151
|
Yan HC, Yu TT, Li J, Qiao YQ, Wang LC, Zhang T, Li Q, Zhou YH, Liu DW. The Delivery of Extracellular Vesicles Loaded in Biomaterial Scaffolds for Bone Regeneration. Front Bioeng Biotechnol 2020; 8:1015. [PMID: 32974327 PMCID: PMC7466762 DOI: 10.3389/fbioe.2020.01015] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 08/03/2020] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles (EVs) are heterogeneous nanoparticles actively released by cells that comprise highly conserved and efficient systems of intercellular communication. In recent years, numerous studies have proven that EVs play an important role in the field of bone tissue engineering (BTE) due to several advantages, such as good biosafety, stability and efficient delivery. However, the application of EVs therapies in bone regeneration has not been widely used. One of the major challenges for the application of EVs is the lack of sufficient scaffolds to load and control the release of EVs. Thus, in this review, we describe the most advanced current strategies for delivering EVs with various biomaterials for the use in bone regeneration, the role of EVs in bone regeneration, the distribution of EVs mediated by biomaterials and common methods of promoting EVs delivery efficacy with a focus on biomaterial properties.
Collapse
Affiliation(s)
- Hui-Chun Yan
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Ting-Ting Yu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Jing Li
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Yi-Qiang Qiao
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lin-Chuan Wang
- Eastman Institute for Oral Health, University of Rochester, Rochester, NY, United States
| | - Ting Zhang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Qian Li
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Yan-Heng Zhou
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Da-Wei Liu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Beijing, China
| |
Collapse
|
152
|
Belhadj Z, He B, Deng H, Song S, Zhang H, Wang X, Dai W, Zhang Q. A combined "eat me/don't eat me" strategy based on extracellular vesicles for anticancer nanomedicine. J Extracell Vesicles 2020; 9:1806444. [PMID: 32944191 PMCID: PMC7480498 DOI: 10.1080/20013078.2020.1806444] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 06/07/2020] [Accepted: 07/05/2020] [Indexed: 12/19/2022] Open
Abstract
A long-term and huge challenge in nanomedicine is the substantial uptake and rapid clearance mediated by the mononuclear phagocyte system (MPS), which enormously hinders the development of nanodrugs. Inspired by the natural merits of extracellular vesicles, we therefore developed a combined "eat me/don't eat me" strategy in an effort to achieve MPS escape and efficient drug delivery. Methodologically, cationized mannan-modified extracellular vesicles derived from DC2.4 cells were administered to saturate the MPS (eat me strategy). Then, nanocarriers fused to CD47-enriched exosomes originated from human serum were administered to evade phagocytosis by MPS (don't eat me strategy). The nanocarriers were also loaded with antitumor drugs and functionalized with a novel homing peptide to promote the tumour tissue accumulation and cancer cell uptake (eat me strategy). The concept was proven in vitro as evidenced by the reduced endocytosis of macrophages and enhanced uptake by tumour cells, whereas prolonged circulation time and increased tumour accumulation were demonstrated in vivo. Specially, the strategy induced a 123.53% increase in tumour distribution compared to conventional nanocarrier. The study both shed light on the challenge overcoming of phagocytic evasion and provided a strategy for significantly improving therapeutic outcomes, potentially permitting active drug delivery via targeted nanomedicines.
Collapse
Affiliation(s)
- Zakia Belhadj
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Bing He
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Hailiang Deng
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Siyang Song
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Hua Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Xueqing Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Wenbing Dai
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Qiang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| |
Collapse
|
153
|
Nazari-Shafti TZ, Neuber S, Garcia Duran A, Xu Z, Beltsios E, Seifert M, Falk V, Stamm C. Human mesenchymal stromal cells and derived extracellular vesicles: Translational strategies to increase their proangiogenic potential for the treatment of cardiovascular disease. Stem Cells Transl Med 2020; 9:1558-1569. [PMID: 32761804 PMCID: PMC7695640 DOI: 10.1002/sctm.19-0432] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 05/14/2020] [Accepted: 05/18/2020] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) offer great potential for the treatment of cardiovascular diseases (CVDs) such as myocardial infarction and heart failure. Studies have revealed that the efficacy of MSCs is mainly attributed to their capacity to secrete numerous trophic factors that promote angiogenesis, inhibit apoptosis, and modulate the immune response. There is growing evidence that MSC‐derived extracellular vesicles (EVs) containing a cargo of lipids, proteins, metabolites, and RNAs play a key role in this paracrine mechanism. In particular, encapsulated microRNAs have been identified as important positive regulators of angiogenesis in pathological settings of insufficient blood supply to the heart, thus opening a new path for the treatment of CVD. In the present review, we discuss the current knowledge related to the proangiogenic potential of MSCs and MSC‐derived EVs as well as methods to enhance their biological activities for improved cardiac tissue repair. Increasing our understanding of mechanisms supporting angiogenesis will help optimize future approaches to CVD intervention.
Collapse
Affiliation(s)
- Timo Z Nazari-Shafti
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany.,German Centre for Cardiovascular Research, Partner Site Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sebastian Neuber
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany.,German Centre for Cardiovascular Research, Partner Site Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ana Garcia Duran
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg School for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Zhiyi Xu
- Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Eleftherios Beltsios
- Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Martina Seifert
- Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt- Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Volkmar Falk
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany.,German Centre for Cardiovascular Research, Partner Site Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Division of Cardiovascular Surgery, University of Zurich, Zurich, Switzerland
| | - Christof Stamm
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany.,German Centre for Cardiovascular Research, Partner Site Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
154
|
Platelets Extracellular Vesicles as Regulators of Cancer Progression-An Updated Perspective. Int J Mol Sci 2020; 21:ijms21155195. [PMID: 32707975 PMCID: PMC7432409 DOI: 10.3390/ijms21155195] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/19/2020] [Accepted: 07/20/2020] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles (EVs) are a diverse group of membrane-bound structures secreted in physiological and pathological conditions by prokaryotic and eukaryotic cells. Their role in cell-to-cell communications has been discussed for more than two decades. More attention is paid to assess the impact of EVs in cancer. Numerous papers showed EVs as tumorigenesis regulators, by transferring their cargo molecules (miRNA, DNA, protein, cytokines, receptors, etc.) among cancer cells and cells in the tumor microenvironment. During platelet activation or apoptosis, platelet extracellular vesicles (PEVs) are formed. PEVs present a highly heterogeneous EVs population and are the most abundant EVs group in the circulatory system. The reason for the PEVs heterogeneity are their maternal activators, which is reflected on PEVs size and cargo. As PLTs role in cancer development is well-known, and PEVs are the most numerous EVs in blood, their feasible impact on cancer growth is strongly discussed. PEVs crosstalk could promote proliferation, change tumor microenvironment, favor metastasis formation. In many cases these functions were linked to the transfer into recipient cells specific cargo molecules from PEVs. The article reviews the PEVs biogenesis, cargo molecules, and their impact on the cancer progression.
Collapse
|
155
|
Fernandes M, Lopes I, Teixeira J, Botelho C, Gomes AC. Exosome-like Nanoparticles: A New Type of Nanocarrier. Curr Med Chem 2020; 27:3888-3905. [PMID: 30706777 DOI: 10.2174/0929867326666190129142604] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 11/23/2018] [Accepted: 01/17/2019] [Indexed: 12/12/2022]
Abstract
Nanoparticles are one of the most commonly used systems for imaging or therapeutic drug delivery. Exosomes are nanovesicular carriers that transport cargo for intercellular communication. These nanovesicles are linked to the pathology of some major diseases, in some cases with a central role in their progression. The use of these carriers to transport therapeutic drugs is a recent and promising approach to treat diseases such as cancer and Alzheimer disease. The physiological production of these structures is limited impairing its collection and subsequent purification. These drawbacks inspired the search for mimetic alternatives. The collection of exosome-like nanoparticles from plants can be a good alternative, since they are easier to extract and do not have the drawbacks of those produced in animal cells. Both natural and synthetic exosome-like nanoparticles, produced from serial extrusion of cells or by bottom up synthesis, are currently some of the most promising, biocompatible, high efficiency systems for drug delivery.
Collapse
Affiliation(s)
- Mário Fernandes
- Centre of Biological Engineering (CEB), Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal.,Centre of Molecular and Environmental Biology (CBMA), Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Ivo Lopes
- Centre of Biological Engineering (CEB), Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal.,Centre of Molecular and Environmental Biology (CBMA), Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - José Teixeira
- Centre of Biological Engineering (CEB), Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Cláudia Botelho
- Centre of Biological Engineering (CEB), Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal.,Centre of Molecular and Environmental Biology (CBMA), Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Andreia C Gomes
- Centre of Molecular and Environmental Biology (CBMA), Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| |
Collapse
|
156
|
Transport of Extracellular Vesicles across the Blood-Brain Barrier: Brain Pharmacokinetics and Effects of Inflammation. Int J Mol Sci 2020; 21:ijms21124407. [PMID: 32575812 PMCID: PMC7352415 DOI: 10.3390/ijms21124407] [Citation(s) in RCA: 329] [Impact Index Per Article: 65.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/08/2020] [Accepted: 06/10/2020] [Indexed: 01/08/2023] Open
Abstract
Extracellular vesicles can cross the blood–brain barrier (BBB), but little is known about passage. Here, we used multiple-time regression analysis to examine the ability of 10 exosome populations derived from mouse, human, cancerous, and non-cancerous cell lines to cross the BBB. All crossed the BBB, but rates varied over 10-fold. Lipopolysaccharide (LPS), an activator of the innate immune system, enhanced uptake independently of BBB disruption for six exosomes and decreased uptake for one. Wheatgerm agglutinin (WGA) modulated transport of five exosome populations, suggesting passage by adsorptive transcytosis. Mannose 6-phosphate inhibited uptake of J774A.1, demonstrating that its BBB transporter is the mannose 6-phosphate receptor. Uptake rates, patterns, and effects of LPS or WGA were not predicted by exosome source (mouse vs. human) or cancer status of the cell lines. The cell surface proteins CD46, AVβ6, AVβ3, and ICAM-1 were variably expressed but not predictive of transport rate nor responses to LPS or WGA. A brain-to-blood efflux mechanism variably affected CNS retention and explains how CNS-derived exosomes enter blood. In summary, all exosomes tested here readily crossed the BBB, but at varying rates and by a variety of vesicular-mediated mechanisms involving specific transporters, adsorptive transcytosis, and a brain-to-blood efflux system.
Collapse
|
157
|
Zhang S, Hou Y, Yang J, Xie D, Jiang L, Hu H, Hu J, Luo C, Zhang Q. Application of mesenchymal stem cell exosomes and their drug-loading systems in acute liver failure. J Cell Mol Med 2020; 24:7082-7093. [PMID: 32492261 PMCID: PMC7339207 DOI: 10.1111/jcmm.15290] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 03/22/2020] [Accepted: 03/26/2020] [Indexed: 12/11/2022] Open
Abstract
Stem cell exosomes are nanoscale membrane vesicles released from stem cells of various origins that can regulate signal transduction pathways between liver cells, and their functions in intercellular communication have been recognized. Due to their natural substance transport properties and excellent biocompatibility, exosomes can also be used as drug carriers to release a variety of substances, which has great prospects in the treatment of critical and incurable diseases. Different types of stem cell exosomes have been used to study liver diseases. Due to current difficulties in the treatment of acute liver failure (ALF), this review will outline the potential of stem cell exosomes for ALF treatment. Specifically, we reviewed the pathogenesis of acute liver failure and the latest progress in the use of stem cell exosomes in the treatment of ALF, including the role of exosomes in inhibiting the ALF inflammatory response and regulating signal transduction pathways, the advantages of stem cell exosomes and their use as a drug‐loading system, and their pre‐clinical application in the treatment of ALF. Finally, the clinical research status of stem cell therapy for ALF and the current challenges of exosome clinical transformation are summarized.
Collapse
Affiliation(s)
- Shuqin Zhang
- Office of Clinical Trial of Drug, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Yu Hou
- Office of Clinical Trial of Drug, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Jing Yang
- Office of Clinical Trial of Drug, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Denghui Xie
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Linrui Jiang
- Office of Clinical Trial of Drug, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Huazhong Hu
- Office of Clinical Trial of Drug, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Jingjing Hu
- Office of Clinical Trial of Drug, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Caizhu Luo
- Office of Clinical Trial of Drug, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Qun Zhang
- Office of Clinical Trial of Drug, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| |
Collapse
|
158
|
Exosomes Derived from Stem Cells from the Apical Papilla Promote Dentine-Pulp Complex Regeneration by Inducing Specific Dentinogenesis. Stem Cells Int 2020; 2020:5816723. [PMID: 32565828 PMCID: PMC7273441 DOI: 10.1155/2020/5816723] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/26/2020] [Accepted: 05/12/2020] [Indexed: 12/16/2022] Open
Abstract
Regenerative endodontic procedures (REPs) are a new option for the treatment of dental pulp or periapical diseases in permanent teeth with open apices. Histologically, the new tissues formed in the root canal after REPs are mainly cementum- or bone-like mineralised tissues, but not the real dentine-pulp complex. Therefore, how to promote dentine-pulp complex regeneration and improve the clinical effects of REPs has become a prominent research topic. Stem cells from apical papilla (SCAP) are derived from the dental papilla that can differentiate into primary odontoblasts and dental pulp cells that produce root dentine and dental pulp. Exosomes are the key regulator for the paracrine activity of stem cells and can influence the function of recipient cells. In this study, SCAP-derived exosomes (SCAP-Exo) were introduced into the root fragment containing bone marrow mesenchymal stem cells (BMMSCs) and transplanted subcutaneously into immunodeficient mice. We observed that dental pulp-like tissues were present and the newly formed dentine was deposited onto the existing dentine in the root canal. Afterwards, the effects of SCAP-Exo on the dentinogenesis of BMMSCs were elucidated in vitro. We found that the gene and protein expression of dentine sialophosphoprotein and mineralised nodule formation in BMMSCs treated with SCAP-Exo were significantly increased. In summary, SCAP-Exo were endocytosed by BMMSCs and obviously improved their specific dentinogenesis. The use of exosomes derived from dental stem cells could comprise a potential therapeutic approach for dentine-pulp complex regeneration in REPs.
Collapse
|
159
|
Haney MJ, Zhao Y, Jin YS, Batrakova EV. Extracellular Vesicles as Drug Carriers for Enzyme Replacement Therapy to Treat CLN2 Batten Disease: Optimization of Drug Administration Routes. Cells 2020; 9:cells9051273. [PMID: 32443895 PMCID: PMC7290714 DOI: 10.3390/cells9051273] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/14/2020] [Accepted: 05/19/2020] [Indexed: 12/11/2022] Open
Abstract
CLN2 Batten disease (BD) is one of a broad class of lysosomal storage disorders that is characterized by the deficiency of lysosomal enzyme, TPP1, resulting in a build-up of toxic intracellular storage material in all organs and subsequent damage. A major challenge for BD therapeutics is delivery of enzymatically active TPP1 to the brain to attenuate progressive loss of neurological functions. To accomplish this daunting task, we propose the harnessing of naturally occurring nanoparticles, extracellular vesicles (EVs). Herein, we incorporated TPP1 into EVs released by immune cells, macrophages, and examined biodistribution and therapeutic efficacy of EV-TPP1 in BD mouse model, using various routes of administration. Administration through intrathecal and intranasal routes resulted in high TPP1 accumulation in the brain, decreased neurodegeneration and neuroinflammation, and reduced aggregation of lysosomal storage material in BD mouse model, CLN2 knock-out mice. Parenteral intravenous and intraperitoneal administrations led to TPP1 delivery to peripheral organs: liver, kidney, spleen, and lungs. A combination of intrathecal and intraperitoneal EV-TPP1 injections significantly prolonged lifespan in BD mice. Overall, the optimization of treatment strategies is crucial for successful applications of EVs-based therapeutics for BD.
Collapse
Affiliation(s)
- Matthew J. Haney
- Center for Nanotechnology in Drug Delivery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.J.H.); (Y.Z.)
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Yuling Zhao
- Center for Nanotechnology in Drug Delivery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.J.H.); (Y.Z.)
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Yeon S. Jin
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Elena V. Batrakova
- Center for Nanotechnology in Drug Delivery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.J.H.); (Y.Z.)
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
- Correspondence: ; Tel.: +919-537-3712
| |
Collapse
|
160
|
Xu B, Chen SS, Liu MZ, Gan CX, Li JQ, Guo GH. Stem cell derived exosomes-based therapy for acute lung injury and acute respiratory distress syndrome: A novel therapeutic strategy. Life Sci 2020; 254:117766. [PMID: 32418895 DOI: 10.1016/j.lfs.2020.117766] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/21/2020] [Accepted: 05/06/2020] [Indexed: 02/07/2023]
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is a common critical disease which can be caused by multiple pathological factors in clinic. However, feasible and effective treatment strategies of ALI/ARDS are limited. At present, the beneficial effect of stem cells (SCs)-based therapeutic strategies for ALI/ARDS can be attributed to paracrine. Exosomes, as a paracrine product, are regarded as a critical regulatory mediator. Furthermore, substantial evidence has indicated that exosomes from SCs can transmit bioactive components including genetic material and protein to the recipient cells and provide a protective effect. The protective role is played through a series of process including inflammation modulation, the reconstruction of alveolar epithelium and endothelium, and pulmonary fibrosis prevention. Therefore, SCs derived exosomes have the potential to be used for therapeutic strategies for ALI/ARDS. In this review, we discuss the present understanding of SCs derived exosomes related to ALI/ARDS and provide insights for developing a cell-free strategy for treating ALI/ARDS.
Collapse
Affiliation(s)
- Bin Xu
- Department of Burns, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Si-Si Chen
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Ming-Zhuo Liu
- Department of Burns, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Chun-Xia Gan
- Department of Burns, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Jia-Qi Li
- Department of Burns, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Guang-Hua Guo
- Department of Burns, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| |
Collapse
|
161
|
Innovative Visualization and Quantification of Extracellular Vesicles Interaction with and Incorporation in Target Cells in 3D Microenvironments. Cells 2020; 9:cells9051180. [PMID: 32397409 PMCID: PMC7291305 DOI: 10.3390/cells9051180] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/30/2020] [Accepted: 05/06/2020] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EVs) showed therapeutic properties in several applications, many in regenerative medicine. A clear example is in the treatment of osteoarthritis (OA), where adipose-derived mesenchymal stem cells (ASCs)-EVs were able to promote regeneration and reduce inflammation in both synovia and cartilage. A still obscure issue is the effective ability of EVs to be internalized by target cells, rather than simply bound to the extracellular matrix (ECM) or plasma membrane, since the current detection or imaging technologies cannot fully decipher it due to technical limitations. In the present study, human articular chondrocytes (ACHs) and fibroblast-like synoviocytes (FLSs) isolated from the same OA patients were cocultured in 2D as well as in 3D conditions with fluorescently labeled ASC-EVs, and analyzed by flow cytometry or confocal microscopy, respectively. In contrast with conventional 2D, in 3D cultures, confocal microscopy allowed a clear detection of the tridimensional morphology of the cells and thus an accurate discrimination of EV interaction with the external and/or internal cell environment. In both 2D and 3D conditions, FLSs were more efficient in interacting with ASC-EVs and 3D imaging demonstrated a faster uptake process. The removal of the hyaluronic acid component from the ECM of both cell types reduced their interaction with ASC-EVs only in the 2D system, showing that 2D and 3D conditions can yield different outcomes when investigating events where ECM plays a key role. These results indicate that studying EVs binding and uptake both in 2D and 3D guarantees a more precise and complementary characterization of the molecular mechanisms involved in the process. The implementation of this strategy can become a valuable tool not only for basic research, but also for release assays and potency prediction for clinical EV batches.
Collapse
|
162
|
Arenaccio C, Chiozzini C, Ferrantelli F, Leone P, Olivetta E, Federico M. Exosomes in Therapy: Engineering, Pharmacokinetics and Future Applications. Curr Drug Targets 2020; 20:87-95. [PMID: 29779478 DOI: 10.2174/1389450119666180521100409] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 05/04/2018] [Accepted: 05/16/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND Eukaryotic cells release vesicles of different sizes under both physiological and pathological conditions. On the basis of the respective biogenesis, extracellular vesicles are classified as apoptotic bodies, microvesicles, and exosomes. Among these, exosomes are considered tools for innovative therapeutic interventions, especially when engineered with effector molecules. The delivery functions of exosomes are favored by a number of typical features. These include their small size (i.e., 50-200 nm), the membrane composition tightly similar to that of producer cells, lack of toxicity, stability in serum as well as other biological fluids, and accession to virtually any organ and tissue including central nervous system. However, a number of unresolved questions still affects the possible use of exosomes in therapy. Among these are the exact identification of both in vitro and ex vivo produced vesicles, their large-scale production and purification, the uploading efficiency of therapeutic macromolecules, and the characterization of their pharmacokinetics. OBJECTIVE Here, we discuss two key aspects to be analyzed before considering exosomes as a tool of delivery for the desired therapeutic molecule, i.e., techniques of engineering, and their in vivo biodistribution/ pharmacokinetics. In addition, an innovative approach aimed at overcoming at least part of the obstacles towards a safe and efficient use of exosomes in therapy will be discussed. CONCLUSION Several biologic features render exosomes an attractive tool for the delivery of therapeutic molecules. They will surely be a part of innovative therapeutic interventions as soon as few still unmet technical hindrances will be overcome.
Collapse
Affiliation(s)
- Claudia Arenaccio
- National Center for Global Health, Istituto Superiore di Sanità (ISS), Viale Regina Elena 299, 00161, Rome, Italy
| | - Chiara Chiozzini
- National Center for Global Health, Istituto Superiore di Sanità (ISS), Viale Regina Elena 299, 00161, Rome, Italy
| | - Flavia Ferrantelli
- National Center for Global Health, Istituto Superiore di Sanità (ISS), Viale Regina Elena 299, 00161, Rome, Italy
| | - Patrizia Leone
- National Center for Global Health, Istituto Superiore di Sanità (ISS), Viale Regina Elena 299, 00161, Rome, Italy
| | - Eleonora Olivetta
- National Center for Global Health, Istituto Superiore di Sanità (ISS), Viale Regina Elena 299, 00161, Rome, Italy
| | - Maurizio Federico
- National Center for Global Health, Istituto Superiore di Sanità (ISS), Viale Regina Elena 299, 00161, Rome, Italy
| |
Collapse
|
163
|
Abstract
Exosomes are membrane-bound cargo measuring 30–140 nm comprised of a lipid bilayer containing various proteins, RNAs, DNAs, and bioactive lipids that can be transferred between cells. They have been shown to be produced and released by many different types of healthy and diseased cells. Exosomes are secreted by all types of cells in culture, and are also found in various body fluids including blood, saliva, urine, and breast milk. Exosomes are essential for healthy physiological as well as pathological processes. In addition to their normal function, exosomes are involved in the development and progression of various diseases, potentiating cellular stress and damage. Pathogens take advantage of exosome release from infected host cells by manipulating host-derived exosomes to evade the immune system responses. Exosomes are involved in other pathological conditions such as neurodegenerative diseases, liver diseases, heart failure, cancer, diabetes, kidney diseases, osteoporosis and atherosclerotic cardiovascular disease. Hence, we can exploit exosomes as biomarkers and vaccines and modify them rationally for therapeutic interventions including tissue engineering. Further studies on exosomes will explore their potential and provide new methodology for effective clinical diagnostics and therapeutic strategies: such uses can be called exosome theragnostics. This chapter reviews the potential theragnostic (diagnostic and therapeutic) application of exosomes in major organ systems in clinical fields.
Collapse
|
164
|
Hood JL. Natural melanoma-derived extracellular vesicles. Semin Cancer Biol 2019; 59:251-265. [DOI: 10.1016/j.semcancer.2019.06.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 06/26/2019] [Accepted: 06/28/2019] [Indexed: 12/20/2022]
|
165
|
Fukuta T, Nishikawa A, Kogure K. Low level electricity increases the secretion of extracellular vesicles from cultured cells. Biochem Biophys Rep 2019; 21:100713. [PMID: 31828227 PMCID: PMC6889636 DOI: 10.1016/j.bbrep.2019.100713] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 10/31/2019] [Accepted: 11/22/2019] [Indexed: 01/01/2023] Open
Abstract
Exosomes, a type of extracellular vesicles, can be collected from the conditioned medium of cultured cells, and are expected to be used in disease therapy and drug delivery systems. However, since the yield of exosomes from conditioned medium is generally low, investigations to develop new methods to increase exosome secretion and to elucidate the secretion mechanism have been performed. Our previous studies demonstrated that activation of intracellular signaling including Rho GTPase and subsequent endocytosis of extraneous molecules in cells could be induced by low level electricity (0.3–0.5 mA/cm2). Since exosomes are produced in the process of endocytosis and secreted by exocytosis via certain signaling pathways, we hypothesized that low level electric treatment (ET) would increase exosome secretion from cultured cells via intracellular signaling activation. In the present study, the influence of ET (0.34 mA/cm2) on extracellular vesicle (EV) secretion from cultured cells was examined by using murine melanoma and murine fibroblast cells. The results showed that the number of EV particles collected by ultracentrifugation was remarkably increased by ET in both cell lines without cellular toxicity or changes in the particle distribution. Also, protein amounts of the collected EVs were significantly increased in both cells by ET without alteration of expression of representative exosome marker proteins. Moreover, in both cells, the ratio of particle numbers to protein amount was not significantly changed by ET. Rho GTPase inhibition significantly suppressed ET-mediated increase of EV secretion in murine melanoma, indicating that Rho GTPase activation could be involved in ET-mediated EV secretion in the cell. Additionally, there were almost no differences in uptake of each EV into each donor cell regardless of whether the cells had been exposed to ET for EV collection. Taken together, these results suggest that ET could increase EV secretion from both cancer and normal cells without apparent changes in EV quality. Low level electric treatment (ET; 0.34 mA/cm2) increased exosome yield from cells. The number of exosome particles was increased by ET without distribution changes. ET increased protein amounts of the collected exosomes from conditioned media. ET did not induce cellular toxicity and change in exosomal marker protein expression. ET-mediated increase of exosome secretion occurred in both cancer and normal cells.
Collapse
Affiliation(s)
- Tatsuya Fukuta
- Department of Pharmaceutical Health Chemistry, Graduate School of Biomedical Sciences, Tokushima University, Shomachi 1, Tokushima, 770-8505, Japan
| | - Akina Nishikawa
- Department of Pharmaceutical Health Chemistry, Graduate School of Biomedical Sciences, Tokushima University, Shomachi 1, Tokushima, 770-8505, Japan
| | - Kentaro Kogure
- Department of Pharmaceutical Health Chemistry, Graduate School of Biomedical Sciences, Tokushima University, Shomachi 1, Tokushima, 770-8505, Japan
| |
Collapse
|
166
|
Chen Z, Chopp M, Zacharek A, Li W, Venkat P, Wang F, Landschoot-Ward J, Chen J. Brain-Derived Microparticles (BDMPs) Contribute to Neuroinflammation and Lactadherin Reduces BDMP Induced Neuroinflammation and Improves Outcome After Stroke. Front Immunol 2019; 10:2747. [PMID: 31993045 PMCID: PMC6968774 DOI: 10.3389/fimmu.2019.02747] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 11/08/2019] [Indexed: 12/28/2022] Open
Abstract
Microparticles (MPs, ~size between 0.1 and 1 mm) are lipid encased containers derived from intact cells which contain antigen from the parent cells. MPs are involved in intercellular communication and regulate inflammation. Stroke increases secretion of brain derived MP (BDMP) which activate macrophages/microglia and induce neuroinflammation. Lactadherin (Milk fat globule–EGF factor-8) binds to anionic phospholipids and extracellular matrices, promotes apoptotic cell clearance and limits pathogenic antigen cross presentation. In this study, we investigate whether BDMP affects stroke-induced neuroinflammation and whether Lactadherin treatment reduces stroke initiated BDMP-induced neuroinflammation, thereby improving functional outcome after stroke. Middle aged (8–9 months old) male C57BL/6J mice were subjected to distal middle cerebral artery occlusion (dMCAo) stroke, and BDMPs were extracted from ischemic brain 24 h after dMCAo by ultracentrifugation. Adult male C57BL/6J mice were subjected to dMCAo and treated via tail vein injection at 3 h after stroke with: (A) +PBS (n = 5/group); (B) +BDMPs (1.5 × 108, n = 6/group); (C) +Lactadherin (400 μg/kg, n = 5/group); (D) +BDMP+Lactadherin (n = 6/group). A battery of neurological function tests were performed and mice sacrificed for immunostaining at 14 days after stroke. Blood plasma was used for Western blot assay. Our data indicate: (1) treatment of Stroke with BDMP significantly increases lesion volume, neurological deficits, blood brain barrier (BBB) leakage, microglial activation, inflammatory cell infiltration (CD45, microglia/macrophages, and neutrophils) into brain, inflammatory factor (TNFα, IL6, and IL1β) expression in brain, increases axon/white matter (WM) damage identified by decreased axon and myelin density, and increases inflammatory factor expression in the plasma when compared to PBS treated stroke mice; (2) when compared to PBS and BDMP treated stroke mice, Lactadherin and BDMP+Lactadherin treatment significantly improves neurological outcome, and decreases lesion volume, BBB leakage, axon/WM injury, inflammatory cell infiltration and inflammatory factor expression in the ischemic brain, respectively. Lactadherin treatment significantly increases anti-inflammatory factor (IL10) expression in ischemic brain and decreases IL1β expression in plasma compared to PBS and BDMP treated stroke mice, respectively. BDMP increases neuroinflammation and aggravates ischemic brain damage after stroke. Thus, Lactadherin exerts anti-inflammatory effects and improves the clearance of MPs to reduce stroke and BDMP induced neurological deficits.
Collapse
Affiliation(s)
- Zhili Chen
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States.,Department of Physics, Oakland University, Rochester, MI, United States
| | - Alex Zacharek
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Wei Li
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Poornima Venkat
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Fenjie Wang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | | | - Jieli Chen
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| |
Collapse
|
167
|
Riau AK, Ong HS, Yam GHF, Mehta JS. Sustained Delivery System for Stem Cell-Derived Exosomes. Front Pharmacol 2019; 10:1368. [PMID: 31798457 PMCID: PMC6868085 DOI: 10.3389/fphar.2019.01368] [Citation(s) in RCA: 161] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 10/28/2019] [Indexed: 12/20/2022] Open
Abstract
Recent literature has ascribed that the paracrine action of stem cells is mediated by exosomes. Exosomes are nano-sized extracellular vesicles (30 to 100 nm) of endocytic origin that play important roles in intercellular communication. They have the ability to deliver various therapeutic effects, e.g., skin regeneration or cardiac function recovery, when applied topically or injected systemically. However, injection of exosomes has been shown to result in rapid clearance from blood circulation and accumulation of the exosomes in the liver, spleen, lung, and gastrointestinal tract can be found as early as 2 h after injection. Topical administration of exosomes on the skin or ocular surface would suffer the same fate due to rapid fluid turnover (sweat or tears). Biodegradable or highly porous hydrogels have been utilized to load exosomes and to deliver a sustained therapeutic effect. They can also prevent the exosomes from being cleared prematurely and allow the delivery of a more localized and concentrated exosome dosage by placing the hydrogel directly at or in the proximity of the target site. In this mini-review, we elaborate on the challenges of conventional exosome administration and highlight the solution to the shortcomings in the form of exosome-incorporated hydrogels. Different techniques to encapsulate exosomes and examples of hydrogels that have been used to create sustained delivery systems of exosomes are also discussed.
Collapse
Affiliation(s)
- Andri K Riau
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore, Singapore
| | - Hon Shing Ong
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore, Singapore.,Corneal and External Eye Disease Department, Singapore National Eye Centre, Singapore, Singapore.,Ophthalmology and Visual Sciences Academic Clinical Programme, Duke-NUS Medical School, Singapore, Singapore
| | - Gary H F Yam
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore, Singapore.,Ophthalmology and Visual Sciences Academic Clinical Programme, Duke-NUS Medical School, Singapore, Singapore
| | - Jodhbir S Mehta
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore, Singapore.,Corneal and External Eye Disease Department, Singapore National Eye Centre, Singapore, Singapore.,Ophthalmology and Visual Sciences Academic Clinical Programme, Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
168
|
Abstract
Extracellular vesicles (EVs) are essential tools for conveying biological information and modulating functions of recipient cells. Implantation of isolated or modulated EVs can be innovative therapeutics for various diseases. Furthermore, EVs could be a biocompatible drug delivery vehicle to carry both endogenous and exogenous biologics. Tracking EVs should play essential roles in understanding the functions of EVs and advancing EV therapeutics. EVs have the characteristic structures consisting of the lipid bilayer and specific membrane proteins, through which they can be labeled efficiently. EVs can be labeled either directly using probes or indirectly by transfection of reporter genes. Optical imaging (fluorescent imaging and bioluminescent imaging), single-photon emission computed tomography (SPECT)/positron emission tomography (PET), and magnetic resonance imaging (MRI) are currently used for imaging EVs. Labeling EVs with superparamagnetic iron oxide (SPIO) nanoparticles for MRI tracking is a promising method that can be translated into clinic. SPIO can be internalized by most of the cell types and then released as SPIO containing EVs, which can be visualized on T2*-weighted imaging. However, this method has limitations in real-time imaging because of the life cycle of SPIO after EV degradation. Further studies will be needed to validate SPIO labeling by other imaging modalities in preclinical studies. The emerging technologies of labeling and imaging EVs with SPIO in comparison with other imaging modalities are reviewed in this paper.
Collapse
|
169
|
Oshchepkova A, Neumestova A, Matveeva V, Artemyeva L, Morozova K, Kiseleva E, Zenkova M, Vlassov V. Cytochalasin-B-Inducible Nanovesicle Mimics of Natural Extracellular Vesicles That Are Capable of Nucleic Acid Transfer. MICROMACHINES 2019; 10:E750. [PMID: 31683842 PMCID: PMC6915531 DOI: 10.3390/mi10110750] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 10/27/2019] [Accepted: 10/31/2019] [Indexed: 12/12/2022]
Abstract
Extracellular vesicles provide cell-to-cell communication and have great potential for use as therapeutic carriers. This study was aimed at the development of an extracellular vesicle-based system for nucleic acid delivery. Three types of nanovesicles were assayed as oligonucleotide carriers: mesenchymal stem cell-derived extracellular vesicles and mimics prepared either by cell treatment with cytochalasin B or by vesicle generation from plasma membrane. Nanovesicles were loaded with a DNA oligonucleotide by freezing/thawing, sonication, or permeabilization with saponin. Oligonucleotide delivery was assayed using HEK293 cells. Extracellular vesicles and mimics were characterized by a similar oligonucleotide loading level but different efficiency of oligonucleotide delivery. Cytochalasin-B-inducible nanovesicles exhibited the highest level of oligonucleotide accumulation in HEK293 cells and a loading capacity of 0.44 ± 0.05 pmol/µg. The loaded oligonucleotide was mostly protected from nuclease action.
Collapse
Affiliation(s)
- Anastasiya Oshchepkova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia.
| | - Alexandra Neumestova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia.
| | - Vera Matveeva
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia.
| | - Lyudmila Artemyeva
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia.
| | - Ksenia Morozova
- Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia.
| | - Elena Kiseleva
- Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia.
| | - Marina Zenkova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia.
| | - Valentin Vlassov
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia.
| |
Collapse
|
170
|
Mansouri N, Willis GR, Fernandez-Gonzalez A, Reis M, Nassiri S, Mitsialis SA, Kourembanas S. Mesenchymal stromal cell exosomes prevent and revert experimental pulmonary fibrosis through modulation of monocyte phenotypes. JCI Insight 2019; 4:128060. [PMID: 31581150 DOI: 10.1172/jci.insight.128060] [Citation(s) in RCA: 159] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 09/27/2019] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stromal/stem cell (MSC) therapy has shown promise in experimental models of idiopathic pulmonary fibrosis (IPF). The aim of this study was to test the therapeutic effects of extracellular vesicles produced by human BM MSCs (MEx) in a bleomycin-induced pulmonary fibrosis model and investigate mechanisms of action. Adult C57BL/6 mice were challenged with endotracheal instillation of bleomycin and treated with MEx concurrently, or for reversal models, at day 7 or 21. Experimental groups were assessed at day 7, 14, or 28. Bleomycin-challenged mice presented with severe septal thickening and prominent fibrosis, and this was effectively prevented or reversed by MEx treatment. MEx modulated lung macrophage phenotypes, shifting the proportions of lung proinflammatory/classical and nonclassical monocytes and alveolar macrophages toward the monocyte/macrophage profiles of control mice. A parallel immunomodulatory effect was demonstrated in the BM. Notably, transplantation of MEx-preconditioned BM-derived monocytes alleviated core features of pulmonary fibrosis and lung inflammation. Proteomic analysis revealed that MEx therapy promotes an immunoregulatory, antiinflammatory monocyte phenotype. We conclude that MEx prevent and revert core features of bleomycin-induced pulmonary fibrosis and that the beneficial actions of MEx may be mediated via systemic modulation of monocyte phenotypes.
Collapse
Affiliation(s)
- Nahal Mansouri
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital (BCH), Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA.,Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.,Division of Pulmonary Medicine, Department of Medicine, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Gareth R Willis
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital (BCH), Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Angeles Fernandez-Gonzalez
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital (BCH), Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Monica Reis
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital (BCH), Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Sina Nassiri
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.,Bioinformatics Core Facility, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - S Alex Mitsialis
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital (BCH), Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Stella Kourembanas
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital (BCH), Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
171
|
Walker S, Busatto S, Pham A, Tian M, Suh A, Carson K, Quintero A, Lafrence M, Malik H, Santana MX, Wolfram J. Extracellular vesicle-based drug delivery systems for cancer treatment. Theranostics 2019; 9:8001-8017. [PMID: 31754377 PMCID: PMC6857056 DOI: 10.7150/thno.37097] [Citation(s) in RCA: 286] [Impact Index Per Article: 47.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 09/13/2019] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles (EVs) are naturally occurring cell-secreted nanoparticles that play important roles in many physiological and pathological processes. EVs enable intercellular communication by serving as delivery vehicles for a wide range of endogenous cargo molecules, such as RNAs, proteins, carbohydrates, and lipids. EVs have also been found to display tissue tropism mediated by surface molecules, such as integrins and glycans, making them promising for drug delivery applications. Various methods can be used to load therapeutic agents into EVs, and additional modification strategies have been employed to prolong circulation and improve targeting. This review gives an overview of EV-based drug delivery strategies in cancer therapy.
Collapse
Affiliation(s)
- Sierra Walker
- Department of Transplantation/Department of Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Sara Busatto
- Department of Transplantation/Department of Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Anthony Pham
- Department of Transplantation/Department of Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Ming Tian
- Department of Transplantation/Department of Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Annie Suh
- Department of Transplantation/Department of Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Kelsey Carson
- Department of Biology, University of North Florida, Jacksonville, FL, 32224, USA
| | - Astrid Quintero
- Department of Biology, University of North Florida, Jacksonville, FL, 32224, USA
| | - Maria Lafrence
- Department of Biology, University of North Florida, Jacksonville, FL, 32224, USA
| | - Hanna Malik
- Department of Biology, University of North Florida, Jacksonville, FL, 32224, USA
| | - Moises X. Santana
- Department of Biology, University of North Florida, Jacksonville, FL, 32224, USA
| | - Joy Wolfram
- Department of Transplantation/Department of Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, FL, 32224, USA
- Department of Biology, University of North Florida, Jacksonville, FL, 32224, USA
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
| |
Collapse
|
172
|
Vidal M. Exosomes: Revisiting their role as "garbage bags". Traffic 2019; 20:815-828. [PMID: 31418976 DOI: 10.1111/tra.12687] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 08/14/2019] [Indexed: 12/17/2022]
Abstract
In recent years, the term "extracellular vesicle" (EV) has been used to define different types of vesicles released by various cells. It includes plasma membrane-derived vesicles (ectosomes/microvesicles) and endosome-derived vesicles (exosomes). Although it remains difficult to evaluate the compartment of origin of the two kinds of vesicles once released, it is critical to discriminate these vesicles because their mode of biogenesis is probably directly related to their physiologic function and/or to the physio-pathologic state of the producing cell. The purpose of this review is to specifically consider exosome secretion and its consequences in terms of a material loss for producing cells, rather than on the effects of exosomes once they are taken up by recipient cells. I especially describe one putative basic function of exosomes, that is, to convey material out of cells for off-site degradation by recipient cells. As illustrated by some examples, these components could be evacuated from cells for various reasons, for example, to promote "differentiation" or enhance homeostatic responses. This basic function might explain why so many diseases have made use of the exosomal pathway during pathogenesis.
Collapse
Affiliation(s)
- Michel Vidal
- LPHI - Université de Montpellier, CNRS, Montpellier, France
| |
Collapse
|
173
|
Patras L, Banciu M. Intercellular Crosstalk Via Extracellular Vesicles in Tumor Milieu as Emerging Therapies for Cancer Progression. Curr Pharm Des 2019; 25:1980-2006. [DOI: 10.2174/1381612825666190701143845] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 06/19/2019] [Indexed: 02/07/2023]
Abstract
:Increasing evidence has suggested that extracellular vesicles (EV) mediated bidirectional transfer of functional molecules (such as proteins, different types of RNA, and lipids) between cancer cells and tumor stromal cells (immune cells, endothelial cells, fibroblasts, stem cells) and strongly contributed to the reinforcement of cancer progression. Thus, intercellular EV-mediated signaling in tumor microenvironment (TME) is essential in the modulation of all processes that support and promote tumor development like immune suppression, angiogenesis, invasion and metastasis, and resistance of tumor cells to anticancer treatments.:Besides EV potential to revolutionize our understanding of the cancer cell-stromal cells crosstalk in TME, their ability to selectively transfer different cargos to recipient cells has created excitement in the field of tumortargeted delivery of specific molecules for anticancer treatments. Therefore, in tight connection with previous findings, this review brought insight into the dual role of EV in modulation of TME. Thus, on one side EV create a favorable phenotype of tumor stromal cells for tumor progression; however, as a future new class of anticancer drug delivery systems EV could re-educate the TME to overcome main supportive processes for malignancy progression.
Collapse
Affiliation(s)
- Laura Patras
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Manuela Banciu
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania
| |
Collapse
|
174
|
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
|
175
|
Extracellular vesicles and their diagnostic potential in amyotrophic lateral sclerosis. Clin Chim Acta 2019; 497:27-34. [PMID: 31301281 DOI: 10.1016/j.cca.2019.07.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 07/09/2019] [Accepted: 07/09/2019] [Indexed: 01/15/2023]
Abstract
Extracellular vesicles, small reservoirs that carry various biomolecules, have gained significant interest from the clinical field in recent years based on the diagnostic, therapeutic and prognostic possibilities they offer. While information abound regarding the clinical potential of such vesicles in diverse conditions, the information demonstrating their likely importance in amyotrophic lateral sclerosis (ALS) is more limited. This review will thus provide a brief introduction to extracellular vesicles, highlight their diagnostic significance in various diseases with a focus on ALS and explore additional applications of extracellular vesicles in the medical field. Overall, this work sheds further light on the clinical importance of extracellular vesicles in diagnostic applications as well as supports the need to better characterize their roles and signatures in patients diagnosed with ALS.
Collapse
|
176
|
Balachandran B, Yuana Y. Extracellular vesicles-based drug delivery system for cancer treatment. COGENT MEDICINE 2019. [DOI: 10.1080/2331205x.2019.1635806] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Banuja Balachandran
- Division of Imaging, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Yuana Yuana
- Division of Imaging, University Medical Centre Utrecht, Utrecht, The Netherlands
- Faculty of Biomedical Engineering, Technical University Eindhoven, Eindhoven, The Netherlands
| |
Collapse
|
177
|
Current Trends and Future Perspective of Mesenchymal Stem Cells and Exosomes in Corneal Diseases. Int J Mol Sci 2019; 20:ijms20122853. [PMID: 31212734 PMCID: PMC6627168 DOI: 10.3390/ijms20122853] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 06/01/2019] [Accepted: 06/10/2019] [Indexed: 02/07/2023] Open
Abstract
The corneal functions (transparency, refractivity and mechanical strength) deteriorate in many corneal diseases but can be restored after corneal transplantation (penetrating and lamellar keratoplasties). However, the global shortage of transplantable donor corneas remains significant and patients are subject to life-long risk of immune response and graft rejection. Various studies have shown the differentiation of multipotent mesenchymal stem cells (MSCs) into various corneal cell types. With the unique properties of immunomodulation, anti-angiogenesis and anti-inflammation, they offer the advantages in corneal reconstruction. These effects are widely mediated by MSC differentiation and paracrine signaling via exosomes. Besides the cell-free nature of exosomes in circumventing the problems of cell-fate control and tumorigenesis, the vesicle content can be genetically modified for optimal therapeutic affinity. The pharmacology and toxicology, xeno-free processing with sustained delivery, scale-up production in compliant to Good Manufacturing Practice regulations, and cost-effectiveness are the current foci of research. Routes of administration via injection, topical and/or engineered bioscaffolds are also explored for its applicability in treating corneal diseases.
Collapse
|
178
|
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
|
179
|
Kang JY, Park H, Kim H, Mun D, Park H, Yun N, Joung B. Human peripheral blood‑derived exosomes for microRNA delivery. Int J Mol Med 2019; 43:2319-2328. [PMID: 30942393 PMCID: PMC6488179 DOI: 10.3892/ijmm.2019.4150] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 03/27/2019] [Indexed: 12/20/2022] Open
Abstract
Exosomes serve important functions in cell-to-cell communication and biological functions by serving as a delivery cargo shuttle for various molecules. The application of an improved delivery method for microRNAs (miRNAs/miRs) may enhance their potential as a therapeutic tool in cardiac diseases. Thus, the present study investigated whether human peripheral blood-derived exosomes may be used as a delivery cargo system for miRNAs, and whether the delivery of miR-21 using a human peripheral blood derived-exosome may influence the degree of remodeling following myocardial infarction (MI). In H9C2 and HL-1 cells, miR-21 expression was successfully regulated by treatment with human peripheral blood derived-exosomes loaded with an miR-21 mimic or inhibitor compared with untreated cells. In addition, the mRNA and protein expression levels of SMAD family member 7 (Smad7), phosphatase and tensin homolog (PTEN) and matrix metalloproteinase 2 (MMP2), which are involved in cardiac fibrosis, were associated with the uptake of miR-21 mimic- or inhibitor-loaded exosomes. Similarly, the in vivo mRNA and protein expression of Smad7, PTEN and MMP2 were altered following treatment with miR-21 mimic- or inhibitor-loaded exosomes. Furthermore, miR-21 mimic-loaded exosomes enhanced fibrosis, whereas miR-21 inhibitor-loaded exosomes reduced fibrosis in a mouse MI model. These results suggested that miRNA-loaded human peripheral blood derived-exosomes may be used as a therapeutic tool for cardiac diseases.
Collapse
Affiliation(s)
- Ji-Young Kang
- Division of Cardiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Hyewon Park
- Division of Cardiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Hyoeun Kim
- Division of Cardiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Dasom Mun
- Division of Cardiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Hyelim Park
- Division of Cardiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Nuri Yun
- Institute of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Boyoung Joung
- Division of Cardiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| |
Collapse
|
180
|
Murphy DE, de Jong OG, Brouwer M, Wood MJ, Lavieu G, Schiffelers RM, Vader P. Extracellular vesicle-based therapeutics: natural versus engineered targeting and trafficking. Exp Mol Med 2019; 51:1-12. [PMID: 30872574 PMCID: PMC6418170 DOI: 10.1038/s12276-019-0223-5] [Citation(s) in RCA: 467] [Impact Index Per Article: 77.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 12/12/2018] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles (EVs) are increasingly being recognized as mediators of intercellular signaling via the delivery of effector molecules. Interestingly, certain types of EVs are also capable of inducing therapeutic responses. For these reasons, the therapeutic potential of EVs is a topic of intense research, both in the context of drug delivery and regenerative medicine. However, to fully utilize EVs for therapeutic purposes, an improved understanding of the mechanisms by which they function would be highly advantageous. Here, the current state of knowledge regarding the cellular uptake and trafficking of EVs is reviewed, along with a consideration of how these pathways potentially influence the functions of therapeutic EVs. Furthermore, the natural cell-targeting abilities, biodistribution profiles, and pharmacokinetics of exogenously administered EVs, along with the components responsible for these features are discussed. An overview of the potential clinical applications and preclinical examples of their successful use is also provided. Finally, examples of EV modifications that have successfully been employed to improve their therapeutic characteristics receive a particular focus. We suggest that, in addition to investigation of EV cell targeting and routes of uptake, future research into the routes of intracellular trafficking in recipient cells is required to optimally utilize EVs for therapeutic purposes. An increased understanding of how extracellular vesicles (EVs) enter cells and deliver molecules will enable promising new therapies, according to researchers in the Netherlands, UK and France. EVs are liquid-filled sacs secreted by cells that transport proteins, lipids and RNA between cells, and therefore have potential for delivering drugs. Pieter Vader at UMC Utrecht and co-workers review recent research into EVs, focusing on how EVs are distributed around the body, and how they target and enter cells. However, there is little known about EV biology once they are inside cells, and it is likely that many EVs simply degrade without delivering their cargo. Further research in this area could help identify features that improve cargo escape from EVs, thus ensuring that future therapies can be effective.
Collapse
Affiliation(s)
- Daniel E Murphy
- Laboratory of Clinical Chemistry and Haematology, UMC Utrecht, Utrecht, The Netherlands
| | - Olivier G de Jong
- Laboratory of Clinical Chemistry and Haematology, UMC Utrecht, Utrecht, The Netherlands.,Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Maarten Brouwer
- Laboratory of Clinical Chemistry and Haematology, UMC Utrecht, Utrecht, The Netherlands
| | - Matthew J Wood
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Grégory Lavieu
- Institut Curie, PSL Research University, INSERM U932, Paris, France
| | - Raymond M Schiffelers
- Laboratory of Clinical Chemistry and Haematology, UMC Utrecht, Utrecht, The Netherlands.
| | - Pieter Vader
- Laboratory of Clinical Chemistry and Haematology, UMC Utrecht, Utrecht, The Netherlands. .,Department of Experimental Cardiology, UMC Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
181
|
Zhang M, Zang X, Wang M, Li Z, Qiao M, Hu H, Chen D. Exosome-based nanocarriers as bio-inspired and versatile vehicles for drug delivery: recent advances and challenges. J Mater Chem B 2019; 7:2421-2433. [PMID: 32255119 DOI: 10.1039/c9tb00170k] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Recent decades have witnessed the fast and impressive development of nanocarriers as a drug delivery system. Considering the safety, delivery efficiency and stability of nanocarriers, there are many obstacles in accomplishing successful clinical translation of these nanocarrier-based drug delivery systems. The gap has urged drug delivery scientists to develop innovative nanocarriers with high compatibility, stability and longer circulation time. Exosomes are nanometer-sized, lipid-bilayer-enclosed extracellular vesicles secreted by many types of cells. Exosomes serving as versatile drug vehicles have attracted increasing attention due to their inherent ability of shuttling proteins, lipids and genes among cells and their natural affinity to target cells. Attractive features of exosomes, such as nanoscopic size, low immunogenicity, high biocompatibility, encapsulation of various cargoes and the ability to overcome biological barriers, distinguish them from other nanocarriers. To date, exosome-based nanocarriers delivering small molecule drugs as well as bioactive macromolecules have been developed for the treatment of many prevalent and obstinate diseases including cancer, CNS disorders and some other degenerative diseases. Exosome-based nanocarriers have a huge prospect in overcoming many hindrances encountered in drug and gene delivery. This review highlights the advances as well as challenges of exosome-based nanocarriers as drug vehicles. Special focus has been placed on the advantages of exosomes in delivering various cargoes and in treating obstinate diseases, aiming to offer new insights for exploring exosomes in the field of drug delivery.
Collapse
Affiliation(s)
- Mengjun Zhang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, P. R. China.
| | | | | | | | | | | | | |
Collapse
|
182
|
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
|
183
|
Verweij FJ, Revenu C, Arras G, Dingli F, Loew D, Pegtel DM, Follain G, Allio G, Goetz JG, Zimmermann P, Herbomel P, Del Bene F, Raposo G, van Niel G. Live Tracking of Inter-organ Communication by Endogenous Exosomes In Vivo. Dev Cell 2019; 48:573-589.e4. [PMID: 30745143 DOI: 10.1016/j.devcel.2019.01.004] [Citation(s) in RCA: 232] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 12/21/2018] [Accepted: 12/31/2018] [Indexed: 01/05/2023]
Abstract
Extracellular vesicles (EVs) are released by most cell types but providing evidence for their physiological relevance remains challenging due to a lack of appropriate model organisms. Here, we developed an in vivo model to study EV function by expressing CD63-pHluorin in zebrafish embryos. A combination of imaging methods and proteomic analysis allowed us to study biogenesis, composition, transfer, uptake, and fate of individual endogenous EVs. We identified a subpopulation of EVs with exosome features, released in a syntenin-dependent manner from the yolk syncytial layer into the blood circulation. These exosomes are captured, endocytosed, and degraded by patrolling macrophages and endothelial cells in the caudal vein plexus (CVP) in a scavenger receptor- and dynamin-dependent manner. Interference with exosome biogenesis affected CVP growth, suggesting a role in trophic support. Altogether, our work represents a system for studying endogenous EV function in vivo with high spatiotemporal accuracy, demonstrating functional inter-organ communication by exosomes.
Collapse
Affiliation(s)
- Frederik J Verweij
- Institut Curie, PSL Research University, CNRS UMR144, Paris 75005, France; Institute for Psychiatry and Neuroscience Paris, Hopital Saint-Anne, Université Descartes, INSERM U894, Paris 75014, France.
| | - Celine Revenu
- Institut Curie, PSL Research University, INSERM U934, CNRS UMR3215, Sorbonne Université, Paris 75005, France
| | - Guillaume Arras
- Institut Curie, PSL Research University, Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, Paris, France
| | - Florent Dingli
- Institut Curie, PSL Research University, Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, Paris, France
| | - Damarys Loew
- Institut Curie, PSL Research University, Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, Paris, France
| | - D Michiel Pegtel
- Department of Pathology, Cancer Center Amsterdam, the Netherlands
| | - Gautier Follain
- INSERM UMR_S1109, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg, France
| | - Guillaume Allio
- INSERM UMR_S1109, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg, France
| | - Jacky G Goetz
- INSERM UMR_S1109, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg, France
| | - Pascale Zimmermann
- Centre de Recherche en Cancérologie de Marseille, Aix-Marseille Université, Marseille 13284, France
| | - Philippe Herbomel
- Institut Pasteur, Department of Developmental & Stem Cell Biology, 25 rue du Dr Roux, Paris 75015, France
| | - Filippo Del Bene
- Institut Curie, PSL Research University, INSERM U934, CNRS UMR3215, Sorbonne Université, Paris 75005, France
| | - Graça Raposo
- Institut Curie, PSL Research University, CNRS UMR144, Paris 75005, France
| | - Guillaume van Niel
- Institut Curie, PSL Research University, CNRS UMR144, Paris 75005, France; Institute for Psychiatry and Neuroscience Paris, Hopital Saint-Anne, Université Descartes, INSERM U894, Paris 75014, France.
| |
Collapse
|
184
|
Moore JB, Tang XL, Zhao J, Fischer AG, Wu WJ, Uchida S, Gumpert AM, Stowers H, Wysoczynski M, Bolli R. Epigenetically modified cardiac mesenchymal stromal cells limit myocardial fibrosis and promote functional recovery in a model of chronic ischemic cardiomyopathy. Basic Res Cardiol 2018; 114:3. [PMID: 30446837 PMCID: PMC6335654 DOI: 10.1007/s00395-018-0710-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 10/31/2018] [Indexed: 01/01/2023]
Abstract
Preclinical investigations support the concept that donor cells more oriented towards a cardiovascular phenotype favor repair. In light of this philosophy, we previously identified HDAC1 as a mediator of cardiac mesenchymal cell (CMC) cardiomyogenic lineage commitment and paracrine signaling potency in vitro-suggesting HDAC1 as a potential therapeutically exploitable target to enhance CMC cardiac reparative capacity. In the current study, we examined the effects of pharmacologic HDAC1 inhibition, using the benzamide class 1 isoform-selective HDAC inhibitor entinostat (MS-275), on CMC cardiomyogenic lineage commitment and CMC-mediated myocardial repair in vivo. Human CMCs pre-treated with entinostat or DMSO diluent control were delivered intramyocardially in an athymic nude rat model of chronic ischemic cardiomyopathy 30 days after a reperfused myocardial infarction. Indices of cardiac function were assessed by echocardiography and left ventricular (LV) Millar conductance catheterization 35 days after treatment. Compared with naïve CMCs, entinostat-treated CMCs exhibited heightened capacity for myocyte-like differentiation in vitro and superior ability to attenuate LV remodeling and systolic dysfunction in vivo. The improvement in CMC therapeutic efficacy observed with entinostat pre-treatment was not associated with enhanced donor cell engraftment, cardiomyogenesis, or vasculogenesis, but instead with more efficient inhibition of myocardial fibrosis and greater increase in myocyte size. These results suggest that HDAC inhibition enhances the reparative capacity of CMCs, likely via a paracrine mechanism that improves ventricular compliance and contraction and augments myocyte growth and function.
Collapse
Affiliation(s)
- Joseph B Moore
- Institute of Molecular Cardiology, Division of Cardiovascular Medicine, University of Louisville, 580 S. Preston Street, Louisville, KY, 40292, USA.
| | - Xian-Liang Tang
- Institute of Molecular Cardiology, Division of Cardiovascular Medicine, University of Louisville, 580 S. Preston Street, Louisville, KY, 40292, USA
| | - John Zhao
- Institute of Molecular Cardiology, Division of Cardiovascular Medicine, University of Louisville, 580 S. Preston Street, Louisville, KY, 40292, USA
| | - Annalara G Fischer
- Institute of Molecular Cardiology, Division of Cardiovascular Medicine, University of Louisville, 580 S. Preston Street, Louisville, KY, 40292, USA
| | - Wen-Jian Wu
- Institute of Molecular Cardiology, Division of Cardiovascular Medicine, University of Louisville, 580 S. Preston Street, Louisville, KY, 40292, USA
| | - Shizuka Uchida
- Department of Medicine, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA
| | - Anna M Gumpert
- Institute of Molecular Cardiology, Division of Cardiovascular Medicine, University of Louisville, 580 S. Preston Street, Louisville, KY, 40292, USA
| | - Heather Stowers
- Institute of Molecular Cardiology, Division of Cardiovascular Medicine, University of Louisville, 580 S. Preston Street, Louisville, KY, 40292, USA
| | - Marcin Wysoczynski
- Institute of Molecular Cardiology, Division of Cardiovascular Medicine, University of Louisville, 580 S. Preston Street, Louisville, KY, 40292, USA
| | - Roberto Bolli
- Institute of Molecular Cardiology, Division of Cardiovascular Medicine, University of Louisville, 580 S. Preston Street, Louisville, KY, 40292, USA
| |
Collapse
|
185
|
Charoenviriyakul C, Takahashi Y, Nishikawa M, Takakura Y. Preservation of exosomes at room temperature using lyophilization. Int J Pharm 2018; 553:1-7. [PMID: 30316791 DOI: 10.1016/j.ijpharm.2018.10.032] [Citation(s) in RCA: 162] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 10/09/2018] [Accepted: 10/10/2018] [Indexed: 01/08/2023]
Abstract
The application of exosomes as a therapeutic reagent or drug delivery vehicle can be expanded by developing a method to preserve exosomes. Although exosomes are generally stored at -80 °C, this temperature is not suitable for their handling or transportation and, therefore, other storage methods are desirable. Lyophilization is a promising storage method that can be used to preserve various substances at room temperature. In this study, we sought to develop a room temperature preservation method for exosomes using lyophilization and compared the properties of the lyophilized exosomes with ones stored at -80 °C. Lyophilization without cryoprotectant resulted in the aggregation of B16BL6 melanoma-derived exosomes, while the addition of trehalose, a cryoprotectant, prevented aggregation during lyophilization. PAGE analysis revealed that the proteins and RNA of exosomes were protected following lyophilization in the presence of trehalose. Lyophilization had little effect on the pharmacokinetics of Gaussia luciferase (gLuc)-labeled exosomes after an intravenous injection into mice. Moreover, it was found that lyophilized exosomes retained the activity of loaded gLuc and immunostimulatory CpG DNA for approximately 4 weeks even when stored at 25 °C. In conclusion, lyophilization with trehalose is an effective method for the storage of exosomes for various applications.
Collapse
Affiliation(s)
- Chonlada Charoenviriyakul
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yuki Takahashi
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan.
| | - Makiya Nishikawa
- Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Yoshinobu Takakura
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
186
|
Yamashita T, Takahashi Y, Takakura Y. Possibility of Exosome-Based Therapeutics and Challenges in Production of Exosomes Eligible for Therapeutic Application. Biol Pharm Bull 2018; 41:835-842. [PMID: 29863072 DOI: 10.1248/bpb.b18-00133] [Citation(s) in RCA: 207] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Exosomes are cell-derived vesicles with a diameter 30-120 nm. Exosomes contain endogenous proteins and nucleic acids; delivery of these molecules to exosome-recipient cells causes biological effects. Exosomes derived from some types of cells such as mesenchymal stem cells and dendritic cells have therapeutic potential and may be biocompatible and efficient agents against various disorders such as organ injury. However, there are many challenges for the development of exosome-based therapeutics. In particular, producing exosomal formulations is the major barrier for therapeutic application because of their heterogeneity and low productivity. Development and optimization of producing methods, including methods for isolation and storage of exosome formulations, are required for realizing exosome-based therapeutics. In addition, improvement of therapeutic potential and delivery efficiency of exosomes are important for their therapeutic application. In this review, we summarize current knowledge about therapeutic application of exosomes and discuss some challenges in their successful use.
Collapse
Affiliation(s)
| | - Yuki Takahashi
- Graduate School of Pharmaceutical Sciences, Kyoto University
| | | |
Collapse
|
187
|
Record M, Silvente-Poirot S, Poirot M, Wakelam MJO. Extracellular vesicles: lipids as key components of their biogenesis and functions. J Lipid Res 2018; 59:1316-1324. [PMID: 29764923 PMCID: PMC6071772 DOI: 10.1194/jlr.e086173] [Citation(s) in RCA: 207] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Indexed: 12/15/2022] Open
Abstract
Intercellular communication has been known for decades to involve either direct contact between cells or to operate via circulating molecules, such as cytokines, growth factors, or lipid mediators. During the last decade, we have begun to appreciate the increasing importance of intercellular communication mediated by extracellular vesicles released by viable cells either from plasma membrane shedding (microvesicles, also named microparticles) or from an intracellular compartment (exosomes). Exosomes and microvesicles circulate in all biological fluids and can trigger biological responses at a distance. Their effects include a large variety of biological processes, such as immune surveillance, modification of tumor microenvironment, or regulation of inflammation. Extracellular vesicles can carry a large array of active molecules, including lipid mediators, such as eicosanoids, proteins, and nucleic acids, able to modify the phenotype of receiving cells. This review will highlight the role of the various lipidic pathways involved in the biogenesis and functions of microvesicles and exosomes.
Collapse
Affiliation(s)
- Michel Record
- UMR INSERM 1037-CRCT (Cancer Research Center of Toulouse), University of Toulouse III Paul Sabatier, Team "Cholesterol Metabolism and Therapeutic Innovations," Toulouse, France
| | - Sandrine Silvente-Poirot
- UMR INSERM 1037-CRCT (Cancer Research Center of Toulouse), University of Toulouse III Paul Sabatier, Team "Cholesterol Metabolism and Therapeutic Innovations," Toulouse, France
| | - Marc Poirot
- UMR INSERM 1037-CRCT (Cancer Research Center of Toulouse), University of Toulouse III Paul Sabatier, Team "Cholesterol Metabolism and Therapeutic Innovations," Toulouse, France
| | | |
Collapse
|
188
|
Obata Y, Kita S, Koyama Y, Fukuda S, Takeda H, Takahashi M, Fujishima Y, Nagao H, Masuda S, Tanaka Y, Nakamura Y, Nishizawa H, Funahashi T, Ranscht B, Izumi Y, Bamba T, Fukusaki E, Hanayama R, Shimada S, Maeda N, Shimomura I. Adiponectin/T-cadherin system enhances exosome biogenesis and decreases cellular ceramides by exosomal release. JCI Insight 2018; 3:99680. [PMID: 29669945 DOI: 10.1172/jci.insight.99680] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 03/14/2018] [Indexed: 12/15/2022] Open
Abstract
Adiponectin, an adipocyte-derived circulating protein, accumulates in vasculature, heart, and skeletal muscles through interaction with a unique glycosylphosphatidylinositol-anchored cadherin, T-cadherin. Recent studies have demonstrated that such accumulation is essential for adiponectin-mediated cardiovascular protection. Here, we demonstrate that the adiponectin/T-cadherin system enhances exosome biogenesis and secretion, leading to the decrease of cellular ceramides. Adiponectin accumulated inside multivesicular bodies, the site of exosome generation, in cultured cells and in vivo aorta, and also in exosomes in conditioned media and in blood, together with T-cadherin. The systemic level of exosomes in blood was significantly affected by adiponectin or T-cadherin in vivo. Adiponectin increased exosome biogenesis from the cells, dependently on T-cadherin, but not on AdipoR1 or AdipoR2. Such enhancement of exosome release accompanied the reduction of cellular ceramides through ceramide efflux in exosomes. Consistently, the ceramide reduction by adiponectin was found in aortas of WT mice treated with angiotensin II, but not in T-cadherin-knockout mice. Our findings provide insights into adiponectin/T-cadherin-mediated organ protection through exosome biogenesis and secretion.
Collapse
Affiliation(s)
| | - Shunbun Kita
- Department of Metabolic Medicine.,Department of Adipose Management, and
| | - Yoshihisa Koyama
- Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | | | - Hiroaki Takeda
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Masatomo Takahashi
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | - Tohru Funahashi
- Department of Metabolic Medicine.,Department of Metabolism and Atherosclerosis, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Barbara Ranscht
- Sanford Burnham Prebys Medical Discovery Institute, NIH-designated Cancer Center, Development, Aging and Regeneration Program, La Jolla, California, USA
| | - Yoshihiro Izumi
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Takeshi Bamba
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Eiichiro Fukusaki
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Rikinari Hanayama
- Department of Immunology, Kanazawa University Graduate School of Medical Sciences, Ishikawa, Japan
| | - Shoichi Shimada
- Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Norikazu Maeda
- Department of Metabolic Medicine.,Department of Metabolism and Atherosclerosis, Graduate School of Medicine, Osaka University, Osaka, Japan
| | | |
Collapse
|
189
|
Mentkowski KI, Snitzer JD, Rusnak S, Lang JK. Therapeutic Potential of Engineered Extracellular Vesicles. AAPS JOURNAL 2018; 20:50. [PMID: 29546642 PMCID: PMC8299397 DOI: 10.1208/s12248-018-0211-z] [Citation(s) in RCA: 143] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 02/27/2018] [Indexed: 12/16/2022]
Abstract
Extracellular vesicles (EVs) comprise a heterogeneous group of small membrane vesicles, including exosomes, which play a critical role in intracellular communication and regulation of numerous physiological processes in health and disease. Naturally released from virtually all cells, these vesicles contain an array of nucleic acids, lipids and proteins which they transfer to target cells within their local milieu and systemically. They have been proposed as a means of “cell-free, cell therapy” for cancer, immune disorders, and more recently cardiovascular disease. In addition, their unique properties of stability, biocompatibility, and low immunogenicity have prompted research into their potential as therapeutic delivery agents for drugs and small molecules. In this review, we aim to provide a comprehensive overview of the current understanding of extracellular vesicle biology as well as engineering strategies in play to improve their therapeutic potential.
Collapse
Affiliation(s)
- Kyle I Mentkowski
- Department of Medicine, Division of Cardiology, Jacobs School of Medicine and Biomedical Sciences, Clinical and Translational Research Center, 895 Ellicott Street, Buffalo, NY, 14203, USA
| | - Jonathan D Snitzer
- Department of Medicine, Division of Cardiology, Jacobs School of Medicine and Biomedical Sciences, Clinical and Translational Research Center, 895 Ellicott Street, Buffalo, NY, 14203, USA
| | - Sarah Rusnak
- Department of Medicine, Division of Cardiology, Jacobs School of Medicine and Biomedical Sciences, Clinical and Translational Research Center, 895 Ellicott Street, Buffalo, NY, 14203, USA
| | - Jennifer K Lang
- Department of Medicine, Division of Cardiology, Jacobs School of Medicine and Biomedical Sciences, Clinical and Translational Research Center, 895 Ellicott Street, Buffalo, NY, 14203, USA.
| |
Collapse
|
190
|
Gangadaran P, Hong CM, Ahn BC. An Update on in Vivo Imaging of Extracellular Vesicles as Drug Delivery Vehicles. Front Pharmacol 2018; 9:169. [PMID: 29541030 PMCID: PMC5835830 DOI: 10.3389/fphar.2018.00169] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 02/15/2018] [Indexed: 12/21/2022] Open
Abstract
Extracellular vesicles (EVs) are currently being considered as promising drug delivery vehicles. EVs are naturally occurring vesicles that exhibit many characteristics favorable to serve as drug delivery vehicles. In addition, EVs have inherent properties for treatment of cancers and other diseases. For research and clinical translation of use of EVs as drug delivery vehicles, in vivo tracking of EVs is essential. The latest molecular imaging techniques enable the tracking of EVs in living animals. However, each molecular imaging technique has its certain advantages and limitations for the in vivo imaging of EVs; therefore, understanding the molecular imaging techniques is essential to select the most appropriate imaging technology to achieve the desired imaging goal. In this review, we summarize the characteristics of EVs as drug delivery vehicles and the molecular imaging techniques used in visualizing and monitoring EVs in in vivo environments. Furthermore, we provide a perceptual vision of EVs as drug delivery vehicles and in vivo monitoring of EVs using molecular imaging technologies.
Collapse
Affiliation(s)
| | | | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University and Hospital, Daegu, South Korea
| |
Collapse
|
191
|
Exosomes: Outlook for Future Cell-Free Cardiovascular Disease Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 998:285-307. [PMID: 28936747 DOI: 10.1007/978-981-10-4397-0_19] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cardiovascular diseases are the number one cause of death globally with an estimated 7.4 million people dying from coronary heart disease. Studies have been conducted to identify the therapeutic utility of exosomes in many diseases, including cardiovascular diseases. It has been demonstrated that exosomes are immune modulators, can be used to treat cardiac ischemic injury, pulmonary hypertension and many other diseases, including cancers. Exosomes can be used as a biomarker for disease and cell-free drug delivery system for targeting the cells. Many studies suggest that exosomes can be used as a cell-free vaccine for many diseases. In this chapter, we explore the possibility of future therapeutic potential of exosomes in various cardiovascular diseases.
Collapse
|
192
|
Charoenviriyakul C, Takahashi Y, Morishita M, Nishikawa M, Takakura Y. Role of Extracellular Vesicle Surface Proteins in the Pharmacokinetics of Extracellular Vesicles. Mol Pharm 2018; 15:1073-1080. [PMID: 29382201 DOI: 10.1021/acs.molpharmaceut.7b00950] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Extracellular vesicles (EVs) are small membrane vesicles secreted from cells and have great potential as drug delivery carriers. Surface proteins on EV membranes might play roles in pharmacokinetics. One method which can be used to study the role of surface membrane of EV is to modify the inner space of EV. In the present study, we constructed a plasmid DNA expressing a fusion protein of Gag protein derived from Moloney murine leukemia virus (Gag) and Gaussia luciferase (gLuc) (Gag-gLuc) to modify the inner space of EVs. EVs were collected from B16BL6 melanoma cells, transfected with the plasmid, and isolated by a differential ultracentrifugation method. Gag-gLuc EVs were negatively charged globular vesicles with a diameter of approximately 100 nm. gLuc labeling of the Gag-gLuc EVs was stable in serum. gLuc activity of Gag-gLuc EVs was minimally decreased by proteinase K (ProK) treatment, indicating that gLuc was modified in the inner space of EV. Then, to evaluate the effect of the surface proteins of EVs on their pharmacokinetics, Gag-gLuc EVs treated with ProK were intravenously administered to mice. Volume of distribution (Vd) was significantly smaller for treated EVs than untreated EVs. Moreover, integrin α6β1, an integrin known to be involved in lung targeting, was degraded after ProK treatment. The ProK treatment significantly reduced the lung distribution of EVs after intravenous injection. These results indicate that the surface proteins of EVs such as integrin α6β1 play some roles in pharmacokinetics in terms of reducing Vd and their distribution to the lung.
Collapse
Affiliation(s)
- Chonlada Charoenviriyakul
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences , Kyoto University , Sakyo-ku , Kyoto 606-8501 , Japan
| | - Yuki Takahashi
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences , Kyoto University , Sakyo-ku , Kyoto 606-8501 , Japan
| | - Masaki Morishita
- Department of Biopharmaceutics , Kyoto Pharmaceutical University , Misasagi, Yamashina-ku , Kyoto 607-8414 , Japan
| | - Makiya Nishikawa
- Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences , Tokyo University of Science , 2641 Yamazaki , Noda , Chiba 278-8510 , Japan
| | - Yoshinobu Takakura
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences , Kyoto University , Sakyo-ku , Kyoto 606-8501 , Japan
| |
Collapse
|
193
|
Borrelli DA, Yankson K, Shukla N, Vilanilam G, Ticer T, Wolfram J. Extracellular vesicle therapeutics for liver disease. J Control Release 2018; 273:86-98. [PMID: 29373816 DOI: 10.1016/j.jconrel.2018.01.022] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 01/19/2018] [Accepted: 01/22/2018] [Indexed: 12/18/2022]
Abstract
Extracellular vesicles (EVs) are endogenous nanoparticles that play important roles in intercellular communication. Unmodified and engineered EVs can be utilized for therapeutic purposes. For instance, mesenchymal stem cell (MSC)-derived EVs have shown promise for tissue repair, while drug-loaded EVs have the potential to be used for cancer treatment. The liver is an ideal target for EV therapy due to the intrinsic regenerative capacity of hepatic tissue and the tropism of systemically injected nanovesicles for this organ. This review will give an overview of the potential of EV therapeutics in liver disease. Specifically, the mechanisms by which MSC-EVs induce liver repair will be covered. Moreover, the use of drug-loaded EVs for the treatment of hepatocellular carcinoma will also be discussed. Although there are several challenges associated with the clinical translation of EVs, these biological nanoparticles represent a promising new therapeutic modality for liver disease.
Collapse
Affiliation(s)
- David A Borrelli
- Department of Transplantation, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Kiera Yankson
- Department of Transplantation, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Neha Shukla
- Department of Transplantation, Mayo Clinic, Jacksonville, FL 32224, USA
| | - George Vilanilam
- Department of Transplantation, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Taylor Ticer
- Department of Transplantation, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Joy Wolfram
- Department of Transplantation, Mayo Clinic, Jacksonville, FL 32224, USA; Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Biology, University of North Florida, Jacksonville, FL 32224, USA; Wenzhou Institute of Biomaterials and Engineering, Ningbo Institute of Industrial Technology, Chinese Academy of Sciences, Wenzhou, China.
| |
Collapse
|
194
|
Gheytanchi E, Madjd Z, Janani L, Rasti A, Ghods R, Atyabi F, Asadi-Lari MH, Babashah S. Exosomal microRNAs as potential circulating biomarkers in gastrointestinal tract cancers: a systematic review protocol. Syst Rev 2017; 6:228. [PMID: 29149908 PMCID: PMC5693515 DOI: 10.1186/s13643-017-0624-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Accepted: 11/08/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Metastasis is the most frequent type of recurrence in gastrointestinal (GI) cancers, and there is an emerging potential for new diagnostic and therapeutic approaches, especially in the cases of metastatic GI carcinomas. The expression profiles of circulating exosomal microRNAs are of particular interest as novel non-invasive diagnostic and prognostic biomarkers for improved detection of GI cancers in body fluids, especially in the serum of patients with recurrent cancers. The aim of this study is to systematically review primary studies and identify the miRNA profiles of serum exosomes of GI cancers. METHODS AND DESIGN This systematic review will be reported in line with the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidance. Relevant studies will be identified through a comprehensive search of the following main electronic databases: PubMed, Web of Science, Embase, Scopus, and Google Scholar, with no language restrictions (up to July 2017). Full copies of articles will be identified by a defined search strategy and will be considered for inclusion against pre-defined criteria. The quality assessment of the included studies will be performed by the Newcastle-Ottawa Scale (NOS). Data will be analyzed using Stata software V.12. Publication bias will be assessed by funnel plots, Beggs' and Eggers' tests. The levels of evidence for primary outcomes will be evaluated using the GRADE criteria. DISCUSSION The analysis of circulating exosomal miRNA profiles provides attractive screening and non-invasive diagnostic tools for the majority of solid tumors including GI cancers. There is limited information regarding the relationship between serum exosomal miRNA profiles and the pathological condition of patients with different GI cancers. Since there is no specific biomarker for GI cancers, we aim to suggest a number of circulating exosomal miRNA candidates as potential multifaceted GI cancer biomarkers for clinical utility. SYSTEMATIC REVIEW REGISTRATION PROSPERO CRD42017057129.
Collapse
Affiliation(s)
- Elmira Gheytanchi
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Madjd
- Oncopathology Research Center, Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Leila Janani
- Department of Biostatistics, School of Public Health, Iran University of Medical Sciences, Tehran, Iran.
| | - Arezoo Rasti
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Roya Ghods
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Atyabi
- Department of Pharmaceutics, Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Asadi-Lari
- Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia (UBC), Vancouver, BC, Canada
| | - Sadegh Babashah
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
195
|
Moore JB, Zhao J, Fischer AG, Keith MCL, Hagan D, Wysoczynski M, Bolli R. Histone Deacetylase 1 Depletion Activates Human Cardiac Mesenchymal Stromal Cell Proangiogenic Paracrine Signaling Through a Mechanism Requiring Enhanced Basic Fibroblast Growth Factor Synthesis and Secretion. J Am Heart Assoc 2017; 6:JAHA.117.006183. [PMID: 28679560 PMCID: PMC5586316 DOI: 10.1161/jaha.117.006183] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND Cardiac mesenchymal cell (CMC) administration improves cardiac function in animal models of heart failure. Although the precise mechanisms remain unclear, transdifferentiation and paracrine signaling are suggested to underlie their cardiac reparative effects. We have shown that histone deacetylase 1 (HDAC1) inhibition enhances CMC cardiomyogenic lineage commitment. Here, we investigated the impact of HDAC1 on CMC cytokine secretion and associated paracrine-mediated activities on endothelial cell function. METHODS AND RESULTS CMCs were transduced with shRNA constructs targeting HDAC1 (shHDAC1) or nontarget (shNT) control. Cytokine arrays were used to assess the expression of secreted proteins in conditioned medium (CM) from shHDAC1 or shNT-transduced CMCs. In vitro functional assays for cell proliferation, protection from oxidative stress, cell migration, and tube formation were performed on human endothelial cells incubated with CM from the various treatment conditions. CM from shHDAC1-transduced CMCs contained more cytokines involved in cell growth/differentiation and more efficiently promoted endothelial cell proliferation and tube formation compared with CM from shNT. After evaluating key cytokines previously implicated in cell-therapy-mediated cardiac repair, we found that basic fibroblast growth factor was significantly upregulated in shHDAC1-transduced CMCs. Furthermore, shRNA-mediated knockdown of basic fibroblast growth factor in HDAC1-depleted CMCs inhibited the effects of shHDAC1 CM in promoting endothelial proliferation and tube formation-indicating that HDAC1 depletion activates CMC proangiogenic paracrine signaling in a basic fibroblast growth factor-dependent manner. CONCLUSIONS These results reveal a hitherto unknown role for HDAC1 in the modulation of CMC cytokine secretion and implicate the targeted inhibition of HDAC1 in CMCs as a means to enhance paracrine-mediated neovascularization in cardiac cell therapy applications.
Collapse
Affiliation(s)
- Joseph B Moore
- Institute of Molecular Cardiology, University of Louisville, KY
| | - John Zhao
- Institute of Molecular Cardiology, University of Louisville, KY
| | | | | | - David Hagan
- Institute of Molecular Cardiology, University of Louisville, KY
| | | | - Roberto Bolli
- Institute of Molecular Cardiology, University of Louisville, KY
| |
Collapse
|
196
|
Morishita M, Takahashi Y, Nishikawa M, Takakura Y. Pharmacokinetics of Exosomes-An Important Factor for Elucidating the Biological Roles of Exosomes and for the Development of Exosome-Based Therapeutics. J Pharm Sci 2017; 106:2265-2269. [PMID: 28283433 DOI: 10.1016/j.xphs.2017.02.030] [Citation(s) in RCA: 167] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 02/28/2017] [Indexed: 12/14/2022]
Abstract
Exosomes are small membrane vesicles containing lipids, proteins, and nucleic acids. Recently, researchers have uncovered that exosomes are involved in various biological events, such as tumor growth, metastasis, and the immune response, by delivering their cargos to exosome-receiving cells. Moreover, exosomes are expected to be used in therapeutic treatments, such as tissue regeneration therapy and antitumor immunotherapy, because exosomes are effective delivery vehicles for proteins, nucleic acids, and other bioactive compounds. To elucidate the biological functions of exosomes, and for the development of exosome-based therapeutics, the pharmacokinetics of exosomes is important. In this review, we aim to summarize current knowledge about the pharmacokinetics and biodistribution of exosomes. The pharmacokinetics of exogenously administered exosomes is discussed based on the tissue distribution, types of cells taking up exosomes, and key molecules in the pharmacokinetics of exosomes. In addition, recent progress in the methods to control the pharmacokinetics of exosomes is reviewed.
Collapse
Affiliation(s)
- Masaki Morishita
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Yuki Takahashi
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Japan.
| | - Makiya Nishikawa
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Yoshinobu Takakura
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Japan
| |
Collapse
|
197
|
Stranford DM, Leonard JN. Delivery of Biomolecules via Extracellular Vesicles. ADVANCES IN GENETICS 2017; 98:155-175. [DOI: 10.1016/bs.adgen.2017.08.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|