201
|
Seras‐Franzoso J, Díaz‐Riascos ZV, Corchero JL, González P, García‐Aranda N, Mandaña M, Riera R, Boullosa A, Mancilla S, Grayston A, Moltó‐Abad M, Garcia‐Fruitós E, Mendoza R, Pintos‐Morell G, Albertazzi L, Rosell A, Casas J, Villaverde A, Schwartz S, Abasolo I. Extracellular vesicles from recombinant cell factories improve the activity and efficacy of enzymes defective in lysosomal storage disorders. J Extracell Vesicles 2021; 10:e12058. [PMID: 33738082 PMCID: PMC7953474 DOI: 10.1002/jev2.12058] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 12/16/2020] [Accepted: 01/05/2021] [Indexed: 12/20/2022] Open
Abstract
In the present study the use of extracellular vesicles (EVs) as vehicles for therapeutic enzymes in lysosomal storage disorders was explored. EVs were isolated from mammalian cells overexpressing alpha-galactosidase A (GLA) or N-sulfoglucosamine sulfohydrolase (SGSH) enzymes, defective in Fabry and Sanfilippo A diseases, respectively. Direct purification of EVs from cell supernatants was found to be a simple and efficient method to obtain highly active GLA and SGSH proteins, even after EV lyophilization. Likewise, EVs carrying GLA (EV-GLA) were rapidly uptaken and reached the lysosomes in cellular models of Fabry disease, restoring lysosomal functionality much more efficiently than the recombinant enzyme in clinical use. In vivo, EVs were well tolerated and distributed among all main organs, including the brain. DiR-labelled EVs were localized in brain parenchyma 1 h after intra-arterial (internal carotid artery) or intravenous (tail vein) administrations. Moreover, a single intravenous administration of EV-GLA was able to reduce globotriaosylceramide (Gb3) substrate levels in clinically relevant tissues, such kidneys and brain. Overall, our results demonstrate that EVs from cells overexpressing lysosomal enzymes act as natural protein delivery systems, improving the activity and the efficacy of the recombinant proteins and facilitating their access to organs neglected by conventional enzyme replacement therapies.
Collapse
|
202
|
Stam J, Bartel S, Bischoff R, Wolters JC. Isolation of extracellular vesicles with combined enrichment methods. J Chromatogr B Analyt Technol Biomed Life Sci 2021; 1169:122604. [PMID: 33713953 DOI: 10.1016/j.jchromb.2021.122604] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 02/12/2021] [Accepted: 02/14/2021] [Indexed: 02/08/2023]
Abstract
Extracellular vesicles (EVs) are currently of tremendous interest in many research disciplines and EVs have potential for development of EV diagnostics or therapeutics. Most well-known single EV isolation methods have their particular advantages and disadvantages in terms of EV purity and EV yield. Combining EV isolation methods provides additional potential to improve the efficacy of both purity and yield. This review assesses the contribution and efficacy of using combined EV isolation methods by performing a two-step systematic literature analysis from all papers applying EV isolation in the year 2019. This resulted in an overview of the various methods being applied for EV isolations. A second database was generated for all studies within the first database that fairly compared multiple EV isolation methods by determining both EV purity and EV yield after isolation. From these databases it is shown that the most used EV isolation methods are not per definition the best methods based on EV purity or EV yield, indicating that more factors play a role in the choice which EV isolation method to choose than only the efficacy of the method. From the included studies it is shown that ~60% of all the included EV isolations were performed with combined EV isolation methods. The majority of EV isolations were performed with differential ultracentrifugation alone or in combination with differential ultrafiltration. When efficacy of EV isolation methods was determined in terms of EV purity and EV yield, combined EV isolation methods clearly outperformed single EV isolation methods, regardless of the type of starting material used. A recommended starting point would be the use of size-exclusion chromatography since this method, especially when combined with low-speed centrifugation, resulted in the highest EV purity, while still providing a reasonable EV yield.
Collapse
Affiliation(s)
- Janine Stam
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, the Netherlands
| | - Sabine Bartel
- Department of Pathology and Medical Biology, GRIAC Research Institute, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Rainer Bischoff
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, the Netherlands
| | - Justina C Wolters
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
203
|
Dalirfardouei R, Gholoobi A, Vahabian M, Mahdipour E, Afzaljavan F. Therapeutic role of extracellular vesicles derived from stem cells in cutaneous wound models: A systematic review. Life Sci 2021; 273:119271. [PMID: 33652035 DOI: 10.1016/j.lfs.2021.119271] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 02/07/2021] [Accepted: 02/18/2021] [Indexed: 02/07/2023]
Abstract
A growing body of evidence has shown that extracellular vesicles can be efficient as experimental therapeutics in pre-clinical models of skin wounds, but there is a significant unmet need to translate this to clinical utilization. The objectives of the current systematic review were to identify the strength of the therapeutic effects of EVs derived from stem cells in cutaneous wounds and to assess which EV-mediated mechanisms could be involved in the therapeutic response. PubMed, ISI Web of Science, and Scopus databases were systematically searched. We retrieved English-language articles published through June 2020. In vivo studies which applied stem cell-derived EVs were included for further analysis. The Risk of bias was assessed by the SYRCLE tool. We identified thirty-nine pre-clinical studies that evaluated the effects of EVs on the wound healing process. The included studies varied greatly in EVs isolation techniques, route of administration, EVs producing cells, and follow-up time. In vivo application revealed beneficial effects of EVs on accelerating wound closure and re-epithelialization in a dose-dependent manner. Elevated angiogenesis was reported in twelve eligible studies through multiple signaling pathways such as PI3K/Akt, MAPK/ERK, and JAK/STAT. The well-known signaling pathway to inhibit scar formation was TGF-β2/SMAD2. However, all included studies were not blinded enough which may have introduced bias. Therefore, the transition of EV's efficacy into the clinics is deeply rooted in the following important factors: 1) pre-clinical studies with a lower risk of bias and longer follow-up time, and 2) consistent, reproducible, and feasible manufacturing of EVs production in a large-scale commercial program.
Collapse
Affiliation(s)
- Razieh Dalirfardouei
- Research center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Aida Gholoobi
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehrangiz Vahabian
- Department of English Language and Persian Literature, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Elahe Mahdipour
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fahimeh Afzaljavan
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
204
|
Kim K, Park J, Jung JH, Lee R, Park JH, Yuk JM, Hwang H, Yeon JH. Cyclic tangential flow filtration system for isolation of extracellular vesicles. APL Bioeng 2021; 5:016103. [PMID: 33688618 PMCID: PMC7932758 DOI: 10.1063/5.0037768] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 01/27/2021] [Indexed: 02/07/2023] Open
Abstract
Size-based filtration techniques have been developed for high-throughput isolation of extracellular vesicles (EVs). Conventional direct filtration systems have limitations in that large particles generally not only block the pores of the membrane but also damage the particles because of the high fluid pressure. Here, we propose a cyclic tangential flow filtration (TFF) system that includes two membranes with pore sizes of 200 and 30 nm, connected to a peristaltic pump that feeds the stream flowing to the membrane for continuous circulation. The cyclic TFF system is better able to isolate the specific 30–200 nm size range in one step through dual cyclic filtration compared with direct filtration (DF) and single cyclic TFF (scTFF). We further introduced a buffer-exchange process to the dcTFF system after filtration to remove contaminants for more efficient purification. As a result of comparative evaluation of dcTFF and ExoQuick, EVs isolated by dcTFF had more abundant exosome markers and active EVs. The cyclic TFF system not only has great potential to separate EVs with high selectivity and separation efficiency in small volumes of samples but can also be used in clinical applications, including medical diagnostic procedures.
Collapse
Affiliation(s)
- Kimin Kim
- Department of Integrative Biosciences, University of Brain Education, Cheonan 31228, Republic of Korea
| | - Jungjae Park
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34051, Republic of Korea
| | - Jik-Han Jung
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34051, Republic of Korea
| | - Ruri Lee
- BBB Inc., Seoul 05637, Republic of Korea
| | - Ji-Ho Park
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34051, Republic of Korea
| | - Jong Min Yuk
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34051, Republic of Korea
| | | | - Ju Hun Yeon
- Department of Integrative Biosciences, University of Brain Education, Cheonan 31228, Republic of Korea
| |
Collapse
|
205
|
Izci M, Maksoudian C, Manshian BB, Soenen SJ. The Use of Alternative Strategies for Enhanced Nanoparticle Delivery to Solid Tumors. Chem Rev 2021; 121:1746-1803. [PMID: 33445874 PMCID: PMC7883342 DOI: 10.1021/acs.chemrev.0c00779] [Citation(s) in RCA: 216] [Impact Index Per Article: 72.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Indexed: 02/08/2023]
Abstract
Nanomaterial (NM) delivery to solid tumors has been the focus of intense research for over a decade. Classically, scientists have tried to improve NM delivery by employing passive or active targeting strategies, making use of the so-called enhanced permeability and retention (EPR) effect. This phenomenon is made possible due to the leaky tumor vasculature through which NMs can leave the bloodstream, traverse through the gaps in the endothelial lining of the vessels, and enter the tumor. Recent studies have shown that despite many efforts to employ the EPR effect, this process remains very poor. Furthermore, the role of the EPR effect has been called into question, where it has been suggested that NMs enter the tumor via active mechanisms and not through the endothelial gaps. In this review, we provide a short overview of the EPR and mechanisms to enhance it, after which we focus on alternative delivery strategies that do not solely rely on EPR in itself but can offer interesting pharmacological, physical, and biological solutions for enhanced delivery. We discuss the strengths and shortcomings of these different strategies and suggest combinatorial approaches as the ideal path forward.
Collapse
Affiliation(s)
- Mukaddes Izci
- NanoHealth
and Optical Imaging Group, Translational Cell and Tissue Research
Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Christy Maksoudian
- NanoHealth
and Optical Imaging Group, Translational Cell and Tissue Research
Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Bella B. Manshian
- Translational
Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Stefaan J. Soenen
- NanoHealth
and Optical Imaging Group, Translational Cell and Tissue Research
Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| |
Collapse
|
206
|
Slyusarenko M, Nikiforova N, Sidina E, Nazarova I, Egorov V, Garmay Y, Merdalimova A, Yevlampieva N, Gorin D, Malek A. Formation and Evaluation of a Two-Phase Polymer System in Human Plasma as a Method for Extracellular Nanovesicle Isolation. Polymers (Basel) 2021; 13:polym13030458. [PMID: 33572666 PMCID: PMC7867002 DOI: 10.3390/polym13030458] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 01/23/2021] [Accepted: 01/27/2021] [Indexed: 12/31/2022] Open
Abstract
The aim of the study was to explore the polyethylene glycol-dextran two-phase polymer system formed in human plasma to isolate the exosome-enriched fraction of plasma extracellular nanovesicles (ENVs). Systematic analysis was performed to determine the optimal combination of the polymer mixture parameters (molecular mass and concentration) that resulted in phase separation. The separated phases were analyzed by nanoparticle tracking analysis and Raman spectroscopy. The isolated vesicles were characterized by atomic force microscopy and dot blotting. In conclusion, the protein and microRNA contents of the isolated ENVs were assayed by flow cytometry and by reverse transcription followed by quantitative polymerase chain reaction (RT-qPCR), respectively. The presented results revealed the applicability of a new method for plasma ENV isolation and further analysis with a diagnostic purpose.
Collapse
Affiliation(s)
- Maria Slyusarenko
- Subcellular Technology Lab, N.N. Petrov National Medical Research Center of Oncology, 197758 St. Petersburg, Russia; (M.S.); (N.N.); (E.S.); (I.N.)
- The Faculty of Physics, Saint-Petersburg State University, 199034 St. Petersburg, Russia;
- Oncosystem Ltd., 121205 Moscow, Russia
| | - Nadezhda Nikiforova
- Subcellular Technology Lab, N.N. Petrov National Medical Research Center of Oncology, 197758 St. Petersburg, Russia; (M.S.); (N.N.); (E.S.); (I.N.)
- Oncosystem Ltd., 121205 Moscow, Russia
| | - Elena Sidina
- Subcellular Technology Lab, N.N. Petrov National Medical Research Center of Oncology, 197758 St. Petersburg, Russia; (M.S.); (N.N.); (E.S.); (I.N.)
- Oncosystem Ltd., 121205 Moscow, Russia
| | - Inga Nazarova
- Subcellular Technology Lab, N.N. Petrov National Medical Research Center of Oncology, 197758 St. Petersburg, Russia; (M.S.); (N.N.); (E.S.); (I.N.)
- Oncosystem Ltd., 121205 Moscow, Russia
| | - Vladimir Egorov
- Department of Molecular and Radiation Biophysics, Petersburg Nuclear Physics Institute Named by B. P. Konstantinov of National Research Center “Kurchatov Institute”, 188300 Gatchina, Russia; (V.E.); (Y.G.)
| | - Yuri Garmay
- Department of Molecular and Radiation Biophysics, Petersburg Nuclear Physics Institute Named by B. P. Konstantinov of National Research Center “Kurchatov Institute”, 188300 Gatchina, Russia; (V.E.); (Y.G.)
| | - Anastasiia Merdalimova
- Center for Photonics and Quantum Materials, Skolkovo Institute of Science and Technology, 121205 Moscow, Russia; (A.M.); (D.G.)
| | - Natalia Yevlampieva
- The Faculty of Physics, Saint-Petersburg State University, 199034 St. Petersburg, Russia;
| | - Dmitry Gorin
- Center for Photonics and Quantum Materials, Skolkovo Institute of Science and Technology, 121205 Moscow, Russia; (A.M.); (D.G.)
| | - Anastasia Malek
- Subcellular Technology Lab, N.N. Petrov National Medical Research Center of Oncology, 197758 St. Petersburg, Russia; (M.S.); (N.N.); (E.S.); (I.N.)
- Oncosystem Ltd., 121205 Moscow, Russia
- Correspondence: ; Tel.: +(7)-960-250-46-80
| |
Collapse
|
207
|
Shi J, Zhao YC, Niu ZF, Fan HJ, Hou SK, Guo XQ, Sang L, Lv Q. Mesenchymal stem cell-derived small extracellular vesicles in the treatment of human diseases: Progress and prospect. World J Stem Cells 2021; 13:49-63. [PMID: 33584979 PMCID: PMC7859991 DOI: 10.4252/wjsc.v13.i1.49] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 11/02/2020] [Accepted: 11/12/2020] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are self-renewing, multipotent cells that could differentiate into multiple tissues. MSC-based therapy has become an attractive and promising strategy for treating human diseases through immune regulation and tissue repair. However, accumulating data have indicated that MSC-based therapeutic effects are mainly attributed to the properties of the MSC-sourced secretome, especially small extracellular vesicles (sEVs). sEVs are signaling vehicles in intercellular communication in normal or pathological conditions. sEVs contain natural contents, such as proteins, mRNA, and microRNAs, and transfer these functional contents to adjacent cells or distant cells through the circulatory system. MSC-sEVs have drawn much attention as attractive agents for treating multiple diseases. The properties of MSC-sEVs include stability in circulation, good biocompatibility, and low toxicity and immunogenicity. Moreover, emerging evidence has shown that MSC-sEVs have equal or even better treatment efficacies than MSCs in many kinds of disease. This review summarizes the current research efforts on the use of MSC-sEVs in the treatment of human diseases and the existing challenges in their application from lab to clinical practice that need to be considered.
Collapse
Affiliation(s)
- Jie Shi
- Institute of Disaster Medicine, Tianjin University, Tianjin 300072, China
- Department of Biomaterials and Regenrative Medicine, Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Yu-Chen Zhao
- Institute of Disaster Medicine, Tianjin University, Tianjin 300072, China
- Department of Biomaterials and Regenrative Medicine, Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Zhi-Fang Niu
- General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Hao-Jun Fan
- Institute of Disaster Medicine, Tianjin University, Tianjin 300072, China
- Department of Biomaterials and Regenrative Medicine, Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Shi-Ke Hou
- Institute of Disaster Medicine, Tianjin University, Tianjin 300072, China
- Department of Biomaterials and Regenrative Medicine, Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Xiao-Qin Guo
- Institute of Disaster Medicine, Tianjin University, Tianjin 300072, China
- Department of Biomaterials and Regenrative Medicine, Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Lu Sang
- Institute of Disaster Medicine, Tianjin University, Tianjin 300072, China
- Department of Biomaterials and Regenrative Medicine, Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Qi Lv
- Institute of Disaster Medicine, Tianjin University, Tianjin 300072, China
- Department of Biomaterials and Regenrative Medicine, Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| |
Collapse
|
208
|
Almiñana C, Rudolf Vegas A, Tekin M, Hassan M, Uzbekov R, Fröhlich T, Bollwein H, Bauersachs S. Isolation and Characterization of Equine Uterine Extracellular Vesicles: A Comparative Methodological Study. Int J Mol Sci 2021; 22:ijms22020979. [PMID: 33478136 PMCID: PMC7835857 DOI: 10.3390/ijms22020979] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 01/15/2021] [Accepted: 01/16/2021] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles (EVs) have been identified in the uterine fluid in different species and have been pointed as key players in the embryo-maternal dialogue, maternal recognition of pregnancy and establishment of pregnancy. However, little is known about the uterine EVs in the mare. Therefore, the present study aimed at characterizing EVs from uterine lavage of cyclic mares by comparing five EVs isolation methods and the combination of them: (1) ultracentrifugation (UC); (2) concentration of lavage volume by Centricon ultrafiltration (CE); (3) the use of CE with different washing steps (phosphate-buffered saline with or without trehalose); (4) size-exclusion chromatography with iZON-qEV columns, and (5) a combination of the methods with best results based on EVs yield, purity, and protein cargo profiles. Transmission electron microscopy and Western blotting confirmed the isolation of EVs by all methods but with quantitative and qualitative differences. Mass spectrometry provided differences in protein profiles between methods, number of identified proteins, and protein classes. Our results indicate that the combination of CE/trehalose/iZON/UC is an optimal method to isolate equine uterine EVs with good yield and purity that can be applied in future studies to determine the role of equine uterine EVs in embryo-maternal interactions.
Collapse
Affiliation(s)
- Carmen Almiñana
- Functional Genomics Group, Institute of Veterinary Anatomy, Vetsuisse Faculty Zurich, University of Zurich, 8315 Lindau, Switzerland; (A.R.V.); (S.B.)
- UMR85 PRC, INRAE, CNRS 7247, Université de Tours, IFCE, 37380 Nouzilly, France
- Correspondence:
| | - Alba Rudolf Vegas
- Functional Genomics Group, Institute of Veterinary Anatomy, Vetsuisse Faculty Zurich, University of Zurich, 8315 Lindau, Switzerland; (A.R.V.); (S.B.)
- Clinic of Reproductive Medicine, Department for Farm Animals, Vetsuisse-Faculty, University of Zurich, 8057 Zurich, Switzerland; (M.T.); (M.H.); (H.B.)
| | - Muhittin Tekin
- Clinic of Reproductive Medicine, Department for Farm Animals, Vetsuisse-Faculty, University of Zurich, 8057 Zurich, Switzerland; (M.T.); (M.H.); (H.B.)
| | - Mubbashar Hassan
- Clinic of Reproductive Medicine, Department for Farm Animals, Vetsuisse-Faculty, University of Zurich, 8057 Zurich, Switzerland; (M.T.); (M.H.); (H.B.)
| | - Rustem Uzbekov
- Laboratoire Biologie Cellulaire et Microscopie Electronique, Faculté de Médecine, Université de Tours, 37032 Tours, France;
- Faculty of Bioengineering and Bioinformatics, Moscow State University, 119992 Moscow, Russia
| | - Thomas Fröhlich
- Gene Center, Laboratory for Functional Genome Analysis, LMU Munich, 81377 Munich, Germany;
| | - Heinrich Bollwein
- Clinic of Reproductive Medicine, Department for Farm Animals, Vetsuisse-Faculty, University of Zurich, 8057 Zurich, Switzerland; (M.T.); (M.H.); (H.B.)
| | - Stefan Bauersachs
- Functional Genomics Group, Institute of Veterinary Anatomy, Vetsuisse Faculty Zurich, University of Zurich, 8315 Lindau, Switzerland; (A.R.V.); (S.B.)
| |
Collapse
|
209
|
Yousif G, Qadri S, Haik M, Haik Y, Parray AS, Shuaib A. Circulating Exosomes of Neuronal Origin as Potential Early Biomarkers for Development of Stroke. Mol Diagn Ther 2021; 25:163-180. [DOI: 10.1007/s40291-020-00508-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/10/2020] [Indexed: 12/11/2022]
|
210
|
Liu J, Chen Y, Pei F, Zeng C, Yao Y, Liao W, Zhao Z. Extracellular Vesicles in Liquid Biopsies: Potential for Disease Diagnosis. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6611244. [PMID: 33506022 PMCID: PMC7814955 DOI: 10.1155/2021/6611244] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/19/2020] [Accepted: 12/23/2020] [Indexed: 02/05/2023]
Abstract
Liquid biopsy is conducted through minimally invasive or noninvasive procedures, and the resulting material can be subjected to genomic, proteomic, and lipidomic analyses for early diagnosis of cancers and other diseases. Extracellular vesicles (EVs), one kind of promising tool for liquid biopsy, are nanosized bilayer particles that are secreted by all kinds of cells and that carry cargoes such as lipids, proteins, and nucleic acids, protecting them from enzymatic degradation in the extracellular environment. In this review, we provide a comprehensive introduction to the properties and applications of EVs, including their biogenesis, contents, sample collection, isolation, and applications in diagnostics based on liquid biopsy.
Collapse
Affiliation(s)
- Jialing Liu
- Department of Orthodontics, West China Hospital of Stomatology, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
| | - Ye Chen
- Department of Orthodontics, West China Hospital of Stomatology, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
| | - Fang Pei
- Department of Orthodontics, West China Hospital of Stomatology, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
| | - Chongmai Zeng
- Department of Orthodontics, West China Hospital of Stomatology, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
| | - Yang Yao
- Department of Implantology, West China Hospital of Stomatology, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
| | - Wen Liao
- Department of Orthodontics, West China Hospital of Stomatology, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
| | - Zhihe Zhao
- Department of Orthodontics, West China Hospital of Stomatology, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
| |
Collapse
|
211
|
Exosome detection via the ultrafast-isolation system: EXODUS. Nat Methods 2021; 18:212-218. [PMID: 33432243 DOI: 10.1038/s41592-020-01034-x] [Citation(s) in RCA: 161] [Impact Index Per Article: 53.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 11/30/2020] [Indexed: 12/17/2022]
Abstract
Exosomes have shown great potential in disease diagnostics and therapeutics. However, current isolation approaches are burdensome and suffer from low speed, yield and purity, limiting basic research and clinical applications. Here, we describe an efficient exosome detection method via the ultrafast-isolation system (EXODUS) that allows automated label-free purification of exosomes from varied biofluids. We obtained the ultra-efficient purification of exosomes by negative pressure oscillation and double coupled harmonic oscillator-enabled membrane vibration. Our two coupled oscillators generate dual-frequency transverse waves on the membranes, enabling EXODUS to outperform other isolation techniques in speed, purity and yield. We demonstrated EXODUS by purifying exosomes from urine samples of 113 patients and validated the practical relevance in exosomal RNA profiling with the high-resolution capability and high-throughput analysis.
Collapse
|
212
|
Gebeyehu A, Kommineni N, Bagde A, Meckes DG, Sachdeva MS. Role of Exosomes for Delivery of Chemotherapeutic Drugs. Crit Rev Ther Drug Carrier Syst 2021; 38:53-97. [PMID: 34375513 PMCID: PMC8691065 DOI: 10.1615/critrevtherdrugcarriersyst.2021036301] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Exosomes are endogenous extracellular vesicles (30-100 nm) composed with membrane lipid bilayer which carry vesicular proteins, enzymes, mRNA, miRNA and nucleic acids. They act as messengers for intra- and inter-cellular communication. In addition to their physiological roles, exosomes have the potential to encapsulate and deliver small chemotherapeutic drugs and biological molecules such as proteins and nucleic acid-based drugs to the recipient tissue or organs. Due to their biological properties, exosomes have better organotropism, homing capacity, cellular uptake and cargo release ability than other synthetic nano-drug carriers such as liposomes, micelles and nanogels. The secretion of tumor-derived exosomes is increased in the hypoxic and acidic tumor microenvironment, which can be used as a target for nontoxic and nonimmunogenic drug delivery vehicles for various cancers. Moreover, exosomes have the potential to carry both hydrophilic and hydrophobic chemotherapeutic drugs, bypass RES effect and bypass BBB. Exosomes can be isolated from other types of EVs and cell debris based on their size, density and specific surface proteins through ultracentrifugation, density gradient separation, precipitation, immunoaffinity interaction and gel filtration. Drugs can be loaded into exosomes at the biogenesis stage or with the isolated exosomes by incubation, electroporation, extrusion or sonication methods. Finally, exosomal cargo vehicles can be characterized by ultrastructural microscopic analysis. In this review we intend to summarize the inception, structure and function of the exosomes, role of exosomes in immunological regulation and cancer, methods of isolation and characterization of exosomes and products under clinical trials. This review will provide an inclusive insight of exosomes in drug delivery.
Collapse
Affiliation(s)
- Aragaw Gebeyehu
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Nagavendra Kommineni
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Arvind Bagde
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - David G. Meckes
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, USA
| | - Mandip Singh Sachdeva
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| |
Collapse
|
213
|
Kumar A, Zhou L, Zhi K, Raji B, Pernell S, Tadrous E, Kodidela S, Nookala A, Kochat H, Kumar S. Challenges in Biomaterial-Based Drug Delivery Approach for the Treatment of Neurodegenerative Diseases: Opportunities for Extracellular Vesicles. Int J Mol Sci 2020; 22:E138. [PMID: 33375558 PMCID: PMC7795247 DOI: 10.3390/ijms22010138] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/19/2020] [Accepted: 12/21/2020] [Indexed: 02/06/2023] Open
Abstract
Biomaterials have been the subject of numerous studies to pursue potential therapeutic interventions for a wide variety of disorders and diseases. The physical and chemical properties of various materials have been explored to develop natural, synthetic, or semi-synthetic materials with distinct advantages for use as drug delivery systems for the central nervous system (CNS) and non-CNS diseases. In this review, an overview of popular biomaterials as drug delivery systems for neurogenerative diseases is provided, balancing the potential and challenges associated with the CNS drug delivery. As an effective drug delivery system, desired properties of biomaterials are discussed, addressing the persistent challenges such as targeted drug delivery, stimuli responsiveness, and controlled drug release in vivo. Finally, we discuss the prospects and limitations of incorporating extracellular vesicles (EVs) as a drug delivery system and their use for biocompatible, stable, and targeted delivery with limited immunogenicity, as well as their ability to be delivered via a non-invasive approach for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Asit Kumar
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (L.Z.); (S.P.); (E.T.); (S.K.)
| | - Lina Zhou
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (L.Z.); (S.P.); (E.T.); (S.K.)
| | - Kaining Zhi
- Plough Center for Sterile Drug Delivery Solutions, University of Tennessee Health Science Center, Memphis, TN 38104, USA; (K.Z.); (B.R.); (H.K.)
| | - Babatunde Raji
- Plough Center for Sterile Drug Delivery Solutions, University of Tennessee Health Science Center, Memphis, TN 38104, USA; (K.Z.); (B.R.); (H.K.)
| | - Shelby Pernell
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (L.Z.); (S.P.); (E.T.); (S.K.)
| | - Erene Tadrous
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (L.Z.); (S.P.); (E.T.); (S.K.)
| | - Sunitha Kodidela
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (L.Z.); (S.P.); (E.T.); (S.K.)
| | | | - Harry Kochat
- Plough Center for Sterile Drug Delivery Solutions, University of Tennessee Health Science Center, Memphis, TN 38104, USA; (K.Z.); (B.R.); (H.K.)
| | - Santosh Kumar
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (L.Z.); (S.P.); (E.T.); (S.K.)
| |
Collapse
|
214
|
Chiabotto G, Pasquino C, Camussi G, Bruno S. Molecular Pathways Modulated by Mesenchymal Stromal Cells and Their Extracellular Vesicles in Experimental Models of Liver Fibrosis. Front Cell Dev Biol 2020; 8:594794. [PMID: 33425900 PMCID: PMC7794013 DOI: 10.3389/fcell.2020.594794] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/06/2020] [Indexed: 12/18/2022] Open
Abstract
End-stage liver fibrosis is common to all chronic liver diseases. Since liver transplantation has several limitations, including lack of donors, immunological rejection, and high medical costs, therapeutic alternatives are needed. The administration of mesenchymal stromal cells (MSCs) has been proven effective in tissue regeneration after damage. However, the risk of uncontrolled side effects, such as cellular rejection and tumorigenesis, should be taken into consideration. A safer alternative to MSC transplantation is represented by the MSC secretome, which retains the same beneficial effect of the cell of origin, without showing any considerable side effect. The paracrine effect of MSCs is mainly carried out by secreted particles in the nanometer range, known as extracellular vesicles (EVs) that play a fundamental role in intercellular communication. In this review, we discuss the current literature on MSCs and MSC-EVs, focusing on their potential therapeutic action in liver fibrosis and on their molecular content (proteins and RNA), which contributes in reverting fibrosis and prompting tissue regeneration.
Collapse
Affiliation(s)
- Giulia Chiabotto
- Department of Medical Sciences, University of Turin, Turin, Italy.,Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - Chiara Pasquino
- Department of Medical Sciences, University of Turin, Turin, Italy.,Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - Giovanni Camussi
- Department of Medical Sciences, University of Turin, Turin, Italy.,Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - Stefania Bruno
- Department of Medical Sciences, University of Turin, Turin, Italy.,Molecular Biotechnology Center, University of Turin, Turin, Italy
| |
Collapse
|
215
|
Liangsupree T, Multia E, Riekkola ML. Modern isolation and separation techniques for extracellular vesicles. J Chromatogr A 2020; 1636:461773. [PMID: 33316564 DOI: 10.1016/j.chroma.2020.461773] [Citation(s) in RCA: 256] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/26/2020] [Accepted: 11/28/2020] [Indexed: 02/07/2023]
Abstract
Extracellular vesicles (EVs) are heterogenous membrane-bound vesicles released from various origins. EVs play a crucial role in cellular communication and mediate several physiological and pathological processes, highlighting their potential therapeutic and diagnostic applications. Due to the rapid increase in interests and needs to elucidate EV properties and functions, numerous isolation and separation approaches for EVs have been developed to overcome limitations of conventional techniques, such as ultracentrifugation. This review focuses on recently emerging and modern EV isolation and separation techniques, including size-, charge-, and affinity-based techniques while excluding ultracentrifugation and precipitation-based techniques due to their multiple limitations. The advantages and drawbacks of each technique are discussed together with insights into their applications. Emerging approaches all share similar features in terms of being time-effective, easy-to-operate, and capable of providing EVs with suitable and desirable purity and integrity for applications of interest. Combination and hyphenation of techniques have been used for EV isolation and separation to yield EVs with the best quality. The most recent development using an automated on-line system including selective affinity-based trapping unit and asymmetrical flow field-flow fractionation allows reliable isolation and fractionation of EV subpopulations from human plasma.
Collapse
Affiliation(s)
| | - Evgen Multia
- Department of Chemistry, P.O. Box 55, FI-00014 University of Helsinki, Finland
| | | |
Collapse
|
216
|
Nam GH, Choi Y, Kim GB, Kim S, Kim SA, Kim IS. Emerging Prospects of Exosomes for Cancer Treatment: From Conventional Therapy to Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2002440. [PMID: 33015883 DOI: 10.1002/adma.202002440] [Citation(s) in RCA: 167] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/29/2020] [Indexed: 05/05/2023]
Abstract
Exosomes are a class of extracellular vesicles of around 100 nm in diameter that are secreted by most cells and contain various bioactive molecules reflecting their cellular origin and mediate intercellular communication. Studies of these exosomal features in tumor pathogenesis have led to the development of therapeutic and diagnostic approaches using exosomes for cancer therapy. Exosomes have many advantages for conveying therapeutic agents such as small interfering RNAs, microRNAs, membrane-associated proteins, and chemotherapeutic compounds; thus, they are considered a prime candidate as a delivery tool for cancer treatment. Since exosomes also provide an optimal microenvironment for the effective function of immunomodulatory factors, exosomes harboring bioactive molecules have been bioengineered as cancer immunotherapies that can effectively activate each stage of the cancer immunity cycle to successfully elicit cancer-specific immunity. This review discusses the advantages of exosomes for treating cancer and the challenges that must be overcome for their successful clinical development.
Collapse
Affiliation(s)
- Gi-Hoon Nam
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Yoonjeong Choi
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Gi Beom Kim
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Seohyun Kim
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Seong A Kim
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - In-San Kim
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| |
Collapse
|
217
|
Ali M, Pham A, Wang X, Wolfram J, Pham S. Extracellular vesicles for treatment of solid organ ischemia-reperfusion injury. Am J Transplant 2020; 20:3294-3307. [PMID: 32594616 DOI: 10.1111/ajt.16164] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/10/2020] [Accepted: 06/17/2020] [Indexed: 01/25/2023]
Abstract
As the incidence of ischemia-reperfusion (I-R) injury has substantially increased, there is a pressing need to develop effective strategies to treat this global health issue. I-R injury can affect all organs and is associated with high morbidity and mortality rates. Pathological settings such as myocardial infarction, stroke, hemorrhagic shock, and solid organ transplant are particularly prone to cause I-R injury. Ischemia (hypoxia) and/or reperfusion (reoxygenation) induces various forms of cellular and structural damage. A major cause of damage is local inflammatory responses, which may spread to produce more advanced systemic inflammation. Management of I-R injury relies primarily on supportive measures, as specific treatment strategies are lacking. Extracellular vesicles (EVs) are cell-secreted nano-scale structures containing various biomolecules involved in cell communication and multiple physiological processes. EVs derived from certain cell types have been shown to exhibit anti-inflammatory, antioxidant, and angiogenic properties. This review provides an overview of EV-based therapeutics for I-R injury in kidneys, liver, heart, lungs, and brain. Additionally, the mechanisms by which EVs protect against I-R injury are discussed. Promising preclinical findings highlight the potential clinical use of EVs for I-R injury.
Collapse
Affiliation(s)
- Mojahid Ali
- Department of Cardiothoracic Surgery, Mayo Clinic, Jacksonville, Florida, USA.,Department of Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, Florida, USA
| | - Anthony Pham
- Department of Cardiothoracic Surgery, Mayo Clinic, Jacksonville, Florida, USA.,Department of Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, Florida, USA
| | - Xinghua Wang
- Department of Cardiothoracic Surgery, Mayo Clinic, Jacksonville, Florida, USA.,Department of Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, Florida, USA
| | - Joy Wolfram
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, Florida, USA.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, Florida, USA.,Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas, USA
| | - Si Pham
- Department of Cardiothoracic Surgery, Mayo Clinic, Jacksonville, Florida, USA
| |
Collapse
|
218
|
Mesenchymal stem cell derived-exosomes: a modern approach in translational medicine. J Transl Med 2020; 18:449. [PMID: 33246476 PMCID: PMC7691969 DOI: 10.1186/s12967-020-02622-3] [Citation(s) in RCA: 207] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 11/18/2020] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have captured great attention in regenerative medicine for over a few decades by virtue of their differentiation capacity, potent immunomodulatory properties, and their ability to be favorably cultured and manipulated. Recent investigations implied that the pleiotropic effects of MSCs is not associated to their ability of differentiation, but rather is mediated by the secretion of soluble paracrine factors. Exosomes, nanoscale extracellular vesicles, are one of these paracrine mediators. Exosomes transfer functional cargos like miRNA and mRNA molecules, peptides, proteins, cytokines and lipids from MSCs to the recipient cells. Exosomes participate in intercellular communication events and contribute to the healing of injured or diseased tissues and organs. Studies reported that exosomes alone are responsible for the therapeutic effects of MSCs in numerous experimental models. Therefore, MSC-derived exosomes can be manipulated and applied to establish a novel cell-free therapeutic approach for treatment of a variety of diseases including heart, kidney, liver, immune and neurological diseases, and cutaneous wound healing. In comparison with their donor cells, MSC-derived exosomes offer more stable entities and diminished safety risks regarding the administration of live cells, e.g. microvasculature occlusion risk. This review discusses the exosome isolation methods invented and utilized in the clinical setting thus far and presents a summary of current information on MSC exosomes in translational medicine.
Collapse
|
219
|
Tiwari S, Kumar V, Randhawa S, Verma SK. Preparation and characterization of extracellular vesicles. Am J Reprod Immunol 2020; 85:e13367. [PMID: 33118232 DOI: 10.1111/aji.13367] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 10/14/2020] [Accepted: 10/16/2020] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles (EVs) are heterogeneous membranous vesicles secreted by every cell type and offer significant potential in therapy and diagnostics. Differential ultracentrifugation is the gold standard for EV isolation, although other techniques including, polyethylene glycol (PEG) precipitation, immunoprecipitation, size exclusion chromatography, and immuno-isolation approaches are common. Purified EVs can be characterized based on their physical characteristics, biochemical composition, or cell of origin. For size and concentration measurement, nanoparticle tracking analysis (NTA), dynamic light scattering (DLS), and electron microscopy are commonly employed methods. Biochemical analyses of EVs are typically performed using flow cytometry, immunoblotting, or proteomic investigation. Based on tissue of origin, EVs have specific markers that can be used to isolate and purify specific cell-associated EVs using an affinity selection approach. Despite existence of several methods for isolation and characterization, major limitations associated with each method hinder the progress of the field. Evolving concepts in EV biology possess great promise for better isolation and characterization leading to a better insight of biological function and have immense clinical implications. In this review, we discuss recent advancements in EV isolation and characterization approaches.
Collapse
Affiliation(s)
- Swasti Tiwari
- Department of Molecular Medicine & Biotechnology, Sanjay Gandhi PGI, Lucknow, India
| | - Vinod Kumar
- Department of Molecular Medicine & Biotechnology, Sanjay Gandhi PGI, Lucknow, India
| | | | - Santosh K Verma
- Department of Molecular Medicine & Biotechnology, Sanjay Gandhi PGI, Lucknow, India
| |
Collapse
|
220
|
Gupta M, Tieu A, Slobodian M, Shorr R, Burger D, Lalu MM, Allan DS. Preclinical Studies of MSC-Derived Extracellular Vesicles to Treat or Prevent Graft Versus Host Disease: a Systematic Review of the Literature. Stem Cell Rev Rep 2020; 17:332-340. [PMID: 33159616 PMCID: PMC7648545 DOI: 10.1007/s12015-020-10058-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2020] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Treating and preventing graft-versus-host disease (GVHD) after allogeneic hematopoietic cell transplant (HCT) remains a significant challenge. The use of mesenchymal stromal cell-derived extracellular vesicles (MSC-EVs) appears promising and a systematic review of preclinical studies is needed to accelerate the design of translational studies. METHODS We identified 4 eligible studies from a systematic review performed on December 1, 2018. In brief, eligible studies included the treatment or prevention of GVHD in animal models and the use of MSC-EVs. Study design and outcome data were extracted and reporting was evaluated using the SYRCLE tool to identify potential bias. RESULTS Two studies assessed the efficacy of MSC-EVs in treatment of GVHD and 2 studies address prevention. Mice treated with MSC-EVs showed improved median survival, GVHD clinical scores and histology scores as compared to untreated mice with GVHD. Prophylactic treatment with MSC-EVs attenuated GVHD severity and improved median survival as compared to no treatment or saline. CONCLUSION Our systematic review provides important insight regarding the potential of MSC-EVs to treat or prevent GVHD. Although few studies were identified, improved survival and attenuated histologic findings of GVHD were observed in mice after MSC-EV administration for the treatment and prevention of GVHD. Dosing of EVs and route of administration remain inconsistent, however, and scalability of EV isolation for clinical studies remains a challenge. Standardized outcome reporting is needed to pool results for metanalysis. Graphical abstract.
Collapse
Affiliation(s)
- Manika Gupta
- Department of Medicine (Blood and Marrow Transplantation), The Ottawa Hospital, 501 Smyth Rd, Box 704, Ottawa, ON, K1H 8L6, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Alvin Tieu
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.,Clinical Epidemiology, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Mitchell Slobodian
- Clinical Epidemiology, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Risa Shorr
- Library Services, The Ottawa Hospital, Ottawa, ON, Canada
| | - Dylan Burger
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.,Chronic Disease Programs, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Manoj M Lalu
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.,Clinical Epidemiology, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - David S Allan
- Department of Medicine (Blood and Marrow Transplantation), The Ottawa Hospital, 501 Smyth Rd, Box 704, Ottawa, ON, K1H 8L6, Canada. .,Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada. .,Clinical Epidemiology, Ottawa Hospital Research Institute, Ottawa, ON, Canada.
| |
Collapse
|
221
|
Brain metastases-derived extracellular vesicles induce binding and aggregation of low-density lipoprotein. J Nanobiotechnology 2020; 18:162. [PMID: 33160390 PMCID: PMC7648399 DOI: 10.1186/s12951-020-00722-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 10/24/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Cancer cell-derived extracellular vesicles (EVs) have previously been shown to contribute to pre-metastatic niche formation. Specifically, aggressive tumors secrete pro-metastatic EVs that travel in the circulation to distant organs to modulate the microenvironment for future metastatic spread. Previous studies have focused on the interface between pro-metastatic EVs and epithelial/endothelial cells in the pre-metastatic niche. However, EV interactions with circulating components such as low-density lipoprotein (LDL) have been overlooked. RESULTS This study demonstrates that EVs derived from brain metastases cells (Br-EVs) and corresponding regular cancer cells (Reg-EVs) display different interactions with LDL. Specifically, Br-EVs trigger LDL aggregation, and the presence of LDL accelerates Br-EV uptake by monocytes, which are key components in the brain metastatic niche. CONCLUSIONS Collectively, these data are the first to demonstrate that pro-metastatic EVs display distinct interactions with LDL, which impacts monocyte internalization of EVs.
Collapse
|
222
|
Noren Hooten N, Yáñez‐Mó M, DeRita R, Russell A, Quesenberry P, Ramratnam B, Robbins PD, Di Vizio D, Wen S, Witwer KW, Languino LR. Hitting the Bullseye: Are extracellular vesicles on target? J Extracell Vesicles 2020; 10:e12032. [PMID: 33708359 PMCID: PMC7890543 DOI: 10.1002/jev2.12032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 10/26/2020] [Accepted: 10/31/2020] [Indexed: 12/12/2022] Open
Affiliation(s)
- Nicole Noren Hooten
- Laboratory of Epidemiology and Population ScienceNational Institute on AgingNational Institutes of HealthBaltimoreMarylandUSA
| | - María Yáñez‐Mó
- Departamento de Biología MolecularUAMCentro de Biología Molecular Severo OchoaIIS‐IPMadridSpain
| | - Rachel DeRita
- Department of Cancer BiologySidney Kimmel Cancer CenterJefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Ashley Russell
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Peter Quesenberry
- Division of Hematology/OncologyThe Warren Alpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| | - Bharat Ramratnam
- Department of MedicineAlpert Medical SchoolBrown UniversityProvidenceRhode IslandUSA
| | - Paul D. Robbins
- Institute on the Biology of Aging and Metabolism, and Department of BiochemistryMolecular Biology and Biophysics University of MinnesotaMinneapolisMinnesotaUSA
| | - Dolores Di Vizio
- Departments of SurgeryBiomedical Sciences, and Pathology & Laboratory MedicineDivision of Cancer Biology and TherapeuticsCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Sicheng Wen
- Division of Hematology/OncologyThe Warren Alpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| | - Kenneth W. Witwer
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of NeurologyRichman Family Precision Medicine Center of Excellence in Alzheimer’s DiseaseJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Lucia R. Languino
- Department of Cancer BiologySidney Kimmel Cancer CenterJefferson UniversityPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
223
|
Recent Advances in Extracellular Vesicles as Drug Delivery Systems and Their Potential in Precision Medicine. Pharmaceutics 2020; 12:pharmaceutics12111006. [PMID: 33105857 PMCID: PMC7690579 DOI: 10.3390/pharmaceutics12111006] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/15/2020] [Accepted: 10/20/2020] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles (EVs) are membrane-bilayered nanoparticles released by most cell types. Recently, an enormous number of studies have been published on the potential of EVs as carriers of therapeutic agents. In contrast to systems such as liposomes, EVs exhibit less immunogenicity and higher engineering potential. Here, we review the most relevant publications addressing the potential and use of EVs as a drug delivery system (DDS). The information is divided based on the key steps for designing an EV-mediated delivery strategy. We discuss possible sources and isolation methods of EVs. We address the administration routes that have been tested in vivo and the tissue distribution observed. We describe the current knowledge on EV clearance, a significant challenge towards enhancing bioavailability. Also, EV-engineering approaches are described as alternatives to improve tissue and cell-specificity. Finally, a summary of the ongoing clinical trials is performed. Although the application of EVs in the clinical practice is still at an early stage, a high number of studies in animals support their potential as DDS. Thus, better treatment options could be designed to precisely increase target specificity and therapeutic efficacy while reducing off-target effects and toxicity according to the individual requirements of each patient.
Collapse
|
224
|
Mallia A, Gianazza E, Zoanni B, Brioschi M, Barbieri SS, Banfi C. Proteomics of Extracellular Vesicles: Update on Their Composition, Biological Roles and Potential Use as Diagnostic Tools in Atherosclerotic Cardiovascular Diseases. Diagnostics (Basel) 2020; 10:diagnostics10100843. [PMID: 33086718 PMCID: PMC7588996 DOI: 10.3390/diagnostics10100843] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/13/2020] [Accepted: 10/16/2020] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EVs) are lipid-bound vesicles released from cells under physiological and pathological conditions. Basing on biogenesis, dimension, content and route of secretion, they can be classified into exosomes, microvesicles (MVs) and apoptotic bodies. EVs have a key role as bioactive mediators in intercellular communication, but they are also involved in other physiological processes like immune response, blood coagulation, and tissue repair. The interest in studying EVs has increased over the years due to their involvement in several diseases, such as cardiovascular diseases (CVDs), and their potential role as biomarkers in diagnosis, therapy, and in drug delivery system development. Nowadays, the improvement of mass spectrometry (MS)-based techniques allows the characterization of the EV protein composition to deeply understand their role in several diseases. In this review, a critical overview is provided on the EV’s origin and physical properties, as well as their emerging functional role in both physiological and disease conditions, focusing attention on the role of exosomes in CVDs. The most important cardiac exosome proteomic studies will be discussed giving a qualitative and quantitative characterization of the exosomal proteins that could be used in future as new potential diagnostic markers or targets for specific therapies.
Collapse
|
225
|
Challenges in the development and establishment of exosome-based drug delivery systems. J Control Release 2020; 329:894-906. [PMID: 33058934 DOI: 10.1016/j.jconrel.2020.10.020] [Citation(s) in RCA: 166] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/09/2020] [Accepted: 10/10/2020] [Indexed: 12/16/2022]
Abstract
Exosomes are extracellular vesicles released from cells and are characterized by a lipid bilayer membrane encapsulating a variety of biological molecules such as nucleic acids or proteins within the lumen or the lipid-bilayer. Under physiological environments, exosomes mediate cell-to-cell communication and cargo transport. Therefore, exosomes have been explored as drug delivery vehicles for improving therapeutic outcomes. Although recent studies have demonstrated promising advances with exosome-based drug delivery systems, several challenges severely hinder further development of exosomes for clinical applications. This review summarizes and emphasizes some of the technical challenges related to the isolation, characterization, and stability testing of exosomes. More importantly, challenges related specifically to the application of exosomes for drug delivery such as cell-uptake, drug loading, drug release, and in vivo distribution will be examined in this article.
Collapse
|
226
|
Quijano LM, Naranjo JD, El-Mossier SO, Turner NJ, Pineda Molina C, Bartolacci J, Zhang L, White L, Li H, Badylak SF. Matrix-Bound Nanovesicles: The Effects of Isolation Method upon Yield, Purity, and Function. Tissue Eng Part C Methods 2020; 26:528-540. [PMID: 33012221 PMCID: PMC7869881 DOI: 10.1089/ten.tec.2020.0243] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 09/25/2020] [Indexed: 12/11/2022] Open
Abstract
Identification of matrix-bound nanovesicles (MBV) as ubiquitous components of the extracellular matrix (ECM) raises questions regarding their biologic functions and their potential theranostic application. Unlike liquid-phase extracellular vesicles (e.g., exosomes), MBV are tightly bound to the ECM, which makes their isolation and harvesting more challenging. The indiscriminate use of different methods to harvest MBV can alter or disrupt their structural and/or functional integrity. The objective of the present study was to compare the effect of various MBV harvesting methods upon yield, purity, and biologic activity. Combinations of four methods to solubilize the ECM (collagenase [COL], liberase [LIB], or proteinase K [PK] and nonenzymatic elution with potassium chloride) and four isolation methods (ultracentrifugation, ultrafiltration [UF], density barrier, and size exclusion chromatography [SEC]) were used to isolate MBV from urinary bladder-derived ECM. All combinations of solubilization and isolation methods allowed for the harvesting of MBV, however, distinct differences were noted. The highest yield, purity, cellular uptake, and biologic activity were seen with MBV isolated by a combination of liberase or collagenase followed by SEC. The combination of proteinase K and UF was shown to have detrimental effects on bioactivity. The results show the importance of selecting appropriate MBV harvesting methods for the characterization and evaluation of MBV and for analysis of their potential theranostic application. Impact statement Identification of matrix-bound nanovesicles (MBV) as ubiquitous components of the extracellular matrix (ECM) has raised questions regarding their biologic functions and their potential theranostic application. This study demonstrates that the harvesting methods used can result in samples with physical and biochemical properties that are unique to the isolation and solubilization methods used. Consequently, developing harvesting methods that minimize sample contamination with ECM remnants and/or solubilization agents will be essential in determining the theranostic potential of MBV in future studies.
Collapse
Affiliation(s)
- Lina M. Quijano
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Juan D. Naranjo
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Salma O. El-Mossier
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Neill J. Turner
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Catalina Pineda Molina
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Joseph Bartolacci
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Li Zhang
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Lisa White
- School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | - Hui Li
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens Research Institute, Pittsburgh, Pennsylvania, USA
| | - Stephen F. Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
227
|
Hu T, Wolfram J, Srivastava S. Extracellular Vesicles in Cancer Detection: Hopes and Hypes. Trends Cancer 2020; 7:122-133. [PMID: 33008796 DOI: 10.1016/j.trecan.2020.09.003] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 09/04/2020] [Accepted: 09/08/2020] [Indexed: 02/06/2023]
Abstract
Early cancer diagnosis is critical for improving patient survival and mortality rates, but most diagnostics on solid tumors rely on imaging tests with limited sensitivity and specificity to identify potential cases, which are then confirmed by tissue biopsies. However, this process is usually not suitable for cancer screening or evaluation of tumor responses to treatment. Liquid biopsies have the potential to bridge this gap, but few such assays have been approved for cancer applications. Extracellular vesicles hold particular promise for liquid biopsy diagnostics but are currently limited by the lack of robust methods for isolation and analysis. New isolation and analysis techniques, however, show promise to improve the clinical utility of extracellular vesicle-based cancer diagnosis.
Collapse
Affiliation(s)
- Tony Hu
- Department of Biochemistry and Molecular Biology Center for Cellular and Molecular Diagnosis, School of Medicine, Tulane University, New Orleans, LA, USA.
| | - Joy Wolfram
- Department of Biochemistry and Molecular Biology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, FL, USA
| | - Sudhir Srivastava
- Cancer Biomarkers Research Group, National Cancer Institute, Rockville, MD, USA.
| |
Collapse
|
228
|
Extracellular Vesicles as an Efficient and Versatile System for Drug Delivery. Cells 2020; 9:cells9102191. [PMID: 33003285 PMCID: PMC7600121 DOI: 10.3390/cells9102191] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/24/2020] [Accepted: 05/30/2020] [Indexed: 12/12/2022] Open
Abstract
Despite the recent advances in drug development, the majority of novel therapeutics have not been successfully translated into clinical applications. One of the major factors hindering their clinical translation is the lack of a safe, non-immunogenic delivery system with high target specificity upon systemic administration. In this respect, extracellular vesicles (EVs), as natural carriers of bioactive cargo, have emerged as a promising solution and can be further modified to improve their therapeutic efficacy. In this review, we provide an overview of the biogenesis pathways, biochemical features, and isolation methods of EVs with an emphasis on their many intrinsic properties that make them desirable as drug carriers. We then describe in detail the current advances in EV therapeutics, focusing on how EVs can be engineered to achieve improved target specificity, better circulation kinetics, and efficient encapsulation of therapeutic payloads. We also identify the challenges and obstacles ahead for clinical translation and provide an outlook on the future perspective of EV-based therapeutics.
Collapse
|
229
|
Song Y, Kim Y, Ha S, Sheller-Miller S, Yoo J, Choi C, Park CH. The emerging role of exosomes as novel therapeutics: Biology, technologies, clinical applications, and the next. Am J Reprod Immunol 2020; 85:e13329. [PMID: 32846024 PMCID: PMC7900947 DOI: 10.1111/aji.13329] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/14/2020] [Accepted: 08/18/2020] [Indexed: 12/21/2022] Open
Abstract
The extracellular vesicles (EVs) research area has grown rapidly because of their pivotal roles in intercellular communications and maintaining homeostasis of individual organism. As a subtype of EVs, exosomes are made via unique biogenesis pathway and exhibit disparate functional and phenotypic characteristics. Functionally, exosomes transfer biological messages from donor cell to recipient cell, which makes exosomes as a novel therapeutic platform delivering therapeutic materials to the target tissue/cell. Currently, both academia and industry try to develop exosome platform‐based therapeutics for disease management, some of which are already in clinical trials. In this review, we will discuss focusing on therapeutic values of exosomes, recent advances in therapeutic exosome platform development, and late development of exosome therapeutics in diverse therapeutic areas.
Collapse
Affiliation(s)
| | | | - Sunhyung Ha
- ILIAS Biologics Inc, Daejeon, Republic of Korea
| | - Samantha Sheller-Miller
- Division of Maternal-Fetal Medicine & Perinatal Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch, Galveston, TX, USA
| | | | - Chulhee Choi
- ILIAS Biologics Inc, Daejeon, Republic of Korea.,Department of Bio and Brain Engineering, KAIST, Daejeon, Republic of Korea
| | | |
Collapse
|
230
|
Adlerz K, Patel D, Rowley J, Ng K, Ahsan T. Strategies for scalable manufacturing and translation of MSC-derived extracellular vesicles. Stem Cell Res 2020; 48:101978. [PMID: 32947235 DOI: 10.1016/j.scr.2020.101978] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 06/25/2020] [Accepted: 08/26/2020] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal Stem/Stromal Cells (MSCs) are a well-studied cellular therapy with many clinical trials over the last few decades to treat a range of therapeutic indications. Recently, extracellular vesicles secreted by MSCs (MSC-EVs) have been shown to recapitulate many of the therapeutic effects of the MSCs themselves. While research in MSC-EVs has exploded, it is still early in their development towards a clinical therapy. One of the main challenges in cellular therapy, which will clearly also be a challenge in MSC-EV manufacturing, is developing a scalable, cGMP-compatible manufacturing paradigm. Therefore, the focus of this review is to identify some key MSC-EV manufacturing considerations such as the selection of critical raw materials, manufacturing platforms, and critical quality attribute assays. Addressing these issues early in research and development will accelerate clinical product development, clinical trials, and commercial therapies of MSC-EVs.
Collapse
Affiliation(s)
- Katrina Adlerz
- RoosterBio, Inc. 5295 Westview Drive, Suite 275, Frederick, MD 21703, USA
| | - Divya Patel
- RoosterBio, Inc. 5295 Westview Drive, Suite 275, Frederick, MD 21703, USA
| | - Jon Rowley
- RoosterBio, Inc. 5295 Westview Drive, Suite 275, Frederick, MD 21703, USA
| | - Kelvin Ng
- Bioprocessing Technology Institute, 20 Biopolis Way, Centros #06-01 138668, Singapore.
| | - Tabassum Ahsan
- RoosterBio, Inc. 5295 Westview Drive, Suite 275, Frederick, MD 21703, USA.
| |
Collapse
|
231
|
McNamara RP, Dittmer DP. Modern Techniques for the Isolation of Extracellular Vesicles and Viruses. J Neuroimmune Pharmacol 2020. [PMID: 31512168 DOI: 10.1007/s11481-%20019-09874-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Extracellular signaling is pivotal to maintain organismal homeostasis. A quickly emerging field of interest within extracellular signaling is the study of extracellular vesicles (EV), which act as messaging vehicles for nucleic acids, proteins, metabolites, lipids, etc. from donor cells to recipient cells. This transfer of biologically active material within a vesicular body is similar to the infection of a cell through a virus particle, which transfers genetic material from one cell to another to preserve an infection state, and viruses are known to modulate EV. Although considerable heterogeneity exists within EV and viruses, this review focuses on those that are small (< 200 nm in diameter) and of relatively low density (< 1.3 g/mL). A multitude of isolation methods for EV and virus particles exist. In this review, we present an update on methods for their isolation, purification, and phenotypic characterization. We hope that the information we provide will be of use to basic science and clinical investigators, as well as biotechnologists in this emerging field. Graphical Abstract.
Collapse
Affiliation(s)
- Ryan P McNamara
- Lineberger Comprehensive Cancer Center, Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Dirk P Dittmer
- Lineberger Comprehensive Cancer Center, Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
232
|
Walker SA, Aguilar Díaz De león JS, Busatto S, Wurtz GA, Zubair AC, Borges CR, Wolfram J. Glycan Node Analysis of Plasma-Derived Extracellular Vesicles. Cells 2020; 9:cells9091946. [PMID: 32842648 PMCID: PMC7563425 DOI: 10.3390/cells9091946] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/06/2020] [Accepted: 08/20/2020] [Indexed: 12/12/2022] Open
Abstract
Blood plasma is a readily accessible source of extracellular vesicles (EVs), i.e., cell-secreted nanosized carriers that contain various biomolecules, including glycans. Previous studies have demonstrated that glycans play a major role in physiological and pathological processes, and certain plasma glycans have been associated with disease conditions. However, glycome studies have been limited by a lack of analytical techniques with the throughput capacity necessary to study hundreds of clinical samples. This study is the first to characterize the EV plasma glycome based on all major glycan classes. The results based on glycan node analysis revealed, as expected, that plasma-derived EVs have distinct glycan features from donor-matched whole plasma. Specifically, glycan nodes corresponding to those observed in chondroitin sulfate, dermatan sulfate, type I keratan sulfate, and type II keratan sulfate were enriched on EVs. The identification of specific differences in glycan features in plasma vs. plasma-derived EVs is relevant for understanding the physiological role of EVs and as a reference for future diagnostic studies. Additionally, the results indicate that EV glycan nodes do not substantially differ among a small set of healthy donors. These results lay the framework for the further evaluation of all EV glycan classes as diagnostic markers, therapeutic targets, and biologically active components in health and disease.
Collapse
Affiliation(s)
- Sierra A. Walker
- Department of Biochemistry and Molecular Biology, Department of Physiology and Biomedical Engineering, Department of Transplantation, Mayo Clinic, Jacksonville, FL 32224, USA; (S.A.W.); (S.B.)
| | - Jesús S. Aguilar Díaz De león
- School of Molecular Sciences and Virginia G. Piper Center for Personalized Diagnostics, The Biodesign Institute at Arizona State University, Tempe, AZ 85287, USA;
| | - Sara Busatto
- Department of Biochemistry and Molecular Biology, Department of Physiology and Biomedical Engineering, Department of Transplantation, Mayo Clinic, Jacksonville, FL 32224, USA; (S.A.W.); (S.B.)
| | - Gregory A. Wurtz
- Department of Physics, University of North Florida, Jacksonville, FL 32224, USA;
| | - Abba C. Zubair
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Jacksonville, FL 32224, USA;
| | - Chad R. Borges
- School of Molecular Sciences and Virginia G. Piper Center for Personalized Diagnostics, The Biodesign Institute at Arizona State University, Tempe, AZ 85287, USA;
- Correspondence: (C.R.B.); (J.W.)
| | - Joy Wolfram
- Department of Biochemistry and Molecular Biology, Department of Physiology and Biomedical Engineering, Department of Transplantation, Mayo Clinic, Jacksonville, FL 32224, USA; (S.A.W.); (S.B.)
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- Correspondence: (C.R.B.); (J.W.)
| |
Collapse
|
233
|
Kumar A, Kodidela S, Tadrous E, Cory TJ, Walker CM, Smith AM, Mukherjee A, Kumar S. Extracellular Vesicles in Viral Replication and Pathogenesis and Their Potential Role in Therapeutic Intervention. Viruses 2020; 12:E887. [PMID: 32823684 PMCID: PMC7472073 DOI: 10.3390/v12080887] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles (EVs) have shown their potential as a carrier of molecular information, and they have been involved in physiological functions and diseases caused by viral infections. Virus-infected cells secrete various lipid-bound vesicles, including endosome pathway-derived exosomes and microvesicles/microparticles that are released from the plasma membrane. They are released via a direct outward budding and fission of plasma membrane blebs into the extracellular space to either facilitate virus propagation or regulate the immune responses. Moreover, EVs generated by virus-infected cells can incorporate virulence factors including viral protein and viral genetic material, and thus can resemble noninfectious viruses. Interactions of EVs with recipient cells have been shown to activate signaling pathways that may contribute to a sustained cellular response towards viral infections. EVs, by utilizing a complex set of cargos, can play a regulatory role in viral infection, both by facilitating and suppressing the infection. EV-based antiviral and antiretroviral drug delivery approaches provide an opportunity for targeted drug delivery. In this review, we summarize the literature on EVs, their associated involvement in transmission in viral infections, and potential therapeutic implications.
Collapse
Affiliation(s)
- Asit Kumar
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (S.K.); (E.T.); (A.M.)
| | - Sunitha Kodidela
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (S.K.); (E.T.); (A.M.)
| | - Erene Tadrous
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (S.K.); (E.T.); (A.M.)
| | - Theodore James Cory
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA;
| | - Crystal Martin Walker
- College of Nursing, University of Tennessee Health Science Center, Memphis, TN 38163, USA;
| | - Amber Marie Smith
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38163, USA;
| | - Ahona Mukherjee
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (S.K.); (E.T.); (A.M.)
| | - Santosh Kumar
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (S.K.); (E.T.); (A.M.)
| |
Collapse
|
234
|
Masaoutis C, Al Besher S, Koutroulis I, Theocharis S. Exosomes in Nephropathies: A Rich Source of Novel Biomarkers. DISEASE MARKERS 2020; 2020:8897833. [PMID: 32849923 PMCID: PMC7441435 DOI: 10.1155/2020/8897833] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/08/2020] [Accepted: 07/24/2020] [Indexed: 12/12/2022]
Abstract
The biomarkers commonly utilized in diagnostic evaluations of kidney disease suffer from low sensitivity, especially in the early stages of renal damage. On the other hand, obtaining a renal biopsy to augment clinical decision making can lead to potentially serious complications. In order to overcome the shortcomings of currently available diagnostic tools, recent studies suggest that exosomes, cell-secreted extracellular vesicles containing a large array of active molecules to facilitate cell-to-cell communication, may represent a rich source of novel disease biomarkers. Because of their endocytic origin, exosomes carry markers typical for their parent cells, which could permit the localization of biochemical cellular alterations in specific kidney compartments. Different types of exosomes can be isolated from noninvasively obtained biofluids; however, in the context of kidney disease, evidence has emerged on the role of urinary exosomes in the diagnostic and predictive modeling of renal pathology. The current review summarizes the potential application of exosomes in the detection of acute and chronic inflammatory, metabolic, degenerative, and genetic renal diseases.
Collapse
Affiliation(s)
- Christos Masaoutis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75, Mikras Asias street, Bld 10, Goudi, 11527 Athens, Greece
| | - Samer Al Besher
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75, Mikras Asias street, Bld 10, Goudi, 11527 Athens, Greece
| | - Ioannis Koutroulis
- Children's National Hospital, Division of Emergency Medicine and Center for Genetic Medicine, George Washington University School of Medicine and Health Sciences, 111 Michigan Ave. NW, Washington, DC 20010, USA
| | - Stamatios Theocharis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75, Mikras Asias street, Bld 10, Goudi, 11527 Athens, Greece
| |
Collapse
|
235
|
Busatto S, Walker SA, Grayson W, Pham A, Tian M, Nesto N, Barklund J, Wolfram J. Lipoprotein-based drug delivery. Adv Drug Deliv Rev 2020; 159:377-390. [PMID: 32791075 PMCID: PMC7747060 DOI: 10.1016/j.addr.2020.08.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 07/01/2020] [Accepted: 08/07/2020] [Indexed: 02/07/2023]
Abstract
Lipoproteins (LPs) are circulating heterogeneous nanoparticles produced by the liver and intestines. LPs play a major role in the transport of dietary and endogenous lipids to target cells through cell membrane receptors or cell surface-bound lipoprotein lipase. The stability, biocompatibility, and selective transport of LPs make them promising delivery vehicles for various therapeutic and imaging agents. This review discusses isolation, manufacturing, and drug loading techniques used for LP-based drug delivery, as well as recent applications for diagnosis and treatment of cancer, atherosclerosis, and other life-threatening diseases.
Collapse
Affiliation(s)
- Sara Busatto
- Department of Biochemistry and Molecular Biology, Department of Physiology and Biomedical Engineering, Department of Transplantation, Mayo Clinic, Jacksonville, FL 32224, USA.
| | - Sierra A Walker
- Department of Biochemistry and Molecular Biology, Department of Physiology and Biomedical Engineering, Department of Transplantation, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Whisper Grayson
- Department of Biology, University of North Florida, Jacksonville, FL 32224, USA
| | - Anthony Pham
- Department of Biochemistry and Molecular Biology, Department of Physiology and Biomedical Engineering, Department of Transplantation, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Ming Tian
- Department of Biochemistry and Molecular Biology, Department of Physiology and Biomedical Engineering, Department of Transplantation, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Nicole Nesto
- Department of Biology, University of North Florida, Jacksonville, FL 32224, USA
| | - Jacqueline Barklund
- Department of Biology, University of North Florida, Jacksonville, FL 32224, USA
| | - Joy Wolfram
- Department of Biochemistry and Molecular Biology, Department of Physiology and Biomedical Engineering, Department of Transplantation, Mayo Clinic, Jacksonville, FL 32224, USA; Department of Biology, University of North Florida, Jacksonville, FL 32224, USA; Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA.
| |
Collapse
|
236
|
Lee JH, Ha DH, Go HK, Youn J, Kim HK, Jin RC, Miller RB, Kim DH, Cho BS, Yi YW. Reproducible Large-Scale Isolation of Exosomes from Adipose Tissue-Derived Mesenchymal Stem/Stromal Cells and Their Application in Acute Kidney Injury. Int J Mol Sci 2020; 21:E4774. [PMID: 32635660 PMCID: PMC7370182 DOI: 10.3390/ijms21134774] [Citation(s) in RCA: 56] [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: 06/03/2020] [Revised: 07/03/2020] [Accepted: 07/03/2020] [Indexed: 12/18/2022] Open
Abstract
Acute kidney injury (AKI) is a fatal medical episode caused by sudden kidney damage or failure, leading to the death of patients within a few hours or days. Previous studies demonstrated that exosomes derived from various mesenchymal stem/stromal cells (MSC-exosomes) have positive effects on renal injuries in multiple experimental animal models of kidney diseases including AKI. However, the mass production of exosomes is a challenge not only in preclinical studies with large animals but also for successful clinical applications. In this respect, tangential flow filtration (TFF) is suitable for good manufacturing practice (GMP)-compliant large-scale production of high-quality exosomes. Until now, no studies have been reported on the use of TFF, but rather ultracentrifugation has been almost exclusively used, to isolate exosomes for AKI therapeutic application in preclinical studies. Here, we demonstrated the reproducible large-scale production of exosomes derived from adipose tissue-derived MSC (ASC-exosomes) using TFF and the lifesaving effect of the ASC-exosomes in a lethal model of cisplatin-induced rat AKI. Our results suggest the possibility of large-scale stable production of ASC-exosomes without loss of function and their successful application in life-threatening diseases.
Collapse
Affiliation(s)
- Jun Ho Lee
- ExoCoBio Exosome Institue (EEI), ExoCoBio Inc., Seoul 08594, Korea; (J.H.L.); (D.H.H.); (J.Y.); (H.-k.K.)
| | - Dae Hyun Ha
- ExoCoBio Exosome Institue (EEI), ExoCoBio Inc., Seoul 08594, Korea; (J.H.L.); (D.H.H.); (J.Y.); (H.-k.K.)
| | | | - Jinkwon Youn
- ExoCoBio Exosome Institue (EEI), ExoCoBio Inc., Seoul 08594, Korea; (J.H.L.); (D.H.H.); (J.Y.); (H.-k.K.)
| | - Hyun-keun Kim
- ExoCoBio Exosome Institue (EEI), ExoCoBio Inc., Seoul 08594, Korea; (J.H.L.); (D.H.H.); (J.Y.); (H.-k.K.)
| | | | | | | | - Byong Seung Cho
- ExoCoBio Exosome Institue (EEI), ExoCoBio Inc., Seoul 08594, Korea; (J.H.L.); (D.H.H.); (J.Y.); (H.-k.K.)
| | - Yong Weon Yi
- ExoCoBio Exosome Institue (EEI), ExoCoBio Inc., Seoul 08594, Korea; (J.H.L.); (D.H.H.); (J.Y.); (H.-k.K.)
| |
Collapse
|
237
|
Popowski K, Lutz H, Hu S, George A, Dinh PU, Cheng K. Exosome therapeutics for lung regenerative medicine. J Extracell Vesicles 2020; 9:1785161. [PMID: 32944172 PMCID: PMC7480570 DOI: 10.1080/20013078.2020.1785161] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 02/06/2023] Open
Abstract
Exosomes are 30 to 100 nm extracellular vesicles that are secreted by many cell types. Initially viewed as cellular garbage with no biological functions, exosomes are now recognized for their therapeutic potential and used in regenerative medicine. Cell-derived exosomes are released into almost all biological fluids, making them abundant and accessible vesicles for a variety of diseases. These naturally occurring nanoparticles have a wide range of applications including drug delivery and regenerative medicine. Exosomes sourced from a specific tissue have been proven to provide greater therapeutic effects to their native tissue, expanding exosome sources beyond traditional cell lines such as mesenchymal stem cells. However, standardizing production and passing regulations remain obstacles, due to variations in methods and quantification techniques across studies. Additionally, obtaining pure exosomes at sufficient quantities remains difficult due to the heterogeneity of exosomes. In this review, we will underline the uses of exosomes as a therapy and their roles in lung regenerative medicine, as well as current challenges in exosome therapies.
Collapse
Affiliation(s)
- Kristen Popowski
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| | - Halle Lutz
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, USA
| | - Shiqi Hu
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, USA
| | - Arianna George
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, NC, USA
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Phuong-Uyen Dinh
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| | - Ke Cheng
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State
University, NC, USA
- Division of Pharmacoengineering and Molecular
Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC,
USA
| |
Collapse
|
238
|
Kluszczyńska K, Czernek L, Cypryk W, Pęczek Ł, Düchler M. Methods for the Determination of the Purity of Exosomes. Curr Pharm Des 2020; 25:4464-4485. [PMID: 31808383 DOI: 10.2174/1381612825666191206162712] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 12/03/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Exosomes open exciting new opportunities for advanced drug transport and targeted release. Furthermore, exosomes may be used for vaccination, immunosuppression or wound healing. To fully utilize their potential as drug carriers or immune-modulatory agents, the optimal purity of exosome preparations is of crucial importance. METHODS Articles describing the isolation and purification of exosomes were retrieved from the PubMed database. RESULTS Exosomes are often separated from biological fluids containing high concentrations of proteins, lipids and other molecules that keep vesicle purification challenging. A great number of purification protocols have been published, however, their outcome is difficult to compare because the assessment of purity has not been standardized. In this review, we first give an overview of the generation and composition of exosomes, as well as their multifaceted biological functions that stimulated various medical applications. Finally, we describe various methods that have been used to purify small vesicles and to assess the purity of exosome preparations and critically compare the quality of these evaluation protocols. CONCLUSION Combinations of various techniques have to be applied to reach the required purity and quality control of exosome preparations.
Collapse
Affiliation(s)
- Katarzyna Kluszczyńska
- Department of Bioorganic Chemistry, Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, 112 Sienkiewicza Street, 90-363 Lodz, Poland
| | - Liliana Czernek
- Department of Bioorganic Chemistry, Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, 112 Sienkiewicza Street, 90-363 Lodz, Poland
| | - Wojciech Cypryk
- Department of Bioorganic Chemistry, Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, 112 Sienkiewicza Street, 90-363 Lodz, Poland
| | - Łukasz Pęczek
- Department of Bioorganic Chemistry, Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, 112 Sienkiewicza Street, 90-363 Lodz, Poland
| | - Markus Düchler
- Department of Bioorganic Chemistry, Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, 112 Sienkiewicza Street, 90-363 Lodz, Poland
| |
Collapse
|
239
|
To K, Romain K, Mak C, Kamaraj A, Henson F, Khan W. The Treatment of Cartilage Damage Using Human Mesenchymal Stem Cell-Derived Extracellular Vesicles: A Systematic Review of in vivo Studies. Front Bioeng Biotechnol 2020; 8:580. [PMID: 32596228 PMCID: PMC7300288 DOI: 10.3389/fbioe.2020.00580] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 05/13/2020] [Indexed: 12/21/2022] Open
Abstract
Damage to joints through injury or disease can result in cartilage loss, which if left untreated can lead to inflammation and ultimately osteoarthritis. There is currently no cure for osteoarthritis and management focusses on symptom control. End-stage osteoarthritis can be debilitating and ultimately requires joint replacement in order to maintain function. Therefore, there is growing interest in innovative therapies for cartilage repair. In this systematic literature review, we sought to explore the in vivo evidence for the use of human Mesenchymal Stem Cell-derived Extracellular Vesicles (MSC-EVs) for treating cartilage damage. We conducted a systematic literature review in accordance with the PRISMA protocol on the evidence for the treatment of cartilage damage using human MSC-EVs. Studies examining in vivo models of cartilage damage were included. A risk of bias analysis of the studies was conducted using the SYRCLE tool. Ten case-control studies were identified in our review, including a total of 159 murine subjects. MSC-EVs were harvested from a variety of human tissues. Five studies induced osteoarthritis, including cartilage loss through surgical joint destabilization, two studies directly created osteochondral lesions and three studies used collagenase to cause cartilage loss. All studies in this review reported reduced cartilage loss following treatment with MSC-EVs, and without significant complications. We conclude that transplantation of MSC-derived EVs into damaged cartilage can effectively reduce cartilage loss in murine models of cartilage injury. Additional randomized studies in animal models that recapitulates human osteoarthritis will be necessary in order to establish findings that inform clinical safety in humans.
Collapse
Affiliation(s)
- Kendrick To
- Division of Trauma and Orthopaedics, Department of Surgery, University of Cambridge, Cambridge, United Kingdom
| | - Karl Romain
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Christopher Mak
- Division of Trauma and Orthopaedics, Department of Surgery, University of Cambridge, Cambridge, United Kingdom
| | - Achi Kamaraj
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Frances Henson
- Division of Trauma and Orthopaedics, Department of Surgery, University of Cambridge, Cambridge, United Kingdom
| | - Wasim Khan
- Division of Trauma and Orthopaedics, Department of Surgery, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
240
|
Wiest EF, Zubair AC. Challenges of manufacturing mesenchymal stromal cell-derived extracellular vesicles in regenerative medicine. Cytotherapy 2020; 22:606-612. [PMID: 32532592 DOI: 10.1016/j.jcyt.2020.04.040] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/23/2020] [Accepted: 04/07/2020] [Indexed: 12/18/2022]
Abstract
The field of regenerative medicine has expanded greatly in the past decade, with more than 1000 current clinical trials involving mesenchymal stromal cell (MSC) treatment. Multiple recent publications have demonstrated that the beneficial effects from MSCs are not simply due to engraftment into the target organ as classically thought but rather are largely attributable to the release of paracrine factors including cytokines, growth factors and extracellular vesicles (EVs). These EVs contain miRNAs, free fatty acids and proteins that promote regeneration, proliferation and cell function and improve inflammation. Although EVs have shown promising results in animal studies, there are many obstacles to the manufacturing of EVs for clinical applications. This review discusses challenges associated with the manufacturing of clinical-grade EVs in regard to identity, purity, reproducibility, sterility, storage, potency and safety. We discuss currently employed methods and approaches for developing clinical Good Manufacturing Practices (GMP)-grade EVs and the limitations for each. We further discuss the best approaches to overcome the current hurdles in developing clinical GMP-grade EVs.
Collapse
Affiliation(s)
- Elani F Wiest
- Department of Laboratory Medicine and Pathology and Center for Regenerative Medicine, Mayo Clinic, Jacksonville, Florida, USA
| | - Abba C Zubair
- Department of Laboratory Medicine and Pathology and Center for Regenerative Medicine, Mayo Clinic, Jacksonville, Florida, USA.
| |
Collapse
|
241
|
Benmoussa A, Michel S, Gilbert C, Provost P. Isolating Multiple Extracellular Vesicles Subsets, Including Exosomes and Membrane Vesicles, from Bovine Milk Using Sodium Citrate and Differential Ultracentrifugation. Bio Protoc 2020; 10:e3636. [PMID: 33659307 DOI: 10.21769/bioprotoc.3636] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/29/2020] [Accepted: 03/16/2020] [Indexed: 12/21/2022] Open
Abstract
Milk is a complex fluid that contains various types of proteins and extracellular vesicles (EVs). Some proteins can mingle with EVs, and interfere with their isolation. Among these proteins, caseins form micelles of a size comparable to milk EVs, and can thus be co-isolated with EVs. Preliminary steps that affect milk are crucial for EV isolation and impact the purity and abundance of isolated EVs. In the course of our previous works on cow's milk EVs, we found that sodium citrate (1% final), which is a biocompatible reagent capable of breaking down casein micelles into 40-nm monomers, allowed the isolation of high quantities of EVs with low coprecipitation of caseins or other contaminating proteins. Using this protocol, we successfully separated different EV subsets, characterized in depth their morphology, protein content and small RNA enrichment patterns. We were also able to describe their biological function in a mouse model of intestinal inflammation. We, hereby, detail the differential ultracentrifugation procedure that leads to high quantify, medium specificity, isolation of different milk EV subsets from the same sample. More specifically, we highlight the use of sodium citrate as a standardized approach to isolate and study milk EVs and its potential for isolation techniques other than differential ultracentrifugation.
Collapse
Affiliation(s)
- Abderrahim Benmoussa
- CHUQ Research Center/CHUL Pavilion, 2705 Blvd Laurier, Quebec, QC, G1V 4G2, Canada, Department of Microbiology, Infectious Diseases and Immunology and Faculty of Medicine, Université Laval, Quebec, QC, G1V 0A6, Canada
| | - Sara Michel
- CHUQ Research Center/CHUL Pavilion, 2705 Blvd Laurier, Quebec, QC, G1V 4G2, Canada, Department of Microbiology, Infectious Diseases and Immunology and Faculty of Medicine, Université Laval, Quebec, QC, G1V 0A6, Canada
| | - Caroline Gilbert
- CHUQ Research Center/CHUL Pavilion, 2705 Blvd Laurier, Quebec, QC, G1V 4G2, Canada, Department of Microbiology, Infectious Diseases and Immunology and Faculty of Medicine, Université Laval, Quebec, QC, G1V 0A6, Canada
| | - Patrick Provost
- CHUQ Research Center/CHUL Pavilion, 2705 Blvd Laurier, Quebec, QC, G1V 4G2, Canada, Department of Microbiology, Infectious Diseases and Immunology and Faculty of Medicine, Université Laval, Quebec, QC, G1V 0A6, Canada
| |
Collapse
|
242
|
Tung SL, Fanelli G, Matthews RI, Bazoer J, Letizia M, Vizcay-Barrena G, Faruqu FN, Philippeos C, Hannen R, Al-Jamal KT, Lombardi G, Smyth LA. Regulatory T Cell Extracellular Vesicles Modify T-Effector Cell Cytokine Production and Protect Against Human Skin Allograft Damage. Front Cell Dev Biol 2020; 8:317. [PMID: 32509778 PMCID: PMC7251034 DOI: 10.3389/fcell.2020.00317] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 04/09/2020] [Indexed: 12/18/2022] Open
Abstract
Regulatory T cells (Tregs) are a subpopulation of CD4+ T cells with a fundamental role in maintaining immune homeostasis and inhibiting unwanted immune responses using several different mechanisms. Recently, the intercellular transfer of molecules between Tregs and their target cells has been shown via trogocytosis and the release of small extracellular vesicles (sEVs). In this study, CD4+CD25+CD127lo human Tregs were found to produce sEVs capable of inhibiting the proliferation of effector T cells (Teffs) in a dose dependent manner. These vesicles also modified the cytokine profile of Teffs leading to an increase in the production of IL-4 and IL-10 whilst simultaneously decreasing the levels of IL-6, IL-2, and IFNγ. MicroRNAs found enriched in the Treg EVs were indirectly linked to the changes in the cytokine profile observed. In a humanized mouse skin transplant model, human Treg derived EVs inhibited alloimmune-mediated skin tissue damage by limiting immune cell infiltration. Taken together, Treg sEVs may represent an exciting cell-free therapy to promote transplant survival.
Collapse
Affiliation(s)
- Sim Lai Tung
- Immunoregulation Laboratory, MRC Centre for Transplantation, School of Immunology & Microbial Sciences, King's College London, Guy's Hospital, London, United Kingdom
| | - Giorgia Fanelli
- Immunoregulation Laboratory, MRC Centre for Transplantation, School of Immunology & Microbial Sciences, King's College London, Guy's Hospital, London, United Kingdom
| | - Robert Ian Matthews
- School of Health, Sport and Bioscience, Stratford Campus, University of East London, London, United Kingdom
| | - Jordan Bazoer
- School of Health, Sport and Bioscience, Stratford Campus, University of East London, London, United Kingdom
| | - Marilena Letizia
- Immunoregulation Laboratory, MRC Centre for Transplantation, School of Immunology & Microbial Sciences, King's College London, Guy's Hospital, London, United Kingdom
| | - Gema Vizcay-Barrena
- Centre for Ultrastructural Imaging, King's College London, London, United Kingdom
| | - Farid N Faruqu
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Christina Philippeos
- Centre for Stem Cells & Regenerative Medicine, King's College London, London, United Kingdom
| | - Rosalind Hannen
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Khuloud T Al-Jamal
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Giovanna Lombardi
- Immunoregulation Laboratory, MRC Centre for Transplantation, School of Immunology & Microbial Sciences, King's College London, Guy's Hospital, London, United Kingdom
| | - Lesley Ann Smyth
- Immunoregulation Laboratory, MRC Centre for Transplantation, School of Immunology & Microbial Sciences, King's College London, Guy's Hospital, London, United Kingdom.,School of Health, Sport and Bioscience, Stratford Campus, University of East London, London, United Kingdom
| |
Collapse
|
243
|
Salem I, Naranjo NM, Singh A, DeRita R, Krishn SR, Sirman LS, Quaglia F, Duffy A, Bowler N, Sayeed A, Languino LR. Methods for extracellular vesicle isolation from cancer cells. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2020; 3:371-384. [PMID: 33062957 PMCID: PMC7556721 DOI: 10.20517/cdr.2019.118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cells are known to release different types of vesicles such as small extracellular vesicles (sEVs) and large extracellular vesicles (LEVs). sEVs and LEVs play important roles in intercellular communication, pre-metastatic niche formation, and disease progression; both can be detected cell culture media and biological fluids. sEVs and LEVs contain a variety of protein and RNA cargo, and they are believed to impact many biological functions of the recipient cells upon their internalization or binding to cell surface proteins. It has recently been established that standard isolation techniques, such as differential ultracentrifugation, yield a mixed population of EVs. However, density gradient ultracentrifugation has been reported to allow the isolation of sEVs without cellular debris. Here, we describe the most common methods used to isolate sEVs from cell culture medium, mouse and human plasma, and a new technique for isolating sEVs from tissues as well. This article also provides detailed procedures to isolate LEVs.
Collapse
Affiliation(s)
- Israa Salem
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Nicole M Naranjo
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Amrita Singh
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA.,(Present Address) Astellas Institute for Regenerative Medicine, Marlborough, MA 01752, USA
| | - Rachel DeRita
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA.,(Present Address) School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shiv Ram Krishn
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Luca S Sirman
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Fabio Quaglia
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Alexander Duffy
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Nicholas Bowler
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Aejaz Sayeed
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA.,(Present Address) Baruch S. Blumberg Institute, Doylestown, PA 18902, USA
| | - Lucia R Languino
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
244
|
Kim KH, Jo JH, Cho HJ, Park TS, Kim TM. Therapeutic potential of stem cell-derived extracellular vesicles in osteoarthritis: preclinical study findings. Lab Anim Res 2020; 36:10. [PMID: 32322556 PMCID: PMC7160998 DOI: 10.1186/s42826-020-00043-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/23/2020] [Indexed: 01/08/2023] Open
Abstract
Extracellular vesicles (EVs) are nano-sized particles secreted by almost all cell types, and they mediate various biological processes via cell-to-cell communication. Compared with parental cells for therapeutic purposes, stem cell-derived EVs have several advantages such as reduced risk of rejection, less oncogenic potential, ease of long-term storage, lower chance of thromboembolism, and readiness for immediate use. Recent studies have demonstrated that EVs from stem cells, mostly from mesenchymal stem cells (MSCs) from various tissues, have anti-inflammatory, anti-oxidative, anti-apoptotic, and proliferative role in injured organs including osteoarthritic lesions. Herein, we provide a review about the up-to-date studies in preclinical application of stem cell-derived EVs in osteoarthritis animal arthritis models.
Collapse
Affiliation(s)
- Ki Hoon Kim
- Graduate School of International Agricultural Technology, Pyeongchang, South Korea
| | - Jeong Hyun Jo
- Graduate School of International Agricultural Technology, Pyeongchang, South Korea
| | - Hye Jin Cho
- 2Institutes of Green-Bio Science and Technology, Seoul National University, Pyeongchang, Gangwon-do 25354 South Korea
| | - Tae Sub Park
- Graduate School of International Agricultural Technology, Pyeongchang, South Korea.,2Institutes of Green-Bio Science and Technology, Seoul National University, Pyeongchang, Gangwon-do 25354 South Korea
| | - Tae Min Kim
- Graduate School of International Agricultural Technology, Pyeongchang, South Korea.,2Institutes of Green-Bio Science and Technology, Seoul National University, Pyeongchang, Gangwon-do 25354 South Korea
| |
Collapse
|
245
|
Busatto S, Zendrini A, Radeghieri A, Paolini L, Romano M, Presta M, Bergese P. The nanostructured secretome. Biomater Sci 2020; 8:39-63. [PMID: 31799977 DOI: 10.1039/c9bm01007f] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The term secretome, which traditionally strictly refers to single proteins, should be expanded to also include the great variety of nanoparticles secreted by cells (secNPs) into the extracellular space, which ranges from high-density lipoproteins of a few nanometers to extracellular vesicles and fat globules of hundreds of nanometers. Widening the definition is urged by the ever-increasing understanding of the role of secNPs as regulators/mediators of key physiological and pathological processes, which also puts them in the running as breakthrough cell-free therapeutics and diagnostics. "Made by cells for cells", secNPs are envisioned as a sweeping paradigm shift in nanomedicine, promising to overcome the limitations of synthetic nanoparticles by unsurpassed circulation and targeting abilities, precision and sustainability. From a longer/wider perspective, advanced manipulation would possibly make secNPs available as building blocks for future "biogenic" nanotechnology. However, the current knowledge is fragmented and sectorial (the majority of the studies being focused on a specific biological and/or medical aspect of a given secNP class or subclass), the understanding of the nanoscale and interfacial properties is limited and the development of bioprocesses and regulatory initiatives is in the early days. We believe that new multidisciplinary competencies and synergistic efforts need to be attracted and augmented to move forward. This review will contribute to the effort by attempting for the first time to rationally gather and elaborate secNPs and their traits into a unique concise framework - from biogenesis to colloidal properties, engineering and clinical translation - disclosing the overall view and easing comparative analysis and future exploitation.
Collapse
Affiliation(s)
- S Busatto
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
| | | | | | | | | | | | | |
Collapse
|
246
|
El Baradie KBY, Nouh M, O'Brien Iii F, Liu Y, Fulzele S, Eroglu A, Hamrick MW. Freeze-Dried Extracellular Vesicles From Adipose-Derived Stem Cells Prevent Hypoxia-Induced Muscle Cell Injury. Front Cell Dev Biol 2020; 8:181. [PMID: 32266262 PMCID: PMC7099601 DOI: 10.3389/fcell.2020.00181] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 03/04/2020] [Indexed: 12/12/2022] Open
Abstract
Cellular therapies have tremendous potential for the successful treatment of major extremity wounds in the combat setting, however, the challenges associated with transplanting stem cells in the prolonged field care (PFC) environment are a critical barrier to progress in treating such injuries. These challenges include not only production and storage but also transport and handling issues. Our goal is to develop a new strategy utilizing extracellular vesicles (EVs) secreted by stem cells that can resolve many of these issues and prevent ischemic tissue injury. While EVs can be preserved by freezing or lyophilization, both processes result in decrease in their bioactivity. Here, we describe optimized procedures for EVs production, isolation, and lyophilization from primary human adipose-derived stem cells (hADSCs). We compared two isolation approaches that were ultrafiltration (UF) using a tangential fluid filtration (TFF) system and differential ultracentrifugation (UC). We also optimized EVs lyophilization in conjunction with trehalose and polyvinylpyrrolidone 40 (PVP40) as lyoprotectants. Bioactivity of EVs was assessed based on reversal of hypoxia-induced muscle cell injury. To this end, primary human myoblasts were subjected to hypoxic conditions for 6 h, and then treated with hADSC-derived EVs at a concentration of 50 μg/mL. Subsequently, muscle cell viability and toxicity were evaluated using MTS and LDH assays, respectively. Overall, nanoparticle tracking data indicated that UF/TFF yields threefold more particles than UC. Lyophilization of EVs resulted in a significantly reduced number of particles, which could be attenuated by adding lyoprotections to the freeze-drying solution. Furthermore, EVs isolated by UF/TFF and freeze-dried in the presence of trehalose significantly increased viability (P < 0.0193). Taken together, our findings suggest that the isolation and preservation methods presented in this study may enhance therapeutic applications of EVs.
Collapse
Affiliation(s)
| | - Mohamed Nouh
- Medical College of Georgia, Augusta University, Augusta, GA, United States.,Tanta Cancer Center, Tanta, Egypt
| | | | - Yutao Liu
- Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Sadanand Fulzele
- Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Ali Eroglu
- Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Mark W Hamrick
- Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
247
|
Tian M, Ticer T, Wang Q, Walker S, Pham A, Suh A, Busatto S, Davidovich I, Al-Kharboosh R, Lewis-Tuffin L, Ji B, Quinones-Hinojosa A, Talmon Y, Shapiro S, Rückert F, Wolfram J. Adipose-Derived Biogenic Nanoparticles for Suppression of Inflammation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1904064. [PMID: 32067382 DOI: 10.1002/smll.201904064] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/30/2019] [Indexed: 06/10/2023]
Abstract
Extracellular vesicles secreted from adipose-derived mesenchymal stem cells (ADSCs) have therapeutic effects in inflammatory diseases. However, production of extracellular vesicles (EVs) from ADSCs is costly, inefficient, and time consuming. The anti-inflammatory properties of adipose tissue-derived EVs and other biogenic nanoparticles have not been explored. In this study, biogenic nanoparticles are obtained directly from lipoaspirate, an easily accessible and abundant source of biological material. Compared to ADSC-EVs, lipoaspirate nanoparticles (Lipo-NPs) take less time to process (hours compared to months) and cost less to produce (clinical-grade cell culture facilities are not required). The physicochemical characteristics and anti-inflammatory properties of Lipo-NPs are evaluated and compared to those of patient-matched ADSC-EVs. Moreover, guanabenz loading in Lipo-NPs is evaluated for enhanced anti-inflammatory effects. Apolipoprotein E and glycerolipids are enriched in Lipo-NPs compared to ADSC-EVs. Additionally, the uptake of Lipo-NPs in hepatocytes and macrophages is higher. Lipo-NPs and ADSC-EVs have comparable protective and anti-inflammatory effects. Specifically, Lipo-NPs reduce toll-like receptor 4-induced secretion of inflammatory cytokines in macrophages. Guanabenz-loaded Lipo-NPs further suppress inflammatory pathways, suggesting that this combination therapy can have promising applications for inflammatory diseases.
Collapse
Affiliation(s)
- Ming Tian
- Department of Biochemistry and Molecular Biology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, FL, 32224, USA
- Department of Surgery, Surgical Lab, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Taylor Ticer
- Department of Biochemistry and Molecular Biology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Qikun Wang
- Department of Biochemistry and Molecular Biology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, FL, 32224, USA
- Department of Surgery, Surgical Lab, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Sierra Walker
- Department of Biochemistry and Molecular Biology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Anthony Pham
- Department of Biochemistry and Molecular Biology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Annie Suh
- Department of Biochemistry and Molecular Biology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Sara Busatto
- Department of Biochemistry and Molecular Biology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Irina Davidovich
- Department of Chemical Engineering and the Russell Berrie Nanotechnology Institute (RBNI), Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Rawan Al-Kharboosh
- Department of Neurosurgery, Mayo Clinic Florida, Jacksonville, FL, 32224, USA
| | | | - Baoan Ji
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | | | - Yeshayahu Talmon
- Department of Chemical Engineering and the Russell Berrie Nanotechnology Institute (RBNI), Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Shane Shapiro
- Department of Orthopedic Surgery, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Felix Rückert
- Department of Surgery, Surgical Lab, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Joy Wolfram
- Department of Biochemistry and Molecular Biology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, FL, 32224, USA
| |
Collapse
|
248
|
Leidal AM, Debnath J. Unraveling the mechanisms that specify molecules for secretion in extracellular vesicles. Methods 2020; 177:15-26. [PMID: 31978536 DOI: 10.1016/j.ymeth.2020.01.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 12/19/2019] [Accepted: 01/16/2020] [Indexed: 12/15/2022] Open
Abstract
Extracellular vesicles (EVs) are small membrane-bound organelles naturally released from cells and potentially function as vehicles of intercellular communication. Cells release numerous sub-species of EVs, including exosomes and microvesicles, which are formed via distinct cellular pathways and molecular machineries and contain specific proteins, RNAs and lipids. Accumulating evidence indicates that the repertoire of molecules packaged into EVs is shaped by both the physiological state of the cell and the EV biogenesis pathway involved. Although these observations intimate that precisely regulated pathways sort molecules into EVs, the underlying molecular mechanisms that direct molecules for secretion remain poorly defined. Recently, with the advancement of mass spectrometry, next-generation sequencing techniques and molecular biology tools, several mechanisms contributing to EV cargo selection are beginning to be unraveled. This review examines strategies employed to reveal how specific proteins, RNAs and lipids are directed for secretion via EVs.
Collapse
Affiliation(s)
- Andrew M Leidal
- Department of Pathology and Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94143, USA.
| | - Jayanta Debnath
- Department of Pathology and Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
249
|
Pang B, Zhu Y, Ni J, Thompson J, Malouf D, Bucci J, Graham P, Li Y. Extracellular vesicles: the next generation of biomarkers for liquid biopsy-based prostate cancer diagnosis. Theranostics 2020; 10:2309-2326. [PMID: 32089744 PMCID: PMC7019149 DOI: 10.7150/thno.39486] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 10/01/2019] [Indexed: 12/13/2022] Open
Abstract
Prostate cancer (PCa) is a leading cause of cancer death for males in western countries. The current gold standard for PCa diagnosis - template needle biopsies - often does not convey a true representation of the molecular profile given sampling error and complex tumour heterogeneity. Presently available biomarker blood tests have limited accuracy. There is a growing demand for novel diagnostic approaches to reduce both the number of men with an abnormal PSA/ DRE who undergo invasive biopsy and the number of cores collected per biopsy. 'Liquid biopsy' is a minimally invasive biofluid-based approach that has the potential to provide information and improve the accuracy of diagnosis for patients' treatment selection, prognostic counselling and development of risk-adjusted follow-up protocols. Extracellular vesicles (EVs) are lipid bilayer-delimited particles released by tumour cells which may provide a real-time snapshot of the entire tumour in a non-invasive way. EVs can regulate physiological processes and mediate systemic dissemination of various types of cancers. Emerging evidence suggests that EVs have crucial roles in PCa development and metastasis. Most importantly, EVs are directly derived from their parent cells with their information. EVs contain components including proteins, mRNAs, DNA fragments, non-coding RNAs and lipids, and play a critical role in intercellular communication. Therefore, EVs hold promise for the discovery of liquid biopsy-based biomarkers for PCa diagnosis. Here, we review the current approaches for EV isolation and analysis, summarise the recent advances in EV protein biomarkers in PCa and focus on liquid biopsy-based EV biomarkers in PCa diagnosis for personalised medicine.
Collapse
Affiliation(s)
- Bairen Pang
- St George and Sutherland Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia
- Cancer Care Centre, St. George Hospital, Sydney, NSW 2217, Australia
| | - Ying Zhu
- St George and Sutherland Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia
- Cancer Care Centre, St. George Hospital, Sydney, NSW 2217, Australia
| | - Jie Ni
- St George and Sutherland Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia
- Cancer Care Centre, St. George Hospital, Sydney, NSW 2217, Australia
| | - James Thompson
- St George and Sutherland Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia
- Department of Urology, St George Hospital, Sydney, NSW 2217, Australia
- Garvan Institute of Medical Research/ APCRC, Sydney, UNSW, 2010, Australia
| | - David Malouf
- Cancer Care Centre, St. George Hospital, Sydney, NSW 2217, Australia
- Department of Urology, St George Hospital, Sydney, NSW 2217, Australia
| | - Joseph Bucci
- St George and Sutherland Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia
- Cancer Care Centre, St. George Hospital, Sydney, NSW 2217, Australia
| | - Peter Graham
- St George and Sutherland Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia
- Cancer Care Centre, St. George Hospital, Sydney, NSW 2217, Australia
| | - Yong Li
- St George and Sutherland Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia
- Cancer Care Centre, St. George Hospital, Sydney, NSW 2217, Australia
- School of Basic Medical Sciences, Zhengzhou University, Henan 450001, China
| |
Collapse
|
250
|
Optimized Isolation of Extracellular Vesicles From Various Organic Sources Using Aqueous Two-Phase System. Sci Rep 2019; 9:19159. [PMID: 31844310 PMCID: PMC6915764 DOI: 10.1038/s41598-019-55477-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 11/18/2019] [Indexed: 12/19/2022] Open
Abstract
From biomarkers to drug carriers, Extracellular Vesicles (EVs) are being used successfully in numerous applications. However, while the subject has been steadily rising in popularity, current methods of isolating EVs are lagging behind, incapable of isolating EVs at a high enough quantity or quality while also requiring expensive, specialized equipment. The “isolation problem” is one of the major obstacles in the field of EV research - and even more so for their potential, widespread use for clinical diagnosis and therapeutic applications. Aqueous Two-Phase Systems (ATPS) has been reported previously as a promising method for isolating EVs quickly and efficiently, and with little contaminants - however, this method has not seen widespread use. In this study, an ATPS-based isolation protocol is used to isolate small EVs from plant, cell culture, and parasite culture sources. Isolated EVs were characterized in surface markers, size, and morphological manner. Additionally, the capacity of ATPS-based EV isolation in removing different contaminants was shown by measuring protein, fatty acid, acid, and phenol red levels of the final isolate. In conclusion, we have shown that EVs originating from different biological sources can be isolated successfully in a cost-effective and user-friendly manner with the use of aqueous two-phase systems.
Collapse
|