251
|
Huang T, Banizs AB, Shi W, Klibanov AL, He J. Size Exclusion HPLC Detection of Small-Size Impurities as a Complementary Means for Quality Analysis of Extracellular Vesicles. J Circ Biomark 2015; 4:6. [PMID: 28936242 PMCID: PMC5572986 DOI: 10.5772/61148] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 06/18/2015] [Indexed: 12/21/2022] Open
Abstract
For extracellular vesicle research, whether for biomarker discoveries or therapeutic applications, it is critical to have high-quality samples. Both microscopy and NanoSight Tracking Analysis (NTA) for size distribution have been used to detect large vesicles. However, there is currently no well-established method that is convenient for routine quality analysis of small-size impurities in vesicle samples. In this paper we report a convenient method, called 'size-exclusion high-performance liquid chromatography' (SE-HPLC), alongside NTA and Microscopy analysis to guide and qualify the isolation and processing of vesicles. First, the SE-HPLC analysis was used to detect impurities of small-size proteins during the ultra-centrifugation process of vesicle isolation; it was then employed to test the changes of vesicles under different pH conditions or integrity after storage. As SE-HPLC is generally accessible in most institutions, it could be used as a routine means to assist researchers in examining the integrity and quality of extracellular vesicles along with other techniques either during isolation/preparation or for further engineering and storage.
Collapse
Affiliation(s)
- Tao Huang
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, USA
| | - Anna B Banizs
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, USA
| | - Weibin Shi
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, USA
| | | | - Jiang He
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
252
|
Nakase I, Kobayashi NB, Takatani-Nakase T, Yoshida T. Active macropinocytosis induction by stimulation of epidermal growth factor receptor and oncogenic Ras expression potentiates cellular uptake efficacy of exosomes. Sci Rep 2015; 5:10300. [PMID: 26036864 PMCID: PMC4453128 DOI: 10.1038/srep10300] [Citation(s) in RCA: 205] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 04/08/2015] [Indexed: 12/18/2022] Open
Abstract
Exosomes are approximately 100-nm vesicles that consist of a lipid bilayer of cellular membranes secreted in large quantities from various types of normal and disease-related cells. Endocytosis has been reported as a major pathway for the cellular uptake of exosomes; however, the detailed mechanisms of their cellular uptake are still unknown. Here, we demonstrate the active induction of macropinocytosis (accompanied by actin reorganisation, ruffling of plasma membrane, and engulfment of large volumes of extracellular fluid) by stimulation of cancer-related receptors and show that the epidermal growth factor (EGF) receptor significantly enhances the cellular uptake of exosomes. We also demonstrate that oncogenic K-Ras-expressing MIA PaCa-2 cells exhibit intensive macropinocytosis that actively transports extracellular exosomes into the cells compared with wild-type K-Ras-expressing BxPC-3 cells. Furthermore, encapsulation of the ribosome-inactivating protein saporin with EGF in exosomes using our simple electroporation method produces superior cytotoxicity via the enhanced cellular uptake of exosomes. Our findings contribute to the biological, pharmaceutical, and medical research fields in terms of understanding the macropinocytosis-mediated cellular uptake of exosomes with applications for exosomal delivery systems.
Collapse
Affiliation(s)
- Ikuhiko Nakase
- Nanoscience and Nanotechnology Research Center, Research Organization for the 21st Century, Osaka Prefecture University, Naka-ku, Sakai, Osaka 599-8570, Japan
| | - Nahoko Bailey Kobayashi
- 1] Keio Advanced Research Centers (KARC), Keio University, Tsukuba, Ibaraki 300-2611, Japan [2] Institute for Advanced Sciences, Toagosei Co., Ltd., Tsukuba, Ibaraki 300-2611, Japan
| | - Tomoka Takatani-Nakase
- Department of Pharmaceutics, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University, 11-68, Koshien Kyuban-cho, Nishinomiya, Hyogo 663-8179, Japan
| | - Tetsuhiko Yoshida
- 1] Keio Advanced Research Centers (KARC), Keio University, Tsukuba, Ibaraki 300-2611, Japan [2] Institute for Advanced Sciences, Toagosei Co., Ltd., Tsukuba, Ibaraki 300-2611, Japan
| |
Collapse
|
253
|
Kutralam-Muniasamy G, Flores-Cotera LB, Perez-Guevara F. Potential of yeast secretory vesicles in biodelivery systems. Drug Discov Today 2015; 20:659-66. [DOI: 10.1016/j.drudis.2015.03.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 02/24/2015] [Accepted: 03/25/2015] [Indexed: 10/23/2022]
|
254
|
Fuhrmann G, Herrmann IK, Stevens MM. Cell-derived vesicles for drug therapy and diagnostics: opportunities and challenges. NANO TODAY 2015; 10:397-409. [PMID: 28458718 PMCID: PMC5409525 DOI: 10.1016/j.nantod.2015.04.004] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Extracellular vesicles are small lipid-based membrane-bound entities shed by cells under both physiological and pathological conditions. Their discovery as intercellular communicators through transfer of nucleic acid- and protein-based cargos between cells locally and at distance in a highly specific manner has created recent excitement. The information they transport and their composition may vary depending on the cell of origin as well as the eliciting stimulus. Such sensitive changes in vesicle characteristics hold significant promise for the improved diagnosis of pathological conditions, including infections and neoplastic lesions in a minimally invasive way. Similarly, these cell-derived vesicles exhibit promising characteristics that could enhance drug targeting efficiencies. Recent developments in the field have aimed at studying EVs as novel drug carriers due to their natural composition, biological function and selective cell interaction. In this review, we discuss new research avenues in diagnostics and drug therapy based on extracellular vesicles. We show how cell-derived vesicles can be harvested and engineered to meet application-specific design requirements. We finally discuss potential risks encountered when translating extracellular vesicle based approaches into (pre)clinical applications.
Collapse
Affiliation(s)
- Gregor Fuhrmann
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, Prince Consort Road, SW7 2AZ London, United Kingdom
| | - Inge K. Herrmann
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, Prince Consort Road, SW7 2AZ London, United Kingdom
| | - Molly M. Stevens
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, Prince Consort Road, SW7 2AZ London, United Kingdom
| |
Collapse
|
255
|
Nakase I, Futaki S. Combined treatment with a pH-sensitive fusogenic peptide and cationic lipids achieves enhanced cytosolic delivery of exosomes. Sci Rep 2015; 5:10112. [PMID: 26011176 PMCID: PMC4443764 DOI: 10.1038/srep10112] [Citation(s) in RCA: 203] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 03/27/2015] [Indexed: 12/19/2022] Open
Abstract
Exosomes, which are approximately 100 nm vesicles secreted by cells, have been studied with respect to cell-to-cell communication, disease diagnosis, and intracellular delivery. The cellular uptake of exosomes occurs by endocytosis; however, the cytosolic release efficiency of encapsulated molecules inside cells is low. To address this issue, here we demonstrate a simple technique for enhancing the cellular uptake and cytosolic release of exosomes by combining a pH-sensitive fusogenic peptide for the fusion of endosomal and exosomal membranes inside cells. This method stimulates the efficient cytosolic release of the exosomal contents with cationic lipids that act as a “glue” to support cellular uptake. Using this simple combined technique, the effective cellular uptake and cytosolic release of an artificially encapsulated dextran macromolecule (70 kDa) in exosomes are achieved, and a marked improvement in bioactivity is attained with the artificially encapsulated ribosome-inactivating protein saporin. Our method will contribute to many biological research fields, including the assessment of the activities of exosomal contents and the development of candidate tools enabling intracellular visualisation and cellular regulation for future therapeutic applications.
Collapse
Affiliation(s)
- Ikuhiko Nakase
- Nanoscience and Nanotechnology Research Center, Research Organization for the 21st Century, Osaka Prefecture University, Naka-ku, Sakai, Osaka 599-8570, Japan
| | - Shiroh Futaki
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| |
Collapse
|
256
|
Fuhrmann G, Serio A, Mazo M, Nair R, Stevens MM. Active loading into extracellular vesicles significantly improves the cellular uptake and photodynamic effect of porphyrins. J Control Release 2015; 205:35-44. [DOI: 10.1016/j.jconrel.2014.11.029] [Citation(s) in RCA: 337] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 11/21/2014] [Accepted: 11/24/2014] [Indexed: 12/24/2022]
|
257
|
Jinnin M. Recent progress in studies of miRNA and skin diseases. J Dermatol 2015; 42:551-8. [PMID: 25917002 DOI: 10.1111/1346-8138.12904] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 03/12/2015] [Indexed: 01/05/2023]
Abstract
miRNA is a family of small non-coding RNA that consists of 22 nucleotides on average. miRNA are implicated in various cellular activities such as cell proliferation or migration via the modulation of gene expression, and also are linked to the pathogenesis of human diseases. This paper reviews recent research progress about the contribution of miRNA to the pathogenesis of various skin diseases, and possible application of miRNA as the disease markers in each disease. For example, downregulated miR-424-5p in psoriatic skin causes the overexpression of MEK1 and cyclin E1 in psoriatic keratinocytes, resulting in the keratinocyte overgrowth and hyperproliferation seen in the disease. Although there was no significant difference in the serum miR-424-5p levels between psoriasis patients and healthy controls, serum miR-1266-5p levels were significantly upregulated in psoriasis patients, and showed weak and inverse correlation with disease activity. Furthermore, combination of serum levels of miR-146a-5p and -203a-3p was more reliable to distinguish psoriasis patients and normal subjects, than each miRNA alone. Hair shaft miR-424-5p levels were significantly higher in psoriasis patients than normal subjects, while hair root miR-19a-3p levels in psoriasis patients were inversely correlated with the duration between symptom onset and the first visit to the hospital. Future researches of miRNA will enable the advances of their clinical applications including the clarification of pathogenesis, disease markers and novel treatments.
Collapse
Affiliation(s)
- Masatoshi Jinnin
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
258
|
Tan S, Wu T, Zhang D, Zhang Z. Cell or cell membrane-based drug delivery systems. Theranostics 2015; 5:863-81. [PMID: 26000058 PMCID: PMC4440443 DOI: 10.7150/thno.11852] [Citation(s) in RCA: 335] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 02/18/2015] [Indexed: 01/14/2023] Open
Abstract
Natural cells have been explored as drug carriers for a long period. They have received growing interest as a promising drug delivery system (DDS) until recently along with the development of biology and medical science. The synthetic materials, either organic or inorganic, are found to be with more or less immunogenicity and/or toxicity. The cells and extracellular vesicles (EVs), are endogenous and thought to be much safer and friendlier. Furthermore, in view of their host attributes, they may achieve different biological effects and/or targeting specificity, which can meet the needs of personalized medicine as the next generation of DDS. In this review, we summarized the recent progress in cell or cell membrane-based DDS and their fabrication processes, unique properties and applications, including the whole cells, EVs and cell membrane coated nanoparticles. We expect the continuing development of this cell or cell membrane-based DDS will promote their clinic applications.
Collapse
Affiliation(s)
- Songwei Tan
- 1. Tongji School of Pharmacy
- 2. National Engineering Research Center for Nanomedicine
- 3. Hubei Engineering Research Center for Novel DDS, Huazhong University of Science and Technology, Wuhan 430030, P R China
| | | | | | - Zhiping Zhang
- 1. Tongji School of Pharmacy
- 2. National Engineering Research Center for Nanomedicine
- 3. Hubei Engineering Research Center for Novel DDS, Huazhong University of Science and Technology, Wuhan 430030, P R China
| |
Collapse
|
259
|
Smith JA, Leonardi T, Huang B, Iraci N, Vega B, Pluchino S. Extracellular vesicles and their synthetic analogues in aging and age-associated brain diseases. Biogerontology 2015; 16:147-85. [PMID: 24973266 PMCID: PMC4578234 DOI: 10.1007/s10522-014-9510-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 06/16/2014] [Indexed: 02/06/2023]
Abstract
Multicellular organisms rely upon diverse and complex intercellular communications networks for a myriad of physiological processes. Disruption of these processes is implicated in the onset and propagation of disease and disorder, including the mechanisms of senescence at both cellular and organismal levels. In recent years, secreted extracellular vesicles (EVs) have been identified as a particularly novel vector by which cell-to-cell communications are enacted. EVs actively and specifically traffic bioactive proteins, nucleic acids, and metabolites between cells at local and systemic levels, modulating cellular responses in a bidirectional manner under both homeostatic and pathological conditions. EVs are being implicated not only in the generic aging process, but also as vehicles of pathology in a number of age-related diseases, including cancer and neurodegenerative and disease. Thus, circulating EVs-or specific EV cargoes-are being utilised as putative biomarkers of disease. On the other hand, EVs, as targeted intercellular shuttles of multipotent bioactive payloads, have demonstrated promising therapeutic properties, which can potentially be modulated and enhanced through cellular engineering. Furthermore, there is considerable interest in employing nanomedicinal approaches to mimic the putative therapeutic properties of EVs by employing synthetic analogues for targeted drug delivery. Herein we describe what is known about the origin and nature of EVs and subsequently review their putative roles in biology and medicine (including the use of synthetic EV analogues), with a particular focus on their role in aging and age-related brain diseases.
Collapse
Affiliation(s)
- J A Smith
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, CB2 0PY, UK
| | | | | | | | | | | |
Collapse
|
260
|
Cocucci E, Meldolesi J. Ectosomes and exosomes: shedding the confusion between extracellular vesicles. Trends Cell Biol 2015; 25:364-72. [PMID: 25683921 DOI: 10.1016/j.tcb.2015.01.004] [Citation(s) in RCA: 1066] [Impact Index Per Article: 106.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 12/29/2014] [Accepted: 01/20/2015] [Indexed: 12/11/2022]
Abstract
Long- and short-distance communication can take multiple forms. Among them are exosomes and ectosomes, extracellular vesicles (EVs) released from the cell to deliver signals to target cells. While most of our understanding of how these vesicles are assembled and work comes from mechanistic studies performed on exosomes, recent studies have begun to shift their focus to ectosomes. Unlike exosomes, which are released on the exocytosis of multivesicular bodies (MVBs), ectosomes are ubiquitous vesicles assembled at and released from the plasma membrane. Here we review the similarities and differences between these two classes of vesicle, suggesting that, despite their considerable differences, the functions of ectosomes may be largely analogous to those of exosomes. Both vesicles appear to be promising targets in the diagnosis and therapy of diseases, especially cancer.
Collapse
Affiliation(s)
- Emanuele Cocucci
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA.
| | - Jacopo Meldolesi
- Vita-Salute San Raffaele University, 20132 Milan, Italy; San Raffaele Scientific Institute, 20132 Milan, Italy.
| |
Collapse
|
261
|
Waiczies S, Lepore S, Sydow K, Drechsler S, Ku MC, Martin C, Lorenz D, Schütz I, Reimann HM, Purfürst B, Dieringer MA, Waiczies H, Dathe M, Pohlmann A, Niendorf T. Anchoring dipalmitoyl phosphoethanolamine to nanoparticles boosts cellular uptake and fluorine-19 magnetic resonance signal. Sci Rep 2015; 5:8427. [PMID: 25673047 PMCID: PMC5389132 DOI: 10.1038/srep08427] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 01/15/2015] [Indexed: 01/19/2023] Open
Abstract
Magnetic resonance (MR) methods to detect and quantify fluorine (19F) nuclei provide the opportunity to study the fate of cellular transplants in vivo. Cells are typically labeled with 19F nanoparticles, introduced into living organisms and tracked by 19F MR methods. Background-free imaging and quantification of cell numbers are amongst the strengths of 19F MR-based cell tracking but challenges pertaining to signal sensitivity and cell detection exist. In this study we aimed to overcome these limitations by manipulating the aminophospholipid composition of 19F nanoparticles in order to promote their uptake by dendritic cells (DCs). As critical components of biological membranes, phosphatidylethanolamines (PE) were studied. Both microscopy and MR spectroscopy methods revealed a striking (at least one order of magnitude) increase in cytoplasmic uptake of 19F nanoparticles in DCs following enrichment with 1,2-dipalmitoyl-sn-glycero-3-phosphoethanolamine (DPPE). The impact of enriching 19F nanoparticles with PE on DC migration was also investigated. By manipulating the nanoparticle composition and as a result the cellular uptake we provide here one way of boosting 19F signal per cell in order to overcome some of the limitations related to 19F MR signal sensitivity. The boost in signal is ultimately necessary to detect and track cells in vivo.
Collapse
Affiliation(s)
- Sonia Waiczies
- Berlin Ultrahigh Field Facility, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Stefano Lepore
- Berlin Ultrahigh Field Facility, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Karl Sydow
- Leibniz-Institut für Molekulare Pharmakologie, Berlin, Germany
| | - Susanne Drechsler
- Berlin Ultrahigh Field Facility, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Min-Chi Ku
- Berlin Ultrahigh Field Facility, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Conrad Martin
- Berlin Ultrahigh Field Facility, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Dorothea Lorenz
- Leibniz-Institut für Molekulare Pharmakologie, Berlin, Germany
| | - Irene Schütz
- Leibniz-Institut für Molekulare Pharmakologie, Berlin, Germany
| | - Henning M Reimann
- Berlin Ultrahigh Field Facility, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Bettina Purfürst
- Electron Microscopy Core Facility, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Matthias A Dieringer
- 1] Berlin Ultrahigh Field Facility, Max Delbrück Center for Molecular Medicine, Berlin, Germany [2] Experimental and Clinical Research Center, Berlin, Germany
| | | | - Margitta Dathe
- Leibniz-Institut für Molekulare Pharmakologie, Berlin, Germany
| | - Andreas Pohlmann
- Berlin Ultrahigh Field Facility, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Thoralf Niendorf
- Berlin Ultrahigh Field Facility, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| |
Collapse
|
262
|
Maas SLN, de Vrij J, van der Vlist EJ, Geragousian B, van Bloois L, Mastrobattista E, Schiffelers RM, Wauben MHM, Broekman MLD, Nolte-'t Hoen ENM. Possibilities and limitations of current technologies for quantification of biological extracellular vesicles and synthetic mimics. J Control Release 2014; 200:87-96. [PMID: 25555362 PMCID: PMC4324667 DOI: 10.1016/j.jconrel.2014.12.041] [Citation(s) in RCA: 216] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 12/27/2014] [Accepted: 12/29/2014] [Indexed: 12/02/2022]
Abstract
Nano-sized extracelullar vesicles (EVs) released by various cell types play important roles in a plethora of (patho)physiological processes and are increasingly recognized as biomarkers for disease. In addition, engineered EV and EV-inspired liposomes hold great potential as drug delivery systems. Major technologies developed for high-throughput analysis of individual EV include nanoparticle tracking analysis (NTA), tunable resistive pulse sensing (tRPS) and high-resolution flow cytometry (hFC). Currently, there is a need for comparative studies on the available technologies to improve standardization of vesicle analysis in diagnostic or therapeutic settings. We investigated the possibilities, limitations and comparability of NTA, tRPS and hFC for analysis of tumor cell-derived EVs and synthetic mimics (i.e. differently sized liposomes). NTA and tRPS instrument settings were identified that significantly affected the quantification of these particles. Furthermore, we detailed the differences in absolute quantification of EVs and liposomes using the three technologies. This study increases our understanding of possibilities and pitfalls of NTA, tRPS and hFC, which will benefit standardized and large-scale clinical application of (engineered) EVs and EV-mimics in the future.
Collapse
Affiliation(s)
- Sybren L N Maas
- Department of Neurosurgery, University Medical Center Utrecht, The Netherlands; Brain Center Rudolf Magnus, University Medical Center Utrecht, The Netherlands
| | - Jeroen de Vrij
- Department of Neurosurgery, University Medical Center Utrecht, The Netherlands; Brain Center Rudolf Magnus, University Medical Center Utrecht, The Netherlands
| | - Els J van der Vlist
- Department of Biochemistry & Cell Biology, Faculty of Veterinary Medicine, Utrecht University, The Netherlands
| | - Biaina Geragousian
- Department of Neurosurgery, University Medical Center Utrecht, The Netherlands; Brain Center Rudolf Magnus, University Medical Center Utrecht, The Netherlands
| | - Louis van Bloois
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Enrico Mastrobattista
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Raymond M Schiffelers
- Department of Clinical Chemistry and Hematology, University Medical Center Utrecht, The Netherlands
| | - Marca H M Wauben
- Department of Biochemistry & Cell Biology, Faculty of Veterinary Medicine, Utrecht University, The Netherlands
| | - Marike L D Broekman
- Department of Neurosurgery, University Medical Center Utrecht, The Netherlands; Brain Center Rudolf Magnus, University Medical Center Utrecht, The Netherlands
| | - Esther N M Nolte-'t Hoen
- Department of Biochemistry & Cell Biology, Faculty of Veterinary Medicine, Utrecht University, The Netherlands.
| |
Collapse
|
263
|
Gu H, Overstreet AMC, Yang Y. Exosomes Biogenesis and Potentials in Disease Diagnosis and Drug Delivery. ACTA ACUST UNITED AC 2014. [DOI: 10.1142/s1793984414410177] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Exosomes were discovered more than 30 years ago. Only recently has their importance been recognized for intercellular communication. Exosomes, with their size ranging from 30 nm to 100 nm, are lipid bilayer nanoparticles and secreted by many different types of cells with versatile functions. Exosomes contain macromolecules and exist in various body fluids, including blood, urine, milk and ascites fluid. Due to their specific property, exosomes are very promising in the fields of disease diagnosis and therapy. Nanotechnology is a great tool that will be helpful in basic research and the application of exosomes. Here, we briefly review the function and potential use of exosomes in nanomedicine.
Collapse
Affiliation(s)
- Haitao Gu
- Department of Pharmacology & Cell Biophysics University of Cincinnati, College of Medicine, Cincinnati, OH 45267, USA
| | - Anne-Marie C. Overstreet
- Department of Cancer and Cell Biology, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, USA
| | - Yongguang Yang
- Department of Cancer and Cell Biology, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, USA
| |
Collapse
|
264
|
Biodistribution and delivery efficiency of unmodified tumor-derived exosomes. J Control Release 2014; 199:145-55. [PMID: 25523519 DOI: 10.1016/j.jconrel.2014.12.013] [Citation(s) in RCA: 539] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 12/02/2014] [Accepted: 12/13/2014] [Indexed: 01/16/2023]
Abstract
The use of exosomes as a drug delivery vehicle has gained considerable interest. To establish if exosomes could be utilized effectively for drug delivery, a better understanding of their in vivo fate must be established. Through comparisons to liposomal formulations, which have been studied extensively for the last thirty years, we were able to make some comprehensive conclusions about the fate of unmodified tumor-derived exosomes in vivo. We observed a comparable rapid clearance and minimal tumor accumulation of intravenously-injected exosomes, PC:Chol liposomes, and liposomes formulated with the lipid extract of exosomes, suggesting that the unique protein and lipid composition of exosomes does not appreciably impact exosomes' rate of clearance and biodistribution. This rapid clearance along with minimal tumor accumulation of unmodified exosomes limits their use as an anti-cancer drug delivery vehicle; however, when delivered intratumorally, exosomes remained associated with tumor tissue to a significantly greater extent than PC:Chol liposomes. Furthermore, experiments utilizing mice with impaired adaptive or innate immune systems, revealed the significance of the innate immune system along with the complement protein C5 on exosomes' rate of clearance.
Collapse
|
265
|
van der Meel R, Krawczyk-Durka M, van Solinge WW, Schiffelers RM. Toward routine detection of extracellular vesicles in clinical samples. Int J Lab Hematol 2014; 36:244-53. [PMID: 24750670 DOI: 10.1111/ijlh.12247] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 03/25/2014] [Indexed: 01/01/2023]
Abstract
The majority, if not all, of human cell types secrete extracellular vesicles (EVs) into their environment, at least partly as a means of intercellular communication. These secreted vesicles can be detected in most bodily fluids including blood, urine, and saliva. The number of secreted vesicles and their composition is altered in various pathological conditions, raising opportunities to exploit EVs as diagnostic and/or prognostic biomarkers. For this to become a reality, it is important to reach consensus regarding the standardization of protocols for sample collection, EV isolation, handling, and storage for valid comparison and interpretation of measurements. Depending on the information required, there are several detection options including EV number and size distribution, molecular surface markers, procoagulation activity, and RNA content. For these purposes, different techniques are currently utilized or under development. This review discusses the techniques that have the potential to become standard EV detection methods in a clinical diagnostic setting. In addition to the accuracy of the detection technique, other factors such as high-throughput, cost-effectiveness, time consumption, and required operator skill are important to consider. A combination of increasing fundamental knowledge, technological progress, standardization of sample collection, and processing protocols is required for EVs to become reliable predictors of altered physiology or development of disease suitable for routine clinical diagnostics. Cancer and (cardio)vascular disorders are examples of pathologies where EV detection may be applied in the near future for diagnosis and/or prognosis.
Collapse
Affiliation(s)
- R van der Meel
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | |
Collapse
|
266
|
Lai FW, Lichty BD, Bowdish DME. Microvesicles: ubiquitous contributors to infection and immunity. J Leukoc Biol 2014; 97:237-45. [PMID: 25473096 DOI: 10.1189/jlb.3ru0513-292rr] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
MVs, which can be subgrouped into exosomes, SVs, and OMVs, are secreted by eukaryotic and prokaryotic cells. Many previously inexplicable phenomena can be explained by the existence of these vesicles, as they appear to be important in a wide range of biologic processes, such as intercellular communication and transfer of functional genetic information. In this review, we discuss the immunologic roles of MVs during sterile insult and infectious disease. MVs contribute to clotting initiation, cell recruitment, and neovascularization during wound healing. In the context of pathogen infection, both the host and the pathogen use MVs for communication and defense. MVs are exploited by various viruses to evade the host immune response and contribute to viral spread. Bacteria produce MVs that contain virulence factors that contribute to disease pathology and antibiotic resistance. This review summarizes the role of MVs in the pathology and resolution of disease.
Collapse
Affiliation(s)
- Frances W Lai
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Brian D Lichty
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Dawn M E Bowdish
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
267
|
Smyth TJ, Redzic JS, Graner MW, Anchordoquy TJ. Examination of the specificity of tumor cell derived exosomes with tumor cells in vitro. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1838:2954-65. [PMID: 25102470 PMCID: PMC5657189 DOI: 10.1016/j.bbamem.2014.07.026] [Citation(s) in RCA: 148] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 07/18/2014] [Accepted: 07/28/2014] [Indexed: 12/11/2022]
Abstract
Small endogenous vesicles called exosomes are beginning to be explored as drug delivery vehicles. The in vivo targets of exosomes are poorly understood; however, they are believed to be important in cell-to-cell communication and may play a prominent role in cancer metastasis. We aimed to elucidate whether cancer derived exosomes can be used as drug delivery vehicles that innately target tumors over normal tissue. Our in vitro results suggest that while there is some specificity towards cancer cells over "immortalized" cells, it is unclear if the difference is sufficient to achieve precise in vivo targeting. Additionally, we found that exosomes associate with their cellular targets to a significantly greater extent (>10-fold) than liposomes of a similar size. Studies on the association of liposomes mimicking the unique lipid content of exosomes revealed that the lipid composition contributes significantly to cellular adherence/internalization. Cleavage of exosome surface proteins yielded exosomes exhibiting reduced association with their cellular targets, demonstrating the importance of proteins in binding/internalization. Furthermore, although acidic conditions are known to augment the metastatic potential of tumors, we found that cells cultured at low pH released exosomes with significantly less potential for cellular association than cells cultured at physiological pH.
Collapse
Affiliation(s)
- Tyson J Smyth
- University of Colorado Denver, Anschutz Medical Campus, Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, CO, USA.
| | - Jasmina S Redzic
- University of Colorado Denver, Anschutz Medical Campus, Department of Neurosurgery, Aurora, CO, USA
| | - Michael W Graner
- University of Colorado Denver, Anschutz Medical Campus, Department of Neurosurgery, Aurora, CO, USA
| | - Thomas J Anchordoquy
- University of Colorado Denver, Anschutz Medical Campus, Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, CO, USA.
| |
Collapse
|
268
|
Morhayim J, Baroncelli M, van Leeuwen JP. Extracellular vesicles: Specialized bone messengers. Arch Biochem Biophys 2014; 561:38-45. [DOI: 10.1016/j.abb.2014.05.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 04/16/2014] [Accepted: 05/08/2014] [Indexed: 12/22/2022]
|
269
|
Morhayim J, van de Peppel J, Demmers JAA, Kocer G, Nigg AL, van Driel M, Chiba H, van Leeuwen JP. Proteomic signatures of extracellular vesicles secreted by nonmineralizing and mineralizing human osteoblasts and stimulation of tumor cell growth. FASEB J 2014; 29:274-85. [PMID: 25359493 DOI: 10.1096/fj.14-261404] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Beyond forming bone, osteoblasts play pivotal roles in various biologic processes, including hematopoiesis and bone metastasis. Extracellular vesicles (EVs) have been implicated in intercellular communication via transfer of proteins and nucleic acids between cells. We focused on the proteomic characterization of nonmineralizing (NMOBs) and mineralizing (MOBs) human osteoblast (SV-HFOs) EVs and investigated their effect on human prostate cancer (PC3) cells by microscopic, proteomic, and gene expression analyses. Proteomic analysis showed that 97% of the proteins were shared among NMOB and MOB EVs, and 30% were novel osteoblast-specific EV proteins. Label-free quantification demonstrated mineralization stage-dependent 5-fold enrichment of 59 and 451 EV proteins in NMOBs and MOBs, respectively. Interestingly, bioinformatic analyses of the osteoblast EV proteomes and EV-regulated prostate cancer gene expression profiles showed that they converged on pathways involved in cell survival and growth. This was verified by in vitro proliferation assays where osteoblast EV uptake led to 2-fold increase in PC3 cell growth compared to cell-free culture medium-derived vesicle controls. Our findings elucidate the mineralization stage-specific protein content of osteoblast-secreted EVs, show a novel way by which osteoblasts communicate with prostate cancer, and open up innovative avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Jess Morhayim
- Department of Internal Medicine and Erasmus MC Stem Cell and Regenerative Medicine Institute
| | - Jeroen van de Peppel
- Department of Internal Medicine and Erasmus MC Stem Cell and Regenerative Medicine Institute
| | | | - Gulistan Kocer
- Department of Internal Medicine and Erasmus MC Stem Cell and Regenerative Medicine Institute
| | - Alex L Nigg
- Department of Pathology, Erasmus University Medical Center, Rotterdam, The Netherlands; and
| | - Marjolein van Driel
- Department of Internal Medicine and Erasmus MC Stem Cell and Regenerative Medicine Institute
| | - Hideki Chiba
- Department of Basic Pathology, Fukushima Medical University School of Medicine, Hikarigaoka, Fukushima, Japan
| | - Johannes P van Leeuwen
- Department of Internal Medicine and Erasmus MC Stem Cell and Regenerative Medicine Institute,
| |
Collapse
|
270
|
Jo W, Kim J, Yoon J, Jeong D, Cho S, Jeong H, Yoon YJ, Kim SC, Gho YS, Park J. Large-scale generation of cell-derived nanovesicles. NANOSCALE 2014; 6:12056-64. [PMID: 25189198 DOI: 10.1039/c4nr02391a] [Citation(s) in RCA: 160] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Exosomes are enclosed compartments that are released from cells and that can transport biological contents for the purpose of intercellular communications. Research into exosomes is hindered by their rarity. In this article, we introduce a device that uses centrifugal force and a filter with micro-sized pores to generate a large quantity of cell-derived nanovesicles. The device has a simple polycarbonate structure to hold the filter, and operates in a common centrifuge. Nanovesicles are similar in size and membrane structure to exosomes. Nanovesicles contain intracellular RNAs ranging from microRNA to mRNA, intracellular proteins, and plasma membrane proteins. The quantity of nanovesicles produced using the device is 250 times the quantity of naturally secreted exosomes. Also, the quantity of intracellular contents in nanovesicles is twice that in exosomes. Nanovesicles generated from murine embryonic stem cells can transfer RNAs to target cells. Therefore, this novel device and the nanovesicles that it generates are expected to be used in exosome-related research, and can be applied in various applications such as drug delivery and cell-based therapy.
Collapse
Affiliation(s)
- W Jo
- Department of Mechanical Engineering, POSTECH, 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk 790-784, Republic of Korea.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
271
|
Maas SLN, De Vrij J, Broekman MLD. Quantification and size-profiling of extracellular vesicles using tunable resistive pulse sensing. J Vis Exp 2014:e51623. [PMID: 25350417 PMCID: PMC4335984 DOI: 10.3791/51623] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Extracellular vesicles (EVs), including ‘microvesicles’ and ‘exosomes’, are highly abundant in bodily fluids. Recent years have witnessed a tremendous increase in interest in EVs. EVs have been shown to play important roles in various physiological and pathological processes, including coagulation, immune responses, and cancer. In addition, EVs have potential as therapeutic agents, for instance as drug delivery vehicles or as regenerative medicine. Because of their small size (50 to 1,000 nm) accurate quantification and size profiling of EVs is technically challenging. This protocol describes how tunable resistive pulse sensing (tRPS) technology, using the qNano system, can be used to determine the concentration and size of EVs. The method, which relies on the detection of EVs upon their transfer through a nano sized pore, is relatively fast, suffices the use of small sample volumes and does not require the purification and concentration of EVs. Next to the regular operation protocol an alternative approach is described using samples spiked with polystyrene beads of known size and concentration. This real-time calibration technique can be used to overcome technical hurdles encountered when measuring EVs directly in biological fluids.
Collapse
Affiliation(s)
- Sybren L N Maas
- Department of Neurosurgery, University Medical Center Utrecht; Brain Center Rudolf Magnus, University Medical Center Utrecht
| | - Jeroen De Vrij
- Department of Neurosurgery, University Medical Center Utrecht; Brain Center Rudolf Magnus, University Medical Center Utrecht
| | - Marike L D Broekman
- Department of Neurosurgery, University Medical Center Utrecht; Brain Center Rudolf Magnus, University Medical Center Utrecht;
| |
Collapse
|
272
|
Feng DQ, Huang B, Li J, Liu J, Chen XM, Xu YM, Chen X, Zhang HB, Hu LH, Wang XZ. Selective miRNA expression profile in chronic myeloid leukemia K562 cell-derived exosomes. Asian Pac J Cancer Prev 2014; 14:7501-8. [PMID: 24460325 DOI: 10.7314/apjcp.2013.14.12.7501] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Chronic myeloid leukemia (CML) is a myeloproliferative disorder of hematopoietic stem cell scarrying the Philadelphia (Ph) chromosome and an oncogenic BCR-ABL1 fusion gene. The tyrosine kinase inhibitor (TKI) of BCR-ABL1 kinase is a treatment of choice for control of CML. OBJECTIVE Recent studies have demonstrated that miRNAs within exosomes from cancer cells play crucial roles in initiation and progression. This study was performed to assess miRNAs within exosomes of K562 cells. METHODS miRNA microarray analysis of K562 cells and K562 cell-derived exosomes was conducted with the 6th generation miRCURYTM LNA Array (v.16.0). Gene ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were also carried out. GO terms and signaling pathways were categorized into 66 classes (including homophilic cell adhesion, negative regulation of apoptotic process, cell adhesion) and 26 signaling pathways (such as Wnt). RESULTS In exosomes, 49 miRNAs were up regulated as compared to K562 cells, and two of them were further confirmed by quantitative real-time PCR. There are differentially expressed miRNAs between K562 cell derived-exosomes and K562 cells. CONCLUSION Selectively expressed miRNAs in exosomes may promote the development of CML via effects on interactions (e.g. adhesion) of CML cells with their microenvironment.
Collapse
Affiliation(s)
- Dan-Qin Feng
- Department of Clinical Laboratory, the Second Affiliated Hospital of Nanchang University, Nanchang, China E-mail :
| | | | | | | | | | | | | | | | | | | |
Collapse
|
273
|
Brambilla D, Luciani P, Leroux JC. Breakthrough discoveries in drug delivery technologies: The next 30 years. J Control Release 2014; 190:9-14. [DOI: 10.1016/j.jconrel.2014.03.056] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Revised: 03/19/2014] [Accepted: 03/21/2014] [Indexed: 12/13/2022]
|
274
|
van der Meel R, Fens MHAM, Vader P, van Solinge WW, Eniola-Adefeso O, Schiffelers RM. Extracellular vesicles as drug delivery systems: lessons from the liposome field. J Control Release 2014; 195:72-85. [PMID: 25094032 DOI: 10.1016/j.jconrel.2014.07.049] [Citation(s) in RCA: 335] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Revised: 07/25/2014] [Accepted: 07/26/2014] [Indexed: 12/18/2022]
Abstract
Extracellular vesicles (EVs) are membrane-derived particles surrounded by a (phospho)lipid bilayer that are released by cells in the human body. In addition to direct cell-to-cell contact and the secretion of soluble factors, EVs function as another mechanism of intercellular communication. These vesicles are able to efficiently deliver their parental cell-derived molecular cargo to recipient cells, which can result in structural changes at an RNA, protein, or even phenotypic level. For this reason, EVs have recently gained much interest for drug delivery purposes. In contrast to these 'natural delivery systems', synthetic (phospho)lipid vesicles, or liposomes, have been employed as drug carriers for decades, resulting in several approved liposomal nanomedicines used in the clinic. This review discusses the similarities and differences between EVs and liposomes with the focus on features that are relevant for drug delivery purposes such as circulation time, biodistribution, cellular interactions and cargo loading. By applying beneficial features of EVs to liposomes and vice versa, improved drug carriers can be developed which will advance the field of nanomedicines and ultimately improve patient outcomes. While the application of EVs for therapeutic drug delivery is still in its infancy, issues regarding the understanding of EV biogenesis, large-scale production and in vivo interactions need to be addressed in order to develop successful and cost-effective EV-based drug delivery systems.
Collapse
Affiliation(s)
- Roy van der Meel
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marcel H A M Fens
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Pieter Vader
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Wouter W van Solinge
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Raymond M Schiffelers
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
275
|
Pascucci L, Coccè V, Bonomi A, Ami D, Ceccarelli P, Ciusani E, Viganò L, Locatelli A, Sisto F, Doglia SM, Parati E, Bernardo ME, Muraca M, Alessandri G, Bondiolotti G, Pessina A. Paclitaxel is incorporated by mesenchymal stromal cells and released in exosomes that inhibit in vitro tumor growth: a new approach for drug delivery. J Control Release 2014; 192:262-70. [PMID: 25084218 DOI: 10.1016/j.jconrel.2014.07.042] [Citation(s) in RCA: 661] [Impact Index Per Article: 60.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Revised: 07/14/2014] [Accepted: 07/20/2014] [Indexed: 12/13/2022]
Abstract
Mesenchymal stromal cells (MSCs) have been proposed for delivering anticancer agents because of their ability to home in on tumor microenvironment. We found that MSCs can acquire strong anti-tumor activity after priming with Paclitaxel (PTX) through their capacity to uptake and then release the drug. Because MSCs secrete a high amount of membrane microvesicles (MVs), we here investigated the role of MVs in the releasing mechanism of PTX. The murine SR4987 line was used as MSC model. The release of PTX from SR4987 in the conditioned medium (CM) was checked by HPLC and the anti-tumor activity of both CM and MVs was tested on the human pancreatic cell line CFPAC-1. MVs were isolated by ultracentrifugation, analyzed by transmission (TEM) and scanning electron microscopy (SEM), and the presence of PTX by the Fourier transformed infrared (FTIR) microspectroscopy. SR4987 loaded with PTX (SR4987PTX) secreted a significant amount of PTX and their CM possessed strong anti-proliferative activity on CFPAC-1. At TEM and SEM, SR4987PTX showed an increased number of "vacuole-like" structures and shed a relevant number of MVs, but did not differ from untreated SR4987. However, SR4987PTX-derived-MVs (SR4987PTX-MVs) demonstrated a strong anti proliferative activity on CFPAC-1. FTIR analysis of SR4987PTX-MVs showed the presence of an absorption spectrum in the corresponding regions of the PTX marker, absent in MVs from SR4987. Our work is the first demonstration that MSCs are able to package and deliver active drugs through their MVs, suggesting the possibility of using MSCs as a factory to develop drugs with a higher cell-target specificity.
Collapse
Affiliation(s)
- Luisa Pascucci
- Department of Veterinary Medicine, University of Perugia, Italy
| | - Valentina Coccè
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Arianna Bonomi
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Diletta Ami
- Department of Biotechnology and Biosciences, University of Milan Bicocca, Milan, Italy
| | | | - Emilio Ciusani
- Laboratory of Clinical Pathology and Neurogenetic Medicine, Fondazione IRCCS Neurological Institute Carlo Besta, Milan, Italy
| | - Lucia Viganò
- Department of Medical Oncology, Ospedale S. Raffaele Scientific Institute, Milan ,Italy
| | - Alberta Locatelli
- Department of Medical Oncology, Ospedale S. Raffaele Scientific Institute, Milan ,Italy
| | - Francesca Sisto
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | | | - Eugenio Parati
- Cellular Neurobiology Laboratory, Department of Cerebrovascular Diseases, IRCCS Neurological Institute C. Besta, Milan, Italy
| | | | - Maurizio Muraca
- Laboratory Medicine, Children's Hospital Bambino Gesù, Roma, Italy
| | - Giulio Alessandri
- Cellular Neurobiology Laboratory, Department of Cerebrovascular Diseases, IRCCS Neurological Institute C. Besta, Milan, Italy
| | - Gianpietro Bondiolotti
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Augusto Pessina
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy.
| |
Collapse
|
276
|
Llauradó M, Majem B, Altadill T, Lanau L, Castellví J, Sánchez-Iglesias JL, Cabrera S, De la Torre J, Díaz-Feijoo B, Pérez-Benavente A, Colás E, Olivan M, Doll A, Alameda F, Matias-Guiu X, Moreno-Bueno G, Carey MS, Del Campo JM, Gil-Moreno A, Reventós J, Rigau M. MicroRNAs as prognostic markers in ovarian cancer. Mol Cell Endocrinol 2014; 390:73-84. [PMID: 24747602 DOI: 10.1016/j.mce.2014.03.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 02/09/2014] [Accepted: 03/25/2014] [Indexed: 01/18/2023]
Abstract
Ovarian cancer (OC) is the most lethal gynecological malignancy among women. Over 70% of women with OC are diagnosed in advanced stages and most of these cases are incurable. Although most patients respond well to primary chemotherapy, tumors become resistant to treatment. Mechanisms of chemoresistance in cancer cells may be associated with mutational events and/or alterations of gene expression through epigenetic events. Although focusing on known genes has already yielded new information, previously unknown non-coding RNAs, such as microRNAs (miRNAs), also lead insight into the biology of chemoresistance. In this review we summarize the current evidence examining the role of miRNAs as biomarkers of response and survival to therapy in OC. Beside their clinical implications, we also discuss important differences between studies that may have limited their use as clinical biomarkers and suggest new approaches.
Collapse
Affiliation(s)
- Marta Llauradó
- Faculty of Medicine, University of British Columbia, Vancouver, Canada; Research Unit in Biomedicine and Translational Oncology, Vall Hebron Research Institute University Hospital, Barcelona, Spain
| | - Blanca Majem
- Research Unit in Biomedicine and Translational Oncology, Vall Hebron Research Institute University Hospital, Barcelona, Spain
| | - Tatiana Altadill
- Research Unit in Biomedicine and Translational Oncology, Vall Hebron Research Institute University Hospital, Barcelona, Spain
| | - Lucia Lanau
- Research Unit in Biomedicine and Translational Oncology, Vall Hebron Research Institute University Hospital, Barcelona, Spain
| | - Josep Castellví
- Department of Pathology, Vall Hebron University Hospital, Barcelona, Spain
| | | | - Silvia Cabrera
- Department of Gynecological Oncology, Vall Hebron University Hospital, Barcelona, Spain
| | - Javier De la Torre
- Department of Gynecological Oncology, Vall Hebron University Hospital, Barcelona, Spain
| | - Berta Díaz-Feijoo
- Department of Gynecological Oncology, Vall Hebron University Hospital, Barcelona, Spain
| | | | - Eva Colás
- Research Unit in Biomedicine and Translational Oncology, Vall Hebron Research Institute University Hospital, Barcelona, Spain
| | - Mireia Olivan
- Research Unit in Biomedicine and Translational Oncology, Vall Hebron Research Institute University Hospital, Barcelona, Spain
| | - Andreas Doll
- Research Unit in Biomedicine and Translational Oncology, Vall Hebron Research Institute University Hospital, Barcelona, Spain
| | - Francesc Alameda
- Department of Pathology, Hospital del Mar, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Xavier Matias-Guiu
- Department of Pathology and Molecular Genetics and Research Laboratory, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLLEIDA, Lleida, Spain
| | - Gema Moreno-Bueno
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), IdiPAZ, 28029, Madrid, Spain & Fundación MD Anderson Internacional, 28033 Madrid, Spain
| | - Mark S Carey
- Division of Gynecologic Oncology, University of British Columbia and BC Cancer Agency, Vancouver, BC, Canada
| | - Josep Maria Del Campo
- Division of Gynecology and Head and Neck, Department of Oncology, Vall Hebron University Hospital, Barcelona, Spain
| | - Antonio Gil-Moreno
- Department of Gynecological Oncology, Vall Hebron University Hospital, Barcelona, Spain; Faculty of Medicine, Autonomous University of Barcelona, Barcelona, Spain
| | - Jaume Reventós
- Research Unit in Biomedicine and Translational Oncology, Vall Hebron Research Institute University Hospital, Barcelona, Spain; Faculty of Medicine, Autonomous University of Barcelona, Barcelona, Spain; Departament de Ciències Bàsiques, Universitat Internacional de Catalunya, Barcelona, Spain; IDIBELL- Bellvitge Biomedical Research Institute, Barcelona, Spain.
| | - Marina Rigau
- Research Unit in Biomedicine and Translational Oncology, Vall Hebron Research Institute University Hospital, Barcelona, Spain
| |
Collapse
|
277
|
Natasha G, Gundogan B, Tan A, Farhatnia Y, Wu W, Rajadas J, Seifalian AM. Exosomes as Immunotheranostic Nanoparticles. Clin Ther 2014; 36:820-9. [DOI: 10.1016/j.clinthera.2014.04.019] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 04/25/2014] [Accepted: 04/28/2014] [Indexed: 11/25/2022]
|
278
|
Giulivi C. Grand challenges in cellular biochemistry: the "next-gen" biochemistry. Front Chem 2014; 2:22. [PMID: 24809045 PMCID: PMC4010732 DOI: 10.3389/fchem.2014.00022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 04/10/2014] [Indexed: 01/10/2023] Open
Affiliation(s)
- Cecilia Giulivi
- Department of Molecular Biosciences, University of California, DavisDavis, CA, USA
- Medical Investigations of Neurodevelopmental Disorders Institute, University of California, DavisDavis, CA, USA
| |
Collapse
|
279
|
Kavanagh DPJ, Robinson J, Kalia N. Mesenchymal Stem Cell Priming: Fine-tuning Adhesion and Function. Stem Cell Rev Rep 2014; 10:587-99. [DOI: 10.1007/s12015-014-9510-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
280
|
Abstract
Exosomes are nanovesicles secreted into the extracellular environment upon internal vesicle fusion with the plasma membrane. The molecular content of exosomes is a fingerprint of the releasing cell type and of its status. For this reason, and because they are released in easily accessible body fluids such as blood and urine, they represent a precious biomedical tool. A growing body of evidence suggests that exosomes may be used as biomarkers for the diagnosis and prognosis of malignant tumors. This article focuses on the exploitation of exosomes as diagnostic tools for human tumors and discusses possible applications of the same strategies to other pathologies, such as neurodegenerative diseases.
Collapse
Affiliation(s)
- Francesca Properzi
- Department of Cell Biology & Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | | | | |
Collapse
|
281
|
Grange C, Tapparo M, Bruno S, Chatterjee D, Quesenberry PJ, Tetta C, Camussi G. Biodistribution of mesenchymal stem cell-derived extracellular vesicles in a model of acute kidney injury monitored by optical imaging. Int J Mol Med 2014; 33:1055-63. [PMID: 24573178 PMCID: PMC4020482 DOI: 10.3892/ijmm.2014.1663] [Citation(s) in RCA: 268] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 01/17/2014] [Indexed: 12/16/2022] Open
Abstract
Mesenchymal stem cells (MSCs) contribute to the recovery of tissue injury, providing a paracrine support. Cell-derived extracellular vesicles (EVs), carrying membrane and cytoplasmatic constituents of the cell of origin, have been described as a fundamental mechanism of intercellular communication. We previously demonstrated that EVs derived from human MSCs accelerated recovery following acute kidney injury (AKI) in vivo. The aim of the present study was to investigate the biodistribution and the renal localization of EVs in AKI. For this purpose, two methods for EV labeling suitable for in vivo tracking with optical imaging (OI), were employed using near infrared (NIR) dye (DiD): i) labeled EVs were generated by MSCs pre-incubated with NIR dye and collected from cell supernatants; ii) purified EVs were directly labeled with NIR dye. EVs obtained with these two procedures were injected intravenously (i.v.) into mice with glycerol-induced AKI and into healthy mice to compare the efficacy of the two labeling methods for in vivo detection of EVs at the site of damage. We found that the labeled EVs accumulated specifically in the kidneys of the mice with AKI compared with the healthy controls. After 5 h, the EVs were detectable in whole body images and in dissected kidneys by OI with both types of labeling procedures. The directly labeled EVs showed a higher and brighter fluorescence compared with the labeled EVs produced by cells. The signal generated by the directly labeled EVs was maintained in time, but provided a higher background than that of the labeled EVs produced by cells. The comparison of the two methods indicated that the latter displayed a greater specificity for the injured kidney.
Collapse
Affiliation(s)
- Cristina Grange
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - Marta Tapparo
- Translational Center for Regenerative Medicine, University of Torino, Torino, Italy
| | - Stefania Bruno
- Department of Molecular Biotechnology and Health Science, University of Torino, Torino, Italy
| | - Devasis Chatterjee
- Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Peter J Quesenberry
- Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Ciro Tetta
- EMEA LA Medical Board, Fresenius Medical Care, Bad Homburg, Germany
| | - Giovanni Camussi
- Department of Medical Sciences, University of Torino, Torino, Italy
| |
Collapse
|
282
|
Gajos-Michniewicz A, Duechler M, Czyz M. MiRNA in melanoma-derived exosomes. Cancer Lett 2014; 347:29-37. [PMID: 24513178 DOI: 10.1016/j.canlet.2014.02.004] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 01/21/2014] [Accepted: 02/03/2014] [Indexed: 02/08/2023]
Abstract
Proteins, RNAs and viruses can be spread through exosomes, therefore transport utilizing these nanovesicles is of the great interest. MiRNAs are common exosomal constituents capable of influencing expression of a variety of target genes. MiRNA signatures of exosomes are unique in cancer patients and differ from those in normal controls. The knowledge about miRNA profiles of tumor-derived exosomes may contribute to better diagnosis, determination of tumor progression and response to treatment, as well as to the development of targeted therapies. We summarize the current knowledge with regard to miRNAs that are found in exosomes derived from tumors, particularly from melanoma.
Collapse
Affiliation(s)
| | - Markus Duechler
- Department of Bioorganic Chemistry, Centre for Molecular and Macromolecular Studies, Polish Academy of Sciences, Lodz, Poland
| | - Malgorzata Czyz
- Department of Molecular Biology of Cancer, Medical University of Lodz, Poland.
| |
Collapse
|
283
|
New considerations in the preparation of nucleic acid-loaded extracellular vesicles. Ther Deliv 2014; 5:105-7. [DOI: 10.4155/tde.13.142] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
284
|
Cho YW, Kim SY, Kwon IC, Kim IS. Complex adaptive therapeutic strategy (CATS) for cancer. J Control Release 2014; 175:43-7. [DOI: 10.1016/j.jconrel.2013.12.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 11/19/2013] [Accepted: 12/16/2013] [Indexed: 12/16/2022]
|
285
|
Jinnin M. Various applications of microRNAs in skin diseases. J Dermatol Sci 2014; 74:3-8. [PMID: 24530178 DOI: 10.1016/j.jdermsci.2014.01.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 01/11/2014] [Accepted: 01/16/2014] [Indexed: 12/21/2022]
Abstract
microRNA (miRNA) is a family of non-coding RNAs, which consists of 19-25 nucleotides and regulates the expression of approximately 30% of human protein-coding mRNAs. miRNAs can bind to complementary sequences of the three prime untranslated regions of target mRNAs, leading to the modulation of gene expression. By altering target expression, miRNAs can affect various cellular activities including cell proliferation and cell development in vitro or carcinogenesis and immune response in vivo. A lot of researches have paid attention to the possibility that miRNAs play a role in the pathogenesis of various human disorders including skin diseases. For example, miR-29a down-regulation is thought to mediate the posttranscriptional up-regulation of collagens, which contributes to the tissue fibrosis in scleroderma. In addition, recent studies indicate that extracellular miRNA levels may be useful for the diagnosis and/or the estimation of disease activity of skin diseases. miR-150 levels were significantly decreased in sera of scleroderma patients, and were inversely correlated with the prevalence of pitting scars/ulcers and the incidence of anti-topoisomerase I antibody. Currently, the therapeutic value of miRNAs for the treatment of human diseases is under evaluation in animal models. let-7a can be overexpressed in the mouse skin by intermittent intraperitoneal miRNA injection, and skin fibrosis induced by bleomycin in mice can be improved by the supplementation of let-7a. This paper discusses the possible applications of miRNAs in the clarification of pathogenesis, diagnosis, evaluation of disease activity and treatment of skin diseases.
Collapse
Affiliation(s)
- Masatoshi Jinnin
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, Japan.
| |
Collapse
|
286
|
Naksuriya O, Okonogi S, Schiffelers RM, Hennink WE. Curcumin nanoformulations: a review of pharmaceutical properties and preclinical studies and clinical data related to cancer treatment. Biomaterials 2014; 35:3365-83. [PMID: 24439402 DOI: 10.1016/j.biomaterials.2013.12.090] [Citation(s) in RCA: 598] [Impact Index Per Article: 54.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 12/22/2013] [Indexed: 12/26/2022]
Abstract
Curcumin, a natural yellow phenolic compound, is present in many kinds of herbs, particularly in Curcuma longa Linn. (turmeric). It is a natural antioxidant and has shown many pharmacological activities such as anti-inflammatory, anti-microbial, anti-cancer, and anti-Alzheimer in both preclinical and clinical studies. Moreover, curcumin has hepatoprotective, nephroprotective, cardioprotective, neuroprotective, hypoglycemic, antirheumatic, and antidiabetic activities and it also suppresses thrombosis and protects against myocardial infarction. Particularly, curcumin has demonstrated efficacy as an anticancer agent, but a limiting factor is its extremely low aqueous solubility which hampers its use as therapeutic agent. Therefore, many technologies have been developed and applied to overcome this limitation. In this review, we summarize the recent works on the design and development of nano-sized delivery systems for curcumin, including liposomes, polymeric nanoparticles and micelles, conjugates, peptide carriers, cyclodextrins, solid dispersions, lipid nanoparticles and emulsions. Efficacy studies of curcumin nanoformulations using cancer cell lines and in vivo models as well as up-to-date human clinical trials are also discussed.
Collapse
Affiliation(s)
- Ornchuma Naksuriya
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Suthep Rd, Mueang, Chiang Mai 50200, Thailand; Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, Utrecht 3805 TB, The Netherlands
| | - Siriporn Okonogi
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Suthep Rd, Mueang, Chiang Mai 50200, Thailand
| | - Raymond M Schiffelers
- Department of Clinical Chemistry and Hematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Wim E Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, Utrecht 3805 TB, The Netherlands.
| |
Collapse
|
287
|
Kim J, Morley S, Le M, Bedoret D, Umetsu DT, Di Vizio D, Freeman MR. Enhanced shedding of extracellular vesicles from amoeboid prostate cancer cells: potential effects on the tumor microenvironment. Cancer Biol Ther 2014; 15:409-18. [PMID: 24423651 DOI: 10.4161/cbt.27627] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The gene encoding the cytoskeletal regulator DIAPH3 is lost at high frequency in metastatic prostate cancer, and DIAPH3 silencing evokes a transition to an amoeboid tumor phenotype in multiple cell backgrounds. This amoeboid transformation is accompanied by increased tumor cell migration, invasion, and metastasis. DIAPH3 silencing also promotes the formation of atypically large (> 1 μm) membrane blebs that can be shed as extracellular vesicles (EV) containing bioactive cargo. Whether loss of DIAPH3 also stimulates the release of nano-sized EV (e.g., exosomes) is not established. Here we examined the mechanism of release and potential biological functions of EV shed from DIAPH3-silenced and other prostate cancer cells. We observed that stimulation of LNCaP cells with the prostate stroma-derived growth factor heparin-binding EGF-like growth factor (HB-EGF), combined with p38MAPK inhibition caused EV shedding, a process mediated by ERK1/2 hyperactivation. DIAPH3 silencing in DU145 cells also increased rates of EV production. EV isolated from DIAPH3-silenced cells activated AKT1 and androgen signaling, increased proliferation of recipient tumor cells, and suppressed proliferation of human macrophages and peripheral blood mononuclear cells. DU145 EV contained miR-125a, which suppressed AKT1 expression and proliferation in recipient human peripheral blood mononuclear cells and macrophages. Our findings suggest that EV produced as a result of DIAPH3 loss or growth factor stimulation may condition the tumor microenvironment through multiple mechanisms, including the proliferation of cancer cells and suppression of tumor-infiltrating immune cells.
Collapse
Affiliation(s)
- Jayoung Kim
- Division of Cancer Biology and Therapeutics; Departments of Surgery, Pathology and Laboratory Medicine, and Biomedical Sciences; Samuel Oschin Comprehensive Cancer Institute; Cedars-Sinai Medical Center; Los Angeles, CA USA; Urological Diseases Research Center; Boston Children's Hospital; Boston, MA USA; Department of Surgery; Harvard Medical School; Boston, MA USA
| | - Samantha Morley
- Urological Diseases Research Center; Boston Children's Hospital; Boston, MA USA; Department of Surgery; Harvard Medical School; Boston, MA USA
| | - Minh Le
- Program in Cellular and Molecular Medicine; Boston Children's Hospital; Harvard Medical School; Boston, MA USA
| | - Denis Bedoret
- Division of Immunology; Boston Children's Hospital; Harvard Medical School; Boston, MA USA
| | - Dale T Umetsu
- Division of Immunology; Boston Children's Hospital; Harvard Medical School; Boston, MA USA
| | - Dolores Di Vizio
- Division of Cancer Biology and Therapeutics; Departments of Surgery, Pathology and Laboratory Medicine, and Biomedical Sciences; Samuel Oschin Comprehensive Cancer Institute; Cedars-Sinai Medical Center; Los Angeles, CA USA; Urological Diseases Research Center; Boston Children's Hospital; Boston, MA USA; Department of Surgery; Harvard Medical School; Boston, MA USA
| | - Michael R Freeman
- Division of Cancer Biology and Therapeutics; Departments of Surgery, Pathology and Laboratory Medicine, and Biomedical Sciences; Samuel Oschin Comprehensive Cancer Institute; Cedars-Sinai Medical Center; Los Angeles, CA USA; Urological Diseases Research Center; Boston Children's Hospital; Boston, MA USA; Department of Surgery; Harvard Medical School; Boston, MA USA
| |
Collapse
|
288
|
Fierabracci A, Del Fattore A, Luciano R, Muraca M, Teti A, Muraca M. Recent advances in mesenchymal stem cell immunomodulation: the role of microvesicles. Cell Transplant 2013; 24:133-149. [PMID: 24268069 DOI: 10.3727/096368913x675728] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Mesenchymal stem cells are the most widely used cell phenotype for therapeutic applications, the main reasons being their well-established abilities to promote regeneration of injured tissues and to modulate immune responses. Efficacy was reported in the treatment of several animal models of inflammatory and autoimmune diseases and, in clinical settings, for the management of disorders such as GVHD, systemic lupus erythematosus, multiple sclerosis, and inflammatory bowel disease. The effects of mesenchymal stem cells are believed to be largely mediated by paracrine signals, and several secreted molecules have been identified as contributors to the net biological effect. Recently, it has been recognized that bioactive molecules can be shuttled from cell to cell packed in microvesicles, tiny portions of cytoplasm surrounded by a membrane. Coding and noncoding RNAs are also carried in such microvesicles, transferring relevant biological activity to target cells. Several reports indicate that the regenerative effect of mesenchymal stem cells can be reproduced by microvesicles isolated from their culture medium. More recent evidence suggests that the immunomodulatory effects of mesenchymal stem cells are also at least partially mediated by secreted microvesicles. These findings allow better understanding of the mechanisms involved in cell-to-cell interaction and may have interesting implications for the development of novel therapeutic tools in place of the parent cells.
Collapse
|
289
|
Takemoto R, Jinnin M, Wang Z, Kudo H, Inoue K, Nakayama W, Ichihara A, Igata T, Kajihara I, Fukushima S, Ihn H. Hair miR-29a levels are decreased in patients with scleroderma. Exp Dermatol 2013; 22:832-3. [DOI: 10.1111/exd.12245] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2013] [Indexed: 12/28/2022]
Affiliation(s)
- Risa Takemoto
- Department of Dermatology and Plastic Surgery; Faculty of Life Sciences; Kumamoto University; Kumamoto Japan
| | - Masatoshi Jinnin
- Department of Dermatology and Plastic Surgery; Faculty of Life Sciences; Kumamoto University; Kumamoto Japan
| | - Zhongzhi Wang
- Department of Dermatology and Plastic Surgery; Faculty of Life Sciences; Kumamoto University; Kumamoto Japan
| | - Hideo Kudo
- Department of Dermatology and Plastic Surgery; Faculty of Life Sciences; Kumamoto University; Kumamoto Japan
| | - Kuniko Inoue
- Department of Dermatology and Plastic Surgery; Faculty of Life Sciences; Kumamoto University; Kumamoto Japan
| | - Wakana Nakayama
- Department of Dermatology and Plastic Surgery; Faculty of Life Sciences; Kumamoto University; Kumamoto Japan
| | - Asako Ichihara
- Department of Dermatology and Plastic Surgery; Faculty of Life Sciences; Kumamoto University; Kumamoto Japan
| | - Toshikatsu Igata
- Department of Dermatology and Plastic Surgery; Faculty of Life Sciences; Kumamoto University; Kumamoto Japan
| | - Ikko Kajihara
- Department of Dermatology and Plastic Surgery; Faculty of Life Sciences; Kumamoto University; Kumamoto Japan
| | - Satoshi Fukushima
- Department of Dermatology and Plastic Surgery; Faculty of Life Sciences; Kumamoto University; Kumamoto Japan
| | - Hironobu Ihn
- Department of Dermatology and Plastic Surgery; Faculty of Life Sciences; Kumamoto University; Kumamoto Japan
| |
Collapse
|
290
|
Jang SC, Kim OY, Yoon CM, Choi DS, Roh TY, Park J, Nilsson J, Lötvall J, Kim YK, Gho YS. Bioinspired exosome-mimetic nanovesicles for targeted delivery of chemotherapeutics to malignant tumors. ACS NANO 2013; 7:7698-710. [PMID: 24004438 DOI: 10.1021/nn402232g] [Citation(s) in RCA: 738] [Impact Index Per Article: 61.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Exosomes, the endogenous nanocarriers that can deliver biological information between cells, were recently introduced as new kind of drug delivery system. However, mammalian cells release relatively low quantities of exosomes, and purification of exosomes is difficult. Here, we developed bioinspired exosome-mimetic nanovesicles that deliver chemotherapeutics to the tumor tissue after systemic administration. The chemotherapeutics-loaded nanovesicles were produced by the breakdown of monocytes or macrophages using a serial extrusion through filters with diminishing pore sizes (10, 5, and 1 μm). These cell-derived nanovesicles have similar characteristics with the exosomes but have 100-fold higher production yield. Furthermore, the nanovesicles have natural targeting ability of cells by maintaining the topology of plasma membrane proteins. In vitro, chemotherapeutic drug-loaded nanovesicles induced TNF-α-stimulated endothelial cell death in a dose-dependent manner. In vivo, experiments in mice showed that the chemotherapeutic drug-loaded nanovesicles traffic to tumor tissue and reduce tumor growth without the adverse effects observed with equipotent free drug. Furthermore, compared with doxorubicin-loaded exosomes, doxorubicin-loaded nanovesicles showed similar in vivo antitumor activity. However, doxorubicin-loaded liposomes that did not carry targeting proteins were inefficient in reducing tumor growth. Importantly, removal of the plasma membrane proteins by trypsinization eliminated the therapeutic effects of the nanovesicles both in vitro and in vivo. Taken together, these studies suggest that the bioengineered nanovesicles can serve as novel exosome-mimetics to effectively deliver chemotherapeutics to treat malignant tumors.
Collapse
Affiliation(s)
- Su Chul Jang
- Department of Life Sciences, Pohang University of Science and Technology , Pohang, Gyeongbuk 790-784, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
291
|
Kooijmans SAA, Stremersch S, Braeckmans K, de Smedt SC, Hendrix A, Wood MJA, Schiffelers RM, Raemdonck K, Vader P. Electroporation-induced siRNA precipitation obscures the efficiency of siRNA loading into extracellular vesicles. J Control Release 2013; 172:229-238. [PMID: 23994516 DOI: 10.1016/j.jconrel.2013.08.014] [Citation(s) in RCA: 468] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2013] [Revised: 08/06/2013] [Accepted: 08/14/2013] [Indexed: 12/25/2022]
Abstract
Extracellular vesicles (EVs) are specialised endogenous carriers of proteins and nucleic acids and are involved in intercellular communication. EVs are therefore proposed as candidate drug delivery systems for the delivery of nucleic acids and other macromolecules. However, the preparation of EV-based drug delivery systems is hampered by the lack of techniques to load the vesicles with nucleic acids. In this work we have now characterised in detail the use of an electroporation method for this purpose. When EVs were electroporated with fluorescently labelled siRNA, siRNA retention was comparable with previously published results (20-25% based on fluorescence spectroscopy and fluorescence fluctuation spectroscopy), and electroporation with unlabelled siRNA resulted in significant siRNA retention in the EV pellet as measured by RT-PCR. Remarkably, when siRNA was electroporated in the absence of EVs, a similar or even greater siRNA retention was measured. Nanoparticle tracking analysis and confocal microscopy showed extensive formation of insoluble siRNA aggregates after electroporation, which could be dramatically reduced by addition of EDTA. Other strategies to reduce aggregate formation, including the use of cuvettes with conductive polymer electrodes and the use of an acidic citrate electroporation buffer, resulted in a more efficient reduction of siRNA precipitation than EDTA. However, under these conditions, siRNA retention was below 0.05% and no significant differences in siRNA retention could be measured between samples electroporated in the presence or absence of EVs. Our results show that electroporation of EVs with siRNA is accompanied by extensive siRNA aggregate formation, which may cause overestimation of the amount of siRNA actually loaded into EVs. Moreover, our data clearly illustrate that electroporation is far less efficient than previously described, and highlight the necessity for alternative methods to prepare siRNA-loaded EVs.
Collapse
Affiliation(s)
- Sander A A Kooijmans
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Stephan Stremersch
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ghent, Belgium
| | - Kevin Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ghent, Belgium; Centre for Nano- and Biophotonics, Ghent University, Ghent, Belgium
| | - Stefaan C de Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ghent, Belgium
| | - An Hendrix
- Department of Radiation Oncology and Experimental Cancer Research, Laboratory of Experimental Cancer Research, Ghent University Hospital, Ghent, Belgium
| | - Matthew J A Wood
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Raymond M Schiffelers
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Koen Raemdonck
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ghent, Belgium.
| | - Pieter Vader
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.
| |
Collapse
|
292
|
Vader P, Fens MHAM, Sachini N, van Oirschot BA, Andringa G, Egberts ACG, Gaillard CAJM, Rasmussen JT, van Wijk R, van Solinge WW, Schiffelers RM. Taxol®-induced phosphatidylserine exposure and microvesicle formation in red blood cells is mediated by its vehicle Cremophor® EL. Nanomedicine (Lond) 2013; 8:1127-35. [DOI: 10.2217/nnm.12.163] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Aim: The conventional clinical formulation of paclitaxel (PTX), Taxol®, consists of Cremophor® EL (CrEL) and ethanol. CrEL-formulated PTX is associated with acute hypersensitivity reactions, anemia and cardiovascular events. In this study, the authors investigated the effects of CrEL-PTX on red blood cells (RBCs) and compared these with the effects observed after exposure to the novel nanoparticle albumin-bound PTX, marketed as Abraxane®. Results: The authors demonstrate that CrEL is primarily responsible for RBC lysis and induction of phosphatidylserine exposure. Phosphatidylserine-exposing RBCs showed increased association with endothelial cells in culture. The authors also identified CrEL as being responsible for vesiculation of RBCs. This is the first time that excipients have been shown to be involved in microvesicle formation. Microvesicles were taken up by endothelial cells. Conclusion: These results offer new insights into the side effect profile of Taxol, which is likely to have implications for patients with erythrocyte disorders. Abraxane did not induce any of these effects on RBCs, indicating that the choice of excipients can have a pronounced influence on the efficacy and side effects of drug molecules. Original submitted 16th April 2012; Revised submitted 24th August 2012; Published online 5 February 2013
Collapse
Affiliation(s)
- Pieter Vader
- Department of Clinical Chemistry & Hematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marcel HAM Fens
- Department of Clinical Chemistry & Hematology, University Medical Center Utrecht, Utrecht, The Netherlands
- Children’s Hospital Oakland Research Institute, Oakland, CA, USA
| | - Nikoleta Sachini
- Department of Clinical Chemistry & Hematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Brigitte A van Oirschot
- Department of Clinical Chemistry & Hematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Grietje Andringa
- Department of Clinical Chemistry & Hematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Antoine CG Egberts
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Carlo AJM Gaillard
- Department of Nephrology, Vrije Universiteit University Medical Center, Amsterdam, The Netherlands
| | - Jan T Rasmussen
- Protein Chemistry Laboratory, Department of Molecular Biology, Aarhus University, Aarhus, Denmark
| | - Richard van Wijk
- Department of Clinical Chemistry & Hematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Wouter W van Solinge
- Department of Clinical Chemistry & Hematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Raymond M Schiffelers
- Department of Clinical Chemistry & Hematology, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
293
|
Silva AKA, Kolosnjaj-Tabi J, Bonneau S, Marangon I, Boggetto N, Aubertin K, Clément O, Bureau MF, Luciani N, Gazeau F, Wilhelm C. Magnetic and photoresponsive theranosomes: translating cell-released vesicles into smart nanovectors for cancer therapy. ACS NANO 2013; 7:4954-4966. [PMID: 23641799 DOI: 10.1021/nn400269x] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Cell-released vesicles are natural carriers that circulate in body fluids and transport biological agents to distal cells. As nature uses vesicles in cell communication to promote tumor progression, we propose to harness their unique properties and exploit these biogenic carriers as Trojan horses to deliver therapeutic payloads to cancer cells. In a theranostic approach, cell-released vesicles were engineered by a top-down procedure from precursor cells, previously loaded with a photosensitizer and magnetic nanoparticles. The double exogenous cargo provided vesicles with magnetic and optical responsiveness allowing therapeutic and imaging functions. This new class of cell-derived smart nanovectors was named "theranosomes". Theranosomes enabled efficient photodynamic tumor therapy in a murine cancer model in vivo. Moreover the distribution of this biogenic vector could be monitored by dual-mode imaging, combining fluorescence and MRI. This study reports the first success in translating a cell communication mediator into a smart theranostic nanovector.
Collapse
Affiliation(s)
- Amanda K A Silva
- Laboratoire Matière et Systèmes Complexes, UMR 7057, CNRS and Université Paris Diderot , 10 Rue Alice Domon et Léonie Duquet, 75205 Paris Cedex 13, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
294
|
Vagapova G, Ibragimova A, Zakharov A, Dobrynin A, Galkina I, Zakharova L, Konovalov A. Novel biomimetic systems based on polyethylene glycols and amphiphilic phosphonium salt. Self-organization and solubilization of hydrophobic guest. Eur Polym J 2013. [DOI: 10.1016/j.eurpolymj.2013.01.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
295
|
Inal JM, Kosgodage U, Azam S, Stratton D, Antwi-Baffour S, Lange S. Blood/plasma secretome and microvesicles. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1834:2317-25. [PMID: 23590876 DOI: 10.1016/j.bbapap.2013.04.005] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 04/04/2013] [Accepted: 04/08/2013] [Indexed: 11/26/2022]
Abstract
A major but hitherto overseen component of the blood/plasma secretome is that of extracellular vesicles (EVs) which are shed from all blood cell types. These EVs are made up of microvesicles (MVs) and exosomes. MVs, 100nm-1μm in diameter, are released from the cell surface, and are a rich source of non-conventionally secreted proteins lacking a conventional signal peptide, and thus not secreted by the classical secretory pathways. Exosomes are smaller vesicles (≤100nm) having an endocytic origin and released upon multivesicular body fusion with the plasma membrane. Both vesicle types play major roles in intercellular cross talk and constitute an important component of the secretome especially in the area of biomarkers for cancer. The release of EVs, which are found in all the bodily fluids, is enhanced in cancer and a major focus of cancer proteomics is therefore targeted at EVs. The blood/plasma secretome is also a source of EVs, potentially diagnostic of infectious disease, whether from EVs released from infected cells or from the pathogens themselves. Despite the great excitement in this field, as is stated here and in other parts of this Special issue entitled: An Updated Secretome, much of the EV research, whether proteomic or functional in nature, urgently needs standardisation both in terms of nomenclature and isolation protocols. This article is part of a Special Issue entitled: An Updated Secretome.
Collapse
Affiliation(s)
- Jameel M Inal
- Cellular and Molecular Immunology Research Centre, School of Human Sciences, London Metropolitan University, 166-220 Holloway Road, London, N7 8DB, UK.
| | | | | | | | | | | |
Collapse
|
296
|
Siegal T. Which drug or drug delivery system can change clinical practice for brain tumor therapy? Neuro Oncol 2013; 15:656-69. [PMID: 23502426 DOI: 10.1093/neuonc/not016] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The prognosis and treatment outcome for primary brain tumors have remained unchanged despite advances in anticancer drug discovery and development. In clinical trials, the majority of promising experimental agents for brain tumors have had limited impact on survival or time to recurrence. These disappointing results are partially explained by the inadequacy of effective drug delivery to the CNS. The impediments posed by the various specialized physiological barriers and active efflux mechanisms lead to drug failure because of inability to reach the desired target at a sufficient concentration. This perspective reviews the leading strategies that aim to improve drug delivery to brain tumors and their likelihood to change clinical practice. The English literature was searched for defined search items. Strategies that use systemic delivery and those that use local delivery are critically reviewed. In addition, challenges posed for drug delivery by combined treatment with anti-angiogenic therapy are outlined. To impact clinical practice and to achieve more than just a limited local control, new drugs and delivery systems must adhere to basic clinical expectations. These include, in addition to an antitumor effect, a verified favorable adverse effects profile, easy introduction into clinical practice, feasibility of repeated or continuous administration, and compatibility of the drug or delivery system with any tumor size and brain location.
Collapse
Affiliation(s)
- Tali Siegal
- Gaffin Center for Neuro-Oncology, Hadassah Hebrew-University Medical Center, Ein Kerem, P.O. Box 12000, Jerusalem 91120, Israel.
| |
Collapse
|
297
|
Koppers-Lalic D, Hogenboom MM, Middeldorp JM, Pegtel DM. Virus-modified exosomes for targeted RNA delivery; a new approach in nanomedicine. Adv Drug Deliv Rev 2013; 65:348-56. [PMID: 22820525 PMCID: PMC7103310 DOI: 10.1016/j.addr.2012.07.006] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Revised: 07/04/2012] [Accepted: 07/08/2012] [Indexed: 12/21/2022]
Abstract
A major goal in biomedical research is to clinically reverse the cause of disease rather than treating the symptoms. Gene therapy has the potential to meet this goal and the discovery of RNA interference (RNAi) has lead to a new class of highly selective therapeutics. However, initial enthusiasm is reduced due to safety concerns associated with virus-based delivery vectors that are used for in vivo delivery. Viral vectors for siRNA delivery into target cells are used because of their high target specificity and delivery efficacy (endosomal escape). Recent discoveries suggest that a specialized form of nano-sized lipid vesicles called exosomes can incorporate and transport functional RNAs into target cells and may serve as an attractive alternative. Evidence is accumulating that most pluricellular organisms sustain exosome-based communications via inter-cellular exchange of mRNA and miRNAs between cells. We discovered that viruses have found ways to exploit this communication pathway and we argue here that adaptations of exosomes imposed by viruses maybe exploited for superior delivery of RNA in vivo. We discuss recent discoveries in exosome biogenesis their physical properties, targeting and delivery strategies and how the knowledge of exosome production in virus infected cells could propel their entry into clinical settings.
Collapse
Key Words
- abc, atp-binding cassette
- ago, argonaute
- bace1, β-site app-cleaving enzyme 1
- bbb, blood brain barrier
- dex, dendritic cell- derived exosome
- ebv, epstein–barr virus
- ee, early endosome
- escrt, endosomal sorting complexes required for transport
- hcc, hepatocellular carcinoma
- ilv, intralumenal vesicles
- imdc, immature dendritic cell
- imdex, immature dendritic cell-derived exosome
- lcl, latency type iii lymphoblastoid b cell line
- le, late endosome
- lmp1, latent membrane protein 1
- mdex, mature dendritic cell-derived exosome
- mdr1, multidrug resistance protein 1
- mirna, microrna
- mrna, messenger rna
- mvb, multivesicular body
- p-body, processing body
- peg, poly(ethylene glycol)
- pirna, piwi-interacting rna
- risc, rna-induced silencing complex
- rnai, rna interference
- sirna, small interference rna
- vlp, virus-like particles
- vsv-g, g protein of vesicular stomatitis virus
- exosomes
- microrna
- genetic-transfer
- virus
- gene-therapy
Collapse
Affiliation(s)
- Danijela Koppers-Lalic
- Department of Pathology, Cancer Center Amsterdam, VU University Medical Center, de Boelelaan 1117HV Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
298
|
Reyes-Izquierdo T, Argumedo R, Shu C, Nemzer B, Pietrzkowski Z. Stimulatory Effect of Whole Coffee Fruit Concentrate Powder on Plasma Levels of Total and Exosomal Brain-Derived Neurotrophic Factor in Healthy Subjects: An Acute Within-Subject Clinical Study. ACTA ACUST UNITED AC 2013. [DOI: 10.4236/fns.2013.49127] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
299
|
Plasma membrane-derived microvesicles released from tip endothelial cells during vascular sprouting. Angiogenesis 2012; 15:761-9. [PMID: 22886085 PMCID: PMC3496552 DOI: 10.1007/s10456-012-9292-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Accepted: 07/18/2012] [Indexed: 01/23/2023]
Abstract
During human foetal brain vascularization, activated CD31+/CD105+ endothelial cells are characterized by the emission of filopodial processes which also decorate the advancing tip of the vascular sprout. Together with filopodia, both the markers also reveal a number of plasma membrane-derived microvesicles (MVs) which are concentrated around the tip cell tuft of processes. At this site, MVs appear in tight contact with endothelial filopodia and follow these long processes, advancing into the surrounding neuropil to a possible cell target. These observations suggest that, like shedding vesicles of many other cell types that deliver signalling molecules and play a role in cell-to-cell communication, MVs sent out from endothelial tip cells could be involved in tip cell guidance and/or act on target cells, regulating cell-to-cell mutual recognition during vessel sprouting and final anastomosis. The results also suggest a new role for tip cell filopodia as conveyor processes for transporting MVs far from the cell of origin in a controlled microenvironment. Additional studies focused on the identification of MV content are needed to ultimately clarify the significance of tip cell MVs during human brain vascularization.
Collapse
|
300
|
Guduric-Fuchs J, O'Connor A, Camp B, O'Neill CL, Medina RJ, Simpson DA. Selective extracellular vesicle-mediated export of an overlapping set of microRNAs from multiple cell types. BMC Genomics 2012; 13:357. [PMID: 22849433 PMCID: PMC3532190 DOI: 10.1186/1471-2164-13-357] [Citation(s) in RCA: 423] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Accepted: 07/16/2012] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) are a class of small RNA molecules that regulate expression of specific mRNA targets. They can be released from cells, often encapsulated within extracellular vesicles (EVs), and therefore have the potential to mediate intercellular communication. It has been suggested that certain miRNAs may be selectively exported, although the mechanism has yet to be identified. Manipulation of the miRNA content of EVs will be important for future therapeutic applications. We therefore wished to assess which endogenous miRNAs are enriched in EVs and how effectively an overexpressed miRNA would be exported. RESULTS Small RNA libraries from HEK293T cells and vesicles before or after transfection with a vector for miR-146a overexpression were analysed by deep sequencing. A subset of miRNAs was found to be enriched in EVs; pathway analysis of their predicted target genes suggests a potential role in regulation of endocytosis. RT-qPCR in additional cell types and analysis of publicly available data revealed that many of these miRNAs tend to be widely preferentially exported. Whilst overexpressed miR-146a was highly enriched both in transfected cells and their EVs, the cellular:EV ratios of endogenous miRNAs were not grossly altered. MiR-451 was consistently the most highly exported miRNA in many different cell types. Intriguingly, Argonaute2 (Ago2) is required for miR-451 maturation and knock out of Ago2 has been shown to decrease expression of other preferentially exported miRNAs (eg miR-150 and miR-142-3p). CONCLUSION The global expression data provided by deep sequencing confirms that specific miRNAs are enriched in EVs released by HEK293T cells. Observation of similar patterns in a range of cell types suggests that a common mechanism for selective miRNA export may exist.
Collapse
Affiliation(s)
- Jasenka Guduric-Fuchs
- Centre for Vision and Vascular Science, Queen's University Belfast, Northern Ireland, UK
| | | | | | | | | | | |
Collapse
|