201
|
Martellucci S, Orefice NS, Angelucci A, Luce A, Caraglia M, Zappavigna S. Extracellular Vesicles: New Endogenous Shuttles for miRNAs in Cancer Diagnosis and Therapy? Int J Mol Sci 2020; 21:ijms21186486. [PMID: 32899898 PMCID: PMC7555972 DOI: 10.3390/ijms21186486] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 09/01/2020] [Accepted: 09/03/2020] [Indexed: 12/16/2022] Open
Abstract
Extracellular Vesicles (EVs) represent a heterogeneous population of membranous cell-derived structures, including cargo-oriented exosomes and microvesicles. EVs are functionally associated with intercellular communication and play an essential role in multiple physiopathological conditions. Shedding of EVs is frequently increased in malignancies and their content, including proteins and nucleic acids, altered during carcinogenesis and cancer progression. EVs-mediated intercellular communication between tumor cells and between tumor and stromal cells can modulate, through cargo miRNA, the survival, progression, and drug resistance in cancer conditions. These consolidated suggestions and EVs’ stability in bodily fluids have led to extensive investigations on the potential employment of circulating EVs-derived miRNAs as tumor biomarkers and potential therapeutic vehicles. In this review, we highlight the current knowledge about circulating EVs-miRNAs in human cancer and the application limits of these tools, discussing their clinical utility and challenges in functions such as in biomarkers and instruments for diagnosis, prognosis, and therapy.
Collapse
Affiliation(s)
- Stefano Martellucci
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.M.); (A.A.)
| | - Nicola Salvatore Orefice
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Correspondence: or ; Tel.: +1-608-262-21-89
| | - Adriano Angelucci
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.M.); (A.A.)
| | - Amalia Luce
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (A.L.); (M.C.); (S.Z.)
| | - Michele Caraglia
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (A.L.); (M.C.); (S.Z.)
- Biogem Scarl, Institute of Genetic Research, Laboratory of Precision and Molecular Oncology, Ariano Irpino, 83031 Avellino, Italy
| | - Silvia Zappavigna
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (A.L.); (M.C.); (S.Z.)
| |
Collapse
|
202
|
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: 60] [Impact Index Per Article: 12.0] [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
|
203
|
Mehryab F, Rabbani S, Shahhosseini S, Shekari F, Fatahi Y, Baharvand H, Haeri A. Exosomes as a next-generation drug delivery system: An update on drug loading approaches, characterization, and clinical application challenges. Acta Biomater 2020; 113:42-62. [PMID: 32622055 DOI: 10.1016/j.actbio.2020.06.036] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 06/23/2020] [Accepted: 06/25/2020] [Indexed: 12/13/2022]
Abstract
Exosomes are small nanoparticles secreted by almost all cells and have a well-known role in intercellular communication. They are found in different body fluids and can also be isolated from cell culture media. They contain a natural cargo including various protein and nucleic acid molecules originated from their donor cells. In recent years, exosomes have emerged as a desired drug delivery system. They are believed to provide a targeted delivery of drug molecules, supplemented with their natural function. Furthermore, they have a membranous structure similar to liposomes, and that motivated researchers to apply their previous experience of drug loading into liposomes for exosomes. Herein, we discuss applied methods for the encapsulation of different drugs into exosomes, parameters affecting the incorporation of drug molecules into exosomes, characterization techniques, recent achievements, commercialization challenges and the potential future developments of exosomal drugs. Overall, while the application of exosomes as a drug delivery system is still in its infancy, they are considered to be a new class of natural nanocarriers with great potential for clinical application. Understanding of their key formulation parameters, pharmaceutical properties, in vivo behavior and applicable scale-up production will pave their way to the market. STATEMENT OF SIGNIFICANCE: Details of loading methods, characterization and biopharmaceutical properties of drug-incorporated exosomes are presented. Most parameters affecting encapsulation of drugs into exosomes are mentioned to serve as a guide for future studies in this field. Moreover, challenges on the way of exosomes to the market and clinic are described.
Collapse
|
204
|
Leggio L, Arrabito G, Ferrara V, Vivarelli S, Paternò G, Marchetti B, Pignataro B, Iraci N. Mastering the Tools: Natural versus Artificial Vesicles in Nanomedicine. Adv Healthc Mater 2020; 9:e2000731. [PMID: 32864899 DOI: 10.1002/adhm.202000731] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/27/2020] [Indexed: 12/12/2022]
Abstract
Naturally occurring extracellular vesicles and artificially made vesicles represent important tools in nanomedicine for the efficient delivery of biomolecules and drugs. Since its first appearance in the literature 50 years ago, the research on vesicles is progressing at a fast pace, with the main goal of developing carriers able to protect cargoes from degradation, as well as to deliver them in a time- and space-controlled fashion. While natural occurring vesicles have the advantage of being fully compatible with their host, artificial vesicles can be easily synthetized and functionalized according to the target to reach. Research is striving to merge the advantages of natural and artificial vesicles, in order to provide a new generation of highly performing vesicles, which would improve the therapeutic index of transported molecules. This progress report summarizes current manufacturing techniques used to produce both natural and artificial vesicles, exploring the promises and pitfalls of the different production processes. Finally, pros and cons of natural versus artificial vesicles are discussed and compared, with special regard toward the current applications of both kinds of vesicles in the healthcare field.
Collapse
Affiliation(s)
- Loredana Leggio
- Department of Biomedical and Biotechnological Sciences University of Catania Torre Biologica, Via S. Sofia 97 Catania 95125 Italy
| | - Giuseppe Arrabito
- Department of Physics and Chemistry – Emilio Segrè University of Palermo Building 17, Viale delle Scienze Palermo 90128 Italy
| | - Vittorio Ferrara
- Department of Chemical Sciences University of Catania Viale Andrea Doria 6 Catania 95125 Italy
| | - Silvia Vivarelli
- Department of Biomedical and Biotechnological Sciences University of Catania Torre Biologica, Via S. Sofia 97 Catania 95125 Italy
| | - Greta Paternò
- Department of Biomedical and Biotechnological Sciences University of Catania Torre Biologica, Via S. Sofia 97 Catania 95125 Italy
| | - Bianca Marchetti
- Department of Biomedical and Biotechnological Sciences University of Catania Torre Biologica, Via S. Sofia 97 Catania 95125 Italy
- Neuropharmacology Section OASI Institute for Research and Care on Mental Retardation and Brain Aging Troina 94018 Italy
| | - Bruno Pignataro
- Department of Physics and Chemistry – Emilio Segrè University of Palermo Building 17, Viale delle Scienze Palermo 90128 Italy
| | - Nunzio Iraci
- Department of Biomedical and Biotechnological Sciences University of Catania Torre Biologica, Via S. Sofia 97 Catania 95125 Italy
| |
Collapse
|
205
|
Guo P, Huang J, Moses MA. Cancer Nanomedicines in an Evolving Oncology Landscape. Trends Pharmacol Sci 2020; 41:730-742. [PMID: 32873407 DOI: 10.1016/j.tips.2020.08.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 07/21/2020] [Accepted: 08/02/2020] [Indexed: 12/12/2022]
Abstract
Nanomedicine represents an important class of cancer therapy. Clinical translation of cancer nanomedicine has significantly reduced the toxicity and adverse consequences of standard-of-care chemotherapy. Recent advances in new cancer treatment modalities (e.g., gene and immune therapies) are profoundly changing the oncology landscape, bringing with them new requirements and challenges for next-generation cancer nanomedicines. We present an overview of cancer nanomedicines in four emerging oncology-associated fields: (i) gene therapy, (ii) immunotherapy, (iii) extracellular vesicle (EV) therapy, and (iv) machine learning-assisted therapy. We discuss the incorporation of nanomedicine into these emerging disciplines, present prominent examples, and evaluate their advantages and challenges. Finally, we discuss future opportunities for next-generation cancer nanomedicines.
Collapse
Affiliation(s)
- Peng Guo
- Vascular Biology Program, Boston Children's Hospital, Boston, MA, USA; Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA.
| | - Jing Huang
- Vascular Biology Program, Boston Children's Hospital, Boston, MA, USA; Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Marsha A Moses
- Vascular Biology Program, Boston Children's Hospital, Boston, MA, USA; Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
206
|
孟 婉, 李 灵, 朱 桂. [Prospects and challenges of exosomes as drug delivery systems]. SHENG WU YI XUE GONG CHENG XUE ZA ZHI = JOURNAL OF BIOMEDICAL ENGINEERING = SHENGWU YIXUE GONGCHENGXUE ZAZHI 2020; 37:714-720. [PMID: 32840090 PMCID: PMC10319532 DOI: 10.7507/1001-5515.201810027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Indexed: 11/03/2022]
Abstract
Exosomes are nanoscale vectors with a diameter of 30~100 nm secreted by living cells, and they are important media for intercellular communication. Recent studies have demonstrated that exosomes can not only serve as biomarkers for diagnosis, but also have great potential as natural drug delivery vectors. Exosomes can be loaded with therapeutic cargos, including small molecules, proteins, and oligonucleotides. Meanwhile, the unique biological compatibility, high stability, and tumor targeting of exosomes make them attractive in future tumor therapy. Though exosomes can effectively deliver bioactive materials to receptor cells, there is a wide gap between our current understanding of exosomes and their application as ideal drug delivery systems. In this review, we will briefly introduce the function and composition of exosomes, and mainly summarize the potential advantages and challenges of exosomes as drug carriers. Finally, this review is expected to provide new ideas for the development of exosome-based drug delivery systems.
Collapse
Affiliation(s)
- 婉蓉 孟
- 电子科技大学 医学院附属肿瘤医院 四川省肿瘤医院(研究所)四川省癌症防治中心 四川省放射肿瘤学重点实验室(成都 610041)Sichuan Cancer Hospital& Institute, Sichuan Cancer Center, Sichuan Key Laboratory of Radiation Oncology School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, P.R. China
| | - 灵 李
- 电子科技大学 医学院附属肿瘤医院 四川省肿瘤医院(研究所)四川省癌症防治中心 四川省放射肿瘤学重点实验室(成都 610041)Sichuan Cancer Hospital& Institute, Sichuan Cancer Center, Sichuan Key Laboratory of Radiation Oncology School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, P.R. China
| | - 桂全 朱
- 电子科技大学 医学院附属肿瘤医院 四川省肿瘤医院(研究所)四川省癌症防治中心 四川省放射肿瘤学重点实验室(成都 610041)Sichuan Cancer Hospital& Institute, Sichuan Cancer Center, Sichuan Key Laboratory of Radiation Oncology School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, P.R. China
| |
Collapse
|
207
|
Gupta D, Liang X, Pavlova S, Wiklander OPB, Corso G, Zhao Y, Saher O, Bost J, Zickler AM, Piffko A, Maire CL, Ricklefs FL, Gustafsson O, Llorente VC, Gustafsson MO, Bostancioglu RB, Mamand DR, Hagey DW, Görgens A, Nordin JZ, El Andaloussi S. Quantification of extracellular vesicles in vitro and in vivo using sensitive bioluminescence imaging. J Extracell Vesicles 2020; 9:1800222. [PMID: 32944187 PMCID: PMC7481830 DOI: 10.1080/20013078.2020.1800222] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Extracellular vesicles (EVs) are naturally occurring nano-sized carriers that are secreted by cells and facilitate cell-to-cell communication by their unique ability to transfer biologically active cargo. Despite the pronounced increase in our understanding of EVs over the last decade, from disease pathophysiology to therapeutic drug delivery, improved molecular tools to track their therapeutic delivery are still needed. Unfortunately, the present catalogue of tools utilised for EV labelling lacks sensitivity or are not sufficiently specific. Here, we have explored the bioluminescent labelling of EVs using different luciferase enzymes tethered to CD63 to achieve a highly sensitive system for in vitro and in vivo tracking of EVs. Using tetraspanin fusions to either NanoLuc or ThermoLuc permits performing highly sensitive in vivo quantification of EVs or real-time imaging, respectively, at low cost and in a semi-high throughput manner. We find that the in vivo distribution pattern of EVs is determined by the route of injection, but that different EV subpopulations display differences in biodistribution patterns. By applying this technology for real-time non-invasive in vivo imaging of EVs, we show that their distribution to different internal organs occurs just minutes after administration.
Collapse
Affiliation(s)
- Dhanu Gupta
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Xiuming Liang
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Svetlana Pavlova
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Oscar P B Wiklander
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Giulia Corso
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ying Zhao
- Experimental Cancer Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Clinical Research Center, Karolinska University Hospital, Stockholm, Sweden
| | - Osama Saher
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Department Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Jeremy Bost
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Antje M Zickler
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Andras Piffko
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Cecile L Maire
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Franz L Ricklefs
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Oskar Gustafsson
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Manuela O Gustafsson
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - R Beklem Bostancioglu
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Doste R Mamand
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Biology, Faculty of Science, Cihan University-Erbil, Iraq
| | - Daniel W Hagey
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - André Görgens
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Joel Z Nordin
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Samir El Andaloussi
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
208
|
Cabeza L, Perazzoli G, Peña M, Cepero A, Luque C, Melguizo C, Prados J. Cancer therapy based on extracellular vesicles as drug delivery vehicles. J Control Release 2020; 327:296-315. [PMID: 32814093 DOI: 10.1016/j.jconrel.2020.08.018] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/11/2020] [Accepted: 08/14/2020] [Indexed: 02/07/2023]
Abstract
Extracellular vesicles (EVs) are lipid bilayer vesicles of nanometric size secreted by cells to communicate with other cells, either nearby or remotely. Their physicochemical properties make them a promising nanomedicine for drug transport and release in cancer therapy. In this review, we present the different types and biogenesis of EVs and highlight the importance of adequately selecting the cell of origin in cancer therapy. Furthermore, the main methodologies followed for the isolation of EVs and drug loading, as well as the modification and functionalization of these vesicles to generate EV-based nanocarriers are discussed. Finally, we review some of the main studies using drug-loaded exosomes in tumor therapy both in in vitro and in vivo models (even in resistant tumors). These investigations show promising results, achieving significant improvement in the antitumor effect of drugs in most cases. However, the number of clinical trials and patents based on these nanoformulations is still low, thus further research is still warranted in this area.
Collapse
Affiliation(s)
- Laura Cabeza
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain; Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
| | - Gloria Perazzoli
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain
| | - Mercedes Peña
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
| | - Ana Cepero
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
| | - Cristina Luque
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
| | - Consolacion Melguizo
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain; Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain.
| | - Jose Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain; Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
| |
Collapse
|
209
|
Jafari D, Malih S, Eini M, Jafari R, Gholipourmalekabadi M, Sadeghizadeh M, Samadikuchaksaraei A. Improvement, scaling-up, and downstream analysis of exosome production. Crit Rev Biotechnol 2020; 40:1098-1112. [PMID: 32772758 DOI: 10.1080/07388551.2020.1805406] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Exosomes are the most researched extracellular vesicles. In many biological, physiological, and pathological studies, they have been identified as suitable candidates for treatment and diagnosis of diseases by acting as the carriers of both drugs and genes. Considerable success has been achieved regarding the use of exosomes for tissue regeneration, cancer diagnosis, and targeted drug/gene delivery to specific tissues. While major progress has been made in exosome extraction and purification, extraction of large quantities of exosomes is still a major challenge. This issue limits the scope of both exosome-based research and therapeutic development. In this review, we have aimed to summarize experimental studies focused at increasing the number of exosomes. Biotechnological studies aimed at identifying the pathways of exosome biogenesis to manipulate some genes in order to increase the production of exosomes. Generally, two major strategies are employed to increase the production of exosomes. First, oogenesis pathways are genetically manipulated to overexpress activator genes of exosome biogenesis and downregulate the genes involved in exosome recycling pathways. Second, manipulation of the cell culture medium, treatment with specific drugs, and limiting certain conditions can force the cell to produce more exosomes. In this study, we have reviewed and categorized these strategies. It is hoped that the information presented in this review will provide a better understanding for expanding biotechnological approaches in exosome-based therapeutic development.
Collapse
Affiliation(s)
- Davod Jafari
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.,Student Research Committee, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sara Malih
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Maryam Eini
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Rasool Jafari
- Department of Medical Parasitology and Mycology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Mazaher Gholipourmalekabadi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Sadeghizadeh
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ali Samadikuchaksaraei
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.,Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
210
|
Bheri S, Hoffman JR, Park HJ, Davis ME. Biomimetic nanovesicle design for cardiac tissue repair. Nanomedicine (Lond) 2020; 15:1873-1896. [PMID: 32752925 DOI: 10.2217/nnm-2020-0097] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease is a major cause of mortality and morbidity worldwide. Exosome therapies are promising for cardiac repair. Exosomes transfer cargo between cells, have high uptake by native cells and are ideal natural carriers for proteins and nucleic acids. Despite their proreparative potential, exosome production is dependent on parent cell state with typically low yields and cargo variability. Therefore, there is potential value in engineering exosomes to maximize their benefits by delivering customized, potent cargo for cardiovascular disease. Here, we outline several methods of exosome engineering focusing on three important aspects: optimizing cargo, homing to target tissue and minimizing clearance. Finally, we put these methods in context of the cardiac field and discuss the future potential of vesicle design.
Collapse
Affiliation(s)
- Sruti Bheri
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, GA 30332, USA
| | - Jessica R Hoffman
- Molecular & Systems Pharmacology Graduate Training Program, Graduate Division of Biological & Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, GA 30322, USA
| | - Hyun-Ji Park
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, GA 30332, USA
| | - Michael E Davis
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, GA 30332, USA.,Department of Pediatrics, Division of Pediatric Cardiology, School of Medicine, Emory University, Atlanta, GA 30322, USA.,Children's Heart Research & Outcomes (HeRO) Center, Children's Healthcare of Atlanta & Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
211
|
Salunkhe S, Dheeraj, Basak M, Chitkara D, Mittal A. Surface functionalization of exosomes for target-specific delivery and in vivo imaging & tracking: Strategies and significance. J Control Release 2020; 326:599-614. [PMID: 32730952 DOI: 10.1016/j.jconrel.2020.07.042] [Citation(s) in RCA: 254] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/24/2020] [Accepted: 07/24/2020] [Indexed: 12/17/2022]
Abstract
Exosomes are natural nanovesicles excreted by many cells for intercellular communication and for transfer of materials including proteins, nucleic acids and even synthetic therapeutic agents. Surface modification of exosomes imparts additional functionality to the exosomes to enable site specific drug delivery and in vivo imaging and tracking and is an emerging area in drug delivery research. The present review focuses upon these modifications on the exosomal surface, the chemistry involved and their impact on targeted drug delivery for the treatment of brain, breast, lung, liver, colon tumors and, heart diseases and for understanding their in vivo fate including their uptake mechanisms, pharmacokinetics and biodistribution. The specific exosomal membrane proteins such as tetraspanins (CD63, CD81, CD9), lactadherin (LA), lysosome associated membrane protein-2b (Lamp-2b) and, glycosyl-phosphatidyl-inositol (GPI) involved in functionalization of exosome surface have also been discussed along with different strategies of surface modification like genetic engineering, covalent modification (click chemistry and metabolic engineering of parent cells of exosomes) and non-covalent modification (multivalent electrostatic interactions, ligand-receptor interaction, hydrophobic interaction, aptamer based modification and modification by anchoring CP05 peptide) along with optical (fluorescent and bioluminescent) and radioactive isotope labelling techniques of exosomes for imaging purpose.
Collapse
Affiliation(s)
- Shubham Salunkhe
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani, Rajasthan 333031, India
| | - Dheeraj
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani, Rajasthan 333031, India
| | - Moumita Basak
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani, Rajasthan 333031, India
| | - Deepak Chitkara
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani, Rajasthan 333031, India
| | - Anupama Mittal
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani, Rajasthan 333031, India.
| |
Collapse
|
212
|
Ma D, Gao X, Liu Z, Lu X, Ju H, Zhang N. Exosome-transferred long non-coding RNA ASMTL-AS1 contributes to malignant phenotypes in residual hepatocellular carcinoma after insufficient radiofrequency ablation. Cell Prolif 2020; 53:e12795. [PMID: 32722884 PMCID: PMC7507479 DOI: 10.1111/cpr.12795] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/20/2020] [Accepted: 02/03/2020] [Indexed: 12/12/2022] Open
Abstract
Objectives Long non‐coding RNAs (lncRNAs) are emerging RNA regulators in cancer progression, including in hepatocellular carcinoma (HCC). Recently, insufficient radiofrequency ablation (RFA) has been reported to lead to recurrence and metastasis of residual HCC tumours. Herein, we aimed to the role of ASMTL‐AS1 in residual HCC after insufficient RFA. Materials and methods In vitro insufficient RFA model was simulated in Huh7 cells and subsequently named Huh7‐H cells. In vitro and in vivo assays were conducted to investigate ASMTL‐AS1 function in HCC. Results LncRNA ASMTL‐AS1 low expressed in normal human liver was found to be highly expressed in HCC tissues and further increased in tumours after insufficient RFA. ASMTL‐AS1 expression was related to stage, metastasis and prognosis in HCC. Huh7‐H possessed higher ASMTL‐AS1 level and more aggressive than Huh7 cells. ASMTL‐AS1 contributed to the malignancy of HCC cells both in vitro and in vivo. Mechanistically, ASMTL‐AS1 was trans‐activated by MYC and promoted NLK expression to activate YAP signalling via sequestering miR‐342‐3p in HCC. Interestingly, ASMTL‐AS1 could be wrapped by exosomes and then convey malignancy through NLK/YAP axis between cells even in residual HCC after insufficient RFA. Conclusions Exosomal ASMTL‐AS1 aggravates the malignancy in residual HCC after insufficient RFA via miR‐342‐3p/NLK/YAP signalling, opening a new road for the treatment of HCC and the prevention of recurrence or metastasis of residual HCC after insufficient RFA.
Collapse
Affiliation(s)
- Dening Ma
- Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, 1 Banshan East Road, Hangzhou, 310022, China.,Department of Colorectal Surgery, Cancer Hospital of the University of Chinese Academy of Science, 1 Banshan East Road, Hangzhou, 310022, China.,Department of Colorectal Surgery, Zhejiang Cancer Hospital, 1 Banshan East Road, Hangzhou, 310022, China
| | - Xinyi Gao
- Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, 1 Banshan East Road, Hangzhou, 310022, China.,Department of Radiology, Cancer Hospital of the University of Chinese Academy of Sciences, 1 Banshan East Road, Hangzhou, 310022, China.,Department of Radiology, Zhejiang Cancer Hospital, 1 Banshan East Road, Hangzhou, 310022, China
| | - Zhuo Liu
- Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, 1 Banshan East Road, Hangzhou, 310022, China.,Department of Colorectal Surgery, Cancer Hospital of the University of Chinese Academy of Science, 1 Banshan East Road, Hangzhou, 310022, China.,Department of Colorectal Surgery, Zhejiang Cancer Hospital, 1 Banshan East Road, Hangzhou, 310022, China
| | - Xingang Lu
- Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, 1 Banshan East Road, Hangzhou, 310022, China.,Department of Colorectal Surgery, Cancer Hospital of the University of Chinese Academy of Science, 1 Banshan East Road, Hangzhou, 310022, China.,Department of Colorectal Surgery, Zhejiang Cancer Hospital, 1 Banshan East Road, Hangzhou, 310022, China
| | - Haixing Ju
- Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, 1 Banshan East Road, Hangzhou, 310022, China.,Department of Colorectal Surgery, Cancer Hospital of the University of Chinese Academy of Science, 1 Banshan East Road, Hangzhou, 310022, China.,Department of Colorectal Surgery, Zhejiang Cancer Hospital, 1 Banshan East Road, Hangzhou, 310022, China
| | - Ning Zhang
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Cancer Hospital, 270 Dong-An Road, Shanghai, 200032, China
| |
Collapse
|
213
|
Tang TT, Wang B, Lv LL, Liu BC. Extracellular vesicle-based Nanotherapeutics: Emerging frontiers in anti-inflammatory therapy. Theranostics 2020; 10:8111-8129. [PMID: 32724461 PMCID: PMC7381724 DOI: 10.7150/thno.47865] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 06/19/2020] [Indexed: 12/19/2022] Open
Abstract
Dysregulated inflammation is a complicated pathological process involved in various diseases, and the treatment of inflammation-linked disorders currently represents an enormous global burden. Extracellular vesicles (EVs) are nanosized, lipid membrane-enclosed vesicles secreted by virtually all types of cells, which act as an important intercellular communicative medium. Considering their capacity to transfer bioactive substances, both unmodified and engineered EVs are increasingly being explored as potential therapeutic agents or therapeutic vehicles. Moreover, as the nature's own delivery tool, EVs possess many desirable advantages, such as stability, biocompatibility, low immunogenicity, low toxicity, and biological barrier permeability. The application of EV-based therapy to combat inflammation, though still in an early stage of development, has profound transformative potential. In this review, we highlight the recent progress in EV engineering for inflammation targeting and modulation, summarize their preclinical applications in the treatment of inflammatory disorders, and present our views on the anti-inflammatory applications of EV-based nanotherapeutics.
Collapse
|
214
|
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: 74] [Impact Index Per Article: 14.8] [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
|
215
|
Cao J, Wang B, Tang T, Lv L, Ding Z, Li Z, Hu R, Wei Q, Shen A, Fu Y, Liu B. Three-dimensional culture of MSCs produces exosomes with improved yield and enhanced therapeutic efficacy for cisplatin-induced acute kidney injury. Stem Cell Res Ther 2020; 11:206. [PMID: 32460853 PMCID: PMC7251891 DOI: 10.1186/s13287-020-01719-2] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/16/2020] [Accepted: 05/08/2020] [Indexed: 02/08/2023] Open
Abstract
Background Exosomes derived from mesenchymal stem cells (MSC-exos) have been demonstrated with great potential in the treatment of multiple human diseases including acute kidney injury (AKI) by virtue of their intrinsic cargoes. However, there are major challenges of low yield and the lack of an established biomanufacturing platform to efficiently produce MSC-exos, thereby limiting their therapeutic application. Here, we aimed to establish a novel strategy to produce MSC-exos with a hollow fiber bioreactor-based three-dimensional (3D) culture system and evaluate the therapeutic efficacy of 3D-exosomes (3D-exos) on AKI. Methods Mesenchymal stem cells (MSCs) were isolated from fresh human umbilical cord and cultured in two-dimensional (2D) flasks. 2 × 108 MSCs were inoculated into the hollow fiber bioreactor for 3D culture. The culture supernatants were collected every 1 or 2 days for isolating exosomes. Exosomes from 2D (2D-exos) and 3D cultures were characterized by transmission electron microscopy, nanoparticle tracking analysis, and western blotting analysis of exosome markers. The yield of exosomes from 2 × 108 MSCs seeded in 2D and 3D culture system was compared, based on protein quantification. The therapeutic efficacy of 2D-exos and 3D-exos was investigated in a murine model of cisplatin-induced AKI in vivo and in vitro. Results 3D culture did not significantly change the surface markers of MSCs, as well as the morphology, size, and exosomal markers of 3D-exos when compared to those of 2D-exos. Compared with conventional 2D culture, the 3D culture system increased total exosome production up to 19.4-fold. 3D-exos were more concentrated in the harvested supernatants (15.5-fold) than 2D-exos, which led to a higher exosome collection efficiency of 3D culture system. In vivo, both 2D-exos and 3D-exos significantly alleviated cisplatin-induced murine AKI evidenced by improved renal function, attenuated pathological changes of renal tubules, reduced inflammatory factors, and repressed T cell and macrophage infiltration. Impressively, 3D-exos were more effective than 2D-exos. Moreover, 3D-exos were taken up by tubular epithelial cells (TECs) with improved efficiency, thereby exhibiting superior anti-inflammatory effect and improved viability of TECs in vitro. Conclusions In summary, our findings demonstrate that the hollow fiber 3D culture system provides an efficient strategy for the continuous production of MSC-exos which has enhanced therapeutic potential for cisplatin-induced AKI.
Collapse
Affiliation(s)
- Jingyuan Cao
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, 210009, Jiangsu Province, China
| | - Bin Wang
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, 210009, Jiangsu Province, China
| | - Taotao Tang
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, 210009, Jiangsu Province, China
| | - Linli Lv
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, 210009, Jiangsu Province, China
| | - Zhaoying Ding
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, 210009, Jiangsu Province, China
| | - Zuolin Li
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, 210009, Jiangsu Province, China
| | - Ruoyu Hu
- Department of Cardiothoracic Surgery, Zhongda Hospital, Southeast University School of Medicine, Nanjing, 210009, Jiangsu Province, China
| | - Qing Wei
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, 210009, Jiangsu Province, China
| | - Anran Shen
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, 210009, Jiangsu Province, China
| | - Yuqi Fu
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, 210009, Jiangsu Province, China
| | - Bicheng Liu
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, 210009, Jiangsu Province, China.
| |
Collapse
|
216
|
Gao J, Dong X, Wang Z. Generation, purification and engineering of extracellular vesicles and their biomedical applications. Methods 2020; 177:114-125. [PMID: 31790730 PMCID: PMC7198327 DOI: 10.1016/j.ymeth.2019.11.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/23/2019] [Accepted: 11/25/2019] [Indexed: 12/20/2022] Open
Abstract
Extracellular vesicles (EVs), derived from cell membranes, demonstrate the potential to be excellent therapeutics and drug carriers. Although EVs are promising, the process to develop high-quality and scalable EVs for their translation is demanding. Within this research, we analyzed the production of EVs, their purification and their post-bioengineering, and we also discussed the biomedical applications of EVs. We focus on the developments of methods in producing EVs including biological, physical, and chemical approaches. Furthermore, we discuss the challenges and the opportunities that arose when we translated EVs in clinic. With the advancements in nanotechnology and immunology, genetically engineering EVs is a new frontier in developing new therapeutics in order to tailor to individuals and different disease stages in treatments of cancer and inflammatory diseases.
Collapse
Affiliation(s)
- Jin Gao
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| | - Xinyue Dong
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| | - Zhenjia Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA.
| |
Collapse
|
217
|
Elsharkasy OM, Nordin JZ, Hagey DW, de Jong OG, Schiffelers RM, Andaloussi SEL, Vader P. Extracellular vesicles as drug delivery systems: Why and how? Adv Drug Deliv Rev 2020; 159:332-343. [PMID: 32305351 DOI: 10.1016/j.addr.2020.04.004] [Citation(s) in RCA: 721] [Impact Index Per Article: 144.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 03/09/2020] [Accepted: 04/14/2020] [Indexed: 12/16/2022]
Abstract
Over the past decades, a multitude of synthetic drug delivery systems has been developed and introduced to the market. However, applications of such systems are limited due to inefficiency, cytotoxicity and/or immunogenicity. At the same time, the field of natural drug carrier systems has grown rapidly. One of the most prominent examples of such natural carriers are extracellular vesicles (EVs). EVs are cell-derived membranous particles which play important roles in intercellular communication. EVs possess a number of characteristics that qualify them as promising vehicles for drug delivery. In order to take advantage of these attributes, an in-depth understanding of why EVs are such unique carrier systems and how we can exploit their qualities is pivotal. Here, we review unique EV features that are relevant for drug delivery and highlight emerging strategies to make use of those features for drug loading and targeted delivery.
Collapse
|
218
|
Takakura Y, Matsumoto A, Takahashi Y. Therapeutic Application of Small Extracellular Vesicles (sEVs): Pharmaceutical and Pharmacokinetic Challenges. Biol Pharm Bull 2020; 43:576-583. [DOI: 10.1248/bpb.b19-00831] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
| | | | - Yuki Takahashi
- Graduate School of Pharmaceutical Sciences, Kyoto University
| |
Collapse
|
219
|
Jeske R, Bejoy J, Marzano M, Li Y. Human Pluripotent Stem Cell-Derived Extracellular Vesicles: Characteristics and Applications. TISSUE ENGINEERING. PART B, REVIEWS 2020; 26:129-144. [PMID: 31847715 PMCID: PMC7187972 DOI: 10.1089/ten.teb.2019.0252] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 12/16/2019] [Indexed: 02/06/2023]
Abstract
Extracellular vesicles (EVs), including exosomes and microvesicles, are found to play an important role in various biological processes and maintaining tissue homeostasis. Because of the protective effects, stem cell-derived EVs can be used to reduce oxidative stress and apoptosis in the recipient cells. In addition, EVs/exosomes have been used as directional communication tools between stem cells and parenchymal cells, giving them the ability to serve as biomarkers. Likewise, altered EVs/exosomes can be utilized for drug delivery by loading with proteins, small interfering RNAs, and viral vectors, in particular, because EVs/exosomes are able to cross the blood-brain barrier. In this review article, the properties of human induced pluripotent stem cell (iPSC)-derived EVs are discussed. The biogenesis, that is, how EVs originate in the endosomal compartment or from the cell layer of microvesicles, EV composition, the available methods of purification, and characterizations of EVs/exosomes are summarized. In particular, EVs/exosomes derived from iPSCs of different lineage specifications and the applications of these stem cell-derived exosomes in neurological diseases are discussed. Impact statement In this review, we summarized the work related to extracellular vesicles (EVs) derived from human pluripotent stem cells (hPSCs). In particular, EVs/exosomes derived from hPSCs of different lineage specifications and the applications of these stem cell-derived exosomes in neurological diseases are discussed. The results highlight the important role of cell-cell interactions in neural cellular phenotype and neurodegeneration. The findings reported in this article are significant for pluripotent stem cell-derived cell-free products toward applications in stem cell-based therapies.
Collapse
Affiliation(s)
- Richard Jeske
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida
| | - Julie Bejoy
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida
| | - Mark Marzano
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida
| |
Collapse
|
220
|
Therapeutic Advances of Stem Cell-Derived Extracellular Vesicles in Regenerative Medicine. Cells 2020; 9:cells9030707. [PMID: 32183102 PMCID: PMC7140663 DOI: 10.3390/cells9030707] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 03/07/2020] [Accepted: 03/10/2020] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles (EVs), which are the main paracrine components of stem cells, mimic the regenerative capacity of these cells. Stem cell-derived EVs (SC-EVs) have been used for the treatment of various forms of tissue injury in preclinical trials through maintenance of their stemness, induction of regenerative phenotypes, apoptosis inhibition, and immune regulation. The efficiency of SC-EVs may be enhanced by selecting the appropriate EV-producing cells and cell phenotypes, optimizing cell culture conditions for the production of optimal EVs, and further engineering the EVs produced to transport therapeutic and targeting molecules.
Collapse
|
221
|
Wang K, Wei Y, Zhang P, Wang J, Hu J, Wang L, Li B. [Progress in extracellular vesicle imaging methods]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2020; 40:279-286. [PMID: 32376541 DOI: 10.12122/j.issn.1673-4254.2020.02.22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Extracellular vesicles (EVs), including exosomes and microvesicles, are lipid bilayer-enclosed nanovesicles secreted by cells. These EVs are important mediators of intercellular communication by serving as vehicles for transfer of proteins, mRNA, miRNA and lipids between cells. Various visualization methods have been established to explore the characteristics of EVs and their role in physiological and pathological processes. The nanoscale size and high heterogeneity of EVs hamper the identification of their biological characteristics and functions. This review presents a comprehensive overview of EV imaging methods in light of the origin, separation and dynamic tracking of EVs, and the advantages and disadvantages of different imaging strategies are discussed. We believe that studies at the levels of single vesicles and single cells will become the frontier of future researches of EVs.
Collapse
Affiliation(s)
- Kaizhe Wang
- CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuhui Wei
- CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China.,Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Ping Zhang
- CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jianhua Wang
- School of Physics Science and Technology, Ningbo University, Ningbo 315211, China
| | - Jun Hu
- CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China.,Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Lihua Wang
- CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China.,Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Bin Li
- CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China.,Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| |
Collapse
|
222
|
Soriani A, Vulpis E, Cuollo L, Santoni A, Zingoni A. Cancer extracellular vesicles as novel regulators of NK cell response. Cytokine Growth Factor Rev 2020; 51:19-26. [DOI: 10.1016/j.cytogfr.2019.11.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 11/28/2019] [Accepted: 11/29/2019] [Indexed: 12/21/2022]
|
223
|
Merckx G, Hosseinkhani B, Kuypers S, Deville S, Irobi J, Nelissen I, Michiels L, Lambrichts I, Bronckaers A. Angiogenic Effects of Human Dental Pulp and Bone Marrow-Derived Mesenchymal Stromal Cells and their Extracellular Vesicles. Cells 2020; 9:cells9020312. [PMID: 32012900 PMCID: PMC7072370 DOI: 10.3390/cells9020312] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/23/2020] [Accepted: 01/25/2020] [Indexed: 12/18/2022] Open
Abstract
Blood vessel formation or angiogenesis is a key process for successful tooth regeneration. Bone marrow-derived mesenchymal stromal cells (BM-MSCs) possess paracrine proangiogenic properties, which are, at least partially, induced by their extracellular vesicles (EVs). However, the isolation of BM-MSCs is associated with several drawbacks, which could be overcome by MSC-like cells of the teeth, called dental pulp stromal cells (DPSCs). This study aims to compare the angiogenic content and functions of DPSC and BM-MSC EVs and conditioned medium (CM). The angiogenic protein profile of DPSC- and BM-MSC-derived EVs, CM and EV-depleted CM was screened by an antibody array and confirmed by ELISA. Functional angiogenic effects were tested in transwell migration and chicken chorioallantoic membrane assays. All secretion fractions contained several pro- and anti-angiogenic proteins and induced in vitro endothelial cell motility. This chemotactic potential was higher for (EV-depleted) CM, compared to EVs with a stronger effect for BM-MSCs. Finally, BM-MSC CM, but not DPSC CM, nor EVs, increased in ovo angiogenesis. In conclusion, we showed that DPSCs are less potent in relation to endothelial cell chemotaxis and in ovo neovascularization, compared to BM-MSCs, which emphasizes the importance of choice of cell type and secretion fraction for stem cell-based regenerative therapies in inducing angiogenesis.
Collapse
Affiliation(s)
- Greet Merckx
- UHasselt - Hasselt University, Faculty of Medicine and Life Sciences, Biomedical Research Institute (BIOMED), Agoralaan, 3590 Diepenbeek, Belgium
| | - Baharak Hosseinkhani
- UHasselt - Hasselt University, Faculty of Medicine and Life Sciences, Biomedical Research Institute (BIOMED), Agoralaan, 3590 Diepenbeek, Belgium
| | - Sören Kuypers
- UHasselt - Hasselt University, Faculty of Medicine and Life Sciences, Biomedical Research Institute (BIOMED), Agoralaan, 3590 Diepenbeek, Belgium
| | - Sarah Deville
- UHasselt - Hasselt University, Faculty of Medicine and Life Sciences, Biomedical Research Institute (BIOMED), Agoralaan, 3590 Diepenbeek, Belgium
- Flemish Institute for Technological Research (VITO), Health Department, Boeretang, 2400 Mol, Belgium
| | - Joy Irobi
- UHasselt - Hasselt University, Faculty of Medicine and Life Sciences, Biomedical Research Institute (BIOMED), Agoralaan, 3590 Diepenbeek, Belgium
| | - Inge Nelissen
- Flemish Institute for Technological Research (VITO), Health Department, Boeretang, 2400 Mol, Belgium
| | - Luc Michiels
- UHasselt - Hasselt University, Faculty of Medicine and Life Sciences, Biomedical Research Institute (BIOMED), Agoralaan, 3590 Diepenbeek, Belgium
| | - Ivo Lambrichts
- UHasselt - Hasselt University, Faculty of Medicine and Life Sciences, Biomedical Research Institute (BIOMED), Agoralaan, 3590 Diepenbeek, Belgium
| | - Annelies Bronckaers
- UHasselt - Hasselt University, Faculty of Medicine and Life Sciences, Biomedical Research Institute (BIOMED), Agoralaan, 3590 Diepenbeek, Belgium
- Correspondence: ; Tel.: +32-(0)-11-26-92-23
| |
Collapse
|
224
|
Imafuku A, Sjoqvist S. Extracellular Vesicle Therapeutics in Regenerative Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1312:131-138. [PMID: 33330962 DOI: 10.1007/5584_2020_599] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Extracellular vesicles (EVs) are nano-sized, cell-released vesicles which contain lipids, proteins, and nucleic acids derived from the parental cells. EVs play an important role in intercellular communication and influence both physiological and pathological conditions. They are increasingly explored as potential therapeutic agents since they can cross biological barriers, their cargo is protected from degradation and they are involved in the transfer of bioactive components. EVs can promote tissue regeneration and might be alternatives to cell therapy. They can be used both in their native form, and as delivery vehicles for therapeutic agents. However, there are many hurdles to overcome for broad clinical application of EVs as therapeutics. Here, we review recent conditions regarding EVs therapeutics in regenerative medicine.
Collapse
Affiliation(s)
| | - Sebastian Sjoqvist
- ExTherea Inc, Fujisawa, Kanagawa, Japan.
- Department of Clinical Sciences, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
225
|
Zhou Y, Xiao Y. The Development of Extracellular Vesicle-Integrated Biomaterials for Bone Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1250:97-108. [PMID: 32601940 DOI: 10.1007/978-981-15-3262-7_7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The clinical need for effective bone regeneration remains in huge demands. Although autologous and allogeneic bone grafts are generally considered "gold standard" treatments for bone defects, these approaches may result in various complications. Furthermore, safety considerations of gene- and cell-based therapies require further clarification and approval from regulatory authorities. Therefore, developing new therapeutic biomaterials that can empower endogenous regenerative properties to accelerate bone repair and regeneration is of great significance. Extracellular vesicles (EVs) comprise a heterogeneous population of naturally derived nanoparticles that play a critical role in mediating cell-cell communication. The vast amount of biological processes that EVs are involved in, such as immune modulation, senescence, and angiogenesis, and the versatility of manner in which they can influence the behavior of recipient cells make EVs an interesting source for both diagnostic and therapeutic applications. Advancement of knowledge in the fields of immunology and cell biology has sparked the exploration of the potential of EVs in the field of regenerative medicine. EVs travel between cells and deliver functional cargoes, such as proteins and RNAs, thereby regulating the recruitment, proliferation, and differentiation of recipient cells. Numerous studies have demonstrated the pivotal role of EVs in tissue regeneration both in vitro and in vivo. In this chapter, we will outline current knowledge surrounding EVs, summarize their functional roles in bone regenerative medicine, and elaborate on potential application and challenges of EV-integrated biomaterials in bone tissue engineering.
Collapse
Affiliation(s)
- Yinghong Zhou
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Brisbane, QLD, Australia. .,Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou, China. .,The Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology (QUT), Brisbane, QLD, Australia.
| | - Yin Xiao
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Brisbane, QLD, Australia.,Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou, China.,The Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology (QUT), Brisbane, QLD, Australia
| |
Collapse
|
226
|
Yan L, Wu X. Exosomes produced from 3D cultures of umbilical cord mesenchymal stem cells in a hollow-fiber bioreactor show improved osteochondral regeneration activity. Cell Biol Toxicol 2019; 36:165-178. [PMID: 31820164 PMCID: PMC7196084 DOI: 10.1007/s10565-019-09504-5] [Citation(s) in RCA: 148] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 11/21/2019] [Indexed: 01/09/2023]
Abstract
Animal and clinical studies have shown that mesenchymal stem cells (MSCs) play an important role in cartilage repair. The therapeutic effect of mesenchymal stem cells based therapies has been increasingly demonstrated to exosome-mediated paracrine secretion. Here, we investigated the cellular processes and mechanism of exosomes produced by conventional 2D culture (2D-Exos) and exosomes produced from 3D culture (3D-Exos) of umbilical MSCs (U-MSCs) in a hollow-fiber bioreactor for the treatment of cartilage repair. We found that the yield of 3D-Exos was 7.5-fold higher than that of 2D-Exos. The in vitro experiments indicated that both 2D-Exos and 3D-Exos can stimulate chondrocyte proliferation, migration, and matrix synthesis, and inhibit apoptosis, with 3D-Exos exerting a stronger effect than 2D-Exos. This effect was partly attributed to the activation of transforming growth factor beta 1 and Smad2/3 signaling. The injection of 2D-Exos and 3D-Exos showed enhanced gross appearance and attenuated cartilage defect; however, 3D-Exos showed a superior therapeutic effect than 2D-Exos. In summary, our study provides novel insights into the chondroprotective effects of exosomes produced from 3D culture of U-MSCs in a hollow-fiber bioreactor. Because of its promising biological function and high yield, 3D-Exos may become a promising therapeutic method for the treatment of cartilage defects.
Collapse
Affiliation(s)
- Litao Yan
- Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, People's Republic of China
| | - Xing Wu
- Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, People's Republic of China.
| |
Collapse
|
227
|
Biodistribution of Mesenchymal Stem Cell-Derived Extracellular Vesicles in a Radiation Injury Bone Marrow Murine Model. Int J Mol Sci 2019; 20:ijms20215468. [PMID: 31684046 PMCID: PMC6861905 DOI: 10.3390/ijms20215468] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 10/29/2019] [Accepted: 10/31/2019] [Indexed: 01/15/2023] Open
Abstract
We have previously shown that injury induced by irradiation to murine marrow can be partially or completely reversed by exposure to human or murine mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs). Investigation of the biodistribution of EVs in vivo is essential for understanding EV biology. In this study, we evaluated the DiD lipid dye labeled MSC-EV biodistribution in mice under different conditions, including different MSC-EV doses and injection schedules, time post MSC-EV injection, and doses of radiation. DiD-labeled MSC-EVs appeared highest in the liver and spleen; lower in bone marrow of the tibia, femur, and spine; and were undetectable in the heart, kidney and lung, while a predominant EV accumulation was detected in the lung of mice infused with human lung fibroblast cell derived EVs. There was significantly increased MSC-EV accumulation in the spleen and bone marrow (tibia and femur) post radiation appearing with an increase of MSC-EV uptake by CD11b+ and F4/80+ cells, but not by B220 cells, compared to those organs from non-irradiated mice. We further demonstrated that increasing levels of irradiation caused a selective increase in vesicle homing to marrow. This accumulation of MSC-EVs at the site of injured bone marrow could be detected as early as 1 h after MSC- EV injection and was not significantly different between 2 and 24 h post MSC-EV injection. Our study indicates that irradiation damage to hematopoietic tissue in the spleen and marrow targets MSC-EVs to these tissues.
Collapse
|
228
|
Kim H, Kim D, Nam H, Moon S, Kwon YJ, Lee JB. Engineered extracellular vesicles and their mimetics for clinical translation. Methods 2019; 177:80-94. [PMID: 31626895 DOI: 10.1016/j.ymeth.2019.10.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/08/2019] [Accepted: 10/11/2019] [Indexed: 12/12/2022] Open
Abstract
Cells secrete extracellular vesicles (EVs) to external environments to achieve cellular homeostasis and cell-to-cell communication. Their therapeutic potential has been constantly spotlighted since they mirror both cytoplasmic and membranous components of parental cells. Meanwhile, growing evidence suggests that EV engineering could further promote EVs with a maximized capacity. In this review, a range of engineering techniques as well as upscaling approaches to exploit EVs and their mimetics are introduced. By laying out the pros and cons of each technique from different perspectives, we sought to provide an overview potentially helpful for understanding the current state of the art EV engineering and a guideline for choosing a suitable technique for engineering EVs. Furthermore, we envision that the advances in each technique will give rise to the combinatorial engineering of EVs, taking us a step closer to a clinical translation of EV-based therapeutics.
Collapse
Affiliation(s)
- Hyejin Kim
- Department of Chemical Engineering, University of Seoul, 163 Seoulsiripdaero, Dongdaemungu, Seoul 02504, Republic of Korea
| | - Dajeong Kim
- Department of Chemical Engineering, University of Seoul, 163 Seoulsiripdaero, Dongdaemungu, Seoul 02504, Republic of Korea
| | - Hyangsu Nam
- Department of Chemical Engineering, University of Seoul, 163 Seoulsiripdaero, Dongdaemungu, Seoul 02504, Republic of Korea
| | - Sunghyun Moon
- Department of Chemical Engineering, University of Seoul, 163 Seoulsiripdaero, Dongdaemungu, Seoul 02504, Republic of Korea
| | - Young Jik Kwon
- Department of Chemical Engineering and Materials Science, University of California, Irvine, CA 92697, United States; Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697, United States; Department of Pharmaceutical Sciences, University of California, Irvine, CA 92697, United States; Department of Biomedical Engineering, University of California, Irvine, CA 92697, United States.
| | - Jong Bum Lee
- Department of Chemical Engineering, University of Seoul, 163 Seoulsiripdaero, Dongdaemungu, Seoul 02504, Republic of Korea.
| |
Collapse
|
229
|
Rashid MH, Borin TF, Ara R, Angara K, Cai J, Achyut BR, Liu Y, Arbab AS. Differential in vivo biodistribution of 131I-labeled exosomes from diverse cellular origins and its implication for theranostic application. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2019; 21:102072. [PMID: 31376572 PMCID: PMC6814553 DOI: 10.1016/j.nano.2019.102072] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 05/21/2019] [Accepted: 07/21/2019] [Indexed: 12/21/2022]
Abstract
Exosomes are critical mediators of intercellular crosstalk and are regulator of the cellular/tumor microenvironment. Exosomes have great prospects for clinical application as a theranostic and prognostic probe. Nevertheless, the advancement of exosomes research has been thwarted by our limited knowledge of the most efficient isolation method and their in vivo trafficking. Here we have shown that a combination of two size-based methods using a 0.20 μm syringe filter and 100 k centrifuge membrane filter followed by ultracentrifugation yields a greater number of uniform exosomes. We also demonstrated the visual representation and quantification of the differential in vivo distribution of radioisotope 131I-labeled exosomes from diverse cellular origins, e.g., tumor cells with or without treatments, myeloid-derived suppressor cells and endothelial progenitor cells. We also determined that the distribution was dependent on the exosomal protein/cytokine contents. The applied in vivo imaging modalities can be utilized to monitor disease progression, metastasis, and exosome-based targeted therapy.
Collapse
Affiliation(s)
- Mohammad H Rashid
- Laboratory of Tumor Angiogenesis, Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA, USA
| | - Thaiz F Borin
- Laboratory of Tumor Angiogenesis, Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA, USA
| | - Roxan Ara
- Laboratory of Tumor Angiogenesis, Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA, USA
| | - Kartik Angara
- Department of Pediatrics & Human Development, Grand Rapids Research Center, Michigan State University, Grand Rapids, MI, USA
| | - Jingwen Cai
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Bhagelu R Achyut
- Laboratory of Tumor Angiogenesis, Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA, USA; Cancer Animal Models Shared Resource, Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Yutao Liu
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Ali S Arbab
- Laboratory of Tumor Angiogenesis, Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA, USA.
| |
Collapse
|
230
|
Wang M, Wang C, Chen M, Xi Y, Cheng W, Mao C, Xu T, Zhang X, Lin C, Gao W, Guo Y, Lei B. Efficient Angiogenesis-Based Diabetic Wound Healing/Skin Reconstruction through Bioactive Antibacterial Adhesive Ultraviolet Shielding Nanodressing with Exosome Release. ACS NANO 2019; 13:10279-10293. [PMID: 31483606 DOI: 10.1021/acsnano.9b03656] [Citation(s) in RCA: 355] [Impact Index Per Article: 59.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Diabetic wound healing and angiogenesis remain a worldwide challenge for both clinic and research. The use of adipose stromal cell derived exosomes delivered by bioactive dressing provides a potential strategy for repairing diabetic wounds with less scar formation and fast healing. In this study, we fabricated an injectable adhesive thermosensitive multifunctional polysaccharide-based dressing (FEP) with sustained pH-responsive exosome release for promoting angiogenesis and diabetic wound healing. The FEP dressing possessed multifunctional properties including efficient antibacterial activity/multidrug-resistant bacteria, fast hemostatic ability, self-healing behavior, and tissue-adhesive and good UV-shielding performance. FEP@exosomes (FEP@exo) can significantly enhance the proliferation, migration, and tube formation of endothelial cells in vitro. In vivo results from a diabetic full-thickness cutaneous wound model showed that FEP@exo dressing accelerated the wound healing by stimulating the angiogenesis process of the wound tissue. The enhanced cell proliferation, granulation tissue formation, collagen deposition, remodeling, and re-epithelialization probably lead to the fast healing with less scar tissue formation and skin appendage regeneration. This study showed that combining bioactive molecules into multifunctional dressing should have great potential in achieving satisfactory healing in diabetic and other vascular-impaired related wounds.
Collapse
Affiliation(s)
- Min Wang
- Frontier Institute of Science and Technology, Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology , Xi'an Jiaotong University , Xi'an 710000 , China
| | - Chenggui Wang
- Key Laboratory of Orthopedics of Zhejiang Province , Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou 325027 , China
- Department of Orthopedics Surgery , Second Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou 310009 , China
| | - Mi Chen
- Frontier Institute of Science and Technology, Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology , Xi'an Jiaotong University , Xi'an 710000 , China
| | - Yuewei Xi
- Frontier Institute of Science and Technology, Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology , Xi'an Jiaotong University , Xi'an 710000 , China
| | - Wei Cheng
- Frontier Institute of Science and Technology, Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology , Xi'an Jiaotong University , Xi'an 710000 , China
| | - Cong Mao
- Key Laboratory of Orthopedics of Zhejiang Province , Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou 325027 , China
| | - Tianzhen Xu
- Key Laboratory of Orthopedics of Zhejiang Province , Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou 325027 , China
| | - Xingxing Zhang
- Department of Burn , First Affiliated Hospital of Wenzhou Medical University , Wenzhou 325000 , China
| | - Cai Lin
- Department of Burn , First Affiliated Hospital of Wenzhou Medical University , Wenzhou 325000 , China
| | - Weiyang Gao
- Key Laboratory of Orthopedics of Zhejiang Province , Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou 325027 , China
| | - Yi Guo
- Department of Biologic and Materials Science , University of Michigan , Ann Arbor , Michigan 48109 , United States
| | - Bo Lei
- Frontier Institute of Science and Technology, Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology , Xi'an Jiaotong University , Xi'an 710000 , China
| |
Collapse
|
231
|
Deng C, Xie Y, Zhang C, Ouyang B, Chen H, Lv L, Yao J, Liang X, Zhang Y, Sun X, Deng C, Liu G. Urine-Derived Stem Cells Facilitate Endogenous Spermatogenesis Restoration of Busulfan-Induced Nonobstructive Azoospermic Mice by Paracrine Exosomes. Stem Cells Dev 2019; 28:1322-1333. [PMID: 31311428 DOI: 10.1089/scd.2019.0026] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Nonobstructive azoospermia (NOA) is a severe form of male infertility, with limited effective treatments. Urine-derived stem cells (USCs) possess multipotent differentiation capacity and paracrine effects, and participate in tissue repair and regeneration. The aim of this study is to investigate whether the transplantation of USCs or USC exosomes (USC-exos) could promote endogenous spermatogenesis restoration in a busulfan-induced NOA mice model. USCs were cultured and characterized by flow cytometry. High-density USCs were cultured in a hollow fiber bioreactor for exosomes collection. USC-exos were isolated from USCs conditional media and identified by transmission electron microscopy, western blotting, and Flow NanoAnalyzer analysis. USC-exos exhibited sphere- or cup-shaped morphology with a mean diameter of 66.5 ± 16.0 nm, and expressed CD63 and CD9. USCs and USC-exos were transplanted into the interstitial space in the testes of NOA mice per the following groups: normal group; groups treated with no injection, phosphate-buffered saline (PBS), USCs or USC-exos on days 3 and 36 after busulfan administration, respectively. Thirty days after USCs and USC-exos transplantation, spermatogenesis was restored by both USCs and USC-exos in NOA mice 36 days after busulfan treatment as confirmed by immunofluorescence staining and hematoxylin and eosin staining. Moreover, spermatogenic genes (Pou5f1, Prm1, SYCP3, and DAZL) and the spermatogenic protein UCHL1 were significantly increased in both the USCs 36 and USC-exos36 groups compared with the PBS group, as demonstrated using quantitative real-time polymerase chain reaction and western blot analysis. However, the transplantation of USCs or USC-exos at day 3 after busulfan treatment did not improve spermatogenesis in NOA mice. Our study demonstrated that USCs could facilitate endogenous spermatogenesis restoration of busulfan-induced NOA mice through paracrine exosomes but could not protect the mouse testicles at the early stage of destruction caused by busulfan. This study provides a novel insight into the treatment of NOA.
Collapse
Affiliation(s)
- Cuncan Deng
- Reproductive Medicine Center, The Sixth Affiliate Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yun Xie
- Department of Andrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, China
| | - Chi Zhang
- Department of Andrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Bin Ouyang
- Department of Andrology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Haicheng Chen
- Department of Andrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Linyan Lv
- Reproductive Medicine Center, The Sixth Affiliate Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jiahui Yao
- Reproductive Medicine Center, The Sixth Affiliate Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaoyan Liang
- Reproductive Medicine Center, The Sixth Affiliate Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yuanyuan Zhang
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, North Carolina
| | - Xiangzhou Sun
- Department of Andrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Chunhua Deng
- Department of Andrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Guihua Liu
- Reproductive Medicine Center, The Sixth Affiliate Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
232
|
Patel DB, Luthers CR, Lerman MJ, Fisher JP, Jay SM. Enhanced extracellular vesicle production and ethanol-mediated vascularization bioactivity via a 3D-printed scaffold-perfusion bioreactor system. Acta Biomater 2019; 95:236-244. [PMID: 30471476 DOI: 10.1016/j.actbio.2018.11.024] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 11/12/2018] [Accepted: 11/15/2018] [Indexed: 01/08/2023]
Abstract
Extracellular vesicles (EVs) have garnered significant interest in the biotechnology field due to their intrinsic therapeutic properties as well as their ability to serve as vehicles for bioactive cargo. However, the lack of an established biomanufacturing platform and limited potency of EVs in vivo remain critical bottlenecks for clinical translation. In this study, we utilized a 3D-printed scaffold-perfusion bioreactor system to assess the response of dynamic culture on extracellular vesicle production from endothelial cells (ECs). We also investigated whether ethanol conditioning, which was previously shown to enhance vascularization bioactivity of EC-derived EVs produced in standard 2D culture conditions, could be employed successfully for the same purpose in a 3D production system. Our results indicate that dynamic culture in a perfusion bioreactor significantly enhances EV production from human ECs. Moreover, the use of ethanol conditioning in conjunction with dynamic culture induces pro-vascularization bioactivity of EC-derived EVs that is correlated with increased EV levels of pro-angiogenic lncRNAs HOTAIR and MALAT1. Thus, this study represents one of the first reports of rationally-designed EV potency enhancement that is conserved between static 2D and dynamic 3D EV production systems, increasing the potential for scalable biomanufacturing of therapeutic EC-derived EVs for a variety of applications. STATEMENT OF SIGNIFICANCE: Extracellular vesicles (EVs) have substantial therapeutic potential in a variety of applications. However, translation of EV-based therapies may be hindered by biomanufacturing challenges. EV production to date has predominantly involved the use of tissue culture flasks. Here, we report, for the first time, the use of a tubular perfusion bioreactor system with an integrated 3D-printed biomaterial scaffold for EV production from human endothelial cells. This system increases EV yield by over 100-fold compared to conventional tissue culture systems. Further, we show that an ethanol-conditioning approach that our group previously developed in 2D culture for enhancing EV potency is compatible with this new system. Thus, potency enhancement of EVs for vascularization applications is possible even with significantly increased production rate.
Collapse
Affiliation(s)
- Divya B Patel
- Fischell Department of Bioengineering, University of Maryland 3102 A. James Clark Hall, College Park, MD 20742, United States
| | - Christopher R Luthers
- Fischell Department of Bioengineering, University of Maryland 3102 A. James Clark Hall, College Park, MD 20742, United States
| | - Max J Lerman
- Department of Materials Science and Engineering, University of Maryland 4418 Stadium Drive, College Park, MD 20742, United States; Center for Engineering Complex Tissues, University of Maryland 3102 A. James Clark Hall, College Park, MD 20742, United States; Surface and Trace Chemical Analysis Group, Materials Measurement Science Division National Institute of Standards and Technology 100 Bureau Drive, Mailstop 6431, Gaithersburg, MD 20899, United States
| | - John P Fisher
- Fischell Department of Bioengineering, University of Maryland 3102 A. James Clark Hall, College Park, MD 20742, United States; Center for Engineering Complex Tissues, University of Maryland 3102 A. James Clark Hall, College Park, MD 20742, United States
| | - Steven M Jay
- Fischell Department of Bioengineering, University of Maryland 3102 A. James Clark Hall, College Park, MD 20742, United States; Greenbaum Comprehensive Cancer Center, University of Maryland - Baltimore, Baltimore, MD 21201, United States; Program in Molecular and Cell Biology, University of Maryland College Park, MD 20742, United States.
| |
Collapse
|
233
|
Differential Effects of Extracellular Vesicles of Lineage-Specific Human Pluripotent Stem Cells on the Cellular Behaviors of Isogenic Cortical Spheroids. Cells 2019; 8:cells8090993. [PMID: 31466320 PMCID: PMC6770916 DOI: 10.3390/cells8090993] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 08/23/2019] [Accepted: 08/27/2019] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EVs) contribute to a variety of signaling processes and the overall physiological and pathological states of stem cells and tissues. Human induced pluripotent stem cells (hiPSCs) have unique characteristics that can mimic embryonic tissue development. There is growing interest in the use of EVs derived from hiPSCs as therapeutics, biomarkers, and drug delivery vehicles. However, little is known about the characteristics of EVs secreted by hiPSCs and paracrine signaling during tissue morphogenesis and lineage specification. Methods: In this study, the physical and biological properties of EVs isolated from hiPSC-derived neural progenitors (ectoderm), hiPSC-derived cardiac cells (mesoderm), and the undifferentiated hiPSCs (healthy iPSK3 and Alzheimer’s-associated SY-UBH lines) were analyzed. Results: Nanoparticle tracking analysis and electron microscopy results indicate that hiPSC-derived EVs have an average size of 100–250 nm. Immunoblot analyses confirmed the enrichment of exosomal markers Alix, CD63, TSG101, and Hsc70 in the purified EV preparations. MicroRNAs including miR-133, miR-155, miR-221, and miR-34a were differently expressed in the EVs isolated from distinct hiPSC lineages. Treatment of cortical spheroids with hiPSC-EVs in vitro resulted in enhanced cell proliferation (indicated by BrdU+ cells) and axonal growth (indicated by β-tubulin III staining). Furthermore, hiPSC-derived EVs exhibited neural protective abilities in Aβ42 oligomer-treated cultures, enhancing cell viability and reducing oxidative stress. Our results demonstrate that the paracrine signaling provided by tissue context-dependent EVs derived from hiPSCs elicit distinct responses to impact the physiological state of cortical spheroids. Overall, this study advances our understanding of cell‒cell communication in the stem cell microenvironment and provides possible therapeutic options for treating neural degeneration.
Collapse
|
234
|
Strategies for the use of Extracellular Vesicles for the Delivery of Therapeutics. J Neuroimmune Pharmacol 2019; 15:422-442. [PMID: 31456107 DOI: 10.1007/s11481-019-09873-y] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 08/13/2019] [Indexed: 12/17/2022]
Abstract
Extracellular vesicles (EVs) are nanosized, membrane-bound vesicles released from eukaryotic and prokaryotic cells that can transport cargo containing DNA, RNA, lipids and proteins, between cells as a means of intercellular communication. Although EVs were initially considered to be cellular debris deprived of any essential biological functions, emerging literature highlights the critical roles of EVs in the context of intercellular signaling, maintenance of tissue homeostasis, modulation of immune responses, inflammation, cancer progression, angiogenesis, and coagulation under both physiological and pathological states. Based on the ability of EVs to shuttle proteins, lipids, carbohydrates, mRNAs, long non-coding RNAs (lncRNAs), microRNAs, chromosomal DNA, and mitochondrial DNA into target cells, the presence and content of EVs in biofluids have been exploited for biomarker research in the context of diagnosis, prognosis and treatment strategies. Additionally, owing to the characteristics of EVs such as stability in circulation, biocompatibility as well as low immunogenicity and toxicity, these vesicles have become attractive systems for the delivery of therapeutics. More recently, EVs are increasingly being exploited as conduits for delivery of therapeutics for anticancer strategies, immunomodulation, targeted drug delivery, tissue regeneration, and vaccination. In this review, we highlight and discuss the multiple strategies that are employed for the use of EVs as delivery vehicles for therapeutic agents, including the potential advantages and challenges involved. Graphical abstract.
Collapse
|
235
|
Monfared H, Jahangard Y, Nikkhah M, Mirnajafi-Zadeh J, Mowla SJ. Potential Therapeutic Effects of Exosomes Packed With a miR-21-Sponge Construct in a Rat Model of Glioblastoma. Front Oncol 2019; 9:782. [PMID: 31482067 PMCID: PMC6710330 DOI: 10.3389/fonc.2019.00782] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 08/01/2019] [Indexed: 01/01/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a grade 4 and the most aggressive form of glioma, with a poor response to current treatments. The expression of microRNAs (miRNAs) is widely dysregulated in various cancers, including GBM. One of the overexpressed miRNAs in GBM is miR-21 which promotes proliferation, invasion and metastatic behaviors of tumor cells. With a size of 30-100 nm, the extracellular vesicles "exosomes" have emerged as a novel and powerful drug delivering systems. Recently, exosomal transfer of miRNAs or anti-miRNAs to tumor cells has introduced a new approach for therapeutic application of miRNAs to combat cancer. Here, we have tried to down-regulate miR-21 expression in glioma cell lines, U87-MG, and C6, by using engineered exosomes, packed with a miR-21-sponge construct. Our data revealed that the engineered exosomes have the potential to suppress miR-21 and consequently to upregulate miR-21 target genes, PDCD4 and RECK. Interestingly, in cells treated with miR-21-sponge exosomes we observed a decline in proliferation and also an elevation in apoptotic rates. Finally, in a rat model of glioblastoma, administrating exosomes loaded with a miR-21-sponge construct leads to a significant reduction in the volume of the tumors. In brief, our findings suggest a new therapeutic strategy to use engineered exosomes to deliver a miR-21-sponge construct to GBM cells, in order to block its malignant behavior.
Collapse
Affiliation(s)
- Hamideh Monfared
- Department of Molecular Genetics, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran
| | - Yavar Jahangard
- Department of Molecular Genetics, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran
| | - Maryam Nikkhah
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Javad Mirnajafi-Zadeh
- Department of Physiology, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran
| | - Seyed Javad Mowla
- Department of Molecular Genetics, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
236
|
Mentkowski KI, Lang JK. Exosomes Engineered to Express a Cardiomyocyte Binding Peptide Demonstrate Improved Cardiac Retention in Vivo. Sci Rep 2019; 9:10041. [PMID: 31296886 PMCID: PMC6624248 DOI: 10.1038/s41598-019-46407-1] [Citation(s) in RCA: 168] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 06/28/2019] [Indexed: 12/19/2022] Open
Abstract
Injury to the heart results in cardiomyocyte cell death and can lead to pathological remodeling of remaining cells, contributing to heart failure. Despite the therapeutic potential of new drugs and small molecules, there remains a gap in the ability to efficiently deliver cardioprotective agents in a cell specific manner while minimizing nonspecific delivery to other organs. Exosomes derived from cardiosphere-derived cells (CDCs) have been shown to stimulate angiogenesis, induce endogenous cardiomyocyte proliferation and modulate cardiomyocyte apoptosis and hypertrophy. While innately cardioprotective at high doses, unmodified CDC-exosomes demonstrate limited cardiac tropism. To generate an efficient exosomal delivery system that can target cardiomyocytes, we engineered CDCs to express Lamp2b, an exosomal membrane protein, fused to a cardiomyocyte specific peptide (CMP), WLSEAGPVVTVRALRGTGSW. Exosomes isolated from engineered CDCs expressed CMP on their surface and retained their native physical properties. Targeted exosomes resulted in increased uptake by cardiomyocytes, decreased cardiomyocyte apoptosis, and higher cardiac retention following intramyocardial injection when compared with non-targeted exosomes. Importantly, we established a novel targeting system to improve exosomal uptake by cardiomyocytes and laid the foundation for cell-specific exosomal delivery of drug and gene therapies to improve the functional capacity of the heart following both ischemic and non-ischemic injury.
Collapse
Affiliation(s)
- Kyle I Mentkowski
- Department of Medicine, Division of Cardiology, Jacobs School of Medicine and Biomedical Sciences, Buffalo, N.Y., 14203, United States of America.,Department of Biomedical Engineering, University at Buffalo, Buffalo, N.Y., 14260, United States of America
| | - Jennifer K Lang
- Department of Medicine, Division of Cardiology, Jacobs School of Medicine and Biomedical Sciences, Buffalo, N.Y., 14203, United States of America. .,Department of Biomedical Engineering, University at Buffalo, Buffalo, N.Y., 14260, United States of America. .,VA WNY Healthcare System, Buffalo, N.Y., 14215, United States of America.
| |
Collapse
|
237
|
Paganini C, Capasso Palmiero U, Pocsfalvi G, Touzet N, Bongiovanni A, Arosio P. Scalable Production and Isolation of Extracellular Vesicles: Available Sources and Lessons from Current Industrial Bioprocesses. Biotechnol J 2019; 14:e1800528. [PMID: 31140717 DOI: 10.1002/biot.201800528] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 05/20/2019] [Indexed: 12/17/2022]
Abstract
Potential applications of extracellular vesicles (EVs) are attracting increasing interest in the fields of medicine, cosmetics, and nutrition. However, the manufacturing of EVs is currently characterized by low yields. This limitation severely hampers progress in research at the laboratory and clinical scales, as well as the realization of successful and cost-effective EV-based products. Moreover, the high level of heterogeneity of EVs further complicates reproducible manufacturing on a large scale. In this review, possible directions toward the scalable production of EVs are discussed. In particular, two strategies are considered: i) the optimization of upstream unit operations and ii) the exploitation of well-established and mature technologies already in use in other industrial bioprocesses.
Collapse
Affiliation(s)
- Carolina Paganini
- Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir Prelog Weg 1, 8093, Zurich, Switzerland
| | - Umberto Capasso Palmiero
- Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir Prelog Weg 1, 8093, Zurich, Switzerland
| | - Gabriella Pocsfalvi
- Institute of Biosciences and Bioresources, National Research Council of Italy, Naples, 80131, Italy
| | - Nicolas Touzet
- Centre for Environmental Research Innovation and Sustainability, Institute of Technology Sligo, Sligo, F91 YW50, Ireland
| | - Antonella Bongiovanni
- Institute for Biomedical Research and Innovation, National Research Council of Italy, Palermo, 90146, Italy
| | - Paolo Arosio
- Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir Prelog Weg 1, 8093, Zurich, Switzerland
| |
Collapse
|
238
|
Gudbergsson JM, Jønsson K, Simonsen JB, Johnsen KB. Systematic review of targeted extracellular vesicles for drug delivery – Considerations on methodological and biological heterogeneity. J Control Release 2019; 306:108-120. [DOI: 10.1016/j.jconrel.2019.06.006] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/03/2019] [Accepted: 06/03/2019] [Indexed: 12/20/2022]
|
239
|
Extracellular Microvesicles as New Industrial Therapeutic Frontiers. Trends Biotechnol 2019; 37:707-729. [DOI: 10.1016/j.tibtech.2018.11.012] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 11/23/2018] [Accepted: 11/30/2018] [Indexed: 12/18/2022]
|
240
|
Monguió-Tortajada M, Gálvez-Montón C, Bayes-Genis A, Roura S, Borràs FE. Extracellular vesicle isolation methods: rising impact of size-exclusion chromatography. Cell Mol Life Sci 2019; 76:2369-2382. [PMID: 30891621 PMCID: PMC11105396 DOI: 10.1007/s00018-019-03071-y] [Citation(s) in RCA: 264] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 03/08/2019] [Accepted: 03/13/2019] [Indexed: 02/07/2023]
Abstract
Extracellular vesicles (EVs) include a variety of nanosized vesicles released to the extracellular microenvironment by the vast majority of cells transferring bioactive lipids, proteins, mRNA, miRNA or non-coding RNA, as means of intercellular communication. Remarkably, among other fields of research, their use has become promising for immunomodulation, tissue repair and as source for novel disease-specific molecular signatures or biomarkers. However, a major challenge is to define accurate, reliable and easily implemented techniques for EV isolation due to their nanoscale size and high heterogeneity. In this context, differential ultracentrifugation (dUC) has been the most widely used laboratory methodology, but alternative procedures have emerged to allow purer EV preparations with easy implementation. Here, we present and discuss the most used of the different EV isolation methods, focusing on the increasing impact of size exclusion chromatography (SEC) on the resulting EV preparations from in vitro cultured cells-conditioned medium and biological fluids. Comparatively, low protein content and cryo-electron microscopy analysis show that SEC removes most of the overabundant soluble plasma proteins, which are not discarded using dUC or precipitating agents, while being more user friendly and less time-consuming than gradient-based EV isolation. Also, SEC highly maintains the major EVs' characteristics, including vesicular structure and content, which guarantee forthcoming applications. In sum, together with scaling-up possibilities to increase EV recovery and manufacturing following high-quality standards, SEC could be easily adapted to most laboratories to assist EV-associated biomarker discovery and to deliver innovative cell-free immunomodulatory and pro-regenerative therapies.
Collapse
Affiliation(s)
- Marta Monguió-Tortajada
- REMAR-IVECAT Group, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Badalona, Spain
- ICREC Research Program, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Badalona, Spain
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Carolina Gálvez-Montón
- ICREC Research Program, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Badalona, Spain
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Antoni Bayes-Genis
- ICREC Research Program, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Badalona, Spain
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
- Cardiology Service, Germans Trias i Pujol University Hospital, Badalona, Spain
- Department of Medicine, UAB, Barcelona, Spain
| | - Santiago Roura
- ICREC Research Program, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Badalona, Spain.
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain.
| | - Francesc E Borràs
- REMAR-IVECAT Group, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Badalona, Spain.
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain.
- Nephrology Service, Germans Trias i Pujol University Hospital, Badalona, Spain.
| |
Collapse
|
241
|
Ouyang B, Xie Y, Zhang C, Deng C, Lv L, Yao J, Zhang Y, Liu G, Deng J, Deng C. Extracellular Vesicles From Human Urine-Derived Stem Cells Ameliorate Erectile Dysfunction in a Diabetic Rat Model by Delivering Proangiogenic MicroRNA. Sex Med 2019; 7:241-250. [PMID: 30910509 PMCID: PMC6522949 DOI: 10.1016/j.esxm.2019.02.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 01/18/2019] [Accepted: 02/11/2019] [Indexed: 12/18/2022] Open
Abstract
INTRODUCTION Stem cell therapies represent a promising new frontier for the treatment of refractory diabetic erectile dysfunction (DED). The use of stem cell-derived extracellular vesicles (EVs) is a novel strategy for cell-free stem cell therapy. We have reported that urine-derived stem cells (USCs) can improve DED; however, the therapeutic effects of EVs secreted by USCs (USC-EVs) remain unknown. AIM To determine the therapeutic effects of USC-EVs on DED in a rat model. METHODS USC-EVs were isolated from conditioned medium by ultracentrifugation. DED was induced in male Sprague-Dawley rats via an intraperitoneal injection of streptozotocin. Sixteen DED rats were divided into phosphate-buffered saline (PBS) and USC-EV groups. Eight normal rats served as the normal control group. PBS or USC-EVs were transplanted into the corpora cavernosa in the corresponding groups. MAIN OUTCOME MEASURE Intracavernosal pressure (ICP), mean arterial pressure (MAP), expression of endothelial markers (CD31), endothelial nitric oxide synthase (eNOS), phospho-eNOS, and neural nitric oxide synthase (nNOS) were assessed in each group. Masson's trichrome staining was used to determine the collagen deposition and ratio of smooth muscle cells to collagen. The microRNA (miRNA) cargo of USC-EVs was characterized by high-throughput RNA sequencing. RESULTS Recovery of erectile function was observed in the USC-EV group, as represented by improved ICP and ICP/MAP ratio. CD31, eNOS, phospho-eNOS, and nNOS expression in the penis was significantly improved in the USC-EV group. In addition, the ratio of smooth muscle to collagen was significantly increased in the USC-EV group. RNA sequencing revealed that USC-EVs were enriched for distinct classes of miRNA (miR-21-5p, let-7 family, miR-10 family, miR-30 family, and miR-148a-3p) that promote angiogenesis. CONCLUSION USC-EV transplantation can ameliorate DED in rats. Its mechanism may involve the delivery of proangiogenic miRNA. Ouyang B, Xie Y, Zhang C, et al. Extracellular Vesicles From Human Urine-Derived Stem Cells Ameliorate Erectile Dysfunction in a Diabetic Rat Model by Delivering Proangiogenic MicroRNA. Sex Med 2019;7:241-250.
Collapse
Affiliation(s)
- Bin Ouyang
- Department of Andrology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China; Department of Andrology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yun Xie
- Department of Andrology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Chi Zhang
- Department of Andrology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Cuncan Deng
- Reproductive Medicine Research Center, the Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Linyan Lv
- Reproductive Medicine Research Center, the Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jiahui Yao
- Department of Andrology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yuanyuan Zhang
- Department of Andrology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China; Department of Andrology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China; Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Guihua Liu
- Reproductive Medicine Research Center, the Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Junhong Deng
- Department of Andrology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China; Department of Andrology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Chunhua Deng
- Department of Andrology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
242
|
Veerman RE, Güçlüler Akpinar G, Eldh M, Gabrielsson S. Immune Cell-Derived Extracellular Vesicles – Functions and Therapeutic Applications. Trends Mol Med 2019; 25:382-394. [DOI: 10.1016/j.molmed.2019.02.003] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/05/2019] [Accepted: 02/08/2019] [Indexed: 12/15/2022]
|
243
|
Palviainen M, Saari H, Kärkkäinen O, Pekkinen J, Auriola S, Yliperttula M, Puhka M, Hanhineva K, Siljander PRM. Metabolic signature of extracellular vesicles depends on the cell culture conditions. J Extracell Vesicles 2019; 8:1596669. [PMID: 31007875 PMCID: PMC6461113 DOI: 10.1080/20013078.2019.1596669] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 03/07/2019] [Accepted: 03/13/2019] [Indexed: 12/21/2022] Open
Abstract
One of the greatest bottlenecks in extracellular vesicle (EV) research is the production of sufficient material in a consistent and effective way using in vitro cell models. Although the production of EVs in bioreactors maximizes EV yield in comparison to conventional cell cultures, the impact of their cell growth conditions on EVs has not yet been established. In this study, we grew two prostate cancer cell lines, PC-3 and VCaP, in conventional cell culture dishes and in two-chamber bioreactors to elucidate how the growth environment affects the EV characteristics. Specifically, we wanted to investigate the growth condition-dependent differences by non-targeted metabolite profiling using liquid chromatography-mass spectrometry (LC-MS) analysis. EVs were also characterized by their morphology, size distribution, and EV protein marker expression, and the EV yields were quantified by NTA. The use of bioreactor increased the EV yield >100 times compared to the conventional cell culture system. Regarding morphology, size distribution and surface markers, only minor differences were observed between the bioreactor-derived EVs (BR-EVs) and the EVs obtained from cells grown in conventional cell cultures (C-EVs). In contrast, metabolomic analysis revealed statistically significant differences in both polar and non-polar metabolites when the BR-EVs were compared to the C-EVs. The results show that the growth conditions markedly affected the EV metabolite profiles and that metabolomics was a sensitive tool to study molecular differences of EVs. We conclude that the cell culture conditions of EV production should be standardized and carefully detailed in publications and care should be taken when EVs from different production platforms are compared with each other for systemic effects.
Collapse
Affiliation(s)
- Mari Palviainen
- EV-group, Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
- EV-core, University of Helsinki, Helsinki, Finland
| | - Heikki Saari
- Drug Research Program, Faculty of Pharmacy, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | - Olli Kärkkäinen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- LC-MS Metabolomics Center, University of Eastern Finland, Kuopio, Finland
| | - Jenna Pekkinen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- LC-MS Metabolomics Center, University of Eastern Finland, Kuopio, Finland
| | - Seppo Auriola
- LC-MS Metabolomics Center, University of Eastern Finland, Kuopio, Finland
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Marjo Yliperttula
- Drug Research Program, Faculty of Pharmacy, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | - Maija Puhka
- EV-core, University of Helsinki, Helsinki, Finland
- Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland
| | - Kati Hanhineva
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- LC-MS Metabolomics Center, University of Eastern Finland, Kuopio, Finland
| | - Pia R.-M. Siljander
- EV-group, Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
- EV-core, University of Helsinki, Helsinki, Finland
- Drug Research Program, Faculty of Pharmacy, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| |
Collapse
|
244
|
Gurunathan S, Kang MH, Jeyaraj M, Qasim M, Kim JH. Review of the Isolation, Characterization, Biological Function, and Multifarious Therapeutic Approaches of Exosomes. Cells 2019; 8:307. [PMID: 30987213 PMCID: PMC6523673 DOI: 10.3390/cells8040307] [Citation(s) in RCA: 756] [Impact Index Per Article: 126.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 03/22/2019] [Accepted: 04/01/2019] [Indexed: 12/18/2022] Open
Abstract
Exosomes are extracellular vesicles that contain a specific composition of proteins, lipids, RNA, and DNA. They are derived from endocytic membranes and can transfer signals to recipient cells, thus mediating a novel mechanism of cell-to-cell communication. They are also thought to be involved in cellular waste disposal. Exosomes play significant roles in various biological functions, including the transfer of biomolecules such as RNA, proteins, enzymes, and lipids and the regulation of numerous physiological and pathological processes in various diseases. Because of these properties, they are considered to be promising biomarkers for the diagnosis and prognosis of various diseases and may contribute to the development of minimally invasive diagnostics and next generation therapies. The biocompatible nature of exosomes could enhance the stability and efficacy of imaging probes and therapeutics. Due to their potential use in clinical applications, exosomes have attracted much research attention on their roles in health and disease. To explore the use of exosomes in the biomedical arena, it is essential that the basic molecular mechanisms behind the transport and function of these vesicles are well-understood. Herein, we discuss the history, biogenesis, release, isolation, characterization, and biological functions of exosomes, as well as the factors influencing their biogenesis and their technical and biological challenges. We conclude this review with a discussion on the future perspectives of exosomes.
Collapse
Affiliation(s)
- Sangiliyandi Gurunathan
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, 1 Hwayang-Dong, Gwangin-gu, Seoul 05029, Korea.
| | - Min-Hee Kang
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, 1 Hwayang-Dong, Gwangin-gu, Seoul 05029, Korea.
| | - Muniyandi Jeyaraj
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, 1 Hwayang-Dong, Gwangin-gu, Seoul 05029, Korea.
| | - Muhammad Qasim
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, 1 Hwayang-Dong, Gwangin-gu, Seoul 05029, Korea.
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, 1 Hwayang-Dong, Gwangin-gu, Seoul 05029, Korea.
| |
Collapse
|
245
|
Abudoureyimu M, Zhou H, Zhi Y, Wang T, Feng B, Wang R, Chu X. Recent progress in the emerging role of exosome in hepatocellular carcinoma. Cell Prolif 2019; 52:e12541. [PMID: 30397975 PMCID: PMC6496614 DOI: 10.1111/cpr.12541] [Citation(s) in RCA: 33] [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: 05/18/2018] [Revised: 08/14/2018] [Accepted: 08/16/2018] [Indexed: 12/22/2022] Open
Abstract
Exosomes are small membrane vesicles 50-150 nm in diameter released by a variety of cells, which contain miRNAs, mRNAs and proteins with the potential to regulate signalling pathways in recipient cells. Exosomes deliver nucleic acids and proteins to participate in orchestrating cell-cell communication and microenvironment modulation. In this review, we summarize recent progress in our understanding of the role of exosomes in hepatocellular carcinoma (HCC). This review focuses on recent studies on HCC exosomes, considering biogenesis, cargo and their effects on the development and progression of HCC, including chemoresistance, epithelial-mesenchymal transition, angiogenesis, metastasis and immune response. Finally, we discuss the clinical application of exosomes as a therapeutic agent for HCC.
Collapse
Affiliation(s)
- Mubalake Abudoureyimu
- Department of Medical Oncology, School of Medicine, Jinling HospitalNanjing UniversityNanjingChina
| | - Hao Zhou
- Department of Medical Oncology, Jinling HospitalNanjing Medical UniversityNanjingChina
| | - Yingru Zhi
- Department of Medical Oncology, School of Medicine, Jinling HospitalNanjing UniversityNanjingChina
| | - Ting Wang
- Department of Medical OncologyJinling HospitalNanjingChina
| | - Bing Feng
- Department of Medical Oncology, School of Medicine, Jinling HospitalNanjing UniversityNanjingChina
| | - Rui Wang
- Department of Medical Oncology, School of Medicine, Jinling HospitalNanjing UniversityNanjingChina
| | - Xiaoyuan Chu
- Department of Medical Oncology, School of Medicine, Jinling HospitalNanjing UniversityNanjingChina
| |
Collapse
|
246
|
Faruqu FN, Wang JTW, Xu L, McNickle L, Chong EMY, Walters A, Gurney M, Clayton A, Smyth LA, Hider R, Sosabowski J, Al-Jamal KT. Membrane Radiolabelling of Exosomes for Comparative Biodistribution Analysis in Immunocompetent and Immunodeficient Mice - A Novel and Universal Approach. Am J Cancer Res 2019; 9:1666-1682. [PMID: 31037130 PMCID: PMC6485196 DOI: 10.7150/thno.27891] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 11/08/2018] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles, in particular exosomes, have recently gained interest as novel drug delivery vectors due to their biological origin and inherent intercellular biomolecule delivery capability. An in-depth knowledge of their in vivo biodistribution is therefore essential. This work aimed to develop a novel, reliable and universal method to radiolabel exosomes to study their in vivo biodistribution. Methods: Melanoma (B16F10) cells were cultured in bioreactor flasks to increase exosome yield. B16F10-derived exosomes (ExoB16) were isolated using ultracentrifugation onto a single sucrose cushion, and were characterised for size, yield, purity, exosomal markers and morphology using nanoparticle tracking analysis (NTA), protein measurements, flow cytometry and electron microscopy. ExoB16 were radiolabelled using 2 different approaches - intraluminal labelling (entrapment of 111Indium via tropolone shuttling); and membrane labelling (chelation of 111Indium via covalently attached bifunctional chelator DTPA-anhydride). Labelling efficiency and stability was assessed using gel filtration and thin layer chromatography. Melanoma-bearing immunocompetent (C57BL/6) and immunodeficient (NSG) mice were injected intravenously with radiolabelled ExoB16 (1x1011 particles/mouse) followed by metabolic cages study, whole body SPECT-CT imaging and ex vivo gamma counting at 1, 4 and 24 h post-injection. Results: Membrane-labelled ExoB16 showed superior radiolabelling efficiency and radiochemical stability (19.2 ± 4.53 % and 80.4 ± 1.6 % respectively) compared to the intraluminal-labelled exosomes (4.73 ± 0.39 % and 14.21 ± 2.76 % respectively). Using the membrane-labelling approach, the in vivo biodistribution of ExoB16 in melanoma-bearing C57Bl/6 mice was carried out, and was found to accumulate primarily in the liver and spleen (~56% and ~38% ID/gT respectively), followed by the kidneys (~3% ID/gT). ExoB16 showed minimal tumour i.e. self-tissue accumulation (~0.7% ID/gT). The membrane-labelling approach was also used to study ExoB16 biodistribution in melanoma-bearing immunocompromised (NSG) mice, to compare with that in the immunocompetent C57Bl/6 mice. Similar biodistribution profile was observed in both C57BL/6 and NSG mice, where prominent accumulation was seen in liver and spleen, apart from the significantly lower tumour accumulation observed in the NSG mice (~0.3% ID/gT). Conclusion: Membrane radiolabelling of exosomes is a reliable approach that allows for accurate live imaging and quantitative biodistribution studies to be performed on potentially all exosome types without engineering parent cells.
Collapse
|
247
|
Accelerating Biologics Manufacturing by Modeling or: Is Approval under the QbD and PAT Approaches Demanded by Authorities Acceptable Without a Digital-Twin? Processes (Basel) 2019. [DOI: 10.3390/pr7020094] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Innovative biologics, including cell therapeutics, virus-like particles, exosomes,recombinant proteins, and peptides, seem likely to substitute monoclonal antibodies as the maintherapeutic entities in manufacturing over the next decades. This molecular variety causes agrowing need for a general change of methods as well as mindset in the process development stage,as there are no platform processes available such as those for monoclonal antibodies. Moreover,market competitiveness demands hyper-intensified processes, including accelerated decisionstoward batch or continuous operation of dedicated modular plant concepts. This indicates gaps inprocess comprehension, when operation windows need to be run at the edges of optimization. Inthis editorial, the authors review and assess potential methods and begin discussing possiblesolutions throughout the workflow, from process development through piloting to manufacturingoperation from their point of view and experience. Especially, the state-of-the-art for modeling inred biotechnology is assessed, clarifying differences and applications of statistical, rigorousphysical-chemical based models as well as cost modeling. “Digital-twins” are described and effortsvs. benefits for new applications exemplified, including the regulation-demanded QbD (quality bydesign) and PAT (process analytical technology) approaches towards digitalization or industry 4.0based on advanced process control strategies. Finally, an analysis of the obstacles and possiblesolutions for any successful and efficient industrialization of innovative methods from processdevelopment, through piloting to manufacturing, results in some recommendations. A centralquestion therefore requires attention: Considering that QbD and PAT have been required byauthorities since 2004, can any biologic manufacturing process be approved by the regulatoryagencies without being modeled by a “digital-twin” as part of the filing documentation?
Collapse
|
248
|
Ng KS, Smith JA, McAteer MP, Mead BE, Ware J, Jackson FO, Carter A, Ferreira L, Bure K, Rowley JA, Reeve B, Brindley DA, Karp JM. Bioprocess decision support tool for scalable manufacture of extracellular vesicles. Biotechnol Bioeng 2019; 116:307-319. [PMID: 30063243 PMCID: PMC6322973 DOI: 10.1002/bit.26809] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 07/22/2018] [Accepted: 07/26/2018] [Indexed: 12/28/2022]
Abstract
Newly recognized as natural nanocarriers that deliver biological information between cells, extracellular vesicles (EVs), including exosomes and microvesicles, provide unprecedented therapeutic opportunities. Large-scale and cost-effective manufacturing is imperative for EV products to meet commercial and clinical demands; successful translation requires careful decisions that minimize financial and technological risks. Here, we develop a decision support tool (DST) that computes the most cost-effective technologies for manufacturing EVs at different scales, by examining the costs of goods associated with using published protocols. The DST identifies costs of labor and consumables during EV harvest as key cost drivers, substantiating a need for larger-scale, higher-throughput, and automated technologies for harvesting EVs. Importantly, we highlight a lack of appropriate technologies for meeting clinical demands, and propose a potentially cost-effective solution. This DST can facilitate decision-making very early on in development and be used to predict, and better manage, the risk of process changes when commercializing EV products.
Collapse
Affiliation(s)
- Kelvin S. Ng
- Harvard‐MIT Division of Health Sciences and TechnologyCambridgeMassachusetts
- Division of Engineering in Medicine, Department of MedicineBrigham & Women’s Hospital, Harvard Medical SchoolBostonMA
- Harvard Stem Cell InstituteCambridgeMassachusetts
- RoosterBioFrederickMaryland
| | - James A. Smith
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal SciencesUniversity of OxfordOxfordUK
- The Oxford‐UCL Centre for the Advancement of Sustainable Medical Innovation, University of OxfordOxfordUK
| | - Matthew P. McAteer
- Department of NeurologyMassachusetts General Hospital, Harvard Medical SchoolCharlestownMassachusetts
| | - Benjamin E. Mead
- Harvard‐MIT Division of Health Sciences and TechnologyCambridgeMassachusetts
- Division of Engineering in Medicine, Department of MedicineBrigham & Women’s Hospital, Harvard Medical SchoolBostonMA
- Harvard Stem Cell InstituteCambridgeMassachusetts
- Broad Institute of Harvard and MITCambridgeMassachusetts
- Koch Institute for Integrative Cancer Research, MITCambridgeMassachusetts
| | - Jamie Ware
- The Oxford‐UCL Centre for the Advancement of Sustainable Medical Innovation, University of OxfordOxfordUK
| | - Felix O. Jackson
- The Oxford‐UCL Centre for the Advancement of Sustainable Medical Innovation, University of OxfordOxfordUK
| | - Alison Carter
- Department of PaediatricsUniversity of OxfordOxfordUK
| | - Lino Ferreira
- University of Coimbra, Center for Neuroscience and Cell BiologyPortugal
| | - Kim Bure
- The Oxford‐UCL Centre for the Advancement of Sustainable Medical Innovation, University of OxfordOxfordUK
| | | | - Brock Reeve
- Harvard Stem Cell InstituteCambridgeMassachusetts
| | - David A. Brindley
- Harvard Stem Cell InstituteCambridgeMassachusetts
- The Oxford‐UCL Centre for the Advancement of Sustainable Medical Innovation, University of OxfordOxfordUK
- Department of PaediatricsUniversity of OxfordOxfordUK
- Centre for Behavioural Medicine, UCL School of Pharmacy, University College LondonLondonUK
- UCSF‐Stanford Center of Excellence in Regulatory Science and InnovationSan FranciscoCalifornia
| | - Jeffrey M. Karp
- Harvard‐MIT Division of Health Sciences and TechnologyCambridgeMassachusetts
- Division of Engineering in Medicine, Department of MedicineBrigham & Women’s Hospital, Harvard Medical SchoolBostonMA
- Harvard Stem Cell InstituteCambridgeMassachusetts
- Broad Institute of Harvard and MITCambridgeMassachusetts
| |
Collapse
|
249
|
Monguió-Tortajada M, Morón-Font M, Gámez-Valero A, Carreras-Planella L, Borràs FE, Franquesa M. Extracellular-Vesicle Isolation from Different Biological Fluids by Size-Exclusion Chromatography. ACTA ACUST UNITED AC 2019; 49:e82. [PMID: 30698351 DOI: 10.1002/cpsc.82] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
This unit describes how to isolate extracellular vesicles (EVs) from different biological fluids using size-exclusion chromatography (SEC) and how to prepare your starting sample and the EV product for downstream applications. EVs are membrane nanovesicles with specific content that reflects the phenotype and functions of the cell of origin, including protected proteins, lipids, metabolites, and nucleic acids. EVs are thus an excellent resource for noninvasive biomarker discovery in a number of pathological situations and are a promising nanotherapeutic tool to overcome the disadvantages associated with cellular therapy. However, there are still no standardized methods to isolate pure EV preparations, as many approaches do not guarantee proper EV purification, free of contaminating non-EV molecules. Currently, SEC is one of the most promising approaches to purify EVs from any biological fluid, as it avoids co-isolation of contaminants and is user friendly and scalable. © 2019 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Marta Monguió-Tortajada
- REMAR-IVECAT Group, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Badalona, Spain.,Department of Cell Biology, Physiology and Immunology, Autonomous University of Barcelona, Bellaterra, Cerdanyola del Vallès, Spain.,ICREC Research Program, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Badalona, Spain
| | - Miriam Morón-Font
- REMAR-IVECAT Group, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Badalona, Spain
| | - Ana Gámez-Valero
- REMAR-IVECAT Group, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Badalona, Spain.,Pathology Department, Germans Trias i Pujol University Hospital, Badalona, Spain
| | - Laura Carreras-Planella
- REMAR-IVECAT Group, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Badalona, Spain.,Department of Cell Biology, Physiology and Immunology, Autonomous University of Barcelona, Bellaterra, Cerdanyola del Vallès, Spain
| | - Francesc E Borràs
- REMAR-IVECAT Group, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Badalona, Spain.,Department of Cell Biology, Physiology and Immunology, Autonomous University of Barcelona, Bellaterra, Cerdanyola del Vallès, Spain.,Nephrology Department, Germans Trias i Pujol University Hospital, Badalona, Spain
| | - Marcella Franquesa
- REMAR-IVECAT Group, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Badalona, Spain.,Nephrology Department, Germans Trias i Pujol University Hospital, Badalona, Spain
| |
Collapse
|
250
|
Stem cell exosomes inhibit angiogenesis and tumor growth of oral squamous cell carcinoma. Sci Rep 2019; 9:663. [PMID: 30679544 PMCID: PMC6345809 DOI: 10.1038/s41598-018-36855-6] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 11/28/2018] [Indexed: 12/11/2022] Open
Abstract
Recently, exosomes secreted by menstrual mesenchymal stem cells have been identified as inhibitory agents of tumor angiogenesis and modulators of the tumor cell secretome in prostate and breast cancer. However, their direct effect on endothelial cells and paracrine mediators have not yet been investigated. Using a carrier-based cell culture system to test the scalability for exosome production, we showed that different types of endothelial cells present specific kinetics for exosomes internalization. Exosome-treatment of endothelial cells increased cytotoxicity and reduced VEGF secretion and angiogenesis in a dose-dependent manner. Using the hamster buccal pouch carcinoma as a preclinical model for human oral squamous cell carcinoma, we demonstrated a significant antitumor effect of intra-tumoral injection of exosomes associated with a loss of tumor vasculature. These results address up-scaling of exosome production, a relevant issue for their clinical application, and also assess menstrual stem cell exosomes as potential anti-angiogenic agents for the treatment of neoplastic conditions.
Collapse
|