551
|
Cheruiyot C, Pataki Z, Ramratnam B, Li M. Proteomic Analysis of Exosomes and Its Application in HIV-1 Infection. Proteomics Clin Appl 2018; 12:e1700142. [PMID: 29687643 DOI: 10.1002/prca.201700142] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 03/23/2018] [Indexed: 12/21/2022]
Abstract
Exosomes are 30-100 nm extracellular vesicles secreted from late endosomes by various types of cells. Numerous studies have suggested that exosomes play significant roles in human immunodeficiency virus 1 (HIV-1) biogenesis. Proteomics coupled with exosome fractionation has been successfully used to identify various exosomal proteins and helped to uncover the interactions between exosomes and HIV-1. To inform the current progress in the intersection of exosome, proteomics, and HIV-1, this review is focused on: i) analyzing different exosome isolation, purification methods, and their implications in HIV-1 studies; ii) evaluating the roles of various proteomic techniques in defining exosomal contents; iii) discussing the research and clinical applications of proteomics and exosome in HIV-1 biology.
Collapse
Affiliation(s)
- Collins Cheruiyot
- Department of Medicine, Division of Infectious Diseases, Laboratory of Retrovirology, Alpert Medical School of Brown University, Providence, RI, 02903, USA
| | - Zemplen Pataki
- Department of Medicine, Division of Infectious Diseases, Laboratory of Retrovirology, Alpert Medical School of Brown University, Providence, RI, 02903, USA
| | - Bharat Ramratnam
- Department of Medicine, Division of Infectious Diseases, Laboratory of Retrovirology, Alpert Medical School of Brown University, Providence, RI, 02903, USA.,Centers of Biomedical Research Excellence, Center for Cancer Research, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI, 02903, USA.,Clinical Research Center of Lifespan, Providence, RI, 02903, USA
| | - Ming Li
- Department of Medicine, Division of Infectious Diseases, Laboratory of Retrovirology, Alpert Medical School of Brown University, Providence, RI, 02903, USA
| |
Collapse
|
552
|
Muhsin-Sharafaldine MR, McLellan AD. Tumor-Derived Apoptotic Vesicles: With Death They Do Part. Front Immunol 2018; 9:957. [PMID: 29780392 PMCID: PMC5952256 DOI: 10.3389/fimmu.2018.00957] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/17/2018] [Indexed: 12/21/2022] Open
Abstract
Tumor cells release lipid particles known as extracellular vesicles (EV) that contribute to cancer metastasis, to the immune response, and to thrombosis. When tumors are exposed to radiation or chemotherapy, apoptotic vesicles (ApoVs) are released in abundance as the plasma membrane delaminates from the cytoskeleton. Recent studies have suggested that ApoVs are distinct from the EVs released from living cells, such as exosomes or microvesicles. Depending on their treatment conditions, tumor-released ApoV have been suggested to either enhance or suppress anti-cancer immunity. In addition, tumor-derived ApoV possess procoagulant activity that could increase the thrombotic state in cancer patients undergoing chemotherapy or radiotherapy. Since ApoVs are one of the least appreciated type of EVs, we focus in this review on the distinctive characterization of tumor ApoVs and their proposed mechanistic effects on cancer immunity, coagulation, and metastasis.
Collapse
Affiliation(s)
| | - Alexander D McLellan
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
553
|
Cancer-derived exosomes from HER2-positive cancer cells carry trastuzumab-emtansine into cancer cells leading to growth inhibition and caspase activation. BMC Cancer 2018; 18:504. [PMID: 29720111 PMCID: PMC5930687 DOI: 10.1186/s12885-018-4418-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 04/22/2018] [Indexed: 12/24/2022] Open
Abstract
Background Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate that carries a cytotoxic drug (DM1) to HER2-positive cancer. The target of T-DM1 (HER2) is present also on cancer-derived exosomes. We hypothesized that exosome-bound T-DM1 may contribute to the activity of T-DM1. Methods Exosomes were isolated from the cell culture medium of HER2-positive SKBR-3 and EFM-192A breast cancer cells, HER2-positive SNU-216 gastric cancer cells, and HER2-negative MCF-7 breast cancer cells by serial centrifugations including two ultracentrifugations, and treated with T-DM1. T-DM1 not bound to exosomes was removed using HER2-coated magnetic beads. Exosome samples were analyzed by electron microscopy, flow cytometry and Western blotting. Binding of T-DM1-containing exosomes to cancer cells and T-DM1 internalization were investigated with confocal microscopy. Effects of T-DM1-containg exosomes on cancer cells were investigated with the AlamarBlue cell proliferation assay and the Caspase-Glo 3/7 caspase activation assay. Results T-DM1 binds to exosomes derived from HER2-positive cancer cells, but not to exosomes derived from HER2-negative MCF-7 cells. HER2-positive SKBR-3 cells accumulated T-DM1 after being treated with T-DM1-containg exosomes, and treatment of SKBR-3 and EFM-192A cells with T-DM1-containing exosomes resulted in growth inhibition and activation of caspases 3 and/or 7. Conclusion T-DM1 binds to exosomes derived from HER2-positive cancer cells, and T-DM1 may be carried to other cancer cells via exosomes leading to reduced viability of the recipient cells. The results suggest a new mechanism of action for T-DM1, mediated by exosomes derived from HER2-positive cancer. Electronic supplementary material The online version of this article (10.1186/s12885-018-4418-2) contains supplementary material, which is available to authorized users.
Collapse
|
554
|
Chen J, Chopp M. Exosome Therapy for Stroke. Stroke 2018; 49:1083-1090. [PMID: 29669873 PMCID: PMC6028936 DOI: 10.1161/strokeaha.117.018292] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 08/28/2017] [Accepted: 08/30/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Jieli Chen
- From the Department of Neurology, Henry Ford Hospital, Detroit, MI (J.C., M.C.)
- Department of Geriatrics, Tianjin Medical University General Hospital, China (J.C.)
- Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education, China (J.C.)
| | - Michael Chopp
- From the Department of Neurology, Henry Ford Hospital, Detroit, MI (J.C., M.C.)
- Department of Physics, Oakland University, Rochester, MI (M.C.)
| |
Collapse
|
555
|
Sedgwick AE, D'Souza-Schorey C. The biology of extracellular microvesicles. Traffic 2018; 19:319-327. [PMID: 29479795 PMCID: PMC6922305 DOI: 10.1111/tra.12558] [Citation(s) in RCA: 170] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 02/16/2018] [Accepted: 02/16/2018] [Indexed: 12/11/2022]
Abstract
The study of extracellular vesicles (EVs) is a rapidly evolving field, owing in large part to recent advances in the realization of their significant contributions to normal physiology and disease. Once discredited as cell debris, these membrane vesicles have now emerged as mediators of intercellular communication by interaction with target cells, drug and gene delivery, and as potentially versatile platforms of clinical biomarkers as a result of their distinctive protein, nucleic acid and lipid cargoes. While there are multiple classes of EVs released from almost all cell types, here we focus primarily on the biogenesis, fate and functional cargoes of microvesicles (MVs). MVs regulate many important cellular processes including facilitating cell invasion, cell growth, evasion of immune response, stimulating angiogenesis, drug resistance and many others.
Collapse
Affiliation(s)
- Alanna E Sedgwick
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana
| | | |
Collapse
|
556
|
Augeri S, Capano S, Morone S, Fissolo G, Giacomino A, Peola S, Drace Z, Rapa I, Novello S, Volante M, Righi L, Ferrero E, Ortolan E, Funaro A. Soluble CD157 in pleural effusions: a complementary tool for the diagnosis of malignant mesothelioma. Oncotarget 2018; 9:22785-22801. [PMID: 29854315 PMCID: PMC5978265 DOI: 10.18632/oncotarget.25237] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 04/07/2018] [Indexed: 12/17/2022] Open
Abstract
Background CD157/Bst1 glycoprotein is expressed in >85% of malignant pleural mesotheliomas and is a marker of enhanced tumor aggressiveness. Results In vitro, mesothelial cells (malignant and non-malignant) released CD157 in soluble form or as an exosomal protein. In vivo, sCD157 is released and can be measured in pleural effusions by ELISA. Significantly higher levels of effusion sCD157 were detected in patients with malignant pleural mesothelioma than in patients with non-mesothelioma tumors or with non-malignant conditions. In our patient cohort, the area under the receiver-operating characteristic curve for sCD157 that discriminated malignant pleural mesothelioma from all other causes of pleural effusion was 0.685, cut-off (determined by the Youden Index) = 23.66 ng/ml (62.3% sensitivity; 73.93% specificity). Using a cut-off that yielded 95.58% specificity, measurement of sCD157 in cytology-negative effusions increased sensitivity of malignant pleural mesothelioma diagnosis from 34.42% to 49.18%. Conclusions Evaluation of soluble CD157 in pleural effusions provides a diagnostic aid in malignant mesothelioma. Methods Soluble CD157 (sCD157) was detected biochemically in culture supernatants of malignant and non-malignant mesothelial cells, and in pleural effusions from various pathological conditions. An ELISA system was established to measure the concentration of sCD157 in fluids, and extended to analyze sCD157 in pleural effusions from a cohort of 295 patients.
Collapse
Affiliation(s)
- Stefania Augeri
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, Torino 10126, Italy
| | - Stefania Capano
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, Torino 10126, Italy
| | - Simona Morone
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, Torino 10126, Italy
| | - Giulia Fissolo
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, Torino 10126, Italy
| | - Alice Giacomino
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, Torino 10126, Italy
| | - Silvia Peola
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, Torino 10126, Italy
| | - Zahida Drace
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, Torino 10126, Italy
| | - Ida Rapa
- Department of Oncology, University of Torino, San Luigi Hospital, Torino 10043, Italy
| | - Silvia Novello
- Department of Oncology, University of Torino, San Luigi Hospital, Torino 10043, Italy
| | - Marco Volante
- Department of Oncology, University of Torino, San Luigi Hospital, Torino 10043, Italy
| | - Luisella Righi
- Department of Oncology, University of Torino, San Luigi Hospital, Torino 10043, Italy
| | - Enza Ferrero
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, Torino 10126, Italy
| | - Erika Ortolan
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, Torino 10126, Italy
| | - Ada Funaro
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, Torino 10126, Italy
| |
Collapse
|
557
|
Cai C, Koch B, Morikawa K, Suda G, Sakamoto N, Rueschenbaum S, Akhras S, Dietz J, Hildt E, Zeuzem S, Welsch C, Lange CM. Macrophage-Derived Extracellular Vesicles Induce Long-Lasting Immunity Against Hepatitis C Virus Which Is Blunted by Polyunsaturated Fatty Acids. Front Immunol 2018; 9:723. [PMID: 29706960 PMCID: PMC5906748 DOI: 10.3389/fimmu.2018.00723] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 03/23/2018] [Indexed: 12/20/2022] Open
Abstract
Extracellular vesicles (EVs) are increasingly recognized as important mediators of intercellular communication. In this study, we aimed to further characterize the role of macrophage-derived EVs in immune responses against hepatitis C virus (HCV) and the potential of polyunsaturated fatty acids (PUFAs) to modulate this modality of innate immunity. To this end, EVs were isolated from interferon-stimulated macrophage cultures or from serum of patients with acute or chronic hepatitis C. EVs were characterized by electron microscopy, flow cytometry, RNA-sequencing, and Western blot analysis. The effect of EVs on replication of HCV was assessed in coculture models. Functional analyses were performed to assess the impact of PUFAs on EV-mediated antiviral immunity. We found that macrophages secreted various cytokines shortly after stimulation with type I and II IFN, which orchestrated a fast but short-lasting antiviral state. This rapid innate immune answer was followed by the production of macrophage-derived EVs, which induced a late, but long-lasting inhibitory effect on HCV replication. Of note, exposure of macrophages to PUFAs, which are important regulators of immune responses, dampened EV-mediated antiviral immune responses. Finally, EVs from patients with hepatitis C exhibited long-lasting antiviral activities during IFN therapy as well. The antiviral effect of EVs from Caucasian and Japanese patients differed, which may be explained by different nutritional uptake of PUFAs. In conclusion, our data indicate that macrophage-derived EVs mediate long-lasting inhibitory effects on HCV replication, which may bridge the time until efficient adaptive immune responses are established, and which can be blunted by PUFAs.
Collapse
Affiliation(s)
- Chengcong Cai
- Department of Medicine 1, Goethe University Hospital, Frankfurt am Main, Germany
| | - Benjamin Koch
- Department of Medicine 3, Goethe University Hospital, Frankfurt am Main, Germany
| | - Kenichi Morikawa
- Department of Gastroenterology and Hepatology, Hokkaido University Hospital, Hokkaido, Japan
| | - Goki Suda
- Department of Gastroenterology and Hepatology, Hokkaido University Hospital, Hokkaido, Japan
| | - Naoya Sakamoto
- Department of Gastroenterology and Hepatology, Hokkaido University Hospital, Hokkaido, Japan
| | - Sabrina Rueschenbaum
- Department of Medicine 1, Goethe University Hospital, Frankfurt am Main, Germany
| | - Sami Akhras
- Department of Virology, Paul-Ehrlich-Institut, Langen, Germany
| | - Julia Dietz
- Department of Medicine 1, Goethe University Hospital, Frankfurt am Main, Germany
| | - Eberhard Hildt
- Department of Virology, Paul-Ehrlich-Institut, Langen, Germany
| | - Stefan Zeuzem
- Department of Medicine 1, Goethe University Hospital, Frankfurt am Main, Germany
| | - Christoph Welsch
- Department of Medicine 1, Goethe University Hospital, Frankfurt am Main, Germany
| | - Christian M Lange
- Department of Medicine 1, Goethe University Hospital, Frankfurt am Main, Germany
| |
Collapse
|
558
|
Migration/Invasion of Malignant Gliomas and Implications for Therapeutic Treatment. Int J Mol Sci 2018; 19:ijms19041115. [PMID: 29642503 PMCID: PMC5979613 DOI: 10.3390/ijms19041115] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 03/22/2018] [Accepted: 04/03/2018] [Indexed: 02/07/2023] Open
Abstract
Malignant tumors of the central nervous system (CNS) are among cancers with the poorest prognosis, indicated by their association with tumors of high-level morbidity and mortality. Gliomas, the most common primary CNS tumors that arise from neuroglial stem or progenitor cells, have estimated annual incidence of 6.6 per 100,000 individuals in the USA, and 3.5 per 100,000 individuals in Taiwan. Tumor invasion and metastasis are the major contributors to the deaths in cancer patients. Therapeutic goals including cancer stem cells (CSC), phenotypic shifts, EZH2/AXL/TGF-β axis activation, miRNAs and exosomes are relevant to GBM metastasis to develop novel targeted therapeutics for GBM and other brain cancers. Herein, we highlight tumor metastasis in our understanding of gliomas, and illustrate novel exosome therapeutic approaches in glioma, thereby paving the way towards innovative therapies in neuro-oncology.
Collapse
|
559
|
Li K, Rodosthenous RS, Kashanchi F, Gingeras T, Gould SJ, Kuo LS, Kurre P, Lee H, Leonard JN, Liu H, Lombo TB, Momma S, Nolan JP, Ochocinska MJ, Pegtel DM, Sadovsky Y, Sánchez-Madrid F, Valdes KM, Vickers KC, Weaver AM, Witwer KW, Zeng Y, Das S, Raffai RL, Howcroft TK. Advances, challenges, and opportunities in extracellular RNA biology: insights from the NIH exRNA Strategic Workshop. JCI Insight 2018; 3:98942. [PMID: 29618663 DOI: 10.1172/jci.insight.98942] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Extracellular RNA (exRNA) has emerged as an important transducer of intercellular communication. Advancing exRNA research promises to revolutionize biology and transform clinical practice. Recent efforts have led to cutting-edge research and expanded knowledge of this new paradigm in cell-to-cell crosstalk; however, gaps in our understanding of EV heterogeneity and exRNA diversity pose significant challenges for continued development of exRNA diagnostics and therapeutics. To unravel this complexity, the NIH convened expert teams to discuss the current state of the science, define the significant bottlenecks, and brainstorm potential solutions across the entire exRNA research field. The NIH Strategic Workshop on Extracellular RNA Transport helped identify mechanistic and clinical research opportunities for exRNA biology and provided recommendations on high priority areas of research that will advance the exRNA field.
Collapse
Affiliation(s)
- Kang Li
- Division of Vascular and Endovascular Surgery, Department of Surgery, University of California, San Francisco, and Veterans Affairs Medical Center, San Francisco, California, USA
| | | | - Fatah Kashanchi
- Laboratory of Molecular Virology, National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, Virginia, USA
| | - Thomas Gingeras
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, USA
| | - Stephen J Gould
- Department of Biological Chemistry, Johns Hopkins University, Baltimore, Maryland, USA
| | - Lillian S Kuo
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Peter Kurre
- Doernbecher Children's Hospital, Department of Pediatrics and Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Hakho Lee
- Center for Systems Biology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Joshua N Leonard
- Department of Chemical and Biological Engineering, Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois, USA
| | - Huiping Liu
- Departments of Pharmacology and Medicine (Hematology and Oncology), Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Tania B Lombo
- NIH, Office of the Director, Environmental Influences on Child Health Outcomes Program, Bethesda, Maryland, USA
| | - Stefan Momma
- Institute of Neurology (Edinger Institute), Frankfurt University Medical School, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Frankfurt, Heidelberg, Germany
| | - John P Nolan
- Scintillon Institute, San Diego, California, USA
| | | | - D Michiel Pegtel
- Department of Pathology, Cancer Center Amsterdam, Vrije Universiteit (VU) University Medical Center, Amsterdam, The Netherlands
| | - Yoel Sadovsky
- Magee-Womens Research Institute, Department of Microbiology and Molecular Genetics, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Francisco Sánchez-Madrid
- Instituto de Investigación Sanitaria Princesa, Hospital Universitario de la Princesa, Universidad Autónoma de Madrid, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Kayla M Valdes
- National Center for Advancing Translational Science, Bethesda, Maryland, USA
| | - Kasey C Vickers
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Alissa M Weaver
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Kenneth W Witwer
- Department of Molecular and Comparative Pathobiology, Department of Neurology, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Yong Zeng
- Department of Chemistry, University of Kansas Cancer Center, Lawrence, Kansas, USA
| | - Saumya Das
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Robert L Raffai
- Division of Vascular and Endovascular Surgery, Department of Surgery, University of California, San Francisco, and Veterans Affairs Medical Center, San Francisco, California, USA
| | - T Kevin Howcroft
- Cancer Immunology, Hematology, and Etiology Branch, Division of Cancer Biology, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
560
|
Cho YE, Song BJ, Akbar M, Baek MC. Extracellular vesicles as potential biomarkers for alcohol- and drug-induced liver injury and their therapeutic applications. Pharmacol Ther 2018; 187:180-194. [PMID: 29621595 DOI: 10.1016/j.pharmthera.2018.03.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 01/18/2018] [Indexed: 02/06/2023]
Abstract
Extracellular vesicles (EVs) are small membranous vesicles originating from various cells and tissues, including the liver parenchymal hepatocytes and nonparenchymal cells such as Kupffer and stellate cells. Recently, the pathophysiological role of EVs, such as exosomes and microvesicles, has been increasingly recognized based on their properties of intercellular communications. These EVs travel through the circulating blood and interact with specific cells and then deliver their cargos such as nucleic acids and proteins into recipient cells. In addition, based on their stabilities, circulating EVs from body fluids such as blood, cerebrospinal fluid, urine, saliva, semen, breast milk and amniotic fluids are being studied as a valuable source of potential biomarkers for providing information about the physiological status of original cells or tissues. In addition, EVs are considered potential therapeutic agents due to their ability for intercellular communications between different cell types within the liver and between various organs through transfer of their cargos. In this review, we have briefly described recent advances in the characteristics and pathophysiological roles of EVs in alcoholic liver disease (ALD) or drug-induced liver injury (DILI) and discuss their advantages in the discovery of potential biomarkers and therapeutic agents.
Collapse
Affiliation(s)
- Young-Eun Cho
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Byoung-Joon Song
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Mohammed Akbar
- Division of Neuroscience and Behavior, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Moon-Chang Baek
- Department of Molecular Medicine, CMRI, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea.
| |
Collapse
|
561
|
Samanta S, Rajasingh S, Drosos N, Zhou Z, Dawn B, Rajasingh J. Exosomes: new molecular targets of diseases. Acta Pharmacol Sin 2018; 39:501-513. [PMID: 29219950 PMCID: PMC5888687 DOI: 10.1038/aps.2017.162] [Citation(s) in RCA: 291] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 09/12/2017] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles (EVs) comprise apoptotic bodies, microvesicles and exosomes, and they perform as key regulators in cell-to-cell communication in normal as well as diseased states. EVs contain natural cargo molecules, such as miRNA, mRNA and proteins, and transfer these functional cargos to neighboring cells or more distant cells through circulation. These functionally active molecules then affect distinct signaling cascades. The message conveyed to the recipient cells is dependent upon the composition of the EV, which is determined by the parent cell and the EV biogenesis. Because of their properties such as increased stability in circulation, biocompatibility, low immunogenicity and toxicity, EVs have drawn attention as attractive delivery systems for therapeutics. This review focuses on the functional use of exosomes in therapy and the potential advantages and challenges in using exosomes for therapeutic purposes.
Collapse
Affiliation(s)
- Saheli Samanta
- Cardiovascular Research Institute, Division of Cardiovascular Diseases, Department of Internal Medicine
| | - Sheeja Rajasingh
- Cardiovascular Research Institute, Division of Cardiovascular Diseases, Department of Internal Medicine
| | - Nicholas Drosos
- Cardiovascular Research Institute, Division of Cardiovascular Diseases, Department of Internal Medicine
| | - Zhigang Zhou
- Cardiovascular Research Institute, Division of Cardiovascular Diseases, Department of Internal Medicine
| | - Buddhadeb Dawn
- Cardiovascular Research Institute, Division of Cardiovascular Diseases, Department of Internal Medicine
| | - Johnson Rajasingh
- Cardiovascular Research Institute, Division of Cardiovascular Diseases, Department of Internal Medicine
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
562
|
MicroRNA Expression Levels Are Altered in the Cerebrospinal Fluid of Patients with Young-Onset Alzheimer's Disease. Mol Neurobiol 2018; 55:8826-8841. [PMID: 29603092 PMCID: PMC6208843 DOI: 10.1007/s12035-018-1032-x] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 02/12/2018] [Indexed: 12/19/2022]
Abstract
Clinical diagnosis of Alzheimer’s disease (AD) prior to the age of 65 years is classified as young-onset (YOAD), whereas diagnosis after the age of 65 years is considered late-onset (LOAD). Although rare autosomal mutations more commonly associate with YOAD, most YOAD and LOAD cases are sporadic. YOAD and LOAD share amyloid and tau pathology, but many YOAD patients show increased disease severity and rate of progression. The current study examined the microRNA (miRNA) expression profile from exosomes isolated from the cerebrospinal fluid (CSF) of YOAD patients with biomarker-confirmed AD. Results uncovered miR-16-5p, miR-125b-5p, miR-451a, and miR-605-5p as differentially expressed in the CSF-derived exosomes of YOAD patients when compared with healthy controls (HC). In a cohort of LOAD patients, miR-125b-5p, miR-451a, and miR-605-5p were similarly altered in expression, but miR-16-5p showed similar expression to control. Analysis of the mRNA targets of these miRNAs revealed transcripts enriched in biological processes relevant to the post-mortem posterior cingulate cortex transcriptome in YOAD from a previously published microarray study, including those related to neuron projections, synaptic signaling, metabolism, apoptosis, and the immune system. Hence, these miRNAs represent novel targets for uncovering disease mechanisms and for biomarker development in both YOAD and LOAD.
Collapse
|
563
|
Abstract
Plasma biomarker discovery necessitates a method for deep proteomic profiling, as well as for highly accurate quantification of the proteins in the sample. Furthermore, to obtain strong candidates for potential biomarkers, the method should be high throughput to enable a large scale analysis. Here we describe in detail PROMIS-Quan (PROteomics of MIcroparticles using Super-SILAC Quantification), a method for a simple and robust fractionation of the plasma samples by extraction of plasma microparticles, followed by SILAC-based relative and absolute quantification.
Collapse
Affiliation(s)
- Michal Harel
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel.
| | - Tamar Geiger
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel.
| |
Collapse
|
564
|
Manandhar S, Kothandan VK, Oh J, Yoo SH, Hwang J, Hwang SR. A pharmaceutical investigation into exosomes. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2018. [DOI: 10.1007/s40005-018-0391-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
565
|
Yang N, Li S, Li G, Zhang S, Tang X, Ni S, Jian X, Xu C, Zhu J, Lu M. The role of extracellular vesicles in mediating progression, metastasis and potential treatment of hepatocellular carcinoma. Oncotarget 2018; 8:3683-3695. [PMID: 27713136 PMCID: PMC5356911 DOI: 10.18632/oncotarget.12465] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 09/28/2016] [Indexed: 12/21/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a major cause of cancer-related death worldwide. As vectors for intercellular information exchange, the potential role of extracellular vesicles (EVs) in HCC formation, progression and therapy has been widely investigated. In this review, we explore the current status of the researches in this field. Altogether there is undeniable evidence that EVs play a crucial role in HCC development, metastasis. Moreover, EVs have shown great potential as drug delivery systems (DDSs) for the treatment of HCC. Exosomal miRNAs derived from HCC cells can enhance transformed cell growth in recipient cells by modulating the expression of transforming growth factor-β activated kinase-1(TAK1) and downstream signaling molecules. Furthermore, vacuolar protein sortin 4 homolog A(VPS4A) and insulin-like growth factor(IGF)-1 regulate exosome-mediated miRNAs transfer. Immune cells- derived EVs containing integrin αMβ2 or CD147 may facilitate HCC metastasis. In addition, EVs-mediated shuttle of long non-coding RNAs (lncRNAs), specifically linc- VLDLR and linc-ROR promote chemoresistance of malignant cells. Heat shock proteins (HSPs)-harboring exosomes derived from HCC tumor cells increase the antitumor effect of natural killer (NK) cells, thus enhancing HCC immunotherapy. Indeed, inhibition of HCC tumor growth has been associated with tumor cell-derived exosomes (TEX)-pulsed dentritic cells (DCs). Exosomes are also essential in liver metastasis during colorectal carcinoma (CRC) and pancreatic ductal adenocarcinomas (PDAC). Therefore, as nucleic acid and drug delivery vehicles, EVs show a tremendous potential for effective treatment against HCC.
Collapse
Affiliation(s)
- Naibin Yang
- Department of Infection and Liver Diseases, Ningbo First Hospital, Ningbo, China
| | - Shanshan Li
- Department of Infection and Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Institute of Liver Research, Wenzhou Medical University, Wenzhou, China
| | - Guoxiang Li
- Department of Infection and Liver Diseases, Ningbo First Hospital, Ningbo, China
| | - Shengguo Zhang
- Department of Infection and Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Institute of Liver Research, Wenzhou Medical University, Wenzhou, China
| | - Xinyue Tang
- Department of Infection and Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Institute of Liver Research, Wenzhou Medical University, Wenzhou, China
| | - Shunlan Ni
- Department of Infection and Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Institute of Liver Research, Wenzhou Medical University, Wenzhou, China
| | - Xiaomin Jian
- Department of The First Clinical Medical, Wenzhou Medical University, Wenzhou, China
| | - Cunlai Xu
- Department of Respiration, Lishui People's Hospital of Wenzhou Medical University, Lishui, China
| | - Jiayin Zhu
- Laboratory Animal Center, Wenzhou Medical University, Wenzhou, China
| | - Mingqin Lu
- Department of Infection and Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Institute of Liver Research, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
566
|
Beit‐Yannai E, Tabak S, Stamer WD. Physical exosome:exosome interactions. J Cell Mol Med 2018; 22:2001-2006. [PMID: 29377463 PMCID: PMC5824382 DOI: 10.1111/jcmm.13479] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 09/25/2017] [Indexed: 12/25/2022] Open
Abstract
Exosomes are extracellular nanovesicles that mediate a number of cellular processes, including intracellular signalling. There are many published examples of exosome-exosome dimers; however, their relevance has not been explored. Here, we propose that cells release exosomes to physically interact with incoming exosomes, forming dimers that we hypothesize attenuate incoming exosome-mediated signalling. We discuss experiments to test this hypothesis and potential relevance in health and disease.
Collapse
Affiliation(s)
- Elie Beit‐Yannai
- Clinical Biochemistry and Pharmacology departmentBen‐Gurion University of the NegevBeer‐ShevaIsrael
| | - Saray Tabak
- Clinical Biochemistry and Pharmacology departmentBen‐Gurion University of the NegevBeer‐ShevaIsrael
| | | |
Collapse
|
567
|
Guix FX, Corbett GT, Cha DJ, Mustapic M, Liu W, Mengel D, Chen Z, Aikawa E, Young-Pearse T, Kapogiannis D, Selkoe DJ, Walsh DM. Detection of Aggregation-Competent Tau in Neuron-Derived Extracellular Vesicles. Int J Mol Sci 2018; 19:E663. [PMID: 29495441 PMCID: PMC5877524 DOI: 10.3390/ijms19030663] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 02/10/2018] [Accepted: 02/20/2018] [Indexed: 11/17/2022] Open
Abstract
Progressive cerebral accumulation of tau aggregates is a defining feature of Alzheimer's disease (AD). A popular theory that seeks to explain the apparent spread of neurofibrillary tangle pathology proposes that aggregated tau is passed from neuron to neuron. Such a templated seeding process requires that the transferred tau contains the microtubule binding repeat domains that are necessary for aggregation. While it is not clear how a protein such as tau can move from cell to cell, previous reports have suggested that this may involve extracellular vesicles (EVs). Thus, measurement of tau in EVs may both provide insights on the molecular pathology of AD and facilitate biomarker development. Here, we report the use of sensitive immunoassays specific for full-length (FL) tau and mid-region tau, which we applied to analyze EVs from human induced pluripotent stem cell (iPSC)-derived neuron (iN) conditioned media, cerebrospinal fluid (CSF), and plasma. In each case, most tau was free-floating with a small component inside EVs. The majority of free-floating tau detected by the mid-region assay was not detected by our FL assays, indicating that most free-floating tau is truncated. Inside EVs, the mid-region assay also detected more tau than the FL assay, but the ratio of FL-positive to mid-region-positive tau was higher inside exosomes than in free solution. These studies demonstrate the presence of minute amounts of free-floating and exosome-contained FL tau in human biofluids. Given the potential for FL tau to aggregate, we conclude that further investigation of these pools of extracellular tau and how they change during disease is merited.
Collapse
Affiliation(s)
- Francesc X. Guix
- Laboratory for Neurodegenerative Disease Research, Ann Romney Center for Neurologic Diseases, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA; (F.X.G.); (G.T.C.); (D.J.C.); (W.L.); (D.M.); (Z.C.); (T.Y.-P.); (D.J.S.)
| | - Grant T. Corbett
- Laboratory for Neurodegenerative Disease Research, Ann Romney Center for Neurologic Diseases, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA; (F.X.G.); (G.T.C.); (D.J.C.); (W.L.); (D.M.); (Z.C.); (T.Y.-P.); (D.J.S.)
| | - Diana J. Cha
- Laboratory for Neurodegenerative Disease Research, Ann Romney Center for Neurologic Diseases, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA; (F.X.G.); (G.T.C.); (D.J.C.); (W.L.); (D.M.); (Z.C.); (T.Y.-P.); (D.J.S.)
| | - Maja Mustapic
- Laboratory of Neurosciences, National Institute on Aging, NIH, Baltimore, MD 21224, USA; (M.M.); (D.K.)
| | - Wen Liu
- Laboratory for Neurodegenerative Disease Research, Ann Romney Center for Neurologic Diseases, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA; (F.X.G.); (G.T.C.); (D.J.C.); (W.L.); (D.M.); (Z.C.); (T.Y.-P.); (D.J.S.)
| | - David Mengel
- Laboratory for Neurodegenerative Disease Research, Ann Romney Center for Neurologic Diseases, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA; (F.X.G.); (G.T.C.); (D.J.C.); (W.L.); (D.M.); (Z.C.); (T.Y.-P.); (D.J.S.)
| | - Zhicheng Chen
- Laboratory for Neurodegenerative Disease Research, Ann Romney Center for Neurologic Diseases, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA; (F.X.G.); (G.T.C.); (D.J.C.); (W.L.); (D.M.); (Z.C.); (T.Y.-P.); (D.J.S.)
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA;
| | - Tracy Young-Pearse
- Laboratory for Neurodegenerative Disease Research, Ann Romney Center for Neurologic Diseases, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA; (F.X.G.); (G.T.C.); (D.J.C.); (W.L.); (D.M.); (Z.C.); (T.Y.-P.); (D.J.S.)
| | - Dimitrios Kapogiannis
- Laboratory of Neurosciences, National Institute on Aging, NIH, Baltimore, MD 21224, USA; (M.M.); (D.K.)
| | - Dennis J. Selkoe
- Laboratory for Neurodegenerative Disease Research, Ann Romney Center for Neurologic Diseases, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA; (F.X.G.); (G.T.C.); (D.J.C.); (W.L.); (D.M.); (Z.C.); (T.Y.-P.); (D.J.S.)
| | - Dominic M. Walsh
- Laboratory for Neurodegenerative Disease Research, Ann Romney Center for Neurologic Diseases, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA; (F.X.G.); (G.T.C.); (D.J.C.); (W.L.); (D.M.); (Z.C.); (T.Y.-P.); (D.J.S.)
| |
Collapse
|
568
|
Gambara G, Gaebler M, Keilholz U, Regenbrecht CRA, Silvestri A. From Chemotherapy to Combined Targeted Therapeutics: In Vitro and in Vivo Models to Decipher Intra-tumor Heterogeneity. Front Pharmacol 2018; 9:77. [PMID: 29491834 PMCID: PMC5817069 DOI: 10.3389/fphar.2018.00077] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 01/23/2018] [Indexed: 12/15/2022] Open
Abstract
Recent advances in next-generation sequencing and other omics technologies capable to map cell fate provide increasing evidence on the crucial role of intra-tumor heterogeneity (ITH) for cancer progression. The different facets of ITH, from genomic to microenvironmental heterogeneity and the hierarchical cellular architecture originating from the cancer stem cell compartment, contribute to the range of tumor phenotypes. Decoding these complex data resulting from the analysis of tumor tissue complexity poses a challenge for developing novel therapeutic strategies that can counteract tumor evolution and cellular plasticity. To achieve this aim, the development of in vitro and in vivo cancer models that resemble the complexity of ITH is crucial in understanding the interplay of cells and their (micro)environment and, consequently, in testing the efficacy of new targeted treatments and novel strategies of tailoring combinations of treatments to the individual composition of the tumor. This challenging approach may be an important cornerstone in overcoming the development of pharmaco-resistances during multiple lines of treatment. In this paper, we report the latest advances in patient-derived 3D (PD3D) cell cultures and patient-derived tumor xenografts (PDX) as in vitro and in vivo models that can retain the genetic and phenotypic heterogeneity of the tumor tissue.
Collapse
Affiliation(s)
- Guido Gambara
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin, Berlin, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Manuela Gaebler
- Department of Interdisciplinary Oncology, HELIOS Klinikum Berlin-Buch GmbH, Berlin, Germany
| | - Ulrich Keilholz
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin, Berlin, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | | |
Collapse
|
569
|
C-Terminal Farnesylation of UCH-L1 Plays a Role in Transport of Epstein-Barr Virus Primary Oncoprotein LMP1 to Exosomes. mSphere 2018; 3:mSphere00030-18. [PMID: 29435490 PMCID: PMC5806207 DOI: 10.1128/msphere.00030-18] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 01/17/2018] [Indexed: 12/14/2022] Open
Abstract
Exosomes are small vesicles that cells secrete into the extracellular space, and there is increasing evidence that they have pivotal roles in cell-to-cell communication in malignancy. It is reported also that EBV-associated malignant cells, including those derived from nasopharyngeal carcinoma (NPC) and B-cell lymphoma, secrete exosomes. These EBV-related exosomes may contain viral products such as latent membrane protein 1 (LMP1) and may contribute to cancer progression. The aim of this study was to investigate the mechanism by which those viral products are loaded in exosomes. In this study, we show for the first time that ubiquitin C-terminal hydrolase-L1 (UCH-L1) and its C-terminal farnesylation, a posttranslational lipid modification, contribute to this mechanism. Our results also suggest that inhibition of UCH-L1 farnesylation is a potential therapeutic target against cancer metastasis and invasion. Increasing evidence shows that exosomes are key regulators in cancer cell-to-cell communication. Several reports on Epstein-Barr virus (EBV)-related malignancies demonstrate that latent membrane protein 1 (LMP1) secreted by exosomes derived from EBV- or LMP1-positive cells can promote cancer progression and metastasis. However, the mechanism by which LMP1 is loaded into exosomes is still poorly understood. Here, we examined whether the process of LMP1 loading into exosomes is linked to the multifunctional molecule of the ubiquitin system—ubiquitin C-terminal hydrolase-L1 (UCH-L1). For the first time, we demonstrate that LMP1 is physically associated with UCH-L1 and that directing of LMP1 to exosomes is mediated by C-terminal farnesylation of UCH-L1. Additionally, we found that the FTI-277 farnesyltransferase inhibitor reduces motility- and anchorage-independent growth of EBV-positive cells in functional assays. On the basis of our results, we conclude that C-terminal farnesylation of UCH-L1 is one of the key mechanisms by which LMP1 is sorted to exosomes. We hypothesize that inhibition of farnesylation with specific small-molecule inhibitors blocks exosome-mediated transfer of prometastatic molecules such as LMP1 during cancer cell-to-cell communications and thereby impedes the process of cancer invasion. IMPORTANCE Exosomes are small vesicles that cells secrete into the extracellular space, and there is increasing evidence that they have pivotal roles in cell-to-cell communication in malignancy. It is reported also that EBV-associated malignant cells, including those derived from nasopharyngeal carcinoma (NPC) and B-cell lymphoma, secrete exosomes. These EBV-related exosomes may contain viral products such as latent membrane protein 1 (LMP1) and may contribute to cancer progression. The aim of this study was to investigate the mechanism by which those viral products are loaded in exosomes. In this study, we show for the first time that ubiquitin C-terminal hydrolase-L1 (UCH-L1) and its C-terminal farnesylation, a posttranslational lipid modification, contribute to this mechanism. Our results also suggest that inhibition of UCH-L1 farnesylation is a potential therapeutic target against cancer metastasis and invasion.
Collapse
|
570
|
Mallardi A, Nuzziello N, Liguori M, Avolio C, Palazzo G. Counting of peripheral extracellular vesicles in Multiple Sclerosis patients by an improved nanoplasmonic assay and dynamic light scattering. Colloids Surf B Biointerfaces 2018; 168:134-142. [PMID: 29428682 DOI: 10.1016/j.colsurfb.2018.02.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 01/27/2018] [Accepted: 02/03/2018] [Indexed: 01/08/2023]
Abstract
Extracellular vesicles (EVs) are vesicles naturally secreted by the majority of human cells. Being composed by a closed phospholipid bilayer secluding proteins and RNAs they are used to transfer molecular information to other cells, thereby influencing the recipient cell functions. Despite the increasingly recognized relevance of EVs, the clarification of their physiological role is hampered by the lack of suitable analytical tools for their quantification and characterization. In this study, we have implemented a nanoplasmonic assay, previously proposed for the purity of the EV fractions, to achieve a robust analytical protocol in order to quantify the total phospholipid concentration (CPL) and the EVs number. We show how the coupling of the nanoplasmonic assay with serial dilutions of the unknown sample allows, by simple visual inspection, to detect deviations from the physiological EVs content. The use of a response that depends on the absorbance values at three wavelengths permits to reduce the limit of detection of CPL to 5 μM (total) and the limit of quantification to 35 μM. We also propose a method that takes into account the spread in EV size when the concentration of phospholipids is turned into a concentration of vesicles. The proposed analytical protocol is successfully applied to a small cohort of Multiple Sclerosis patients examined in different stages of their clinical diseases.
Collapse
Affiliation(s)
- Antonia Mallardi
- CNR-IPCF, National Research Council of Italy, Institute for the Chemical Physics Processes, Division of Bari, Bari, Italy
| | - Nicoletta Nuzziello
- National Research Council of Italy, Institute of Biomedical Technologies, Section of Bari, Bari, Italy; Department of Basic Sciences, Neurosciences and Sense Organs, University of Bari, Bari, Italy
| | - Maria Liguori
- National Research Council of Italy, Institute of Biomedical Technologies, Section of Bari, Bari, Italy
| | - Carlo Avolio
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Gerardo Palazzo
- Department of Chemistry and Center for colloid and surface science (CSGI), University of Bari, Bari, Italy; CNR-NANOTEC, National Research Council of Italy, Istitute of Nanotechnology, Bari, Italy.
| |
Collapse
|
571
|
Review: Microfluidics technologies for blood-based cancer liquid biopsies. Anal Chim Acta 2018; 1012:10-29. [PMID: 29475470 DOI: 10.1016/j.aca.2017.12.050] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 12/29/2017] [Accepted: 12/30/2017] [Indexed: 12/19/2022]
Abstract
Blood-based liquid biopsies provide a minimally invasive alternative to identify cellular and molecular signatures that can be used as biomarkers to detect early-stage cancer, predict disease progression, longitudinally monitor response to chemotherapeutic drugs, and provide personalized treatment options. Specific targets in blood that can be used for detailed molecular analysis to develop highly specific and sensitive biomarkers include circulating tumor cells (CTCs), exosomes shed from tumor cells, cell-free circulating tumor DNA (cfDNA), and circulating RNA. Given the low abundance of CTCs and other tumor-derived products in blood, clinical evaluation of liquid biopsies is extremely challenging. Microfluidics technologies for cellular and molecular separations have great potential to either outperform conventional methods or enable completely new approaches for efficient separation of targets from complex samples like blood. In this article, we provide a comprehensive overview of blood-based targets that can be used for analysis of cancer, review microfluidic technologies that are currently used for isolation of CTCs, tumor derived exosomes, cfDNA, and circulating RNA, and provide a detailed discussion regarding potential opportunities for microfluidics-based approaches in cancer diagnostics.
Collapse
|
572
|
Konoshenko MY, Lekchnov EA, Vlassov AV, Laktionov PP. Isolation of Extracellular Vesicles: General Methodologies and Latest Trends. BIOMED RESEARCH INTERNATIONAL 2018; 2018:8545347. [PMID: 29662902 PMCID: PMC5831698 DOI: 10.1155/2018/8545347] [Citation(s) in RCA: 784] [Impact Index Per Article: 112.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 12/28/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND Extracellular vesicles (EVs) play an essential role in the communication between cells and transport of diagnostically significant molecules. A wide diversity of approaches utilizing different biochemical properties of EVs and a lack of accepted protocols make data interpretation very challenging. SCOPE OF REVIEW This review consolidates the data on the classical and state-of-the-art methods for isolation of EVs, including exosomes, highlighting the advantages and disadvantages of each method. Various characteristics of individual methods, including isolation efficiency, EV yield, properties of isolated EVs, and labor consumption are compared. MAJOR CONCLUSIONS A mixed population of vesicles is obtained in most studies of EVs for all used isolation methods. The properties of an analyzed sample should be taken into account when planning an experiment aimed at studying and using these vesicles. The problem of adequate EVs isolation methods still remains; it might not be possible to develop a universal EV isolation method but the available protocols can be used towards solving particular types of problems. GENERAL SIGNIFICANCE With the wide use of EVs for diagnosis and therapy of various diseases the evaluation of existing methods for EV isolation is one of the key problems in modern biology and medicine.
Collapse
Affiliation(s)
- Maria Yu. Konoshenko
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Novosibirsk 630090, Russia
- Meshalkin Siberian Federal Biomedical Research Center, Ministry of Public Health of the Russian Federation, Novosibirsk 630055, Russia
| | - Evgeniy A. Lekchnov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Novosibirsk 630090, Russia
- Meshalkin Siberian Federal Biomedical Research Center, Ministry of Public Health of the Russian Federation, Novosibirsk 630055, Russia
| | - Alexander V. Vlassov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Pavel P. Laktionov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Novosibirsk 630090, Russia
- Meshalkin Siberian Federal Biomedical Research Center, Ministry of Public Health of the Russian Federation, Novosibirsk 630055, Russia
| |
Collapse
|
573
|
Ramirez MI, Amorim MG, Gadelha C, Milic I, Welsh JA, Freitas VM, Nawaz M, Akbar N, Couch Y, Makin L, Cooke F, Vettore AL, Batista PX, Freezor R, Pezuk JA, Rosa-Fernandes L, Carreira ACO, Devitt A, Jacobs L, Silva IT, Coakley G, Nunes DN, Carter D, Palmisano G, Dias-Neto E. Technical challenges of working with extracellular vesicles. NANOSCALE 2018; 10:881-906. [PMID: 29265147 DOI: 10.1039/c7nr08360b] [Citation(s) in RCA: 363] [Impact Index Per Article: 51.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Extracellular Vesicles (EVs) are gaining interest as central players in liquid biopsies, with potential applications in diagnosis, prognosis and therapeutic guidance in most pathological conditions. These nanosized particles transmit signals determined by their protein, lipid, nucleic acid and sugar content, and the unique molecular pattern of EVs dictates the type of signal to be transmitted to recipient cells. However, their small sizes and the limited quantities that can usually be obtained from patient-derived samples pose a number of challenges to their isolation, study and characterization. These challenges and some possible options to overcome them are discussed in this review.
Collapse
Affiliation(s)
- Marcel I Ramirez
- Fundação Instituto Oswaldo Cruz, Rio de Janeiro, RJ, Brazil and Universidade Federal do Paraná, Curitiba, PR, Brazil
| | | | - Catarina Gadelha
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Ivana Milic
- School of Life and Health Sciences, Aston University, England, UK
| | | | | | - Muhammad Nawaz
- Universidade de São Paulo, São Paulo, SP, Brazil and University of Gothenburg, Sweden
| | - Naveed Akbar
- Division of Cardiovascular Medicine, University of Oxford, Oxford, England, UK
| | - Yvonne Couch
- Acute Stroke Programme, RDM-Investigative Medicine, University of Oxford, Oxford, England, UK
| | - Laura Makin
- Sir William Dunn School of Pathology, University of Oxford, Oxford, England, UK
| | - Fiona Cooke
- University of St Andrews, St Andrews, Fife, Scotland, UK
| | - Andre L Vettore
- Federal University of São Paulo campus Diadema, Diadema, Brazil
| | | | | | - Julia A Pezuk
- Universidade Anhanguera de São Paulo, São Paulo, Brazil
| | - Lívia Rosa-Fernandes
- Universidade de São Paulo, São Paulo, SP, Brazil and University of Southern Denmark, Odense, Denmark
| | | | - Andrew Devitt
- School of Life and Health Sciences, Aston University, England, UK
| | | | | | - Gillian Coakley
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, Scotland, UK
| | - Diana N Nunes
- CIPE, A.C.Camargo Cancer Center, São Paulo, SP, Brazil.
| | - Dave Carter
- Oxford Brookes University, Oxford, England, UK
| | - Giuseppe Palmisano
- Universidade de São Paulo, São Paulo, SP, Brazil and IRCCS, Fondazione Santa Lucia, Rome, Italy
| | - Emmanuel Dias-Neto
- CIPE, A.C.Camargo Cancer Center, São Paulo, SP, Brazil. and Universidade de São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
574
|
Prospects in non-invasive assessment of liver fibrosis: Liquid biopsy as the future gold standard? Biochim Biophys Acta Mol Basis Dis 2018; 1864:1024-1036. [PMID: 29329986 DOI: 10.1016/j.bbadis.2018.01.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 01/04/2018] [Accepted: 01/07/2018] [Indexed: 12/11/2022]
Abstract
Liver fibrosis is the result of persistent liver injury, and is characterized by sustained scar formation and disruption of the normal liver architecture. The extent of fibrosis is considered as an important prognostic factor for the patient outcome, as an absence of (early) treatment can lead to the development of liver cirrhosis and hepatocellular carcinoma. Till date, the most sensitive and specific way for the diagnosis and staging of liver fibrosis remains liver biopsy, an invasive diagnostic tool, which is associated with high costs and discomfort for the patient. Over time, non-invasive scoring systems have been developed, of which the measurements of serum markers and liver stiffness are validated for use in the clinic. These tools lack however the sensitivity and specificity to detect small changes in the progression or regression of both early and late stages of fibrosis. Novel non-invasive diagnostic markers with the potential to overcome these limitations have been developed, but often lack validation in large patient cohorts. In this review, we will summarize novel trends in non-invasive markers of liver fibrosis development and will discuss their (dis-)advantages for use in the clinic.
Collapse
|
575
|
Qiu J, Yang G, Feng M, Zheng S, Cao Z, You L, Zheng L, Zhang T, Zhao Y. Extracellular vesicles as mediators of the progression and chemoresistance of pancreatic cancer and their potential clinical applications. Mol Cancer 2018; 17:2. [PMID: 29304816 PMCID: PMC5756395 DOI: 10.1186/s12943-017-0755-z] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Accepted: 12/27/2017] [Indexed: 12/17/2022] Open
Abstract
Pancreatic cancer is one of the most lethal cancers worldwide due to its insidious symptoms, early metastasis, and chemoresistance. Hence, the underlying mechanisms contributing to pancreatic cancer progression require further exploration. Based on accumulating evidence, extracellular vesicles, including exosomes and microvesicles, play a crucial role in pancreatic cancer progression and chemoresistance. Furthermore, they also possess the potential to be promising biomarkers, therapy targets and tools for treating pancreatic cancer. Therefore, in-depth studies on the role of extracellular vesicles in pancreatic cancer are meaningful. In this review, we focus on the regulatory effects of extracellular vesicles on pancreatic cancer progression, metastasis, cancer-related immunity and chemoresistance, particularly their potential roles as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Jiangdong Qiu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Gang Yang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Mengyu Feng
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Suli Zheng
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Zhe Cao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Lianfang Zheng
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China. .,Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China.
| |
Collapse
|
576
|
Abstract
This chapter will discuss the potential use of microRNAs, particularly those located in peripherally-isolated exosomes, as biomarkers in neuropsychiatric disorders. These extracellular vesicles are released as a form of cell-to-cell communication and may mediate the soma-to-germline transmission of brain-relevant information, thereby potentially contributing to the inter- or transgenerational transmission of behavioral traits. Recent novel methods allow for the enrichment of peripheral exosomes specifically released by neurons and astrocytes and may provide valuable brain-relevant biosignatures of disease.
Collapse
Affiliation(s)
- Gabriel R Fries
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.
| | - Joao Quevedo
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Neuroscience Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA
- Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| |
Collapse
|
577
|
Šibíková M, Živný J, Janota J. Cell Membrane-Derived Microvesicles in Systemic Inflammatory Response. Folia Biol (Praha) 2018; 64:113-124. [PMID: 30724157 DOI: 10.14712/fb2018064040113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Human body reacts to physical, chemical and biological insults with a complex inflammatory reaction. Crucial components and executors of this response are endothelial cells, platelets, white blood cells, plasmatic coagulation system, and complement. Endothelial injury and inflammation are associated with elevated blood levels of cell membrane-derived microvesicles. Increased concentrations of microvesicles were found in several inflammatory reactions and diseases including acute coronary syndromes, stroke, vasculitis, venous thromboembolism, multiple sclerosis, rheumatoid arthritis, systemic lupus erythematosus, anti-phospholipid antibody syndrome, inflammatory bowel disease, thrombotic thrombocytopenic purpura, viral myocarditis, sepsis, disseminated intravascular coagulation, polytrauma, and burns. Microvesicles can modulate a variety of cellular processes, thereby having an impact on pathogenesis of diseases associated with inflammation. Microvesicles are important mediators and potential biomarkers of systemic inflammation. Measurement of inflammatory cell-derived microvesicles may be utilized in diagnostic algorithms and used for detection and determination of severity in diseases associated with inflammatory responses, as well as for prediction of their outcome. This review focuses on the mechanisms of release of microvesicles in diseases associated with systemic inflammation and their potential role in the regulation of cellular and humoral interactions.
Collapse
Affiliation(s)
- M Šibíková
- Third Faculty of Medicine, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - J Živný
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - J Janota
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
- Department of Neonatology, Thomayer Hospital, Prague, Czech Republic
| |
Collapse
|
578
|
Bag BG, Ghorai S, Panja SK, Dinda SK, Paul K. First in situ vesicular self-assembly of ‘binols’ generated by a two-component aerobic oxidation reaction. RSC Adv 2018; 8:29155-29163. [PMID: 35547996 PMCID: PMC9084448 DOI: 10.1039/c8ra06488a] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 08/07/2018] [Indexed: 11/25/2022] Open
Abstract
Generation of vesicular self-assemblies from natural and synthetic components has been in the frontiers of research in recent years for an improved understanding of the self-assembly process and also because of its prospective and realized applications in the areas of advanced materials, biotechnology and medicine. In the present work, we report the first example of the in situ generation of vesicular self-assemblies during an aerobic coupling reaction. The two precursor 2-naphthol units, having hydrogen bond donor–acceptor groups with appended alkyl chains, yielded binol (2,2′-dihydroxy-1,1′-binaphthyl) derivatives by aerobic coupling that spontaneously self-assembled in situ, yielding vesicular self-assemblies and gels. The morphology of the self-assemblies has been characterized by various optical, electron and atomic force microscopic techniques. The vesicular self-assemblies obtained in the liquids were capable of entrapping fluorophores such as rhodamine-B and carboxy fluorescein including the anticancer drug doxorubicin. The entrapped fluorophores could also be released by sonication or by rupture of vesicles. The supramolecular gels obtained in binary solvent mixtures showed improved gelation abilities with increase in the alkyl chain lengths as reflected by their minimum gelator concentration (mgcs) values, gel to sol transition temperatures (Tgel) and rheology properties. The results described here are also the first demonstration of gelation during an aerobic coupling reaction. Binol derivatives, obtained by aerobic coupling of two 2-naphthol derivatives having H-bond donor–acceptor groups and appended alkyl chains, spontaneously self-assembled in situ yielding vesicular self-assemblies and gels.![]()
Collapse
Affiliation(s)
- Braja G. Bag
- Department of Chemistry and Chemical Technology
- Vidyasagar University
- Midnapore 721102
- India
| | - Subrata Ghorai
- Department of Chemistry and Chemical Technology
- Vidyasagar University
- Midnapore 721102
- India
| | - Saikat K. Panja
- Department of Chemistry and Chemical Technology
- Vidyasagar University
- Midnapore 721102
- India
| | - Shaishab K. Dinda
- Department of Chemistry and Chemical Technology
- Vidyasagar University
- Midnapore 721102
- India
| | - Koushik Paul
- Department of Chemistry and Chemical Technology
- Vidyasagar University
- Midnapore 721102
- India
| |
Collapse
|
579
|
Schneider DJ, Speth JM, Penke LR, Wettlaufer SH, Swanson JA, Peters-Golden M. Mechanisms and modulation of microvesicle uptake in a model of alveolar cell communication. J Biol Chem 2017; 292:20897-20910. [PMID: 29101235 PMCID: PMC5743066 DOI: 10.1074/jbc.m117.792416] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 10/27/2017] [Indexed: 12/31/2022] Open
Abstract
Extracellular vesicles, including exosomes and shed microvesicles (MVs), can be internalized by recipient cells to modulate function. Although the mechanism by which extracellular vesicles are internalized is incompletely characterized, it is generally considered to involve endocytosis and an initial surface-binding event. Furthermore, modulation of uptake by microenvironmental factors is largely unstudied. Here, we used flow cytometry, confocal microscopy, and pharmacologic and molecular targeting to address these gaps in knowledge in a model of pulmonary alveolar cell-cell communication. Alveolar macrophage-derived MVs were fully internalized by alveolar epithelial cells in a time-, dose-, and temperature-dependent manner. Uptake was dependent on dynamin and actin polymerization. However, it was neither saturable nor dependent on clathrin or receptor binding. Internalization was enhanced by extracellular proteins but was inhibited by cigarette smoke extract via oxidative disruption of actin polymerization. We conclude that MV internalization occurs via a pathway more consistent with fluid-phase than receptor-dependent endocytosis and is subject to bidirectional modulation by relevant pathologic perturbations.
Collapse
Affiliation(s)
| | | | - Loka R Penke
- From the Division of Pulmonary and Critical Care Medicine
| | | | - Joel A Swanson
- Department of Microbiology and Immunology, and
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Marc Peters-Golden
- From the Division of Pulmonary and Critical Care Medicine,
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| |
Collapse
|
580
|
Chiarello DI, Salsoso R, Toledo F, Mate A, Vázquez CM, Sobrevia L. Foetoplacental communication via extracellular vesicles in normal pregnancy and preeclampsia. Mol Aspects Med 2017; 60:69-80. [PMID: 29222068 DOI: 10.1016/j.mam.2017.12.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 11/28/2017] [Accepted: 12/04/2017] [Indexed: 02/06/2023]
Abstract
Intercellular communication is a critical process in biological mechanisms. During pregnancy foetoplacental tissues release a heterogeneous group of extracellular vesicles (EVs) that include exosomes, microvesicles, apoptotic bodies, and syncytial nuclear aggregates. These vesicles contain a complex cargo (proteins, DNA, mRNA transcripts, microRNAs, noncoding RNA, lipids, and other molecules) that actively participate in the maternal-foetal communication by modulating different processes during gestation for a successful foetal development. Each stage of human gestation is marked by events such as immunomodulation, proliferation, invasion, migration, and differentiation, among others, requiring EVs-mediated signalling to be nearby or distant target cells. Furthermore, EVs also associate with pregnancy pathologies such as preeclampsia and intrauterine growth restriction. This review addresses the role of EVs in human foetomaternal communication in normal pregnancy and preeclampsia.
Collapse
Affiliation(s)
- Delia I Chiarello
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile.
| | - Rocío Salsoso
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, Seville E-41012, Spain
| | - Fernando Toledo
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Department of Basic Sciences, Faculty of Sciences, Universidad Del Bío-Bío, Chillán 3780000, Chile
| | - Alfonso Mate
- Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, Seville E-41012, Spain
| | - Carmen M Vázquez
- Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, Seville E-41012, Spain
| | - Luis Sobrevia
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, Seville E-41012, Spain; University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston, QLD 4029 Queensland, Australia.
| |
Collapse
|
581
|
Macías M, Alegre E, Díaz-Lagares A, Patiño A, Pérez-Gracia JL, Sanmamed M, López-López R, Varo N, González A. Liquid Biopsy: From Basic Research to Clinical Practice. Adv Clin Chem 2017; 83:73-119. [PMID: 29304904 DOI: 10.1016/bs.acc.2017.10.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Liquid biopsy refers to the molecular analysis in biological fluids of nucleic acids, subcellular structures, especially exosomes, and, in the context of cancer, circulating tumor cells. In the last 10 years, there has been an intensive research in liquid biopsy to achieve a less invasive and more precise personalized medicine. Molecular assessment of these circulating biomarkers can complement or even surrogate tissue biopsy. Because of this research, liquid biopsy has been introduced in clinical practice, especially in oncology, prenatal screening, and transplantation. Here we review the biology, methodological approaches, and clinical applications of the main biomarkers involved in liquid biopsy.
Collapse
Affiliation(s)
| | - Estibaliz Alegre
- Clínica Universidad de Navarra, Pamplona, Spain; The Health Research Institute of Navarra (IDISNA), Pamplona, Spain
| | - Angel Díaz-Lagares
- Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), University Clinical Hospital of Santiago (CHUS), CIBERONC, Santiago de Compostela, Spain; Roche-CHUS Joint Unit, University Clinical Hospital of Santiago (CHUS), Santiago de Compostela, Spain
| | - Ana Patiño
- Clínica Universidad de Navarra, Pamplona, Spain; The Health Research Institute of Navarra (IDISNA), Pamplona, Spain
| | - Jose L Pérez-Gracia
- Clínica Universidad de Navarra, Pamplona, Spain; The Health Research Institute of Navarra (IDISNA), Pamplona, Spain
| | - Miguel Sanmamed
- Yale University School of Medicine, New Haven, CT, United States
| | - Rafael López-López
- Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), University Clinical Hospital of Santiago (CHUS), CIBERONC, Santiago de Compostela, Spain; Roche-CHUS Joint Unit, University Clinical Hospital of Santiago (CHUS), Santiago de Compostela, Spain
| | - Nerea Varo
- Clínica Universidad de Navarra, Pamplona, Spain; The Health Research Institute of Navarra (IDISNA), Pamplona, Spain
| | - Alvaro González
- Clínica Universidad de Navarra, Pamplona, Spain; The Health Research Institute of Navarra (IDISNA), Pamplona, Spain.
| |
Collapse
|
582
|
Target-induced proximity ligation triggers recombinase polymerase amplification and transcription-mediated amplification to detect tumor-derived exosomes in nasopharyngeal carcinoma with high sensitivity. Biosens Bioelectron 2017; 102:204-210. [PMID: 29145073 DOI: 10.1016/j.bios.2017.11.033] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Revised: 10/29/2017] [Accepted: 11/07/2017] [Indexed: 01/09/2023]
Abstract
Tumor-derived exosomes (TEXs) are extracellular vesicles that are continuously released into the blood by tumor cells and carry specific surface markers of the original tumor cells. Substantial evidence has implicated TEXs as attractive diagnostic markers for cancer. However, the detection of TEXs in blood at an early tumor stage is challenging due to their very low concentration. Here, we established a method called PLA-RPA-TMA assay that allows TEXs to be detected with high sensitivity and specificity. Based on two proximity ligation assay (PLA) probes that recognize a biomarker on a TEX, we generated a unique surrogate DNA signal for the specific biomarker, which was synchronously amplified twice by recombinase polymerase amplification (RPA) coupled with transcription-mediated amplification (TMA), and then the products of the RPA-TMA reaction were quantitatively detected using a gold nanoparticle-based colorimetric assay. We established proof-of-concept evidence for this approach using TEXs from nasopharyngeal carcinoma (NPC) cells, with a detection limit of 102 particles/mL, and reported the measurement of plasma Epstein-Barr virus latent membrane protein 1 (LPM1)-positive (LMP1+, accuracy: 0.956) and epidermal growth factor receptor (EGFR)-positive (EGFR+, accuracy: 0.906) TEXs as potent early diagnostic biomarkers for NPC.
Collapse
|
583
|
Adamiak M, Cheng G, Bobis-Wozowicz S, Zhao L, Kedracka-Krok S, Samanta A, Karnas E, Xuan YT, Skupien-Rabian B, Chen X, Jankowska U, Girgis M, Sekula M, Davani A, Lasota S, Vincent RJ, Sarna M, Newell KL, Wang OL, Dudley N, Madeja Z, Dawn B, Zuba-Surma EK. Induced Pluripotent Stem Cell (iPSC)-Derived Extracellular Vesicles Are Safer and More Effective for Cardiac Repair Than iPSCs. Circ Res 2017; 122:296-309. [PMID: 29118058 DOI: 10.1161/circresaha.117.311769] [Citation(s) in RCA: 202] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 11/03/2017] [Accepted: 11/07/2017] [Indexed: 12/21/2022]
Abstract
RATIONALE Extracellular vesicles (EVs) are tiny membrane-enclosed droplets released by cells through membrane budding or exocytosis. The myocardial reparative abilities of EVs derived from induced pluripotent stem cells (iPSCs) have not been directly compared with the source iPSCs. OBJECTIVE To examine whether iPSC-derived EVs can influence the biological functions of cardiac cells in vitro and to compare the safety and efficacy of iPSC-derived EVs (iPSC-EVs) and iPSCs for cardiac repair in vivo. METHODS AND RESULTS Murine iPSCs were generated, and EVs isolated from culture supernatants by sequential centrifugation. Atomic force microscopy, high-resolution flow cytometry, real-time quantitative RT-PCR, and mass spectrometry were used to characterize EV morphology and contents. iPSC-EVs were enriched in miRNAs and proteins with proangiogenic and cytoprotective properties. iPSC-EVs enhanced angiogenic, migratory, and antiapoptotic properties of murine cardiac endothelial cells in vitro. To compare the cardiac reparative capacities in vivo, vehicle, iPSCs, and iPSC-EVs were injected intramyocardially at 48 hours after a reperfused myocardial infarction in mice. Compared with vehicle-injected mice, both iPSC- and iPSC-EV-treated mice exhibited improved left ventricular function at 35 d after myocardial infarction, albeit iPSC-EVs rendered greater improvement. iPSC-EV injection also resulted in reduction in left ventricular mass and superior perfusion in the infarct zone. Both iPSCs and iPSC-EVs preserved viable myocardium in the infarct zone, whereas reduction in apoptosis was significant with iPSC-EVs. iPSC injection resulted in teratoma formation, whereas iPSC-EV injection was safe. CONCLUSIONS iPSC-derived EVs impart cytoprotective properties to cardiac cells in vitro and induce superior cardiac repair in vivo with regard to left ventricular function, vascularization, and amelioration of apoptosis and hypertrophy. Because of their acellular nature, iPSC-EVs represent a safer alternative for potential therapeutic applications in patients with ischemic myocardial damage.
Collapse
Affiliation(s)
- Marta Adamiak
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Guangming Cheng
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Sylwia Bobis-Wozowicz
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Lin Zhao
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Sylwia Kedracka-Krok
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Anweshan Samanta
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Elzbieta Karnas
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Yu-Ting Xuan
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Bozena Skupien-Rabian
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Xing Chen
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Urszula Jankowska
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Magdy Girgis
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Malgorzata Sekula
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Arash Davani
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Slawomir Lasota
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Robert J Vincent
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Michal Sarna
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Kathy L Newell
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Ou-Li Wang
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Nathaniel Dudley
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Zbigniew Madeja
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna)
| | - Buddhadeb Dawn
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna).
| | - Ewa K Zuba-Surma
- From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute (G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna).
| |
Collapse
|
584
|
Effect of the irradiation on Neuroblastoma-derived microvesicles: A physical and biological investigation. Colloids Surf A Physicochem Eng Asp 2017. [DOI: 10.1016/j.colsurfa.2017.05.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
585
|
Lv LL, Wu WJ, Feng Y, Li ZL, Tang TT, Liu BC. Therapeutic application of extracellular vesicles in kidney disease: promises and challenges. J Cell Mol Med 2017; 22:728-737. [PMID: 29083099 PMCID: PMC5783839 DOI: 10.1111/jcmm.13407] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 08/28/2017] [Indexed: 12/22/2022] Open
Abstract
Extracellular vesicles (EVs) are nanosized, membrane‐bound vesicles released from different cells. Recent studies have revealed that EVs may participate in renal tissue damage and regeneration through mediating inter‐nephron communication. Thus, the potential use of EVs as therapeutic vector has gained considerable interest. In this review, we will discuss the basic characteristics of EVs and its role in nephron cellular communication. Then, the application of EVs as therapeutic vector based on its natural content or as carriers of drug, in acute and chronic kidney injury, was discussed. Finally, perspectives and challenges of EVs in therapy of kidney disease were described.
Collapse
Affiliation(s)
- Lin-Li Lv
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Wei-Jun Wu
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Ye Feng
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Zuo-Lin Li
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Tao-Tao Tang
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Bi-Cheng Liu
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| |
Collapse
|
586
|
Alhomrani M, Correia J, Zavou M, Leaw B, Kuk N, Xu R, Saad MI, Hodge A, Greening DW, Lim R, Sievert W. The Human Amnion Epithelial Cell Secretome Decreases Hepatic Fibrosis in Mice with Chronic Liver Fibrosis. Front Pharmacol 2017; 8:748. [PMID: 29114223 PMCID: PMC5660722 DOI: 10.3389/fphar.2017.00748] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 10/03/2017] [Indexed: 12/22/2022] Open
Abstract
Background: Hepatic stellate cells (HSCs) are the primary collagen-secreting cells in the liver. While HSCs are the major cell type involved in the pathogenesis of liver fibrosis, hepatic macrophages also play an important role in mediating fibrogenesis and fibrosis resolution. Previously, we observed a reduction in HSC activation, proliferation, and collagen synthesis following exposure to human amnion epithelial cells (hAEC) and hAEC-conditioned media (hAEC-CM). This suggested that specific factors secreted by hAEC might be effective in ameliorating liver fibrosis. hAEC-derived extracellular vesicles (hAEC-EVs), which are nanosized (40–100 nm) membrane bound vesicles, may act as novel cell–cell communicators. Accordingly, we evaluated the efficacy of hAEC-EV in modulating liver fibrosis in a mouse model of chronic liver fibrosis and in human HSC. Methods: The hAEC-EVs were isolated and characterized. C57BL/6 mice with CCl4-induced liver fibrosis were administered hAEC-EV, hAEC-CM, or hAEC-EV depleted medium (hAEC-EVDM). LX2 cells, a human HSC line, and bone marrow-derived mouse macrophages were exposed to hAEC-EV, hAEC-CM, and hAEC-EVDM. Mass spectrometry was used to examine the proteome profile of each preparation. Results: The extent of liver fibrosis and number of activated HSCs were reduced significantly in CCl4-treated mice given hAEC-EVs, hAEC-CM, and hAEC EVDM compared to untreated controls. Hepatic macrophages were significantly decreased in all treatment groups, where a predominant M2 phenotype was observed. Human HSCs cultured with hAEC-EV and hAEC-CM displayed a significant reduction in collagen synthesis and hAEC-EV, hAEC-CM, and hAEC-EVDM altered macrophage polarization in bone marrow-derived mouse macrophages. Proteome analysis showed that 164 proteins were unique to hAEC-EV in comparison to hAEC-CM and hAEC-EVDM, and 51 proteins were co-identified components with the hAEC-EV fraction. Conclusion: This study provides novel data showing that hAEC-derived EVs significantly reduced liver fibrosis and macrophage infiltration to an extent similar to hAEC-EVDM and hAEC-CM. hAEC-EV-based therapy may be a potential therapeutic option for liver fibrosis.
Collapse
Affiliation(s)
- Majid Alhomrani
- Centre for Inflammatory Diseases, Monash University, Clayton, VIC, Australia.,Hudson Institute of Medical Research, Clayton, VIC, Australia.,Medical College, Taif University, Taif, Saudi Arabia
| | - Jeanne Correia
- Centre for Inflammatory Diseases, Monash University, Clayton, VIC, Australia.,Gastroenterology and Hepatology Unit, Monash Health, Clayton, VIC, Australia
| | - Marcus Zavou
- Centre for Inflammatory Diseases, Monash University, Clayton, VIC, Australia.,Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Bryan Leaw
- Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Nathan Kuk
- Centre for Inflammatory Diseases, Monash University, Clayton, VIC, Australia.,Gastroenterology and Hepatology Unit, Monash Health, Clayton, VIC, Australia
| | - Rong Xu
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Mohamed I Saad
- Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
| | - Alexander Hodge
- Centre for Inflammatory Diseases, Monash University, Clayton, VIC, Australia.,Gastroenterology and Hepatology Unit, Monash Health, Clayton, VIC, Australia
| | - David W Greening
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Rebecca Lim
- Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - William Sievert
- Centre for Inflammatory Diseases, Monash University, Clayton, VIC, Australia.,Gastroenterology and Hepatology Unit, Monash Health, Clayton, VIC, Australia
| |
Collapse
|
587
|
Lobb RJ, Hastie ML, Norris EL, van Amerongen R, Gorman JJ, Möller A. Oncogenic transformation of lung cells results in distinct exosome protein profile similar to the cell of origin. Proteomics 2017; 17. [DOI: 10.1002/pmic.201600432] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 06/09/2017] [Indexed: 01/01/2023]
Affiliation(s)
- Richard J. Lobb
- Tumour Microenvironment Laboratory; QIMR Berghofer Medical Research Institute; Herston Australia
| | - Marcus L. Hastie
- Protein Discovery Centre; QIMR Berghofer Medical Research Institute; Herston Australia
| | - Emma L. Norris
- Protein Discovery Centre; QIMR Berghofer Medical Research Institute; Herston Australia
| | - Rosa van Amerongen
- Tumour Microenvironment Laboratory; QIMR Berghofer Medical Research Institute; Herston Australia
| | - Jeffrey J. Gorman
- Protein Discovery Centre; QIMR Berghofer Medical Research Institute; Herston Australia
- Institute for Molecular Bioscience; University of Queensland; Brisbane Australia
| | - Andreas Möller
- Tumour Microenvironment Laboratory; QIMR Berghofer Medical Research Institute; Herston Australia
- School of Medicine; University of Queensland; Brisbane Australia
| |
Collapse
|
588
|
Kim SM, Kim HS. Engineering of extracellular vesicles as drug delivery vehicles. Stem Cell Investig 2017; 4:74. [PMID: 29057246 DOI: 10.21037/sci.2017.08.07] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 08/21/2017] [Indexed: 12/31/2022]
Abstract
Extracellular vesicles (EVs) are secreted membrane-enclosed nano-sized particles (40-1,000 nm) that deliver biological information between cells. The molecular composition of these subcellular particles includes growth factor receptors, ligands adhesion proteins, mRNA, miRNAs, lncRNA and lipids that are derived from donor cells. A number of studies demonstrated that stem cell-derived EVs are the key mediator of tissue repair and regeneration in multiple animal disease models. In addition, the composition of these particles is known to be altered in cancer and disease pathology suggesting them for useful in diagnostic and therapeutic purposes. Their endogenous origin and biological properties offer benefits over conventional drug delivery systems (DDS), such as liposome, synthetic nanoparticles and prompted the further application of EVs as drug delivery vehicles for chemical drugs, genetic materials and proteins. The contents of EVs can be efficiently modified by chemical, biological or physical means. Thus, EVs can be an innovative DDS as it can overcome physical and biological barriers and safely deliver therapeutic drugs to target tissues. In this minireview, we summarized current progress on the strategies of drug loading onto EVs; ex vivo and in vivo loading.
Collapse
Affiliation(s)
- Sung-Man Kim
- Medical Management Department, College of Medical Convergence, Catholic Kwandong University, Gangneung-si, Gangwon-do, Republic of Korea
| | - Han-Soo Kim
- Department of Biomedical Sciences, College of Medical Convergence, Catholic Kwandong University, Gangneung-si, Gangwon-do, Republic of Korea
| |
Collapse
|
589
|
Rufino-Ramos D, Albuquerque PR, Carmona V, Perfeito R, Nobre RJ, Pereira de Almeida L. Extracellular vesicles: Novel promising delivery systems for therapy of brain diseases. J Control Release 2017; 262:247-258. [DOI: 10.1016/j.jconrel.2017.07.001] [Citation(s) in RCA: 166] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 06/30/2017] [Accepted: 07/01/2017] [Indexed: 12/11/2022]
|
590
|
de la Cuesta F, Baldan-Martin M, Moreno-Luna R, Alvarez-Llamas G, Gonzalez-Calero L, Mourino-Alvarez L, Sastre-Oliva T, López JA, Vázquez J, Ruiz-Hurtado G, Segura J, Vivanco F, Ruilope LM, Barderas MG. Kalirin and CHD7: novel endothelial dysfunction indicators in circulating extracellular vesicles from hypertensive patients with albuminuria. Oncotarget 2017; 8:15553-15562. [PMID: 28152519 PMCID: PMC5362505 DOI: 10.18632/oncotarget.14948] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 12/05/2016] [Indexed: 11/25/2022] Open
Abstract
Despite of the great advances in anti-hypertensive therapies, many patients under Renin-Angiotensin- System (RAS) suppression develop albuminuria, which is a clear indicator of therapeutic inefficiency. Hence, indicators of vascular function are needed to assess patients’ condition and help deciding future therapies. Proteomic analysis of circulating extracellular vesicles (EVs) showed two proteins, kalirin and chromodomain-helicase-DNA-binding protein 7 (CHD7), increased in albuminuric patients. A positive correlation of both with the expression of the endothelial activation marker E-selectin was found in EVs. In vitro analysis using TNFα-treated adult human endothelial cells proved their involvement in endothelial cell activation. Hence, we propose protein levels of kalirin and CHD7 in circulating EVs as novel endothelial dysfunction markers to monitor vascular condition in hypertensive patients with albuminuria.
Collapse
Affiliation(s)
- Fernando de la Cuesta
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos (HNP), SESCAM, Toledo, Spain.,Current address: Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Montserrat Baldan-Martin
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos (HNP), SESCAM, Toledo, Spain
| | - Rafael Moreno-Luna
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos (HNP), SESCAM, Toledo, Spain
| | | | | | - Laura Mourino-Alvarez
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos (HNP), SESCAM, Toledo, Spain
| | - Tamara Sastre-Oliva
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos (HNP), SESCAM, Toledo, Spain
| | | | | | - Gema Ruiz-Hurtado
- Unidad de Hipertension, Instituto de Investigacion i + 12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Julian Segura
- Unidad de Hipertension, Instituto de Investigacion i + 12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Fernando Vivanco
- Department of Immunology, IIS-Fundacion Jimenez Diaz, Madrid, Spain.,Departamento de Bioquimica y Biologia Molecular I, Universidad Complutense, Madrid, Spain
| | - Luis M Ruilope
- Unidad de Hipertension, Instituto de Investigacion i + 12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Maria G Barderas
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos (HNP), SESCAM, Toledo, Spain
| |
Collapse
|
591
|
Potential Therapeutic Mechanisms and Tracking of Transplanted Stem Cells: Implications for Stroke Treatment. Stem Cells Int 2017; 2017:2707082. [PMID: 28904531 PMCID: PMC5585684 DOI: 10.1155/2017/2707082] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Revised: 07/08/2017] [Accepted: 07/30/2017] [Indexed: 02/06/2023] Open
Abstract
Stem cell therapy is a promising potential therapeutic strategy to treat cerebral ischemia in preclinical and clinical trials. Currently proposed treatments for stroke employing stem cells include the replacement of lost neurons and integration into the existing host circuitry, the release of growth factors to support and promote endogenous repair processes, and the secretion of extracellular vesicles containing proteins, noncoding RNA, or DNA to regulate gene expression in recipient cells and achieve immunomodulation. Progress has been made to elucidate the precise mechanisms underlying stem cell therapy and the homing, migration, distribution, and differentiation of transplanted stem cells in vivo using various imaging modalities. Noninvasive and safe tracer agents with high sensitivity and image resolution must be combined with long-term monitoring using imaging technology to determine the optimal therapy for stroke in terms of administration route, dosage, and timing. This review discusses potential therapeutic mechanisms of stem cell transplantation for the treatment of stroke and the limitations of current therapies. Methods to label transplanted cells and existing imaging systems for stem cell labeling and in vivo tracking will also be discussed.
Collapse
|
592
|
Coumans FAW, Brisson AR, Buzas EI, Dignat-George F, Drees EEE, El-Andaloussi S, Emanueli C, Gasecka A, Hendrix A, Hill AF, Lacroix R, Lee Y, van Leeuwen TG, Mackman N, Mäger I, Nolan JP, van der Pol E, Pegtel DM, Sahoo S, Siljander PRM, Sturk G, de Wever O, Nieuwland R. Methodological Guidelines to Study Extracellular Vesicles. Circ Res 2017; 120:1632-1648. [PMID: 28495994 DOI: 10.1161/circresaha.117.309417] [Citation(s) in RCA: 722] [Impact Index Per Article: 90.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Owing to the relationship between extracellular vesicles (EVs) and physiological and pathological conditions, the interest in EVs is exponentially growing. EVs hold high hopes for novel diagnostic and translational discoveries. This review provides an expert-based update of recent advances in the methods to study EVs and summarizes currently accepted considerations and recommendations from sample collection to isolation, detection, and characterization of EVs. Common misconceptions and methodological pitfalls are highlighted. Although EVs are found in all body fluids, in this review, we will focus on EVs from human blood, not only our most complex but also the most interesting body fluid for cardiovascular research.
Collapse
Affiliation(s)
- Frank A W Coumans
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Alain R Brisson
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Edit I Buzas
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Françoise Dignat-George
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Esther E E Drees
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Samir El-Andaloussi
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Costanza Emanueli
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Aleksandra Gasecka
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - An Hendrix
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Andrew F Hill
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Romaric Lacroix
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Yi Lee
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Ton G van Leeuwen
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Nigel Mackman
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Imre Mäger
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - John P Nolan
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Edwin van der Pol
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - D Michiel Pegtel
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Susmita Sahoo
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Pia R M Siljander
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Guus Sturk
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Olivier de Wever
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.)
| | - Rienk Nieuwland
- From the Biomedical Engineering and Physics (F.A.W.C., T.G.v.L., E.v.d.P.), Vesicle Observation Centre (F.A.W.C., A.G., T.G.v.L., E.v.d.P., G.S., R.N.), and Laboratory of Experimental Clinical Chemistry (A.G., G.S., R.N.), Academic Medical Center, University of Amsterdam, The Netherlands; Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS, University of Bordeaux, IPB, Pessac, France (A.R.B.); Department of Genetics, Cell- and Immunobology, Semmelweis University, Budapest, Hungary (E.I.B.); VRCM, UMRS-1076, INSERM, Aix-Marseille University, UFR de Pharmacie, Marseille, France (F.D.-G., R.L.); Haematology and vascular biology department, CHU La Conception, APHM, Marseille, France (F.D.-G., R.L.); Exosomes Research Group, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands (E.E.E.D., D.M.P.); Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (S.E.-A., Y.L.); Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (S.E.-A., I.M.); Bristol Heart Institute, University of Bristol, United Kingdom (C.E.); National Heart & Lung Institute, Imperial College London, United Kingdom (C.E.); 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland (A.G.); Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Belgium (A.H., O.d.W.); Cancer Research Institute Ghent, Belgium (A.H., O.d.W.); Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia (A.F.H.); Department of Medicine, University of North Carolina at Chapel Hill (N.M.); Institute of Technology, University of Tartu, Estonia (I.M.); Scintillon Institute, San Diego, CA (J.P.N.); Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (S.S.); and EV Core Facility, University of Helsinki and EV-Group, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Finland (P.R.M.S.).
| |
Collapse
|
593
|
Lee H, Zhang D, Wu J, Otterbein LE, Jin Y. Lung Epithelial Cell-Derived Microvesicles Regulate Macrophage Migration via MicroRNA-17/221-Induced Integrin β 1 Recycling. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 199:1453-1464. [PMID: 28674181 PMCID: PMC5561736 DOI: 10.4049/jimmunol.1700165] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 06/11/2017] [Indexed: 12/26/2022]
Abstract
Robust lung inflammation is one of the prominent features in the pathogenesis of acute lung injury (ALI). Macrophage migration and recruitment are often seen at the early stage of lung inflammatory responses to noxious stimuli. Using an acid inhalation-induced lung injury model, we explored the mechanisms by which acid exposure initiates macrophage recruitment and migration during development of ALI. The lung epithelium comprises a large surface area and functions as a first-line defense against noxious insults. We found that acid exposure induced a remarkable microvesicle (MV) release from lung epithelium as detected in bronchoalveolar lavage fluid. Significantly elevated RNA, rather than protein, was found in these epithelium-derived MVs after acid and included several highly elevated microRNAs, including microRNA (miR)-17 and miR-221. Acid-induced epithelial MV release promoted macrophage migration in vitro and recruitment into the lung in vivo and required, in part, MV shuttling of miR-17 and/or miR-221. Mechanistically, acid-induced epithelial MV miR-17/221 promoted β1 integrin recycling and presentation back onto the surface of macrophages, in part via a Rab11-mediated pathway. Integrin β1 is known to play an essential role in regulating macrophage migration. Taken together, acid-induced ALI results in epithelial MV shuttling of miR-17/221 that in turn modulates macrophage β1 integrin recycling, promoting macrophage recruitment and ultimately contributing to lung inflammation.
Collapse
Affiliation(s)
- Heedoo Lee
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, MA 02118; and
| | - Duo Zhang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, MA 02118; and
| | - Jingxuan Wu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, MA 02118; and
| | - Leo E Otterbein
- Department of Surgery, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA 02215
| | - Yang Jin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, MA 02118; and
| |
Collapse
|
594
|
McBride JD, Rodriguez-Menocal L, Guzman W, Candanedo A, Garcia-Contreras M, Badiavas EV. Bone Marrow Mesenchymal Stem Cell-Derived CD63 + Exosomes Transport Wnt3a Exteriorly and Enhance Dermal Fibroblast Proliferation, Migration, and Angiogenesis In Vitro. Stem Cells Dev 2017; 26:1384-1398. [PMID: 28679315 DOI: 10.1089/scd.2017.0087] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Wnts are secreted glycoproteins that regulate stem cell self-renewal, differentiation, and cell-to-cell communication during embryonic development and in adult tissues. Bone marrow mesenchymal stem cells (BM-MSCs) have been shown to stimulate dermis repair and regeneration; however, it is unclear how BM-MSCs may modulate downstream Wnt signaling. While recent reports implicate that Wnt ligands and Wnt messenger RNAs (such as Wnt4) exist within the interior compartment of exosomes, it has been debated whether or not Wnts exist on the exterior surface of exosomes to travel in the extracellular space. To help answer this question, we utilized flow cytometry of magnetic beads coated with anti-CD63 antibodies and found, for the first time, that Wnt3a protein is detectable exteriorly on CD63+ exosomes derived from BM-MSCs over-secreting Wnt3a into serum-free conditioned media (Wnt3a CM). Our data suggest that CD63+ exosomes significantly help transport exterior Wnt3a signal to recipient cells to promote fibroblast and endothelial functions. During purification of exosomes, we unexpectedly found that use of ultracentrifugation alone significantly decreased the ability to detect exteriorly bound Wnt3a on CD63+ exosomes, however, polyethylene glycol (PEG)-mediated exosome-enrichment before exosome-purification (with ultracentrifugation into a sucrose cushion) resulted in exosomes more likely to retain exterior Wnt3a detectability and downstream Wnt/beta-catenin activity. Our findings indicate the important role that purification methods may have on stem cell-derived Wnt-exosome activity in downstream assays. The ability for BM-MSC Wnt3a CM and exosomes to stimulate dermal fibroblast proliferation and migration, and endothelial angiogenesis in vitro, was significantly decreased after CD63+-exosome depletion or knockdown of Wnt coreceptor LRP6 in recipient cells, suggesting both are required for optimal Wnt-exosome activity in our system. Thus, BM-MSC-derived CD63+ exosomes are a significant carrier of exterior Wnt3a within high Wnt environments, resulting in downstream fibroblast proliferation, migration, and angiogenesis in vitro.
Collapse
Affiliation(s)
- Jeffrey D McBride
- 1 Department of Dermatology and Cutaneous Surgery, University of Miami School of Medicine , Miami, Florida.,2 Interdisciplinary Stem Cell Institute , Miami, Florida
| | - Luis Rodriguez-Menocal
- 1 Department of Dermatology and Cutaneous Surgery, University of Miami School of Medicine , Miami, Florida.,2 Interdisciplinary Stem Cell Institute , Miami, Florida
| | - Wellington Guzman
- 1 Department of Dermatology and Cutaneous Surgery, University of Miami School of Medicine , Miami, Florida.,2 Interdisciplinary Stem Cell Institute , Miami, Florida
| | - Ambar Candanedo
- 1 Department of Dermatology and Cutaneous Surgery, University of Miami School of Medicine , Miami, Florida.,2 Interdisciplinary Stem Cell Institute , Miami, Florida
| | | | - Evangelos V Badiavas
- 1 Department of Dermatology and Cutaneous Surgery, University of Miami School of Medicine , Miami, Florida.,2 Interdisciplinary Stem Cell Institute , Miami, Florida
| |
Collapse
|
595
|
Abstract
OBJECTIVE Exosomes are small, cell-released vesicles (40-100 nm in size) with the potential to transfer proteins, lipids, small RNAs, messenger RNAs, or DNA between cells via interstitial fluids. Due to their role in tissue homeostasis, exosomes have emerged as a new type of therapeutic and diagnostic (theranostic) tool in the noninvasive assessment of organ response to injury or treatment and in the development of reliable organ-protective intensive therapy. Our review provides current insights into the role of exosomes in the personalized management of injury and repair responses in critical illness. DATA SOURCE Data were obtained from a PubMed search of the most recent medical literature, including the PubMed "related articles" search methodology. STUDY SELECTION Articles considered include original articles, review articles and conference proceedings. DATA EXTRACTION A detailed review of scientific, peer-reviewed data was performed. Relevant pre-clinical and clinical studies were included and summarized. DATA SYNTHESIS Current scientific evidence is focused on the following: 1) Frontiers in the management of critical illness; 2) Biogenesis, characterization, and function of circulating exosomes; 3) The role of exosomes in acute lung injury; 4) The role of exosomes in acute cardiac injury; 5) The role of exosomes in acute kidney injury; 6) The role of exosomes in sepsis; 7) Limitations of exosome isolation protocols; and 8) Perspectives in the theranostic use of exosomes. CONCLUSIONS Circulating levels of exosomes are associated with the onset and clinical course of critical illness. Exosomes released from cells with different phenotypes exert different functions in order to protect tissue and preserve organ function. Therefore, multifunctional exosomes with combined diagnostic and therapeutic functions show great promise in terms of personalized nanomedicine for patient-specific diagnosis and treatment of critical illness.
Collapse
|
596
|
Lu M, Xing H, Yang Z, Sun Y, Yang T, Zhao X, Cai C, Wang D, Ding P. Recent advances on extracellular vesicles in therapeutic delivery: Challenges, solutions, and opportunities. Eur J Pharm Biopharm 2017; 119:381-395. [PMID: 28739288 DOI: 10.1016/j.ejpb.2017.07.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 06/24/2017] [Accepted: 07/20/2017] [Indexed: 12/21/2022]
Abstract
Extracellular vesicles (EVs) are intrinsic mediators of intercellular communication in our body, allowing functional transfer of biomolecules (lipids, proteins, and nucleic acid) between diverse locations. Such an instrumental role evokes a surge of interest within the drug delivery community in tailoring EVs for therapeutic delivery. These vesicles represent a novel generation of drug delivery systems, providing high delivery efficiency, intrinsic targeting properties, and low immunogenicity. In the recent years, considerable research efforts have been directed toward developing safe and efficient EV-based delivery vehicles. Although EVs are shown to harbor great promise in therapeutic delivery, substantial improvements in exploring standardized isolation techniques with high efficiency and robust yield, scalable production, standard procedures for EV storage, efficient loading methods without damaging EV integrity, understanding their in vivo trafficking, and developing novel EV-based nanocarriers are still required before their clinical transformation. In this review, we seek to summarize the recent advance on harnessing EVs for drug delivery with focus on state-of-the-art solutions for overcoming major challenges.
Collapse
Affiliation(s)
- Mei Lu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Haonan Xing
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Zhen Yang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Yanping Sun
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Tianzhi Yang
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, Husson University, Bangor, ME, USA
| | - Xiaoyun Zhao
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Cuifang Cai
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Dongkai Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China.
| | - Pingtian Ding
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China.
| |
Collapse
|
597
|
Mourino-Alvarez L, Baldan-Martin M, Rincon R, Martin-Rojas T, Corbacho-Alonso N, Sastre-Oliva T, Barderas MG. Recent advances and clinical insights into the use of proteomics in the study of atherosclerosis. Expert Rev Proteomics 2017; 14:701-713. [PMID: 28689450 DOI: 10.1080/14789450.2017.1353912] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION The application of new proteomics methods may help to identify new diagnostic/predictive molecular markers in an attempt to improve the clinical management of atherosclerosis. Areas covered: Technological advances in proteomics have enhanced its sensitivity and multiplexing capacity, as well as the possibility of studying protein interactions and tissue structure. These advances will help us better understand the molecular mechanisms at play in atherosclerosis as a biological system. Moreover, this should help identify new predictive/diagnostic biomarkers and therapeutic targets that may facilitate effective risk stratification and early diagnosis, with the ensuing rapid implementation of treatment. This review provides a comprehensive overview of the novel methods in proteomics, including state-of-the-art techniques, novel biological samples and applications for the study of atherosclerosis. Expert commentary: Collaboration between clinicians and researchers is crucial to further validate and introduce new molecular markers to manage atherosclerosis that are identified using the most up to date proteomic approaches.
Collapse
Affiliation(s)
- Laura Mourino-Alvarez
- a Department of Vascular Physiopathology , Hospital Nacional de Paraplejicos , Toledo , Spain
| | | | - Raul Rincon
- a Department of Vascular Physiopathology , Hospital Nacional de Paraplejicos , Toledo , Spain
| | - Tatiana Martin-Rojas
- a Department of Vascular Physiopathology , Hospital Nacional de Paraplejicos , Toledo , Spain
| | - Nerea Corbacho-Alonso
- a Department of Vascular Physiopathology , Hospital Nacional de Paraplejicos , Toledo , Spain
| | - Tamara Sastre-Oliva
- a Department of Vascular Physiopathology , Hospital Nacional de Paraplejicos , Toledo , Spain
| | - Maria G Barderas
- a Department of Vascular Physiopathology , Hospital Nacional de Paraplejicos , Toledo , Spain
| |
Collapse
|
598
|
Proteomic Analysis of Blood Extracellular Vesicles in Cardiovascular Disease by LC-MS/MS Analysis. Methods Mol Biol 2017. [PMID: 28674883 DOI: 10.1007/978-1-4939-7057-5_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Extracellular vesicles are membrane vesicles related to cell communication. These vesicles consist of proteins, RNA, and microRNA and are an interesting and important tool to understand the processes taking place in the secreting cell, especially in diseases in which its release is often enhanced. The used of blood extracellular vesicles in cardiovascular disease as a low invasive, easily accessible source of circulating markers could give us important information related to pathological process even more with the use of proteomic analysis. In this chapter, we describe a protocol to isolate and proteomic analyze extracellular vesicles from blood associated with cardiovascular disease.
Collapse
|
599
|
Khalyfa A, Kheirandish-Gozal L, Gozal D. Circulating exosomes in obstructive sleep apnea as phenotypic biomarkers and mechanistic messengers of end-organ morbidity. Respir Physiol Neurobiol 2017; 256:143-156. [PMID: 28676332 DOI: 10.1016/j.resp.2017.06.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 06/13/2017] [Accepted: 06/19/2017] [Indexed: 02/08/2023]
Abstract
Obstructive sleep apnea (OSA), the most severe form of sleep disordered breathing, is characterized by intermittent hypoxia during sleep (IH), sleep fragmentation, and episodic hypercapnia. OSA is associated with increased risk for morbidity and mortality affecting cardiovascular, metabolic, and neurocognitive systems, and more recently with non-alcoholic fatty liver disease (NAFLD) and cancer-related deaths. Substantial variability in OSA outcomes suggests that genetically-determined and environmental and lifestyle factors affect the phenotypic susceptibility to OSA. Furthermore, OSA and obesity often co-exist and manifest activation of shared molecular end-organ injury mechanisms that if properly identified may represent potential therapeutic targets. A challenge in the development of non-invasive diagnostic assays in body fluids is the ability to identify clinically relevant biomarkers. Circulating extracellular vesicles (EVs) include a heterogeneous population of vesicular structures including exosomes, prostasomes, microvesicles (MVs), ectosomes and oncosomes, and are classified based on their size, shape and membrane surface composition. Of these, exosomes (30-100nm) are very small membrane vesicles derived from multi-vesicular bodies or from the plasma membrane and play important roles in mediating cell-cell communication via cargo that includes lipids, proteins, mRNAs, miRNAs and DNA. We have recently identified a unique cluster of exosomal miRNAs in both humans and rodents exposed to intermittent hypoxia as well as in patients with OSA with divergent morbid phenotypes. Here we summarize such recent findings, and will focus on exosomal miRNAs in both adult and children which mediate intercellular communication relevant to OSA and endothelial dysfunction, and their potential value as diagnostic and prognostic biomarkers.
Collapse
Affiliation(s)
- Abdelnaby Khalyfa
- Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, USA.
| | - Leila Kheirandish-Gozal
- Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| | - David Gozal
- Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
600
|
Lobb RJ, Lima LG, Möller A. Exosomes: Key mediators of metastasis and pre-metastatic niche formation. Semin Cell Dev Biol 2017; 67:3-10. [DOI: 10.1016/j.semcdb.2017.01.004] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 12/22/2016] [Accepted: 01/06/2017] [Indexed: 12/21/2022]
|