Abstract
The study of extracellular vesicles (EVs) is a rapidly evolving field, owing in large part to recent advances in the realization of their significant contributions to normal physiology and disease. Once discredited as cell debris, these membrane vesicles have now emerged as mediators of intercellular communication by interaction with target cells, drug and gene delivery, and as potentially versatile platforms of clinical biomarkers as a result of their distinctive protein, nucleic acid and lipid cargoes. While there are multiple classes of EVs released from almost all cell types, here we focus primarily on the biogenesis, fate and functional cargoes of microvesicles (MVs). MVs regulate many important cellular processes including facilitating cell invasion, cell growth, evasion of immune response, stimulating angiogenesis, drug resistance and many others.
Keywords: cancer, cell invasion, extracellular vesicles, microvesicles
1 |. INTRODUCTION
Extracellular vesicles (EVs) comprise a varied and heterogeneous group of particles released from cells originating largely from endosomes and/or the plasma membrane.1,2 Initially described as the disposal mechanisms used by cells to discard unwanted material, ensuing research has shown that EVs are important mediators of intercellular communication in normal physiology and pathophysiology.3–7 Recent studies have shown that EVs contain various proteins, lipids, glycolipids, glycoproteins and nucleic acids including DNA, mRNA and noncoding RNAs.8–18 Thus, EVs have the potential to deliver multiplexed information to surrounding tissues and systemically through the body.
The prevalence of EVs in disease has made them an attractive focus for the development of diagnostics and as a noninvasive or minimally invasive screening tool for detecting pathology in diseases such as cancer,19,20 diabetes21,22 and cardiovascular disease.23,24 EVs have been detected in multiple body fluids including urine, saliva and blood, making them readily available for analysis.22,25–27 Efforts have also been directed at investigating the therapeutic potential of EVs, both by preventing their formation,28 and as tools to package and deliver disease modulating proteins or drugs, for enhanced uptake by target cells. This could be in the form of harvested vesicles with innate therapeutic properties,29–31 vesicles which are harvested from cells which have been altered to enhance the presence of particular cargo,32–34 or vesicles which are loaded with cargo artificially, after their isolation from cells.33–35 Despite intense research in the biology and therapeutic implications of EVs in recent years, categorization of these vesicles into various subtypes is still rather tenuous. Here, we will describe this expanding and complex field of research by first outlining the subtypes of EVs with subsequent focus primarily on the biogenesis, cargo and fate of 1 EV subtype, namely, microvesicles (MVs).
2 |. TYPES OF EXTRACELLULAR VESICLES
Currently assigned into the broad categories of exosomes, MVs, or apoptotic bodies based upon their mechanism of formation, mode of release from cells, and size2 (Table 1), these classifications somewhat belie the heterogeneity that may exist within vesicle classes in terms of both cargo and functionality.36 Exosomes, the best characterized of the EV subtypes, are formed first as intraluminal vesicles within a multivesicular body (MVB), which are released into the extracellular space upon MVB fusion with the plasma membrane.37 MVs, sometimes referred to as ectosomes or microparticles, are formed by outward blebbing of the plasma membrane and subsequent fission of plasma membrane blebs.2 MVs derived from human cancer cells (Figure 1A) have received a good deal of attention because of their ability to participate in the horizontal transfer of signaling proteins between cancer cells and to contribute to their invasive activity. Apoptotic bodies are formed during cellular blebbing and fragmentation upon cell death2 (Figure 1B), and their impact upon other cells is not well-studied. Vesicles may also be released from nanotubular structures radiating from the plasma membrane.38 At present it appears that most, if not all, cell types form EVs,25,39 however, in general, cell stress and disease frequently upregulate their formation and alter the cargo contained within.40
TABLE 1.
Characteristics of natural and synthetic biological vesicles
| Vesicle type | Size | Origin | Function |
|---|---|---|---|
| Nanovesicle | 8–12 nm | • Unknown at present | • Intercellular communication • Promote cancer progression |
| Exosome | 50–100 nm | • Intraluminal vesicles of multivesicular body • Released upon MVB fusion with plasma membrane |
• Intercellular communication in healthy and diseased cells • Condition/educate the extracellular microenvironment • May promote disease pathogenesis |
| Microvesicle | 200–1000 nm+ | • Direct budding from plasma membrane • Released via acto-myosin-driven fission into extracellular space |
• Intercellular communication in healthy and diseased cells • Condition/educate the extracellular microenvironment • May promote disease pathogenesis |
| Apoptotic body | 500 nm+ | • Random plasma membrane blebs of cytoplasm and organelle fragments | • Breakdown of apoptotic cells • Potential intercellular signal transduction |
| Large oncosome | 1–10 μm | • Direct budding from plasma membrane | • Intercellular communication sent from cancer cells • Condition/educate the extracellular microenvironment • May promote disease pathogenesis |
| Giant plasma membrane vesicle | 1 μm+ | • Formed by artificially stimulating cells to shed blebs of lipid-rich plasma membrane | • Study plasma membrane protein behavior • Study plasma membrane lipid organization |
| Uni- and multilamellar vesicles | Unilamellar: Small: <100 nm; Large: 100–400 nm; Giant: 1–100 μm; Multilamellar: Small: 80–120 nm; Large: 200–3000 nm | • Synthesized in vitro as mixtures of cholesterol and phospholipids in an organic solvent. May require sonic or mechanical processing to form the single lipid bilayer (unilamellar) or multi-bilayered (multilamellar) vesicles | • Study properties of the plasma membrane such as lipid rafts and phase separation • Delivery of therapeutics |
FIGURE 1.
Micrographs of EVs. A, Microvesicles (arrows) are shed by invasive LOX melanoma cells invading fibrillar collagen matrix (confocal reflection of matrix shown as red). Cells were labeled for β1 integrin (green). B, EVs (arrows) released by melanoma cells following apoptosis induction with hydrogen peroxide. Cells were labeled for β1 integrin in red, and cleaved caspase-3 is shown in green
Overall, EVs comprise a wide spectrum of vesicles ranging from 8 nm to several microns. MVs generally range from 200 nm to several microns in diameter, whereas exosomes are smaller, and range from 50 to 100 nm in diameter.2,41 Nanovesicles ranging from 8 to 12 nm in peripheral blood have also been recently described,42 and these very small vesicles may be errantly isolated with exosomes using many of the currently utilized isolation protocols. Due to the considerable heterogeneity in the size of MVs, there are likely to be subpopulations within this category of vesicles. The term “oncosomes” has been used to describe MVs or even all EVs that have been released from cancer cells.43 However, of note, large oncosomes (LO) refer to a subtype of plasma membrane-derived MVs, 1 to 10 μm in diameter, which are released from tumor cells.44,45 Nomenclature, as well as sizes attributed to various EV subtypes, has not been without debate.46,47 Complicating this matter is the fact that isolation methods for EV subtypes are still in development and consensus best-practices are not yet reached, leading to differences in the populations of vesicles being evaluated between studies.36,48 As a result of the lack of clear distinction between EV subtypes, in many studies collective EV populations have been utilized. For the purposes of this review, we use the term “extracellular vesicles” for all studies that have not been attributed to a given population or when the isolation methods suggest that multiple subtypes were likely being investigated. We have attributed characteristics to “microvesicles” if the methodologies for vesicle isolation have enriched for the MV fraction.
It is likely that mode of biogenesis and EV cargo will play a critical role in defining criteria for classification. In this vein, recently, subpopulations of EVs have been described by characterization of cargo, size and density,36,49,50 as well their effects upon recipient cells.49 It is possible, if not likely, that as the field moves forward additional vesicle classes, or modifications to the current classification systems, will emerge.
Although not classified along with the above, additional vesicle types have been described primarily as research tools to study lipid and protein behavior. These may be formed upon artificial stimulation of cells, as in giant plasma membrane vesicles (GPMVs), which are formed upon the treatment of live cells with stimuli such as laser irradiation, salt treatment or the chemicals paraformaldehyde and dithiothreitol.51–53 This results in the formation of large (micron sized) vesicles which are frequently used to study lipid and protein organization at the plasma membrane52,54,55 (Figure 2). Additional vesicle types are prepared fully synthetically in vitro, as in small, large and giant uni- and multilamellar lipid vesicles,56 including variants referred to as artificial plasma membrane vesicles.57 These synthetic vesicles have also been used for therapeutic purposes, providing improved stability and uptake of drugs and nucleic acids,58,59 likely exploiting the same mechanisms utilized by naturally-derived vesicles.
FIGURE 2.
Artificially-stimulated EVs. Phase contrast image of GPMVs (arrows) formed by live HeLa cells after stimulation with paraformaldehyde and dithiothrietol. Scale bar is 20 μm
3 |. MICROVESICLE BIOGENESIS
How cargo is trafficked to MVs is still an area of ongoing investigation, and is less well characterized relative to exosomes. However, several mechanisms of cargo trafficking have emerged from investigations in tumor cells. In general, MVs appear to form by the outward pinching of the plasma membrane. ARF6-positive recycling vesicles have been found to traffic cargo to the cell surface for incorporation into MVs.3 The v-SNARE VAMP3 has been shown to regulate the delivery of cargo such as MT1-MMP to facilitate matrix proteolysis during tumor cell invasion, in a process dependent on the association of VAMP3 with CD9.60
The biogenesis and release of MVs from the cell may be regulated by multiple mechanisms. MVs are generated from sites of high membrane blebbing,2,45 and their formation is stimulated in cells invading through compliant matrices.61 Interestingly in this regard, cells invading more rigid matrices frequently utilize a different invasive structure termed invadopodia at their ventral surface to mediate matrix proteolysis, and studies have indicated that exosomes are released at invadopodia to facilitate invasion,62 with the integral invadopodia protein cortactin regulating the trafficking and docking of MVBs.63 Plasma membrane bleb formation is regulated by a host of factors which modulate deformability and bending of the membrane, including the lipid composition and organization of the peripheral cytoskeleton, to alter membrane fluidity and deformability. MVs display unique lipid characteristics, such as the externalization of the phospholipid phosphatidylserine (PS),64 which has been demonstrated to promote uptake by recipient cells.65 The phospholipid lysophosphatidylcholines, the sphingolipid sphingomyelins, and acylcarnitines, the fatty acyl esters of L-carnitine, have also been found to be enriched in MVs.66 The prominent plasma membrane lipid cholesterol is thought to play an important role in MV formation, as its depletion reduces MV formation.67 Ceramide, a cone-shaped lipid which promotes membrane bending, has also been shown to regulate MV formation.68 Once loaded with cargo, the bleb must be pinched free from the cell in a process governed by acto-myosin contraction. This requires a tightly regulated balance of cytoskeletal elements which promote or suppress the required blebbing and pinching. Rho family GTPases have emerged as critical mediators of MV formation, which is not surprising, given their integral roles in regulating actin reorganization and cortical contractility.61,69 RhoA activity has been demonstrated to promote MV formation, through the downstream kinases ROCK (Rho-associated coiled-coil containing kinases) and ERK (extracellular signal-regulated kinases).61 In tumor cells this is antagonized by an elevation in Rac1 activity when cells encounter firm extracellular matrix conditions that are not conducive to MV-mediated invasion. Activation of myosin light chain (MLC) at MV “necks” is required for MV fission into the extracellular space, and this is regulated by increased activity of MLC kinase and the suppression of MLC phosphatase,3,61 mediated by RhoA activity, to promote enhanced myosin contractility. Cofilin phosphorylation downstream of Rho also promotes MV generation.69
Several additional mechanisms have been shown to affect MV formation. The extracellular concentration of calcium can impact MV formation, with increased calcium eliciting increased vesiculation.70 This is particularly interesting in light of the fact that calcium signaling is frequently dysregulated in cancer,71,72 and multiple oncogenes and tumor suppressors impact calcium regulation.72 Increased calcium levels have been demonstrated to induce membrane phospholipid scrambling and increased MV formation in erythrocytes and platelets,73 and a similar mechanism may be at work in cancer cells. G-protein coupled receptor 30 (GRP30) has been shown to stimulate the formation of EMMPRIN (extracellular matrix metalloproteinase inducer)-containing MVs from uterine cells,74 which may contribute to tumorigenesis or cancer progression. Hypoxia, a common occurrence within solid tumors which is associated with tumor progression and therapy resistance,75 has also been shown to promote MV release via a cellular process mediated by hypoxia-inducible factors (HIFs) and Rab22a.76 Actin deimination, whereupon protein arginine deiminases (PADs) catalyze the hydrolysis of peptidyl-arginine to peptidyl-citrulline, has also been associated with facilitating MV biogenesis.77 Additional work has shown a role for the ESCRT protein TSG101, in concert with arrestin domain-containing protein 1 - (ARRDC1), in regulating MV formation at the plasma membrane, in a process that also requires the VPS4 ATPase for vesicle release.78
Suppression of the actin-nucleator diaphanous-related formin 3 - (DIAPH3) enhances formation of LO.79 Interestingly in this regard, analysis of primary and metastatic human prostate tumors reveals a significantly higher frequency of deletion of the locus encoding DIAPH3 in metastatic tumors in comparison to organ-confined tumors. Additionally, the activation of EGFR (epidermal growth factor receptor),79 overexpression of membrane-targeted Akt1,79 caveolin-1 overexpression,45 and stimulation with heparin-binding EGF (epidermal growth factor)-like growth factor combined with p38MAPK inhibition80 have also been found to stimulate LO formation.
It is important to mention that there is overlap in the molecules which mediate the biogenesis of multiple vesicle types, best studied in exosomes and MVs. In these populations, proteins including ARF6 and ESCRT complex components such as TSG101, and lipids such as ceramide, have been shown to regulate the formation of both vesicle types,3,27,49,66,68,78,81 indicating that there may be coupling between the biogenesis of different vesicle families. Further work is required to determine if the aforementioned regulators work in concert or if the cellular machinery engaged is dependent on the content of the EVs being released.
4 |. UPTAKE OF EVS INTO CELLS
Modes of MV cargo uptake by recipient cells include simple plasma membrane-EV fusion with direct cargo deposition into the cytoplasm, and the uptake of intact vesicles via multiple mechanisms for trafficking to an endosomal or lysosomal compartment.82–84 EV uptake by recipient cells is carried out by a variety of modes, including clathrin and caveolin-mediated endocytosis,85 lipid raft endocytosis,86,87 phagocytosis83 and micropinocytosis,85,88 likely guided by vesicle membrane composition and surface protein profile.83 Further, low pH as provided by tumor microenvironments would be conducive to the fusion of MV membranes with recipient cells. Studies have shown that the cellular uptake of MVs is enhanced by the inclusion of a pH-sensitive peptide to promote membrane fusion of MV and endosomal membranes upon internalization.89 Low pH has also been shown to promote exosome release and uptake,90 indicating that this may be a conserved EV characteristic. Recent studies have indicated an importance for actin polymerization and dynamin, a key protein involved in membrane budding and fission, in macropinocytic-like MV uptake by alveolar epithelial cells.88 An important regulator in EV internalization is heparan sulfate proteoglycans (HSPGs), and treating EVs with heparin reduces their uptake.91,92 Blocking the scavenger receptor type B1 (SR-B1), a receptor for HDL (high-density lipoproteins) which mediates the uptake of extracellular material, with HDL nanoparticles also blocks exosome uptake by altering lipid raft composition and SR-B1 clustering,93 and similar mechanisms may regulate MV uptake as well. It appears that the mode of uptake is likely dependent on the cell type and the ligands on the recipient cells, although much remains to be learned in this regard. Interactions of EVs with target cells have been shown to trigger a host of cellular signaling cascades, and their uptake in recipient cells may facilitate transfer of functional cargo or its degradation.83
5 |. FUNCTIONAL CARGOES IN MICROVESICLES DERIVED FROM TUMOR CELLS
The cargoes contained within EVs reflect both the intracellular origin of the cargo as well as the cell type from which the vesicle was derived. We refer readers to the many profiling studies which have been published on this subject for further information.36,66,94–100 Prototypical exosome markers include tetraspannins such as CD9, CD63 and CD81, and ESCRT proteins Alix and TSG101.37,101 Unique and universal MV markers are somewhat less well-defined. At present they are best studied in tumor cells, and frequently include selectins, flotillin-2, ARF6 and CD40.3,102 As indicated above, an ever-evolving body of work reveals that many of these markers may not be exclusive to a particular class of vesicle,36 underscoring the heterogeneity of EVs and the potential overlap in functions between vesicle classes.
In normal cells, EVs have been implicated in regulating many vital physiological activities, ranging from the maintenance of stem cell plasticity,103 communication between mother and fetus during pregnancy,104 and blood clotting105; however, it is their role in disease which has garnered the most attention. MVs have been shown to play a role in a wide variety of pathologies, including cardiovascular disease,24,106–111 diabetes,21,107,111–113 and neurodegeneration.68,114–116 The functional roles of EV cargoes have been best characterized in cancer progression and the rest of this section will focus on tumor-derived EVs. EV cargo from invasive tumor cells contains cargoes such as matrix metalloproteases to enable extracellular matrix proteolysis and subsequent cell invasion.2,62,64 Cancer cell-derived EVs have also been characterized to contain extracellular matrix proteins such as fibronectin, which may mediate interaction between the vesicles and target cells,117 as well as facilitate anchorage-independent growth in recipient cells,118 and promote the induction of pro-inflammatory cytokines in immune cells.119 Fibronectin in exosomes has been shown to be an important cargo to promote directional motility and chemotaxis in tumor cells,120,121 and the abundance of fibronectin on EVs in the plasma of individuals with breast cancer has indicated its potential as a diagnostic biomarker.122
The chemoattractant potential of EV cargoes provides the potential for invasive cells to deposit cues to facilitate the directed movement of subsequent cells, and the recruitment of other associated cell types such as stromal and endothelial cells.123 Oncogenic cargo, such as the mutant EGFR variant EGFRvIII can stimulate transformation of naïve cells.124 Notably absent from tumor cell MVs are cortactin and Tks5,2 prominent markers of the aforementioned invadopodia invasive structures.
One of the key roles of MVs in tumor progression is the ability to alter the behavior of other cell types to facilitate tumor cell survival. In this regard, MVs may facilitate tumor progression by directing cytotoxic cargo toward circulating immune cells, as demonstrated by melanoma cells inciting apoptosis in lymphocytes via proapoptotic Fas ligand containing MVs.125 A similar effect was induced by melanoma FasL and TRAIL-containing MVs on T-cell blasts,126 again promoting tumor cell survival. MiR-214 has been shown to be released in tumor cell MVs, resulting in downregulation of PTEN in T regulatory cells while promoting Treg expansion and host immune tolerance.127 MVs isolated from esophageal cancer cells have been shown to promote the differentiation of naïve B cells into TGFβ-producing regulatory B cells, which suppress immunity derived from CD8+ T cells.128 MVs have also been shown to promote angiogenesis, carrying cargo such as TGFβ, stimulating migration, proliferation and tube formation in endothelial cells.129 A recent study similarly found that MVs released from oral cancer cells stimulate angiogenesis via sonic hedgehog and RhoA.130
Furthering the profound implications of MVs in tumor survival and progression are their roles in drug resistance. MVs have been noted to mediate the efflux of chemotherapeutics,28,131 promoting cell survival, and the number of MVs formed upon drug treatment has been shown to correlate with their chemotherapeutic resistance.131 MVs have also been demonstrated to transfer the drug resistance molecule p-glycoprotein in ovarian cancer cells, mediating drug-resistance in previously drug-sensitive cells.132
Additionally, MVs are thought to prepare a niche for metastatic tumor cells, allowing for the colonization of a secondary site. This can be accomplished utilizing a complex set of cargo, including proteases for matrix degradation and impairment of vascular integrity, molecules which alter stromal stiffness and organization, deposition of chemoattractant molecules, angiogenic stimulation and changes in immune cell behavior.133–136
EVs, including MVs, have been characterized to contain a broad range of nucleic acids, including nuclear and mitochondrial DNA, mRNA and noncoding RNAs such as miRNA, circular RNA and long noncoding RNA.8–17 This presents the potential for wide-ranging implications in cancer as a potential efflux mechanism for “unwanted” nucleic acids as well as to transfer DNA and RNA which directly support oncogenesis and tumor progression. Packaging within EVs likely provides protection from degradation as the cargo is shuttled from the cell of origin to the recipient, as well as enhanced uptake by targeted cells.137 Potential diagnostic implications of this nucleic acid content are revealed in multiple cancers. miRNA expression has been noted to be altered in the EVs isolated from a host of cancer types, including lung, prostate, pancreatic, colorectal, melanoma, glioblastoma, hepatocellular, leukemic and ovarian.138 For example, in chronic myelogenous leukemia, an upregulation of miRNAs associated with tumorigenesis was noted in blast-derived MVs.139 The therapeutic use of nucleic acids in EVs is also an area of investigation, to evaluate their potential as a tool for the delivery of gene therapy.137
A recent analysis of MVs in the blood of patients with locally advanced or metastatic lung, breast, colorectal or head & neck cancers indicated that the profile of the protein markers MUC1, EGFR and EpCAM correlated with tumor subtype, whereas the MMP-inducer EMMPRIN was increased regardless of tumor type and the increased abundance of EMMPRIN-positive MVs correlated strongly with poor overall survival.19 Another recent study demonstrated the differential phosphorylation of an array of proteins found in exosomes and MVs isolated from plasma in breast cancer patients as compared to healthy controls.140 In plasma cell dyscrasias such as multiple myeloma, free antibody light chains in the blood are a prominent characteristic of disease, and these may be circulated through the bloodstream within MVs that also contain Hsp70, annexin V and c-src,141 presenting diagnostic possibilities and indicating the mode of cargo transfer between cells. Other studies have indicated that the overall prevalence of MVs in blood alone may be a diagnostic indicator, such as the increase in MVs that has been noted in multiple myeloma142 and in gastric cancer.143
6 |. CONCLUDING STATEMENTS
Increasing evidence supports a role for MVs, as well as other types of EVs, in intercellular communication. This, coupled with the findings that EVs derived from many sites in the body can make their way into body fluids, have led to exciting possibilities of their use in diagnostic and therapeutic applications. However, there is need for general consensus on classification and content of these shed vesicles. Further research on biogenesis and cargoes of EVs will greatly facilitate more precise discrimination between EV populations and functional characterization of EV subtypes.
ACKNOWLEDGMENTS
We acknowledge support for related research from the National Cancer Institute, The Catherine Peachey Fund, and 100 Voices of Hope through the Indiana University Melvin and Bren Simon Cancer Center to CDS.
Funding information
National Cancer Institute, Grant/Award number: R01CA115316
Footnotes
Editorial Process File: is available in the online version of this article.
REFERENCES
- 1.Akers JC, Gonda D, Kim R, Carter BS, Chen CC. Biogenesis of extracellular vesicles (EV): exosomes, microvesicles, retrovirus-like vesicles, and apoptotic bodies. J Neuro-Oncol. 2013;113(1):1–11. 10.1007/s11060-013-1084-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.D’Souza-Schorey C, Clancy JW. Tumor-derived microvesicles: shedding light on novel microenvironment modulators and prospective cancer biomarkers. Genes Dev. 2012;26(12):1287–1299. 10.1101/gad.192351.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Muralidharan-Chari V, Clancy J, Plou C, et al. ARF6-regulated shedding of tumor cell-derived plasma membrane microvesicles. Curr Biol. 2009;19(22):1875–1885. 10.1016/j.cub.2009.09.059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Cocucci E, Racchetti G, Meldolesi J. Shedding microvesicles: artefacts no more. Trends Cell Biol. 2009;19(2):43–51. 10.1016/j.tcb.2008.11.003. [DOI] [PubMed] [Google Scholar]
- 5.Barteneva NS, Maltsev N, Vorobjev IA. Microvesicles and intercellular communication in the context of parasitism. Front Cell Infect Microbiol. 2013;3:49 10.3389/fcimb.2013.00049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Schorey JS, Bhatnagar S. Exosome function: from tumor immunology to pathogen biology. Traffic. 2008;9(6):871–881. 10.1111/j.1600-0854.2008.00734.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Regev-Rudzki N, Wilson DW, Carvalho TG, et al. Cell-cell communication between malaria-infected red blood cells via exosome-like vesicles. Cell. 2013;153(5):1120–1133. 10.1016/j.cell.2013.04.029. [DOI] [PubMed] [Google Scholar]
- 8.Abels ER, Breakefield XO. Introduction to extracellular vesicles: biogenesis, RNA cargo selection, content, release, and uptake. Cell Mol Neurobiol. 2016;36(3):301–312. 10.1007/s10571-016-0366-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Waldenström A, Gennebäck N, Hellman U, Ronquist G. Cardiomyocyte microvesicles contain DNA/RNA and convey biological messages to target cells. PLoS One. 2012;7(4):e34653 10.1371/journal.pone.0034653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Skog J, Würdinger T, van Rijn S, et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol. 2008;10(12):1470–1476. 10.1038/ncb1800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Pan Q, Ramakrishnaiah V, Henry S, et al. Hepatic cell-to-cell transmission of small silencing RNA can extend the therapeutic reach of RNA interference (RNAi). Gut. 2012;61(9):1330–1339. 10.1136/gutjnl-2011-300449. [DOI] [PubMed] [Google Scholar]
- 12.Smalheiser NR. Exosomal transfer of proteins and RNAs at synapses in the nervous system. Biol Direct. 2007;2(1):35 10.1186/1745-6150-2-35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Kim KM, Abdelmohsen K, Mustapic M, Kapogiannis D, Gorospe M. RNA in extracellular vesicles. Wiley Interdiscip Rev RNA. 2017;8(4): e1413 10.1002/wrna.1413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Noerholm M, Balaj L, Limperg T, et al. RNA expression patterns in serum microvesicles from patients with glioblastoma multiforme and controls. BMC Cancer. 2012;12(1):22 10.1186/1471-2407-12-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Yuan A, Farber EL, Rapoport AL, et al. Transfer of microRNAs by embryonic stem cell microvesicles. PLoS One. 2009;4(3):e4722 10.1371/journal.pone.0004722. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Ma P, Pan Y, Li W, et al. Extracellular vesicles-mediated noncoding RNAs transfer in cancer. J Hematol Oncol. 2017;10(1):57 10.1186/s13045-017-0426-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Guescini M, Genedani S, Stocchi V, Agnati LF. Astrocytes and Glioblastoma cells release exosomes carrying mtDNA. J Neural Transm. 2010;117(1):1–4. 10.1007/s00702-009-0288-8. [DOI] [PubMed] [Google Scholar]
- 18.Desrochers LM, Antonyak MA, Cerione RA. Extracellular vesicles: satellites of information transfer in cancer and stem cell biology. Dev Cell. 2016;37(4):301–309. 10.1016/j.devcel.2016.04.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Menck K, Bleckmann A, Wachter A, et al. Characterisation of tumour-derived microvesicles in cancer patients’ blood and correlation with clinical outcome. J Extracell Vesicles. 2017;6(1):1340745 10.1080/20013078.2017.1340745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Lee TH, D’Asti E, Magnus N, Al-Nedawi K, Meehan B, Rak J. Microvesicles as mediators of intercellular communication in cancer—the emerging science of cellular “debris”. Semin Immunopathol. 2011; 33(5):455–467. 10.1007/s00281-011-0250-3. [DOI] [PubMed] [Google Scholar]
- 21.Lakhter AJ, Sims EK. Minireview: emerging roles for extracellular vesicles in diabetes and related metabolic disorders. Mol Endocrinol. 2015;29(11):1535–1548. 10.1210/me.2015-1206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.La MV, Fierabracci A. Insights into the diagnostic potential of extracellular vesicles and their miRNA signature from liquid biopsy as early biomarkers of diabetic micro/macrovascular complications. Int J Mol Sci. 2017;18(9):1974 10.3390/ijms18091974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Ailawadi S, Wang X, Gu H, Fan G-C. Pathologic function and therapeutic potential of exosomes in cardiovascular disease. Biochim Biophys Acta. 2015;1852(1):1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Jansen F, Nickenig G, Werner N. Extracellular vesicles in cardiovascular disease. Circ Res. 2017;120(10):1649–1657. 10.1161/CIRCRESAHA.117.310752. [DOI] [PubMed] [Google Scholar]
- 25.Ciardiello C, Cavallini L, Spinelli C, et al. Focus on extracellular vesicles: new Frontiers of cell-to-cell communication in cancer. Int J Mol Sci. 2016;17(2):175 10.3390/ijms17020175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Xu R, Greening DW, Zhu H-J, Takahashi N, Simpson RJ. Extracellular vesicle isolation and characterization: toward clinical application. J Clin Invest. 2016;126(4):1152–1162. 10.1172/JCI81129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol. 2013;200(4):373–383. 10.1083/jcb.201211138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Kosgodage US, Trindade RP, Thompson PR, Inal JM, Lange S. Chloramidine/Bisindolylmaleimide-I-mediated inhibition of exosome and microvesicle release and enhanced efficacy of cancer chemotherapy. Int J Mol Sci. 2017;18(5). 10.3390/ijms18051007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Kalimuthu S, Gangadaran P, Li XJ, et al. In vivo therapeutic potential of mesenchymal stem cell-derived extracellular vesicles with optical imaging reporter in tumor mice model. Sci Rep. 2016;6(1):30418 10.1038/srep30418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Wu S, Ju G-Q, Du T, Zhu Y-J, Liu G-H. Microvesicles derived from human umbilical cord wharton’s jelly mesenchymal stem cells attenuate bladder tumor cell growth in vitro and in vivo. PLoS One. 2013; 8(4):e61366 10.1371/journal.pone.0061366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Bruno S, Collino F, Deregibus MC, Grange C, Tetta C, Camussi G. Microvesicles derived from human bone marrow mesenchymal stem cells inhibit tumor growth. Stem Cells Dev. 2013;22(5):758–771. 10.1089/scd.2012.0304. [DOI] [PubMed] [Google Scholar]
- 32.Mizrak A, Bolukbasi MF, Ozdener GB, et al. Genetically engineered microvesicles carrying suicide mRNA/protein inhibit schwannoma tumor growth. Mol Ther. 2013;21(1):101–108. 10.1038/mt.2012.161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Zhu L, Dong D, Yu Z-L, Zhao Y-F, Pang D-W, Zhang Z-L. Folate-engineered microvesicles for enhanced target and synergistic therapy toward breast cancer. ACS Appl Mater Interfaces. 2017;9(6): 5100–5108. 10.1021/acsami.6b14633. [DOI] [PubMed] [Google Scholar]
- 34.Armstrong JPK, Holme MN, Stevens MM. Re-engineering extracellular vesicles as smart nanoscale therapeutics. ACS Nano. 2017;11(1): 69–83. 10.1021/acsnano.6b07607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Saari H, Lázaro-Ibáñez E, Viitala T, Vuorimaa-Laukkanen E, Siljander P, Yliperttula M. Microvesicle- and exosome-mediated drug delivjery enhances the cytotoxicity of Paclitaxel in autologous prostate cancer cells. J Control Release. 2015;220(pt B):727–737. 10.1016/j.jconrel.2015.09.031. [DOI] [PubMed] [Google Scholar]
- 36.Kowal J, Arras G, Colombo M, et al. Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes. Proc Natl Acad Sci USA. 2016;113(8): E968–E977. 10.1073/pnas.1521230113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Hessvik NP, Llorente A. Current knowledge on exosome biogenesis and release. Cell Mol Life Sci. 2017;75:1–16. 10.1007/s00018-017-2595-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Rilla K, Siiskonen H, Tammi M, Tammi R. Hyaluronan-coated extracellular vesicles—a novel link between Hyaluronan and cancer. Adv Cancer Res. 2014;123:121–148. 10.1016/B978-0-12-800092-2.00005-8. [DOI] [PubMed] [Google Scholar]
- 39.Pegtel DM, Peferoen L, Amor S. Extracellular vesicles as modulators of cell-to-cell communication in the healthy and diseased brain. Philos Trans R Soc Lond Ser B Biol Sci. 2014;369(1652). 10.1098/rstb.2013.0516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Kreger BT, Dougherty AL, Greene KS, Cerione RA, Antonyak MA. Microvesicle cargo and function changes upon induction of cellular transformation. J Biol Chem. 2016;291(38):19774–19785. 10.1074/jbc.M116.725705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Colombo M, Moita C, van Niel G, et al. Analysis of ESCRT functions in exosome biogenesis, composition and secretion highlights the heterogeneity of extracellular vesicles. J Cell Sci. 2013;126(24): 5553–5565. [DOI] [PubMed] [Google Scholar]
- 42.Zhang H-G, Cao P, Teng Y, et al. Isolation, identification, and characterization of novel nanovesicles. Oncotarget. 2016;7(27): 41346–41362. 10.18632/oncotarget.9325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Meehan B, Rak J, Di Vizio D. Oncosomes - large and small: what are they, where they came from? J Extracell Vesicles. 2016;5:33109 10.3402/JEV.V5.33109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Minciacchi VR, You S, Spinelli C, et al. Large oncosomes contain distinct protein cargo and represent a separate functional class of tumor-derived extracellular vesicles. Oncotarget. 2015;6(13): 11327–11341. 10.18632/oncotarget.3598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Di Vizio D, Morello M, Dudley AC, et al. Large Oncosomes in human prostate cancer tissues and in the circulation of mice with metastatic disease. Am J Pathol. 2012;181(5):1573–1584. 10.1016/j.ajpath.2012.07.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Witwer KW, Buzás EI, Bemis LT, et al. Standardization of sample collection, isolation and analysis methods in extracellular vesicle research. J Extracell Vesicles. 2013;2(1):20360 10.3402/jev.v2i0.20360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Gould SJ, Raposo G. As we wait: coping with an imperfect nomenclature for extracellular vesicles. J Extracell Vesicles. 2013;2 10.3402/jev.v2i0.20389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Furi I, Momen-Heravi F, Szabo G. Extracellular vesicle isolation: present and future. Ann Transl Med. 2017;5(12):263 10.21037/atm.2017.03.95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Willms E, Johansson HJ, Mäger I, et al. Cells release subpopulations of exosomes with distinct molecular and biological properties. Sci Rep. 2016;6(1):22519 10.1038/srep22519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Bobrie A, Colombo M, Krumeich S, Raposo G, Théry C. Diverse subpopulations of vesicles secreted by different intracellular mechanisms are present in exosome preparations obtained by differential ultracentrifugation. J Extracell Vesicles. 2012;1(1):18397 10.3402/jev.v1i0.18397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Sarabipour S, Chan RB, Zhou B, Di Paolo G, Hristova K. Analytical characterization of plasma membrane-derived vesicles produced via osmotic and chemical vesiculation. Biochim Biophys Acta. 2015; 1848(7):1591–1598. 10.1016/j.bbamem.2015.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Sezgin E, Kaiser H-J, Baumgart T, Schwille P, Simons K, Levental I. Elucidating membrane structure and protein behavior using giant plasma membrane vesicles. Nat Protoc. 2012;7(6):1042–1051. 10.1038/nprot.2012.059. [DOI] [PubMed] [Google Scholar]
- 53.Del Piccolo N, Placone J, He L, Agudelo SC, Hristova K. Production of plasma membrane vesicles with chloride salts and their utility as a cell membrane mimetic for biophysical characterization of membrane protein interactions. Anal Chem. 2012;84(20):8650–8655. 10.1021/ac301776j. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Baumgart T Large-scale fluid/fluid phase separation of proteins and lipids in giant plasma membrane vesicles. Proc Natl Acad Sci USA. 2007;104(9):3165–3170. http://www.pnas.org/content/104/9/3165.short. Accessed July 11, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Levental I, Byfield FJ, Chowdhury P, Gai F, Baumgart T, Janmey PA. Cholesterol-dependent phase separation in cell-derived giant plasma-membrane vesicles. Biochem J. 2009;424(2):163–167. 10.1042/BJ20091283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Schmitt C, Lippert AH, Bonakdar N, Sandoghdar V, Voll LM. Compartmentalization and transport in synthetic vesicles. Front Bioeng Biotechnol. 2016;4:19 10.3389/fbioe.2016.00019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Lin Q, London E. Preparation of artificial plasma membrane mimicking vesicles with lipid asymmetry. PLoS One. 2014;9(1):e87903 10.1371/journal.pone.0087903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Akbarzadeh A, Rezaei-Sadabady R, Davaran S, et al. Liposome: classification, preparation, and applications. Nanoscale Res Lett. 2013; 8(1):102 10.1186/1556-276X-8-102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Allen TM, Cullis PR. Liposomal drug delivery systems: From concept to clinical applications. Adv Drug Deliv Rev. 2013;65(1):36–48. 10.1016/j.addr.2012.09.037. [DOI] [PubMed] [Google Scholar]
- 60.Clancy JW, Sedgwick A, Rosse C, et al. Regulated delivery of molecular cargo to invasive tumour-derived microvesicles. Nat Commun. 2015;6:6919 10.1038/ncomms7919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Sedgwick AE, Clancy JW, Olivia Balmert M, D’Souza-Schorey C. Extracellular microvesicles and invadopodia mediate non-overlapping modes of tumor cell invasion. Sci Rep. 2015;5 10.1038/srep14748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Hoshino D, Kirkbride KC, Costello K, et al. Exosome secretion is enhanced by invadopodia and drives invasive behavior. Cell Rep. 2013;5(5):1159–1168. 10.1016/j.celrep.2013.10.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Sinha S, Hoshino D, Hong NH, et al. Cortactin promotes exosome secretion by controlling branched actin dynamics. J Cell Biol. 2016; 214(2):197–213. 10.1083/jcb.201601025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Muralidharan-Chari V, Clancy JW, Sedgwick A, D’Souza-Schorey C. Microvesicles: mediators of extracellular communication during cancer progression. J Cell Sci. 2010;123(pt 10):1603–1611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Wei X, Liu C, Wang H, et al. Surface Phosphatidylserine is responsible for the internalization on microvesicles derived from hypoxia-induced human bone marrow mesenchymal stem cells into human endothelial cells. PLoS One. 2016;11(1):e0147360 10.1371/journal.pone.0147360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Haraszti RA, Didiot M-C, Sapp E, et al. High-resolution proteomic and lipidomic analysis of exosomes and microvesicles from different cell sources. J Extracell Vesicles. 2016;5:32570 10.3402/JEV.V5.32570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Del Conde I, Shrimpton CN, Thiagarajan P, López JA. Tissue-factor-bearing microvesicles arise from lipid rafts and fuse with activated platelets to initiate coagulation. Blood. 2005;106(5): 1604–1611. 10.1182/blood-2004-03-1095. [DOI] [PubMed] [Google Scholar]
- 68.Budnik V, Ruiz-Cañada C, Wendler F. Extracellular vesicles round off communication in the nervous system. Nat Rev Neurosci. 2016;17(3): 160–172. 10.1038/nrn.2015.29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Li B, Antonyak MA, Zhang J, Cerione RA. RhoA triggers a specific signaling pathway that generates transforming microvesicles in cancer cells. Oncogene. 2012;31(45):4740–4749. 10.1038/onc.2011.636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Crawford S, Diamond D, Brustolon L, Penarreta R. Effect of increased extracellular ca on microvesicle production and tumor spheroid formation. Cancer Microenviron. 2010;4(1):93–103. 10.1007/s12307-010-0049-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Stewart TA, Yapa KTDS, Monteith GR. Altered calcium signaling in cancer cells. Biochim Biophys Acta Biomembr. 2015;1848(10): 2502–2511. 10.1016/j.bbamem.2014.08.016. [DOI] [PubMed] [Google Scholar]
- 72.Monteith GR, Prevarskaya N, Roberts-Thomson SJ. The calcium–cancer signalling nexus. Nat Rev Cancer. 2017;17(6):367–380. 10.1038/nrc.2017.18. [DOI] [PubMed] [Google Scholar]
- 73.Bucki R, Bachelot-Loza C, Zachowski A, Giraud F, Sulpice JC. Calcium induces phospholipid redistribution and microvesicle release in human erythrocyte membranes by independent pathways. Biochemistry. 1998;37(44):15383–15391. 10.1021/bi9805238. [DOI] [PubMed] [Google Scholar]
- 74.Burnett LA, Light MM, Mehrotra P, Nowak RA. Stimulation of GPR30 increases release of EMMPRIN-containing microvesicles in human uterine epithelial cells. J Clin Endocrinol Metab. 2012;97(12): 4613–4622. 10.1210/jc.2012-2098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Hockel M, Vaupel P. Tumor hypoxia: definitions and current clinical, biologic, and molecular aspects. J Natl Cancer Inst. 2001;93(4): 266–276. 10.1093/jnci/93.4.266. [DOI] [PubMed] [Google Scholar]
- 76.Wang T, Gilkes DM, Takano N, et al. Hypoxia-inducible factors and RAB22A mediate formation of microvesicles that stimulate breast cancer invasion and metastasis. Proc Natl Acad Sci USA. 2014; 111(31):E3234–E3242. 10.1073/pnas.1410041111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Kholia S, Jorfi S, Thompson PR, et al. A novel role for peptidylarginine deiminases in microvesicle release reveals therapeutic potential of PAD inhibition in sensitizing prostate cancer cells to chemotherapy. J Extracell Vesicles. 2015;4:26192 10.3402/JEV.V4.26192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Nabhan JF, Hu R, Oh RS, Cohen SN, Lu Q. Formation and release of arrestin domain-containing protein 1-mediated microvesicles (ARMMs) at plasma membrane by recruitment of TSG101 protein. Proc Natl Acad Sci USA. 2012;109(11):4146–4151. 10.1073/pnas.1200448109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Di Vizio D, Kim J, Hager MH, et al. Oncosome formation in prostate cancer: association with a region of frequent chromosomal deletion in metastatic disease. Cancer Res. 2009;69(13):5601–5609. 10.1158/0008-5472.CAN-08-3860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Kim J, Morley S, Le M, et al. Enhanced shedding of extracellular vesicles from amoeboid prostate cancer cells: potential effects on the tumor microenvironment. Cancer Biol Ther. 2014;15(4):409–418. 10.4161/cbt.27627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Ghossoub R, Lembo F, Rubio A, et al. Syntenin-ALIX exosome biogenesis and budding into multivesicular bodies are controlled by ARF6 and PLD2. Nat Commun. 2014;5:3477 10.1038/ncomms4477. [DOI] [PubMed] [Google Scholar]
- 82.McKelvey KJ, Powell KL, Ashton AW, Morris JM, McCracken SA. Exosomes: mechanisms of uptake. J Circ Biomark. 2015;4:7 10.5772/61186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Mulcahy LA, Pink RC, Carter DRF. Routes and mechanisms of extracellular vesicle uptake. J Extracell Vesicles. 2014;3 10.3402/jev.v3.24641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.French KC, Antonyak MA, Cerione RA. Extracellular vesicle docking at the cellular port: extracellular vesicle binding and uptake. Semin Cell Dev Biol. 2017;67:48–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Costa Verdera H, Gitz-Francois JJ, Schiffelers RM, Vader P. Cellular uptake of extracellular vesicles is mediated by clathrin-independent endocytosis and macropinocytosis. J Control Release. 2017;266: 100–108. 10.1016/j.jconrel.2017.09.019. [DOI] [PubMed] [Google Scholar]
- 86.Svensson KJ, Christianson HC, Wittrup A, et al. Exosome uptake depends on ERK1/2-heat shock protein 27 signaling and lipid raft-mediated endocytosis negatively regulated by Caveolin-1. J Biol Chem. 2013;288(24):17713–17724. 10.1074/jbc.M112.445403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Koumangoye RB, Sakwe AM, Goodwin JS, Patel T, Ochieng J, Srivastava RK. Detachment of breast tumor cells induces rapid secretion of exosomes which subsequently mediate cellular adhesion and spreading. PLoS One. 2011;6(9):e24234 10.1371/journal.pone.0024234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Schneider DJ, Speth JM, Penke LR, Wettlaufer SH, Swanson JA, Peters-Golden M. Mechanisms and modulation of microvesicle uptake in a model of alveolar cell communication. J Biol Chem. 2017; 292(51):20897–20910. 10.1074/jbc.M117.792416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.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(1):10112 10.1038/srep10112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Parolini I, Federici C, Raggi C, et al. Microenvironmental pH is a key factor for exosome traffic in tumor cells. J Biol Chem. 2009;284(49): 34211–34222. 10.1074/jbc.M109.041152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Atai NA, Balaj L, van Veen H, et al. Heparin blocks transfer of extracellular vesicles between donor and recipient cells. J Neuro-Oncol. 2013;115(3):343–351. 10.1007/s11060-013-1235-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Christianson HC, Svensson KJ, van Kuppevelt TH, Li J-P, Belting M. Cancer cell exosomes depend on cell-surface heparan sulfate proteoglycans for their internalization and functional activity. Proc Natl Acad Sci. 2013;110(43):17380–17385. 10.1073/pnas.1304266110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Plebanek MP, Mutharasan RK, Volpert O, Matov A, Gatlin JC, Thaxton CS. Nanoparticle targeting and cholesterol flux through scavenger receptor type B-1 inhibits cellular exosome uptake. Sci Rep. 2015;5(1):15724 10.1038/srep15724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Sandvig K, Llorente A. Proteomic analysis of microvesicles released by the human prostate cancer cell line PC-3. Mol Cell Proteomics. 2012; 11(7):M111.012914. 10.1074/mcp.M111.012914. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Witas R, Chaput D, Khan H, Stevens SM, Kang D. Isolation and proteomic analysis of microvesicles and exosomes from HT22 cells and primary neurons. Methods Mol Biol. 2017;1598:255–267. 10.1007/978-1-4939-6952-4_12. [DOI] [PubMed] [Google Scholar]
- 96.Tissot J-D, Canellini G, Rubin O, et al. Blood microvesicles: from proteomics to physiology. Transl Proteom. 2013;1(1):38–52. 10.1016/J.TRPROT.2013.04.004. [DOI] [Google Scholar]
- 97.Kim H-S, Choi D-Y, Yun SJ, et al. Proteomic analysis of microvesicles derived from human mesenchymal stem cells. J Proteome Res. 2012; 11(2):839–849. 10.1021/pr200682z. [DOI] [PubMed] [Google Scholar]
- 98.Kumar N, Gupta DG, Kumar S, et al. Exploring packaged microvesicle proteome composition of Chinese hamster ovary secretome. J Bioprocess Biotech. 2016;6:274 10.4172/2155-9821.1000274. [DOI] [Google Scholar]
- 99.Baig S, Lim JY, Fernandis AZ, et al. Lipidomic analysis of human placental Syncytiotrophoblast microvesicles in adverse pregnancy outcomes. Placenta. 2013;34(5):436–442. 10.1016/J.PLACENTA.2013.02.004. [DOI] [PubMed] [Google Scholar]
- 100.Pienimaeki-Roemer A, Kuhlmann K, Böttcher A, et al. Lipidomic and proteomic characterization of platelet extracellular vesicle subfractions from senescent platelets. Transfusion. 2015;55(3):507–521. 10.1111/trf.12874. [DOI] [PubMed] [Google Scholar]
- 101.Yáñez-Mó M, Siljander PR-M, Andreu Z, et al. Biological properties of extracellular vesicles and their physiological functions. J Extracell Vesicles. 2015;4:27066 http://www.journalofextracellularvesicles.net/index.php/jev/article/view/27066/xml_13. Accessed May 3, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Giusti I, D’Ascenzo S, Dolo V. Microvesicles as potential ovarian cancer biomarkers. Biomed Res Int. 2013;2013:703048 10.1155/2013/703048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Lee Y, Andaloussi S EL, Wood MJA. Exosomes and microvesicles: extracellular vesicles for genetic information transfer and gene therapy. Hum Mol Genet. 2012;21(R1):R125–R134. 10.1093/hmg/dds317. [DOI] [PubMed] [Google Scholar]
- 104.Tong M, Chamley LW. Placental extracellular vesicles and feto-maternal communication. Cold Spring Harb Perspect Med. 2015; 5(3):a023028 10.1101/cshperspect.a023028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Melki I, Tessandier N, Zufferey A, Boilard E. Platelet microvesicles in health and disease. Platelets. 2017;28(3):214–221. 10.1080/09537104.2016.1265924. [DOI] [PubMed] [Google Scholar]
- 106.Loyer X, Vion A-C, Tedgui A, Boulanger CM. Microvesicles as cell-cell messengers in cardiovascular diseases. Circ Res. 2014;114(2):345–353. 10.1161/CIRCRESAHA.113.300858. [DOI] [PubMed] [Google Scholar]
- 107.Lawson C, Vicencio JM, Yellon DM, Davidson SM. Microvesicles and exosomes: new players in metabolic and cardiovascular disease. J Endocrinol. 2016;228(2):R57–R71. 10.1530/JOE-15-0201. [DOI] [PubMed] [Google Scholar]
- 108.Tomaniak M, Gąsecka A, Filipiak KJ. Cell-derived microvesicles in cardiovascular diseases and antiplatelet therapy monitoring - a lesson for future trials? Current evidence, recent progresses and perspectives of clinical application. Int J Cardiol. 2017;226:93–102. 10.1016/j.ijcard.2016.10.007. [DOI] [PubMed] [Google Scholar]
- 109.Izumi Y Therapeutical potential of microvesicles in cardiovascular diseases. J Genet Syndr Gene Ther. 2012;3(4):1–2. 10.4172/2157-7412.1000e107. [DOI] [Google Scholar]
- 110.Zaldivia MTK, McFadyen JD, Lim B, Wang X, Peter K. Platelet-derived microvesicles in cardiovascular diseases. Front Cardiovasc Med. 2017;4:74 10.3389/fcvm.2017.00074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Wang Y, Chen L, Liu M. Microvesicles and diabetic complications — novel mediators, potential biomarkers and therapeutic targets. Acta Pharmacol Sin. 2014;35(4):433–443. 10.1038/aps.2013.188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Müller G Microvesicles/exosomes as potential novel biomarkers of metabolic diseases. Diabetes Metab Syndr Obes. 2012;5:247–282. 10.2147/DMSO.S32923. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Zhang W, Chen S, Liu M-L. Pathogenic roles of microvesicles in diabetic retinopathy. Acta Pharmacol Sin. 2018;39(1):1–11. 10.1038/aps.2017.77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Thompson AG, Gray E, Heman-Ackah SM, et al. Extracellular vesicles in neurodegenerative disease — pathogenesis to biomarkers. Nat Rev Neurol. 2016;12(6):346–357. 10.1038/nrneurol.2016.68. [DOI] [PubMed] [Google Scholar]
- 115.Colombo E, Borgiani B, Verderio C, Furlan R. Microvesicles: novel biomarkers for neurological disorders. Front Physiol. 2012;3:63 10.3389/fphys.2012.00063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Janas AM, Sapoń K, Janas T, Stowell MHB, Janas T. Exosomes and other extracellular vesicles in neural cells and neurodegenerative diseases. Biochim Biophys Acta Biomembr. 2016;1858(6):1139–1151. 10.1016/J.BBAMEM.2016.02.011. [DOI] [PubMed] [Google Scholar]
- 117.Purushothaman A, Bandari SK, Liu J, Mobley JA, Brown EE, Sanderson RD. Fibronectin on the surface of myeloma cell-derived exosomes mediates exosome-cell interactions. J Biol Chem. 2016; 291(4):1652–1663. 10.1074/jbc.M115.686295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Antonyak MA, Li B, Boroughs LK, et al. Cancer cell-derived microvesicles induce transformation by transferring tissue transglutaminase and fibronectin to recipient cells. Proc Natl Acad Sci USA. 2011; 108(12):4852–4857. 10.1073/pnas.1017667108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Atay SDC, Gercel-Taylor CDD. Ovarian cancer-derived exosomal fibronectin induces pro-inflammatory IL-1beta. Exosomes Microvesicles. January 2013;1 10.5772/56180. [DOI] [Google Scholar]
- 120.Sung BH, Ketova T, Hoshino D, Zijlstra A, Weaver AM. Directional cell movement through tissues is controlled by exosome secretion. Nat Commun. 2015;6(1):7164 10.1038/ncomms8164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Sung BH, Weaver AM. Exosome secretion promotes chemotaxis of cancer cells. Cell Adhes Migr. 2017;11(2):187–195. 10.1080/19336918.2016.1273307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Moon P-G, Lee J-E, Cho Y-E, et al. Fibronectin on circulating extracellular vesicles as a liquid biopsy to detect breast cancer. Oncotarget. 2016;7(26):40189–40199. 10.18632/oncotarget.9561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Wysoczynski M, Ratajczak MZ. Lung cancer secreted microvesicles: underappreciated modulators of microenvironment in expanding tumors. Int J Cancer. 2009;125(7):1595–1603. 10.1002/ijc.24479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Al-Nedawi K, Meehan B, Micallef J, et al. Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles derived from tumour cells. Nat Cell Biol. 2008;10(5):619–624. 10.1038/ncb1725. [DOI] [PubMed] [Google Scholar]
- 125.Andreola G, Rivoltini L, Castelli C, et al. Induction of lymphocyte apoptosis by tumor cell secretion of FasL-bearing microvesicles. J Exp Med. 2002;195(10):1303–1316. 10.1084/JEM.20011624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Martínez-Lorenzo MJ, Anel A, Alava MA, et al. The human melanoma cell line MelJuSo secretes bioactive FasL and APO2L/TRAIL on the surface of microvesicles. Possible contribution to tumor counterattack. Exp Cell Res. 2004;295(2):315–329. 10.1016/j.yexcr.2003.12.024. [DOI] [PubMed] [Google Scholar]
- 127.Chen X, Liang H, Zen K, Zhang C-Y. Secreted microRNAs from tumor cells can suppress immune function. Oncoimmunology. 2016; 5(4):e982407 10.4161/2162402X.2014.982407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Li Y, An J, Huang S, He J, Zhang J. Esophageal cancer-derived microvesicles induce regulatory B cells. Cell Biochem Funct. 2015;33(5): 308–313. 10.1002/cbf.3115. [DOI] [PubMed] [Google Scholar]
- 129.Yamada N, Tsujimura N, Kumazaki M, et al. Colorectal cancer cell-derived microvesicles containing microRNA-1246 promote angiogenesis by activating Smad 1/5/8 signaling elicited by PML down-regulation in endothelial cells. Biochim Biophys Acta. 2014; 1839(11):1256–1272. 10.1016/j.bbagrm.2014.09.002. [DOI] [PubMed] [Google Scholar]
- 130.Huaitong X, Yuanyong F, Yueqin T, Peng Z, Wei S, Kai S. Microvesicles releasing by oral cancer cells enhance endothelial cell angiogenesis via Shh/RhoA signaling pathway. Cancer Biol Ther. 2017;18:1–9. 10.1080/15384047.2017.1373213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Muralidharan-Chari V, Gilzad Kohan H, Asimakopoulos AG, et al. Microvesicle removal of anticancer drugs contributes to drug resistance in human pancreatic cancer cells. Oncotarget. 2014;7(31): 50365–50379. 10.18632/oncotarget.10395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Zhang F, Zhu Y, Zhao Q, et al. Microvesicles mediate transfer of P-glycoprotein to paclitaxel-sensitive A2780 human ovarian cancer cells, conferring paclitaxel-resistance. Eur J Pharmacol. 2014;738: 83–90. 10.1016/j.ejphar.2014.05.026. [DOI] [PubMed] [Google Scholar]
- 133.Peinado H, Zhang H, Matei IR, et al. Pre-metastatic niches: organ-specific homes for metastases. Nat Rev Cancer. 2017;17(5): 302–317. 10.1038/nrc.2017.6. [DOI] [PubMed] [Google Scholar]
- 134.Grange C, Tapparo M, Collino F, et al. Microvesicles released from human renal cancer stem cells stimulate angiogenesis and formation of lung premetastatic niche. Cancer Res. 2011;71(15):5346–5356. 10.1158/0008-5472.CAN-11-0241. [DOI] [PubMed] [Google Scholar]
- 135.Castellana D, Zobairi F, Martinez MC, et al. Membrane microvesicles as actors in the establishment of a favorable prostatic tumoral niche: a role for activated fibroblasts and CX3CL1-CX3CR1 axis. Cancer Res. 2009; 69(3):785–793. 10.1158/0008-5472.CAN-08-1946. [DOI] [PubMed] [Google Scholar]
- 136.Fujita Y, Yoshioka Y, Ochiya T. Extracellular vesicle transfer of cancer pathogenic components. Cancer Sci. 2016;107(4):385–390. 10.1111/cas.12896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Jiang L, Vader P, Schiffelers RM. Extracellular vesicles for nucleic acid delivery: progress and prospects for safe RNA-based gene therapy. Gene Ther. 2017;24(3):157–166. 10.1038/gt.2017.8. [DOI] [PubMed] [Google Scholar]
- 138.Kinoshita T, Yip KW, Spence T, Liu F-F. MicroRNAs in extracellular vesicles: potential cancer biomarkers. J Hum Genet. 2017;62(1): 67–74. 10.1038/jhg.2016.87. [DOI] [PubMed] [Google Scholar]
- 139.Zhang J, Zhao A, Sun L, et al. Selective surface marker and miRNA profiles of CD34+ blast-derived microvesicles in chronic myelogenous leukemia. Oncol Lett. 2017;14(2):1866–1874. 10.3892/ol.2017.6336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Chen I-H, Xue L, Hsu C-C, et al. Phosphoproteins in extracellular vesicles as candidate markers for breast cancer. Proc Natl Acad Sci USA. 2017;114(12):3175–3180. 10.1073/pnas.1618088114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Di Noto G, Paolini L, Zendrini A, Radeghieri A, Caimi L, Ricotta D. C-src enriched serum microvesicles are generated in malignant plasma cell dyscrasia. PLoS One. 2013;8(8):e70811 10.1371/journal.pone.0070811. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Zarfati M, Katz T, Avivi I, Brenner B, Aharon A. PO-45 - the role of microvesicles in multiple myeloma progression. Thromb Res. 2016; 140:S193 10.1016/S0049-3848(16)30178-5. [DOI] [PubMed] [Google Scholar]
- 143.Baran J, Baj-Krzyworzeka M, Weglarczyk K, et al. Circulating tumour-derived microvesicles in plasma of gastric cancer patients. Cancer Immunol Immunother. 2010;59(6):841–850. 10.1007/s00262-009-0808-2. [DOI] [PMC free article] [PubMed] [Google Scholar]


