery, University of Washington , 850 Republican Street, Seattle, Washington 98195-0005, United States
| | - James D Bryers
- Department of Bioengineering, University of Washington , 3720 15th Avenue NE, Seattle, Washington 98195, United States
| | - Eugenia Girda
- Division of Gynecologic Oncology, University of California Davis Medical Center , Sacramento, California 98517, United States
| | - Gary S Leiserowitz
- Division of Gynecologic Oncology, University of California Davis Medical Center , Sacramento, California 98517, United States
| | - Zachary J Smith
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China , Hefei, Anhui 230027 China
| | - Kit S Lam
- Department of Biochemistry and Molecular Medicine, University of California Davis , Sacramento, California 95817, United States.,Division of Hematology/Oncology, University of California Davis Cancer Center , Sacramento, California 95817, United States
| |
Collapse
|
454
|
Piffoux M, Silva AKA, Lugagne JB, Hersen P, Wilhelm C, Gazeau F. Extracellular Vesicle Production Loaded with Nanoparticles and Drugs in a Trade-off between Loading, Yield and Purity: Towards a Personalized Drug Delivery System. ACTA ACUST UNITED AC 2017; 1:e1700044. [DOI: 10.1002/adbi.201700044] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Indexed: 12/17/2022]
Affiliation(s)
- Max Piffoux
- Laboratoire Matière et Systèmes Complexes; UMR 7057; CNRS and Université Paris Diderot; 10 rue Alice Domon et Léonie Duquet 75205 Paris Cedex 13 France
| | - Amanda K. A. Silva
- Laboratoire Matière et Systèmes Complexes; UMR 7057; CNRS and Université Paris Diderot; 10 rue Alice Domon et Léonie Duquet 75205 Paris Cedex 13 France
| | - Jean-Baptiste Lugagne
- Laboratoire Matière et Systèmes Complexes; UMR 7057; CNRS and Université Paris Diderot; 10 rue Alice Domon et Léonie Duquet 75205 Paris Cedex 13 France
| | - Pascal Hersen
- Laboratoire Matière et Systèmes Complexes; UMR 7057; CNRS and Université Paris Diderot; 10 rue Alice Domon et Léonie Duquet 75205 Paris Cedex 13 France
| | - Claire Wilhelm
- Laboratoire Matière et Systèmes Complexes; UMR 7057; CNRS and Université Paris Diderot; 10 rue Alice Domon et Léonie Duquet 75205 Paris Cedex 13 France
| | - Florence Gazeau
- Laboratoire Matière et Systèmes Complexes; UMR 7057; CNRS and Université Paris Diderot; 10 rue Alice Domon et Léonie Duquet 75205 Paris Cedex 13 France
| |
Collapse
|
455
|
Cheruiyot C, Pataki Z, Williams R, Ramratnam B, Li M. SILAC Based Proteomic Characterization of Exosomes from HIV-1 Infected Cells. J Vis Exp 2017. [PMID: 28287540 DOI: 10.3791/54799] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Proteomics is the large-scale analysis of proteins. Proteomic techniques, such as liquid chromatography tandem mass spectroscopy (LC-MS/MS), can characterize thousands of proteins at a time. These powerful techniques allow us to have a systemic understanding of cellular changes, especially when cells are subjected to various stimuli, such as infections, stresses, and specific test conditions. Even with recent developments, analyzing the exosomal proteome is time-consuming and often involves complex methodologies. In addition, the resultant large dataset often needs robust and streamlined analysis in order for researchers to perform further downstream studies. Here, we describe a SILAC-based protocol for characterizing the exosomal proteome when cells are infected with HIV-1. The method is based on simple isotope labeling, isolation of exosomes from differentially labeled cells, and mass spectrometry analysis. This is followed by detailed data mining and bioinformatics analysis of the proteomic hits. The resultant datasets and candidates are easy to understand and often offer a wealth of information that is useful for downstream analysis. This protocol is applicable to other subcellular compartments and a wide range of test conditions.
Collapse
Affiliation(s)
| | | | | | - Bharat Ramratnam
- COBRE Center for Cancer Research, Lifespan Laboratories, Rhode Island and Miriam Hospitals; Division of Infectious Diseases, Department of Medicine, Warren Alpert Medical School, Brown University
| | - Ming Li
- Division of Infectious Diseases, Department of Medicine, Warren Alpert Medical School, Brown University;
| |
Collapse
|
456
|
Koritzinsky EH, Street JM, Star RA, Yuen PST. Quantification of Exosomes. J Cell Physiol 2017; 232:1587-1590. [PMID: 27018079 DOI: 10.1002/jcp.25387] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 03/23/2016] [Indexed: 12/11/2022]
Abstract
Exosomes are released by cells as self-contained vesicles with an intact lipid bilayer that encapsulates a small portion of the parent cell. Exosomes have been studied widely as information-rich sources of potential biomarkers that can reveal cellular physiology. We suggest that quantification is essential to understand basic biological relationships between exosomes and their parent cells and hence the underlying interpretation of exosome signals. The number of methods for quantifying exosomes has expanded as interest in exosomes has increased. However, a consensus on proper quantification has not developed, making each study difficult to compare to another. Overcoming this ad hoc approach will require widely available standards that have been adequately characterized, and multiple comparative studies across platforms. We outline the current status of these technical approaches and our view of how they can become more coherent. J. Cell. Physiol. 232: 1587-1590, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - Jonathan M Street
- Renal Diagnostics and Therapeutics Unit, NIDDK, NIH, Bethesda, Maryland
| | - Robert A Star
- Renal Diagnostics and Therapeutics Unit, NIDDK, NIH, Bethesda, Maryland
| | - Peter S T Yuen
- Renal Diagnostics and Therapeutics Unit, NIDDK, NIH, Bethesda, Maryland
| |
Collapse
|
457
|
Kang H, Kim J, Park J. Methods to isolate extracellular vesicles for diagnosis. MICRO AND NANO SYSTEMS LETTERS 2017. [DOI: 10.1186/s40486-017-0049-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
458
|
A Comparative Study of Serum Exosome Isolation Using Differential Ultracentrifugation and Three Commercial Reagents. PLoS One 2017; 12:e0170628. [PMID: 28114422 PMCID: PMC5256994 DOI: 10.1371/journal.pone.0170628] [Citation(s) in RCA: 424] [Impact Index Per Article: 60.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 01/06/2017] [Indexed: 12/21/2022] Open
Abstract
Exosomes play a role in cell-to-cell signaling and serve as possible biomarkers. Isolating exosomes with reliable quality and substantial concentration is a major challenge. Our purpose is to compare the exosomes extracted by three different exosome isolation kits (miRCURY, ExoQuick, and Invitrogen Total Exosome Isolation Reagent) and differential ultracentrifugation (UC) using six different volumes of a non-cancerous human serum (5 ml, 1 ml, 500 μl, 250 μl, 100 μl, and 50 μl) and three different volumes (1 ml, 500 μl and 100 μl) of six individual commercial serum samples collected from human donors. The smaller starting volumes (100 μl and 50 μl) are used to mimic conditions of limited availability of heterogeneous biological samples. The isolated exosomes were characterized based upon size, quantity, zeta potential, CD63 and CD9 protein expression, and exosomal RNA (exRNA) quality and quantity using several complementary methods: nanoparticle tracking analysis (NTA) with ZetaView, western blot, transmission electron microscopy (TEM), the Agilent Bioanalyzer system, and droplet digital PCR (ddPCR). Our NTA results showed that all isolation techniques produced exosomes within the expected size range (40–150 nm). The three kits, though, produced a significantly higher yield (80–300 fold) of exosomes as compared to UC for all serum volumes, except 5 mL. We also found that exosomes isolated by the different techniques and serum volumes had similar zeta potentials to previous studies. Western blot analysis and TEM immunogold labelling confirmed the expression of two common exosomal protein markers, CD63 and CD9, in samples isolated by all techniques. All exosome isolations yielded high quality exRNA, containing mostly small RNA with a peak between 25 and 200 nucleotides in size. ddPCR results indicated that exosomes isolated from similar serum volumes but different isolation techniques rendered similar concentrations of two selected exRNA: hsa-miR-16 and hsa-miR-451. In summary, the three commercial exosome isolation kits are viable alternatives to UC, even when limited amounts of biological samples are available.
Collapse
|
459
|
Soung YH, Ford S, Zhang V, Chung J. Exosomes in Cancer Diagnostics. Cancers (Basel) 2017; 9:cancers9010008. [PMID: 28085080 PMCID: PMC5295779 DOI: 10.3390/cancers9010008] [Citation(s) in RCA: 236] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 01/06/2017] [Accepted: 01/09/2017] [Indexed: 12/21/2022] Open
Abstract
Exosomes are endosome derived extracellular vesicles of 30–120 nm size ranges. Exosomes have been identified as mediators of cell-to-cell communication by transferring bioactive molecules such as nucleic acids, proteins and lipids into recipient cells. While exosomes are secreted by multiple cell types, cancer derived exosomes not only influence the invasive potentials of proximally located cells, but also affect distantly located tissues. Based on their ability to alter tumor microenvironment by regulating immunity, angiogenesis and metastasis, there has been growing interest in defining the clinical relevance of exosomes in cancers. In particular, exosomes are valuable sources for biomarkers due to selective cargo loading and resemblance to their parental cells. In this review, we summarize the recent findings to utilize exosomes as cancer biomarkers for early detection, diagnosis and therapy selection.
Collapse
Affiliation(s)
- Young Hwa Soung
- Department of Pathology, Stony Brook Medicine, 101 Nicolls Road, Stony Brook, NY 11794, USA.
| | - Shane Ford
- Department of Pathology, Stony Brook Medicine, 101 Nicolls Road, Stony Brook, NY 11794, USA.
| | - Vincent Zhang
- Department of Pathology, Stony Brook Medicine, 101 Nicolls Road, Stony Brook, NY 11794, USA.
| | - Jun Chung
- Department of Pathology, Stony Brook Medicine, 101 Nicolls Road, Stony Brook, NY 11794, USA.
| |
Collapse
|
460
|
Abstract
Exosomes are nanometer-scale, membrane-enclosed vesicles that can potentially be used to detect nephrotoxicity, and reveal the subsequent response of the kidney. Epithelial cells of every nephron segment can contribute to the urinary exosome population, which is rich in potential biomarkers, including membrane proteins such as transporters and receptors, transcription factors, and microRNAs. These exosomal biomarkers may be up- or downregulated upon nephrotoxicant exposure. Exosome isolation is an area of ongoing research. Although faster and simpler methods have been developed, ultracentrifugation remains a mainstay for purification. A single ultracentrifugation step provides an enriched preparation suitable for biomarker discovery, and a second ultracentrifugation on a sucrose/D2O cushion provides the purest exosome preparation currently available and may be preferred for bioactivity assays. The concentration of exosomes can be determined using Nanosight Nanoparticle Tracking Analysis and their contents studied with a variety of approaches including western blots for proteins and RT-qPCR for microRNAs.
Collapse
|
461
|
|
462
|
Huang T, He J. Characterization of Extracellular Vesicles by Size-Exclusion High-Performance Liquid Chromatography (HPLC). Methods Mol Biol 2017; 1660:191-199. [PMID: 28828657 DOI: 10.1007/978-1-4939-7253-1_15] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Extracellular vesicles (EVs) have recently attracted substantial attention due to the potential diagnostic and therapeutic relevance. Although a variety of techniques have been used to isolate and analyze EVs, it is still far away from satisfaction. Size-exclusion chromatography (SEC), which separates subjects by size, has been widely applied in protein purification and analysis. The purpose of this chapter is to show the applications of size-exclusion high-performance liquid chromatography (HPLC) as methods for EV characterization of impurities or contaminants of small size, and thus for quality assay for the purity of the samples of EVs.
Collapse
Affiliation(s)
- Tao Huang
- Department of Radiology and Medical Imaging, University of Virginia, PO BOX 801339, Fontaine Research Park, Rm180, 480 Ray C Hunt Drive, Charlottesville, 22903, VA, USA
| | - Jiang He
- Department of Radiology and Medical Imaging, University of Virginia, PO BOX 801339, Fontaine Research Park, Rm. 282, 480 Ray C. Hunt Drive, Charlottesville, VA, 22903, USA.
| |
Collapse
|
463
|
Greening DW, Xu R, Gopal SK, Rai A, Simpson RJ. Proteomic insights into extracellular vesicle biology - defining exosomes and shed microvesicles. Expert Rev Proteomics 2016; 14:69-95. [PMID: 27838931 DOI: 10.1080/14789450.2017.1260450] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Extracellular vesicles (EVs) are critical mediators of intercellular communication, capable of regulating the transcriptional landscape of target cells through horizontal transmission of biological information, such as proteins, lipids, and RNA species. This capability highlights their potential as novel targets for disease intervention. Areas covered: This review focuses on the emerging importance of discovery proteomics (high-throughput, unbiased quantitative protein identification) and targeted proteomics (hypothesis-driven quantitative protein subset analysis) mass spectrometry (MS)-based strategies in EV biology, especially exosomes and shed microvesicles. Expert commentary: Recent advances in MS hardware, workflows, and informatics provide comprehensive, quantitative protein profiling of EVs and EV-treated target cells. This information is seminal to understanding the role of EV subtypes in cellular crosstalk, especially when integrated with other 'omics disciplines, such as RNA analysis (e.g., mRNA, ncRNA). Moreover, high-throughput MS-based proteomics promises to provide new avenues in identifying novel markers for detection, monitoring, and therapeutic intervention of disease.
Collapse
Affiliation(s)
- David W Greening
- a Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science , La Trobe University , Melbourne , Australia
| | - Rong Xu
- a Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science , La Trobe University , Melbourne , Australia
| | - Shashi K Gopal
- a Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science , La Trobe University , Melbourne , Australia
| | - Alin Rai
- a Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science , La Trobe University , Melbourne , Australia
| | - Richard J Simpson
- a Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science , La Trobe University , Melbourne , Australia
| |
Collapse
|
464
|
Reis M, Ogonek J, Qesari M, Borges NM, Nicholson L, Preußner L, Dickinson AM, Wang XN, Weissinger EM, Richter A. Recent Developments in Cellular Immunotherapy for HSCT-Associated Complications. Front Immunol 2016; 7:500. [PMID: 27895644 PMCID: PMC5107577 DOI: 10.3389/fimmu.2016.00500] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 10/26/2016] [Indexed: 12/13/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation is associated with serious complications, and improvement of the overall clinical outcome of patients with hematological malignancies is necessary. During the last decades, posttransplant donor-derived adoptive cellular immunotherapeutic strategies have been progressively developed for the treatment of graft-versus-host disease (GvHD), infectious complications, and tumor relapses. To date, the common challenge of all these cell-based approaches is their implementation for clinical application. Establishing an appropriate manufacturing process, to guarantee safe and effective therapeutics with simultaneous consideration of economic requirements is one of the most critical hurdles. In this review, we will discuss the recent scientific findings, clinical experiences, and technological advances for cell processing toward the application of mesenchymal stromal cells as a therapy for treatment of severe GvHD, virus-specific T cells for targeting life-threating infections, and of chimeric antigen receptors-engineered T cells to treat relapsed leukemia.
Collapse
Affiliation(s)
- Monica Reis
- Haematological Sciences, Institute of Cellular Medicine, Newcastle University , Newcastle upon Tyne , UK
| | - Justyna Ogonek
- Transplantation Biology, Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School , Hannover , Germany
| | | | - Nuno M Borges
- Haematological Sciences, Institute of Cellular Medicine, Newcastle University , Newcastle upon Tyne , UK
| | - Lindsay Nicholson
- Haematological Sciences, Institute of Cellular Medicine, Newcastle University , Newcastle upon Tyne , UK
| | | | - Anne Mary Dickinson
- Haematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK; Alcyomics Ltd., Newcastle upon Tyne, UK
| | - Xiao-Nong Wang
- Haematological Sciences, Institute of Cellular Medicine, Newcastle University , Newcastle upon Tyne , UK
| | - Eva M Weissinger
- Transplantation Biology, Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School , Hannover , Germany
| | - Anne Richter
- Miltenyi Biotec GmbH , Bergisch Gladbach , Germany
| |
Collapse
|
465
|
Bosch S, de Beaurepaire L, Allard M, Mosser M, Heichette C, Chrétien D, Jegou D, Bach JM. Trehalose prevents aggregation of exosomes and cryodamage. Sci Rep 2016; 6:36162. [PMID: 27824088 PMCID: PMC5099918 DOI: 10.1038/srep36162] [Citation(s) in RCA: 231] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 10/07/2016] [Indexed: 12/19/2022] Open
Abstract
Exosomes are important mediators in intercellular communication. Released by many cell types, they transport proteins, lipids, and nucleic acids to distant recipient cells and contribute to important physiopathological processes. Standard current exosome isolation methods based on differential centrifugation protocols tend to induce aggregation of particles in highly concentrated suspensions and freezing of exosomes can induce damage and inconsistent biological activity. Trehalose is a natural, non-toxic sugar widely used as a protein stabilizer and cryoprotectant by the food and drug industry. Here we report that addition of 25 mM trehalose to pancreatic beta-cell exosome-like vesicle isolation and storage buffer narrows the particle size distribution and increases the number of individual particles per microgram of protein. Repeated freeze-thaw cycles induce an increase in particle concentration and in the width of the size distribution for exosome-like vesicles stored in PBS, but not in PBS 25 mM trehalose. No signs of lysis or incomplete vesicles were observed by cryo-electron tomography in PBS and trehalose samples. In macrophage immune assays, beta-cell extracellular vesicles in trehalose show consistently higher TNF-alpha cytokine secretion stimulation indexes suggesting improved preservation of biological activity. The addition of trehalose might be an attractive means to standardize experiments in the field of exosome research and downstream applications.
Collapse
Affiliation(s)
- Steffi Bosch
- IECM, EA4644 Nantes University, ONIRIS, USC1383 INRA, Nantes, France
| | | | - Marie Allard
- IECM, EA4644 Nantes University, ONIRIS, USC1383 INRA, Nantes, France
| | - Mathilde Mosser
- IECM, EA4644 Nantes University, ONIRIS, USC1383 INRA, Nantes, France
| | | | - Denis Chrétien
- IGDR, UMR6290 CNRS, University of Rennes 1, Rennes, France.,MRIC-Biosit, UMS3480 CNRS, University of Rennes 1, Rennes, France
| | - Dominique Jegou
- IECM, EA4644 Nantes University, ONIRIS, USC1383 INRA, Nantes, France
| | - Jean-Marie Bach
- IECM, EA4644 Nantes University, ONIRIS, USC1383 INRA, Nantes, France
| |
Collapse
|
466
|
Gardiner C, Di Vizio D, Sahoo S, Théry C, Witwer KW, Wauben M, Hill AF. Techniques used for the isolation and characterization of extracellular vesicles: results of a worldwide survey. J Extracell Vesicles 2016; 5:32945. [PMID: 27802845 PMCID: PMC5090131 DOI: 10.3402/jev.v5.32945] [Citation(s) in RCA: 691] [Impact Index Per Article: 86.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 09/13/2016] [Accepted: 09/21/2016] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles (EVs) represent an important mode of intercellular communication. Research in this field has grown rapidly in the last few years, and there is a plethora of techniques for the isolation and characterization of EVs, many of which are poorly standardized. EVs are heterogeneous in size, origin and molecular constituents, with considerable overlap in size and phenotype between different populations of EVs. Little is known about current practices for the isolation, purification and characterization of EVs. We report here the first large, detailed survey of current worldwide practices for the isolation and characterization of EVs. Conditioned cell culture media was the most widely used material (83%). Ultracentrifugation remains the most commonly used isolation method (81%) with 59% of respondents use a combination of methods. Only 9% of respondents used only 1 characterization method, with others using 2 or more methods. Sample volume, sample type and downstream application all influenced the isolation and characterization techniques employed.
Collapse
Affiliation(s)
- Chris Gardiner
- Haemostasis Research Unit, Research Department of Haematology, University College London, London, UK;
| | - Dolores Di Vizio
- Icahn School of Medicine, Cardiovascular Research Center, Cedars-Sinai, Los Angeles, CA, USA
| | - Susmita Sahoo
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Clotilde Théry
- Institut Curie, PSL Research University, INSERM U932, Paris, France
| | | | - Marca Wauben
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Andrew F Hill
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Australia
| |
Collapse
|
467
|
Nguyen HPT, Simpson RJ, Salamonsen LA, Greening DW. Extracellular Vesicles in the Intrauterine Environment: Challenges and Potential Functions. Biol Reprod 2016; 95:109. [PMID: 27655784 PMCID: PMC5333933 DOI: 10.1095/biolreprod.116.143503] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 08/08/2016] [Accepted: 09/13/2016] [Indexed: 01/07/2023] Open
Abstract
Extracellular vesicles (EVs), including exosomes (30–150 nm) and microvesicles (100–1500 nm), play important roles in mediating cell-cell communication. Such particles package distinct cargo elements, including lipids, proteins, mRNAs, microRNAs, and DNA, that vary depending on the cell of origin and its phenotype. This cargo can be horizontally transferred to target cells where its components can reprogram the recipient cell to modify its function. EVs have been identified within the uterine cavity of women, sheep, and mice, where they contribute to the microenvironment of sperm transport, and of blastocyst and endometrial preparation for implantation. It is likely that exosomes and microvesicles carry different cargo and coordinate different roles in this intrauterine environment. Understanding and defining these subtypes of EVs is important for future functional studies and clinical translation. Here we critically review the various purification and validation procedures for extracellular vesicle analysis and discuss what is known of endometrial-derived exosome cargo and of their hormonal regulation. The current knowledge of the functions of uterine exosomes, with respect to sperm transport and function, and of their actions on trophectodermal cells to promote implantation are summarized and evaluated in their physiological context. Given the potential importance of this form of cell-cell interactions within the reproductive tract, the critical issues discussed will guide new insights in this rapidly expanding field.
Collapse
Affiliation(s)
- Hong P T Nguyen
- Hudson Institute of Medical Research (previously Prince Henry's Institute), Clayton, Victoria, Australia
| | - Richard J Simpson
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Lois A Salamonsen
- Hudson Institute of Medical Research (previously Prince Henry's Institute), Clayton, Victoria, Australia
| | - David W Greening
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| |
Collapse
|
468
|
Rupert DLM, Claudio V, Lässer C, Bally M. Methods for the physical characterization and quantification of extracellular vesicles in biological samples. Biochim Biophys Acta Gen Subj 2016; 1861:3164-3179. [PMID: 27495390 DOI: 10.1016/j.bbagen.2016.07.028] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 07/06/2016] [Accepted: 07/27/2016] [Indexed: 01/09/2023]
Abstract
BACKGROUND Our body fluids contain a multitude of cell-derived vesicles, secreted by most cell types, commonly referred to as extracellular vesicles. They have attracted considerable attention for their function as intercellular communication vehicles in a broad range of physiological processes and pathological conditions. Extracellular vesicles and especially the smallest type, exosomes, have also generated a lot of excitement in view of their potential as disease biomarkers or as carriers for drug delivery. In this context, state-of-the-art techniques capable of comprehensively characterizing vesicles in biological fluids are urgently needed. SCOPE OF REVIEW This review presents the arsenal of techniques available for quantification and characterization of physical properties of extracellular vesicles, summarizes their working principles, discusses their advantages and limitations and further illustrates their implementation in extracellular vesicle research. MAJOR CONCLUSIONS The small size and physicochemical heterogeneity of extracellular vesicles make their physical characterization and quantification an extremely challenging task. Currently, structure, size, buoyant density, optical properties and zeta potential have most commonly been studied. The concentration of vesicles in suspension can be expressed in terms of biomolecular or particle content depending on the method at hand. In addition, common quantification methods may either provide a direct quantitative measurement of vesicle concentration or solely allow for relative comparison between samples. GENERAL SIGNIFICANCE The combination of complementary methods capable of detecting, characterizing and quantifying extracellular vesicles at a single particle level promises to provide new exciting insights into their modes of action and to reveal the existence of vesicle subpopulations fulfilling key biological tasks.
Collapse
Affiliation(s)
- Déborah L M Rupert
- Department of Physics, Chalmers University of Technology, Gothenburg, Sweden
| | - Virginia Claudio
- Department of Physics, Chalmers University of Technology, Gothenburg, Sweden; Center for Brain Repair and Rehabilitation, Institute for Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Cecilia Lässer
- Krefting Research Centre, Department of Internal Medicine and Clinical Nutrition, University of Gothenburg, Gothenburg, Sweden
| | - Marta Bally
- Department of Physics, Chalmers University of Technology, Gothenburg, Sweden; Institut Curie, Centre de Recherche, CNRS, UMR168, Physico-Chimie Curie, Paris, France.
| |
Collapse
|
469
|
Watson DC, Bayik D, Srivatsan A, Bergamaschi C, Valentin A, Niu G, Bear J, Monninger M, Sun M, Morales-Kastresana A, Jones JC, Felber BK, Chen X, Gursel I, Pavlakis GN. Efficient production and enhanced tumor delivery of engineered extracellular vesicles. Biomaterials 2016; 105:195-205. [PMID: 27522254 DOI: 10.1016/j.biomaterials.2016.07.003] [Citation(s) in RCA: 261] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 07/01/2016] [Accepted: 07/05/2016] [Indexed: 12/20/2022]
Abstract
Extracellular vesicles (EV), including exosomes and microvesicles, are nano-sized intercellular communication vehicles that participate in a multitude of physiological processes. Due to their biological properties, they are also promising candidates for the systemic delivery of therapeutic compounds, such as cytokines, chemotherapeutic drugs, siRNAs and viral vectors. However, low EV production yield and rapid clearance of administered EV by liver macrophages limit their potential use as therapeutic vehicles. We have used a hollow-fiber bioreactor for the efficient production of bioactive EV bearing the heterodimeric cytokine complex Interleukin-15:Interleukin-15 receptor alpha. Bioreactor culture yielded ∼40-fold more EV per mL conditioned medium, as compared to conventional cell culture. Biophysical analysis and comparative proteomics suggested a more diverse population of EV in the bioreactor preparations, while serum protein contaminants were detectable only in conventional culture EV preparations. We also identified the Scavenger Receptor Class A family (SR-A) as a novel monocyte/macrophage uptake receptor for EV. In vivo blockade of SR-A with dextran sulfate dramatically decreased EV liver clearance in mice, while enhancing tumor accumulation. These findings facilitate development of EV therapeutic methods.
Collapse
Affiliation(s)
- Dionysios C Watson
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, United States; Department of Medicine, University of Patras, Greece
| | - Defne Bayik
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, United States; Department of Molecular Biology and Genetics, Bilkent University, Ankara, 06800 Turkey
| | - Avinash Srivatsan
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, United States
| | - Cristina Bergamaschi
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, United States
| | - Antonio Valentin
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, United States
| | - Gang Niu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, United States
| | - Jenifer Bear
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, United States
| | - Mitchell Monninger
- Pathology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, United States
| | - Mei Sun
- Pathology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, United States
| | - Aizea Morales-Kastresana
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, United States
| | - Jennifer C Jones
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, United States
| | - Barbara K Felber
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, United States
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, United States
| | - Ihsan Gursel
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, 06800 Turkey
| | - George N Pavlakis
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, United States.
| |
Collapse
|
470
|
Welton JL, Brennan P, Gurney M, Webber JP, Spary LK, Carton DG, Falcón-Pérez JM, Walton SP, Mason MD, Tabi Z, Clayton A. Proteomics analysis of vesicles isolated from plasma and urine of prostate cancer patients using a multiplex, aptamer-based protein array. J Extracell Vesicles 2016; 5:31209. [PMID: 27363484 PMCID: PMC4929354 DOI: 10.3402/jev.v5.31209] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 04/11/2016] [Accepted: 04/17/2016] [Indexed: 12/28/2022] Open
Abstract
Proteomics analysis of biofluid-derived vesicles holds enormous potential for discovering non-invasive disease markers. Obtaining vesicles of sufficient quality and quantity for profiling studies has, however, been a major problem, as samples are often replete with co-isolated material that can interfere with the identification of genuine low abundance, vesicle components. Here, we used a combination of ultracentrifugation and size-exclusion chromatography to isolate and analyse vesicles of plasma or urine origin. We describe a sample-handling workflow that gives reproducible, quality vesicle isolations sufficient for subsequent protein profiling. Using a semi-quantitative aptamer-based protein array, we identified around 1,000 proteins, of which almost 400 were present at comparable quantities in plasma versus urine vesicles. Significant differences were, however, apparent with elements like HSP90, integrin αVβ5 and Contactin-1 more prevalent in urinary vesicles, while hepatocyte growth factor activator, prostate-specific antigen–antichymotrypsin complex and many others were more abundant in plasma vesicles. This was also applied to a small set of specimens collected from men with metastatic prostate cancer, highlighting several proteins with the potential to indicate treatment refractory disease. The study provides a practical platform for furthering protein profiling of vesicles in prostate cancer, and, hopefully, many other disease scenarios.
Collapse
Affiliation(s)
- Joanne Louise Welton
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom.,Velindre Cancer Centre, Cardiff, United Kingdom.,Cardiff School of Health Sciences, Cardiff Metropolitan University, Cardiff, United Kingdom
| | - Paul Brennan
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Mark Gurney
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom.,Velindre Cancer Centre, Cardiff, United Kingdom
| | - Jason Paul Webber
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom.,Velindre Cancer Centre, Cardiff, United Kingdom
| | - Lisa Kate Spary
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom.,Velindre Cancer Centre, Cardiff, United Kingdom
| | - David Gil Carton
- Metabolomics Unit, CIC bioGUNE, CIBERehd, Bizkaia Technology Park, Derio, Spain
| | | | - Sean Peter Walton
- Department of Computer Science, College of Science, Swansea University, United Kingdom
| | - Malcolm David Mason
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom.,Velindre Cancer Centre, Cardiff, United Kingdom
| | - Zsuzsanna Tabi
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom.,Velindre Cancer Centre, Cardiff, United Kingdom
| | - Aled Clayton
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom.,Velindre Cancer Centre, Cardiff, United Kingdom;
| |
Collapse
|
471
|
Huleihel L, Hussey GS, Naranjo JD, Zhang L, Dziki JL, Turner NJ, Stolz DB, Badylak SF. Matrix-bound nanovesicles within ECM bioscaffolds. SCIENCE ADVANCES 2016; 2:e1600502. [PMID: 27386584 PMCID: PMC4928894 DOI: 10.1126/sciadv.1600502] [Citation(s) in RCA: 232] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 05/19/2016] [Indexed: 05/13/2023]
Abstract
Biologic scaffold materials composed of extracellular matrix (ECM) have been used in a variety of surgical and tissue engineering/regenerative medicine applications and are associated with favorable constructive remodeling properties including angiogenesis, stem cell recruitment, and modulation of macrophage phenotype toward an anti-inflammatory effector cell type. However, the mechanisms by which these events are mediated are largely unknown. Matrix-bound nanovesicles (MBVs) are identified as an integral and functional component of ECM bioscaffolds. Extracellular vesicles (EVs) are potent vehicles of intercellular communication due to their ability to transfer RNA, proteins, enzymes, and lipids, thereby affecting physiologic and pathologic processes. Formerly identified exclusively in biologic fluids, the presence of EVs within the ECM of connective tissue has not been reported. In both laboratory-produced and commercially available biologic scaffolds, MBVs can be separated from the matrix only after enzymatic digestion of the ECM scaffold material, a temporal sequence similar to the functional activity attributed to implanted bioscaffolds during and following their degradation when used in clinical applications. The present study shows that MBVs contain microRNA capable of exerting phenotypical and functional effects on macrophage activation and neuroblastoma cell differentiation. The identification of MBVs embedded within the ECM of biologic scaffolds provides mechanistic insights not only into the inductive properties of ECM bioscaffolds but also into the regulation of tissue homeostasis.
Collapse
Affiliation(s)
- Luai Huleihel
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - George S. Hussey
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Juan Diego Naranjo
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Li Zhang
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Jenna L. Dziki
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Neill J. Turner
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Donna B. Stolz
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Center of Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Stephen F. Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Corresponding author.
| |
Collapse
|
472
|
Rider MA, Hurwitz SN, Meckes DG. ExtraPEG: A Polyethylene Glycol-Based Method for Enrichment of Extracellular Vesicles. Sci Rep 2016; 6:23978. [PMID: 27068479 PMCID: PMC4828635 DOI: 10.1038/srep23978] [Citation(s) in RCA: 451] [Impact Index Per Article: 56.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 03/17/2016] [Indexed: 12/11/2022] Open
Abstract
Initially thought to be a means for cells to eliminate waste, secreted extracellular vesicles, known as exosomes, are now understood to mediate numerous healthy and pathological processes. Though abundant in biological fluids, purifying exosomes has been challenging because their biophysical properties overlap with other secreted cell products. Easy-to-use commercial kits for harvesting exosomes are now widely used, but the relative low-purity and high-cost of the preparations restricts their utility. Here we describe a method for purifying exosomes and other extracellular vesicles by adapting methods for isolating viruses using polyethylene glycol. This technique, called ExtraPEG, enriches exosomes from large volumes of media rapidly and inexpensively using low-speed centrifugation, followed by a single small-volume ultracentrifugation purification step. Total protein and RNA harvested from vesicles is sufficient in quantity and quality for proteomics and sequencing analyses, demonstrating the utility of this method for biomarker discovery and diagnostics. Additionally, confocal microscopy studies suggest that the biological activity of vesicles is not impaired. The ExtraPEG method can be easily adapted to enrich for different vesicle populations, or as an efficient precursor to subsequent purification techniques, providing a means to harvest exosomes from many different biological fluids and for a wide variety of purposes.
Collapse
Affiliation(s)
- Mark A. Rider
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, 32306, FL, USA
| | - Stephanie N. Hurwitz
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, 32306, FL, USA
| | - David G. Meckes
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, 32306, FL, USA
| |
Collapse
|
473
|
Xu R, Greening DW, Zhu HJ, Takahashi N, Simpson RJ. Extracellular vesicle isolation and characterization: toward clinical application. J Clin Invest 2016; 126:1152-62. [PMID: 27035807 DOI: 10.1172/jci81129] [Citation(s) in RCA: 620] [Impact Index Per Article: 77.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Two broad categories of extracellular vesicles (EVs), exosomes and shed microvesicles (sMVs), which differ in size distribution as well as protein and RNA profiles, have been described. EVs are known to play key roles in cell-cell communication, acting proximally as well as systemically. This Review discusses the nature of EV subtypes, strategies for isolating EVs from both cell-culture media and body fluids, and procedures for quantifying EVs. We also discuss proteins selectively enriched in exosomes and sMVs that have the potential for use as markers to discriminate between EV subtypes, as well as various applications of EVs in clinical diagnosis.
Collapse
|
474
|
Paolini L, Zendrini A, Di Noto G, Busatto S, Lottini E, Radeghieri A, Dossi A, Caneschi A, Ricotta D, Bergese P. Residual matrix from different separation techniques impacts exosome biological activity. Sci Rep 2016; 6:23550. [PMID: 27009329 PMCID: PMC4806376 DOI: 10.1038/srep23550] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 03/07/2016] [Indexed: 02/05/2023] Open
Abstract
Exosomes are gaining a prominent role in research due to their intriguing biology and several therapeutic opportunities. However, their accurate purification from body fluids and detailed physicochemical characterization remain open issues. We isolated exosomes from serum of patients with Multiple Myeloma by four of the most popular purification methods and assessed the presence of residual contaminants in the preparations through an ad hoc combination of biochemical and biophysical techniques - including Western Blot, colloidal nanoplasmonics, atomic force microscopy (AFM) and scanning helium ion microscopy (HIM). The preparations obtained by iodixanol and sucrose gradients were highly pure. To the contrary, those achieved with limited processing (serial centrifugation or one step precipitation kit) resulted contaminated by a residual matrix, embedding the exosomes. The contaminated preparations showed lower ability to induce NfkB nuclear translocation in endothelial cells with respect to the pure ones, probably because the matrix prevents the interaction and fusion of the exosomes with the cell membrane. These findings suggest that exosome preparation purity must be carefully assessed since it may interfere with exosome biological activity. Contaminants can be reliably probed only by an integrated characterization approach aimed at both the molecular and the colloidal length scales.
Collapse
Affiliation(s)
- Lucia Paolini
- Department of Molecular and Translational Medicine and INSTM, University of Brescia, Brescia, Italy
| | - Andrea Zendrini
- Department of Molecular and Translational Medicine and INSTM, University of Brescia, Brescia, Italy
| | - Giuseppe Di Noto
- Department of Molecular and Translational Medicine and INSTM, University of Brescia, Brescia, Italy
| | - Sara Busatto
- Department of Molecular and Translational Medicine and INSTM, University of Brescia, Brescia, Italy
| | - Elisabetta Lottini
- Department of Chemistry and INSTM, Laboratory of Molecular Magnetism, University of Firenze, Sesto Fiorentino (Firenze), Italy
| | - Annalisa Radeghieri
- Department of Molecular and Translational Medicine and INSTM, University of Brescia, Brescia, Italy
| | - Alessandra Dossi
- Department of Molecular and Translational Medicine and INSTM, University of Brescia, Brescia, Italy
| | - Andrea Caneschi
- Department of Chemistry and INSTM, Laboratory of Molecular Magnetism, University of Firenze, Sesto Fiorentino (Firenze), Italy
| | - Doris Ricotta
- Department of Molecular and Translational Medicine and INSTM, University of Brescia, Brescia, Italy
| | - Paolo Bergese
- Department of Molecular and Translational Medicine and INSTM, University of Brescia, Brescia, Italy
| |
Collapse
|
475
|
Ibrahim SH, Hirsova P, Tomita K, Bronk SF, Werneburg NW, Harrison SA, Goodfellow VS, Malhi H, Gores GJ. Mixed lineage kinase 3 mediates release of C-X-C motif ligand 10-bearing chemotactic extracellular vesicles from lipotoxic hepatocytes. Hepatology 2016; 63:731-44. [PMID: 26406121 PMCID: PMC4764421 DOI: 10.1002/hep.28252] [Citation(s) in RCA: 184] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 08/28/2015] [Accepted: 09/22/2015] [Indexed: 12/11/2022]
Abstract
UNLABELLED Mixed lineage kinase 3 (MLK3) deficiency reduces macrophage-associated inflammation in a murine model of nonalcoholic steatohepatitis (NASH). However, the mechanistic links between MLK3 activation in hepatocytes and macrophage-driven inflammation in NASH are uncharted. Herein, we report that MLK3 mediates the release of (C-X-C motif) ligand 10 (CXCL10)-laden extracellular vesicles (EVs) from lipotoxic hepatocytes, which induce macrophage chemotaxis. Primary mouse hepatocytes (PMHs) and Huh7 cells were treated with palmitate or lysophosphatidylcholine (LPC). Released EVs were isolated by differential ultracentrifugation. LPC treatment of PMH or Huh7 cells induced release of EVs, which was prevented by either genetic or pharmacological inhibition of MLK3. Mass spectrometry identified the potent chemokine, CXCL10, in the EVs, which was markedly enriched in EVs isolated from LPC-treated hepatocytes versus untreated cells. Green fluorescent protein (GFP)-tagged CXCL10 was present in vesicular structures and colocalized with the red fluorescent protein (RFP)-tagged EV marker, CD63, after LPC treatment of cotransfected Huh-7 cells. Either genetic deletion or pharmacological inhibition of MLK3 prevented CXCL10 enrichment in EVs. Treatment of mouse bone-marrow-derived macrophages with lipotoxic hepatocyte-derived EVs induced macrophage chemotaxis, an effect blocked by incubation with CXCL10-neutralizing antisera. MLK3-deficient mice fed a NASH-inducing diet had reduced concentrations of total plasma EVs and CXCL10 containing EVs compared to wild-type mice. CONCLUSIONS During hepatocyte lipotoxicity, activated MLK3 induces the release of CXCL10-bearing vesicles from hepatocytes, which are chemotactic for macrophages.
Collapse
Affiliation(s)
- Samar H. Ibrahim
- Division of Pediatric Gastroenterology, Mayo Clinic, Rochester, Minnesota
| | - Petra Hirsova
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Kyoko Tomita
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Steven F. Bronk
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Nathan W. Werneburg
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, Minnesota
| | | | | | - Harmeet Malhi
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Gregory J. Gores
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
476
|
Vallabhaneni KC, Penfornis P, Dhule S, Guillonneau F, Adams KV, Mo YY, Xu R, Liu Y, Watabe K, Vemuri MC, Pochampally R. Extracellular vesicles from bone marrow mesenchymal stem/stromal cells transport tumor regulatory microRNA, proteins, and metabolites. Oncotarget 2016; 6:4953-67. [PMID: 25669974 PMCID: PMC4467126 DOI: 10.18632/oncotarget.3211] [Citation(s) in RCA: 256] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 12/27/2014] [Indexed: 12/20/2022] Open
Abstract
Human mesenchymal stem/stromal cells (hMSCs) have been shown to support breast cancer cell proliferation and metastasis, partly through their secretome. hMSCs have a remarkable ability to survive for long periods under stress, and their secretome is tumor supportive. In this study, we have characterized the cargo of extracellular vesicular (EV) fraction (that is in the size range of 40-150nm) of serum deprived hMSCs (SD-MSCs). Next Generation Sequencing assays were used to identify small RNA secreted in the EVs, which indicated presence of tumor supportive miRNA. Further assays demonstrated the role of miRNA-21 and 34a as tumor supportive miRNAs. Next, proteomic assays revealed the presence of ≈150 different proteins, most of which are known tumor supportive factors such as PDGFR-β, TIMP-1, and TIMP-2. Lipidomic assays verified presence of bioactive lipids such as sphingomyelin. Furthermore, metabolite assays identified the presence of lactic acid and glutamic acid in EVs. The co-injection xenograft assays using MCF-7 breast cancer cells demonstrated the tumor supportive function of these EVs. To our knowledge this is the first comprehensive -omics based study that characterized the complex cargo of extracellular vesicles secreted by hMSCs and their role in supporting breast cancers.
Collapse
Affiliation(s)
| | - Patrice Penfornis
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA
| | - Santosh Dhule
- Department of Chemical and Biomolecular Engineering, New Orleans, LA, USA
| | - Francois Guillonneau
- 3P5 Proteomic Platform of the Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Kristen V Adams
- Department of Pathology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Yin Yuan Mo
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA
| | - Rui Xu
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, MS, USA
| | - Yiming Liu
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, MS, USA
| | - Kounosuke Watabe
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA
| | - Mohan C Vemuri
- Stem Cell Biology, Thermo Fisher Scientific, Frederick, MD, USA
| | - Radhika Pochampally
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA.,Department of Biochemistry, University of Mississippi Medical Center, Jackson, MS, USA
| |
Collapse
|
477
|
Kalra H, Drummen GPC, Mathivanan S. Focus on Extracellular Vesicles: Introducing the Next Small Big Thing. Int J Mol Sci 2016; 17:170. [PMID: 26861301 PMCID: PMC4783904 DOI: 10.3390/ijms17020170] [Citation(s) in RCA: 569] [Impact Index Per Article: 71.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 11/12/2015] [Indexed: 12/20/2022] Open
Abstract
Intercellular communication was long thought to be regulated exclusively through direct contact between cells or via release of soluble molecules that transmit the signal by binding to a suitable receptor on the target cell, and/or via uptake into that cell. With the discovery of small secreted vesicular structures that contain complex cargo, both in their lumen and the lipid membrane that surrounds them, a new frontier of signal transduction was discovered. These “extracellular vesicles” (EV) were initially thought to be garbage bags through which the cell ejected its waste. Whilst this is a major function of one type of EV, i.e., apoptotic bodies, many EVs have intricate functions in intercellular communication and compound exchange; although their physiological roles are still ill-defined. Additionally, it is now becoming increasingly clear that EVs mediate disease progression and therefore studying EVs has ignited significant interests among researchers from various fields of life sciences. Consequently, the research effort into the pathogenic roles of EVs is significantly higher even though their protective roles are not well established. The “Focus on extracellular vesicles” series of reviews highlights the current state of the art regarding various topics in EV research, whilst this review serves as an introductory overview of EVs, their biogenesis and molecular composition.
Collapse
Affiliation(s)
- Hina Kalra
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia.
| | - Gregor P C Drummen
- Cellular Stress and Ageing Program, Bionanoscience and Bio-Imaging Program, Bio&Nano-Solutions, D-33647 Bielefeld, Germany.
| | - Suresh Mathivanan
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia.
| |
Collapse
|
478
|
Tong M, Kleffmann T, Pradhan S, Johansson CL, DeSousa J, Stone PR, James JL, Chen Q, Chamley LW. Proteomic characterization of macro-, micro- and nano-extracellular vesicles derived from the same first trimester placenta: relevance for feto-maternal communication. Hum Reprod 2016; 31:687-99. [PMID: 26839151 DOI: 10.1093/humrep/dew004] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 01/02/2016] [Indexed: 12/11/2022] Open
Abstract
STUDY QUESTION What proteins are carried by extracellular vesicles (EVs) released from normal first trimester placentae? SUMMARY ANSWER One thousand five hundred and eighty-five, 1656 and 1476 proteins were characterized in macro-, micro- and nano-vesicles, respectively, from first trimester placentae, with all EV fractions being enriched for proteins involved in vesicle transport and inflammation. WHAT IS KNOWN ALREADY Placental EVs are being increasingly recognized as important mediators of both healthy and pathological pregnancies. However, current research has focused on detecting changes in specific proteins in particular fractions of vesicles during disease. This is the first study to investigate the full proteome of different-sized fractions of EVs from the same first trimester placenta and highlights the differences/similarities between the vesicle fractions. STUDY DESIGN, SIZE, DURATION A well-established ex vivo placental explant culture model was used to generate macro-, micro- and nano-vesicles from 56 first trimester placentae. Vesicle fractions were collected by differential ultracentrifugation, quantified and characterized. PARTICIPANTS/MATERIALS, SETTING, METHODS Placental macro-, micro- and nano-vesicles were characterized by microscopy, dynamic light scattering and nanoparticle tracking analysis. The proteome of each EV fraction was interrogated using liquid chromatography-coupled tandem mass spectrometry. Results were validated by semi-quantitative western blotting. MAIN RESULTS AND THE ROLE OF CHANCE A total of 1585, 1656 and 1476 proteins were identified in macro-, micro- and nano-vesicles, respectively. One thousand one hundred and twenty-five proteins were shared between all three fractions while up to 223 proteins were unique to each fraction. Gene Ontology pathway analysis showed an enrichment of proteins involved in vesicle transport and inflammation in all three fractions of EVs. The expression levels of proteins involved in internalization of vesicles (annexin V, calreticulin, CD31, CD47), the complement pathway [C3, decay-accelerating factor (DAF), membrane cofactor protein (MCP), protectin] and minor histocompatibility antigens [ATP-dependent RNA helicase (DDX3), ribosomal protein S4 (RPS4)] were different between different-sized EVs. LIMITATIONS, REASONS FOR CAUTION This study is largely hypothesis-generating in nature. It is important to validate these findings using EVs isolated from maternal plasma and the function of the different EV fractions would need further investigation. WIDER IMPLICATIONS OF THE FINDINGS Our results support the concept that various EV factions can interact with different maternal cells and have unique effects to mediate feto-maternal communication during early pregnancy. This study also provides a list of candidate proteins, which may inform the identification of robust markers that can be used to isolate placental vesicles from the maternal blood in the future. STUDY FUNDING/COMPETING INTERESTS M.T. is a recipient of the University of Auckland Health Research Doctoral Scholarship and the Freemasons Postgraduate Scholarship. This project was supported by a School of Medicine Performance-based research fund (PBRF) grant awarded to L.W.C. No authors have any conflicts of interest to disclose.
Collapse
Affiliation(s)
- Mancy Tong
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, 85 Park Road, Auckland 1023, New Zealand
| | - Torsten Kleffmann
- Centre for Protein Research, Department of Biochemistry, University of Otago, Dunedin 9016, New Zealand
| | - Shantanu Pradhan
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, 85 Park Road, Auckland 1023, New Zealand
| | - Caroline L Johansson
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, 85 Park Road, Auckland 1023, New Zealand Faculty of Medicine and Health Sciences, Linköping University, Linköping SE-581 83, Sweden
| | - Joana DeSousa
- Maternal Fetal Medicine, Auckland City Hospital, Auckland 1023, New Zealand
| | - Peter R Stone
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, 85 Park Road, Auckland 1023, New Zealand Maternal Fetal Medicine, Auckland City Hospital, Auckland 1023, New Zealand
| | - Joanna L James
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, 85 Park Road, Auckland 1023, New Zealand
| | - Qi Chen
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, 85 Park Road, Auckland 1023, New Zealand
| | - Larry W Chamley
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, 85 Park Road, Auckland 1023, New Zealand
| |
Collapse
|
479
|
Vituret C, Gallay K, Confort MP, Ftaich N, Matei CI, Archer F, Ronfort C, Mornex JF, Chanson M, Di Pietro A, Boulanger P, Hong SS. Transfer of the Cystic Fibrosis Transmembrane Conductance Regulator to Human Cystic Fibrosis Cells Mediated by Extracellular Vesicles. Hum Gene Ther 2016; 27:166-83. [DOI: 10.1089/hum.2015.144] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Affiliation(s)
- Cyrielle Vituret
- Viral Infections & Comparative Pathology, UMR-754 UCBL-INRA-EPHE, Université Lyon 1, Lyon Cedex 07, France
- Institut de Biologie et Chimie des Protéines, Unité BMSSI, UMR 5086 CNRS-Université Lyon 1, Lyon Cedex 07, France
| | - Kathy Gallay
- Viral Infections & Comparative Pathology, UMR-754 UCBL-INRA-EPHE, Université Lyon 1, Lyon Cedex 07, France
| | - Marie-Pierre Confort
- Viral Infections & Comparative Pathology, UMR-754 UCBL-INRA-EPHE, Université Lyon 1, Lyon Cedex 07, France
| | - Najate Ftaich
- Viral Infections & Comparative Pathology, UMR-754 UCBL-INRA-EPHE, Université Lyon 1, Lyon Cedex 07, France
| | - Constantin I. Matei
- Centre Technologique des Microstructures, Université Claude Bernard—Lyon, Villeurbanne, France
| | - Fabienne Archer
- Viral Infections & Comparative Pathology, UMR-754 UCBL-INRA-EPHE, Université Lyon 1, Lyon Cedex 07, France
| | - Corinne Ronfort
- Viral Infections & Comparative Pathology, UMR-754 UCBL-INRA-EPHE, Université Lyon 1, Lyon Cedex 07, France
| | - Jean-François Mornex
- Viral Infections & Comparative Pathology, UMR-754 UCBL-INRA-EPHE, Université Lyon 1, Lyon Cedex 07, France
| | - Marc Chanson
- Département de Physiologie Cellulaire & Métabolisme, Centre Médical Universitaire, Geneva, Switzerland
| | - Attilio Di Pietro
- Institut de Biologie et Chimie des Protéines, Unité BMSSI, UMR 5086 CNRS-Université Lyon 1, Lyon Cedex 07, France
| | - Pierre Boulanger
- Viral Infections & Comparative Pathology, UMR-754 UCBL-INRA-EPHE, Université Lyon 1, Lyon Cedex 07, France
| | - Saw See Hong
- Viral Infections & Comparative Pathology, UMR-754 UCBL-INRA-EPHE, Université Lyon 1, Lyon Cedex 07, France
- Institut National de la Santé et de la Recherche Médicale, Paris, France
| |
Collapse
|
480
|
Abstract
Extracellular vesicles (ECV) are membrane compartments shed from all types of cells in various physiological and pathological states. In recent years, ECV have gained an increasing interest from the scientific community for their role as an intercellular communicator that plays important roles in modifying the tumor microenvironment. Multiple techniques have been established to collect ECV from conditioned media of cell culture or physiological fluids. The gold standard methodology is differential centrifugation. Although alternative techniques exist to collect ECV, these techniques have not proven suitable as a substitution for the ultracentrifugation procedure.
Collapse
Affiliation(s)
- Khalid Al-Nedawi
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, ON, Canada. .,Hamilton Centre for Kidney Research (HCKR), St. Joseph's Hospital, Hamilton, ON, Canada.
| | - Jolene Read
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, ON, Canada.,Hamilton Centre for Kidney Research (HCKR), St. Joseph's Hospital, Hamilton, ON, Canada
| |
Collapse
|
481
|
Smith ZJ, Lee C, Rojalin T, Carney RP, Hazari S, Knudson A, Lam K, Saari H, Ibañez EL, Viitala T, Laaksonen T, Yliperttula M, Wachsmann-Hogiu S. Single exosome study reveals subpopulations distributed among cell lines with variability related to membrane content. J Extracell Vesicles 2015; 4:28533. [PMID: 26649679 PMCID: PMC4673914 DOI: 10.3402/jev.v4.28533] [Citation(s) in RCA: 220] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 10/26/2015] [Accepted: 11/06/2015] [Indexed: 12/21/2022] Open
Abstract
Current analysis of exosomes focuses primarily on bulk analysis, where exosome-to-exosome variability cannot be assessed. In this study, we used Raman spectroscopy to study the chemical composition of single exosomes. We measured spectra of individual exosomes from 8 cell lines. Cell-line-averaged spectra varied considerably, reflecting the variation in total exosomal protein, lipid, genetic, and cytosolic content. Unexpectedly, single exosomes isolated from the same cell type also exhibited high spectral variability. Subsequent spectral analysis revealed clustering of single exosomes into 4 distinct groups that were not cell-line specific. Each group contained exosomes from multiple cell lines, and most cell lines had exosomes in multiple groups. The differences between these groups are related to chemical differences primarily due to differing membrane composition. Through a principal components analysis, we identified that the major sources of spectral variation among the exosomes were in cholesterol content, relative expression of phospholipids to cholesterol, and surface protein expression. For example, exosomes derived from cancerous versus non-cancerous cell lines can be largely separated based on their relative expression of cholesterol and phospholipids. We are the first to indicate that exosome subpopulations are shared among cell types, suggesting distributed exosome functionality. The origins of these differences are likely related to the specific role of extracellular vesicle subpopulations in both normal cell function and carcinogenesis, and they may provide diagnostic potential at the single exosome level.
Collapse
Affiliation(s)
- Zachary J Smith
- Center for Biophotonics, University of California Davis, Sacramento, CA, USA.,Department of Precision Mechanics and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui, China
| | - Changwon Lee
- Center for Biophotonics, University of California Davis, Sacramento, CA, USA
| | - Tatu Rojalin
- Center for Biophotonics, University of California Davis, Sacramento, CA, USA.,Division of Pharmaceutical Biosciences, Centre for Drug Research, University of Helsinki, Helsinki, Finland
| | - Randy P Carney
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, USA
| | - Sidhartha Hazari
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, USA
| | - Alisha Knudson
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, USA
| | - Kit Lam
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, USA
| | - Heikki Saari
- Division of Pharmaceutical Biosciences, Centre for Drug Research, University of Helsinki, Helsinki, Finland
| | - Elisa Lazaro Ibañez
- Division of Pharmaceutical Biosciences, Centre for Drug Research, University of Helsinki, Helsinki, Finland
| | - Tapani Viitala
- Division of Pharmaceutical Biosciences, Centre for Drug Research, University of Helsinki, Helsinki, Finland
| | - Timo Laaksonen
- Division of Pharmaceutical Biosciences, Centre for Drug Research, University of Helsinki, Helsinki, Finland
| | - Marjo Yliperttula
- Division of Pharmaceutical Biosciences, Centre for Drug Research, University of Helsinki, Helsinki, Finland
| | - Sebastian Wachsmann-Hogiu
- Center for Biophotonics, University of California Davis, Sacramento, CA, USA.,Department of Pathology and Laboratory Medicine, University of California Davis, Sacramento, CA, USA;
| |
Collapse
|
482
|
Konala VBR, Mamidi MK, Bhonde R, Das AK, Pochampally R, Pal R. The current landscape of the mesenchymal stromal cell secretome: A new paradigm for cell-free regeneration. Cytotherapy 2015; 18:13-24. [PMID: 26631828 DOI: 10.1016/j.jcyt.2015.10.008] [Citation(s) in RCA: 316] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 10/10/2015] [Accepted: 10/13/2015] [Indexed: 12/13/2022]
Abstract
The unique properties of mesenchymal stromal/stem cells (MSCs) to self-renew and their multipotentiality have rendered them attractive to researchers and clinicians. In addition to the differentiation potential, the broad repertoire of secreted trophic factors (cytokines) exhibiting diverse functions such as immunomodulation, anti-inflammatory activity, angiogenesis and anti-apoptotic, commonly referred to as the MSC secretome, has gained immense attention in the past few years. There is enough evidence to show that the one important pathway by which MSCs participate in tissue repair and regeneration is through its secretome. Concurrently, a large body of MSC research has focused on characterization of the MSC secretome; this includes both soluble factors and factors released in extracellular vesicles, for example, exosomes and microvesicles. This review provides an overview of our current understanding of the MSC secretome with respect to their potential clinical applications.
Collapse
Affiliation(s)
- Vijay Bhaskar Reddy Konala
- Department of Marine Biotechnology, AMET University, Kanathur, Chennai, India; Genes & Life Health Care Pvt. Ltd, Punjagutta, Hyderabad, India
| | | | - Ramesh Bhonde
- School of Regenerative Medicine, Manipal University, Bangalore, India
| | - Anjan Kumar Das
- Department of Surgery, Taylor's University School of Medicine, Sungai Buloh Hospital, Selangor, Malaysia
| | - Radhika Pochampally
- Department of Biochemistry, Cancer Institute, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Rajarshi Pal
- School of Regenerative Medicine, Manipal University, Bangalore, India.
| |
Collapse
|
483
|
Adamczyk M, Griffiths R, Dewitt S, Knäuper V, Aeschlimann D. P2X7 receptor activation regulates rapid unconventional export of transglutaminase-2. J Cell Sci 2015; 128:4615-28. [PMID: 26542019 PMCID: PMC4696497 DOI: 10.1242/jcs.175968] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 10/29/2015] [Indexed: 12/24/2022] Open
Abstract
Transglutaminases (denoted TG or TGM) are externalized from cells via an unknown unconventional secretory pathway. Here, we show for the first time that purinergic signaling regulates active secretion of TG2 (also known as TGM2), an enzyme with a pivotal role in stabilizing extracellular matrices and modulating cell–matrix interactions in tissue repair. Extracellular ATP promotes TG2 secretion by macrophages, and this can be blocked by a selective antagonist against the purinergic receptor P2X7 (P2X7R, also known as P2RX7). Introduction of functional P2X7R into HEK293 cells is sufficient to confer rapid, regulated TG2 export. By employing pharmacological agents, TG2 release could be separated from P2X7R-mediated microvesicle shedding. Neither Ca2+ signaling alone nor membrane depolarization triggered TG2 secretion, which occurred only upon receptor membrane pore formation and without pannexin channel involvement. A gain-of-function mutation in P2X7R associated with autoimmune disease caused enhanced TG2 externalization from cells, and this correlated with increased pore activity. These results provide a mechanistic explanation for a link between active TG2 secretion and inflammatory responses, and aberrant enhanced TG2 activity in certain autoimmune conditions. Summary: Purinergic signaling regulates unconventional secretion of transglutaminase-2 (TG2) and explains the link between aberrant protein modifications and inflammatory responses in TG2-dependent autoimmunity.
Collapse
Affiliation(s)
- Magdalena Adamczyk
- Matrix Biology & Tissue Repair Research Unit and Arthritis Research UK Biomechanics and Bioengineering Center of Excellence, College of Biomedical and Life Sciences, Cardiff University, Cardiff CF14 4XY, UK
| | - Rhiannon Griffiths
- Matrix Biology & Tissue Repair Research Unit and Arthritis Research UK Biomechanics and Bioengineering Center of Excellence, College of Biomedical and Life Sciences, Cardiff University, Cardiff CF14 4XY, UK
| | - Sharon Dewitt
- Matrix Biology & Tissue Repair Research Unit and Arthritis Research UK Biomechanics and Bioengineering Center of Excellence, College of Biomedical and Life Sciences, Cardiff University, Cardiff CF14 4XY, UK
| | - Vera Knäuper
- Matrix Biology & Tissue Repair Research Unit and Arthritis Research UK Biomechanics and Bioengineering Center of Excellence, College of Biomedical and Life Sciences, Cardiff University, Cardiff CF14 4XY, UK
| | - Daniel Aeschlimann
- Matrix Biology & Tissue Repair Research Unit and Arthritis Research UK Biomechanics and Bioengineering Center of Excellence, College of Biomedical and Life Sciences, Cardiff University, Cardiff CF14 4XY, UK
| |
Collapse
|
484
|
Chowdhury R, Webber JP, Gurney M, Mason MD, Tabi Z, Clayton A. Cancer exosomes trigger mesenchymal stem cell differentiation into pro-angiogenic and pro-invasive myofibroblasts. Oncotarget 2015; 6:715-31. [PMID: 25596732 PMCID: PMC4359250 DOI: 10.18632/oncotarget.2711] [Citation(s) in RCA: 200] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 11/11/2014] [Indexed: 12/25/2022] Open
Abstract
Stromal fibroblasts become altered in response to solid cancers, to exhibit myofibroblastic characteristics, with disease promoting influence. Infiltrating mesenchymal stem cells (MSC) may contribute towards these changes, but the factors secreted by cancer cells that impact MSC differentiation are poorly understood. We investigated the role of nano-metre sized vesicles (exosomes), secreted by prostate cancer cells, on the differentiation of bone-marrow MSC (BM-MSC), and the subsequent functional consequences of such changes. Purified exosomes impaired classical adipogenic differentiation, skewing differentiation towards alpha-smooth muscle actin (αSMA) positive myofibroblastic cells. A single exosomes treatment generated myofibroblasts secreting high levels of VEGF-A, HGF and matrix regulating factors (MMP-1, −3 and −13). Differentiated MSC had pro-angiogenic functions and enhanced tumour proliferation and invasivity assessed in a 3D co-culture model. Differentiation was dependent on exosomal-TGFβ, but soluble TGFβ at matched dose could not generate the same phenotype. Exosomes present in the cancer cell secretome were the principal factors driving this phenotype. Prostate cancer exosomes dominantly dictate a programme of MSC differentiation generating myofibroblasts with functional properties consistent with disease promotion.
Collapse
Affiliation(s)
- Ridwana Chowdhury
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Velindre Cancer Centre, Whitchurch, Cardiff, United Kingdom, CF14 2TL
| | - Jason P Webber
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Velindre Cancer Centre, Whitchurch, Cardiff, United Kingdom, CF14 2TL.,Cardiff Institute for Tissue Engineering and Repair, Cardiff University
| | - Mark Gurney
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Velindre Cancer Centre, Whitchurch, Cardiff, United Kingdom, CF14 2TL
| | - Malcolm D Mason
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Velindre Cancer Centre, Whitchurch, Cardiff, United Kingdom, CF14 2TL
| | - Zsuzsanna Tabi
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Velindre Cancer Centre, Whitchurch, Cardiff, United Kingdom, CF14 2TL
| | - Aled Clayton
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Velindre Cancer Centre, Whitchurch, Cardiff, United Kingdom, CF14 2TL.Cardiff Institute for Tissue Engineering and Repair, Cardiff University.,Institute of Cancer and Genetics, School of Medicine, Cardiff University, Velindre Cancer Centre, Whitchurch, Cardiff, United Kingdom, CF14 2TL.Cardiff Institute for Tissue Engineering and Repair, Cardiff University
| |
Collapse
|
485
|
Sotillo J, Pearson M, Potriquet J, Becker L, Pickering D, Mulvenna J, Loukas A. Extracellular vesicles secreted by Schistosoma mansoni contain protein vaccine candidates. Int J Parasitol 2015; 46:1-5. [PMID: 26460238 DOI: 10.1016/j.ijpara.2015.09.002] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 09/26/2015] [Accepted: 09/28/2015] [Indexed: 12/18/2022]
Abstract
Herein we show for the first time that Schistosoma mansoni adult worms secrete exosome-like extracellular vesicles ranging from 50 to 130nm in size. Extracellular vesicles were collected from the excretory/secretory products of cultured adult flukes and purified by Optiprep density gradient, resulting in highly pure extracellular vesicle preparations as confirmed by transmission electron microscopy and Nanosight tracking analysis. Extracellular vesicle proteomic analysis showed numerous known vaccine candidates, potential virulence factors and molecules implicated in feeding. These findings provide new avenues for the exploration of host-schistosome interactions and offer a potential mechanism by which some vaccine antigens exert their protective efficacy.
Collapse
Affiliation(s)
- Javier Sotillo
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia.
| | - Mark Pearson
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia.
| | - Jeremy Potriquet
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Luke Becker
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Darren Pickering
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Jason Mulvenna
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Alex Loukas
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia.
| |
Collapse
|
486
|
Phinney DG, Di Giuseppe M, Njah J, Sala E, Shiva S, St Croix CM, Stolz DB, Watkins SC, Di YP, Leikauf GD, Kolls J, Riches DWH, Deiuliis G, Kaminski N, Boregowda SV, McKenna DH, Ortiz LA. Mesenchymal stem cells use extracellular vesicles to outsource mitophagy and shuttle microRNAs. Nat Commun 2015; 6:8472. [PMID: 26442449 PMCID: PMC4598952 DOI: 10.1038/ncomms9472] [Citation(s) in RCA: 669] [Impact Index Per Article: 74.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 08/26/2015] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) and macrophages are fundamental components of the stem cell niche and function coordinately to regulate haematopoietic stem cell self-renewal and mobilization. Recent studies indicate that mitophagy and healthy mitochondrial function are critical to the survival of stem cells, but how these processes are regulated in MSCs is unknown. Here we show that MSCs manage intracellular oxidative stress by targeting depolarized mitochondria to the plasma membrane via arrestin domain-containing protein 1-mediated microvesicles. The vesicles are then engulfed and re-utilized via a process involving fusion by macrophages, resulting in enhanced bioenergetics. Furthermore, we show that MSCs simultaneously shed micro RNA-containing exosomes that inhibit macrophage activation by suppressing Toll-like receptor signalling, thereby de-sensitizing macrophages to the ingested mitochondria. Collectively, these studies mechanistically link mitophagy and MSC survival with macrophage function, thereby providing a physiologically relevant context for the innate immunomodulatory activity of MSCs.
Collapse
Affiliation(s)
- Donald G Phinney
- Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Michelangelo Di Giuseppe
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, USA
| | - Joel Njah
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, USA
| | - Ernest Sala
- Hospital Son Espases, Palma Mallorca 07010, Spain
| | - Sruti Shiva
- Department of Pharmacology, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, USA
| | - Claudette M St Croix
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, USA.,Department of Pharmacology, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, USA.,Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, USA
| | - Donna B Stolz
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, USA
| | - Simon C Watkins
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, USA
| | - Y Peter Di
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, USA
| | - George D Leikauf
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, USA
| | - Jay Kolls
- Mellon Foundation Institute for Pediatric Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, USA
| | - David W H Riches
- Department of Pediatrics, National Jewish Health, Denver, Colorado 80206, USA
| | - Giuseppe Deiuliis
- Department of Medicine, Yale University, New Haven, Connecticut 06510, USA
| | - Naftali Kaminski
- Department of Medicine, Yale University, New Haven, Connecticut 06510, USA
| | - Siddaraju V Boregowda
- Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - David H McKenna
- Department of Laboratory Medicine and Pathology, University of Minnesota, Saint Paul, Minnesota 55108, USA
| | - Luis A Ortiz
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, USA
| |
Collapse
|
487
|
Systematic review of factors influencing extracellular vesicle yield from cell cultures. Cytotechnology 2015; 68:579-92. [PMID: 26433593 DOI: 10.1007/s10616-015-9913-6] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 08/16/2015] [Indexed: 12/28/2022] Open
Abstract
The potential therapeutic utility of extracellular vesicles (EVs) has spawned an interest into a scalable production, where the quantity and purity of EV samples is sufficient for clinical applications. EVs can be isolated using several different protocols; however, these isolation protocols and the subsequent methods of quantifying the resulting EV yield have not been sufficiently standardized. Therefore, the possibility of comparing different studies with respect to these parameters is limited. In this review, we have presented factors that might influence the yield and function of EVs from cell culture supernatants. The methods of isolation, downstream quantification, and culture conditions of the EV producing cells have been discussed. In order to examine the inter-study coherency of EV yields, 259 studies were initially screened, and 46 studies were included for extensive downstream analysis of EV yields where information pertaining to the isolation protocols and quantification methods was obtained from each study. Several other factors influencing yield were compared, such as cell type producing EVs, cell confluence level, and cell stimulation. In conclusion, various factors may impact the resulting EV yield, including technical aspects such as EV isolation and quantification procedures, and biological aspects such as cell type and culture conditions. The reflections presented in this review might aid in future standardization of the workflow in EV research.
Collapse
|
488
|
Muth DC, McAlexander MA, Ostrenga LJ, Pate NM, Izzi JM, Adams RJ, Pate KAM, Beck SE, Karim BO, Witwer KW. Potential role of cervicovaginal extracellular particles in diagnosis of endometriosis. BMC Vet Res 2015; 11:187. [PMID: 26253321 PMCID: PMC4529722 DOI: 10.1186/s12917-015-0513-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 07/28/2015] [Indexed: 01/11/2023] Open
Abstract
Background Macaques are an excellent model for many human diseases, including reproductive diseases such as endometriosis. A long-recognized need for early biomarkers of endometriosis has not yet resulted in consensus. While biomarker studies have examined many bodily fluids and targets, cervicovaginal secretions have been relatively under-investigated. Extracellular vesicles (EVs, including exosomes and microvesicles) are found in every biofluid examined, carry cargo including proteins and RNA, and may participate in intercellular signaling. Little is known about EVs in the cervicovaginal compartment, including the effects of reproductive tract disease on quantity and quality of EVs. Case presentation In September 2014, a 9-year-old rhesus macaque was diagnosed with endometriosis at The Johns Hopkins University School of Medicine. Ultrasound-guided fine needle aspiration of a cyst and subsequent laparotomy confirmed diagnosis. The animal was sent to necropsy following euthanasia for humane reasons. Perimortem vaginal swabs and cervicovaginal lavages were obtained. Using a combination of methods, including ultracentrifugation and NanoSight visualization technology, approximate numbers of EVs from each sample were calculated and compared to populations of EVs from other, reproductively normal macaques. Fewer EVs were recovered from the endometriosis samples as compared with those from reproductively healthy individuals. Conclusion To our knowledge, this is the first examination of EVs in primate cervicovaginal secretions, including those of a macaque with endometriosis. This case study suggests that additional research is justified to determine whether quantification of EVs—or their molecular cargo—in cervicovaginal lavage and vaginal swabs may provide a novel, relatively non-invasive diagnostic for primate endometrial disease or other reproductive tract diseases.
Collapse
Affiliation(s)
- Dillon C Muth
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Melissa A McAlexander
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Lauren J Ostrenga
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Nathan M Pate
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Jessica M Izzi
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Robert J Adams
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Kelly A Metcalf Pate
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Sarah E Beck
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Baktiar O Karim
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Kenneth W Witwer
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
489
|
Lobb RJ, Becker M, Wen SW, Wong CSF, Wiegmans AP, Leimgruber A, Möller A. Optimized exosome isolation protocol for cell culture supernatant and human plasma. J Extracell Vesicles 2015; 4:27031. [PMID: 26194179 PMCID: PMC4507751 DOI: 10.3402/jev.v4.27031] [Citation(s) in RCA: 1151] [Impact Index Per Article: 127.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 06/02/2015] [Accepted: 06/08/2015] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles represent a rich source of novel biomarkers in the diagnosis and prognosis of disease. However, there is currently limited information elucidating the most efficient methods for obtaining high yields of pure exosomes, a subset of extracellular vesicles, from cell culture supernatant and complex biological fluids such as plasma. To this end, we comprehensively characterize a variety of exosome isolation protocols for their efficiency, yield and purity of isolated exosomes. Repeated ultracentrifugation steps can reduce the quality of exosome preparations leading to lower exosome yield. We show that concentration of cell culture conditioned media using ultrafiltration devices results in increased vesicle isolation when compared to traditional ultracentrifugation protocols. However, our data on using conditioned media isolated from the Non-Small-Cell Lung Cancer (NSCLC) SK-MES-1 cell line demonstrates that the choice of concentrating device can greatly impact the yield of isolated exosomes. We find that centrifuge-based concentrating methods are more appropriate than pressure-driven concentrating devices and allow the rapid isolation of exosomes from both NSCLC cell culture conditioned media and complex biological fluids. In fact to date, no protocol detailing exosome isolation utilizing current commercial methods from both cells and patient samples has been described. Utilizing tunable resistive pulse sensing and protein analysis, we provide a comparative analysis of 4 exosome isolation techniques, indicating their efficacy and preparation purity. Our results demonstrate that current precipitation protocols for the isolation of exosomes from cell culture conditioned media and plasma provide the least pure preparations of exosomes, whereas size exclusion isolation is comparable to density gradient purification of exosomes. We have identified current shortcomings in common extracellular vesicle isolation methods and provide a potential standardized method that is effective, reproducible and can be utilized for various starting materials. We believe this method will have extensive application in the growing field of extracellular vesicle research.
Collapse
Affiliation(s)
- Richard J Lobb
- Tumour Microenvironment Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia.,School of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Melanie Becker
- Tumour Microenvironment Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Shu Wen Wen
- Tumour Microenvironment Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Christina S F Wong
- Tumour Microenvironment Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Adrian P Wiegmans
- Tumour Microenvironment Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Antoine Leimgruber
- Department of Medical Imaging, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland.,Service of Nuclear Medicine and Molecular Imaging, Department of Medical Imaging, University Hospital of Lausanne, Switzerland
| | - Andreas Möller
- Tumour Microenvironment Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia.,School of Medicine, University of Queensland, Brisbane, QLD, Australia;
| |
Collapse
|
490
|
Kim J, Tan Z, Lubman DM. Exosome enrichment of human serum using multiple cycles of centrifugation. Electrophoresis 2015; 36:2017-26. [PMID: 26010067 DOI: 10.1002/elps.201500131] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 03/12/2015] [Accepted: 05/06/2015] [Indexed: 12/21/2022]
Abstract
In this work, we compared the use of repeated cycles of centrifugation at conventional speeds for enrichment of exosomes from human serum compared to the use of ultracentrifugation (UC). After removal of cells and cell debris, a speed of 110 000 × g or 40 000 × g was used for the UC or centrifugation enrichment process, respectively. The enriched exosomes were analyzed using the bicinchoninic acid assay, 1D gel separation, transmission electron microscopy, Western blotting, and high-resolution LC-MS/MS analysis. It was found that a five-cycle repetition of UC or centrifugation is necessary for successful removal of nonexosomal proteins in the enrichment of exosomes from human serum. More significantly, 5× centrifugation enrichment was found to provide similar or better performance than 5× UC enrichment in terms of enriched exosome protein amount, Western blot band intensity for detection of CD-63, and numbers of identified exosome-related proteins and cluster of differentiation (CD) proteins. A total of 478 proteins were identified in the LC-MS/MS analyses of exosome proteins obtained from 5× UCs and 5× centrifugations including many important CD membrane proteins. The presence of previously reported exosome-related proteins including key exosome protein markers demonstrates the utility of this method for analysis of proteins in human serum.
Collapse
Affiliation(s)
- Jeongkwon Kim
- Department of Chemistry, Chungnam National University, Daejeon, Republic of Korea
| | - Zhijing Tan
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - David M Lubman
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI, USA
| |
Collapse
|
491
|
Huang T, Banizs AB, Shi W, Klibanov AL, He J. Size Exclusion HPLC Detection of Small-Size Impurities as a Complementary Means for Quality Analysis of Extracellular Vesicles. J Circ Biomark 2015; 4:6. [PMID: 28936242 PMCID: PMC5572986 DOI: 10.5772/61148] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 06/18/2015] [Indexed: 12/21/2022] Open
Abstract
For extracellular vesicle research, whether for biomarker discoveries or therapeutic applications, it is critical to have high-quality samples. Both microscopy and NanoSight Tracking Analysis (NTA) for size distribution have been used to detect large vesicles. However, there is currently no well-established method that is convenient for routine quality analysis of small-size impurities in vesicle samples. In this paper we report a convenient method, called 'size-exclusion high-performance liquid chromatography' (SE-HPLC), alongside NTA and Microscopy analysis to guide and qualify the isolation and processing of vesicles. First, the SE-HPLC analysis was used to detect impurities of small-size proteins during the ultra-centrifugation process of vesicle isolation; it was then employed to test the changes of vesicles under different pH conditions or integrity after storage. As SE-HPLC is generally accessible in most institutions, it could be used as a routine means to assist researchers in examining the integrity and quality of extracellular vesicles along with other techniques either during isolation/preparation or for further engineering and storage.
Collapse
Affiliation(s)
- Tao Huang
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, USA
| | - Anna B Banizs
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, USA
| | - Weibin Shi
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, USA
| | | | - Jiang He
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
492
|
Balaj L, Atai NA, Chen W, Mu D, Tannous BA, Breakefield XO, Skog J, Maguire CA. Heparin affinity purification of extracellular vesicles. Sci Rep 2015; 5:10266. [PMID: 25988257 PMCID: PMC4437317 DOI: 10.1038/srep10266] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 04/02/2015] [Indexed: 01/05/2023] Open
Abstract
Extracellular vesicles (EVs) are lipid membrane vesicles released by cells. They carry active biomolecules including DNA, RNA, and protein which can be transferred to recipient cells. Isolation and purification of EVs from culture cell media and biofluids is still a major challenge. The most widely used isolation method is ultracentrifugation (UC) which requires expensive equipment and only partially purifies EVs. Previously we have shown that heparin blocks EV uptake in cells, supporting a direct EV-heparin interaction. Here we show that EVs can be purified from cell culture media and human plasma using ultrafiltration (UF) followed by heparin-affinity beads. UF/heparin-purified EVs from cell culture displayed the EV marker Alix, contained a diverse RNA profile, had lower levels of protein contamination, and were functional at binding to and uptake into cells. RNA yield was similar for EVs isolated by UC. We were able to detect mRNAs in plasma samples with comparable levels to UC samples. In conclusion, we have discovered a simple, scalable, and effective method to purify EVs taking advantage of their heparin affinity.
Collapse
Affiliation(s)
- Leonora Balaj
- Department of Neurology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, MA
| | - Nadia A Atai
- 1] Department of Neurology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, MA. [2] Department of Cell Biology and Histology, Academic Medical Center (AMC), University of Amsterdam, The Netherlands
| | - Weilin Chen
- Department of Neurology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, MA
| | - Dakai Mu
- Department of Neurology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, MA
| | - Bakhos A Tannous
- Department of Neurology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, MA
| | - Xandra O Breakefield
- 1] Department of Neurology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, MA. [2] Departments of Radiology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, MA
| | | | - Casey A Maguire
- Department of Neurology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, MA
| |
Collapse
|
493
|
González E, Falcón-Pérez JM. Cell-derived extracellular vesicles as a platform to identify low-invasive disease biomarkers. Expert Rev Mol Diagn 2015; 15:907-23. [DOI: 10.1586/14737159.2015.1043272] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
494
|
Welton JL, Webber JP, Botos LA, Jones M, Clayton A. Ready-made chromatography columns for extracellular vesicle isolation from plasma. J Extracell Vesicles 2015; 4:27269. [PMID: 25819214 PMCID: PMC4376847 DOI: 10.3402/jev.v4.27269] [Citation(s) in RCA: 175] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 02/23/2015] [Accepted: 03/04/2015] [Indexed: 11/14/2022] Open
Abstract
Proteomic studies of circulating vesicles are hampered by difficulties in purifying vesicles from plasma and serum. Isolations are contaminated with high-abundance blood proteins that may mask genuine vesicular-associated proteins and/or simply provide misleading data. In this brief report, we explored the potential utility of a commercially available size exclusion chromatography column for rapid vesicle purification. We evaluated the performance of the column, with cancer cell line conditioned medium or healthy donor plasma, in terms of removing non-vesicular protein and enriching for vesicles exhibiting exosome characteristics. Serial fractions revealed a peak for typical exosomal proteins (CD9, CD81 etc.) that preceded the peak for highly abundant proteins, including albumin, for either sample type, and harvesting only this peak would represent elimination of >95% of protein from the sample. The columns showed good reproducibility, and streamlining the workflow would allow the exosome-relevant material to be collected in less than 10 minutes. Surprisingly, however, subsequent post-column vesicle concentration steps whilst resulting in some protein loss also lead to low vesicle recoveries, with a net effect of reducing sample purity (assessed by the particle-to-protein ratio). The columns provide a convenient, reproducible and highly effective means of eliminating >95% of non-vesicular protein from biological fluid samples such as plasma.
Collapse
Affiliation(s)
- Joanne Louise Welton
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Velindre Cancer Centre, Whitchurch, Cardiff, United Kingdom
| | - Jason Paul Webber
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Velindre Cancer Centre, Whitchurch, Cardiff, United Kingdom
| | | | | | - Aled Clayton
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Velindre Cancer Centre, Whitchurch, Cardiff, United Kingdom;
| |
Collapse
|
495
|
Ridder K, Sevko A, Heide J, Dams M, Rupp AK, Macas J, Starmann J, Tjwa M, Plate KH, Sültmann H, Altevogt P, Umansky V, Momma S. Extracellular vesicle-mediated transfer of functional RNA in the tumor microenvironment. Oncoimmunology 2015; 4:e1008371. [PMID: 26155418 PMCID: PMC4485784 DOI: 10.1080/2162402x.2015.1008371] [Citation(s) in RCA: 211] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 01/09/2015] [Accepted: 01/10/2015] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles (EVs) have been shown to transfer various molecules, including functional RNA between cells and this process has been suggested to be particularly relevant in tumor-host interactions. However, data on EV-mediated RNA transfer has been obtained primarily by in vitro experiments or involving ex vivo manipulations likely affecting its biology, leaving their physiological relevance unclear. We engineered glioma and carcinoma tumor cells to express Cre recombinase showing their release of EVs containing Cre mRNA in various EV subfractions including exosomes. Transplantation of these genetically modified tumor cells into mice with a Cre reporter background leads to frequent recombination events at the tumor site. In both tumor models the majority of recombined cells are CD45+ leukocytes, predominantly Gr1+CD11b+ myeloid-derived suppressor cells (MDSCs). In addition, multiple lineages of recombined cells can be observed in the glioma model. In the lung carcinoma model, recombined MDSCs display an enhanced immunosuppressive phenotype and an altered miRNA profile compared to their non-recombined counterparts. Cre-lox based tracing of tumor EV RNA transfer in vivo can therefore be used to identify individual target cells in the tumor microenvironment for further mechanistical or functional analysis.
Collapse
Affiliation(s)
- Kirsten Ridder
- Institute of Neurology (Edinger Institute); Frankfurt University Medical School; German Cancer Consortium (DKTK); German Cancer Research Center (DKFZ) ; Frankfurt, Heidelberg, Germany
| | - Alexandra Sevko
- Skin Cancer Unit; German Cancer Research Center; Heidelberg and Department of Dermatology, Venereology and Allergology; University Medical Center Mannheim; Ruprecht-Karl University of Heidelberg ; Mannheim, Heidelberg, Germany
| | - Janina Heide
- Institute of Neurology (Edinger Institute); Frankfurt University Medical School; German Cancer Consortium (DKTK); German Cancer Research Center (DKFZ) ; Frankfurt, Heidelberg, Germany
| | - Maria Dams
- Institute of Neurology (Edinger Institute); Frankfurt University Medical School; German Cancer Consortium (DKTK); German Cancer Research Center (DKFZ) ; Frankfurt, Heidelberg, Germany
| | - Anne-Kathleen Rupp
- Tumor Immunology Program; German Cancer Research Center ; Heidelberg, Germany
| | - Jadranka Macas
- Institute of Neurology (Edinger Institute); Frankfurt University Medical School; German Cancer Consortium (DKTK); German Cancer Research Center (DKFZ) ; Frankfurt, Heidelberg, Germany
| | - Julia Starmann
- Division of Molecular Genome Analysis; German Cancer Research Center ; Heidelberg, Germany
| | - Marc Tjwa
- Laboratory of Vascular Hematology/Angiogenesis; Institute for Transfusion Medicine; Frankfurt University Medical School ; Frankfurt, Germany
| | - Karl H Plate
- Institute of Neurology (Edinger Institute); Frankfurt University Medical School; German Cancer Consortium (DKTK); German Cancer Research Center (DKFZ) ; Frankfurt, Heidelberg, Germany
| | - Holger Sültmann
- Division of Molecular Genome Analysis; German Cancer Research Center ; Heidelberg, Germany
| | - Peter Altevogt
- Tumor Immunology Program; German Cancer Research Center ; Heidelberg, Germany
| | - Viktor Umansky
- Skin Cancer Unit; German Cancer Research Center; Heidelberg and Department of Dermatology, Venereology and Allergology; University Medical Center Mannheim; Ruprecht-Karl University of Heidelberg ; Mannheim, Heidelberg, Germany
| | - Stefan Momma
- Institute of Neurology (Edinger Institute); Frankfurt University Medical School; German Cancer Consortium (DKTK); German Cancer Research Center (DKFZ) ; Frankfurt, Heidelberg, Germany
| |
Collapse
|
496
|
Rani S, Ryan AE, Griffin MD, Ritter T. Mesenchymal Stem Cell-derived Extracellular Vesicles: Toward Cell-free Therapeutic Applications. Mol Ther 2015; 23:812-823. [PMID: 25868399 DOI: 10.1038/mt.2015.44] [Citation(s) in RCA: 808] [Impact Index Per Article: 89.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 02/20/2015] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem (stromal) cells (MSCs) are multipotent cells with the ability to differentiate into several cell types, thus serving as a cell reservoir for regenerative medicine. Much of the current interest in therapeutic application of MSCs to various disease settings can be linked to their immunosuppressive and anti-inflammatory properties. One of the key mechanisms of MSC anti-inflammatory effects is the secretion of soluble factors with paracrine actions. Recently it has emerged that the paracrine functions of MSCs could, at least in part, be mediated by extracellular vesicles (EVs). EVs are predominantly released from the endosomal compartment and contain a cargo that includes miRNA, mRNA, and proteins from their cells of origin. Recent animal model-based studies suggest that EVs have significant potential as a novel alternative to whole cell therapies. Compared to their parent cells, EVs may have a superior safety profile and can be safely stored without losing function. In this article, we review current knowledge related to the potential use of MSC-derived EVs in various diseases and discuss the promising future for EVs as an alternative, cell-free therapy.
Collapse
Affiliation(s)
- Sweta Rani
- Regenerative Medicine Institute (REMEDI), College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland.
| | - Aideen E Ryan
- Discipline of Pharmacology and Therapeutics, College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland
| | - Matthew D Griffin
- Regenerative Medicine Institute (REMEDI), College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland
| | - Thomas Ritter
- Regenerative Medicine Institute (REMEDI), College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland
| |
Collapse
|
497
|
Maiolo D, Paolini L, Di Noto G, Zendrini A, Berti D, Bergese P, Ricotta D. Colorimetric nanoplasmonic assay to determine purity and titrate extracellular vesicles. Anal Chem 2015; 87:4168-76. [PMID: 25674701 DOI: 10.1021/ac504861d] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Extracellular Vesicles (EVs) - cell secreted vesicles that carry rich molecular information of the parental cell and constitute an important mode of intercellular communication - are becoming a primary topic in translational medicine. EVs (that comprise exosomes and microvesicles/microparticles) have a size ranging from 40 nm to 1 μm and share several physicochemical proprieties, including size, density, surface charge, and light interaction, with other nano-objects present in body fluids, such as single and aggregated proteins. This makes separation, titration, and characterization of EVs challenging and time-consuming. Here we present a cost-effective and fast colorimetric assay for probing by eye protein contaminants and determine the concentration of EV preparations, which exploits the synergy between colloidal gold nanoplasmonics, nanoparticle-protein corona, and nanoparticle-membrane interaction. The assay hits a limit of detection of protein contaminants of 5 ng/μL and has a dynamic range of EV concentration ranging from 35 fM to 35 pM, which matches the typical range of EV concentration in body fluids. This work provides the first example of the exploitation of the nanoparticle-protein corona in analytical chemistry.
Collapse
Affiliation(s)
- Daniele Maiolo
- †Chemistry for Technologies Laboratory and INSTM, Department of Mechanical and Industrial Engineering, University of Brescia, via Branze 38, 25123 Brescia, Brescia, Italy
| | - Lucia Paolini
- ‡Department of Molecular and Translational Medicine, Faculty of Medicine, University of Brescia, 25123 Brescia, Brescia, Italy
| | - Giuseppe Di Noto
- ‡Department of Molecular and Translational Medicine, Faculty of Medicine, University of Brescia, 25123 Brescia, Brescia, Italy
| | - Andrea Zendrini
- ‡Department of Molecular and Translational Medicine, Faculty of Medicine, University of Brescia, 25123 Brescia, Brescia, Italy
| | - Debora Berti
- §Department of Chemistry "Ugo Schiff" and CSGI, University of Florence, via della Lastruccia 3, 50019 Sesto Fiorentino, Florence Italy
| | - Paolo Bergese
- †Chemistry for Technologies Laboratory and INSTM, Department of Mechanical and Industrial Engineering, University of Brescia, via Branze 38, 25123 Brescia, Brescia, Italy
| | - Doris Ricotta
- ‡Department of Molecular and Translational Medicine, Faculty of Medicine, University of Brescia, 25123 Brescia, Brescia, Italy
| |
Collapse
|
498
|
Sáenz-Cuesta M, Arbelaiz A, Oregi A, Irizar H, Osorio-Querejeta I, Muñoz-Culla M, Banales JM, Falcón-Pérez JM, Olascoaga J, Otaegui D. Methods for extracellular vesicles isolation in a hospital setting. Front Immunol 2015; 6:50. [PMID: 25762995 PMCID: PMC4327731 DOI: 10.3389/fimmu.2015.00050] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 01/27/2015] [Indexed: 12/27/2022] Open
Abstract
The research in extracellular vesicles (EVs) has been rising during the last decade. However, there is no clear consensus on the most accurate protocol to isolate and analyze them. Besides, most of the current protocols are difficult to implement in a hospital setting due to being very time-consuming or to requirements of specific infrastructure. Thus, our aim is to compare five different protocols (comprising two different medium-speed differential centrifugation protocols; commercially polymeric precipitation – exoquick – acid precipitation; and ultracentrifugation) for blood and urine samples to determine the most suitable one for the isolation of EVs. Nanoparticle tracking analysis, flow cytometry, western blot (WB), electronic microscopy, and spectrophotometry were used to characterize basic aspects of EVs such as concentration, size distribution, cell-origin and transmembrane markers, and RNA concentration. The highest EV concentrations were obtained using the exoquick protocol, followed by both differential centrifugation protocols, while the ultracentrifugation and acid-precipitation protocols yielded considerably lower EV concentrations. The five protocols isolated EVs of similar characteristics regarding markers and RNA concentration; however, standard protocol recovered only small EVs. EV isolated with exoquick presented difficult to be analyzed with WB. The RNA concentrations obtained from urine-derived EVs were similar to those obtained from blood-derived ones, despite the urine EV concentration being 10–20 times lower. We consider that a medium-speed differential centrifugation could be suitable to be applied in a hospital setting as it requires the simplest infrastructure and recovers higher concentration of EV than standard protocol. A workflow from sampling to characterization of EVs is proposed.
Collapse
Affiliation(s)
- Matías Sáenz-Cuesta
- Multiple Sclerosis Unit, Neuroscience Area, Biodonostia Health Research Institute , San Sebastián , Spain ; Spanish Network on Multiple Sclerosis , Madrid , Spain
| | - Ander Arbelaiz
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute, Donostia University Hospital , San Sebastián , Spain ; University of the Basque Country , San Sebastián , Spain
| | - Amaia Oregi
- Multiple Sclerosis Unit, Neuroscience Area, Biodonostia Health Research Institute , San Sebastián , Spain
| | - Haritz Irizar
- Multiple Sclerosis Unit, Neuroscience Area, Biodonostia Health Research Institute , San Sebastián , Spain ; Spanish Network on Multiple Sclerosis , Madrid , Spain
| | - Iñaki Osorio-Querejeta
- Multiple Sclerosis Unit, Neuroscience Area, Biodonostia Health Research Institute , San Sebastián , Spain ; Spanish Network on Multiple Sclerosis , Madrid , Spain
| | - Maider Muñoz-Culla
- Multiple Sclerosis Unit, Neuroscience Area, Biodonostia Health Research Institute , San Sebastián , Spain ; Spanish Network on Multiple Sclerosis , Madrid , Spain
| | - Jesus M Banales
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute, Donostia University Hospital , San Sebastián , Spain ; University of the Basque Country , San Sebastián , Spain ; National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, Instituto de Salud Carlos III) , Madrid , Spain ; Ikerbasque - Basque Foundation for Science , Bilbao , Spain ; Asociación Española Contra el Cáncer , Madrid , Spain
| | - Juan M Falcón-Pérez
- National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, Instituto de Salud Carlos III) , Madrid , Spain ; Ikerbasque - Basque Foundation for Science , Bilbao , Spain ; Metabolomics Unit, CIC bioGUNE , Derio , Spain
| | - Javier Olascoaga
- Multiple Sclerosis Unit, Neuroscience Area, Biodonostia Health Research Institute , San Sebastián , Spain ; Spanish Network on Multiple Sclerosis , Madrid , Spain ; Department of Neurology, Donostia University Hospital , San Sebastián , Spain
| | - David Otaegui
- Multiple Sclerosis Unit, Neuroscience Area, Biodonostia Health Research Institute , San Sebastián , Spain ; Spanish Network on Multiple Sclerosis , Madrid , Spain
| |
Collapse
|
499
|
Webber J, Yeung V, Clayton A. Extracellular vesicles as modulators of the cancer microenvironment. Semin Cell Dev Biol 2015; 40:27-34. [PMID: 25662446 DOI: 10.1016/j.semcdb.2015.01.013] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 01/22/2015] [Accepted: 01/28/2015] [Indexed: 12/14/2022]
Abstract
The tumour microenvironment is a highly complex and dynamic tissue. It comprises not only neoplastic cells, but also other resident cells within the milieu such as stroma and vascular cells in addition to a variable cellular infiltrate from the periphery. A host of soluble factors such as growth factors, chemokines, eicosanoids soluble metabolites and extracellular matrix components have been extensively documented as factors which modulate this environment. However, in recent years there has also been growing interests in the potential roles of extracellular vesicles (EV) in many of the processes governing the nature of cancerous tissue. In this brief review, we have assembled evidence describing several distinct functions for extracellular vesicles in modulating the microenvironment with examples that include immune evasion, angiogenesis and stromal activation. Whilst there remains a great deal to be learnt about the interplay between vesicles and the cancerous environment, it is becoming clear that vesicle-mediated communication has a major influence on key aspects of cancer growth and progression. We conclude that the design of future therapeutics should acknowledge the existence and roles of extracellular vesicles, and seriously consider strategies for circumventing their effects in vivo.
Collapse
Affiliation(s)
- Jason Webber
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Velindre Cancer Centre, Whitchurch, Cardiff CF14 2TL, United Kingdom
| | - Vincent Yeung
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Velindre Cancer Centre, Whitchurch, Cardiff CF14 2TL, United Kingdom
| | - Aled Clayton
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Velindre Cancer Centre, Whitchurch, Cardiff CF14 2TL, United Kingdom.
| |
Collapse
|
500
|
Nordin JZ, Lee Y, Vader P, Mäger I, Johansson HJ, Heusermann W, Wiklander OPB, Hällbrink M, Seow Y, Bultema JJ, Gilthorpe J, Davies T, Fairchild PJ, Gabrielsson S, Meisner-Kober NC, Lehtiö J, Smith CIE, Wood MJA, El Andaloussi S. Ultrafiltration with size-exclusion liquid chromatography for high yield isolation of extracellular vesicles preserving intact biophysical and functional properties. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 11:879-83. [PMID: 25659648 DOI: 10.1016/j.nano.2015.01.003] [Citation(s) in RCA: 444] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 12/22/2014] [Accepted: 01/04/2015] [Indexed: 12/12/2022]
Abstract
UNLABELLED Extracellular vesicles (EVs) are natural nanoparticles that mediate intercellular transfer of RNA and proteins and are of great medical interest; serving as novel biomarkers and potential therapeutic agents. However, there is little consensus on the most appropriate method to isolate high-yield and high-purity EVs from various biological fluids. Here, we describe a systematic comparison between two protocols for EV purification: ultrafiltration with subsequent liquid chromatography (UF-LC) and differential ultracentrifugation (UC). A significantly higher EV yield resulted from UF-LC as compared to UC, without affecting vesicle protein composition. Importantly, we provide novel evidence that, in contrast to UC-purified EVs, the biophysical properties of UF-LC-purified EVs are preserved, leading to a different in vivo biodistribution, with less accumulation in lungs. Finally, we show that UF-LC is scalable and adaptable for EV isolation from complex media types such as stem cell media, which is of huge significance for future clinical applications involving EVs. FROM THE CLINICAL EDITOR Recent evidence suggests extracellular vesicles (EVs) as another route of cellular communication. These EVs may be utilized for future therapeutics. In this article, the authors compared ultrafiltration with size-exclusion liquid chromatography (UF-LC) and ultra-centrifugation (UC) for EV recovery.
Collapse
Affiliation(s)
- Joel Z Nordin
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Yi Lee
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Pieter Vader
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Imre Mäger
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom; Institute of Technology, University of Tartu, Tartu, Estonia
| | - Henrik J Johansson
- Cancer Proteomics Mass Spectrometry, Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Wolf Heusermann
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | | | - Mattias Hällbrink
- Department of Neurochemistry, Stockholm University, Stockholm, Sweden
| | - Yiqi Seow
- Molecular Engineering Laboratory, Proteos, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Jarred J Bultema
- Department of Medicine Solna, Translational Immunology Unit, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | | | - Tim Davies
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Paul J Fairchild
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Susanne Gabrielsson
- Department of Medicine Solna, Translational Immunology Unit, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | | | - Janne Lehtiö
- Cancer Proteomics Mass Spectrometry, Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - C I Edvard Smith
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Matthew J A Wood
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom.
| | - Samir El Andaloussi
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|